06.04.2013 Views

Sarcoplasmic Reticulum Function in Smooth Muscle - Physiological ...

Sarcoplasmic Reticulum Function in Smooth Muscle - Physiological ...

Sarcoplasmic Reticulum Function in Smooth Muscle - Physiological ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

<strong>Sarcoplasmic</strong> <strong>Reticulum</strong> <strong>Function</strong> <strong>in</strong> <strong>Smooth</strong> <strong>Muscle</strong><br />

SUSAN WRAY AND THEODOR BURDYGA<br />

Physiol Rev 90: 113–178, 2010;<br />

doi:10.1152/physrev.00018.2008.<br />

Department of Physiology, School of Biomedical Sciences, University of Liverpool, Liverpool, United K<strong>in</strong>gdom<br />

I. Introduction and Brief Historical Overview 114<br />

II. Structure and Location of <strong>Sarcoplasmic</strong> <strong>Reticulum</strong> <strong>in</strong> <strong>Smooth</strong> <strong>Muscle</strong> 116<br />

A. Introduction 116<br />

B. Electron microscopic studies 116<br />

C. Confocal imag<strong>in</strong>g of the SR 117<br />

D. SR, caveolae, microdoma<strong>in</strong>s, and superficial buffer barrier 119<br />

E. SR and the mitochondria 120<br />

F. SR and the nucleus 121<br />

G. Lysosomes 121<br />

III. Sarco/Endoplasmic <strong>Reticulum</strong> Calcium-Adenos<strong>in</strong>etriphosphatase 122<br />

A. Introduction 122<br />

B. Enzyme type and activity 122<br />

C. SERCA structure and catalytic cycle 122<br />

D. SERCA isoforms and expression <strong>in</strong> smooth muscles 123<br />

E. Regulators of SERCA expression and activity 124<br />

F. Summary 125<br />

IV. Prote<strong>in</strong>s Associated With the <strong>Sarcoplasmic</strong> <strong>Reticulum</strong> 125<br />

A. Introduction 125<br />

B. The SR membrane 125<br />

C. Phospholamban 126<br />

D. Sarcolip<strong>in</strong> 128<br />

E. Calsequestr<strong>in</strong> 128<br />

F. Calreticul<strong>in</strong> 129<br />

G. Triad<strong>in</strong> and junct<strong>in</strong> 130<br />

H. Summary 130<br />

V. Calcium Release Channels 130<br />

A. Introduction 130<br />

B. Ryanod<strong>in</strong>e receptors 130<br />

C. Ip 3-sensitive Ca release channels 132<br />

D. Modulation of SR Ca release 133<br />

E. SR Ca leak 135<br />

F. Summary 136<br />

VI. Lum<strong>in</strong>al Calcium 136<br />

A. Introduction 136<br />

B. Fluorescent <strong>in</strong>dicators for SR lum<strong>in</strong>al Ca measurement 136<br />

C. Lum<strong>in</strong>al Ca buffers 137<br />

D. Measurements 137<br />

E. Effects of SR Ca load 138<br />

F. Conclusions 139<br />

VII. Calcium Reuptake Mechanisms 139<br />

A. Introduction 139<br />

B. SERCA uptake of Ca and Ca homeostasis 139<br />

C. SERCA and Ca signal<strong>in</strong>g and contractility 139<br />

D. SERCA Ca efflux and relaxation 140<br />

E. Store-operated Ca entry 140<br />

VIII. Common or Separate Calcium Pools 144<br />

A. Introduction 144<br />

B. One store with one type of receptor 144<br />

C. One store with two types of receptors 146<br />

D. Two stores, two types of receptors 146<br />

E. Role of Ca release channels and store type <strong>in</strong> Ca signal<strong>in</strong>g 146<br />

www.prv.org 0031-9333/10 $18.00 Copyright © 2010 the American <strong>Physiological</strong> Society<br />

113


114 SUSAN WRAY AND THEODOR BURDYGA<br />

F. Summary 148<br />

IX. Elemental Calcium Signals From <strong>Smooth</strong> <strong>Muscle</strong> <strong>Sarcoplasmic</strong> <strong>Reticulum</strong> 148<br />

A. Ca sparks 148<br />

B. Ca puffs 152<br />

X. <strong>Sarcoplasmic</strong> <strong>Reticulum</strong> and Plasmalemmal Ion Channels 153<br />

A. Overview of Ca-activated ion channels 153<br />

B. BK channels 153<br />

C. SK and IK channels 154<br />

D. Ca-activated Cl channels 155<br />

XI. <strong>Sarcoplasmic</strong> <strong>Reticulum</strong> and Development and Ag<strong>in</strong>g 156<br />

XII. Gender 156<br />

XIII. pH, Metabolism, and <strong>Sarcoplasmic</strong> <strong>Reticulum</strong> <strong>Function</strong> 157<br />

A. Introduction 157<br />

B. pH 157<br />

C. Metabolism 158<br />

XIV. Mathematical Model<strong>in</strong>g 158<br />

XV. Conclusions and Future Directions 158<br />

Wray S, Burdyga T. <strong>Sarcoplasmic</strong> <strong>Reticulum</strong> <strong>Function</strong> <strong>in</strong> <strong>Smooth</strong> <strong>Muscle</strong>. Physiol Rev 90: 113–178, 2010;<br />

doi:10.1152/physrev.00018.2008.—The sarcoplasmic reticulum (SR) of smooth muscles presents many <strong>in</strong>trigu<strong>in</strong>g<br />

facets and questions concern<strong>in</strong>g its roles, especially as these change with development, disease, and modulation of<br />

physiological activity. The SR’s function was orig<strong>in</strong>ally perceived to be synthetic and then that of a Ca store for the<br />

contractile prote<strong>in</strong>s, act<strong>in</strong>g as a Ca amplification mechanism as it does <strong>in</strong> striated muscles. Gradually, as <strong>in</strong>vestigators<br />

have struggled to f<strong>in</strong>d a conv<strong>in</strong>c<strong>in</strong>g role for Ca-<strong>in</strong>duced Ca release <strong>in</strong> many smooth muscles, a role <strong>in</strong> controll<strong>in</strong>g<br />

excitability has emerged. This is the Ca spark/spontaneous transient outward current coupl<strong>in</strong>g mechanism which<br />

reduces excitability and limits contraction. Release of SR Ca occurs <strong>in</strong> response to <strong>in</strong>ositol 1,4,5-trisphosphate, Ca,<br />

and nicot<strong>in</strong>ic acid aden<strong>in</strong>e d<strong>in</strong>ucleotide phosphate, and depletion of SR Ca can <strong>in</strong>itiate Ca entry, the mechanism of<br />

which is be<strong>in</strong>g <strong>in</strong>vestigated but seems to <strong>in</strong>volve Stim and Orai as found <strong>in</strong> nonexcitable cells. The contribution of<br />

the elemental Ca signals from the SR, sparks and puffs, to global Ca signals, i.e., Ca waves and oscillations, is<br />

becom<strong>in</strong>g clearer but is far from established. The dynamics of SR Ca release and uptake mechanisms are reviewed<br />

along with the control of lum<strong>in</strong>al Ca. We review the grow<strong>in</strong>g list of the SR’s functions that still <strong>in</strong>cludes Ca storage,<br />

contraction, and relaxation but has been expanded to encompass Ca homeostasis, generat<strong>in</strong>g local and global Ca<br />

signals, and contribut<strong>in</strong>g to cellular microdoma<strong>in</strong>s and signal<strong>in</strong>g <strong>in</strong> other organelles, <strong>in</strong>clud<strong>in</strong>g mitochondria,<br />

lysosomes, and the nucleus. For an <strong>in</strong>tegrated approach, a review of aspects of the SR <strong>in</strong> health and disease and<br />

dur<strong>in</strong>g development and ag<strong>in</strong>g are also <strong>in</strong>cluded. While the sheer versatility of smooth muscle makes it foolish to<br />

have a “one model fits all” approach to this subject, we have tried to synthesize conclusions wherever possible.<br />

I. INTRODUCTION AND BRIEF<br />

HISTORICAL OVERVIEW<br />

For an excellent general historical overview of the<br />

sarcoplasmic reticulum (SR) and endoplasmic reticulum<br />

(ER), the recent review by Verkhratsky (728) can be<br />

consulted. For other general references to the SR (or ER),<br />

Ca homeostasis and SR/ER Ca-ATPase (SERCA), the follow<strong>in</strong>g<br />

reviews are recommended: Laporte et al. (384),<br />

Pozzan et al. (573), Karaki et al. (336), Strehler and<br />

Treiman (665), Floyd and Wray (184), Rossi and Dirksen<br />

(597), Carafoli and Br<strong>in</strong>i (106), Carafoli (105), and Endo<br />

(167).<br />

An <strong>in</strong>ternal store of Ca <strong>in</strong> smooth muscle cells was<br />

postulated follow<strong>in</strong>g demonstrations of Ca rema<strong>in</strong><strong>in</strong>g <strong>in</strong><br />

the cells after immersion <strong>in</strong> Ca-free solutions. This followed<br />

older observations that contractile responses,<br />

which were known to be Ca dependent, could cont<strong>in</strong>ue<br />

for vary<strong>in</strong>g periods <strong>in</strong> Ca-free solutions, and that if external<br />

[Ca] had been elevated before the switch to Ca-free<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

solution, this period was extended (278). Although we<br />

now have a more sophisticated view of Ca handl<strong>in</strong>g and<br />

Ca sensitivity (e.g., Refs. 13, 201, 271, 612, 769), which<br />

could also account for contraction <strong>in</strong> Ca-free solutions,<br />

the conclusion reached, i.e., there must be an <strong>in</strong>ternal Ca<br />

store, was correct. Electron microscopy (EM) of smooth<br />

muscle revealed a membranous system of tubules and<br />

sacs, the SR, that was both close to the periphery and<br />

caveolae as well as deep with<strong>in</strong> the cell. Close apposition<br />

of the SR to mitochondria and the nucleus was also noted.<br />

This membrane system excluded extracellular markers<br />

such as ferrit<strong>in</strong> and horseradish peroxidase, but was contiguous<br />

with the lumen of the nuclear envelope. Although<br />

orig<strong>in</strong>ally described as sparse, follow<strong>in</strong>g improvements <strong>in</strong><br />

fixation and microscopy, these earliest accounts were<br />

replaced with terms such as “well developed” and “rich<br />

reticulum.” It is now appreciated that the SR’s distribution,<br />

amount, and shape is not only smooth muscle specific,<br />

but also changes with physiological stimuli and developmental<br />

stage and <strong>in</strong> disease.


One of the earliest suggestions that the SR <strong>in</strong> smooth<br />

muscle was the major <strong>in</strong>tracellular Ca store and that it<br />

acts as <strong>in</strong> striated muscle was made by Giorgio Gabella<br />

and was based on EM studies (194–196). In the same year,<br />

Avril and Andrew Somlyo demonstrated that strontium,<br />

used as a surrogate for Ca, was accumulated <strong>in</strong> the SR of<br />

smooth muscle (aorta and pulmonary artery), and therefore,<br />

it followed that the SR could store Ca and be an<br />

activator of contraction (656). Many further important<br />

contributions were made by the Somlyos, <strong>in</strong>clud<strong>in</strong>g the<br />

demonstration us<strong>in</strong>g electron-probe X-ray microanalysis<br />

that [Ca] decreased <strong>in</strong> the SR follow<strong>in</strong>g agonist stimulation<br />

(e.g., Refs. 95, 371) and that Ca cycl<strong>in</strong>g to and from<br />

the SR occurred dur<strong>in</strong>g contraction and relaxation (69).<br />

The demonstration of a rich reticular formation <strong>in</strong><br />

smooth muscle renewed <strong>in</strong>terest <strong>in</strong> exam<strong>in</strong>ation of the<br />

role of the SR <strong>in</strong> excitation-contraction (E-C) coupl<strong>in</strong>g.<br />

Much of this work was look<strong>in</strong>g for or assum<strong>in</strong>g similarity<br />

of mechanisms to those of striated muscle. It is only <strong>in</strong> the<br />

last decade that the dist<strong>in</strong>ct role of the SR <strong>in</strong> smooth<br />

SMOOTH MUSCLE SR 115<br />

muscle has been elucidated. Work from Nelson’s group<br />

(70, 306, 507) and others (76, 682) showed that local Ca<br />

release from the SR can control plasma membrane excitability.<br />

We built on this and showed that there is a fundamental<br />

relationship between SR Ca release and the<br />

action potential <strong>in</strong> ureteric smooth muscle (87).<br />

We now appreciate that the SR can have the follow<strong>in</strong>g<br />

roles as demonstrated <strong>in</strong> Figure 1: 1) contribut<strong>in</strong>g to<br />

Ca homeostasis by ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g low rest<strong>in</strong>g [Ca], via action<br />

of the SERCA; 2) restoration of [Ca] and relaxation<br />

follow<strong>in</strong>g stimulation, via the action of SERCA; 3) augment<strong>in</strong>g<br />

contraction through release of Ca and produc<strong>in</strong>g<br />

or modify<strong>in</strong>g global Ca signals <strong>in</strong> response to agonist<br />

stimulation; 4) modulat<strong>in</strong>g membrane excitability through<br />

activation of Ca-sensitive ion channels; 5) contribut<strong>in</strong>g to<br />

the efficacy of plasma membrane Ca extrusion mechanisms<br />

by vectorially releas<strong>in</strong>g Ca to them; 6) contribut<strong>in</strong>g to the<br />

ma<strong>in</strong>tenance of signal<strong>in</strong>g microdoma<strong>in</strong>s around the plasma<br />

membrane and other organelles; and 7) <strong>in</strong>fluenc<strong>in</strong>g development,<br />

ag<strong>in</strong>g, and the health of smooth muscle tissues.<br />

FIG. 1. <strong>Function</strong>s of the SR <strong>in</strong> smooth muscle. A cartoon to demonstrate the functions of the SR. 1: Contribut<strong>in</strong>g to Ca homeostasis and<br />

ma<strong>in</strong>tenance of low rest<strong>in</strong>g level of <strong>in</strong>tracellular [Ca] via sarco/endoplasmic reticulum Ca-ATPase (SERCA) activity. 2: Contribut<strong>in</strong>g to relaxation<br />

of the smooth muscle cell by tak<strong>in</strong>g up Ca via SERCA. 3: Contribut<strong>in</strong>g to Ca signals and contraction by Ca release via IP 3 receptor (IP 3R)- or<br />

ryanod<strong>in</strong>e receptor (RyR)-gated Ca release channels and Ca puffs and Ca sparks. 4: Contribut<strong>in</strong>g to excitability via Ca-activated K and Cl channels.<br />

5: Facilitation of Ca efflux on plasma membrane Ca-ATPase (Ca pump) or Na/Ca exchanger, via vectorial release of Ca. 6: Contribut<strong>in</strong>g to subcellular<br />

microdoma<strong>in</strong>s and organellar Ca homeostasis via Ca uptake and release between organelles. 7: Correct SR function needed for normal development<br />

and health, and SR functional change is associated with disease and ag<strong>in</strong>g.<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org


116 SUSAN WRAY AND THEODOR BURDYGA<br />

This is an extensive job description for an organelle only<br />

occupy<strong>in</strong>g 5% of the cell. We highlight these physiological<br />

and pathological functions <strong>in</strong>ter alia.<br />

Our approach <strong>in</strong> this review has been to synthesize a<br />

holistic view of how the SR works <strong>in</strong> smooth muscle and<br />

then discuss how these components are put together <strong>in</strong><br />

different smooth muscles lead<strong>in</strong>g to tissue specificity.<br />

There is a daunt<strong>in</strong>g amount of literature <strong>in</strong> some areas,<br />

not all of which can be cited, and for this we apologize.<br />

There are areas of the literature where clarity is slowly<br />

emerg<strong>in</strong>g and others where unfortunately it rema<strong>in</strong>s elusive.<br />

Thus the conclusions we make to clarify, summarize,<br />

and hopefully provide <strong>in</strong>sight will <strong>in</strong>clude a degree of<br />

speculation and author bias, and perhaps not all will stand<br />

the test of time. We offer them as discussion po<strong>in</strong>ts <strong>in</strong> the<br />

quest of understand<strong>in</strong>g SR function <strong>in</strong> smooth muscle. We<br />

have also focused wherever possible on literature obta<strong>in</strong>ed<br />

on <strong>in</strong>tact tissues, <strong>in</strong>tact muscle strips, and native<br />

smooth muscle cells, i.e., freshly dissociated and isolated.<br />

The effects of cell cultur<strong>in</strong>g on almost every aspect of<br />

smooth muscle E-C are so well known and long stand<strong>in</strong>g<br />

that <strong>in</strong>vestigators ignore them at their peril (116, 533).<br />

II. STRUCTURE AND LOCATION OF<br />

SARCOPLASMIC RETICULUM IN<br />

SMOOTH MUSCLE<br />

A. Introduction<br />

Although it may sound out-moded to be concerned<br />

with the structure and location of the SR, recent work, for<br />

example, on membrane microdoma<strong>in</strong>s and SR compartmentalization,<br />

make it important that we have a clear<br />

factual understand<strong>in</strong>g of SR structure and distribution, to<br />

better judge what is feasible both with<strong>in</strong> the SR and <strong>in</strong> its<br />

relation to other organelles and the plasma membrane.<br />

B. Electron Microscopic Studies<br />

Much of our structural <strong>in</strong>formation concern<strong>in</strong>g smooth<br />

muscle has come from EM studies. These studies addressed<br />

the size of the SR, usually compared with striated<br />

muscle and <strong>in</strong> terms of cell volume, and SR location<br />

with<strong>in</strong> myocytes. By reveal<strong>in</strong>g these features EM made a<br />

significant contribution not just to the understand<strong>in</strong>g of<br />

SR structure, but also to E-C coupl<strong>in</strong>g <strong>in</strong> smooth muscle.<br />

Before fluorescent probes sensitive to ion concentration<br />

were developed, electron X-ray microprobe analysis provided<br />

spatial <strong>in</strong>formation about the elemental composition<br />

of different areas <strong>in</strong> the myocytes, <strong>in</strong> relaxed and<br />

contracted stages. Determ<strong>in</strong>ations of SR Ca content are<br />

considered <strong>in</strong> section VI.<br />

Consistent between different workers and smooth<br />

muscles is the location of the SR: close to, but not touch-<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

<strong>in</strong>g the plasma membrane. The SR was noted to encircle<br />

the <strong>in</strong>vag<strong>in</strong>ations of the surface membrane, the caveolae.<br />

Sheets of reticulum runn<strong>in</strong>g along the longitud<strong>in</strong>al axis of<br />

the cell are a feature of all the tissues exam<strong>in</strong>ed, e.g.,<br />

vascular (152), airway (375), gastro<strong>in</strong>test<strong>in</strong>al tract and<br />

bladder (197), human blood vessels (677), develop<strong>in</strong>g<br />

blood vessels (693), and portal ve<strong>in</strong> (69). The SR is described<br />

as peripheral (or superficial) when it is close to<br />

the plasma membrane and central (or deep) when it is<br />

located away from this membrane. The peripheral SR has<br />

been associated with Ca homeostasis, local Ca release,<br />

and <strong>in</strong>teractions with plasma membrane ion channels and<br />

hence excitability, whereas the central SR has been suggested<br />

to be more directly <strong>in</strong>volved <strong>in</strong> contraction by<br />

supply<strong>in</strong>g Ca to the myofilaments. This is discussed further<br />

<strong>in</strong> section V.<br />

Given the different activity and mechanisms of excitation<br />

between phasic and tonic smooth muscles, differences<br />

<strong>in</strong> the amount and localization of the SR have been<br />

sought. In vas deferens, a phasic muscle, the SR at EM<br />

level was described as be<strong>in</strong>g located predom<strong>in</strong>antly at the<br />

periphery, whereas the tonic aorta had proportionally<br />

more SR <strong>in</strong> a central location (515). This paper also noted<br />

a central distribution for the tonic pulmonary artery and<br />

trachealis.<br />

The smooth muscle cell’s <strong>in</strong>terior is packed with myofilaments<br />

and <strong>in</strong>termediate filaments. It is estimated that<br />

they, along with associated dense bodies, occupy up to 90%<br />

of the cell’s volume. The <strong>in</strong>tracellular organelles share the<br />

rema<strong>in</strong><strong>in</strong>g volume (195). Although the earliest EM accounts<br />

described the SR of smooth muscles as be<strong>in</strong>g poorly developed<br />

(e.g., Refs. 194, 548, 577), this view gradually changed<br />

and is borne out by recent studies us<strong>in</strong>g confocal microscopy<br />

(described later). In one of the best comparative studies,<br />

Dev<strong>in</strong>e et al. (152) compared rabbit smooth muscles and<br />

reported that 5% of the cell volume was accounted for by<br />

the SR <strong>in</strong> aorta and pulmonary artery and that this fell to<br />

2% <strong>in</strong> mesenteric ve<strong>in</strong> and artery and portal ve<strong>in</strong>. These<br />

f<strong>in</strong>d<strong>in</strong>gs also correlated with the vary<strong>in</strong>g ability of these<br />

preparations to cont<strong>in</strong>ue contract<strong>in</strong>g <strong>in</strong> zero-Ca solutions,<br />

i.e., longer <strong>in</strong> aorta than mesenteric and portal ve<strong>in</strong>. However,<br />

when low values for SR volume were reported <strong>in</strong> some<br />

(vas deferens and taenia coli, 1.7%) but not all (uterus, 7%)<br />

(596) phasic smooth muscles, the correlation with contraction<br />

time <strong>in</strong> zero Ca fell away. Somlyo’s group po<strong>in</strong>ted out<br />

that osmium fixation can damage smooth muscle SR and<br />

that this could have contributed to reports of be<strong>in</strong>g sparse<br />

(652). Thus the view gradually changed from “However, we<br />

must bear <strong>in</strong> m<strong>in</strong>d that a sarcoplasmic reticulum, as we<br />

understand the structure from skeletal muscle work, is def<strong>in</strong>itely<br />

not present <strong>in</strong> smooth muscles, and it seems most<br />

unlikely that the m<strong>in</strong>ute fraction of reticulum present <strong>in</strong><br />

some smooth muscles is capable of modulat<strong>in</strong>g calcium<br />

levels” (275) to “Our major conclusion is that there is <strong>in</strong><br />

mammalian smooth muscles a sarcoplasmic reticulum that,


while be<strong>in</strong>g variable <strong>in</strong> different smooth muscles, is sufficiently<br />

well developed and organised.......tofunction as a<br />

Ca store <strong>in</strong> the process of excitation-contraction and <strong>in</strong>hibition-relaxation<br />

coupl<strong>in</strong>g” (152).<br />

Notwithstand<strong>in</strong>g the fact that smooth muscle SR is<br />

well developed, its volume is less than that of striated<br />

muscles (10% of cell volume). Therefore, there are reasonable<br />

grounds to question whether the SR <strong>in</strong> smooth<br />

muscle can have the same role as <strong>in</strong> striated muscles,<br />

particularly with respect to it be<strong>in</strong>g the amplifier of a<br />

small Ca entry signal to provide the necessary rise of [Ca]<br />

required for contractile activation by Ca-<strong>in</strong>duced Ca release<br />

(CICR). This question<strong>in</strong>g is also suggested by the<br />

generally smaller SR volume <strong>in</strong> visceral smooth muscles,<br />

where Ca entry is predom<strong>in</strong>ant, compared with vascular<br />

smooth muscles, where SR Ca release is more prom<strong>in</strong>ent.<br />

It seems to us that the pendulum swung from <strong>in</strong>itially<br />

dismiss<strong>in</strong>g the SR’s role <strong>in</strong> modulat<strong>in</strong>g Ca signals<br />

and CICR <strong>in</strong> smooth muscle to view<strong>in</strong>g the SR as play<strong>in</strong>g<br />

a very similar role to that occurr<strong>in</strong>g <strong>in</strong> striated<br />

muscles. We are perhaps now able to readjust the<br />

pendulum and see more clearly that smooth muscle SR<br />

is important to E-C coupl<strong>in</strong>g, but <strong>in</strong> its own dist<strong>in</strong>ctive<br />

way.<br />

C. Confocal Imag<strong>in</strong>g of the SR<br />

Confocal imag<strong>in</strong>g can show the extent and three-dimensional<br />

distribution of the SR <strong>in</strong> smooth muscle and identify<br />

Ca release and uptake sites, as well as allow<strong>in</strong>g changes <strong>in</strong><br />

its Ca content to be determ<strong>in</strong>ed. Follow<strong>in</strong>g from the development<br />

of Ca-sensitive fluorescent <strong>in</strong>dicators to monitor<br />

cytoplasmic [Ca] changes, efforts were directed at develop<strong>in</strong>g<br />

similar probes for <strong>in</strong>tracellular organelles. One of the<br />

earliest methods was to use dicarbocyan<strong>in</strong>e dye, DiOC6.<br />

This has been used <strong>in</strong> a variety of cell types as a label for<br />

SR/ER (696). Although some authors have noted its specificity<br />

for the ER (605), others considered that DiOC6 probably<br />

labels other <strong>in</strong>tracellular membranes (696). Another<br />

approach to SR imag<strong>in</strong>g is to target specific components<br />

associated with the SR membrane us<strong>in</strong>g antibodies, e.g.,<br />

aga<strong>in</strong>st ryanod<strong>in</strong>e receptors (RyRs), <strong>in</strong>ositol 1,4,5-trisphosphate<br />

(IP 3) receptors (IP 3Rs), or SERCA pumps. The Ca with<strong>in</strong><br />

the SR can also be targeted by fluorescent <strong>in</strong>dicators. If<br />

these can be preferentially loaded <strong>in</strong>to the SR lumen (as<br />

opposed to other organelles and/or cytoplasm), then<br />

these too can provide a picture of the SR, assum<strong>in</strong>g Ca is<br />

not compartmentalized with<strong>in</strong> the lumen. Another useful<br />

range of markers for the SR/ER has been developed<br />

from ceramide analogs bear<strong>in</strong>g the fluorophore boron<br />

dipyrromethene difluoride (BODIPY) (540). To confer<br />

SR organellar specificity, this backbone is added to<br />

fluorescent ryanod<strong>in</strong>e or thapsigarg<strong>in</strong> (an <strong>in</strong>hibitor of<br />

SERCA) (635).<br />

SMOOTH MUSCLE SR 117<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

Techniques of molecular biology to specifically target<br />

the SR prote<strong>in</strong>s and fuse them with fluorescent prote<strong>in</strong>s<br />

such as aequor<strong>in</strong> can also be used to image the SR,<br />

although these are rarely suitable or successful <strong>in</strong> smooth<br />

muscle cells. Unlike fluorescent <strong>in</strong>dicators that can be<br />

used directly <strong>in</strong> <strong>in</strong>tact tissues or s<strong>in</strong>gle cells, us<strong>in</strong>g genetically<br />

manufactured SR-targeted probes requires transfection<br />

of the recomb<strong>in</strong>ant prote<strong>in</strong>s <strong>in</strong> the cells, and hence<br />

cultur<strong>in</strong>g. This poses two problems for smooth muscle<br />

studies. First, the well-appreciated problems of phenotypic<br />

change that occur with even short periods of cultur<strong>in</strong>g.<br />

Second, conventional transfection has proven difficult<br />

<strong>in</strong> most smooth muscles. However, methods based<br />

on organ-cultur<strong>in</strong>g and nonviral delivery, such as reversible<br />

permeabilization, get around both these difficulties.<br />

They have been shown to work well <strong>in</strong> some smooth<br />

muscles, to <strong>in</strong>troduce plasmids, antisense oligonucleotides,<br />

and small <strong>in</strong>terfer<strong>in</strong>g RNAs (siRNA), e.g., focal adhesion<br />

k<strong>in</strong>ase depletion with antisense <strong>in</strong>tracheal myocytes<br />

(690), calpon<strong>in</strong> depletion with antisense <strong>in</strong> aorta<br />

(311), Rho knockdown <strong>in</strong> cerebral arteries (131), and<br />

siRNA aga<strong>in</strong>st arterial TASK-1 K channel (234). These<br />

methods have added to our understand<strong>in</strong>g of the complex<br />

signal<strong>in</strong>g pathways <strong>in</strong> smooth muscle, e.g., downregulation<br />

of profil<strong>in</strong> with antisense oligonucleotides was shown<br />

to <strong>in</strong>hibit force <strong>in</strong> carotid smooth muscle (691), and transfection<br />

with plasmids encod<strong>in</strong>g a paxill<strong>in</strong> mutant was<br />

employed to explore its role <strong>in</strong> tension development <strong>in</strong><br />

tracheal myocytes (529). However, these and related<br />

methods have been little used to target the SR and its<br />

regulators. There appears to be only one study, which<br />

used adenoviral vectors and target<strong>in</strong>g of apoaequor<strong>in</strong> <strong>in</strong><br />

native tail artery cells (588). This paper should also be<br />

consulted for an <strong>in</strong>formed discussion and evaluation of<br />

methods. The use of fluorescent probes to measure lum<strong>in</strong>al<br />

SR content is discussed <strong>in</strong> section VIB.<br />

Several studies us<strong>in</strong>g fluorescent BODIPY l<strong>in</strong>ked to<br />

ryanod<strong>in</strong>e or thapsigarg<strong>in</strong> and DiOC6 have been used <strong>in</strong><br />

native smooth muscle: stomach (753), portal ve<strong>in</strong> (221),<br />

and uterus (635, 636). Recently, the value of BODIPY<br />

ryanod<strong>in</strong>e has been questioned as it may not block RyR<br />

activity, perhaps due to the presence of the fluorophore<br />

(231, 455). Us<strong>in</strong>g fluorescently labeled thapsigarg<strong>in</strong> and<br />

ryanod<strong>in</strong>e <strong>in</strong> uter<strong>in</strong>e myocytes, we found that both<br />

were distributed nonhomogeneously, but <strong>in</strong> the same<br />

pattern; there was abundant label<strong>in</strong>g around the nucleus<br />

and peripherally. The close apposition of the SR<br />

to plasma membrane leads to the cell shape be<strong>in</strong>g<br />

clearly outl<strong>in</strong>ed.<br />

Figure 2 shows examples of the SR <strong>in</strong> live smooth<br />

muscle cells, imaged us<strong>in</strong>g a variety of techniques.<br />

The SR <strong>in</strong> uter<strong>in</strong>e myocytes can be loaded with the<br />

low-aff<strong>in</strong>ity Ca <strong>in</strong>dicator mag-fluo 4. With the use of this<br />

area of <strong>in</strong>creased [Ca], “hot spots” can be seen. This<br />

distribution was similar to that of SERCA and RyRs


118 SUSAN WRAY AND THEODOR BURDYGA<br />

FIG. 2. Morphological and functional demonstration of the SR <strong>in</strong> smooth muscle. A and B: <strong>in</strong> freshly isolated smooth muscle cells from gu<strong>in</strong>ea<br />

pig ureter, 3-dimensional live sta<strong>in</strong><strong>in</strong>g of the SR with BODIPY FL-X ryanod<strong>in</strong>e (A) and 2-dimensional pseudo-color image of the SR loaded with<br />

Mag-fluo 4 <strong>in</strong> a -esc<strong>in</strong> chemically sk<strong>in</strong>ned myocyte (B). C and D: pseudo-color images of fluo 4-loaded myocyte show<strong>in</strong>g a spontaneous Ca spark<br />

(C) and a Ca wave (D) <strong>in</strong>duced by 10 mM caffe<strong>in</strong>e. Images were obta<strong>in</strong>ed on Ultraview confocal system with Hamamatsu CCD camera. (From T.<br />

Burdyga, L. Borisova, and S. Wray, unpublished work.)<br />

(635). With the use of three-dimensional image reconstruction<br />

techniques, a network of <strong>in</strong>terconnect<strong>in</strong>g, spirally<br />

shaped tubules, ma<strong>in</strong>ly runn<strong>in</strong>g along the longitud<strong>in</strong>al<br />

axis of the cell, can be seen. This is rem<strong>in</strong>iscent of<br />

earlier descriptions <strong>in</strong> fixed tissue with EM. It is notable<br />

that when uter<strong>in</strong>e cells <strong>in</strong> culture were studied with<br />

antibodies to IP 3Rs and RyRs, a different distribution<br />

was found; they had an homogeneous (diffuse) distribution<br />

and hot spots detected by fluo 3FF, were scattered<br />

throughout the cytoplasm (792). In stomach myocytes<br />

from Bufo mar<strong>in</strong>us, Steenbergen and Fay (661)<br />

used mag-fura 2 and found the SR to be distributed <strong>in</strong> a<br />

punctate manner, but aga<strong>in</strong> with a concentration peripherally<br />

and around the nucleus. Other studies describ<strong>in</strong>g<br />

more or less similar structures, i.e., <strong>in</strong>terlac<strong>in</strong>g<br />

tubules, sacs, and cisternae, and location of the SR,<br />

<strong>in</strong>clude cultured A7r5 (706), mesenteric artery (214),<br />

and portal ve<strong>in</strong> (221). It appears that when different<br />

fluorescent <strong>in</strong>dicators are used, the same pattern of SR<br />

distribution <strong>in</strong> smooth muscle myocytes is observed.<br />

As found with EM studies, these images of the SR<br />

show it to be abundant around mitochondria and cont<strong>in</strong>uous<br />

with the nucleus (221, 635). There may be more<br />

SR <strong>in</strong> a peripheral location <strong>in</strong> phasic muscles (portal<br />

ve<strong>in</strong> and uterus), compared with mesenteric artery and<br />

A7r5 cells, aga<strong>in</strong> consistent with suggestions from EM<br />

studies. Immunosta<strong>in</strong>s and immuno-EM of RyRs <strong>in</strong> vas<br />

deferens and ur<strong>in</strong>ary bladder myocytes also revealed a<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

largely discrete, peripheral distribution for the SR, assum<strong>in</strong>g<br />

that the RyRs represent the entire SR (394, 525).<br />

Ohi et al. (525) also noted colocalization with largeconductance,<br />

Ca-activated K (BK) channels, discussed<br />

further <strong>in</strong> section XB. In a study of aortic SR, Lesh et al.<br />

(394) found a patchy distribution of RyR throughout the<br />

cell but absent from the nuclear region. The occurrence<br />

of frequent discharge sites or hot spots on the SR and<br />

their relation to Ca release events are discussed <strong>in</strong><br />

section IX.<br />

In summary, recent data obta<strong>in</strong>ed by live sta<strong>in</strong><strong>in</strong>g of<br />

native smooth muscle cells have added confidence to<br />

earlier data on SR structure and distribution obta<strong>in</strong>ed<br />

with EM. There is no certa<strong>in</strong>ty about what the dist<strong>in</strong>ction<br />

between peripheral and central SR means <strong>in</strong> terms of SR<br />

components or functional consequences for any particular<br />

smooth muscle. Perhaps because of the considerable<br />

<strong>in</strong>terest <strong>in</strong> local Ca signals from the SR and their relation<br />

to plasma membrane ion channels, the literature<br />

tends to emphasize the peripheral SR, and few studies<br />

have addressed the role of central SR. All phasic<br />

smooth muscles exam<strong>in</strong>ed with confocal techniques<br />

appear to have predom<strong>in</strong>antly peripheral SR. As well as<br />

the <strong>in</strong>teractions between the SR and plasma membrane<br />

and caveolae, there is also <strong>in</strong>terest <strong>in</strong> elucidat<strong>in</strong>g the<br />

functional importance for smooth muscles of hav<strong>in</strong>g<br />

their SR envelop<strong>in</strong>g mitochondria and appear<strong>in</strong>g con-


t<strong>in</strong>uous with the nuclear envelope, and these aspects<br />

are discussed next.<br />

D. SR, Caveolae, Microdoma<strong>in</strong>s, and Superficial<br />

Buffer Barrier<br />

1. Introduction<br />

An excellent review of Ca microdoma<strong>in</strong>s <strong>in</strong> smooth<br />

muscle was provided recently by McCarron et al. (451).<br />

This topic <strong>in</strong> other cell types has also been recently<br />

reviewed by Rizzuto and Pozzan (592).<br />

Many f<strong>in</strong>e structural studies have noted the close<br />

(15–30 nm) apposition of the SR to the plasma membrane,<br />

particularly around caveolae (565, 677). The close contact<br />

between caveolae and the SR membrane (195, 197) has<br />

led some <strong>in</strong>vestigators to consider if caveolae also possess<br />

Ca release mechanisms, i.e., IP 3R or RyRs. In mouse<br />

ileal smooth muscle, Fujimoto et al. (191) reported IP 3<br />

type 1 receptors on caveolae. This observation has, however,<br />

never been confirmed to the best of our knowledge.<br />

It has been reported that there is a regularity <strong>in</strong> the close<br />

apposition of the two membranes (565), lead<strong>in</strong>g to the<br />

suggestion that there may be a structural anchor between<br />

the two (688). However, it is generally concluded from<br />

EM studies that the foot structures composed of clusters<br />

of RyR reach<strong>in</strong>g to the plasma membrane and found <strong>in</strong><br />

skeletal muscles are not a feature of smooth muscle (652),<br />

although a case for this has been made by some (490). No<br />

structural rearrangements of the plasma-SR membranes<br />

have been observed with stimulation (although this has<br />

been reported for SR-mitochondrial junctions, see sect.<br />

IIE; Ref. 140).<br />

2. Caveolae<br />

That the membrane <strong>in</strong>vag<strong>in</strong>ations that are caveolae<br />

approach very close to the SR membrane rema<strong>in</strong>ed<br />

largely a structural note of little <strong>in</strong>terest to physiologists,<br />

until it became clear that caveolae were specialized regions<br />

of the plasma membrane and could facilitate cell<br />

signal<strong>in</strong>g and transduction pathways (85). It is now appreciated<br />

that the prote<strong>in</strong> caveol<strong>in</strong> is responsible for the<br />

<strong>in</strong>vag<strong>in</strong>ation of the surface membrane and that caveolae<br />

are a form of membrane lipid rafts. Lipid rafts, rich <strong>in</strong><br />

cholesterol (and sph<strong>in</strong>gomyel<strong>in</strong>), are less fluid than surround<strong>in</strong>g<br />

areas (hence “raft”) and have certa<strong>in</strong> prote<strong>in</strong>s<br />

concentrated or excluded from them, <strong>in</strong> a dynamic fashion.<br />

For a recent review of caveolae <strong>in</strong> smooth muscle,<br />

see References 298 and 520. That these biochemical specializations<br />

of the raft/caveolar membrane are functionally<br />

important has now been shown <strong>in</strong> several smooth<br />

muscles by disrupt<strong>in</strong>g caveolae, usually by extract<strong>in</strong>g cholesterol<br />

[rat uterus (648) and ureter (26), human myometrium<br />

(798), blood vessels (155), and bladder (273)]. A l<strong>in</strong>k<br />

SMOOTH MUSCLE SR 119<br />

with obesity, through elevated cholesterol and other<br />

smooth muscle pathologies, has recently been demonstrated<br />

(186, 797).<br />

A variety of techniques have shown that BK channels<br />

localize <strong>in</strong> caveolae (26, 80, 634). As will be discussed <strong>in</strong><br />

section XB, BK channels are targeted by local SR Ca<br />

release, Ca sparks, and their activation can cause hyperpolarization<br />

and therefore changes <strong>in</strong> excitability <strong>in</strong><br />

smooth muscles. Thus any disruption of the close l<strong>in</strong>k<br />

between the SR and caveolae would be expected to<br />

have functional significance (406). The Na-K-ATPase<br />

(Na pump) is also preferentially located <strong>in</strong> caveolae<br />

(491). As this has profound effects on sett<strong>in</strong>g ionic<br />

gradients for Na and K and thereby also membrane<br />

potential (11), anyth<strong>in</strong>g which modulates its activity<br />

will be expected to <strong>in</strong>fluence smooth muscle excitability<br />

(e.g., Refs. 56, 183, 399). Recently, it has been shown<br />

that the 2 isoform may directly regulate Ca transport<br />

and signal<strong>in</strong>g, due to its spatial proximity to the Na/Ca<br />

exchanger (NCX) (184, 326, 445). In vascular smooth<br />

muscles, a l<strong>in</strong>k between the 2 Na pump subunit and<br />

relaxation pathways has been demonstrated (801). As<br />

with the BK channels, these components of ionic homeostasis<br />

and Ca signal<strong>in</strong>g are located <strong>in</strong> caveolae and<br />

close to the SR (57). From these close appositions of SR<br />

and caveolae, and congregation of signal<strong>in</strong>g components<br />

associated with (but not exclusively limited to)<br />

Ca regulation, it has been proposed that the SR <strong>in</strong> this<br />

region is part of a specialized signal<strong>in</strong>g complex, sometimes<br />

referred to as a signalosome (e.g., Refs. 280, 423).<br />

3. Microdoma<strong>in</strong>s<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

Although it is unlikely that there is a structural anchor<br />

between caveolae and SR, or that there is a Ca<br />

release function <strong>in</strong> caveolae, there is evidence that these<br />

regions <strong>in</strong> the smooth muscle myocytes form a functional<br />

unit affect<strong>in</strong>g Ca signal<strong>in</strong>g. This microdoma<strong>in</strong> arises between<br />

the plasma membrane and SR and has the conditions<br />

and components necessary to 1) have higher [Ca]<br />

than the bulk cytoplasm, 2) vectorially release Ca, and<br />

3) act as a barrier or buffer to the free diffusion of Ca <strong>in</strong><br />

this doma<strong>in</strong>.<br />

The existence of Ca microdoma<strong>in</strong>s <strong>in</strong> smooth muscle,<br />

where the [Ca] may be several orders of magnitude<br />

greater than <strong>in</strong> the bulk cytoplasm, overcomes objections<br />

to the NCX and Ca-activated membrane channels hav<strong>in</strong>g<br />

too low a Ca aff<strong>in</strong>ity to contribute to Ca homeostasis<br />

dur<strong>in</strong>g physiological processes. If [Ca] can also change<br />

rapidly <strong>in</strong> these microdoma<strong>in</strong>s, then those prote<strong>in</strong>s with<br />

high association constants will be activated preferentially,<br />

even when they have similar K d values (451). The relatively<br />

slow diffusion constant of Ca <strong>in</strong> smooth muscle<br />

cytoplasm 2.2 10 6 cm 2 /s (37), compared with water,<br />

will contribute to the ma<strong>in</strong>tenance of the microdoma<strong>in</strong>.


120 SUSAN WRAY AND THEODOR BURDYGA<br />

Many of the features that led to the postulation of a<br />

specialized space and environment between the SR and<br />

plasma membrane will also hold for regions between the<br />

SR and other organelles.<br />

4. Superficial buffer barrier<br />

Van Breemen and colleagues (713, 715) hypothesized<br />

<strong>in</strong> the 1980s that there is a subplasmalemmal doma<strong>in</strong> that<br />

is functionally separated from the cytosolic constituents<br />

by the superficial SR and that this would be important <strong>in</strong><br />

modulat<strong>in</strong>g Ca signal<strong>in</strong>g and force production <strong>in</strong> smooth<br />

muscle. This peripheral SR, be<strong>in</strong>g so close to the sites of<br />

Ca entry, would act to scavenge and buffer extracellular<br />

Ca entry. This buffer<strong>in</strong>g ability would be overcome dur<strong>in</strong>g<br />

agonist stimulation or depolarization but could act to help<br />

ma<strong>in</strong>ta<strong>in</strong> rest<strong>in</strong>g [Ca] low between stimuli. Follow<strong>in</strong>g<br />

stimulation, the superficial SR will release Ca back to the<br />

plasma membrane Ca extrusion mechanisms. Because of<br />

the difficulty of directly measur<strong>in</strong>g [Ca] <strong>in</strong> microdoma<strong>in</strong>s,<br />

direct experimental support for them was lack<strong>in</strong>g. However,<br />

with the advent of near-membrane Ca <strong>in</strong>dicators<br />

(170, 809), evidence support<strong>in</strong>g or consistent with the<br />

superficial buffer barrier role of the SR <strong>in</strong> smooth muscles<br />

has been provided <strong>in</strong> a variety of preparations [vascular<br />

(587, 713), gastric (754), bladder (789), uterus (631, 793)].<br />

The sites on the SR of Ca release and uptake dur<strong>in</strong>g these<br />

processes may be functionally and spatially separate<br />

(635). The estimates of subplasmalemma [Ca] have been<br />

between 10 and 50 M [gastric (170), colonic (77), arterial<br />

(809)], and the elevations of [Ca] last up to 100–200 ms<br />

(339).<br />

Inhibition of SERCA <strong>in</strong>creases bulk Ca signals produced<br />

<strong>in</strong> response to agonists, which is consistent with<br />

the superficial buffer barrier hypothesis. Bradley et al.<br />

(77) calculated that the buffer accounted for b<strong>in</strong>d<strong>in</strong>g of<br />

50% of the Ca enter<strong>in</strong>g colonic myocytes. Recently however,<br />

this group has argued that the SR should be viewed<br />

more as a “Ca trap” and that SERCA activity cannot<br />

curtail the rise of cytosolic [Ca] but rather contributes to<br />

its decl<strong>in</strong>e when efflux ends (451; see also Ref. 635).<br />

Although not generally considered <strong>in</strong> accounts of<br />

smooth muscle microdoma<strong>in</strong>s, a role for calmodul<strong>in</strong> <strong>in</strong><br />

contribut<strong>in</strong>g to localiz<strong>in</strong>g Ca signals has been proposed<br />

(764). This arises from the f<strong>in</strong>d<strong>in</strong>g that, even at low [Ca],<br />

a specific “contractile” pool of calmodul<strong>in</strong> with zero or<br />

two of its four Ca-b<strong>in</strong>d<strong>in</strong>g sites occupied is tightly bound<br />

to the contractile mach<strong>in</strong>ery. Local changes of [Ca] near<br />

the myofilaments could then activate this specific calmodul<strong>in</strong><br />

pool by saturat<strong>in</strong>g the four Ca-b<strong>in</strong>d<strong>in</strong>g sites to activate<br />

myos<strong>in</strong> light-cha<strong>in</strong> k<strong>in</strong>ase (MLCK) and cause contraction,<br />

while more distant local [Ca] changes need not<br />

be associated with contraction (764).<br />

E. SR and the Mitochondria<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

Both EM and confocal imag<strong>in</strong>g studies have shown<br />

the SR and mitochondrial membranes to be <strong>in</strong> very close<br />

proximity (175, 677). The distance between them may be<br />

as small as 10 nm (515). As with caveolae, the SR may<br />

appear to engulf mitochondria, and thereby create a specialized<br />

space for Ca signal<strong>in</strong>g, discussed below. Although<br />

close association between ER and mitochondria has been<br />

described <strong>in</strong> other cell types (489, 561), few have reported<br />

the almost complete envelop<strong>in</strong>g, which appears to be a<br />

feature of smooth muscles. This aspect has been exam<strong>in</strong>ed<br />

<strong>in</strong> detail at the EM level by Dai et al. (140) <strong>in</strong> pig<br />

tracheal muscle. At rest, all (99.4%) mitochondria were<br />

with<strong>in</strong> 30 nm of SR membrane, and the average distance<br />

was 22 nm. The majority (82%) of mitochondria were<br />

completely encircled or enwrapped by the SR network.<br />

The regions of close contact lacked the regular spac<strong>in</strong>g<br />

sometimes seen between SR and plasma membrane, but<br />

specific anchors have been suggested <strong>in</strong> other cell types<br />

(175, 241).<br />

What makes the above data particularly <strong>in</strong>terest<strong>in</strong>g is<br />

that the association of mitochondria and SR changed with<br />

acetylchol<strong>in</strong>e (ACh) stimulation. The number of mitochondria<br />

surrounded by SR fell to 12%, and fewer were <strong>in</strong><br />

close contact. The authors suggest the SR unwraps from<br />

mitochondria with stimulation and extends more <strong>in</strong>to the<br />

cytoplasm. These structural arrangements between SR<br />

and mitochondria have been reported <strong>in</strong> Mad<strong>in</strong>-Darby<br />

can<strong>in</strong>e kidney (MDCK) II cells, but it is elevated [Ca] that<br />

keeps the close association and between them and low<br />

[Ca] causes dissociation (735). Lateral movements of mitochondria<br />

relative to the SR of up to 3 m/s have been<br />

reported <strong>in</strong> neuroblastoma cells (405).<br />

1. Mitochondria and smooth muscle Ca signal<strong>in</strong>g<br />

There is now a large amount of literature on mitochondrial<br />

Ca signal<strong>in</strong>g and the <strong>in</strong>teractions between the<br />

SR and mitochondria. It is beyond the scope of this review<br />

to assess all the literature on mitochondrial Ca signal<strong>in</strong>g,<br />

but we will give an account of recent data <strong>in</strong>vestigat<strong>in</strong>g<br />

SR-mitochondrial signal<strong>in</strong>g <strong>in</strong> smooth muscle. The follow<strong>in</strong>g<br />

reviews provide a good perspective on this area (296,<br />

451, 565).<br />

Mitochondrial Ca uptake has enjoyed a resurgence of<br />

<strong>in</strong>terest (53) largely accounted for by new techniques<br />

allow<strong>in</strong>g for monitor<strong>in</strong>g their role <strong>in</strong> Ca signal<strong>in</strong>g and the<br />

weight of accumulated evidence show<strong>in</strong>g that Ca uptake<br />

occurs dur<strong>in</strong>g physiological conditions (e.g., Refs. 49, 159,<br />

590). Due to the low aff<strong>in</strong>ity of their Ca transporter (46),<br />

the capacity of mitochondria to rapidly accumulate Ca<br />

was considered only relevant dur<strong>in</strong>g pathologically high<br />

[Ca]. That there could be microdoma<strong>in</strong>s of high [Ca]<br />

around mitochondria, to which the SR (or ER) would


contribute, removed the “problem” of the mitochondrial<br />

transporter hav<strong>in</strong>g such a low aff<strong>in</strong>ity for Ca (591).<br />

In smooth muscles, the follow<strong>in</strong>g studies have shown<br />

that mitochondria can contribute to Ca signal<strong>in</strong>g (114,<br />

156, 454, 488, 515, 589). Elevations of mitochondrial [Ca]<br />

with stimulation have been demonstrated <strong>in</strong> pulmonary<br />

arterial cells and considered to curtail the cytosolic [Ca]<br />

<strong>in</strong>creases (157, 488). If the mitochondrial membrane potential<br />

(usually about 180 mV with respect to cytoplasm)<br />

is dissipated to disable the mitochondria, the decay<br />

of the cytosolic Ca transient was slowed <strong>in</strong> myocytes<br />

from portal ve<strong>in</strong> (225), tail artery (675), pulmonary artery<br />

(157), and cultured aortic cells (678). Although not extensively<br />

studied <strong>in</strong> smooth muscles so far, it may be that<br />

mitochondrial uptake of SR Ca is a mechanism of promot<strong>in</strong>g<br />

maximal Ca release from the SR, perhaps by remov<strong>in</strong>g<br />

Ca-<strong>in</strong>duced feedback on the SR Ca release (128,<br />

603, 675). In their study of anococcygeus smooth muscle,<br />

Rest<strong>in</strong>i et al. (589) describe cross-talk between the SR and<br />

mitochondria and show EM figures of the close contact<br />

between the two. Interactions have also been found with<br />

smooth muscle mitochondria when SR Ca release is<br />

through RyR (124, 603). These studies found that when<br />

mitochondria were <strong>in</strong>hibited there was a slow<strong>in</strong>g of the<br />

rate of Ca release and decay of the Ca transient, and a<br />

decrease <strong>in</strong> Ca sparks. The work of Chalmers and McCarron<br />

(114) suggests that <strong>in</strong> colonic myocytes mitochondrial<br />

activity promotes Ca release through IP 3Rs and that some<br />

mitochondria are situated particularly close to these receptors.<br />

They also addressed the important issue of<br />

whether the Ca entry <strong>in</strong>to mitochodria dur<strong>in</strong>g smooth<br />

muscle activity causes significant mitochondrial <strong>in</strong>ner<br />

membrane depolarization, which would impair their function<br />

and ATP production. They found no change dur<strong>in</strong>g<br />

normal Ca transients but suggest depolarization may occur<br />

dur<strong>in</strong>g repetitive Ca oscillations or oxidative stress.<br />

Further work is required <strong>in</strong> smooth muscles to determ<strong>in</strong>e<br />

if the above data can be generalized to other smooth<br />

muscles, and if cross-talk or functional outcomes differ,<br />

depend<strong>in</strong>g on whether Ca is released via RyR or IP 3R.<br />

More work is also needed to establish the effects of<br />

mitochondria on smooth muscle local and global Ca signals.<br />

F<strong>in</strong>ally, there is emerg<strong>in</strong>g data <strong>in</strong>dicat<strong>in</strong>g that mitochondria<br />

may also play a role <strong>in</strong> pacemak<strong>in</strong>g <strong>in</strong> smooth<br />

muscle cells (345, 374, 741, 742), which deserves further<br />

attention (see also Refs. 160, 456, 503).<br />

F. SR and the Nucleus<br />

The nucleus and the SR are commonly associated <strong>in</strong><br />

smooth muscle cells. For example <strong>in</strong> aorta, Lesh et al.<br />

(394) describe the SR form<strong>in</strong>g a cont<strong>in</strong>uous network and<br />

be<strong>in</strong>g contiguous with the outer nuclear envelope. Over<br />

the last decade, the evidence has shown that the nucleus<br />

SMOOTH MUSCLE SR 121<br />

can regulate its own [Ca] and that the nuclear pores are<br />

regulated (208, 353, 436). Furthermore, identification of<br />

IP 3Rs, RyRs, and SERCA on the nuclear membrane has<br />

added to our understand<strong>in</strong>g of how it may generate and<br />

regulate its Ca signals (2, 276, 415).<br />

The SR may contribute to Ca-regulated gene transcription,<br />

via CREB or NFAT (31, 110, 217, 662). The term<br />

excitation-transcription coupl<strong>in</strong>g has been used to highlight<br />

that the changes <strong>in</strong> [Ca] occurr<strong>in</strong>g dur<strong>in</strong>g E-C coupl<strong>in</strong>g<br />

will also be <strong>in</strong>volved <strong>in</strong> regulat<strong>in</strong>g gene expression<br />

and phenotypic modulation (733).<br />

There is only a sparse literature on <strong>in</strong>teractions between<br />

the SR and nucleus <strong>in</strong> smooth muscles. In cultured<br />

aortic cells us<strong>in</strong>g BODIPY-RY and thapsigarg<strong>in</strong>, Abrenica<br />

et al. (3) reported a discrete localization of SERCA and<br />

RyR per<strong>in</strong>uclearly and suggested that they could contribute<br />

to regulat<strong>in</strong>g nucleoplasmic [Ca]. In turn, a role for the<br />

nucleus as a Ca s<strong>in</strong>k was also suggested by the authors. In<br />

their study of Ca sparks <strong>in</strong> portal ve<strong>in</strong> myocytes, Gordienko<br />

et al. (221) reported that the majority of spark<br />

<strong>in</strong>itiation sites were with<strong>in</strong> 1–2 m of the nuclear envelope<br />

(mitochondria were not particularly associated with<br />

sparks <strong>in</strong> this study). The frequency of sparks <strong>in</strong> these<br />

per<strong>in</strong>uclear regions often <strong>in</strong>creased to produce a Ca wave<br />

and contraction.<br />

In cultured aortic (A7r5) cells, Missiaen et al. (482)<br />

noted Ca release and uptake <strong>in</strong> a thapsigarg<strong>in</strong>-<strong>in</strong>sensitive<br />

and nonmitochondrial store, which could have been the<br />

Golgi or nucleus. However, there seems to be little or no<br />

<strong>in</strong>formation on such <strong>in</strong>teractions <strong>in</strong> native smooth muscles,<br />

so noth<strong>in</strong>g further can be said at present.<br />

G. Lysosomes<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

Interest <strong>in</strong> lysosomes as Ca storage organelles was<br />

stimulated by the f<strong>in</strong>d<strong>in</strong>g that <strong>in</strong> sea urch<strong>in</strong> eggs a recently<br />

identified new Ca mobilizer, nicot<strong>in</strong>ic acid aden<strong>in</strong>e d<strong>in</strong>ucleotide<br />

phosphate (NAADP), was stored <strong>in</strong> organelles<br />

equivalent to lysosomes <strong>in</strong> mammalian cells (126).<br />

NAADP is discussed further <strong>in</strong> section V. In smooth muscle,<br />

a close association between the SR and per<strong>in</strong>uclear<br />

lysosomes has been shown (350, 650), and this region is<br />

described as a “trigger zone.” Lysosomes are acidic organelles<br />

dependent on vacuolar H-ATPase activity. Drugs<br />

that <strong>in</strong>hibit the transporter, such as bacliomyc<strong>in</strong>, have<br />

therefore been used to determ<strong>in</strong>e the importance of<br />

this Ca store to cells <strong>in</strong> which this type of Ca mobilizer<br />

has been found [myometrial (650), pulmonary (61), and<br />

coronary myocytes (800)]. In these studies a decrease<br />

<strong>in</strong> the Ca signal to agonists (endothel<strong>in</strong>-1, histam<strong>in</strong>e)<br />

was found. To date, no studies <strong>in</strong> smooth muscle have<br />

tried to assess the size of the lysosomal Ca pool or<br />

looked at the dynamics of its <strong>in</strong>teraction with the SR. It<br />

appears at least <strong>in</strong> some cells that the NAADP release


122 SUSAN WRAY AND THEODOR BURDYGA<br />

can trigger SR Ca release from RyRs, but not IP 3R,<br />

suggest<strong>in</strong>g a cluster<strong>in</strong>g of RyRs with the release channels<br />

on the lysosomes (61).<br />

III. SARCO/ENDOPLASMIC RETICULUM<br />

CALCIUM-ADENOSINETRIPHOSPHATASE<br />

A. Introduction<br />

SERCA is the major prote<strong>in</strong> associated with the SR. It<br />

is a P-type ATPase responsible for transport<strong>in</strong>g Ca <strong>in</strong>to<br />

the SR at the expense of ATP. Calcium b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s<br />

enable this process to cont<strong>in</strong>ue with an SR free ionized Ca<br />

content of 500 M, only 0.5–1% of the total Ca stored<br />

<strong>in</strong> the SR (463). This sequester<strong>in</strong>g of lum<strong>in</strong>al Ca also<br />

means that <strong>in</strong>hibitory feedback of Ca on the ATPase is<br />

m<strong>in</strong>imized. Auxiliary prote<strong>in</strong>s, most notably phospholamban,<br />

as well as metabolites and pH, regulate SERCA activity.<br />

Several isoforms of SERCA exist, and expression<br />

varies between smooth muscles.<br />

<strong>Function</strong>ally SERCA creates an <strong>in</strong>ternal Ca reservoir.<br />

As a by-product of this activity it will also contribute to Ca<br />

homeostasis and basal [Ca] <strong>in</strong>creases with<strong>in</strong> the cytoplasm<br />

when its activity is <strong>in</strong>hibited. SERCA may be considered<br />

a Ca buffer, transferr<strong>in</strong>g bound Ca from the cytosol<br />

to the lumen of the SR (105). SERCA also functions<br />

to expedite the removal of Ca from the cytoplasm follow<strong>in</strong>g<br />

stimulation and thereby contribut<strong>in</strong>g to relaxation.<br />

Pharmacological <strong>in</strong>hibitors of SERCA allowed progress to<br />

be made <strong>in</strong> understand<strong>in</strong>g its structure and roles <strong>in</strong><br />

smooth muscle, and fluorescent <strong>in</strong>dicators have permitted<br />

the monitor<strong>in</strong>g of its effects on <strong>in</strong>tracellular [Ca] and<br />

direct measurements of lum<strong>in</strong>al [Ca]. Molecular studies<br />

have given a precise understand<strong>in</strong>g of many aspects of<br />

how SERCA structure relates to function and the consequences<br />

of genetic mutations. The structure of SERCA<br />

will be described next, followed by its catalytic cycle and<br />

then isoform types and function <strong>in</strong> smooth muscle. Features<br />

of its regulation and function are shown <strong>in</strong> Figure 3.<br />

B. Enzyme Type and Activity<br />

Active transport is required to drive Ca <strong>in</strong>to the SR.<br />

This is brought about by the P-type (ion-motive) ATPase<br />

of the SR membrane, SERCA, which is <strong>in</strong> the same enzymatic<br />

class as the Na-K-ATPase and the plasma membrane<br />

Ca-ATPase (PMCA). Am<strong>in</strong>o acid sequence similarity<br />

to the Na pump is more extensive than with PMCA.<br />

Recent studies us<strong>in</strong>g chimeras have begun to elucidate<br />

how SERCA and PMCA are targeted to their respective<br />

membranes (36, 509); SERCA has an ER retention signal<br />

close to the NH 2 term<strong>in</strong>us, which is cytoplasmic fac<strong>in</strong>g.<br />

SERCA transports two Ca for every ATP hydrolyzed. This<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

FIG. 3. Summary of SERCA functions and regulation. SERCA is<br />

known to be regulated by changes <strong>in</strong> Ca, pH, ATP/ADP ratio, phospholamban,<br />

and hormones, e.g., thyrox<strong>in</strong>e. SERCA activity plays a role <strong>in</strong> Ca<br />

homeostasis, establishment and ma<strong>in</strong>tenance of Ca microdoma<strong>in</strong>s, determ<strong>in</strong><strong>in</strong>g<br />

lum<strong>in</strong>al [Ca], buffer<strong>in</strong>g Ca, transport<strong>in</strong>g Ca and countertransport<strong>in</strong>g<br />

H , regulat<strong>in</strong>g IP 3R and RyR Ca release, and relaxation via Ca<br />

uptake.<br />

also dist<strong>in</strong>guishes it from PMCA, which has a 1:1 stoichiometry<br />

(290). For further details on the structure of<br />

SERCA and its similarity to other type ATPases, see the<br />

follow<strong>in</strong>g reviews (106, 676).<br />

The demonstration that SR membrane fractions<br />

could accumulate Ca and hydrolyzed ATP was the first<br />

evidence of SERCA (161). Such <strong>in</strong>vestigations were<br />

helped by the abundance of SERCA <strong>in</strong> striated muscle SR,<br />

as were the subsequent structural studies. The uptake and<br />

release of Ca <strong>in</strong>to the SR by SERCA occurs as a cycle of<br />

reactions and conformational changes.<br />

C. SERCA Structure and Catalytic Cycle<br />

The follow<strong>in</strong>g account is taken largely from several<br />

excellent reviews (487, 573, 777, 791). The pump was purified<br />

<strong>in</strong> 1970 (428) and the crystal structure resolved <strong>in</strong> 2000<br />

by Toyoshima (701), and the conformational changes dur<strong>in</strong>g<br />

Ca release were elucidated 2 years later by the same<br />

group (702). The structural determ<strong>in</strong>ation of SERCA (isoform<br />

1a) was the first for any active transporter. The<br />

SERCAs are complex prote<strong>in</strong>s with s<strong>in</strong>gle-stranded,<br />

polypeptide cha<strong>in</strong>s of 110 kDa and dist<strong>in</strong>ct doma<strong>in</strong>s<br />

(676). These doma<strong>in</strong>s undergo considerable movement as<br />

the enzyme performs its function, which has the effect of<br />

limit<strong>in</strong>g the amount of regulation that can occur without


compromis<strong>in</strong>g function. Folded <strong>in</strong>to the molecule are<br />

four doma<strong>in</strong>s: M, transmembrane doma<strong>in</strong>; P, phosphorylation<br />

doma<strong>in</strong>; N, nucleotide-bid<strong>in</strong>g doma<strong>in</strong>; and A, the<br />

actuator doma<strong>in</strong>. Figure 4 shows diagrammatically<br />

SERCA structure and its catalytic cycle. The Ca b<strong>in</strong>d<strong>in</strong>g<br />

sites are <strong>in</strong> the M doma<strong>in</strong>, with the two Ca ions be<strong>in</strong>g 5.7A˚<br />

apart. There are 10 transmembrane sections <strong>in</strong> the M doma<strong>in</strong><br />

(M1-M10), and this doma<strong>in</strong> forms the channel through<br />

which Ca moves. The <strong>in</strong>teraction site for phospholamban, a<br />

major regulator of SERCA1 (see sect. IVC) is located <strong>in</strong> the N<br />

doma<strong>in</strong>, around Lys-397, although other b<strong>in</strong>d<strong>in</strong>g sites on the<br />

M doma<strong>in</strong> have been described (19, 20). Thapsigarg<strong>in</strong> and<br />

cyclopiazonic acid (CPA), specific <strong>in</strong>hibitors of SERCA, b<strong>in</strong>d<br />

to the M doma<strong>in</strong> (Phe-256 of M3) (521) and helped with the<br />

elucidation of SERCA structure and function (102). From<br />

topological models, the structure of SERCA can also be<br />

described as hav<strong>in</strong>g three parts: a cytoplasmic head, a stalk<br />

doma<strong>in</strong>, and a transmembrane doma<strong>in</strong> (442). The cytoplasmic<br />

head region constitutes around half of the total SERCA<br />

mass, and it encompasses the A, N, and P doma<strong>in</strong>s. The A<br />

and P doma<strong>in</strong>s are connected to the M doma<strong>in</strong>, and the N<br />

doma<strong>in</strong> is connected to the P doma<strong>in</strong>.<br />

The simplest catalytic cycle for SERCA can be described<br />

as follows: E 132Ca-E 1,32Ca-E 1-ATP32Ca-E 1P i32Ca-E 2-<br />

P i3E 2-P i3E 23E 1 (see also Fig. 4).<br />

The two Ca b<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong>s exist <strong>in</strong> either a high- or<br />

low-aff<strong>in</strong>ity form, known as E 1 and E 2 states, and allow<br />

access only from the cytosolic and lum<strong>in</strong>al side, respec-<br />

SMOOTH MUSCLE SR 123<br />

tively. Their K d values are 10 7 and 10 3 M, respectively.<br />

Work has suggested that two Ca ions from the cytosolic<br />

side b<strong>in</strong>d, and the enzyme is subsequently phosphorylated<br />

by ATP at Asp-351. The formation of an “energy-rich”<br />

aspartyl-phosphorylated <strong>in</strong>termediate is why SERCA is<br />

classified as a P-type pump. The phosphorylation reaction<br />

is highly pH sensitive and H facilitate the reaction of<br />

<strong>in</strong>organic phosphate (P i) with the Ca pump (291). Phospholamban<br />

b<strong>in</strong>ds preferentially to the E 2 state, lower<strong>in</strong>g<br />

its apparent Ca aff<strong>in</strong>ity. It is as this high-energy <strong>in</strong>termediate<br />

(2Ca-E 1P i) that the translocation of Ca occurs.<br />

Follow<strong>in</strong>g translocation and release of the two Ca ions<br />

<strong>in</strong>to the SR lumen, the enzyme becomes a low-energy<br />

<strong>in</strong>termediary. Two or possibly three (395) protons are<br />

countertransported dur<strong>in</strong>g the step E 2-P i3E 2, which aids,<br />

but does not complete, electroneutrality across the SR<br />

membrane. The protons are released <strong>in</strong>to the cytoplasm<br />

follow<strong>in</strong>g b<strong>in</strong>d<strong>in</strong>g of the two Ca ions.<br />

D. SERCA Isoforms and Expression<br />

<strong>in</strong> <strong>Smooth</strong> <strong>Muscle</strong>s<br />

Three genes (ATP2A1, ATP2A2, and ATP2A3) encode<br />

the three mammalian isoforms of SERCA known as<br />

SERCA1, -2, or -3. In addition, alternative splic<strong>in</strong>g occurs<br />

giv<strong>in</strong>g rise to SERCA1a, SERCA1b, SERCA2a, and SERCA2b.<br />

SERCA1a is found <strong>in</strong> adult fast-twitch skeletal muscle, and<br />

FIG. 4. Schematic representation of the structural changes <strong>in</strong> SERCA and the accompany<strong>in</strong>g catalytic cycle. Folded <strong>in</strong>to the molecule are four<br />

doma<strong>in</strong>s: M, transmembrane doma<strong>in</strong>; P, phosphorylation doma<strong>in</strong>; N, nucleotide-b<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong>; A, actuator doma<strong>in</strong>. The two Ca b<strong>in</strong>d<strong>in</strong>g doma<strong>in</strong>s<br />

exist <strong>in</strong> a either a high- or low-aff<strong>in</strong>ity form, known as E 1 and E 2 states, and allow access only from the cytosolic and lum<strong>in</strong>al side, respectively.<br />

(Adapted from Petsko GA, R<strong>in</strong>ge D. Prote<strong>in</strong> Structure and <strong>Function</strong>. New Science Press, 2004; on-l<strong>in</strong>e update 2006–2007.)<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org


124 SUSAN WRAY AND THEODOR BURDYGA<br />

SERCA1b is <strong>in</strong> neonatal muscles. Brody’s disease is a rare<br />

<strong>in</strong>herited muscle disorder <strong>in</strong> which relaxation rates of<br />

skeletal muscle are impaired, lead<strong>in</strong>g to muscle stiffness.<br />

Patients have high rates of mutation <strong>in</strong> the SERCA1 gene<br />

which lower its Ca transport activity (43). SERCA3 appears<br />

to be the most diverse form, and an ever-grow<strong>in</strong>g<br />

number of isoforms are be<strong>in</strong>g discovered (15, 58, 534,<br />

566). The first was SERCA3a, found <strong>in</strong> nonmuscle cells<br />

and which has a low apparent Ca aff<strong>in</strong>ity, lead<strong>in</strong>g to<br />

question<strong>in</strong>g of its functional role <strong>in</strong> Ca signal<strong>in</strong>g (573),<br />

although it may be that it ma<strong>in</strong>ta<strong>in</strong>s high [Ca] <strong>in</strong> microdoma<strong>in</strong>s.<br />

Subsequently, seven additional SERCA3 gene<br />

products have been described (58, 441). SERCA3 has<br />

been found <strong>in</strong> epithelial and endothelial cells, platelets,<br />

megakaryocytes, many endocr<strong>in</strong>e glands, cerebellum,<br />

pancreas, and spleen (59). It seems that SERCA3 is not<br />

found <strong>in</strong> smooth muscle cells but could appear as a<br />

contam<strong>in</strong>ant from other cell types when whole smooth<br />

muscle tissues are analyzed. For example, SERCA3-deficient<br />

mice have impaired vascular and tracheal relaxation,<br />

but the effects are mediated via endothelial and<br />

epithelial cells, respectively, which express SERCA3b.<br />

SERCA2 is the isoform found <strong>in</strong> smooth muscles, and<br />

this will be discussed <strong>in</strong> detail.<br />

1. SERCA2<br />

SERCA2 is 84% identical <strong>in</strong> sequence to SERCA1a.<br />

The gene for SERCA2 is located on human chromosome<br />

12 at 12q24.11. SERCA2b has a longer (lum<strong>in</strong>al) COOHterm<strong>in</strong>al<br />

tail than that of SERCA2a, which is cytoplasmic<br />

(726). SERCA2a is expressed <strong>in</strong> heart, slow-twitch skeletal<br />

muscle, and many types of smooth muscles (25).<br />

SERCA2b appears be ubiquitously expressed, lead<strong>in</strong>g to it<br />

be<strong>in</strong>g labeled as the housekeep<strong>in</strong>g form of SERCA. This<br />

designation, while also consistent with it be<strong>in</strong>g the phylogenetically<br />

earliest isoform, is likely to be too simplistic,<br />

as different tissues express different levels of SERCA2b<br />

(777). From expression studies <strong>in</strong> COS and cardiac cells it<br />

has been suggested that, compared with SERCA2a, the 2b<br />

isoform has a lower turnover rate of ATP hydrolysis, a<br />

10-fold lower vanadate sensitivity and Ca transport rate,<br />

but a higher apparent aff<strong>in</strong>ity for Ca (425, 723, 724).<br />

Removal of the last 12 am<strong>in</strong>o acids from SERCA2b <strong>in</strong><br />

mouse cardiac cells abolished these differences, suggest<strong>in</strong>g<br />

the long tail <strong>in</strong> 2b is affect<strong>in</strong>g SERCA activity (723).<br />

Another difference between SERCA2a and -2b is the<br />

greater stability of 2a’s mRNA, perhaps expla<strong>in</strong><strong>in</strong>g its<br />

<strong>in</strong>creased expression levels. Both isoforms have the same<br />

sensitivity to phospholamban and thapsigarg<strong>in</strong> (426).<br />

Although the differences between 2a and 2b are not<br />

extreme, their functional and developmental importance,<br />

at least <strong>in</strong> cardiac muscle, is great. Mice eng<strong>in</strong>eered to<br />

have SERCA2a replaced with SERCA2b suffer from embryonic/neonatal<br />

mortality due to malformations of their<br />

hearts (40%), and if the switch is made <strong>in</strong> adults, they<br />

develop cardic hypertrophy (723). Heterozygous mice developed<br />

squamous cell tumors (560). The human disease<br />

Darier-White condition, <strong>in</strong> which there are foci of separation<br />

between kerat<strong>in</strong>ocytes, has been associated with mutations<br />

of the SERCA2 gene, but no cardiac symptoms are<br />

apparent (608).<br />

In smooth muscles, when comparisons have been<br />

attempted, SERCA2b expression is often greater than that<br />

of 2a. Both 2a and 2b have been described <strong>in</strong> most smooth<br />

muscles: stomach (162), aorta (15, 434, 774), uterus (94,<br />

707), mesenteric artery (15), trachea (14, 458), ileum<br />

(162), and pulmonary (162).<br />

Follow<strong>in</strong>g an <strong>in</strong>vestigation of splice variants, a third<br />

isoform, SERCA2c, has been described <strong>in</strong> a number of cell<br />

types: epithelial, mesenchymal, hematopoietic, and cardiac<br />

(141, 207), but its expression <strong>in</strong>, or relevance to,<br />

smooth muscle is unknown.<br />

E. Regulators of SERCA Expression and Activity<br />

Given the importance of SERCA to SR function and<br />

hence cellular Ca homeostasis, Ca signal<strong>in</strong>g, and ultimately<br />

function, it was anticipated that its function would<br />

be modulated under physiological conditions and perhaps<br />

altered <strong>in</strong> pathophysiological states. However, the structure<br />

of SERCA has evolved to translocate Ca, and this is<br />

accomplished by large conformational changes <strong>in</strong> the<br />

molecule. This requirement for substantial conformational<br />

change to function is considered to impose limitations<br />

upon how much modification, prote<strong>in</strong>-prote<strong>in</strong> <strong>in</strong>teractions,<br />

and other coupl<strong>in</strong>g to effectors can occur.<br />

1. Accessory prote<strong>in</strong>s<br />

The best known regulator of SERCA is phospholamban,<br />

a small membrane prote<strong>in</strong> discussed <strong>in</strong> section IVC,<br />

along with other prote<strong>in</strong> modulators that work from the<br />

lum<strong>in</strong>al side of the SR. Other cytoplasmic regulators of<br />

SERCA activity are emerg<strong>in</strong>g and were well reviewed by<br />

Wuytack and colleagues (719). The list <strong>in</strong>cludes the <strong>in</strong>sul<strong>in</strong>-receptor<br />

substrate IRS1/2, the Ca b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong><br />

S100A1, acylphosphatase, and the antiapoptotic prote<strong>in</strong><br />

Bc1-2. IRS1/2 has been found to <strong>in</strong>teract with SERCA2 <strong>in</strong><br />

an <strong>in</strong>sul<strong>in</strong>-dependent manner <strong>in</strong> cultured aortic cells (9).<br />

2. Calcium and protons<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

In smooth muscles (and other cell types), regulation<br />

of SERCA expression has been l<strong>in</strong>ked to cytoplasmic<br />

(774) and lum<strong>in</strong>al [Ca] (376). This latter study noted the<br />

coord<strong>in</strong>ated regulation of SERCA and the PMCA, e.g., <strong>in</strong><br />

response to thapsigarg<strong>in</strong>. It is not apparent that the l<strong>in</strong>ked<br />

regulation of SERCA and PMCA are functionally synergistic,<br />

i.e., <strong>in</strong>creased plasmalemmal Ca extrusion on PMCA


will not aid SR Ca refill<strong>in</strong>g, although cytoplasmic [Ca] will<br />

fall when both pumps act. Further studies are required to<br />

confirm this. Protons (2 or 3) are translocated from the SR<br />

to the cytoplasm dur<strong>in</strong>g each cycle of SERCA activity.<br />

Cytoplasmic pH can also affect SERCA activity. Although<br />

pH alteration has not been studied very much <strong>in</strong> smooth<br />

muscle, it appears that alkal<strong>in</strong>ization will decrease and<br />

acidification will <strong>in</strong>crease SERCA activity (22, 645, 772).<br />

There appears to be no data on which SERCA isoforms<br />

are most sensitive to pH alteration.<br />

3. Development, hormones, and gestation<br />

While there is considerable developmental regulation<br />

of SERCA1 and SERCA2a <strong>in</strong> striated muscle (559), and<br />

SERCA3 also shows clear ontological differences <strong>in</strong> expression,<br />

SERCA2b, which predom<strong>in</strong>ates <strong>in</strong> smooth muscle,<br />

appears not to be developmentally regulated.<br />

Cultur<strong>in</strong>g has been shown to change SERCA isoform<br />

expression, as the cells become less differentiated (387,<br />

534). Thus the relative <strong>in</strong>crease <strong>in</strong> SERCA2a with development<br />

<strong>in</strong> aorta is reversed with cultur<strong>in</strong>g (387). It is<br />

worth not<strong>in</strong>g here that more work is required on native<br />

smooth muscle cells if clarity concern<strong>in</strong>g modulators of<br />

SERCA expression and activity is to be obta<strong>in</strong>ed.<br />

Thyrox<strong>in</strong>e is the best recognized hormonal modulator<br />

of SERCA expression, and the SERCA2 gene has promoter<br />

elements that b<strong>in</strong>d thyroid receptors. In neonatal hypothyroidism,<br />

SERCA2a is downregulated. SERCA2a and -2b were<br />

reported to be <strong>in</strong>creased <strong>in</strong> pregnant compared with nonpregnant<br />

rat myometrium (344) and <strong>in</strong> labor<strong>in</strong>g compared<br />

with nonlabor<strong>in</strong>g human myometrium (707). The mechanism<br />

for this upregulation is unclear, although endocr<strong>in</strong>e<br />

changes might well be suspected. A recent paper by Liu et al.<br />

(401) showed that 17-estradiol could upregulate SERCA2a<br />

<strong>in</strong> cultured cells, along with the Na-K-ATPase subunit.<br />

Tamoxifen, the anti-estrogen drug, decreased SERCA activity.<br />

In the male urogenital system, testosterone may <strong>in</strong>fluence<br />

SERCA activity, given that castration <strong>in</strong> rabbits decreased<br />

its activity <strong>in</strong> corpus cavernosum (325). However,<br />

these authors also reported no change <strong>in</strong> SERCA activity <strong>in</strong><br />

urethral myocytes.<br />

F. Summary<br />

There is a very large amount of literature concern<strong>in</strong>g<br />

many aspects of SERCA, but it is often not obta<strong>in</strong>ed on<br />

smooth muscle. We have a detailed understand<strong>in</strong>g of<br />

SERCA structure and catalytic mode of action. We are<br />

also aware of the different SERCA isoforms and their<br />

expression <strong>in</strong> smooth muscle. However, we can still say<br />

very little about how these differences <strong>in</strong>fluence the f<strong>in</strong>etun<strong>in</strong>g<br />

of smooth muscle Ca stores and hence Ca signal<strong>in</strong>g.<br />

Further work <strong>in</strong> smooth muscles on how SERCA<br />

SMOOTH MUSCLE SR 125<br />

activity is affected by developmental or hormonal<br />

changes is needed.<br />

IV. PROTEINS ASSOCIATED WITH THE<br />

SARCOPLASMIC RETICULUM<br />

A. Introduction<br />

SERCA, a prote<strong>in</strong> associated with the SR and of<br />

pivotal importance to its function<strong>in</strong>g, has been described<br />

above. We will now discuss other prote<strong>in</strong>s associated<br />

with the SR and SERCA, especially phospholamban, and<br />

those prote<strong>in</strong>s whose prime role is to b<strong>in</strong>d Ca with<strong>in</strong> the<br />

SR lumen, i.e., calreticul<strong>in</strong> and calsequestr<strong>in</strong>. We also<br />

briefly mention other prote<strong>in</strong>s that may have a role to play<br />

such as junct<strong>in</strong>. We have not considered prote<strong>in</strong>s whose<br />

role is not specific to the SR. However, it is important that<br />

we know about the environment <strong>in</strong> which these prote<strong>in</strong>s<br />

(and SERCA) are operat<strong>in</strong>g; therefore, the SR membrane<br />

and the gradients across it, a topic that has received scant<br />

attention <strong>in</strong> many discussions of the SR, will be discussed<br />

first.<br />

B. The SR Membrane<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

While much attention is given to the plasma membrane<br />

surround<strong>in</strong>g the smooth muscle cell, e.g., its composition,<br />

fluidity, and microdoma<strong>in</strong>s, less attention has<br />

been given to that of the SR. However, it is reasonable to<br />

suggest that the activity of SR channels, transporters, and<br />

associated prote<strong>in</strong>s will also be affected by the same<br />

parameters.<br />

The SR (and ER) has a unique lipid membrane. It is<br />

the most fluid of all membranes, a direct consequence of<br />

it also be<strong>in</strong>g cholesterol-poor and hav<strong>in</strong>g many unsaturated<br />

phospholipids (719). Thus SERCA has evolved to<br />

function <strong>in</strong> this specific membrane, and its optimal activity<br />

<strong>in</strong> artificial lipid bilayers, for example, occurs with a<br />

different composition than that required for maximal Na<br />

pump activity (390). The high fluidity of the SR/ER membrane<br />

presumably helps nascent prote<strong>in</strong>s to fold correctly.<br />

The low cholesterol content also ensures that the<br />

SR membrane is relatively th<strong>in</strong>.<br />

The SR membrane is notably unsaturated <strong>in</strong> nature.<br />

The major phospholipids of the SR are chol<strong>in</strong>e (65%),<br />

ethanolam<strong>in</strong>e (15%), and <strong>in</strong>ositol (7%) phospholipids;<br />

together they constitute 80% of total lipids. Of the rema<strong>in</strong><strong>in</strong>g<br />

neutral lipids, cholesterol makes up 95%. Cholesterol<br />

is excluded from the phospholipid annulus around<br />

SERCA (743). The <strong>in</strong>formative reviews by Tada et al.<br />

(681), Lee et al. (390), and Vangheluwe et al. (719) should<br />

be consulted for further read<strong>in</strong>g on SR lipid and prote<strong>in</strong><br />

composition.


126 SUSAN WRAY AND THEODOR BURDYGA<br />

SERCA is <strong>in</strong> direct contact with several SR lipids but<br />

<strong>in</strong> a cholesterol-poor environment, and this is essential for<br />

its function, as a highly fluid lipid membrane is needed for<br />

the large conformational changes required for SERCA<br />

function. Maximal activity of SERCA is affected by the<br />

thickness and head group composition of the SR phospholipids;<br />

phosphochol<strong>in</strong>e conta<strong>in</strong><strong>in</strong>g 18C fatty acids<br />

yielded maximal ATPase activity, while longer or shorter<br />

fatty acids lowered its activity (261, 499). Dietary fatty<br />

acid changes have been shown to change the composition<br />

of the SR membrane (674), and its cholesterol content<br />

may <strong>in</strong>crease with age (373) and contribute to the decreased<br />

SERCA function found <strong>in</strong> older animals (198) as<br />

the SR membrane becomes <strong>in</strong>creas<strong>in</strong>gly rigid.<br />

Variation <strong>in</strong> the lipid composition of the SR from<br />

different skeletal muscles has been described and related<br />

to differences <strong>in</strong> SERCA activity (73), but noth<strong>in</strong>g is<br />

known about how composition may differ between<br />

smooth muscles. In a study of macrophage SERCA, Li<br />

et al. (397) showed that enrichment with cholesterol <strong>in</strong>hibited<br />

SERCA2b activity. This loss of fluidity may expla<strong>in</strong><br />

SERCA decreased activity, as the large conformational<br />

changes required for Ca transport are limited. In terms of<br />

macrophage cell function, the authors noted that dysfunction<br />

of SERCA could lead to prote<strong>in</strong> unfold<strong>in</strong>g <strong>in</strong> the ER<br />

and contribute to atherosclerotic lesions <strong>in</strong> foam cells<br />

(cholesterol-loaded macrophages).<br />

C. Phospholamban<br />

1. Introduction<br />

Phospholamban is a 52-am<strong>in</strong>o acid homopentameric<br />

prote<strong>in</strong>, which, depend<strong>in</strong>g on its phosphorylation state,<br />

reversibly b<strong>in</strong>ds to SERCA and affects its activity (431,<br />

639, 680). The phosphorylation of phospholamban occurs<br />

by both cAMP-dependent prote<strong>in</strong> k<strong>in</strong>ase (PKA) at Ser-16<br />

and Ca/calmodul<strong>in</strong>-dependent prote<strong>in</strong> k<strong>in</strong>ase II (CaM k<strong>in</strong>ase<br />

II) at Thr-17, and the name phospholamban was<br />

given to mean “phosphate receptor” (679; see also Katz,<br />

1998 and associated articles <strong>in</strong> the same volume). Phospholamban<br />

<strong>in</strong>hibits SERCA by lower<strong>in</strong>g its apparent aff<strong>in</strong>ity<br />

for Ca through direct prote<strong>in</strong>-prote<strong>in</strong> <strong>in</strong>teractions,<br />

probably of a s<strong>in</strong>gle phospholamban molecule associated<br />

with two SERCA molecules, and thus phospholamban<br />

may restrict the large doma<strong>in</strong> movements needed for<br />

SERCA activity (21, 790). Upon phosphorylation there is a<br />

large change <strong>in</strong> phospholamban charge, and this greatly<br />

decreases its <strong>in</strong>hibitory effect on SERCA. It is clear <strong>in</strong><br />

cardiac muscle that phosphorylation of phospholamban is<br />

a highly important part of the mechanism whereby -agonists<br />

via cAMP <strong>in</strong>crease cardiac contractility; by reliev<strong>in</strong>g<br />

the <strong>in</strong>hibition of phospholamban on SERCA, more Ca can<br />

enter the SR and contribute to an <strong>in</strong>creased Ca release on<br />

the next heart beat, and relaxation occurs at a faster rate.<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

In heart, chronic <strong>in</strong>hibition of SERCA, as can occur with<br />

some human phospholamban null mutations (447, 621), is<br />

associated with dilated cardiomyopathy, as contractility<br />

is dim<strong>in</strong>ished.<br />

2. Distribution <strong>in</strong> smooth muscle<br />

Phospholamban is expressed <strong>in</strong> smooth muscle, although<br />

at lower levels than occur <strong>in</strong> cardiac myocytes<br />

(378, 643). Raeymaekers and Jones (584) first reported the<br />

presence of phospholamban <strong>in</strong> smooth muscle. They<br />

found it <strong>in</strong> pig stomach and rabbit and dog aorta but not<br />

pig aorta. This group then used pig stomach to cDNA<br />

clone and sequence phospholamban (725) and concluded<br />

it was 100% sequence identical to cardiac phospholamban<br />

and was the product of the same gene. Thus any differences<br />

<strong>in</strong> effects of phospholamban between cardiac and<br />

smooth muscle cannot be expla<strong>in</strong>ed by isoform diversity.<br />

An immunogold EM study of phospholamban showed a<br />

patchy SR distribution of it <strong>in</strong> a variety of smooth muscles<br />

(ileum, iliac artery, and aorta), aga<strong>in</strong> suggest<strong>in</strong>g that its<br />

density was lower than that of cardiac SR (172). Interest<strong>in</strong>gly,<br />

phospholamban label<strong>in</strong>g of the outer nuclear envelope<br />

was also seen, although this observation does not<br />

appear to have been further <strong>in</strong>vestigated. These early<br />

studies concluded that whereas SERCA expression was<br />

approximately comparable between smooth muscles,<br />

phospholamban mRNA levels varied 12-fold, with aorta<br />

express<strong>in</strong>g low levels compared with ileum and stomach<br />

(162).<br />

Given the importance of PKA and CaM k<strong>in</strong>ase II to<br />

smooth muscle force modulation, phosphorylation of<br />

phospholamban would be predicted to be one of their<br />

targets. It has also been suggested that <strong>in</strong> smooth muscle<br />

cGMP is an important mediator of phospholamban phosphorylation<br />

(129, 583) act<strong>in</strong>g at Ser-16. A recent report<br />

(363) has found that <strong>in</strong> tracheal smooth muscle phospholamban<br />

is associated with a PKA signal<strong>in</strong>g complex, <strong>in</strong><br />

addition to its expected association with SERCA. If this<br />

were also to be the case <strong>in</strong> other smooth muscles, then<br />

this may lead to facilitated IP3-<strong>in</strong>duced Ca release, rather<br />

than effects via SERCA Ca pump<strong>in</strong>g, i.e., phospholamban<br />

may have multiple functions and effects <strong>in</strong> smooth muscles.<br />

As the target for several second messenger-dependent<br />

k<strong>in</strong>ases, phospholamban may also play a role <strong>in</strong> Ca<br />

signal<strong>in</strong>g events associated with vascular smooth muscle<br />

migration and hyperplasia, or <strong>in</strong>fluence endothelial Ca<br />

signals, but a discussion of this is beyond the scope of this<br />

review (117, 563, 672).<br />

3. <strong>Function</strong>al effects<br />

The earliest studies performed <strong>in</strong> smooth muscle to<br />

<strong>in</strong>vestigate phospholamban’s role <strong>in</strong>volved vesicle studies<br />

of Ca uptake (582, 745). Sarcevic et al. (615), us<strong>in</strong>g cultured<br />

aortic smooth muscle cells, suggested that the


cGMP-mediated atrial natriuretic peptide transduction<br />

pathway <strong>in</strong>volved phosphorylation of phospholamban. In<br />

1992, Karczewski et al. (338) suggested a role for phospholamban<br />

<strong>in</strong> the nitric oxide (NO)/endothelium-derived<br />

relax<strong>in</strong>g factor (EDRF)-<strong>in</strong>duced relaxation of rat aorta.<br />

This work was followed up with a paper demonstrat<strong>in</strong>g<br />

more directly phosphorylation of phospholamban and<br />

coronary artery relaxation (337). However, no difference<br />

<strong>in</strong> blood pressure was reported when phospholamban<br />

knockout animals were made (418), and little effect of<br />

modulators of cGMP on the k<strong>in</strong>etics of aortic force was<br />

seen (379). Thus phospholamban may play a role <strong>in</strong> the<br />

actions of endogenous vasodilators, but its effects appear<br />

to be specific to different vascular beds.<br />

The development of the phospholamban knockout<br />

mouse has led to <strong>in</strong>vestigation of its role <strong>in</strong> smooth muscles.<br />

An <strong>in</strong>crease <strong>in</strong> the rate of force development to<br />

phenylephr<strong>in</strong>e stimulation <strong>in</strong> aorta was observed, compared<br />

with wild-type controls (378). However, these authors<br />

reported no differences when KCl was the stimulant<br />

and, importantly, no significant difference <strong>in</strong> relaxation<br />

rates. As noted above, this was also the case when cyclic<br />

nucleotides were used (379). Given the role of SERCA <strong>in</strong><br />

facilitat<strong>in</strong>g Ca efflux from smooth muscle myocytes, these<br />

data do not support a prom<strong>in</strong>ent role for phospholamban<br />

<strong>in</strong> this process, with two caveats. First, as with all knockout<br />

studies, other compensatory mechanisms can occur,<br />

e.g., changes <strong>in</strong> plasma membrane Ca transporters or<br />

other SR prote<strong>in</strong>s such as sarcolip<strong>in</strong> (see below). Second,<br />

force measurements can be an unreliable surrogate for<br />

[Ca] changes, and no simultaneous [Ca] and force measurements<br />

have been made.<br />

The study by Lalli et al. (378) on aorta of knockout<br />

mice described above did however note that greater phenylephr<strong>in</strong>e-<strong>in</strong>duced<br />

force was produced, compared with<br />

controls suggest<strong>in</strong>g, but not prov<strong>in</strong>g, that an <strong>in</strong>crease <strong>in</strong><br />

SR Ca uptake may be occurr<strong>in</strong>g. In subsequent studies,<br />

the same group has exam<strong>in</strong>ed portal ve<strong>in</strong>, a phasic, blood<br />

vessel, which may be considered to have its [Ca] changes<br />

controlled and regulated <strong>in</strong> a different manner from the<br />

tonic aorta (671). This study found an alteration of the<br />

contractile pattern <strong>in</strong> the knockout mice, but no difference<br />

<strong>in</strong> sensitivity to ACh, and no difference <strong>in</strong> relaxation<br />

rate to isoproterenol.<br />

It appears then that there are some differences <strong>in</strong><br />

blood vessels from phospholamban knockout mice compared<br />

with wild type, but there is no consistent f<strong>in</strong>d<strong>in</strong>g<br />

and perhaps only subtle differences <strong>in</strong> the response to<br />

agonists or relaxation rates. One drawback to conclusions<br />

drawn from the studies to date is that no measurements of<br />

SR Ca uptake or release have been made on smooth<br />

muscle cells taken from these animals to directly test<br />

suggestions, such as <strong>in</strong>creased SR Ca uptake or faster Ca<br />

release. <strong>Function</strong>ally no differences <strong>in</strong> blood pressure<br />

have been found <strong>in</strong> these studies.<br />

SMOOTH MUSCLE SR 127<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

Several visceral smooth muscles have also been used<br />

to help elucidate phospholamban’s functional contribution<br />

to Ca signal<strong>in</strong>g and contractility. In a series of studies<br />

on gastric smooth muscle, Kim and colleagues (346–348)<br />

have <strong>in</strong>vestigated the role of phospholamban <strong>in</strong> NO-mediated<br />

relaxation. Their work suggests that CaM k<strong>in</strong>ase II<br />

is responsible for the phosphorylation of phospholamban<br />

<strong>in</strong> this tissue. They conclude that the major mechanism<br />

<strong>in</strong>volved <strong>in</strong> phospholamban’s effect on gastric relaxation<br />

is an <strong>in</strong>crease <strong>in</strong> SERCA activity <strong>in</strong>fluenc<strong>in</strong>g Ca removal,<br />

rather than an <strong>in</strong>crease of spark-spontaneous transient<br />

outward current (STOC) coupl<strong>in</strong>g mechanism, although<br />

neither was measured. PKA-stimulated phosphorylation<br />

of phospholamban is postulated as one of the mechanisms<br />

whereby 2-adrenergic receptors ( 2-AR) act to<br />

relax airway smooth muscle (263). Stimulated by the<br />

observation that asthmatics chronically treated with<br />

2-AR agonist develop enhanced sensitivity to bronchoconstrictors<br />

(372), transgenic mice overexpress<strong>in</strong>g the<br />

2-AR were studied (458). The authors noted that phospholamban<br />

transcripts and prote<strong>in</strong> were markedly reduced<br />

<strong>in</strong> airway smooth muscle cells from these mice.<br />

These mice also had reduced constriction to methachol<strong>in</strong>e.<br />

Thus they suggest that downregulation of phospholamban<br />

is a target of 2-AR agonists and protects aga<strong>in</strong>st<br />

airway hyperactivity.<br />

We have prelim<strong>in</strong>ary data on knockout mice <strong>in</strong>dicat<strong>in</strong>g<br />

that <strong>in</strong> the uterus phospholamban can <strong>in</strong>fluence the<br />

pattern of contractility. Thus the pattern of spontaneous<br />

contractions compared with controls was less frequent<br />

but longer last<strong>in</strong>g, and with many of the contractions<br />

hav<strong>in</strong>g a spiked appearance (518). This pattern was also<br />

found <strong>in</strong> portal ve<strong>in</strong> from knockout mice (671).<br />

Phospholamban is expressed <strong>in</strong> bladder smooth muscle<br />

(517). Measurements of force and [Ca] have been<br />

made <strong>in</strong> phospholamban knockout (KO) and phospholamban<br />

overexpress<strong>in</strong>g (OE) mice. When stimulated by carbachol,<br />

compared with wild types, maximal <strong>in</strong>creases of<br />

force and [Ca] were depressed <strong>in</strong> KOs and <strong>in</strong>creased <strong>in</strong><br />

OE mice (517). These differences were abolished when<br />

SERCA was <strong>in</strong>hibited by CPA. Rest<strong>in</strong>g [Ca] was also<br />

elevated <strong>in</strong> the OE mice. It was concluded that Ca uptake<br />

<strong>in</strong>to the SR, rather than content, is the mechanism by<br />

which phospholamban is affect<strong>in</strong>g force. Interest<strong>in</strong>gly,<br />

the phospholamban OE mice also had a significant decrease<br />

<strong>in</strong> SERCA expression.<br />

In summary, it appears to us that despite some elegant<br />

studies there are <strong>in</strong>sufficient mechanistic and functional<br />

studies to date to draw many firm conclusions<br />

about the role of phospholamban <strong>in</strong> smooth muscle. Some<br />

of the expected f<strong>in</strong>d<strong>in</strong>gs of knock<strong>in</strong>g out phospholamban,<br />

e.g., slow<strong>in</strong>g of Ca relaxation rates, do not occur and the<br />

reported changes <strong>in</strong> force are subtle rather than obvious;<br />

certa<strong>in</strong>ly the profound effects on contractility seen <strong>in</strong><br />

cardiac muscle with phospholamban alteration appear


128 SUSAN WRAY AND THEODOR BURDYGA<br />

not to occur <strong>in</strong> most smooth muscles. Few studies on<br />

smooth muscle have considered the more complex role of<br />

the SR <strong>in</strong> this tissue, e.g., provid<strong>in</strong>g negative feedback on<br />

Ca entry via Ca sparks, as opposed to it act<strong>in</strong>g as a Ca<br />

store to augment contraction. We tentatively conclude<br />

that phospholamban is not a major physiological regulator<br />

of SERCA <strong>in</strong> most smooth muscles; its effects on<br />

SERCA, for example, are not as large as those of lum<strong>in</strong>al<br />

[Ca] alteration. This difference between smooth and cardiac<br />

muscles may be due to the relative expression of<br />

SERCA isoforms between the two muscles, but we suggest<br />

it also reflects the differences <strong>in</strong> the role of SERCA<br />

between them.<br />

D. Sarcolip<strong>in</strong><br />

This 31-am<strong>in</strong>o acid prote<strong>in</strong> appears to be a homolog<br />

of phospholamban (522); it associates with the SR membrane,<br />

and its transmembrane doma<strong>in</strong>s are structurally<br />

similar. Sarcolip<strong>in</strong> <strong>in</strong>hibits SERCA by lower<strong>in</strong>g its apparent<br />

Ca aff<strong>in</strong>ity (<strong>in</strong>creas<strong>in</strong>g K m) and V max (27, 205, 435,<br />

523). It has been found <strong>in</strong> greatest abundance <strong>in</strong> fast- (199,<br />

425, 511) and slow-twitch muscles (523) as well as cardiac<br />

muscles (720). To date, we can f<strong>in</strong>d no data describ<strong>in</strong>g<br />

sarcolip<strong>in</strong> presence <strong>in</strong> any smooth muscle.<br />

E. Calsequestr<strong>in</strong><br />

1. Introduction<br />

Calsequestr<strong>in</strong> and calreticul<strong>in</strong> (discussed next) are<br />

the two most important lum<strong>in</strong>al buffers of Ca <strong>in</strong> the SR.<br />

They share many prote<strong>in</strong> properties, as well as fulfill<strong>in</strong>g<br />

the requirement of be<strong>in</strong>g a Ca buffer, i.e., b<strong>in</strong>d<strong>in</strong>g Ca with<br />

a high capacity and low aff<strong>in</strong>ity. Calsequestr<strong>in</strong> was the<br />

first SR Ca b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> to be identified (103, 429, 432).<br />

Its crystal structure has been elucidated from rabbit skeletal<br />

muscle (734). These studies suggest that three very<br />

negative thioredox<strong>in</strong>-like doma<strong>in</strong>s underlie the high Ca<br />

b<strong>in</strong>d<strong>in</strong>g capacity of calsequestr<strong>in</strong>.<br />

2. Isoforms and expression<br />

There is evidence for calsequestr<strong>in</strong> expression, at<br />

vary<strong>in</strong>g levels, <strong>in</strong> the follow<strong>in</strong>g smooth muscles: stomach<br />

(585, 730, 778), vas deferens (729, 730), aorta (730), ileum<br />

(585), and trachea (585). It may not be present <strong>in</strong> bladder<br />

(730) or pulmonary artery (585) and is either absent or <strong>in</strong><br />

trace amounts <strong>in</strong> uterus (472).<br />

It is now appreciated that two genes exist for calsequestr<strong>in</strong><br />

expression, lead<strong>in</strong>g to two isoforms which are<br />

65% identical (622). These isoforms are often referred to<br />

as the fast-twitch skeletal and cardiac forms (182, 622),<br />

but slow skeletal muscles express the fast form (181) and<br />

it appears that many smooth muscles express both iso-<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

forms (730). The functional significance of smooth muscles<br />

express<strong>in</strong>g the two isoforms of calsequestr<strong>in</strong> or neither<br />

rema<strong>in</strong>s to be established.<br />

Calsequestr<strong>in</strong> <strong>in</strong> skeletal muscle b<strong>in</strong>ds 40–50 Ca<br />

ions with a b<strong>in</strong>d<strong>in</strong>g constant of 1 mM and a high off-rate<br />

(430). The cardiac isoform b<strong>in</strong>ds 20 Ca ions with an<br />

aff<strong>in</strong>ity of 0.5 mM (51). In striated muscles calsequestr<strong>in</strong><br />

b<strong>in</strong>ds to other prote<strong>in</strong>s, <strong>in</strong>clud<strong>in</strong>g triad<strong>in</strong> and junct<strong>in</strong> (see<br />

sect. IVG), lead<strong>in</strong>g to a physical association with RyRs<br />

(233, 443). It may be that this arrangement enables the Ca<br />

of the SR to be <strong>in</strong> the right place, i.e., close to the release<br />

channel (187). Regular arrays of calsequestr<strong>in</strong> with Ca<br />

attached appear as crystall<strong>in</strong>e structures <strong>in</strong> the SR lumen<br />

(607). Calsequestr<strong>in</strong> has been shown to <strong>in</strong>crease the RyR<br />

open probability <strong>in</strong> skeletal muscle (343).<br />

Wuytack et al. (778) were the first to report that<br />

calsequestr<strong>in</strong> was present <strong>in</strong> SR from smooth muscle.<br />

They exam<strong>in</strong>ed pig antrum and concluded it conta<strong>in</strong>ed<br />

the cardiac form of calsequestr<strong>in</strong> <strong>in</strong> low amounts compared<br />

with heart, although commensurate with the lower<br />

SERCA expression <strong>in</strong> smooth muscle compared with<br />

heart. These authors also noted that calsequestr<strong>in</strong> <strong>in</strong> portal<br />

ve<strong>in</strong> smooth muscles had first been proposed, on<br />

morphological grounds, by Somlyo (652) and Franz<strong>in</strong>i-<br />

Armstrong (187). Vas deferens expresses both calsequestr<strong>in</strong><br />

isoforms <strong>in</strong> about equal amounts (730). Its distribution<br />

was reported to be clustered at discrete lum<strong>in</strong>al sites<br />

and on SR located superficially below the plasma membrane<br />

and rich <strong>in</strong> IP 3R (729). Thus a specific distribution<br />

of calsequestr<strong>in</strong> <strong>in</strong> smooth muscle as reported also for<br />

striated muscles may occur (322, 323). This contrasts with<br />

the nondiscrete distribution of calreticul<strong>in</strong> (see below).<br />

The colocalization of IP 3R with calsequestr<strong>in</strong> presumably<br />

aids rapid release of messenger Ca <strong>in</strong> smooth muscles<br />

express<strong>in</strong>g both these elements. Moore et al. (491) demonstrated<br />

<strong>in</strong> toad stomach that calsequestr<strong>in</strong> was not only<br />

present <strong>in</strong> the SR <strong>in</strong> a discrete manner, but that it was<br />

associated with the superficial SR. They also demonstrated<br />

a close association between calsequestr<strong>in</strong> and the<br />

NCX and Na pump distribution on the plasma membrane,<br />

<strong>in</strong> caveolae. Thus not only is calsequestr<strong>in</strong> close to SR Ca<br />

release channels but also to exchangers important to Ca<br />

homeostasis <strong>in</strong> smooth muscle, and contribut<strong>in</strong>g to microdoma<strong>in</strong>s<br />

(as discussed <strong>in</strong> sect. IID).<br />

Despite these several publications from a number of<br />

different laboratories, a paper appeared stat<strong>in</strong>g that calreticul<strong>in</strong><br />

and not calsequestr<strong>in</strong> was the major Ca b<strong>in</strong>d<strong>in</strong>g<br />

prote<strong>in</strong> <strong>in</strong> smooth muscle SR (472). This study however<br />

only exam<strong>in</strong>ed one type of smooth muscle, pig uterus, and<br />

actually reported that calsequestr<strong>in</strong> could be detected <strong>in</strong><br />

uter<strong>in</strong>e tissue extracts but not SR vesicles. They also used<br />

calsequestr<strong>in</strong> antibodies on cultured uter<strong>in</strong>e cells and<br />

reported negligible sta<strong>in</strong><strong>in</strong>g, but given the cultur<strong>in</strong>g and a<br />

switch to a noncontractile, synthetic state, these data are<br />

difficult to <strong>in</strong>terpret. This publication appears to have


stimulated Meldolesi’s laboratory to reexam<strong>in</strong>e, confirm,<br />

and extend their earlier f<strong>in</strong>d<strong>in</strong>gs of calsequestr<strong>in</strong> expression<br />

<strong>in</strong> vas deferens (730). They aga<strong>in</strong> demonstrated both<br />

isoforms of calsequestr<strong>in</strong> are present <strong>in</strong> vas deferens and<br />

also expressed <strong>in</strong> stomach and aorta, particularly the<br />

skeletal muscle isoform. In another study comb<strong>in</strong><strong>in</strong>g ultrastructure<br />

and antibody label<strong>in</strong>g, the presence of calsequestr<strong>in</strong><br />

<strong>in</strong> gu<strong>in</strong>ea pig vas deferens was confirmed, as was<br />

its absence from aorta (515). Aga<strong>in</strong> the calsequestr<strong>in</strong> distribution<br />

was similar to that of IP 3R.<br />

3. <strong>Function</strong><br />

Although calsequestr<strong>in</strong> has been demonstrated <strong>in</strong><br />

many smooth muscles, there appears to have been no<br />

recent work <strong>in</strong>vestigat<strong>in</strong>g its function and role <strong>in</strong> Ca signal<strong>in</strong>g.<br />

Thus its functions, as a b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> for Ca, as<br />

a part of an SR complex with Ca release channels (particularly<br />

RyRs), and triad<strong>in</strong> and junct<strong>in</strong> (see sect. IVG), and<br />

as a sensor of lum<strong>in</strong>al Ca, can largely only be assumed by<br />

analogy to studies <strong>in</strong> striated muscles, with all the caveats<br />

that this requires.<br />

In summary, it seems reasonable to conclude that<br />

many, but not all, smooth muscles express calsequestr<strong>in</strong><br />

and that those that do express the two different isoforms<br />

<strong>in</strong> a tissue-specific manner. It is not possible to do anyth<strong>in</strong>g<br />

other than speculate about how these differences<br />

may relate to differences <strong>in</strong> Ca signal<strong>in</strong>g or function and<br />

pathology <strong>in</strong> different smooth muscles.<br />

F. Calreticul<strong>in</strong><br />

1. Introduction<br />

Calreticul<strong>in</strong> is a 46-kDa prote<strong>in</strong>, the product of a<br />

s<strong>in</strong>gle gene, and has been the subject of recent reviews<br />

focused on its role with<strong>in</strong> the ER and skeletal muscle<br />

(164, 320, 419, 468, 761). It was first described <strong>in</strong> 1974 as<br />

a high-aff<strong>in</strong>ity Ca b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> of the SR b<strong>in</strong>d<strong>in</strong>g 25 mol<br />

Ca/mol prote<strong>in</strong> (531) and its cDNA code was revealed <strong>in</strong><br />

1989 (180, 647). Unlike calsequestr<strong>in</strong> it conta<strong>in</strong>s an ER<br />

retention signal. It was <strong>in</strong>itially isolated from the SR and<br />

its Ca b<strong>in</strong>d<strong>in</strong>g ability noted, which led to Ca b<strong>in</strong>d<strong>in</strong>g be<strong>in</strong>g<br />

viewed as its major function. As discussed <strong>in</strong> the reviews<br />

cited above, it is now appreciated that calreticul<strong>in</strong> has<br />

many functions, <strong>in</strong>clud<strong>in</strong>g chaperon<strong>in</strong>g of ER prote<strong>in</strong>s,<br />

and that it is also expressed <strong>in</strong> other cellular organelles<br />

and at the cell surface. A list of its functions would now<br />

<strong>in</strong>clude cell adhesion (e.g., Ref. 320), antithrombotic activity,<br />

<strong>in</strong>duction of NO <strong>in</strong> endothelial cells, long-term<br />

memory <strong>in</strong> Aplysia, and development (see Refs. 320 and<br />

469 for review of these other functions of calreticul<strong>in</strong>).<br />

Our focus is on its role <strong>in</strong> Ca homeostasis with<strong>in</strong> the SR.<br />

SMOOTH MUSCLE SR 129<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

2. Expression<br />

In smooth muscle, calreticul<strong>in</strong> has been found to be<br />

expressed <strong>in</strong> the SR lumen of several different tissues. In<br />

1992 Thar<strong>in</strong> et al. (697) reported its widespread distribution<br />

<strong>in</strong> rabbit tissues, especially smooth muscle (aorta,<br />

uterus). Calreticul<strong>in</strong> has been described as the major Ca<br />

buffer <strong>in</strong> smooth muscle (472), but as already noted, this<br />

study only exam<strong>in</strong>ed uter<strong>in</strong>e tissue, which expresses calreticul<strong>in</strong><br />

and only trace amounts of calsequestr<strong>in</strong>. In their<br />

study of vas deferens, Villa et al. (729) found both Cab<strong>in</strong>d<strong>in</strong>g<br />

prote<strong>in</strong>s present and reported that calreticul<strong>in</strong><br />

was widely distributed throughout the SR, whereas calsequestr<strong>in</strong><br />

had a more discrete distribution and overlapped<br />

with IP3Rs. A recent study of human esophageal smooth<br />

muscle has detected a particularly strong calreticul<strong>in</strong> signal<br />

(stronger than for cardiac muscle), as well as calsequestr<strong>in</strong><br />

but not phospholamban (177). Both Ca b<strong>in</strong>d<strong>in</strong>g<br />

prote<strong>in</strong>s decreased <strong>in</strong> patients with reduced gastro<strong>in</strong>test<strong>in</strong>al<br />

motility and elevated esophageal pressures.<br />

3. <strong>Function</strong><br />

Calreticul<strong>in</strong> may specifically affect the activity of<br />

SERCA2b, suggest<strong>in</strong>g some specificity of function <strong>in</strong><br />

smooth muscles (318). SERCA2b has an additional transmembrane<br />

segment (see sect. IIIC) that is thought to conta<strong>in</strong><br />

a calreticul<strong>in</strong> b<strong>in</strong>d<strong>in</strong>g site. It has been suggested that<br />

when SR [Ca] is low, calreticul<strong>in</strong> is not bound to SERCA<br />

and thus Ca enters the SR as SERCA is maximally active.<br />

When SR [Ca] is high, calreticul<strong>in</strong> b<strong>in</strong>ds to the lum<strong>in</strong>al tail<br />

of SERCA2b and decreases its activity (468).<br />

In vascular smooth muscle (aorta), calreticul<strong>in</strong> <strong>in</strong>creased<br />

when glucose concentration was elevated, both<br />

<strong>in</strong> vitro and <strong>in</strong> vivo (699). These effects however may be<br />

due to calreticul<strong>in</strong>’s chaperone role and a destabilization<br />

of GLUT1 mRNA, rather than direct effects on Ca signal<strong>in</strong>g<br />

<strong>in</strong> the myocytes. Daniel’s group work<strong>in</strong>g on airway<br />

and esophageal smooth muscle (142, 143) have presented<br />

evidence that calreticul<strong>in</strong> and calsequestr<strong>in</strong> are colocalized<br />

and precipitated <strong>in</strong> the detergent-resistant membrane<br />

fraction <strong>in</strong>dicative of caveolae, along with L-type Ca channels.<br />

While these data await confirmation with additional<br />

techniques to ensure that the immunoprecipitation with<br />

caveol<strong>in</strong> is not due to their loss from the SR dur<strong>in</strong>g<br />

membrane isolation, they <strong>in</strong>dicate that these Ca-b<strong>in</strong>d<strong>in</strong>g<br />

prote<strong>in</strong>s may <strong>in</strong>fluence caveolar function and <strong>in</strong>teractions<br />

with the peripheral SR and may contribute to Ca microdoma<strong>in</strong>s<br />

with<strong>in</strong> the smooth muscle cell. The f<strong>in</strong>d<strong>in</strong>g of calreticul<strong>in</strong><br />

outside the SR/ER has been reported <strong>in</strong> other cell<br />

types and expla<strong>in</strong>s its role <strong>in</strong> processes such as cell<br />

adhesion (320, 468).<br />

Us<strong>in</strong>g a Xenopus oocyte expression system, calreticul<strong>in</strong><br />

was found to suppress IP3-<strong>in</strong>duced Ca oscillations,<br />

suggestive of it act<strong>in</strong>g to <strong>in</strong>hibit Ca uptake by SERCA2b<br />

(318). It has been estimated that 50% of Ca <strong>in</strong> the ER is


130 SUSAN WRAY AND THEODOR BURDYGA<br />

bound to calreticul<strong>in</strong> (501), and if its expression is deficient,<br />

then the ER Ca storage capacity is decreased and<br />

agonist-evoked Ca release is <strong>in</strong>hibited (467, 501). Conversely,<br />

overexpression leads to <strong>in</strong>creased Ca storage<br />

capacity and a decrease <strong>in</strong> store-operated Ca <strong>in</strong>flux (466).<br />

Empty<strong>in</strong>g of the ER Ca store stimulates expression of<br />

calreticul<strong>in</strong> (744). Thus changes <strong>in</strong> calreticul<strong>in</strong> concentration<br />

may be expected to have a direct effect on Ca signal<strong>in</strong>g,<br />

as well as <strong>in</strong>direct ones, as its chaperone function<br />

is also affected (470). Cardiac development is so impaired<br />

<strong>in</strong> calreticul<strong>in</strong> knockout mice that they die at the embryonic<br />

stage (467). However, <strong>in</strong> adult heart, the level of<br />

calreticul<strong>in</strong> expression is low, and its overexpression produces<br />

arrhythmias, heart block, and death (500).<br />

G. Triad<strong>in</strong> and Junct<strong>in</strong><br />

While many prote<strong>in</strong>s have been identified <strong>in</strong> the SR<br />

lumen (51, 424), two that are abundant and <strong>in</strong>tr<strong>in</strong>sic to the<br />

SR membrane are triad<strong>in</strong> and junct<strong>in</strong> (38, 340). They<br />

appear to anchor calsequestr<strong>in</strong> to the junctional face of<br />

the SR membrane, perhaps to keep it close to Ca release<br />

channels. The b<strong>in</strong>d<strong>in</strong>g to calsequestr<strong>in</strong> is disrupted by low<br />

(10 M) and high (10 mM) [Ca] (803). It is also possible<br />

that these two prote<strong>in</strong>s are SR lum<strong>in</strong>al Ca sensors<br />

and modulators of RyR open probability (51, 359, 803). To<br />

date, triad<strong>in</strong> and junct<strong>in</strong> do not appear to have been<br />

described <strong>in</strong> smooth muscle. This may be a consequence<br />

of the lack of triad and diad arrangement of the SR <strong>in</strong><br />

smooth muscle, but it may also be due to lower expression<br />

levels, methodological difficulties (443), or that different<br />

isoforms occur. For example, recent data have<br />

identified new triad<strong>in</strong> isoforms, which colocalize with<br />

IP 3R (721). Thus there may be an IP 3R-specific complex<br />

that could be <strong>in</strong> smooth muscle. Similarly triad<strong>in</strong> and<br />

junction by mediat<strong>in</strong>g <strong>in</strong>teractions with calsequestr<strong>in</strong><br />

could have function <strong>in</strong> smooth muscle if expressed there<br />

(237).<br />

H. Summary<br />

In summary, while it is clear that for the SR to<br />

function as a Ca store the Ca b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s calreticul<strong>in</strong><br />

and calsequestr<strong>in</strong> are required, little data are available<br />

<strong>in</strong>vestigat<strong>in</strong>g the functional effects of alter<strong>in</strong>g their expression<br />

<strong>in</strong> smooth muscle. Nor is it clear what advantages<br />

or specificity is conferred on smooth muscles by<br />

their express<strong>in</strong>g the different prote<strong>in</strong>s or their isoforms.<br />

Little recent data have been obta<strong>in</strong>ed <strong>in</strong> this field despite<br />

the <strong>in</strong>creased <strong>in</strong>terest <strong>in</strong> how lum<strong>in</strong>al Ca content affects<br />

Ca signal<strong>in</strong>g and functions <strong>in</strong> smooth muscle. Triad<strong>in</strong> and<br />

junct<strong>in</strong> may not be expressed <strong>in</strong> smooth muscle, but<br />

verification of this would be helpful. We concur with the<br />

words of Volpe et al. (730) written more than a decade<br />

ago on this issue that “it is therefore possible that what at<br />

present appears to be no more than a complex pleiotropism<br />

could ultimately be attributed to specific physiological<br />

characteristics of the various smooth muscles.” We<br />

have however come no closer to exam<strong>in</strong><strong>in</strong>g this possibility<br />

<strong>in</strong> the <strong>in</strong>terven<strong>in</strong>g years.<br />

V. CALCIUM RELEASE CHANNELS<br />

A. Introduction<br />

Ca release from the SR <strong>in</strong> smooth muscle cells occurs<br />

though activation of two families of Ca release channels:<br />

RyR and IP 3R channels, which have substantial similarities<br />

<strong>in</strong> structure (113, 657). These channels are <strong>in</strong>volved <strong>in</strong><br />

control of various functions <strong>in</strong> smooth muscle cells <strong>in</strong>clud<strong>in</strong>g<br />

contraction, relaxation, proliferation, and differentiation<br />

(31). The functional role of RyR and IP 3R channels<br />

<strong>in</strong> these processes critically depends on molecular<br />

identity, level and proportion of expression, subcellular<br />

distribution, and the types of functional units they form<br />

with other cellular structures. <strong>Smooth</strong> muscles differ by<br />

types (vascular and visceral), location (different vascular<br />

beds or various hollow organs), size (resistance versus<br />

conduit arteries), orientation (circular versus longitud<strong>in</strong>al),<br />

and function (phasic versus tonic), and it is not<br />

surpris<strong>in</strong>g that the data obta<strong>in</strong>ed so far <strong>in</strong>dicate marked<br />

differences <strong>in</strong> expression, spatial distribution, and subcellular<br />

location of different isoforms of RyRs and IP 3Rs and<br />

that these underlie the generation of tissue- or cell-specific<br />

Ca responses (224, 292, 493, 765, 794). NAADP is an<br />

agonist at RyRs, but there may also be a third Ca release<br />

channel, which is discussed <strong>in</strong> the section VD.<br />

B. Ryanod<strong>in</strong>e Receptors<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

The RyR is a homotetramer with the (565 kDa)<br />

subunits surround<strong>in</strong>g a central Ca pore. The subunits act<br />

<strong>in</strong> a coord<strong>in</strong>ated way to gate the Ca channel and are also<br />

each associated with an important 12-kDa regulatory<br />

b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> FKBP (427, 461). Cryo-EM and threedimensional<br />

reconstruction reveal that RyR is a symmetrical<br />

mushroom like structure with a large cytosolic assembly<br />

and a short region that traverses the SR membrane<br />

(610, 624). The ion channel-form<strong>in</strong>g, membranespann<strong>in</strong>g<br />

regions are highly conserved between different<br />

RyR isoforms and are localized to the COOH term<strong>in</strong>us.<br />

The cytosolic doma<strong>in</strong> consists of 80% of the mass of the<br />

RyRs. It assumes a quatrefoil shape and is the modulatory<br />

region of the molecule, conta<strong>in</strong><strong>in</strong>g b<strong>in</strong>d<strong>in</strong>g sites for Ca,<br />

aden<strong>in</strong>e nucleotides, calmodul<strong>in</strong>, FKBPs, as well as phosphorylation<br />

sites (624). The massive size of RyRs makes<br />

them physically the largest ion channels (almost twice as


large as the IP 3R, their closest relatives). Because of its<br />

short and wide channel region, the RyR is suitable for<br />

sudden and large release of Ca from <strong>in</strong>tracellular stores.<br />

The Ca conductance of RyR is on the order of 100 pS,<br />

and Ca is their pr<strong>in</strong>cipal endogenous effector. Studies<br />

describ<strong>in</strong>g the Ca dependence of channel activity <strong>in</strong> isolated<br />

preparations showed that there are sites with<strong>in</strong> the<br />

prote<strong>in</strong> where Ca can b<strong>in</strong>d and <strong>in</strong>hibit as well as activate<br />

the channels (460). Data obta<strong>in</strong>ed us<strong>in</strong>g purified channels<br />

reconstituted <strong>in</strong>to planar lipid bilayers and from [ 3 H]ryanod<strong>in</strong>e<br />

b<strong>in</strong>d<strong>in</strong>g studies, demonstrate that RyR1 and RyR2<br />

are activated by sub- to low micromolar levels of [Ca] and<br />

<strong>in</strong>activated by micromolar to millimolar [Ca] <strong>in</strong> the cytoplasm.<br />

The RyR2 isoform is less sensitive to <strong>in</strong>activation<br />

by calcium than RyR1 (130, 460). The steady-state Ca<br />

dependencies of isolated s<strong>in</strong>gle RyRs are nearly identical<br />

(238). Similar to RyR1, RyR3 was shown to also be sensitive<br />

to caffe<strong>in</strong>e, aden<strong>in</strong>e nucleotides, and ryanod<strong>in</strong>e.<br />

However, it has the lowest Ca sensitivity of the RyR<br />

family. This led to the suggestion that it serves as a<br />

high-threshold Ca release channel (687). The lum<strong>in</strong>al sensitivity<br />

of RyR3 to Ca was also lower than that of RyR1<br />

and RyR2 (669).<br />

1. Expression<br />

The RyR specifically b<strong>in</strong>ds the plant alkaloid ryanod<strong>in</strong>e,<br />

which is the reason for its name. Each of the three<br />

isoforms of RyRs are encoded by a dist<strong>in</strong>ct gene (670).<br />

RyR1 and RyR2 are predom<strong>in</strong>antly expressed <strong>in</strong> skeletal<br />

(812) and cardiac (532, 599) muscle, respectively, while<br />

RyR3 appears to be expressed <strong>in</strong> a variety of tissues (211,<br />

243, 389). Unlike cardiac and skeletal muscle, which express<br />

only one RyR isoform, all three RyR isoforms can be<br />

expressed <strong>in</strong> smooth muscles, with the type and the relative<br />

proportion of expression of each be<strong>in</strong>g tissue and<br />

species dependent. Expression of all three isoforms has<br />

been reported for smooth muscles of the pulmonary and<br />

systemic vasculatures (133, 475, 511, 787, 805) and <strong>in</strong><br />

pregnant human and rat myometrium (439, 440). Some<br />

types of smooth muscles coexpress only two RyR isoforms:<br />

RyR1 and RyR3 are subtypes identified <strong>in</strong> airway<br />

(158), and RyR2 and RyR3 are subtypes found <strong>in</strong> rat aortic<br />

myocytes (340, 351) and duodenum (138). Expression of<br />

just the RyR2 isoform was reported for smooth muscle<br />

cells of ur<strong>in</strong>ary bladder (115, 490), toad stomach (781),<br />

and cerebral artery (213), and only the RyR3 isoform is<br />

expressed <strong>in</strong> rat ureteric (60) and human nonpregnant<br />

(24) and mouse pregnant uter<strong>in</strong>e (358) smooth muscle<br />

cells. An alternative splic<strong>in</strong>g of RyR3 subtype was recently<br />

reported for some types of smooth muscles (138,<br />

139). In mouse, spliced short isoform (RyR3s), without<br />

channel function, are coexpressed with nonspliced functional<br />

full-length isoforms of RyR3 (RyR3L) (139). Mouse<br />

duodenum myocytes express RyR2 and spliced and non-<br />

SMOOTH MUSCLE SR 131<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

spliced RyR3 isoforms (138). The functional significance<br />

of these nonfunction<strong>in</strong>g RyRs is discussed below.<br />

Cultur<strong>in</strong>g of smooth muscle cells (132, 444, 711) or<br />

tissues (154) alters the patterns and levels of RyR expression<br />

<strong>in</strong> smooth muscle cells. Changes <strong>in</strong> RyR expression<br />

have been reported <strong>in</strong> some smooth muscle pathologies,<br />

e.g., a loss of RyR2 expression was reported for hyperactive<br />

detrusor smooth muscle (316).<br />

2. Localization<br />

The spatial arrangement of RyRs <strong>in</strong> smooth muscle<br />

cells is <strong>in</strong> marked contrast to the regular distribution of<br />

RyRs <strong>in</strong> sarcomeric muscles. A helical arrangement of<br />

well-developed superficial SR was reported <strong>in</strong> live smooth<br />

muscle cells double sta<strong>in</strong>ed with fluorescently labeled<br />

<strong>in</strong>dicators for the SR and RyRs (221). Immunofluorescence<br />

and immuno-EM studies also show that the subcellular<br />

distribution of RyRs closely follows that of the SR<br />

(394, 490). Both a peripheral and cytoplasmic distribution<br />

of RyRs is found <strong>in</strong> tonic smooth muscle (394), whereas<br />

the distribution is largely peripheral <strong>in</strong> phasic smooth<br />

muscles [vas deferens (394), ur<strong>in</strong>ary bladder (490)]. A<br />

transition from a diffuse cytoplasmic distribution of functional,<br />

but nonspark<strong>in</strong>g, RyR2 <strong>in</strong> neonatal cerebral arteries,<br />

to a dist<strong>in</strong>ct distribution <strong>in</strong> peripheral clusters <strong>in</strong> adult<br />

arteries, with the production of Ca sparks, <strong>in</strong>dicates that<br />

Ca spark-generat<strong>in</strong>g units of RyRs appear late <strong>in</strong> tissue<br />

differentiation and are conf<strong>in</strong>ed to peripheral regions of<br />

the cell (213). Dist<strong>in</strong>ct RyR2 sta<strong>in</strong><strong>in</strong>g <strong>in</strong> close proximity to<br />

the plasma membrane colocalized with voltage-gated Ltype<br />

Ca channels and BK has been reported for gu<strong>in</strong>ea pig<br />

ur<strong>in</strong>ary bladder myocytes (490) and vas deferens (525).<br />

Rat pulmonary artery myocytes that express all three<br />

subtypes of RyRs have RyR2, their predom<strong>in</strong>ant isoform<br />

located ma<strong>in</strong>ly <strong>in</strong> peripheral sites, and RyR3 <strong>in</strong> the per<strong>in</strong>uclear<br />

region and RyR1s <strong>in</strong> both regions (787). In rat portal<br />

ve<strong>in</strong>, the antibody-sta<strong>in</strong><strong>in</strong>g pattern of RyR3 is diffuse,<br />

unlike that of RyR1 and RyR2 which exhibit discrete areas<br />

of higher density sta<strong>in</strong><strong>in</strong>g (475). A diffuse distribution of<br />

RyR3 was also reported for smooth muscle cells of duodenum<br />

(138) and nonpregnant mouse myometrium (476).<br />

The exist<strong>in</strong>g histological data would fit a model of<br />

peripherally located collections of RyR2 and RyR1 and<br />

more diffuse distribution of RyR3 <strong>in</strong> smooth muscle cells.<br />

3. RyR function<br />

The physiological contribution of different RyR subtypes<br />

to Ca signal<strong>in</strong>g <strong>in</strong> smooth muscle cells has been<br />

recently addressed us<strong>in</strong>g RyR knockout mice (314, 805)<br />

and antisense oligonucleotides specifically target<strong>in</strong>g each<br />

of the subtypes (138, 139, 407, 476). In portal ve<strong>in</strong>, <strong>in</strong>jection<br />

of myocytes with either RyR1 or RyR2 antisense<br />

oligonucleotides abolished Ca sparks, suggest<strong>in</strong>g RyR1<br />

and RyR2 may colocalize to form Ca spark sites, while


132 SUSAN WRAY AND THEODOR BURDYGA<br />

suppression of RyR3 did not alter Ca spark activity. In<br />

ur<strong>in</strong>ary bladder myocytes, RyR2s are required for generation<br />

of Ca sparks (314). Native RyR3s are either not<br />

<strong>in</strong>volved (133, 314) or may even have an <strong>in</strong>hibitory effect<br />

on Ca sparks (407). However, when both RyR1 and RyR2<br />

are <strong>in</strong>hibited with antisense oligonucleotides, and under<br />

conditions of <strong>in</strong>creased SR Ca load<strong>in</strong>g, RyR3 can be activated<br />

by caffe<strong>in</strong>e or agonists (475, 476). RyR3 gene knockout<br />

significantly <strong>in</strong>hibits the contractile response to hypoxia<br />

but not the norep<strong>in</strong>ephr<strong>in</strong>e-<strong>in</strong>duced Ca and contractile<br />

responses, <strong>in</strong> pulmonary artery smooth muscle cells<br />

(805).<br />

The function of RyR3 <strong>in</strong> Ca signal<strong>in</strong>g is complicated<br />

by alternative splic<strong>in</strong>g of RyR3. The expression of a short<br />

isoform of RyR3 (RyR3s) <strong>in</strong> HEK293 cells can <strong>in</strong>hibit both<br />

the full-length RyR3 (RyR3L) and RyR2 subtypes (315). In<br />

native smooth muscle, the dom<strong>in</strong>ant negative effect of the<br />

spliced isoform of RyR3 <strong>in</strong>hibits RyR2 activation and Ca<br />

signals <strong>in</strong> duodenum (138) and the activity of RyR3 <strong>in</strong><br />

mouse pregnant myometrium (139). These observations<br />

reveal a novel mechanism by which a splice variant of one<br />

RyR3 isoform may heteromerize with the other RyR variants<br />

and suppress the activity of these RyR isoforms via a<br />

dom<strong>in</strong>ant negative effect. Thus tissue-specific expression<br />

of RyR3 splice variants is likely to account for some of the<br />

pharmacological and functional heterogeneities of RyR3,<br />

for example, the lack of Ca sparks and caffe<strong>in</strong>e sensitivity<br />

<strong>in</strong> rat myometrium, despite expression of all three forms<br />

of RyRs (88).<br />

On the basis of the available <strong>in</strong>formation, RyR2 appears<br />

to be the most important component of Ca sparks <strong>in</strong><br />

smooth muscle, and recent f<strong>in</strong>d<strong>in</strong>gs on RyR3 splice variants<br />

have helped unravel some of the perplexity <strong>in</strong> Ca<br />

signal<strong>in</strong>g <strong>in</strong> smooth muscle. Calcium sparks are discussed<br />

<strong>in</strong> detail <strong>in</strong> section IX.<br />

C. IP 3-Sensitive Ca release channels<br />

B<strong>in</strong>d<strong>in</strong>g of G prote<strong>in</strong>-coupled receptor agonists to their<br />

receptors <strong>in</strong> smooth muscles leads to activation of phosphatidyl<strong>in</strong>ositol-specific<br />

phospholipase C (PLC), which catalyzes<br />

the hydrolysis of phosphatidyl<strong>in</strong>ositol 4,5-bisphosphate<br />

(PIP 2) to yield IP 3 and diacylglycerol (DAG) (48). The<br />

IP 3 releases Ca from <strong>in</strong>tracellular stores by b<strong>in</strong>d<strong>in</strong>g to IP 3R,<br />

which are IP 3-gated Ca release channels, while DAG activates<br />

prote<strong>in</strong> k<strong>in</strong>ase C (PKC). Activation of IP 3Rs via activation<br />

of L-type Ca channels <strong>in</strong>dependent of Ca entry, socalled<br />

metabotropic Ca channel-<strong>in</strong>duced Ca release, has<br />

been suggested <strong>in</strong> arterial myocytes but requires confirmation<br />

by other studies (149), and is not discussed further.<br />

1. Expression<br />

Three dist<strong>in</strong>ct IP3R gene products (types 1–3) have<br />

been identified <strong>in</strong> different mammalian species (283, 546).<br />

All three isoforms of IP 3R are structurally and functionally<br />

related (193). The IP 3R isoforms differ <strong>in</strong> their sensitivities<br />

to breakdown by cellular proteases (IP 3R2 relatively<br />

more resistant that IP 3R1 and IP 3R3) (765). These<br />

receptors exist as tetrameric structures, with a monomeric<br />

molecular mass of 300 kDa. Unlike RyRs, IP 3Rs<br />

are poorly expressed <strong>in</strong> skeletal and cardiac muscle but<br />

extensively expressed <strong>in</strong> bra<strong>in</strong> and other tissues <strong>in</strong>clud<strong>in</strong>g<br />

smooth muscles (317, 471, 498, 546, 695, 760, 794). In<br />

smooth muscles, the density of IP 3Ris100 times less<br />

than <strong>in</strong> bra<strong>in</strong> (437, 802). In visceral smooth muscles (356,<br />

379–381), IP 3Rs are expressed <strong>in</strong> greater quantity than<br />

RyRs with an overall stoichiometric ratio of IP 3Rs to RyRs<br />

of 10–12:1, while <strong>in</strong> vascular smooth muscles the ratio<br />

is 3-4:1 (62).<br />

<strong>Smooth</strong> muscle cells express multiple IP 3R isoforms,<br />

although as with RyRs, the level of expression of different<br />

subtypes is tissue and species dependent. Us<strong>in</strong>g reversetranscriptase<br />

PCR analysis, Morel et al. (493) showed that<br />

rat portal ve<strong>in</strong> expressed IP 3R1 and IP 3R2, whereas rat<br />

ureter expressed predom<strong>in</strong>antly IP 3R1 and IP 3R3 (493),<br />

although <strong>in</strong> an earlier study it was reported that rat ureteric<br />

myocytes expressed all three isoforms of IP 3Rs (60).<br />

In the thoracic aorta and mesenteric arteries, IP 3R1 was<br />

the only isoform found, while IP 3R1 and weak expression<br />

of IP 3R2 were reported for smooth muscle cells of basilar<br />

artery (324). The type 1 isoform predom<strong>in</strong>ates <strong>in</strong> gu<strong>in</strong>ea<br />

pig <strong>in</strong>test<strong>in</strong>al smooth muscle cells (222) as well as <strong>in</strong> adult<br />

porc<strong>in</strong>e (297) and rat aorta (693), whereas type 3 predom<strong>in</strong>ates<br />

<strong>in</strong> neonatal aorta (693). Subtype-specific IP 3R antibodies<br />

revealed that the expression of IP 3R1 was similar<br />

<strong>in</strong> cultured aortic cells and aorta homogenate, but expression<br />

of IP 3R2 and IP 3R3 types was <strong>in</strong>creased threefold <strong>in</strong><br />

cultured cells (693). Immunosta<strong>in</strong><strong>in</strong>g and functional studies<br />

performed on isolated cells and cells <strong>in</strong> situ <strong>in</strong> rat<br />

portal ve<strong>in</strong> showed two subpopulations of cells, one<br />

which predom<strong>in</strong>antly expressed IP 3R1 and a second predom<strong>in</strong>antly<br />

express<strong>in</strong>g IP 3R2. Dist<strong>in</strong>ct patterns of the Ca<br />

responses were generated by the two subpopulations of<br />

myocytes <strong>in</strong> response to agonist (493).<br />

2. Localization<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

In some smooth muscles, IP 3R1 has been localized<br />

throughout the cell, i.e., central and peripheral SR, as<br />

visualized us<strong>in</strong>g EM (191, 515, 693, 729), and <strong>in</strong> the others<br />

it had a ma<strong>in</strong>ly subplasmalemmal location (222). IP 3R2<br />

has a diffuse cytoplasmic distribution, similar to IP 3R1<br />

(and IP 3R3), but also occurs as dense patches <strong>in</strong> the<br />

peripheral cytoplasm, a pattern that IP 3R1 did not exhibit<br />

(668). IP 3Rs were also arranged <strong>in</strong> clusters <strong>in</strong> rat ureteric<br />

myocytes, as well as throughout the cell (60). In vascular<br />

myocytes, IP 3R2 is distributed peripherally and associated<br />

with the nucleus <strong>in</strong> proliferat<strong>in</strong>g cells (668, 694). In<br />

portal ve<strong>in</strong>, IP 3R2 was also found closely associated with


the nucleus and at the plasma membrane, whereas the<br />

IP 3R3 was distributed predom<strong>in</strong>antly around the nucleus<br />

(209).<br />

Interest<strong>in</strong>gly, IP 3Rs are also localized <strong>in</strong> structures<br />

other than the ER/SR. For <strong>in</strong>stance, they have been found<br />

<strong>in</strong> the nuclear envelope <strong>in</strong> many cell types, <strong>in</strong>clud<strong>in</strong>g<br />

vascular (694) and visceral (729) smooth muscles. This<br />

may be important <strong>in</strong> regulat<strong>in</strong>g <strong>in</strong>tracellular calcium release<br />

around the nucleus when vascular smooth muscle<br />

cells switch to a more proliferat<strong>in</strong>g phenotype. In myocytes<br />

from basilar artery, IP 3R1 sta<strong>in</strong><strong>in</strong>g was detected<br />

close to both the nuclear and plasma membranes. IP 3R1<br />

was also observed throughout the cytoplasm <strong>in</strong>term<strong>in</strong>gled<br />

with the act<strong>in</strong> filaments. This observation suggests that an<br />

extensive SR network spreads from the nuclear region,<br />

throughout the cell, and comes <strong>in</strong>to close association with<br />

the plasma membrane. Localization of IP 3Rs close to the<br />

cell membrane may account for the activation of Ca<strong>in</strong>duced<br />

Ca entry via nonselective cation channels (381),<br />

calcium-dependent potassium (34), or chloride channels<br />

(239).<br />

3. Role of IP 3Rs<br />

Interaction of IP 3 with the IP 3R is a pr<strong>in</strong>cipal mechanism<br />

for mediat<strong>in</strong>g the release of Ca from the ER/SR <strong>in</strong><br />

response to agonist stimulation. The IP 3R has a s<strong>in</strong>gle<br />

high-aff<strong>in</strong>ity b<strong>in</strong>d<strong>in</strong>g site (K D value of 80 nM), and it is<br />

estimated that half-maximal release of Ca from the SR<br />

requires 40 nM InsP 3. The InsP 3 sensitivities of the isoforms<br />

vary to a limited extent and are ranked IP 3R1 <br />

IP 3R2 IP 3R3 (K D values of 1.5, 2.5, and 40 nM, respectively)<br />

(766). IP 3R activity not only depends on the concentration<br />

of IP 3 but also on the cytosolic concentration<br />

of Ca (283, 546). This property allows Ca release activated<br />

by the IP 3R to regulate further Ca release. IP 3R1 activity<br />

can be stimulated at [Ca] below 300 nM, while at 300 nM<br />

Ca <strong>in</strong>hibits activity. This type of biphasic Ca dependence<br />

has been reported <strong>in</strong> smooth muscle cells (52, 282) <strong>in</strong><br />

which the IP 3R1 isoform is predom<strong>in</strong>ant (283). The IP 3R2<br />

and IP 3R3 do not exhibit such marked Ca concentrationdependent<br />

<strong>in</strong>hibition (240, 586, 586). The positive regulation<br />

of the IP 3R by Ca may be largely due to a direct<br />

b<strong>in</strong>d<strong>in</strong>g of Ca to the receptor (484). The sensitivity of IP 3R<br />

is also controlled by the SR lum<strong>in</strong>al Ca content such that<br />

a reduced content also reduces IP 3-<strong>in</strong>duced Ca release<br />

(481), although recent data po<strong>in</strong>t to the cytoplasmic aspect<br />

of the channel be<strong>in</strong>g important (114).<br />

Recent studies have shown that Ca oscillations <strong>in</strong><br />

cultured rat portal ve<strong>in</strong> myocytes and rat adrenal chromaff<strong>in</strong><br />

cells depend on IP 3R2 or an <strong>in</strong>teraction between<br />

IP 3R1 and IP 3R2 (323, 326). It has been shown that coexpression<br />

of IP 3R types 1 and 2 facilitates Ca oscillations,<br />

whereas expression of IP 3R3 alone generates monophasic<br />

Ca transients (493). S<strong>in</strong>ce IP 3R types 1 and 2 are more<br />

SMOOTH MUSCLE SR 133<br />

sensitive to IP 3 and Ca, it was postulated that they are<br />

required for generation of spontaneous SR Ca release<br />

known as Ca puffs (i.e., local Ca release through IP 3Rs)<br />

and Ca oscillations (60, 493). In contrast, the IP 3R3 is<br />

activated by higher [Ca] and may participate <strong>in</strong> agonist<strong>in</strong>duced<br />

Ca waves (493).<br />

The cluster<strong>in</strong>g of IP 3Rs provides spatially discrete<br />

release sites and enables Ca signals that arise from IP 3Rs<br />

to exist with various amplitudes and locations throughout<br />

the cell (60). These localized <strong>in</strong>creases produce an <strong>in</strong>crease<br />

<strong>in</strong> [Ca] throughout the cell when puffs coalesce to<br />

form transient [Ca] <strong>in</strong>creases (Ca waves) that propagate<br />

through the cell <strong>in</strong> a regenerative manner (60, 72). Calcium<br />

puffs are discussed further <strong>in</strong> section IX.<br />

A cautionary note on us<strong>in</strong>g pharmacological <strong>in</strong>hibitors<br />

to study SR release mechanisms was made by Mac-<br />

Millan et al. (433). They found that dantrolene and tetraca<strong>in</strong>e,<br />

thought to be specific RyR blockers, could affect<br />

IP 3R Ca release, i.e., they may not be as specific as previously<br />

thought, and this will have implications when<br />

<strong>in</strong>terpret<strong>in</strong>g such data.<br />

4. Conclusions<br />

The variety, distribution, and comb<strong>in</strong>ation of IP 3Rs<br />

and RyRs <strong>in</strong> smooth muscle and their contribution to<br />

different types of signals will underlie the specificity of<br />

mechanisms, as well as the spatial and temporal parameters<br />

that are required for specificity of function <strong>in</strong> different<br />

smooth muscles. These functions <strong>in</strong>clude not just<br />

tissue-specific contraction and relaxation parameters, but<br />

also secretion, proliferation, and phenotypic change. In<br />

the follow<strong>in</strong>g section we address how these SR release<br />

processes can be modulated.<br />

D. Modulation of SR Ca Release<br />

Recently several excellent reviews have been published<br />

on the pharmacology of the RyRs and IP 3R channels<br />

(231, 384, 670); thus here for completeness we will<br />

briefly overview the basic pharmacology of the Ca release<br />

channels <strong>in</strong> smooth muscle cells. We will then consider <strong>in</strong><br />

a little more detail the roles of cADP ribose and NAADP,<br />

as evidence grows for their physiological role <strong>in</strong> smooth<br />

muscle.<br />

1. RyRs<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

RyRs have b<strong>in</strong>d<strong>in</strong>g sites for many agents, reflect<strong>in</strong>g<br />

the huge size of these channels. Methylxanth<strong>in</strong>es, imidazoles<br />

and imidazol<strong>in</strong>es, perchlorates, suram<strong>in</strong>, and volatile<br />

anesthetics all activate RyR. Inhibitors of RyRs <strong>in</strong>clude<br />

ryanod<strong>in</strong>e, ruthenium red, proca<strong>in</strong>e, tetraca<strong>in</strong>e,<br />

dantrolene, and octanol (for more details, see references<br />

<strong>in</strong> Ref. 384).


134 SUSAN WRAY AND THEODOR BURDYGA<br />

A) CAFFEINE. Caffe<strong>in</strong>e, an alkaloid methylxanth<strong>in</strong>e, <strong>in</strong>creases<br />

the RyR Ca sensitivity by <strong>in</strong>creas<strong>in</strong>g its open<br />

probability (P o) without chang<strong>in</strong>g its conductance, as<br />

shown <strong>in</strong> s<strong>in</strong>gle-channel experiments on RyR purified<br />

from cardiac (601), skeletal (600), and smooth muscles<br />

(258) reconstituted <strong>in</strong>to planar lipid bilayers. The threshold<br />

caffe<strong>in</strong>e concentration for Ca release is 250 M (404).<br />

The caffe<strong>in</strong>e-<strong>in</strong>duced Ca release is a two-step process: a<br />

small Ca release and then a second regenerative phenomenon,<br />

<strong>in</strong> which released Ca acts on a cluster of RyRs,<br />

trigger<strong>in</strong>g further Ca release. Caffe<strong>in</strong>e has long been used<br />

to deplete SR Ca stores <strong>in</strong> smooth muscle; this is frequently<br />

done <strong>in</strong> a Ca-free medium conta<strong>in</strong><strong>in</strong>g Ca chelators<br />

such as EGTA. The transient contractile response obta<strong>in</strong>ed<br />

under such conditions is a qualitative estimate of<br />

the average size of the Ca stores <strong>in</strong> the SR. Caffe<strong>in</strong>e,<br />

however, does not deplete the SR Ca store <strong>in</strong> those tissues<br />

express<strong>in</strong>g the RyR2 dom<strong>in</strong>ant negative splice variant,<br />

e.g., the uterus.<br />

In the gu<strong>in</strong>ea pig ureter, which expresses effectively<br />

only RyR (91, 92), concentrations of caffe<strong>in</strong>e for Ca release<br />

ranged from 0.5 to 20 mM (89). The use of caffe<strong>in</strong>e<br />

<strong>in</strong> smooth muscle comes with several concerns: 1) it<br />

<strong>in</strong>hibits phosphodiesterase and therefore raises cAMP<br />

concentration (341), which at concentrations exceed<strong>in</strong>g 1<br />

mM <strong>in</strong>hibits voltage-gated Ca channels; 2) it has the potential<br />

to augment capacitative Ca entry by depletion of<br />

the SR; and 3) it <strong>in</strong>hibits the SR Ca release <strong>in</strong>duced by IP 3<br />

(480) and <strong>in</strong>ositol phosphate formation (576).<br />

B) RYANODINE. Ryanod<strong>in</strong>e is a poisonous alkaloid found<br />

<strong>in</strong> the plant Ryania speciosa (670). The compound has an<br />

extremely high aff<strong>in</strong>ity for the RyR. The pharmacology of<br />

RyRs has been elegantly reviewed (231). Ryanod<strong>in</strong>e has<br />

complex concentration-dependent effects on the conductance<br />

and gat<strong>in</strong>g of s<strong>in</strong>gle RyR channels (670). It b<strong>in</strong>ds<br />

with such high aff<strong>in</strong>ity to the receptor that it was used as<br />

a label for the first purification of RyRs and gave its name<br />

to them. At nanomolar to low micromolar concentrations,<br />

ryanod<strong>in</strong>e locks the receptors <strong>in</strong> a half-open state,<br />

whereas it closes them at micromolar concentration, irreversibly<br />

<strong>in</strong>hibit<strong>in</strong>g channel open<strong>in</strong>g. There is an agreement<br />

that high-aff<strong>in</strong>ity b<strong>in</strong>d<strong>in</strong>g results <strong>in</strong> channel activation<br />

or subconductivity, whereas low-aff<strong>in</strong>ity b<strong>in</strong>d<strong>in</strong>g<br />

leads to channel <strong>in</strong>hibition (670). Ryanod<strong>in</strong>e at low concentration<br />

is frequently used to deplete the SR by caus<strong>in</strong>g<br />

the Ca release channels to rema<strong>in</strong> <strong>in</strong> a semiconduct<strong>in</strong>g<br />

state. This leak from the SR has several consequences,<br />

such as prevent<strong>in</strong>g the SR from stor<strong>in</strong>g any Ca it may<br />

accumulate, loss of Ca sparks and their effects on Casensitive<br />

ion channels, and loss of ability to generate<br />

IP 3R-mediated Ca waves and Ca oscillations <strong>in</strong> many<br />

types of smooth muscle. This last effect is due to ryanod<strong>in</strong>e<br />

deplet<strong>in</strong>g common Ca stores, i.e., shared by RyR and<br />

IP 3Rs. The b<strong>in</strong>d<strong>in</strong>g of ryanod<strong>in</strong>e to RyRs is use-dependent,<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

that is, the channels have to be <strong>in</strong> the activated state for<br />

it to be effective.<br />

C) TETRACAINE. Tetraca<strong>in</strong>e is a potent local anesthetic<br />

and an allosteric blocker of RyR channel function. At low<br />

concentrations, tetraca<strong>in</strong>e causes an <strong>in</strong>itial <strong>in</strong>hibition of<br />

spontaneous Ca release events (137), while at high concentrations<br />

it blocks release completely (236).<br />

D) RUTHENIUM RED. Ruthenium red is an organic polycationic<br />

dye that tightly b<strong>in</strong>ds to tubul<strong>in</strong> dimers and RyRs.<br />

It is a potent (nM) <strong>in</strong>hibitor of RyR (783), but also <strong>in</strong>hibits<br />

mitochondrial Ca uptake (242).<br />

2. IP3Rs Pharmacological <strong>in</strong>hibitors of IP3Rs are less abundant<br />

compared with RyRs, and unfortunately, agents used<br />

to block these receptors are nonselective. The <strong>in</strong>hibitors<br />

most commonly used <strong>in</strong>clude 2-am<strong>in</strong>oethoxy-diphenylborate<br />

(2-APB), hepar<strong>in</strong>, and xestospong<strong>in</strong>s (for a more<br />

detailed review, see Ref. 384).<br />

A) 2-APB. 2-APB is a synthetic monomer that can form<br />

a five-membered boroxazolid<strong>in</strong>e heterocyclic r<strong>in</strong>g (boroxazolidone),<br />

when an <strong>in</strong>ternal coord<strong>in</strong>ate bond is formed<br />

between the nitrogen <strong>in</strong> the ethanolam<strong>in</strong>e side cha<strong>in</strong> and<br />

the tricoord<strong>in</strong>ated boron (664). 2-APB <strong>in</strong>hibits the IP3R channel open<strong>in</strong>g without affect<strong>in</strong>g IP3 synthesis or b<strong>in</strong>d<strong>in</strong>g.<br />

Despite earlier claims to the contrary, 2-APB is not<br />

selective for IP3Rs as it reduces capacitative Ca entry and<br />

Ca efflux from mitochondria by <strong>in</strong>hibition of the NCX, and<br />

it may also block gap junctions (reviewed <strong>in</strong> detail <strong>in</strong> Ref.<br />

384).<br />

B) HEPARIN. Hepar<strong>in</strong> is a cell-impermeant <strong>in</strong>hibitor of<br />

IP3R and acts as a competitive <strong>in</strong>hibitor of IP3 <strong>in</strong> permeabilized<br />

smooth muscles (91). At low concentrations (up<br />

to 2 mM), hepar<strong>in</strong> may be specific, but at higher concentrations<br />

(20 mM), it chelates Ca and <strong>in</strong>hibits contraction<br />

(358).<br />

C) XESTOSPONGINS. The xestospong<strong>in</strong>s A, C, and D, araguspong<strong>in</strong>e<br />

B, and demethylxestospong<strong>in</strong> B are alkaloids<br />

from the Australian mar<strong>in</strong>e sponge Xestospongia sp.<br />

(199). They are cell-permeant, potent <strong>in</strong>hibitors of IP3Rs. Xestospong<strong>in</strong> C is the most potent and widely used <strong>in</strong><br />

smooth muscle studies (34, 45, 169, 483). Although more<br />

studies are required, accumulated data suggest that the<br />

various isoforms of IP3Rs differ <strong>in</strong> their sensitivities to<br />

xestospong<strong>in</strong>s. Xestospong<strong>in</strong>s are not selective and can<br />

<strong>in</strong>hibit voltage-gated Ca channels, which complicates<br />

their use.<br />

3. cADPR<br />

Cyclic ADP ribose (cADPR) is a cellular messenger<br />

for calcium signal<strong>in</strong>g (235). It is derived from nicot<strong>in</strong>amide<br />

aden<strong>in</strong>e d<strong>in</strong>ucleotide (NAD) and ADP-ribosyl cyclase<br />

(392). It is a physiological allosteric modulator of<br />

RyRs and helps stimulate CICR at low cytosolic [Ca]. RyR


activation with high concentrations of caffe<strong>in</strong>e is partly<br />

due to caffe<strong>in</strong>e mimick<strong>in</strong>g the b<strong>in</strong>d<strong>in</strong>g of cADPR to RyRs.<br />

Whether the action is by direct b<strong>in</strong>d<strong>in</strong>g to RyR or <strong>in</strong>direct<br />

(through b<strong>in</strong>d<strong>in</strong>g to FKBP) is debated. Some reports suggest<br />

that cADPR b<strong>in</strong>d<strong>in</strong>g makes FKBP, which normally<br />

b<strong>in</strong>ds RyR2, fall off RyR2. Involvement of cADPR <strong>in</strong> Ca<br />

release via RyRs has been suggested for some vascular<br />

and visceral smooth muscles (262, 757, 776), but not<br />

colonic myocytes, where Ca efflux by the Ca-ATPase was<br />

stimulated (78). Firm conclusions are difficult to draw<br />

until further studies are performed <strong>in</strong> smooth muscle and<br />

cADPR synthesis <strong>in</strong> response to agonist stimulation is<br />

demonstrated.<br />

4. NAADP<br />

NAADP was first suggested as a Ca-mobiliz<strong>in</strong>g agent<br />

after studies <strong>in</strong> sea urch<strong>in</strong> eggs (125, 391). While NAADP<br />

is similar to cADPR and may be synthesized from the<br />

same enzyme, ADP-ribosyl cyclase CD38 (but see Ref.<br />

650), it is a dist<strong>in</strong>ct Ca mobilizer, us<strong>in</strong>g RyR to amplify its<br />

signals after releas<strong>in</strong>g Ca, rather than act<strong>in</strong>g to modulate<br />

RyR release as cADPR appears to do.<br />

It has been shown that the enzymes for its synthesis<br />

and metabolism are present <strong>in</strong> smooth muscle (cultured<br />

A7r5 and mesangial cells) (795, 796). It appears to be<br />

unimportant <strong>in</strong> terms of Ca signals l<strong>in</strong>ked to contraction<br />

<strong>in</strong> some smooth muscles [tracheal, bronchial, and ileum<br />

(501)] but functionally important <strong>in</strong> others [coronary<br />

(799) and pulmonary (61) arterial, myometrial (650)]. Elucidation<br />

of its signal<strong>in</strong>g pathway <strong>in</strong> smooth muscle has<br />

been best worked out <strong>in</strong> pulmonary myocytes. In elegant<br />

experiments where myocytes were freshly isolated and<br />

then cannulated with a micropipette, NAADP was <strong>in</strong>troduced<br />

<strong>in</strong>to the cytoplasm and produced Ca bursts, which<br />

on occasion produced global Ca waves and cell shorten<strong>in</strong>g<br />

(61). The full effects of NAADP on Ca signals <strong>in</strong><br />

smooth muscle cells appear to need re<strong>in</strong>forcement from<br />

CICR via RyRs, recently identified as be<strong>in</strong>g RyR3 subtype,<br />

at least <strong>in</strong> pulmonary myocytes (350).<br />

Recent <strong>in</strong>terest has turned to identify<strong>in</strong>g the cell<br />

compartment that NAAPD receptors are located with<strong>in</strong>.<br />

Although <strong>in</strong>itially assumed to be the ER/SR and then<br />

nuclear envelope, there is grow<strong>in</strong>g evidence for a lysosomal,<br />

acidic organellar source <strong>in</strong> smooth muscle, and other<br />

tissues. The first evidence came from sea urch<strong>in</strong>s, where<br />

lysosomal-related organelles were shown to be the store<br />

released by NAADP (149). Zhang and Li (799) have s<strong>in</strong>ce<br />

demonstrated that the NAADP channel can be reconstituted<br />

<strong>in</strong> liver lysosomes. K<strong>in</strong>near et al. (350) <strong>in</strong> pulmonary<br />

arterial cells used bafilomyc<strong>in</strong>, an <strong>in</strong>hibitor of lysosomes,<br />

and showed that the Ca response to endothel<strong>in</strong>-1 but not<br />

PGF 2 was reduced. They suggested that this lysosomal<br />

store was per<strong>in</strong>uclear and situated <strong>in</strong> close proximity (0.4<br />

m) to RyRs on the SR, a “trigger zone.” NAADP alone<br />

SMOOTH MUSCLE SR 135<br />

was unable to generate global Ca waves. Similar f<strong>in</strong>d<strong>in</strong>gs,<br />

i.e., bafilomyc<strong>in</strong> decreases NAADP-<strong>in</strong>duced Ca release<br />

and contraction, were obta<strong>in</strong>ed <strong>in</strong> coronary arterial myocytes<br />

(800). These authors also used lipid bilayers to<br />

demonstrate that NAADP does not act directly on RyRs.<br />

In myometrial cells, the microsomal fraction releas<strong>in</strong>g Ca<br />

<strong>in</strong> response to NAADP was <strong>in</strong>hibited by a drug glycyl-Lphenylalan<strong>in</strong>e--naphthylamide<br />

(GPN) that lyses lysosomes,<br />

but SR <strong>in</strong>hibitors had no effect (650). This study<br />

also showed close apposition of fluorescent signals for<br />

lysosomes (lysotracker red) and the SR (BODIPY-TG) <strong>in</strong><br />

myometrial cells. In peritubular myocytes, the NAADP<br />

signal<strong>in</strong>g pathway has been shown to act via endothel<strong>in</strong>-1<br />

receptor subtype B (ET B), and lipid raft <strong>in</strong>tegrity is<br />

needed for the endothel<strong>in</strong>-NAADP signal<strong>in</strong>g pathway to<br />

operate (200), possibly due to the location of both ET B<br />

and CD38 <strong>in</strong> this compartment. Recently, Calcraft et al.<br />

(100) identified the NAADP release channel as two pore<br />

channels (TRCPs) and showed an <strong>in</strong>volvement of IP 3Rs <strong>in</strong><br />

amplify<strong>in</strong>g the NAADP Ca signals.<br />

E. SR Ca Leak<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

As with the plasma membrane, there will be some<br />

low level leak of Ca through the SR membrane, i.e., a<br />

constitutive nonregulated process. Once lum<strong>in</strong>al Ca <strong>in</strong>dicators<br />

had been developed it was possible to directly<br />

monitor the decrease <strong>in</strong> SR [Ca] that occurred <strong>in</strong> the<br />

absence of stimulation. Before this the leak was assumed<br />

to be the mechanism of SR Ca depletion follow<strong>in</strong>g <strong>in</strong>hibition<br />

of SERCA activity, and surrogates such as spark<br />

frequency were used to monitor it. That the leak of Ca<br />

from the SR or ER is not via Ca release channels (573) has<br />

been shown by, for example, block<strong>in</strong>g these channels and<br />

monitor<strong>in</strong>g a decrease <strong>in</strong> lum<strong>in</strong>al Ca content follow<strong>in</strong>g<br />

SERCA <strong>in</strong>hibition, often <strong>in</strong> permeabilized cells. Indeed, it<br />

is easy to argue that this leak, or passive efflux, must be<br />

present to balance the basal SERCA activity (266). The<br />

nature of this leak is still an enigma (101).<br />

Although the SR Ca leak will not be energy dependent,<br />

Hofer et al. (266) presented data <strong>in</strong> a fibroblast cell<br />

l<strong>in</strong>e that shows it to be ATP regulated; more Ca was lost<br />

at higher ATP concentration. This effect was very slow<br />

compared with Ca loss stimulated by IP 3. Clearly then,<br />

there can be a l<strong>in</strong>k between the metabolic state of the cell<br />

and SR [Ca]. We can speculate that if ATP is compromised,<br />

e.g., dur<strong>in</strong>g hypoxia and ischemia, then a feedback<br />

regulator such as ATP, which would reduce leak, would<br />

also therefore reduce the need for SERCA activity. It has<br />

been calculated that SERCA activity is responsible for<br />

1–3% of a cell’s energy demand (573).<br />

In the smooth muscle cell l<strong>in</strong>e A7r5, Missiaen and<br />

colleagues (478, 480) <strong>in</strong>vestigated the k<strong>in</strong>etics of the leak<br />

<strong>in</strong> the absence of IP 3. They found a nonexponential leak


136 SUSAN WRAY AND THEODOR BURDYGA<br />

that persisted after reduc<strong>in</strong>g SR Ca content to 40% of its<br />

<strong>in</strong>itial value. At extremely low [IP 3], Ca efflux via the leak<br />

exceeded IP 3-<strong>in</strong>duced Ca efflux. These authors also speculated<br />

that there is heterogeneity of Ca stores with respect<br />

to this leak, perhaps between peripheral and central<br />

SR. Additional support for a nonrelease channel leak has<br />

come from experiments <strong>in</strong> permeabilized pancreatic ac<strong>in</strong>ar<br />

cells, where <strong>in</strong>hibitors of RyR, IP 3R, and Ca release by<br />

NAADP were used, and still an unchanged basal leak<br />

occurred from the ER (408). This group also found a<br />

consistent leak rate over a broad range of SR Ca loads<br />

(486). Although it may be SERCA isoform specific, there is<br />

little reason to th<strong>in</strong>k that the leak is due to reverse mode<br />

of SERCA <strong>in</strong> smooth muscle cells, as has been suggested<br />

for cardiac muscle (627, 646).<br />

To date, there appear to have been but a handful of<br />

papers exam<strong>in</strong><strong>in</strong>g the nature and extent of the SR Ca leak<br />

<strong>in</strong> smooth muscles. The amount of Ca reported upon<br />

block<strong>in</strong>g SERCA has varied, with substantial depletion of<br />

the SR seen <strong>in</strong> cells from uterus (633), stomach (753), and<br />

(cultured) aorta (478, 706) and very little depletion <strong>in</strong><br />

bladder (218), (toad) gastric (661), and (cultured) uterus<br />

(792). These differences are consistent with estimates<br />

made <strong>in</strong> other cell types of leak rates from 10 to 200<br />

mol/m<strong>in</strong> (see Camello et al., Ref. 101).<br />

For A7r5 cells the rate of loss was, expressed as a<br />

maximal fractional Ca content loss, 22%/m<strong>in</strong> (478). Thus it<br />

is clear that <strong>in</strong> several smooth muscles, the SR leak could<br />

rapidly (few m<strong>in</strong>utes) deplete the SR of Ca. However, it<br />

should be cautioned that none of these measurements has<br />

been made under physiological conditions, as isolated,<br />

permeabilized cells, with a pharmacopeia of <strong>in</strong>hibitors<br />

are required for these studies. Thus, <strong>in</strong> smooth muscle<br />

tissues, the contribution of the leak to sett<strong>in</strong>g lum<strong>in</strong>al<br />

Ca levels and its modulation dur<strong>in</strong>g stimulation to affect<br />

functional responses rema<strong>in</strong>s to be better <strong>in</strong>vestigated.<br />

F. Summary<br />

In this section we have reviewed SR Ca release mechanisms,<br />

their modulation by pharmacological and endogenous<br />

agents, and the process of “leak.” Molecular and<br />

pharmacological tools along with knockout animals have<br />

greatly <strong>in</strong>creased our understand<strong>in</strong>g of how Ca is released<br />

from the SR of smooth muscle cells. With the existence of<br />

these different Ca-releas<strong>in</strong>g mechanisms, along with the<br />

different mechanisms of modulation, isoform expression,<br />

and cellular distribution, smooth muscle myocytes appear<br />

to be the most complex of all cells. Certa<strong>in</strong>ly striated<br />

myocytes as well as neuronal and secretory cell types are<br />

not as rich <strong>in</strong> their Ca release processes. As we have<br />

noted elsewhere, we consider this to be a reflection of the<br />

wide range of functions and phenotypic lability <strong>in</strong> smooth<br />

muscle cells.<br />

VI. LUMINAL CALCIUM<br />

A. Introduction<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

Some of the first evidence that the SR <strong>in</strong> smooth<br />

muscle is a Ca storage site was obta<strong>in</strong>ed histochemically<br />

after calcium had been precipitated (147, 259, 567). Better<br />

spatial resolution and the ability to look simultaneously at<br />

several ions came with the technique of electron-probe<br />

microanalysis, used to determ<strong>in</strong>e elemental concentrations<br />

<strong>in</strong> different regions of the myocyte. This approach<br />

led to measurements of [Ca] <strong>in</strong> the SR of 30–50 mmol/kg<br />

dry wt or 2–5 mM wet wt (69, 371), i.e., considerably<br />

higher than <strong>in</strong> the cytoplasm. These studies also detected<br />

a decrease <strong>in</strong> SR [Ca] with stimulation (655). There are,<br />

however, numerous limitations to this approach, such as<br />

the need for fixation and specialized apparatus and the<br />

small dynamic range. Determ<strong>in</strong>ation of 45 Ca efflux can<br />

provide some <strong>in</strong>dication of SR function (393, 477), but<br />

large background counts from 45 Ca bound to Ca-b<strong>in</strong>d<strong>in</strong>g<br />

prote<strong>in</strong>s, and nonspecific leaks, severely limit its usefulness.<br />

In cardiac muscle, 19 F-NMR has been used with a<br />

difluor<strong>in</strong>ated version of BAPTA. The advantage of this<br />

technique is that by alternat<strong>in</strong>g the obta<strong>in</strong><strong>in</strong>g of 19 F data<br />

with 31 P spectra, near-simultaneous <strong>in</strong>formation on ATP<br />

and phosphocreat<strong>in</strong>e and pH can also be obta<strong>in</strong>ed.<br />

B. Fluorescent Indicators for SR Lum<strong>in</strong>al<br />

Ca Measurement<br />

Other methods have been sought for measur<strong>in</strong>g SR<br />

[Ca], of which fluorescent <strong>in</strong>dicators appear the most<br />

promis<strong>in</strong>g. As Zou et al. (813) put it, “there is a strong<br />

need to develop Ca sensors capable of real-time quantitative<br />

Ca concentration measurements <strong>in</strong> specific subcellular<br />

environments without us<strong>in</strong>g natural Ca b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s<br />

such as calmodul<strong>in</strong>, which themselves participate as<br />

signal<strong>in</strong>g molecules <strong>in</strong> cells.” In this paper they describe<br />

the development of one such set of sensors with K d values<br />

rang<strong>in</strong>g from 0.4 to 2 mM. The [Ca] <strong>in</strong> the SR is such that<br />

K d values <strong>in</strong> this range are required. This along with<br />

selectivity (for Ca and the SR) and considerations of what<br />

is bound and what is free Ca, make measur<strong>in</strong>g SR/ER Ca<br />

nontrivial. For a commentary on these issues around SR<br />

[Ca] determ<strong>in</strong>ation, see Bygrave and Benedetti (96), and<br />

for a general review of [Ca] measurement, see Takahashi<br />

et al. (684).<br />

Lum<strong>in</strong>al [Ca] measurements to date <strong>in</strong> smooth muscles<br />

have only been reported for a limited number of<br />

tissues. Most of the studies have used the approach of<br />

fluorescent <strong>in</strong>dicators such as Mag-fura 2 (K d 49 M;<br />

Ref. 667), which was orig<strong>in</strong>ally developed as a probe for<br />

magnesium, and fluo 5-N, Furaptra (e.g., Ref. 667). Some<br />

<strong>in</strong>dicators orig<strong>in</strong>ally considered for cytoplasmic studies


were found to preferentially enter organelles, and do so <strong>in</strong><br />

a reasonably selective manner <strong>in</strong> some cell types. For<br />

example, rhod 2 will accumulate <strong>in</strong> mitochondria. In the<br />

case of the SR <strong>in</strong>dicators orig<strong>in</strong>ally developed to monitor<br />

Mg, they load relatively easily <strong>in</strong>to the SR and can be used<br />

to report lum<strong>in</strong>al [Ca]. These <strong>in</strong>dicators <strong>in</strong>clude Mag-fura<br />

5, Mag-fura 2, Mag-<strong>in</strong>do 1, and Furaptra, and it may be<br />

because of the particular efficiency of hydrolysis with<strong>in</strong><br />

the SR that they are accumulated with<strong>in</strong> it (667). Indicators<br />

with less Mg dependence <strong>in</strong>clude fluo 3FF and fura<br />

2FF, both of which have been used <strong>in</strong> smooth muscles<br />

(214, 792). Little is known about SR [Mg] <strong>in</strong> smooth<br />

muscle or <strong>in</strong>deed many other cell types; Sugiyama and<br />

Goldman (667) estimated it to be 1 mM. These authors<br />

also concluded that Furaptra (another probe sensitive to<br />

Mg, with a K d of 6 mM and for Ca of 50 M), could be<br />

used to determ<strong>in</strong>e SR [Ca], without Mg impact<strong>in</strong>g on its<br />

usefulness. In the aortic cell l<strong>in</strong>e A7r5, they estimated<br />

lum<strong>in</strong>al [Ca] to be 75–130 M.<br />

Another approach used to <strong>in</strong>vestigate lum<strong>in</strong>al Ca levels<br />

is to modulate its buffer capacity by us<strong>in</strong>g compounds<br />

that can quickly diffuse <strong>in</strong>to the SR and b<strong>in</strong>d Ca. As with<br />

Ca <strong>in</strong>dicators for the SR, the K d for such compounds has<br />

to be <strong>in</strong> the high micromolar range, i.e., low aff<strong>in</strong>ity. The<br />

heavy metal chelator TPEN with a K d around 200–500 M<br />

is such a compound (108, 571). However, TPEN may also<br />

<strong>in</strong>crease the P o of RyRs (327).<br />

C. Lum<strong>in</strong>al Ca Buffers<br />

Calcium b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s are present <strong>in</strong> the cytosol<br />

and <strong>in</strong>tracellular organelles (703). Their role <strong>in</strong> ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g<br />

a low cytosolic [Ca] and compartments capable of<br />

stor<strong>in</strong>g Ca at relatively high concentration, respectively, is<br />

crucial to Ca act<strong>in</strong>g as a ubiquitous second messenger.<br />

Soluble (nonmembranous) Ca-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s buffer Ca<br />

but are limited by their f<strong>in</strong>ite amount. Calcium pumps<br />

such as SERCA may also be viewed as membrane-bound<br />

Ca-b<strong>in</strong>d<strong>in</strong>gs prote<strong>in</strong>s, as they act to b<strong>in</strong>d Ca, transport it<br />

across their constituent membrane, release it, and are<br />

then ready to complex Ca aga<strong>in</strong>. As such, SERCA makes<br />

a large contribution as a Ca-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> and a very<br />

important and efficient regulator of [Ca] <strong>in</strong>side cells (105).<br />

It has been estimated that cytoplasmic Ca buffer<strong>in</strong>g takes<br />

up 84 of every 85 Ca ions enter<strong>in</strong>g gastric smooth muscle<br />

cells (230) and 30–46:1 <strong>in</strong> bladder myocytes (145, 204).<br />

Although cells conta<strong>in</strong> millimolar [Ca], only 0.01%<br />

is free ionized Ca. High-aff<strong>in</strong>ity Ca b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s, such<br />

as calmodul<strong>in</strong>, are present <strong>in</strong> large amounts <strong>in</strong> the cytoplasm<br />

of all mammalian cells and contribute to Ca sensitiz<strong>in</strong>g<br />

and exchange. Calmodul<strong>in</strong> is the best studied of the<br />

endogenous Ca sensors (prote<strong>in</strong>s with high aff<strong>in</strong>ity and<br />

low capacity) <strong>in</strong> smooth muscle (653). It b<strong>in</strong>ds Ca with a<br />

K d of 2 M (319) and is present at 40 M (336). In<br />

SMOOTH MUSCLE SR 137<br />

early studies of [Ca] with<strong>in</strong> smooth muscle cells (rabbit<br />

portal ve<strong>in</strong>) us<strong>in</strong>g electron-probe microanalysis, it was<br />

concluded that there was not sufficient calmodul<strong>in</strong> to<br />

account for the [Ca] measurements made (66). In a recent<br />

study of cytosolic Ca buffer<strong>in</strong>g <strong>in</strong> s<strong>in</strong>gle smooth muscle<br />

cells from gu<strong>in</strong>ea pig bladder, it was concluded that the<br />

Ca buffers present have a low aff<strong>in</strong>ity (EF-hand) for free<br />

Ca, are present at 530 M, and have a calculated b<strong>in</strong>d<strong>in</strong>g<br />

ratio of 40 (145). In this study the volume of the SR was<br />

taken to be 2%, which may be an underestimate. Other<br />

studies <strong>in</strong> smooth muscles have yielded similar values for<br />

the ratio of Ca b<strong>in</strong>d<strong>in</strong>g: airways (178), bladder (204),<br />

larger portal ve<strong>in</strong> (329), stomach (349).<br />

With<strong>in</strong> the SR lumen, specific prote<strong>in</strong>s are expressed<br />

to b<strong>in</strong>d Ca. These SR lum<strong>in</strong>al Ca storage prote<strong>in</strong>s can be<br />

considered to act as buffers, as they keep Ca relatively<br />

loosely bound and thus available for quick release, and<br />

help prevent <strong>in</strong>soluble calcium phosphate precipitat<strong>in</strong>g.<br />

By reduc<strong>in</strong>g the amount of free or ionized Ca <strong>in</strong> the SR,<br />

they also reduce <strong>in</strong>hibition by Ca of SERCA activity. Thus<br />

the expression of these Ca b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s is critical to<br />

the SR’s ability to act as a Ca store, <strong>in</strong> all cells.<br />

Although many prote<strong>in</strong>s capable of b<strong>in</strong>d<strong>in</strong>g Ca may<br />

be expressed <strong>in</strong> the SR, the consideration here is of those<br />

which b<strong>in</strong>d considerable quantities of Ca and which appear<br />

to have this as their prime role with<strong>in</strong> the SR lumen.<br />

There are just two prote<strong>in</strong> families that fulfil this description:<br />

calreticul<strong>in</strong> and calsequestr<strong>in</strong>. As described already<br />

<strong>in</strong> section IV, E and F, they are both high-capacity (25–50<br />

mol/mol), low-aff<strong>in</strong>ity (1–4 mM) Ca b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s that<br />

function only or primarily to dynamically store Ca (384,<br />

597). These two prote<strong>in</strong>s have similar Ca-b<strong>in</strong>d<strong>in</strong>g properties,<br />

molecular weights, and other prote<strong>in</strong> properties,<br />

such as acidic isoelectric po<strong>in</strong>ts (573). As noted above,<br />

specific data for either prote<strong>in</strong> <strong>in</strong> smooth muscle are not<br />

abundant, or there have been controversies <strong>in</strong> the literature<br />

(472, 730) and little recent work.<br />

D. Measurements<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

Not withstand<strong>in</strong>g caveats around <strong>in</strong>dicator specificities,<br />

K d values, Mg sensitivity, and calibration methods, a<br />

few <strong>in</strong>vestigators have estimated/measured the SR lum<strong>in</strong>al<br />

[Ca] (214, 636, 667, 755, 811). Not unexpectedly, there<br />

is a range of values, from 60 to 150 M, but this range is<br />

perhaps surpris<strong>in</strong>gly small given the difficulties <strong>in</strong>volved.<br />

It would therefore appear that the SR free [Ca] is less<br />

<strong>in</strong> smooth muscles compared with other cell types, where<br />

values of 200–1,500 M have been reported (33, 118, 121,<br />

268). However, until more measurements <strong>in</strong> native<br />

smooth muscle cells are made, it is unproductive to draw<br />

further conclusions.


138 SUSAN WRAY AND THEODOR BURDYGA<br />

E. Effects of SR Ca Load<br />

1. Introduction<br />

How lum<strong>in</strong>al SR Ca content relates to a change <strong>in</strong><br />

cytosolic [Ca] is not easy to predict or determ<strong>in</strong>e. This is<br />

because there is feedback from cytosolic [Ca] onto the SR<br />

release channels (282) and a difference <strong>in</strong> buffer<strong>in</strong>g capacity<br />

between the SR and the cytoplasm (230). In cardiac<br />

muscles where a proper quantitative study can be made,<br />

the nonl<strong>in</strong>earity between the two can be steep (704). This<br />

has not been as well studied <strong>in</strong> smooth muscle, but the<br />

follow<strong>in</strong>g study also po<strong>in</strong>ts to a steep relation.<br />

In rat uter<strong>in</strong>e cells we have shown that <strong>in</strong>creas<strong>in</strong>g<br />

external [Ca] from 2 to 10 mM <strong>in</strong>creases cytosolic [Ca],<br />

from 120 to 270 nM, and is associated with a substantial<br />

rise (17% <strong>in</strong>crease compared with steady-state value) <strong>in</strong><br />

SR [Ca] (636). This elevation of SR [Ca], however, did not<br />

lead to any potentiation of the Ca transient elicited <strong>in</strong><br />

response to the IP3-generat<strong>in</strong>g agonist ATP. If the SR load<br />

was decreased (but not emptied) from its steady-state<br />

level, the ATP-<strong>in</strong>duced Ca transients fell. If the SR load<br />

was decreased below 80% of normal, no Ca transients to<br />

ATP could be elicited. Thus there is a steep dependence<br />

of IP3-<strong>in</strong>duced SR Ca release with SR load as it depletes,<br />

but not as it overloads, at least <strong>in</strong> uter<strong>in</strong>e myocytes.<br />

2. Relation between SR Ca load and Ca signals<br />

The effects of alter<strong>in</strong>g SERCA activity on global Ca<br />

signals and function <strong>in</strong> smooth muscle cells are discussed<br />

<strong>in</strong> section VII, C and D. Here we will focus on how SERCA<br />

activity affects SR lum<strong>in</strong>al [Ca] and then Ca release<br />

events. Block<strong>in</strong>g SERCA, e.g., with thapsigarg<strong>in</strong> or CPA,<br />

has been shown by us and others to produce a decrease <strong>in</strong><br />

SR Ca content, which is accelerated with agonist stimulation<br />

(16, 633, 661). In our study of uter<strong>in</strong>e cells, we<br />

simultaneously measured cytosolic and lum<strong>in</strong>al [Ca]<br />

changes and compared the depletion of SR Ca <strong>in</strong> response<br />

to agonist stimulation, with and without SERCA activity<br />

(633). We found that the depletion was greater when<br />

SERCA was <strong>in</strong>hibited and the <strong>in</strong>tracellular Ca transients<br />

were smaller. In a follow up study we found that the rise<br />

<strong>in</strong> cytosolic [Ca] seen with <strong>in</strong>creas<strong>in</strong>g external [Ca] was<br />

significantly <strong>in</strong>creased, from 270 to 320 nM, when<br />

SERCA was <strong>in</strong>hibited with CPA (636). Removal of external<br />

Ca produced a fall <strong>in</strong> cytosolic [Ca] and SR [Ca]. The<br />

effects of repetitive agonist applications under conditions<br />

of low and high SR [Ca] and with and without SERCA are<br />

also described by Shmygol and Wray (636).<br />

From these and other studies we can confidently say<br />

that agonists lower SR [Ca] and SERCA activity is required<br />

to restore, and presumably ma<strong>in</strong>ta<strong>in</strong>, the ability of<br />

smooth muscle cells to respond to agonists. As a rundown<br />

of lum<strong>in</strong>al [Ca] would be anticipated to rapidly curtail<br />

agonist-<strong>in</strong>duced Ca signals, it can be assumed that under<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

normal physiological conditions SERCA is active dur<strong>in</strong>g<br />

stimulation to refill the store, although as noted by Gomez-<br />

Viquez et al. (218), the turnover rate of SERCA is slower<br />

than that of the release channels, be<strong>in</strong>g 3.5 Ca/s for<br />

SERCA2b compared with 1 10 6 Ca/s through RyR<br />

(462). Given the available data on SERCA and RyR expression,<br />

the number of SERCA pumps is not sufficient to<br />

compensate for the RyR Ca efflux.<br />

In their <strong>in</strong>terest<strong>in</strong>g study, Gomez-Viquez et al. (218)<br />

explored the relation between SERCA pump activity and<br />

SR Ca release channels <strong>in</strong> ur<strong>in</strong>ary bladder myocytes. They<br />

<strong>in</strong>vestigated whether SERCA activity per se rather than<br />

lum<strong>in</strong>al [Ca] could <strong>in</strong>fluence Ca release events, or if uptake<br />

and release operate <strong>in</strong>dependently of each other,<br />

assum<strong>in</strong>g there is sufficient [Ca] <strong>in</strong> the SR lumen. They<br />

rapidly and completely <strong>in</strong>hibited SERCA (thapsigarg<strong>in</strong>) so<br />

that lum<strong>in</strong>al [Ca] would be ma<strong>in</strong>ta<strong>in</strong>ed (as verified from<br />

the Mag-fura 2 signals). With SERCA <strong>in</strong>hibited, the AChor<br />

caffe<strong>in</strong>e-<strong>in</strong>duced Ca release <strong>in</strong>to the cytoplasm, as<br />

measured with fura 2, was reduced <strong>in</strong> both amplitude and<br />

rate of rise. Thus SERCA2b <strong>in</strong> bladder smooth muscle<br />

cells can modulate both IP 3R and RyR Ca release events;<br />

SERCA activity is required for optimal Ca release. The<br />

authors speculate that SERCA2b pumps are modulat<strong>in</strong>g<br />

Ca availability <strong>in</strong> the SR. As described earlier, the<br />

SERCA2b isoform conta<strong>in</strong>s a carboxy tail extension that<br />

<strong>in</strong>teracts with lum<strong>in</strong>al Ca b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s, suggest<strong>in</strong>g a<br />

molecular and structural mechanism for this effect.<br />

SERCA activity can <strong>in</strong>directly modulate Ca release<br />

events through sett<strong>in</strong>g the [Ca] load <strong>in</strong> the SR. Both IP 3Rs<br />

and RyRs have been shown, <strong>in</strong> lipid bilayer experiments,<br />

to have their activity affected by lum<strong>in</strong>al SR [Ca] (477,<br />

642; see sect. V). In turn, it is SERCA activity that loads the<br />

SR with Ca (661). ZhuGe et al. (811) work<strong>in</strong>g on gastric<br />

myocytes noted the effects of lum<strong>in</strong>al load by detect<strong>in</strong>g<br />

Ca sparks and STOCs at different Ca loads. They found<br />

the frequency of both <strong>in</strong>creased with load. A similar relation<br />

between the SR Ca load (altered by knock<strong>in</strong>g out<br />

phospholamban) and Ca sparks had previously been reported<br />

<strong>in</strong> cardiac muscle (614). The effects of SR Ca load<br />

on STOCS was also confirmed by others (123, 452). It was<br />

found that a relatively small fall <strong>in</strong> SR [Ca] (16%) had a<br />

profound effect on STOC frequency (decreased by 70%)<br />

(452). As mentioned earlier, <strong>in</strong> uter<strong>in</strong>e myocytes, overload<strong>in</strong>g<br />

the SR with Ca did not <strong>in</strong>crease the simultaneously<br />

measured cytosolic Ca transient <strong>in</strong> response to<br />

ATP stimulation (636). In marked contrast, partial depletion<br />

of the SR Ca substantially reduced the amplitude of<br />

the ATP-<strong>in</strong>duced Ca release: a reduction to 80% of normal<br />

lum<strong>in</strong>al SR Ca content abolished responses. Previous<br />

work had shown that uptake of Ca <strong>in</strong>to the SR occurred<br />

even <strong>in</strong> the absence of external Ca, although this only<br />

replenished lum<strong>in</strong>al Ca to 50% of its rest<strong>in</strong>g value (633).<br />

Thapsigarg<strong>in</strong> abolished both subsequent ATP-evoked Ca<br />

release and SR refill<strong>in</strong>g. Interest<strong>in</strong>gly, this study found no


changes <strong>in</strong> lum<strong>in</strong>al [Ca] dur<strong>in</strong>g spontaneous activity, suggest<strong>in</strong>g<br />

no role for the SR <strong>in</strong> this process, but rather<br />

support<strong>in</strong>g the view that L-type Ca entry is both necessary<br />

and sufficient for normal phasic activity <strong>in</strong> the uterus, and<br />

store-operated Ca <strong>in</strong>flux is not necessary. When the SR<br />

was overloaded, spontaneous activity was abolished, consistent<br />

with a negative feedback from the SR on E-C<br />

coupl<strong>in</strong>g <strong>in</strong> the uterus, despite the lack of Ca sparks (88).<br />

F. Conclusions<br />

While it is well accepted that lum<strong>in</strong>al [Ca] is a very<br />

important control regulator of both SR function and Ca<br />

signal<strong>in</strong>g, the field would be well served by some additional<br />

studies <strong>in</strong> this area. For example, additional calibrated<br />

measurements of lum<strong>in</strong>al [Ca] would clarify if the<br />

value <strong>in</strong> smooth muscles is 100 M, i.e., considerably<br />

lower than striated muscles. Will values be lower <strong>in</strong> tissues<br />

like uterus where the SR may not contribute to<br />

rhythmic activity, compared for example to blood vessels,<br />

which rely more heavily on the SR Ca store? Similarly,<br />

more simultaneous measurements of cytosolic and lum<strong>in</strong>al<br />

[Ca] would add to our knowledge of how SR Ca load<br />

affects Ca signal<strong>in</strong>g. Does the SR release Ca dur<strong>in</strong>g phasic<br />

activity? Is SR Ca overload <strong>in</strong>hibitory to contractility <strong>in</strong><br />

smooth muscles? How do changes <strong>in</strong> the expression of SR<br />

Ca buffers affect SR Ca load? Although these experiments<br />

are technically challeng<strong>in</strong>g, especially when comb<strong>in</strong>ed<br />

with electrophysiological studies, the <strong>in</strong>sight ga<strong>in</strong>ed will<br />

be important and significant to the field.<br />

VII. CALCIUM REUPTAKE MECHANISMS<br />

A. Introduction<br />

In section III, the structure of SERCA and its isoforms<br />

and catalytic cycle were described. The ability of SERCA<br />

to take up Ca is of course crucial to the ability of the SR<br />

to act as a Ca store. In this section we review the effect of<br />

Ca uptake by SERCA on cytosolic [Ca] levels and Ca<br />

signals, its role <strong>in</strong> plasma membrane Ca extrusion, and the<br />

effects of <strong>in</strong>hibit<strong>in</strong>g its activity either pharmacologically<br />

or molecularly. (Discussions of how phospholamban and<br />

related prote<strong>in</strong>s affect SERCA were dealt with <strong>in</strong> section<br />

IV.) This is then followed by a review of store-operated Ca<br />

entry <strong>in</strong> smooth muscle, explor<strong>in</strong>g the relation between<br />

lum<strong>in</strong>al Ca levels and Ca entry mechanisms that replenish<br />

the SR with Ca.<br />

B. SERCA Uptake of Ca and Ca Homeostasis<br />

For the cell to ma<strong>in</strong>ta<strong>in</strong> low cytosolic [Ca] with respect<br />

to both the extracellular fluid and the SR, requires<br />

SMOOTH MUSCLE SR 139<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

not just SERCA activity, but also that of the PMCA and<br />

NCX. For the cell to be <strong>in</strong> a steady state, Ca enter<strong>in</strong>g the<br />

cell across the plasma membrane must be removed by<br />

PMCA and NCX, and Ca released from the SR must be<br />

retaken up by SERCA. This will also be the case for Ca<br />

uptake <strong>in</strong>to organelles such as the nucleus and mitochondria.<br />

There will be basal or “rest<strong>in</strong>g” SERCA activity,<br />

which presumably balances SR Ca leaks, as well as spontaneous<br />

Ca releases from the SR, Ca puffs, and sparks.<br />

SERCA activity is stimulated when cytosolic [Ca], from<br />

whatever source, rises and the presence of the lum<strong>in</strong>al Ca<br />

buffers, calsequestr<strong>in</strong> and calreticul<strong>in</strong>, ensures that <strong>in</strong>hibition<br />

of SERCA by lum<strong>in</strong>al Ca does not occur. The SR<br />

will contribute to plasma membrane Ca efflux by vectorially<br />

releas<strong>in</strong>g Ca towards efflux sites. As with many<br />

other aspects of smooth muscle physiology, the exact<br />

contribution of SERCA, PMCA, and NCX will differ between<br />

them, depend<strong>in</strong>g on expression levels and distribution.<br />

Perhaps the easiest way to uncover the role of<br />

SERCA <strong>in</strong> ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g cytosolic Ca levels is to determ<strong>in</strong>e<br />

the effects of its <strong>in</strong>hibition. As mentioned already, there<br />

are two specific <strong>in</strong>hibitors of SERCA: thapsigarg<strong>in</strong>, which<br />

is irreversible, and CPA, which is reversible. When<br />

SERCA is <strong>in</strong>hibited <strong>in</strong> smooth muscle cells there is a rise<br />

<strong>in</strong> cytoplasmic [Ca]. This rise of [Ca] can be fast or slow<br />

and then plateaus, lead<strong>in</strong>g to an elevated rest<strong>in</strong>g [Ca]. Due<br />

to the difficulties of calibrat<strong>in</strong>g Ca signals, many studies<br />

have simply reported a rise of [Ca] with SERCA <strong>in</strong>hibition,<br />

perhaps expressed relative to high-K depolarization or<br />

some other calibrator. Some of the first studies to explore<br />

the effect of CPA were on vascular myocytes (1, 120) and<br />

used aequor<strong>in</strong> to monitor [Ca]. With CPA there was a<br />

slow, i.e., m<strong>in</strong>utes, <strong>in</strong>crease <strong>in</strong> the Ca signal and basal<br />

tension. The response to norep<strong>in</strong>ephr<strong>in</strong>e was decreased.<br />

Rises <strong>in</strong> [Ca] <strong>in</strong> response to SERCA <strong>in</strong>hibition have also<br />

been shown <strong>in</strong> bladder myocytes (from 137 to 471 nM;<br />

Ref. 789) and uter<strong>in</strong>e myocytes (from 120 to 320 nM; Ref.<br />

636). A study of airway myocytes, however, found no<br />

significant change <strong>in</strong> rest<strong>in</strong>g [Ca] (130 nM) after 25 m<strong>in</strong><br />

<strong>in</strong> 0-Ca and CPA solution (603), perhaps due to <strong>in</strong>creased<br />

Ca efflux under these conditions.<br />

C. SERCA and Ca Signal<strong>in</strong>g and Contractility<br />

Both pharmaceutical SERCA <strong>in</strong>hibition and to a more<br />

limited extent gene ablation have been used to <strong>in</strong>vestigate<br />

the role of SERCA <strong>in</strong> Ca signal<strong>in</strong>g <strong>in</strong> smooth muscles.<br />

When SERCA2 was reduced to 50% and aortic contractility<br />

measured, no changes from wild-type mice were<br />

found <strong>in</strong> response to KCl and phenylephr<strong>in</strong>e (294). This<br />

contrasts with the significant effects found on cardiac<br />

contractility, <strong>in</strong>clud<strong>in</strong>g decreases <strong>in</strong> Ca transients and cell<br />

shorten<strong>in</strong>g (558). It is not clear whether the lack of effect


140 SUSAN WRAY AND THEODOR BURDYGA<br />

<strong>in</strong> the vascular preparation is due to <strong>in</strong>creased SERCA<br />

reserve compared with the heart or some form of compensatory<br />

regulation of other parameters. When CPA is<br />

used to <strong>in</strong>hibit SERCA <strong>in</strong> blood vessels, clear effects are<br />

seen (504). When SERCA3-deficient mice were exam<strong>in</strong>ed,<br />

the relaxation to ACh was reduced <strong>in</strong> aorta (402) and to<br />

substance P <strong>in</strong> trachea (334); however, as discussed earlier,<br />

these effects are not direct on the myocytes, but<br />

rather are due to endothelial- and epithelial-dependent<br />

effects. No overt phenotype is associated with the<br />

SERCA3 knockout (402). This appears to be the extent of<br />

the literature on gene target<strong>in</strong>g studies of SERCA function<br />

<strong>in</strong> smooth muscle. There is, however, a more extensive<br />

literature concern<strong>in</strong>g effects obta<strong>in</strong>ed with CPA and thapsigarg<strong>in</strong>.<br />

In most smooth muscles, a clear elevation of<br />

basel<strong>in</strong>e tension is found with SERCA <strong>in</strong>hibition, and<br />

when measured, this can be seen to be accompanied by<br />

<strong>in</strong>creased global Ca signals [ileal (710), aorta (417, 629),<br />

fundus (562), esophageal (640), uterus (377, 446, 682)].<br />

Before the l<strong>in</strong>k between SR Ca release and BK<br />

channels and membrane hyperpolarizations were elucidated,<br />

many authors <strong>in</strong>terpreted their data, i.e., SERCA<br />

<strong>in</strong>hibition <strong>in</strong>creases [Ca] and contraction, <strong>in</strong> terms of<br />

the SR block<strong>in</strong>g or buffer<strong>in</strong>g the flow of Ca to the<br />

myofilaments. The spark-STOC coupl<strong>in</strong>g mechanisms,<br />

however, can directly account for the effect of SERCA<br />

<strong>in</strong>hibition <strong>in</strong> many, but importantly, not all smooth<br />

muscles. The effects of SERCA <strong>in</strong>hibition on Ca signal<strong>in</strong>g<br />

and contractility <strong>in</strong> phasic smooth muscles can be<br />

very pronounced. For example, <strong>in</strong> the neonatal uterus<br />

(519) or term myometrium (707), the normal phasic<br />

activity becomes tonic-like.<br />

D. SERCA Ca Efflux and Relaxation<br />

Although SERCA takes up Ca and aids restoration of<br />

[Ca] to rest<strong>in</strong>g levels follow<strong>in</strong>g smooth muscle stimulation,<br />

there is evidence from uter<strong>in</strong>e smooth muscle to<br />

show that act<strong>in</strong>g on its own, it does not significantly alter<br />

the decay of the Ca transient, but rather, it enhances<br />

efflux mechanisms. In isolated uter<strong>in</strong>e myocytes (631)<br />

when PMCA and NCX were blocked, <strong>in</strong>tracellular [Ca] did<br />

not fall after stimulation (644). However, if SERCA was<br />

blocked, but not PMCA and NCX, then the decay of the Ca<br />

transient was slowed. This was the case whether Ca efflux<br />

was via PMCA or NCX or both. We concluded that when<br />

SERCA is active, it takes up a portion of Ca enter<strong>in</strong>g the<br />

cell, and the SR then releases Ca <strong>in</strong> a directed manner<br />

towards the plasma membrane extruders, thereby maximiz<strong>in</strong>g<br />

their efficiency (see also Refs. 184, 445). A study <strong>in</strong><br />

airway myocytes also concluded that the decay of the Ca<br />

transient was not altered by SERCA, although it is active<br />

(603). These authors suggested that mitochondria take up<br />

Ca and contribute to the decrease <strong>in</strong> [Ca]. In an earlier<br />

study <strong>in</strong> isolated tracheal myocytes, SERCA had been<br />

shown to contribute to the decay of agonist-<strong>in</strong>duced Ca<br />

transients as the decay was slower when SERCA was<br />

<strong>in</strong>hibited (640).<br />

E. Store-Operated Ca Entry<br />

1. Introduction<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

The Ca content of the SR acts as a powerful factor<br />

controll<strong>in</strong>g the open probabilities of both RyR and IP 3R<br />

channels (see sects. IV and VII). Depletion of Ca stores <strong>in</strong><br />

smooth muscle cells by SERCA pump <strong>in</strong>hibitors results <strong>in</strong><br />

an <strong>in</strong>hibition of Ca sparks and STOCs <strong>in</strong> those tissues<br />

produc<strong>in</strong>g them. This <strong>in</strong> turn can lead to depolarization of<br />

the cell membrane and activation of Ca entry via L-type<br />

Ca channels, as was reported for rat cerebral arteries<br />

(507). However, <strong>in</strong> addition to activation of L-type Ca<br />

channels, depletion of SR Ca can also activate nifedip<strong>in</strong>eresistant<br />

Ca entry, through open<strong>in</strong>g of Ca release-activated<br />

Ca channels (CRAC). In nonexcitable cells, ER Ca<br />

depletion opens voltage-<strong>in</strong>dependent plasma membrane Ca<br />

channels through which extracellular Ca enters the cell to<br />

refill the store. These are termed “capacitative calcium entry”<br />

(CCE) channels (542, 579, 580). The role of CCE, also<br />

known as store-operated Ca entry (SOCE) and Ca entry<br />

mechanism across the sarcolemma of the smooth muscles,<br />

is still be<strong>in</strong>g determ<strong>in</strong>ed.<br />

Recent work has identified the specific molecular<br />

components, Stim and Orai, which constitute the Ca sensor<br />

<strong>in</strong> the ER/SR and the Ca channel, respectively, and<br />

their discovery has helped to clarify the mechanisms of<br />

CCE. The process and mechanisms of SOCE have been<br />

best studied <strong>in</strong> nonexcitable cells, where the process was<br />

first discovered, and studies <strong>in</strong> smooth muscle are relatively<br />

sparse. Therefore, an overview of current knowledge<br />

taken from the literature on nonexcitable cells will<br />

be given to set the scene for smooth muscle.<br />

2. Overview of store depletion, Orai, STIM1,<br />

and TRPCs<br />

This account is based on <strong>in</strong>formation <strong>in</strong> several recent<br />

reviews that have appeared s<strong>in</strong>ce the discovery of<br />

Stim (stromal <strong>in</strong>teraction molecule 1) prote<strong>in</strong>s (99, 173,<br />

189, 260, 524, 569, 581, 649).<br />

Agonist b<strong>in</strong>d<strong>in</strong>g to cell membranes and subsequent<br />

generation of IP 3 leads not only to ER Ca release, but also<br />

to activation of Ca channels <strong>in</strong> the plasma membrane.<br />

This connected depletion and load<strong>in</strong>g of the ER Ca store<br />

was termed capacitative Ca entry by Putney <strong>in</strong> 1986 (578)<br />

and is synonymous with the term store-operated Ca entry.<br />

This form of Ca entry is the major route of Ca entry <strong>in</strong><br />

nonexcitable cells, which lack functional voltage-gated Ca<br />

channels. It was subsequently demonstrated that the Ca


entry could be stimulated without agonist or IP 3, if the ER<br />

Ca was depleted, e.g., us<strong>in</strong>g SERCA <strong>in</strong>hibitors, particularly<br />

thapsigarg<strong>in</strong> (545, 686). Detect<strong>in</strong>g the rise of cytoplasmic<br />

[Ca] and the associated small Ca release-activated<br />

Ca current (I crac) (272) result<strong>in</strong>g from this store<br />

depletion-activated Ca entry was difficult. This led to the<br />

suggestion from experiments on arterial myocytes that<br />

the refill<strong>in</strong>g of the SR occurred by-pass<strong>in</strong>g the cytoplasm,<br />

i.e., direct coupl<strong>in</strong>g between the plasma membrane and<br />

SR, or that the Ca was <strong>in</strong> a restricted/protected space<br />

between the two, and not accessible by the myofilaments,<br />

as no contraction occurred (111). However, given the<br />

existence of I crac, <strong>in</strong> both excitable and nonexcitable<br />

cells, Ca clearly is enter<strong>in</strong>g the cytoplasm, and the failure<br />

to detect a rise <strong>in</strong> [Ca] <strong>in</strong> most cell types is now attributed<br />

to the close proximity between ER/SR and the plasma<br />

membrane and the rapid buffer<strong>in</strong>g of the Ca entry by the<br />

ER/SR.<br />

Much experimental effort was put <strong>in</strong>to elucidat<strong>in</strong>g<br />

the molecular events between the ER and plasma membrane,<br />

which could activate the capacitative store-operated<br />

Ca entry. Until a few years ago, the discussion would<br />

be focused around canonical TRPCs, which were discovered<br />

<strong>in</strong> Drosophila and shown to be activated after an<br />

<strong>in</strong>crease <strong>in</strong> PLC activity (245). These channels, expressed<br />

<strong>in</strong> a large variety of isoforms <strong>in</strong> mammalian cells, almost<br />

became accepted as “the” store-operated channels responsible<br />

for capacitative Ca entry. However, as discussed<br />

by Putney (581), these reports were often from<br />

overexpress<strong>in</strong>g or constitutively active preparations and<br />

the TRPC channels did not reproduce the known properties<br />

of I crac and were usually cationic rather than Caselective<br />

channels.<br />

While it is too early to be categorical, the evidence<br />

that TRPC channels are responsible for capacitative Ca<br />

entry is not strong. It is also the case that s<strong>in</strong>ce 2006 new<br />

putative channels <strong>in</strong> the form of Orai prote<strong>in</strong>s have been<br />

discovered, which appear to be much better candidates.<br />

Orai is a plasma membrane prote<strong>in</strong> which when mutated<br />

results <strong>in</strong> abrogation of I crac and severe comb<strong>in</strong>ed<br />

immunodeficiency syndrome (174). It appears to be unlike<br />

other previously described Ca channels. Mammalian<br />

cells can express three types of Orai, 1–3, and all are<br />

capable of form<strong>in</strong>g Ca channels, probably as homotetramers.<br />

A recent paper suggests that Orai is primarily a dimer<br />

<strong>in</strong> the plasma membrane under rest<strong>in</strong>g conditions, and<br />

upon activation by the COOH term<strong>in</strong>us of Stim, the<br />

dimers dimerize, form<strong>in</strong>g tetramers that constitute the<br />

Ca-selective channel (553). These and other data make it<br />

hard to propose that TRPC channels may contribute to<br />

capacitative Ca entry via <strong>in</strong>clusion <strong>in</strong> an Orai/TRPC Ca<br />

channel (153, 398). Orai1 forms the most conductive Ca<br />

channels, but Orai2 and -3 are also able to perform this<br />

function (464). Orai prote<strong>in</strong>s have four transmembrane<br />

doma<strong>in</strong>s, and the first of these may be most important for<br />

SMOOTH MUSCLE SR 141<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

channel function (575, 788). When Orai1 is expressed with<br />

another prote<strong>in</strong>, Stim1, large I crac currents are observed<br />

(552, 804).<br />

Stim1 was discovered as a cell surface prote<strong>in</strong> (530)<br />

and was related to the ER <strong>in</strong> 2005 (400, 595) as its Ca<br />

sensor, responsible for activat<strong>in</strong>g capacitative Ca entry.<br />

Thus Stim1 knockout reduces I crac and SOCE. Stim1 is a<br />

phosphoprote<strong>in</strong> with a s<strong>in</strong>gle transmembrane doma<strong>in</strong>.<br />

The NH 2 term<strong>in</strong>us of Stim has an EF hand doma<strong>in</strong> <strong>in</strong> the<br />

ER lumen that is considered to be the sensor of lum<strong>in</strong>al<br />

[Ca]. It has been calculated that the EC 50 for Stim1 redistribution<br />

is between 200 and 400 M lum<strong>in</strong>al Ca (82, 651,<br />

660) and that oligomerization of about four Stim1 prote<strong>in</strong>s<br />

occurs with the conformational change <strong>in</strong> the NH 2 term<strong>in</strong>us<br />

as Ca dissociates. This oligomerization is necessary<br />

for activation of I crac (575, 788), and mutation of this<br />

region produces constitutive activation of Ca entry (464).<br />

Both Stim and Orai are required to produce I crac. Although<br />

not completely clear, the mechanism of activation appears<br />

to <strong>in</strong>volve a redistribution of Stim1 <strong>in</strong> the ER when<br />

Ca dissociates from it, i.e., as the ER depletes. This redistribution<br />

br<strong>in</strong>gs Stim closer to the plasma membrane<br />

(with<strong>in</strong> 10–25 m) <strong>in</strong> a punctate pattern on the ER, with<br />

some workers (541) but not all (148) suggest<strong>in</strong>g that it<br />

takes up a lipid raft distribution (541). This redistribution<br />

that precedes I crac by 6–10 s (775) presumably enables<br />

Stim1 to directly <strong>in</strong>teract with Orai channels, and Ca<br />

enters <strong>in</strong> these discrete regions of the plasma membrane<br />

(416). Evidence for this comes from a variety of biophysical<br />

and imag<strong>in</strong>g techniques as well as coimmunoprecipitation.<br />

It may be, however, that there is no direct coupl<strong>in</strong>g<br />

between Orai and Stim and another factor, such as “Ca<strong>in</strong>flux<br />

factor” (CIF), is stimulated by Stim redistribution<br />

and CIF (or another messenger) is needed for Orai activation,<br />

perhaps <strong>in</strong>volv<strong>in</strong>g activation of phospholipase A 2<br />

by CIF displac<strong>in</strong>g calmodul<strong>in</strong> (63). Another prote<strong>in</strong> identified<br />

<strong>in</strong> the Stim family, Stim2, is structurally 60% identical<br />

to Stim1, but does not appear to play any major role<br />

<strong>in</strong> capacitative Ca entry (595), but may contribute to<br />

regulat<strong>in</strong>g basal [Ca] via ER Ca sens<strong>in</strong>g (82).<br />

Most of the studies mentioned above were performed<br />

on immune or exocr<strong>in</strong>e cells. How much evidence is there<br />

for these mechanisms and prote<strong>in</strong>s <strong>in</strong> smooth muscle?<br />

Although only a few years from their discovery, the excitement<br />

around Stim and Orai has already led to studies<br />

<strong>in</strong>vestigat<strong>in</strong>g their expression and role <strong>in</strong> smooth muscle<br />

tissues.<br />

3. Orai, Stim, and TRPCs <strong>in</strong> smooth muscle<br />

The study of capacitative/store-operated Ca entry <strong>in</strong><br />

smooth muscles is more complicated than it is <strong>in</strong> nonexcitable<br />

cells, due to the greater variety of Ca entry mechanisms,<br />

i.e., voltage-gated and receptor-operated channels,<br />

<strong>in</strong> addition to channels that are opened <strong>in</strong> response


142 SUSAN WRAY AND THEODOR BURDYGA<br />

to a depletion of SR Ca. Previous work had <strong>in</strong>vestigated<br />

TRPC channels as the putative Ca entry mechanism<br />

opened by store Ca depletion, but the grow<strong>in</strong>g consensus<br />

is that they are not responsible for I crac. Therefore, it may<br />

be argued that TRPCs are unlikely to be responsible for<br />

SOCE <strong>in</strong> smooth muscles. This has already been suggested<br />

by some (153, 722, 738) but contested by others<br />

(e.g., Ref. 7). TRPC1, the TRP most thought to be <strong>in</strong>volved<br />

<strong>in</strong> SOCE and highly expressed <strong>in</strong> smooth muscles, had no<br />

effect on vascular function or SOCE when knocked out<br />

(12). In TRPC6 knockout mice, enhanced DAG and agonist-<strong>in</strong>duced<br />

current <strong>in</strong> smooth muscle cells was reported,<br />

i.e., the opposite result expected if TRPC6 is <strong>in</strong>volved <strong>in</strong><br />

SOCE (188). RNA <strong>in</strong>terference (RNAi)-based high-throughput<br />

screens also revealed no effect on SOCE activation of<br />

TRP channel knockdown, <strong>in</strong> contrast to Stim (400). As<br />

with studies on nonexcitable cells, caution must be applied<br />

to studies where genes have been knocked out or<br />

overexpressed, and <strong>in</strong> smooth muscle, when cells have<br />

been cultured, the physiological relevance of such studies<br />

is even harder to <strong>in</strong>terpret (274). Nevertheless, while it is<br />

premature to rule TRPCs out of <strong>in</strong>volvement <strong>in</strong> SOCE <strong>in</strong><br />

smooth muscle cells, especially as much of the Orai work<br />

has been conducted on nonexcitable cells, it nevertheless<br />

does not appear profitable to cover <strong>in</strong> great depth previous<br />

studies related to a role <strong>in</strong> SOCE of TRPC, especially<br />

as this has recently been reviewed (7). A recent report by<br />

Dehaven et al. (148) demonstrated that TRPC functions<br />

<strong>in</strong>dependently of Orai1 <strong>in</strong> vascular myocytes (A7r5 and<br />

A10 cell l<strong>in</strong>es) and suggests that TRPCs are activated by<br />

PLC and do not <strong>in</strong>volve Stim1 as the Ca sensor and TRCPs<br />

open nonselective cation channels when stimulated by<br />

arg<strong>in</strong><strong>in</strong>e vasopress<strong>in</strong>. Roles for TRCPs other than <strong>in</strong> storeoperated<br />

Ca entry <strong>in</strong> smooth muscle have been well demonstrated<br />

for receptor-operated channels, where they<br />

constitute nonspecific cation channels (39, 457) and<br />

stretch channels (658), but these are outside the scope of<br />

this review.<br />

There is already evidence to support the Stim-Orai<br />

model for SOCE <strong>in</strong> some smooth muscles: airway, Stim-1<br />

(549), Orai (550); vascular, Stim (153, 413, 685), Stim and<br />

Orai (47, 210); and uterus, Stim and Orai (611). The list of<br />

smooth muscles express<strong>in</strong>g Stim and Orai is expected to<br />

grow rapidly as more studies are performed. To date<br />

however, there is already evidence that capacitative Ca<br />

entry/SOCE <strong>in</strong> some smooth muscles at least occurs via<br />

the Stim-Orai mechanism. Dietrich et al. (153) <strong>in</strong>hibited<br />

SOCE us<strong>in</strong>g siRNA aga<strong>in</strong>st Stim1, whereas TRCP1 knockout<br />

did not affect it. Further support for a physiological<br />

function for these prote<strong>in</strong>s <strong>in</strong> smooth muscles comes<br />

from data show<strong>in</strong>g that their expression levels are altered<br />

at different sites and <strong>in</strong> disease. Thus Orai and Stim were<br />

upregulated <strong>in</strong> proliferat<strong>in</strong>g arterial myocytes, lead<strong>in</strong>g the<br />

authors to suggest that they play a critical role <strong>in</strong> the<br />

altered Ca handl<strong>in</strong>g that occurs dur<strong>in</strong>g vascular growth<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

and remodel<strong>in</strong>g (47). Stim1 expression was greater <strong>in</strong> the<br />

distal than proximal part of the pulmonary artery, where<br />

SOCE is also greatest (413). Stim1 and Orai1 were also<br />

found to be upregulated <strong>in</strong> aortas from hypertensive rats<br />

(210). Knockdown of Orai1 reduced thapsigarg<strong>in</strong>-<strong>in</strong>duced<br />

Ca <strong>in</strong>flux and reduced I crac (210).<br />

Thus, <strong>in</strong> summary, Orai and Stim appear to constitute<br />

the molecular mechanism underly<strong>in</strong>g capacitative Ca entry<br />

and I crac <strong>in</strong> nonexcitable cells, and there is mount<strong>in</strong>g<br />

evidence for this also to be the case <strong>in</strong> smooth muscle. To<br />

us, the most urgent questions revolve around whether<br />

there is only one form of SOCE <strong>in</strong> smooth muscle and if<br />

this <strong>in</strong>volves only the Stim-Orai mechanism. The relative<br />

simplicity of Ca entry <strong>in</strong> nonexcitable cells falls away <strong>in</strong><br />

studies of smooth muscle, as SOCE is a variable feature<br />

between myocytes, and several other Ca entry pathways<br />

exist. In addition, there are only relatively few studies that<br />

have gone beyond demonstrat<strong>in</strong>g a putative SOCE, i.e.,<br />

mechanistic details, measurement of I crac, and biophysical<br />

and imag<strong>in</strong>g studies are sparse, lead<strong>in</strong>g to <strong>in</strong>terpretation<br />

based on only a few myocyte preparations. The <strong>in</strong>teraction<br />

of TRPCs and Orai channels, if any, also requires<br />

resolution.<br />

4. Evidence for store-operated Ca entry<br />

<strong>in</strong> smooth muscle<br />

As one might anticipate from know<strong>in</strong>g that the contribution<br />

of voltage-gated Ca entry varies between smooth<br />

muscles, be<strong>in</strong>g high for example <strong>in</strong> uter<strong>in</strong>e (771) and<br />

<strong>in</strong>test<strong>in</strong>al (64) and low <strong>in</strong> many large blood vessels and<br />

tracheal myocytes (7), so the contribution of Ca entry<br />

through SOCE and/or other voltage-<strong>in</strong>dependent routes<br />

such as nonspecific receptor-operated cation channels<br />

(ROCs) shows considerable variation between smooth<br />

muscles (44). Although ROC activity is a feature of agonist<br />

b<strong>in</strong>d<strong>in</strong>g, e.g., norep<strong>in</strong>ephr<strong>in</strong>e, and is important for<br />

their physiological effects (39), it does not <strong>in</strong>volve SR Ca<br />

depletion, and therefore, these channels are not discussed<br />

<strong>in</strong> detail further.<br />

In smooth muscles, SOCE has been associated with a<br />

second, delayed Ca response to agonist applications; the<br />

IP 3 formed causes an <strong>in</strong>itial fast and transient release of<br />

Ca from the SR, followed by the slower process of SOCE<br />

which produces a more or less susta<strong>in</strong>ed Ca signal, often<br />

of lower amplitude than the <strong>in</strong>itial transient rise. If Ca<br />

sensitization mechanisms are also stimulated by the agonist,<br />

then the force responses will not be directly related<br />

to the Ca signal and force may cont<strong>in</strong>ue long after [Ca]<br />

has fallen.<br />

A susta<strong>in</strong>ed <strong>in</strong>crease <strong>in</strong> [Ca] due to extracellular Ca<br />

entry follow<strong>in</strong>g agonist application has been taken as<br />

evidence for SOCE by many <strong>in</strong>vestigators, sometimes augmented<br />

by data from putative blockers of SOCE such as<br />

SKF96365 and La 3 . It is generally accepted that there are


no specific blockers of SOCE, so pharmacological approaches<br />

are helpful but not def<strong>in</strong>itive. Separat<strong>in</strong>g SOCE<br />

from voltage-gated Ca channel and ROC-mediated Ca entry<br />

must also be accomplished. This is often done by not<br />

us<strong>in</strong>g agonists, deplet<strong>in</strong>g the SR of Ca us<strong>in</strong>g SERCA <strong>in</strong>hibitors,<br />

and prevent<strong>in</strong>g L-type Ca entry, and <strong>in</strong>terpretation<br />

is further simplified by the use of s<strong>in</strong>gle cells. Such<br />

conditions make it hard to relate the f<strong>in</strong>d<strong>in</strong>gs to physiological<br />

conditions. With the use of these approaches, rises<br />

<strong>in</strong> [Ca] considered to be due to SOCE have been reported<br />

<strong>in</strong> many smooth muscles, e.g., colonic (370), portal ve<strong>in</strong><br />

(538), cultured A10 cells (784), <strong>in</strong>ferior vena cava (120),<br />

aorta (265), coronary arteries (731), ileal myocytes (526),<br />

pulmonary (216), esophagus (740), and gallbladder myocytes<br />

(492).<br />

There have been few studies <strong>in</strong> smooth muscle of the<br />

membrane currents or conductance changes associated<br />

with SOCE. Record<strong>in</strong>gs of what would be called I crac <strong>in</strong><br />

nonexcitable cells are often denoted as I soc <strong>in</strong> smooth<br />

muscle, underl<strong>in</strong><strong>in</strong>g the uncerta<strong>in</strong>ty around whether<br />

SOCE <strong>in</strong> smooth muscle uses the same entry mechanism<br />

as <strong>in</strong> nonexcitable cells. An added complexity to the study<br />

of I crac/I soc <strong>in</strong> smooth muscle is that the plethora of ion<br />

channels means that any changes <strong>in</strong> cytoplasmic [Ca] due<br />

to SOCE could evoke changes <strong>in</strong> other currents, e.g.,<br />

open<strong>in</strong>g of Ca-activated Cl channels, giv<strong>in</strong>g rise to <strong>in</strong>ward<br />

current and therefore complicat<strong>in</strong>g <strong>in</strong>terpretation. Studies<br />

of I crac should ideally therefore be performed with heavy<br />

buffer<strong>in</strong>g of Ca, e.g., 10 mM EGTA or BAPTA. From<br />

experiments us<strong>in</strong>g whole cell and s<strong>in</strong>gle-channel record<strong>in</strong>gs,<br />

evidence for SOCE has been provided <strong>in</strong> a variety of<br />

smooth muscles. The first record<strong>in</strong>g of I crac was made <strong>in</strong><br />

mouse anococcygeous myocytes <strong>in</strong> 1996 (746). Subsequent<br />

record<strong>in</strong>gs of a conductance elicited by store Ca<br />

depletion were made <strong>in</strong> portal ve<strong>in</strong> (5, 403), mesenteric<br />

(609) and pulmonary arteries (216, 705), and airway (549).<br />

Thus, as expected, most evidence for significant SOCE<br />

comes from blood vessels, where reliance on L-type Ca<br />

entry is less than <strong>in</strong> many phasic muscles.<br />

Work particularly from Large’s group (7) has questioned<br />

whether SOCE is the same process <strong>in</strong> smooth<br />

muscle as it is <strong>in</strong> nonexcitable cells and if different forms<br />

of SOCE occur <strong>in</strong> smooth muscle cells. As discussed<br />

above, there is mount<strong>in</strong>g evidence for Orai and Stim <strong>in</strong><br />

smooth muscles, <strong>in</strong>clud<strong>in</strong>g vascular and airway, although<br />

this does not prove that I crac and SOCE are identical to<br />

those described <strong>in</strong> nonexcitable cells. In a careful review<br />

of the biophysical properties and activation mechanism<br />

for SOCE <strong>in</strong> a small number of different smooth muscles,<br />

Albert et al. (7) concluded from measurements of unitary<br />

conductance (not current) that the Ca permeability of<br />

SOCs <strong>in</strong> smooth muscle myocytes was much smaller than<br />

measured for I crac <strong>in</strong> nonexcitable cells and that therefore<br />

<strong>in</strong> myocytes SOCE is via nonspecific cation channels.<br />

Such channels will contribute to SR Ca refill<strong>in</strong>g but also to<br />

SMOOTH MUSCLE SR 143<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

depolarization of the myocytes. Albert et al. (7) argue that<br />

such channels may also have TRPCs associated with them<br />

and be activated by factors other than Stim; mention has<br />

been made of Ca-<strong>in</strong>dependent phospholipase A 2 (iPLA 2)<br />

activat<strong>in</strong>g SOCE (644), and there is also evidence for a<br />

role for PKC (6, 609). F<strong>in</strong>ally, store-<strong>in</strong>dependent activation<br />

of SOCE <strong>in</strong>volv<strong>in</strong>g PKC has been described by this<br />

group <strong>in</strong> portal ve<strong>in</strong> (6), with the suggestion that there is<br />

a constitutively active driver of SOCE.<br />

What the relation of these channels is to classical<br />

SOCE/capacitative Ca entry rema<strong>in</strong>s to be established,<br />

but clearly such channels fall outside the conventional<br />

def<strong>in</strong>ition of SOCE. The use of s<strong>in</strong>gle-channel record<strong>in</strong>g <strong>in</strong><br />

isolated patches of membrane clearly provides experimental<br />

clarity, but it is difficult to relate these events to<br />

physiological activity <strong>in</strong> <strong>in</strong>tact cells or tissue, where [Ca]<br />

would not be clamped and Stim-Orai activity would be<br />

present.<br />

There are data which <strong>in</strong>dicate that refill<strong>in</strong>g of the<br />

store occurs directly from the myoplasm (267, 486). It was<br />

shown that when <strong>in</strong>tracellular [Ca] was clamped around<br />

rest<strong>in</strong>g levels, us<strong>in</strong>g Ca chelators, store refill<strong>in</strong>g occurred<br />

<strong>in</strong> the absence of external Ca, suggest<strong>in</strong>g that mechanisms<br />

directly l<strong>in</strong>k<strong>in</strong>g Ca <strong>in</strong>flux to store refill<strong>in</strong>g might not<br />

be necessary (54, 267, 486). Certa<strong>in</strong>ly, SR Ca uptake when<br />

cytoplasmic [Ca] rises (<strong>in</strong> the absence of external Ca) has<br />

been directly demonstrated (636).<br />

If the plasma membrane Ca leak is small and SERCA<br />

activity large, then many cycles of SR Ca release and<br />

uptake could occur without the need for Ca entry. In<br />

those smooth muscles where L-type Ca entry is large and<br />

agonists produce depolarization, then there is little need<br />

for a separate SOCE/CCE mechanism. For smooth muscles<br />

such as the uterus, SOCE may be a mechanism that is<br />

only important dur<strong>in</strong>g the strong agonist-driven contractions<br />

of labor, and that is absent/unused at other times<br />

(518, 707). The heterogeneous nature of the stores (see<br />

sect. VII) may also contribute to differences <strong>in</strong> SOCE<br />

between smooth muscles. For example, <strong>in</strong> arterial myocytes,<br />

the ryanod<strong>in</strong>e/caffe<strong>in</strong>e-sensitive store was not sensitive<br />

to either thapsigarg<strong>in</strong> or CPA, whereas the IP 3sensitive<br />

Ca store was depleted by both of them (307).<br />

From the results obta<strong>in</strong>ed on colonic myocytes, which<br />

have two functionally dist<strong>in</strong>ct SR Ca stores, it was suggested<br />

that the common store (S ) express<strong>in</strong>g both IP 3R<br />

and RyR was dependent on an external Ca source for<br />

replenishment, and the second store conta<strong>in</strong><strong>in</strong>g only RyR<br />

(S ) refills directly from the myoplasm (185). Also <strong>in</strong><br />

colonic myocytes, the mechanisms that control Ca entry<br />

<strong>in</strong>to the cytoplasm <strong>in</strong>volve two types of channels, one of<br />

which is voltage sensitive and the other is voltage <strong>in</strong>sensitive<br />

(453). It is also suggested that the <strong>in</strong>flux of Ca via<br />

these two channels produces a high subplasmalemmal<br />

[Ca] accessed by the common store (453). However, other<br />

authors have found that the magnitude of SOCE <strong>in</strong> colonic


144 SUSAN WRAY AND THEODOR BURDYGA<br />

myocytes was the same irrespective of whether just IP 3R<br />

or IP 3R and RyR stores were depleted (370). The refill<strong>in</strong>g<br />

mechanisms may depend on the location of the SR. Peripherally<br />

located stores may be functionally more closely<br />

l<strong>in</strong>ked to Ca entry via SOCE channels (215), while those<br />

located centrally could be refilled from the myoplasm.<br />

Another consideration is the type of SERCA associated<br />

with each of the stores. The Ca b<strong>in</strong>d<strong>in</strong>g aff<strong>in</strong>ity (K m) for<br />

SERCA2a is 0.31 M and for SERCA2b is 0.17 M (479).<br />

SERCA2b activity can be modulated by calmodul<strong>in</strong> (229)<br />

and phospholamban (162), and aga<strong>in</strong> this could affect<br />

cross-talk between the store and plasma membrane. If<br />

different isoforms of SERCA are associated with different<br />

stores, this could expla<strong>in</strong> differences <strong>in</strong> their ability to<br />

refill the SR <strong>in</strong> the presence of different [Ca].<br />

6. Summary<br />

It seems to us, based on the current state of knowledge,<br />

that SOCE has been demonstrated <strong>in</strong> some but not<br />

all smooth muscles and that this will occur through a<br />

Stim-Orai mechanism when the channel <strong>in</strong>volved gives<br />

rise to I crac. Variations on this mechanism may occur <strong>in</strong><br />

some smooth muscles, but it rema<strong>in</strong>s to be established if<br />

these are true variations on SOCE, or different channels<br />

such as ROCs. TRPCs may contribute to variation of<br />

SOCE <strong>in</strong> smooth muscle, lead<strong>in</strong>g to decreased Ca selectivity,<br />

but this is far from certa<strong>in</strong>. More studies, both<br />

molecular and physiological, are clearly called for, and it<br />

is important to have more <strong>in</strong>formation from nonvascular<br />

myocytes. This rema<strong>in</strong>s a controversial and complex area<br />

(396) but one of some importance. The role of SOCE<br />

pathways may change with disease states, e.g., hypertension<br />

and bronchoconstriction, as well as phenotype, e.g.,<br />

changes <strong>in</strong> arterial myocytes with proliferation, migration,<br />

or contraction (396), and thus an <strong>in</strong>creased understand<strong>in</strong>g<br />

could lead to new therapeutic targets.<br />

There is little doubt that TRP channels are expressed<br />

<strong>in</strong> many smooth muscles, particularly, but not exclusively<br />

tonic ones. As discussed, this expression does not necessarily<br />

equate to SOCE function. One recent <strong>in</strong>terest<strong>in</strong>g<br />

paper has provided evidence that TRPC channels become<br />

converted to SOCE channels as Stim1 leads to their movement<br />

from non-raft (TRCP) to raft (SOCE) membrane<br />

locations (10), whereas another recent study had TRCP<br />

channels <strong>in</strong> raft doma<strong>in</strong>s not Orai1 (148). There is evidence<br />

from only a limited number of smooth muscles of<br />

I crac; whether this scarcity is due to the difficulties of<br />

record<strong>in</strong>g these t<strong>in</strong>y currents or their absence from most<br />

smooth muscles is impossible to determ<strong>in</strong>e at the moment.<br />

Studies on cultured cells are limited models for<br />

native smooth muscle cells, as the downregulation of the<br />

normal components of E-C coupl<strong>in</strong>g and changes <strong>in</strong> SOCE<br />

will distort f<strong>in</strong>d<strong>in</strong>gs (512). The recent discoveries of Orai<br />

and Stim should help better study store-operated Ca entry<br />

<strong>in</strong> all cell types. Firm conclusions must await the ability to<br />

demonstrate store-operated entry under physiological<br />

conditions, e.g., agonist depletion, along with direct measures<br />

of lum<strong>in</strong>al SR Ca content. It is worth not<strong>in</strong>g that <strong>in</strong><br />

cardiac muscle where the SR plays a dom<strong>in</strong>ant role <strong>in</strong> E-C<br />

coupl<strong>in</strong>g, no such SOCE mechanism exists. Perhaps <strong>in</strong><br />

those smooth muscles <strong>in</strong> which Ca entry is the dom<strong>in</strong>ant<br />

feature of force production, SOCE will also not be important.<br />

In contrast, <strong>in</strong> those smooth muscles that are more<br />

reliant on SR Ca contributions to contraction and have<br />

significant neurohormonal stimulation, then store-operated<br />

Ca entry may have an important role.<br />

VIII. COMMON OR SEPARATE CALCIUM POOLS<br />

A. Introduction<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

The presence of two types (at least) of Ca release<br />

channels on the SR, which can <strong>in</strong>teract with each other<br />

to control local and global Ca signals, adds another<br />

level of complexity to the regulation of Ca signal<strong>in</strong>g by<br />

the SR <strong>in</strong> smooth muscles. The scope for such <strong>in</strong>teractions<br />

<strong>in</strong> any particular smooth muscle will depend on<br />

the morphological arrangement of the store(s), isoform<br />

expression, receptor distribution, and the cytoplasmic<br />

and lum<strong>in</strong>al [Ca] (Fig. 5). There is reasonable evidence<br />

to support the view that the SR has heterogeneous<br />

compartments of releasable Ca <strong>in</strong> smooth muscle cells.<br />

In view of this, it is helpful to frame the discussion that<br />

follows accord<strong>in</strong>g to the type of Ca store. These have<br />

been classified <strong>in</strong>to three subtypes: S (conta<strong>in</strong><strong>in</strong>g both<br />

IP 3 and RyRs), S (conta<strong>in</strong><strong>in</strong>g only IP 3 receptors) (281,<br />

285), and S (conta<strong>in</strong><strong>in</strong>g only RyRs) (307). The relative<br />

distribution of each of these stores can vary considerably,<br />

not only among different types of smooth muscles<br />

but also among different species and with development.<br />

In the studies described below, a couple of general<br />

po<strong>in</strong>ts should be borne <strong>in</strong> m<strong>in</strong>d. First, a variety of<br />

different agonists, appropriate to particular smooth<br />

muscles, have been used as a mechanism to generate<br />

IP 3, and any differences <strong>in</strong> their other actions are often<br />

ignored. Second, when conclusions are based on pharmacological<br />

data, the specificity or otherwise of the<br />

drugs must not be overlooked. Mention has already<br />

been made of the problems associated with this approach<br />

to IP 3 receptor blockade, and similar concerns<br />

over some agents used to <strong>in</strong>hibit RyRs have also been<br />

raised (433).<br />

B. One Store With One Type of Receptor<br />

One of the clearest examples of cells hav<strong>in</strong>g just one<br />

type of Ca release channel is provided by the ureter. In rat


ureteric myocytes, SR Ca release was shown to be agonist<br />

sensitive but not caffe<strong>in</strong>e sensitive. In the gu<strong>in</strong>ea pig<br />

ureter, Ca release was caffe<strong>in</strong>e sensitive but agonist <strong>in</strong>sensitive.<br />

These data suggest that IP 3R plays the key role<br />

<strong>in</strong> control of Ca release <strong>in</strong> rat and RyRs <strong>in</strong> the gu<strong>in</strong>ea pig<br />

ureteric cells (91, 92). With<strong>in</strong> the above classification, rat<br />

has an S store and gu<strong>in</strong>ea pig ureter S .<br />

It was subsequently shown that rat ureteric myocytes<br />

generated ryanod<strong>in</strong>e-resistant, hepar<strong>in</strong>-sensitive Ca puffs<br />

and Ca waves when stimulated by IP 3 or ACh (60, 91),<br />

SMOOTH MUSCLE SR 145<br />

FIG. 5. Diagram of proposed SR arrangements and possible function <strong>in</strong> smooth muscle cells. A, a–e: schematic diagram show<strong>in</strong>g proposed<br />

arrangements of the SR Ca stores reported for different types of smooth muscles. The Ca store <strong>in</strong> many types of smooth muscle cells can be<br />

compartmentalized <strong>in</strong>to smaller SR elements that might conta<strong>in</strong> either RyR (green) or IP 3R (red) Ca release channels or a mixture of these channels<br />

(blue). RyRs can be clustered to form specialized Ca release units located <strong>in</strong> close proximity to the cell membrane where they generate Ca sparks.<br />

IP 3Rs <strong>in</strong> most types of smooth muscle are homogeneously distributed <strong>in</strong> the cell and are ma<strong>in</strong>ly <strong>in</strong>volved <strong>in</strong> generation of Ca waves and Ca<br />

oscillations, with or without <strong>in</strong>volvement of the RyR channels. B: schematic diagram illustrat<strong>in</strong>g Ca spark/spontaneous transient outward current<br />

(STOC) coupl<strong>in</strong>g mechanism discovered by Nelson et al. (507), which acts as a powerful negative-feedback mechanism controll<strong>in</strong>g excitability and<br />

electrical activity and has been reported <strong>in</strong> the majority of smooth muscles. Ca sparks can be activated by Ca enter<strong>in</strong>g the cell via L-type Ca channels<br />

(Ca-<strong>in</strong>duced Ca release; CICR) or an <strong>in</strong>crease <strong>in</strong> the lum<strong>in</strong>al level of Ca by SERCa pump. BK Ca, Ca-activated K channels; VOC, voltage-operated Ca<br />

channels. C: possible mechanisms for the generation of global Ca waves <strong>in</strong> smooth muscle cells. In a, the Ca wave is produced by activation of RyR<br />

channels via CICR triggered by Ca. In b, the Ca wave is triggered by IP 3 but propagated and amplified by RyR channels (cooperative action between<br />

two types of Ca release channels). In c, the Ca wave is triggered, amplified, and propagated by IP 3R channels.<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

while gu<strong>in</strong>ea pig ureteric cells generated ryanod<strong>in</strong>e-sensitive<br />

Ca sparks enhanced by low concentrations of caffe<strong>in</strong>e<br />

(76, 87). The functional data obta<strong>in</strong>ed on rat ureter<br />

were also supported by the data obta<strong>in</strong>ed us<strong>in</strong>g molecular<br />

biology and immunocytochemistry techniques (60, 493).<br />

Thus rat myocytes expressed predom<strong>in</strong>antly IP 3Rs (all<br />

three isoforms) and only a trace of one isoform of RyR<br />

(RyR3) (60).<br />

In s<strong>in</strong>gle myometrial cells from pregnant rats (17),<br />

oxytoc<strong>in</strong> and ACh evoked an <strong>in</strong>itial peak <strong>in</strong> [Ca] followed


146 SUSAN WRAY AND THEODOR BURDYGA<br />

by a smaller susta<strong>in</strong>ed rise. The transient <strong>in</strong>crease <strong>in</strong> [Ca]<br />

was abolished by hepar<strong>in</strong>, an <strong>in</strong>hibitor of IP 3-<strong>in</strong>duced Ca<br />

release (IICR) and thapsigarg<strong>in</strong>. In contrast, the transient<br />

[Ca] response <strong>in</strong>duced by oxytoc<strong>in</strong> was unaffected by<br />

ryanod<strong>in</strong>e. In permeabilized fibers of pregnant rat myometrium,<br />

caffe<strong>in</strong>e did not produce contraction, whereas<br />

both IP 3 and the ionophore A23187 evoked contractile<br />

responses (620). These data show that myometrial cells<br />

possess an S but not S store.<br />

C. One Store With Two Types of Receptors<br />

The majority of smooth muscle cells are both agonist<br />

sensitive and caffe<strong>in</strong>e sensitive and have one common<br />

store (S ) shared by both IP 3R and RyR (62, 449, 537, 755).<br />

In addition, these cells may have additional, i.e., separate,<br />

agonist- or caffe<strong>in</strong>e-sensitive Ca stores (673, 687). For<br />

example, <strong>in</strong> rat mesenteric artery myocytes (32), norep<strong>in</strong>ephr<strong>in</strong>e<br />

and caffe<strong>in</strong>e produced a transient <strong>in</strong>crease <strong>in</strong><br />

[Ca] <strong>in</strong> Ca-free solution. In the presence of norep<strong>in</strong>ephr<strong>in</strong>e,<br />

caffe<strong>in</strong>e or thapsigarg<strong>in</strong> elevated [Ca]. However, if<br />

thapsigarg<strong>in</strong> or caffe<strong>in</strong>e were added first, the subsequent<br />

application of norep<strong>in</strong>ephr<strong>in</strong>e did not <strong>in</strong>crease [Ca].<br />

These results suggest the existence of two types of Ca<br />

stores: one store sensitive to both caffe<strong>in</strong>e and agonist<br />

(S ) and one sensitive to caffe<strong>in</strong>e and thapsigarg<strong>in</strong> but not<br />

to agonists (S ). Evidence that <strong>in</strong> some smooth muscles<br />

there is just one functional store but that it possesses both<br />

receptor types comes from direct imag<strong>in</strong>g of the store Ca<br />

(755). One difficulty <strong>in</strong> demonstrat<strong>in</strong>g either one or multiple<br />

Ca stores comes from the reciprocal actions of both<br />

the release channels on lum<strong>in</strong>al Ca, i.e., if the Ca store is<br />

shared, as both IP 3R and RyR are sensitive to cytosolic<br />

and lum<strong>in</strong>al [Ca], the release of Ca from one population<br />

will affect the P o of the other. There are many examples<br />

described below of reciprocal and synergistic effects on<br />

Ca signal<strong>in</strong>g aris<strong>in</strong>g from hav<strong>in</strong>g both IP 3R and RyRs <strong>in</strong><br />

the same smooth muscle cells. This does not, however, by<br />

itself prove that they access entirely the same Ca store.<br />

Identical patterns of store depletion upon exposure to<br />

caffe<strong>in</strong>e or agonist, consistent with the idea that IP 3Rs<br />

and RyRs release Ca from the same store, have been<br />

demonstrated, highly suggestive of a s<strong>in</strong>gle Ca store (755).<br />

D. Two Stores, Two Types of Receptors<br />

Early experiments based on the ability of caffe<strong>in</strong>e<br />

and IP 3 to release Ca from permeabilized strips of taenia<br />

caeci, portal ve<strong>in</strong>, and pulmonary artery demonstrated the<br />

existence of two separate Ca stores, with one gated by<br />

both RyRs and IP 3Rs (S ) and the other by IP 3Rs alone<br />

(S ) (281, 282). In taenia caecum and pulmonary artery,<br />

IP 3 alone or <strong>in</strong> comb<strong>in</strong>ation with caffe<strong>in</strong>e-<strong>in</strong>duced Ca<br />

release, which was about twofold larger than that <strong>in</strong>duced<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

by caffe<strong>in</strong>e alone. Inhibition of S store via its depletion<br />

produced by caffe<strong>in</strong>e and ryanod<strong>in</strong>e did not affect IICR<br />

from the S store (281). The contribution of the S store<br />

to total Ca release <strong>in</strong>duced by IP 3 was 60% <strong>in</strong> taenia<br />

caecum, 40% <strong>in</strong> pulmonary artery, and only 6% <strong>in</strong> portal<br />

ve<strong>in</strong> (281). In experiments performed on <strong>in</strong>tact pulmonary<br />

artery myocytes, depletion of Ca stores with ryanod<strong>in</strong>e<br />

and caffe<strong>in</strong>e elim<strong>in</strong>ated subsequent caffe<strong>in</strong>e-<strong>in</strong>duced <strong>in</strong>tracellular<br />

Ca transients, but had little or no effect on the<br />

IP 3-mediated <strong>in</strong>tracellular Ca transient <strong>in</strong>duced by agonists<br />

(307). Experiments performed on <strong>in</strong>tact term<strong>in</strong>al<br />

arterioles also revealed the existence of two separate<br />

caffe<strong>in</strong>e-sensitive and agonist-sensitive Ca stores. In these<br />

blood vessels, Ca oscillations <strong>in</strong>duced by agonists were<br />

resistant to the comb<strong>in</strong>ed action of caffe<strong>in</strong>e and ryanod<strong>in</strong>e<br />

(74, 75). Recent work from McCarron and Olson<br />

(455) has highlighted the difficulty of us<strong>in</strong>g functional<br />

data to deduce <strong>in</strong>formation about the existence of two<br />

stores. They note that the appearance of two stores may<br />

arise because of partial depletion of SR Ca and lum<strong>in</strong>al Ca<br />

regulation of the receptors term<strong>in</strong>at<strong>in</strong>g release from one<br />

receptor only.<br />

E. Role of Ca Release Channels and Store Type<br />

<strong>in</strong> Ca Signal<strong>in</strong>g<br />

There are two key questions to be addressed here.<br />

First, can the complete set of Ca signals associated with<br />

contraction arise <strong>in</strong> smooth cells us<strong>in</strong>g just IP 3Rs or just<br />

RyRs? Second, how are IP 3-<strong>in</strong>itiated Ca signals <strong>in</strong>fluenced<br />

by RyR Ca release and vice versa?<br />

There is general agreement that the <strong>in</strong>itiation of Ca<br />

signal<strong>in</strong>g is a response to agonists caused by the release<br />

of Ca from the SR via IP 3Rs (212). However, whether Ca<br />

released from the IP 3R then activates RyR to generate<br />

further release by CICR to amplify the signal and support<br />

propagation of Ca waves <strong>in</strong> a regenerative way, is still a<br />

controversial issue. As mentioned above, rat ureteric<br />

myocytes do not express RyR1 or RyR2, the subtypes<br />

which play the major role <strong>in</strong> CICR <strong>in</strong> smooth muscle<br />

(133). The ability of these myocytes to generate Ca puffs<br />

and Ca waves <strong>in</strong> response to agonists or IP 3, which are<br />

resistant to ryanod<strong>in</strong>e and antibodies to RyR (60), demonstrates<br />

that the entire Ca release and signal<strong>in</strong>g process<br />

<strong>in</strong> smooth muscles can arise from IP 3R activity alone, i.e.,<br />

without RyR <strong>in</strong>volvement (60, 91, 92). Thus the answer to<br />

the first question posed above is yes; normal agonist<strong>in</strong>duced<br />

Ca signals and contractions can be produced by<br />

IP 3Rs alone.<br />

There are examples of <strong>in</strong>teractions between IP 3Rs<br />

and RyRs with the most noted be<strong>in</strong>g an amplification of<br />

IP 3Rs responses by RyRs. With the use of a variety of<br />

approaches, <strong>in</strong>clud<strong>in</strong>g block<strong>in</strong>g of the RyR with ryanod<strong>in</strong>e<br />

(or ruthenium red, which may have additional actions) or


us<strong>in</strong>g antibodies to RyRs (62), effects on IP 3 Ca signals<br />

have been demonstrated <strong>in</strong> a wide variety of smooth<br />

muscles. These effects <strong>in</strong>clude abolition of agonist-dependent<br />

Ca oscillations (333, 535, 638) and <strong>in</strong>hibition of Ca<br />

waves (34, 62, 303, 331). There are also many examples of<br />

IP 3 modulat<strong>in</strong>g RyR events, and affect<strong>in</strong>g both local and<br />

global Ca signals (299). Receptor activation lead<strong>in</strong>g to the<br />

generation of IP 3, <strong>in</strong>creased spark discharge from frequent<br />

discharge sites (FDS) <strong>in</strong> vascular (18, 62, 219) and<br />

visceral (755) smooth muscle cells. It has been postulated<br />

<strong>in</strong> portal ve<strong>in</strong> that IP 3 produced by the basal activity of<br />

PLC activates IP 3Rs to recruit neighbor<strong>in</strong>g clusters of<br />

RyRs with<strong>in</strong> the FDS to generate large Ca sparks (219).<br />

Inhibition of IP 3R results <strong>in</strong> a decrease <strong>in</strong> the frequency of<br />

Ca sparks, Ca oscillations, and waves <strong>in</strong> response to<br />

agonists <strong>in</strong> visceral and vascular smooth muscles (18, 62,<br />

299, 333, 535, 638).<br />

A physiological consequence of <strong>in</strong>teraction between<br />

IP 3- and RyR-sensitive Ca channels will be positive<br />

feedback amplify<strong>in</strong>g Ca release. In porc<strong>in</strong>e tracheal<br />

smooth muscle cells, ryanod<strong>in</strong>e and ruthenium<br />

red <strong>in</strong>hibited ACh-<strong>in</strong>duced Ca oscillations (333, 574).<br />

Similarly, the P2Y receptor agonists UTP and 2-methylthio-ATP<br />

each <strong>in</strong>creased the occurrence of Ca waves <strong>in</strong><br />

rat cerebral artery and mur<strong>in</strong>e colonic myocytes, effects<br />

which were blocked by ryanod<strong>in</strong>e (34, 303). In<br />

rabbit cerebral artery (332) and renal resistance arteries<br />

(307), agonist-<strong>in</strong>duced Ca oscillations were blocked<br />

by ryanod<strong>in</strong>e, suggest<strong>in</strong>g that RyR-dependent CICR<br />

contributed to the [Ca] rise.<br />

Agonist-<strong>in</strong>duced propagat<strong>in</strong>g Ca oscillations that<br />

orig<strong>in</strong>ate from FDSs have been demonstrated <strong>in</strong> several<br />

smooth muscles. For example, ACh-<strong>in</strong>duced Ca oscillations<br />

<strong>in</strong> tracheal smooth muscle orig<strong>in</strong>ate from areas of<br />

the cell display<strong>in</strong>g the highest frequency of Ca sparks but<br />

require SR Ca release through IP 3 receptor channels for<br />

<strong>in</strong>itiation (333, 535, 638). In gastric myocytes, Ca waves<br />

evoked by carbachol were <strong>in</strong>itiated from the same sites as<br />

those evoked by caffe<strong>in</strong>e, and these sites also colocalized<br />

with spontaneous spark sites. These observations suggest<br />

that RyRs were <strong>in</strong>volved <strong>in</strong> both cases, and the carbachol<br />

responses were <strong>in</strong>hibited by both IP 3R and RyR blockers<br />

(755). Inhibition of RyRs with antibodies or with ryanod<strong>in</strong>e<br />

abolished agonist-dependent Ca transients and<br />

waves, <strong>in</strong> both visceral and vascular smooth muscles with<br />

one functional store shared by both types of receptor<br />

channels (62, 223, 300, 755, 756).<br />

These f<strong>in</strong>d<strong>in</strong>gs suggest that agonists can stimulate Ca<br />

release <strong>in</strong> smooth muscle via mechanisms that are both<br />

IP 3R and RyR dependent and support a model <strong>in</strong> which<br />

activation of IP 3Rs acts as a trigger lead<strong>in</strong>g to secondary<br />

activation of RyRs. Does this mean that all IP 3R-activated<br />

responses are amplified by RyRs when they are expressed?<br />

Recent observations strongly suggest the scope<br />

for such <strong>in</strong>teractions varies among different types of<br />

SMOOTH MUSCLE SR 147<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

smooth muscles and depends on receptor distribution<br />

and isoform expression.<br />

There is much evidence that <strong>in</strong> some smooth muscles<br />

<strong>in</strong> which IP 3R and RyR share the same store, there is a<br />

noncooperative relationship between the two types of<br />

channels (29, 185, 449). For example, adrenergic activation<br />

decreased the frequency of Ca sparks <strong>in</strong> <strong>in</strong>tact rat<br />

resistance arteries; this was suggested to be due to a<br />

decrease <strong>in</strong> the SR Ca content (449). Blockade of IP 3Rs <strong>in</strong><br />

smooth muscle cells from both the gastric antrum and vas<br />

deferens did not decrease, but <strong>in</strong> fact <strong>in</strong>creased, both<br />

caffe<strong>in</strong>e-releasable store content and spontaneous spark<br />

activity (755, 756). However, <strong>in</strong> rat resistance artery myocytes,<br />

IP 3R blockade did not affect the temporal or spatial<br />

characteristics of Ca sparks (380).<br />

There is, therefore, clear evidence that <strong>in</strong>creases<br />

and decreases <strong>in</strong> IP 3R activity can lead to reciprocal<br />

changes <strong>in</strong> the activity of RyRs through changes <strong>in</strong><br />

lum<strong>in</strong>al SR [Ca]. However, some <strong>in</strong>hibition of Ca sparks<br />

by agonists could be expla<strong>in</strong>ed by activation of PKC,<br />

which also decreases Ca spark frequency, by directly<br />

<strong>in</strong>hibit<strong>in</strong>g RyRs (70).<br />

In equ<strong>in</strong>e tracheal myocytes, the Ca release evoked by<br />

the IP 3-generat<strong>in</strong>g muscar<strong>in</strong>ic agonist methachol<strong>in</strong>e was unaffected,<br />

while that evoked by caffe<strong>in</strong>e was blocked by<br />

ruthenium red (739). In colonic myocytes, ryanod<strong>in</strong>e alone<br />

did not <strong>in</strong>hibit IP 3-evoked [Ca] <strong>in</strong>creases, but when applied<br />

with caffe<strong>in</strong>e allow<strong>in</strong>g ryanod<strong>in</strong>e to lock RyR channels <strong>in</strong> a<br />

subconduct<strong>in</strong>g state, IP 3-mediated [Ca] <strong>in</strong>creases were<br />

blocked. These results were attributed to the drug’s <strong>in</strong>direct<br />

<strong>in</strong>hibition of the IP 3-mediated response by store depletion of<br />

Ca, rather than to <strong>in</strong>volvement of RyR <strong>in</strong> Ca release (185).<br />

Similar data have been reported for the smooth muscle cells<br />

of <strong>in</strong>terpulmonary bronchioles and arterioles, <strong>in</strong> which under<br />

normal conditions the RyR was not essential for generat<strong>in</strong>g<br />

agonist-<strong>in</strong>duced Ca oscillations (29, 556, 557). These<br />

results were consistent with the observation that the release<br />

of caged Ca was able to <strong>in</strong>itiate CICR via RyR <strong>in</strong> the smooth<br />

muscle of the <strong>in</strong>terpulmonary bronchioles only when the<br />

cells were previously depolarized with KCl to overload the<br />

Ca stores and sensitize the RyR.<br />

Acetylchol<strong>in</strong>e, norep<strong>in</strong>ephr<strong>in</strong>e, and nerve stimulation<br />

each released Ca <strong>in</strong> porc<strong>in</strong>e coronary, rabbit mesenteric,<br />

and rat tail arteries, respectively (284, 299, 342, 449). The<br />

release of Ca by the neurotransmitters was <strong>in</strong>hibited by<br />

ryanod<strong>in</strong>e <strong>in</strong> each study and attributed to the drug’s ability<br />

to open RyRs and deplete the Ca store. Norep<strong>in</strong>ephr<strong>in</strong>e-evoked<br />

Ca waves persisted <strong>in</strong> tail artery segments<br />

ma<strong>in</strong>ta<strong>in</strong>ed <strong>in</strong> organ culture for 3 days with ryanod<strong>in</strong>e,<br />

while Ca sparks and Ca release evoked by caffe<strong>in</strong>e were<br />

lost, suggest<strong>in</strong>g that RyR does not directly contribute to<br />

Ca waves <strong>in</strong> this preparation (154). Ruthenium red and<br />

tetraca<strong>in</strong>e (each of which abolished the [Ca] <strong>in</strong>crease<br />

evoked by caffe<strong>in</strong>e without deplet<strong>in</strong>g the store) did not<br />

affect the Ca oscillations <strong>in</strong>duced by agonists <strong>in</strong> pulmo-


148 SUSAN WRAY AND THEODOR BURDYGA<br />

nary arterial myocytes (279). In gu<strong>in</strong>ea pig colonic myocytes<br />

<strong>in</strong> the absence of RyR activity, repetitive Ca release<br />

events <strong>in</strong>duced by photolysis of caged IP 3 were not <strong>in</strong>hibited<br />

by tetraca<strong>in</strong>e or ryanod<strong>in</strong>e (185). This suggests that<br />

IP 3 receptor activity alone accounted for the Ca waves<br />

and loss of the ability of agonists to <strong>in</strong>duce SR Ca release<br />

after caffe<strong>in</strong>e results from Ca depletion <strong>in</strong> a common<br />

store (185).<br />

F. Summary<br />

From the above we propose that the functional implications<br />

of RyRs <strong>in</strong> neurotransmitter-<strong>in</strong>duced Ca waves<br />

or oscillations depend on their density relative to IP 3R<br />

and that they share the same store. In smooth muscles<br />

display<strong>in</strong>g a higher density of IP 3Rs than of RyRs (e.g.,<br />

colonic smooth muscles, Ref. 758), the Ca responses to<br />

neurotransmitters will ma<strong>in</strong>ly depend on activation of IP 3<br />

receptors alone (e.g., Ref. 366). In smooth muscles display<strong>in</strong>g<br />

a higher density of RyRs than IP 3Rs (e.g., rat<br />

portal ve<strong>in</strong>), the Ca waves and oscillations <strong>in</strong>duced by<br />

neurotransmitters will depend on activation of both IP 3Rs<br />

and RyRs. However, comparative functional experiments<br />

correlated with expression and distribution of IP 3R and<br />

RyR channels <strong>in</strong> other types of smooth muscles are<br />

needed to expand this conclusion to all types of smooth<br />

muscles.<br />

To summarize these data, we can conclude that the<br />

functional studies that employed blockers of SERCAs <strong>in</strong><br />

conjunction with activators of IP 3Rs and RyRs shed some<br />

light on the morphological organization of <strong>in</strong>tracellular Ca<br />

stores. In some types of smooth muscles, morphological<br />

studies employ<strong>in</strong>g immunofluorescence and molecular biology<br />

techniques provided <strong>in</strong>formation that could expla<strong>in</strong><br />

the functional data. However, to fully understand the<br />

functional role of the SR <strong>in</strong> smooth muscles, confocal<br />

microscopy and immuno-EM are needed to exam<strong>in</strong>e the<br />

relationship between IP 3R and RyR isoforms and SERCA,<br />

to substantiate conclusions drawn from these functional<br />

studies.<br />

IX. ELEMENTAL CALCIUM SIGNALS<br />

FROM SMOOTH MUSCLE<br />

SARCOPLASMIC RETICULUM<br />

A. Ca Sparks<br />

1. Introduction<br />

Calcium sparks were first identified <strong>in</strong> cardiac (122,<br />

544) and skeletal (354, 708) muscles, where their fundamental<br />

role <strong>in</strong> the generation of global Ca signals underly<strong>in</strong>g<br />

phasic contractions has been well established. In<br />

smooth muscle cells Ca sparks were first observed <strong>in</strong><br />

cerebral artery and showed biophysical and pharmacological<br />

characteristics similar to those of cardiac muscle<br />

cells (507). S<strong>in</strong>ce then, similar events have been described<br />

<strong>in</strong> a wide variety of smooth muscle types <strong>in</strong>clud<strong>in</strong>g different<br />

types of arteries and arterioles (70, 90, 192, 305, 473,<br />

568, 749, 751), portal ve<strong>in</strong> (219, 221, 474), ur<strong>in</strong>ary bladder<br />

(127, 254–256, 288, 312), gastro<strong>in</strong>test<strong>in</strong>al tract (34, 35, 220,<br />

223, 351, 808, 809, 811), airways (369, 810), gallbladder<br />

(572), and gu<strong>in</strong>ea pig ureter (76, 87). A lack of Ca sparks<br />

has been reported for smooth muscle cells of neonatal<br />

cerebral arteries (213), nonpregnant mouse (476), and<br />

pregnant and nonpregnant rat myometrium (88).<br />

Calcium sparks <strong>in</strong> smooth muscle can occur spontaneously<br />

(76, 87, 220, 507) or after activation by one of the<br />

follow<strong>in</strong>g factors: 1) low concentrations (1 mM) of caffe<strong>in</strong>e<br />

(76, 87, 213, 307, 807), 2) elevation of global [Ca]<br />

caused by Ca enter<strong>in</strong>g through L-type Ca channels (312,<br />

508), 3) <strong>in</strong>crease <strong>in</strong> the SR Ca content (87, 811), and<br />

4) stretch (312).<br />

2. Frequent discharge sites<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

Irrespective of the mechanism of activation, Ca<br />

sparks repeatedly arise from a few specialized regions<br />

adjacent to the superficially located SR with<strong>in</strong> the myocytes;<br />

these have been termed FDS (65, 87, 220, 221, 288,<br />

312, 508). These areas are enriched with clusters of RyR2<br />

channels (133, 213, 525). It should, however, also be noted<br />

that frequent discharge events away from the cell membrane<br />

have also been reported <strong>in</strong> smooth muscle cells<br />

from rat portal ve<strong>in</strong> (18, 65) and gastro<strong>in</strong>test<strong>in</strong>al tract<br />

(220, 807). The number of FDS varies not only among<br />

different types of smooth muscle cells (65, 87, 219, 220,<br />

807), but also among different populations of the same<br />

type of smooth muscle (65). The number of FDS is <strong>in</strong>creased<br />

<strong>in</strong> the presence of low concentrations of caffe<strong>in</strong>e<br />

(76, 307, 807) or after <strong>in</strong>creas<strong>in</strong>g the SR Ca content (87,<br />

811). Such a non-uniform distribution of FDS <strong>in</strong> smooth<br />

muscle may arise from either the irregular cluster<strong>in</strong>g of<br />

RyR Ca release channels, or nonuniform distribution of<br />

the SERCA pump on the SR. Cluster<strong>in</strong>g of RyR2 <strong>in</strong> peripheral<br />

zones of SR was an absolute requirement for the<br />

formation of sites generat<strong>in</strong>g spontaneous Ca sparks coupled<br />

to STOCs <strong>in</strong> arteriolar myocytes (213).<br />

The properties of Ca sparks <strong>in</strong> isolated cells appear<br />

to be relatively similar <strong>in</strong> different types of smooth<br />

muscle cells (for details, see reviews <strong>in</strong> Refs. 536, 750)<br />

and do not differ from those observed <strong>in</strong> <strong>in</strong>tact preparations<br />

(70, 87, 305, 507, 568). Thus, although variations<br />

<strong>in</strong> their biophysical parameters have been noted, Ca<br />

sparks appear to be more stereotypic <strong>in</strong> different types<br />

of smooth muscles than the global Ca signal<strong>in</strong>g controll<strong>in</strong>g<br />

contractility.


3. <strong>Function</strong>al role of Ca sparks<br />

The physiological role of Ca sparks has been a subject<br />

of <strong>in</strong>tense <strong>in</strong>vestigation <strong>in</strong> a variety of smooth muscles.<br />

Calcium sparks are the fundamental units of SR Ca<br />

release dur<strong>in</strong>g E-C coupl<strong>in</strong>g <strong>in</strong> cardiac muscle, synchronized<br />

<strong>in</strong> time by the action potential. Cardiac sparks occur<br />

through the CICR process, and the physical and functional<br />

coupl<strong>in</strong>g between Ca <strong>in</strong>flux and RyR channels is tight and<br />

<strong>in</strong>timate (50, 122, 127). RyR channels are positioned <strong>in</strong> junctional<br />

SR elements with<strong>in</strong> short distances (20 nm) of voltage-dependent<br />

Ca channels <strong>in</strong> the transverse tubules (50,<br />

759). Pharmacologically CICR <strong>in</strong> cardiac muscle can be<br />

easily tested us<strong>in</strong>g ryanod<strong>in</strong>e, or a low concentration of<br />

caffe<strong>in</strong>e. In cardiac muscle, ryanod<strong>in</strong>e produces a strong<br />

negative <strong>in</strong>otropic effect associated with marked <strong>in</strong>hibition<br />

of the global Ca transient, but with no effect on the action<br />

potential. Caffe<strong>in</strong>e produces a transient positive <strong>in</strong>otropic<br />

effect associated with an <strong>in</strong>crease <strong>in</strong> systolic [Ca] with no<br />

change of the Ca current.<br />

Cardiac myocytes express RyR2. The majority of<br />

smooth muscles also express RyR2 and 1C L-type Ca<br />

channels (511), which colocalize with them (490, 525). It<br />

is perhaps not surpris<strong>in</strong>g then that a cardiac-like mechanism<br />

of CICR was expected to be present, act<strong>in</strong>g as an<br />

amplify<strong>in</strong>g system for the global Ca transient, especially<br />

<strong>in</strong> phasic smooth muscle cells. However, it should be<br />

recalled that the highly organized junctional arrangement<br />

between plasma membrane and SR seen <strong>in</strong> striated muscles<br />

does not occur <strong>in</strong> smooth muscles, and junctional<br />

prote<strong>in</strong>s such as junct<strong>in</strong> and triad<strong>in</strong> may be absent (see<br />

sect. IV).<br />

Even more importantly, there are significant differences<br />

<strong>in</strong> the activation by Ca between cardiac and smooth<br />

muscles. In cardiac muscle, global Ca is the determ<strong>in</strong>ant<br />

of contraction under normal physiological conditions.<br />

However, <strong>in</strong> smooth muscle cells, myos<strong>in</strong> light cha<strong>in</strong><br />

phosphorylation catalyzed by MLCK and activated by the<br />

Ca-calmodul<strong>in</strong> complex is a key factor <strong>in</strong> the control of<br />

mechanical activity. Data obta<strong>in</strong>ed <strong>in</strong> some types of<br />

smooth muscle cells show that MLC phosphorylation can<br />

achieve its maximal level at submaximal levels of Ca<br />

(654). Thus, unlike cardiac muscle where high cytoplasmic<br />

[Ca] is required to achieve a high level of force, <strong>in</strong><br />

smooth muscle cells a high level of force can be achieved<br />

at moderate levels of Ca, especially dur<strong>in</strong>g agonist-<strong>in</strong>duced<br />

stimulation, when sensitiz<strong>in</strong>g pathways <strong>in</strong>volv<strong>in</strong>g<br />

PKC or Rho-associated k<strong>in</strong>ase <strong>in</strong>hibition of myos<strong>in</strong> lightcha<strong>in</strong><br />

phosphatase (MLCP) can also be activated (625,<br />

692).<br />

F<strong>in</strong>ally, Ca-activated ion channels are conspicuously<br />

expressed <strong>in</strong> smooth muscle cells but are largely absent<br />

from the heart. Thus, as will be described below, a role for<br />

Ca sparks <strong>in</strong> affect<strong>in</strong>g excitability, as well as modulat<strong>in</strong>g<br />

SMOOTH MUSCLE SR 149<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

Ca signals, has emerged as their prime function <strong>in</strong> many<br />

smooth muscles.<br />

The first work <strong>in</strong> which smooth muscle Ca sparks<br />

were observed (507), <strong>in</strong> myocytes of cerebral arteries,<br />

showed that sparks did not contribute to contraction.<br />

Rather, the authors demonstrated that sparks were <strong>in</strong>volved<br />

<strong>in</strong> relaxation of tone via activation of Ca-activated<br />

large-conductance K channels (BK). The discovery of Ca<br />

sparks provided the miss<strong>in</strong>g l<strong>in</strong>k <strong>in</strong> expla<strong>in</strong><strong>in</strong>g the orig<strong>in</strong><br />

and functional role of STOCs, which had been observed<br />

earlier <strong>in</strong> a number of smooth muscle cells (41). The role<br />

of a Ca sparks/STOCs coupl<strong>in</strong>g mechanism and its functional<br />

role <strong>in</strong> tonic smooth muscles has now been thoroughly<br />

<strong>in</strong>vestigated (507), and well reviewed (304, 750).<br />

In contrast, the role of Ca sparks <strong>in</strong> phasic smooth<br />

muscles, particularly their possible role <strong>in</strong> amplification<br />

of the global Ca signal via CICR, is still under <strong>in</strong>vestigation,<br />

and there is no consensus on this issue. It appears to<br />

us that different types of smooth muscles utilize Ca<br />

sparks <strong>in</strong> a variety of ways to match their physiological<br />

function. Unlike cardiac muscle, smooth muscle cells express<br />

Ca-activated K and Cl channels, which can be targeted<br />

by Ca sparks, and <strong>in</strong> this way control excitability.<br />

4. Ca sparks <strong>in</strong> phasic smooth muscles<br />

In cardiac myocytes, each action potential results <strong>in</strong><br />

a contraction that derives from RyR-mediated calcium<br />

release, triggered by an <strong>in</strong>crease <strong>in</strong> [Ca] due to <strong>in</strong>flux via<br />

L-type Ca channels. The signal ga<strong>in</strong> is quite high, s<strong>in</strong>ce<br />

each channel open<strong>in</strong>g results <strong>in</strong> a Ca spark (activation of<br />

several RyRs), the duration of which is longer than the<br />

L-type Ca channel open<strong>in</strong>g (104). Phasic smooth muscles<br />

are also activated by Ca transients controlled by the action<br />

potential. However, <strong>in</strong>formation on the Ca signal <strong>in</strong><br />

the cytosol follow<strong>in</strong>g the action potential is limited, even<br />

though it is crucial to the understand<strong>in</strong>g of the coupl<strong>in</strong>g of<br />

membrane excitation to Ca-sensitive cellular functions.<br />

Thus, despite the broad expression of L-type Ca channels<br />

and RyR <strong>in</strong> many phasic smooth muscles, the existence<br />

and nature of CICR <strong>in</strong> these nonsarcomeric cells, <strong>in</strong> which<br />

the distribution of L-type Ca channels and RyR differs<br />

substantially from an orderly dyadic pattern <strong>in</strong> sarcomeric<br />

muscles, is not well established.<br />

Simultaneous record<strong>in</strong>g of electrical activity and <strong>in</strong>tracellular<br />

[Ca] have been performed <strong>in</strong> some <strong>in</strong>tact phasic<br />

smooth muscles, e.g., gu<strong>in</strong>ea pig ureter (87), pregnant<br />

rat myometrium (88), and ur<strong>in</strong>ary bladder (288) and isolated<br />

cells from gu<strong>in</strong>ea pig ileum (247, 248, 254), gu<strong>in</strong>ea<br />

pig vas deferens (361), and mouse ur<strong>in</strong>ary bladder (494).<br />

In ur<strong>in</strong>ary bladder, vas deferens, uterus, and ileum, the<br />

action potential usually appears as brief (20–40 ms)<br />

spikes, while <strong>in</strong> ureter it consists of a spike followed by a<br />

long-last<strong>in</strong>g (400–800 ms) plateau component. In all these<br />

four smooth muscles, the Ca transient is triggered by the


150 SUSAN WRAY AND THEODOR BURDYGA<br />

action potential, and both are <strong>in</strong>hibited by L-type Ca<br />

channel blockers, <strong>in</strong>dicat<strong>in</strong>g that Ca <strong>in</strong>flux is a key factor<br />

<strong>in</strong> the generation of the global Ca transients. Is CICR<br />

contribut<strong>in</strong>g to the global Ca transients associated with<br />

the action potential <strong>in</strong> smooth muscles?<br />

In the gu<strong>in</strong>ea pig ileum where Ca sparks have been<br />

observed (220), ryanod<strong>in</strong>e and thapsigarg<strong>in</strong> produced<br />

stimulant rather than <strong>in</strong>hibitory effects on the Ca transients<br />

associated with the action potential by <strong>in</strong>creas<strong>in</strong>g<br />

the amplitude and duration of the spike (203, 288, 312,<br />

494). Voltage-clamp experiments performed on ileum<br />

(361), cerebral arteries (330), portal ve<strong>in</strong> (133), vas deferens<br />

and bladder (288), and pregnant myometrium (630)<br />

demonstrated that Ca entry via L-type Ca channels can<br />

trigger some Ca release via RyRs. At the same time, other<br />

workers have failed to detect CICR <strong>in</strong> voltage-clamped<br />

smooth muscle cells isolated from other or even the same<br />

type of smooth muscles. For example, CICR was not<br />

detected <strong>in</strong> voltage-clamped smooth muscle cells of portal<br />

ve<strong>in</strong> (329), gastro<strong>in</strong>test<strong>in</strong>al tract (79, 781), and airways<br />

(178).<br />

A discussion of the physiological role of CICR <strong>in</strong><br />

ur<strong>in</strong>ary bladder smooth muscle cells illustrates the complexity<br />

<strong>in</strong> this area and the data which are hard to unify.<br />

Elegant work performed on voltage-clamped gu<strong>in</strong>ea pig<br />

ur<strong>in</strong>ary bladder myocytes by Ganitkevich and Isenberg<br />

(203) showed that Ca transients <strong>in</strong>duced by long depolariz<strong>in</strong>g<br />

voltage steps were <strong>in</strong>hibited 70% by ryanod<strong>in</strong>e.<br />

This suggested that a relatively high ga<strong>in</strong> “cardiac type” of<br />

CICR operates <strong>in</strong> these myocytes, at least under these<br />

experimental conditions. The importance of a functional<br />

contribution of CICR to E-C coupl<strong>in</strong>g and contraction <strong>in</strong><br />

ur<strong>in</strong>ary bladder smooth muscles has been assessed by<br />

several research groups, us<strong>in</strong>g different species and experimental<br />

models (127, 247, 254, 490, 494, 525). Results<br />

from these studies address<strong>in</strong>g the susceptibility to contraction<br />

<strong>in</strong> bladder myocytes treated with ryanod<strong>in</strong>e are<br />

not <strong>in</strong> agreement. For example, Hashitani and Brad<strong>in</strong>g<br />

(247) showed the <strong>in</strong>tracellular [Ca] rise <strong>in</strong>duced by spontaneous<br />

action potentials <strong>in</strong> the gu<strong>in</strong>ea pig was reduced to<br />

60% by ryanod<strong>in</strong>e. In a later publication (494), the stimulant<br />

action of ryanod<strong>in</strong>e on the mechanical activity of the<br />

gu<strong>in</strong>ea pig bladder was reported. Reduction of spontaneous<br />

or electrically evoked contractions by ryanod<strong>in</strong>e have<br />

been reported for mouse bladder myocytes (494). However,<br />

ryanod<strong>in</strong>e has no significant effect on the [Ca] and<br />

spontaneous contractions <strong>in</strong> rat bladder myocytes (252).<br />

These data suggest that the contribution of RyRs and<br />

CICR to E-C coupl<strong>in</strong>g <strong>in</strong> the bladder differs between<br />

species and experimental conditions.<br />

Imaizumi et al. (288) <strong>in</strong> gu<strong>in</strong>ea pig ur<strong>in</strong>ary bladder<br />

myocytes us<strong>in</strong>g brief (50 ms) depolariz<strong>in</strong>g voltage steps,<br />

obta<strong>in</strong>ed the first direct evidence that Ca “hot spots”<br />

(equivalent to Ca sparks) could be transiently activated <strong>in</strong><br />

cells depolarized to 20 mV. These events lasted for 0.5<br />

s and spread as Ca waves when depolarization was to 0<br />

mV. The <strong>in</strong>crease <strong>in</strong> global [Ca] elicited by 50 ms depolarization<br />

to 0 mV reached a peak 60–100 ms after the<br />

start of depolarization, <strong>in</strong>dicat<strong>in</strong>g that the spread of Ca<br />

hot spots cont<strong>in</strong>ued even after most voltage-dependent<br />

Ca channels had closed. Recently, Kotlikoff and co-workers<br />

(127) have also <strong>in</strong>vestigated the temporal relationship<br />

between the Ca current and local and global Ca signal<strong>in</strong>g<br />

<strong>in</strong> rabbit ur<strong>in</strong>ary bladder myocytes, under voltage- and<br />

current-clamp conditions with physiologically relevant<br />

protocols of stimulation. Localized photolysis of caged Ca<br />

<strong>in</strong>itiated CICR, but multiple excitation pulses were<br />

needed and IP 3R channels were also activated (313). Calcium<br />

channels were shown to activate RyRs to produce<br />

CICR <strong>in</strong> the form of Ca sparks and propagated Ca waves.<br />

Both the <strong>in</strong>itial Ca spark and the subsequent Ca waves<br />

occurred through the open<strong>in</strong>g of RyRs, s<strong>in</strong>ce both were<br />

elim<strong>in</strong>ated by ryanod<strong>in</strong>e, and neither was affected by<br />

dialysis with hepar<strong>in</strong> (127). They concluded that 1) L-type<br />

Ca channels could open without trigger<strong>in</strong>g Ca sparks,<br />

2) triggered Ca sparks could be observed after channel<br />

closure, 3) the trigger stimulus for the Ca release process<br />

is not a local but a global rise <strong>in</strong> <strong>in</strong>tracellular [Ca], and<br />

4) this results <strong>in</strong> a functional uncoupl<strong>in</strong>g of a s<strong>in</strong>gle action<br />

potential from Ca release.<br />

5. Loose coupl<strong>in</strong>g<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

The small number of Ca spark sites evoked by the Ca<br />

current, the delay between Ca channel and RyR channel<br />

open<strong>in</strong>gs, as well as the <strong>in</strong>hibitory effect of BAPTA on Ca<br />

sparks <strong>in</strong> ur<strong>in</strong>ary bladder smooth muscle cells suggested<br />

that compared with cardiac myocytes, a fundamentally<br />

different coupl<strong>in</strong>g process operates <strong>in</strong> smooth muscle,<br />

which was termed “loose coupl<strong>in</strong>g” (127, 368). Features of<br />

the loose coupl<strong>in</strong>g system are low ga<strong>in</strong> (multiple Ca channels<br />

must open to produce sparks), discrim<strong>in</strong>ated responses<br />

(release takes the form of local Ca sparks or<br />

globally propagated Ca waves), and a marked lengthen<strong>in</strong>g<br />

of signal duration (Ca waves last far longer than the<br />

action potential). The delayed activation of CICR and the<br />

subsequent propagation of a Ca wave, which could last up<br />

to 1 s, suggests that CICR <strong>in</strong> contrast to cardiac muscle<br />

(122, 127) cannot be controlled by a brief (40–60 ms)<br />

s<strong>in</strong>gle action potential, and its functional role is yet to be<br />

established (368). Thus, <strong>in</strong> ur<strong>in</strong>ary bladder myocytes paradoxically,<br />

global rises of [Ca] are required to activate Ca<br />

sparks. The concept of loose coupl<strong>in</strong>g can also be expla<strong>in</strong>ed<br />

by an <strong>in</strong>crease <strong>in</strong> the SR [Ca] and activation of<br />

[Ca] sparks. An <strong>in</strong>crease <strong>in</strong> the lum<strong>in</strong>al [Ca] produces<br />

profound effects on both the frequency and the amplitude<br />

of Ca sparks <strong>in</strong> smooth muscle (811).<br />

Both cytosolic and SR Ca levels would be expected to<br />

<strong>in</strong>crease with enhanced Ca entry. The proximity of the SR<br />

to the plasma membrane and the existence of the uptake


mechanisms by the SERCA pump provide the structure<br />

for what has been termed by van Breemen the “superficial<br />

buffer barrier” (714), discussed also <strong>in</strong> section II. S<strong>in</strong>ce<br />

<strong>in</strong>hibition of SERCA has little effect on the global rise of<br />

[Ca] <strong>in</strong>duced by depolariz<strong>in</strong>g voltage steps (e.g., Refs. 77,<br />

507), it can be suggested that only a small portion of the<br />

SR is <strong>in</strong>volved <strong>in</strong> active ATP-dependent accumulation of<br />

Ca, which does not affect the global rise of [Ca]. Similarly,<br />

many myocytes have FDS which, when releas<strong>in</strong>g Ca <strong>in</strong> the<br />

form of Ca sparks, have little effect on the global rise of<br />

[Ca] (330). However, this suggestion should be experimentally<br />

tested by study<strong>in</strong>g the spatial distribution of<br />

SERCA pumps <strong>in</strong> smooth muscle cells. S<strong>in</strong>ce the effects<br />

of ryanod<strong>in</strong>e on the gu<strong>in</strong>ea pig ur<strong>in</strong>ary bladder were mimicked<br />

by CPA, the BK channel <strong>in</strong>hibitor iberiotox<strong>in</strong>, and<br />

BAPTA (247), it was suggested that CICR targets BK<br />

channels via Ca sparks and <strong>in</strong> this manner controls excitability<br />

of this smooth muscle.<br />

Another well-studied smooth muscle with respect to<br />

CICR and the role of Ca sparks is the gu<strong>in</strong>ea pig ureter. In<br />

contrast to bladder smooth muscles, Ca transients and<br />

phasic contraction <strong>in</strong> the gu<strong>in</strong>ea pig ureter are controlled<br />

by a s<strong>in</strong>gle long-last<strong>in</strong>g (400–800 ms) plateau-type action<br />

potential of myogenic orig<strong>in</strong>, which gives rise to a longlast<strong>in</strong>g<br />

Ca transient (76, 87). Parameters of the Ca transients<br />

are tightly coupled to the parameters of the action<br />

potentials (76, 87, 93) and thus meet the criteria of both<br />

tight and loose coupl<strong>in</strong>g. Early contractile studies <strong>in</strong> <strong>in</strong>tact<br />

gu<strong>in</strong>ea pig ureteric smooth muscle preparations identified<br />

ryanod<strong>in</strong>e-sensitive calcium release by caffe<strong>in</strong>e (93,<br />

287) but not agonists (89, 91, 92), and thus these myocytes<br />

serve as an excellent experimental model to <strong>in</strong>vestigate<br />

the role of CICR <strong>in</strong> phasic smooth muscles. The STOCS,<br />

associated with spontaneous transient hyperpolarizations<br />

and sensitive to caffe<strong>in</strong>e, were recorded <strong>in</strong> myocytes under<br />

voltage- and current-clamp conditions (91, 92). We<br />

observed Ca sparks aris<strong>in</strong>g from several FDS <strong>in</strong> isolated<br />

cells and <strong>in</strong>tact preparations (87). In gu<strong>in</strong>ea pig ureteric<br />

smooth muscle cells, low caffe<strong>in</strong>e concentration <strong>in</strong>creased<br />

the frequency of Ca sparks and <strong>in</strong>hibited the<br />

action potential, Ca transients, and phasic contractions.<br />

These components of E-C coupl<strong>in</strong>g were restored by ryanod<strong>in</strong>e,<br />

CPA or TEA, and iberiotox<strong>in</strong> (blockers of BK<br />

channels) (76). Under voltage-clamp conditions, caffe<strong>in</strong>e<br />

at low concentrations had no <strong>in</strong>hibitory effect on the Ca<br />

current and slightly potentiated global rises of [Ca], suggest<strong>in</strong>g<br />

some contribution of CICR to the generation of<br />

depolarization-evoked <strong>in</strong>creases <strong>in</strong> global [Ca]. However,<br />

caffe<strong>in</strong>e markedly <strong>in</strong>creased the frequency and amplitude<br />

of STOCs and <strong>in</strong>creased the overlap between Ca and BK<br />

channel currents when voltage-clamped ureteric myocytes<br />

were depolarized to 0 mV (76). Caffe<strong>in</strong>e had no<br />

effect on the action potential and Ca transients <strong>in</strong> the<br />

presence of TEA, aga<strong>in</strong> suggest<strong>in</strong>g that CICR had little<br />

contribution to global rises of [Ca] controlled by the<br />

SMOOTH MUSCLE SR 151<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

action potential. The lack of effect of ryanod<strong>in</strong>e and CPA<br />

on the Ca transients and the reversal of the <strong>in</strong>hibitory<br />

effects of caffe<strong>in</strong>e by these agents suggest that the contribution<br />

of CICR to global Ca signal<strong>in</strong>g is m<strong>in</strong>imal, and<br />

the major source of Ca is action potential-stimulated Ca<br />

entry.<br />

In the gu<strong>in</strong>ea pig ureter, the duration of the plateau<br />

component of the action potential is affected by <strong>in</strong>ward<br />

Ca current (I Ca) and outward BK (I BK) currents (287, 382).<br />

Normally there is a substantial delay between I Ca and<br />

STOCs <strong>in</strong> ureteric myocytes (287, 382), which enables the<br />

generation of the long-last<strong>in</strong>g plateau. However, if this<br />

delay is shortened or elim<strong>in</strong>ated, as is the case with<br />

caffe<strong>in</strong>e, the action potential is blocked or markedly reduced<br />

<strong>in</strong> duration lead<strong>in</strong>g to the <strong>in</strong>hibition or reduction <strong>in</strong><br />

the duration of the Ca transient. Thus these data fit the<br />

hypothesis of loose coupl<strong>in</strong>g between I Ca and CICR. This<br />

mechanism is an important factor <strong>in</strong> controll<strong>in</strong>g the duration<br />

of the plateau component of the action potential,<br />

which <strong>in</strong> turn plays a key role <strong>in</strong> controll<strong>in</strong>g the amplitude<br />

of phasic contractions (93). A decrease or elim<strong>in</strong>ation of<br />

the delay between I Ca and STOCs, and a slight <strong>in</strong>crease <strong>in</strong><br />

I Ca-<strong>in</strong>dependent amplitude of the global Ca transient occur<br />

under voltage-clamp conditions <strong>in</strong> the presence of<br />

caffe<strong>in</strong>e, suggest<strong>in</strong>g that I Ca can be tightly coupled to Ca<br />

sparks. The mechanism of loose coupl<strong>in</strong>g between I Ca and<br />

STOCs (Ca sparks) <strong>in</strong> gu<strong>in</strong>ea pig ureteric cells can be<br />

expla<strong>in</strong>ed by a prom<strong>in</strong>ent role of lum<strong>in</strong>al Ca <strong>in</strong> the activation<br />

of Ca sparks/STOCs coupl<strong>in</strong>g mechanism. Indeed,<br />

we and others have recently shown that the Ca content of<br />

the SR follow<strong>in</strong>g a global Ca signal is substantially <strong>in</strong>creased<br />

(77, 633), and this was well correlated with a<br />

long-last<strong>in</strong>g discharge of Ca sparks after L-type Ca channels<br />

were closed (87, 93).<br />

The Ca content of the SR was also shown to play a<br />

critical role <strong>in</strong> the generation of spontaneous Ca sparks<br />

and STOCs <strong>in</strong> smooth muscle of the gastro<strong>in</strong>test<strong>in</strong>al tract<br />

(811). We have established that the transient eruption of<br />

Ca sparks plays a key role <strong>in</strong> the control of the longlast<strong>in</strong>g<br />

period of refractor<strong>in</strong>ess, which serves as an important<br />

pac<strong>in</strong>g and protect<strong>in</strong>g mechanism <strong>in</strong> the ureter<br />

(87). In the gu<strong>in</strong>ea pig bladder, the repolariz<strong>in</strong>g phase of<br />

the action potential, as well as the frequency of spike<br />

discharge <strong>in</strong> one burst, are also potentiated by <strong>in</strong>hibitors<br />

of the SR or BK channels (247, 248, 254, 256). These data<br />

would also fit a loose coupl<strong>in</strong>g between I Ca and STOCs,<br />

act<strong>in</strong>g as an important mechanism controll<strong>in</strong>g the parameters<br />

of the action potential and excitability of these types<br />

of smooth muscles, as observed by Collier et al. (127).<br />

To summarize these data, we conclude that the large<br />

contribution of BK channels activated by superficial CICR<br />

to the negative feedback of membrane excitability and/or<br />

the regulation of action potential shape, is well-established<br />

and a characteristic of many types of smooth muscles.<br />

The question of a possible role of CICR as a contrib-


152 SUSAN WRAY AND THEODOR BURDYGA<br />

utor to the global Ca signal is still a matter for future<br />

experiments. That <strong>in</strong>hibition of SR function by SERCA<br />

<strong>in</strong>hibitors results <strong>in</strong> 70% <strong>in</strong>hibition of the [Ca] rises <strong>in</strong><br />

the area of discharge sites <strong>in</strong> ur<strong>in</strong>ary bladder and vas<br />

deferens strongly suggests that CICR is present <strong>in</strong> these<br />

tissues and can produce a significant contribution to the<br />

cytoplasmic [Ca] rise. However, as under the same conditions,<br />

that global rises of [Ca] were not <strong>in</strong>hibited, but <strong>in</strong><br />

fact were slightly potentiated, as also reported for a number<br />

of <strong>in</strong>tact phasic smooth muscles, suggests that CICR<br />

is conf<strong>in</strong>ed to microdoma<strong>in</strong>s. Studies us<strong>in</strong>g other types of<br />

smooth muscles and different species should be carried<br />

out. High temporal and spatial resolution and analyses of<br />

the Ca release from the SR should be correlated with the<br />

isoform expression and spatial distribution of different<br />

types of RyRs.<br />

6. Ca sparks and myogenic tone<br />

Many resistance arteries possess an <strong>in</strong>tr<strong>in</strong>sic “myogenic”<br />

tone, activated <strong>in</strong> response to elevation of <strong>in</strong>travascular<br />

pressure by a graded membrane depolarization,<br />

open<strong>in</strong>g Ca channels, and lead<strong>in</strong>g to elevation <strong>in</strong> [Ca] and<br />

constriction (507). It has been proposed that Ca sparks,<br />

by target<strong>in</strong>g BK channels, generate STOCs, which cause a<br />

tonic hyperpolariz<strong>in</strong>g <strong>in</strong>fluence on the membrane potential<br />

of small pressurized arteries with myogenic tone<br />

(507). Inhibition of Ca sparks or BK channels causes<br />

membrane depolarization, lead<strong>in</strong>g to further elevation of<br />

[Ca], and additional <strong>in</strong>creases <strong>in</strong> muscle tone <strong>in</strong> pressurized<br />

cerebral arteries (330, 356, 507). Thus, <strong>in</strong> resistance<br />

arteries, a Ca sparks/STOCs coupl<strong>in</strong>g mechanism can act<br />

as a negative-feedback mechanism to oppose myogenic<br />

Ca entry via Ca channels and promote relaxation (507).<br />

However, under voltage-clamp conditions, global rises of<br />

[Ca] <strong>in</strong>duced by depolariz<strong>in</strong>g voltage step <strong>in</strong> myocytes<br />

isolated from these arteries were significantly <strong>in</strong>hibited by<br />

ryanod<strong>in</strong>e. This suggested that CICR is also present <strong>in</strong><br />

these cells and can act as an amplify<strong>in</strong>g mechanism (507).<br />

However, under normal physiological conditions, this<br />

mechanism may contribute little, as block<strong>in</strong>g Ca sparks <strong>in</strong><br />

the presence of blockers of BK channels has little effect<br />

on global [Ca] and arterial tone (330). Myogenic tone is<br />

associated with moderate depolarization (356, 357, 507),<br />

and it was suggested that L-type Ca channels operate <strong>in</strong> a<br />

“low-activity mode,” i.e., the myocytes never depolarize<br />

sufficiently to achieve a high channel P o.<br />

We conclude that the Ca sparks/STOCs coupl<strong>in</strong>g<br />

mechanism acts as a powerful vasodilator <strong>in</strong> vascular<br />

beds where membrane potential depolarization acts as an<br />

activat<strong>in</strong>g mechanism to generate tone. If muscle tone is<br />

activated via stimulation of Ca entry by SOC or ROC<br />

channels, this mechanism will be <strong>in</strong>effective, as it will be<br />

<strong>in</strong> controll<strong>in</strong>g tone produced by the Ca oscillations associated<br />

with activation of IP 3Rs. However, this mechanism<br />

can play an important role <strong>in</strong> disabl<strong>in</strong>g the mechanisms of<br />

generation of the action potential, which is a feature of<br />

most of the arteriolar smooth muscle cells and enables<br />

them to be regulated by local factors.<br />

B. Ca Puffs<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

The term Ca puffs refers to the small local <strong>in</strong>creases<br />

<strong>in</strong> [Ca] that occur when IP 3Rs open spontaneously or <strong>in</strong><br />

response to agonists. Unlike sparks, Ca puffs have not<br />

been detected <strong>in</strong> vascular myocytes despite the obvious<br />

presence of IP 3Rs. So far, Ca puffs have been detected <strong>in</strong><br />

only two smooth muscles: rat ureter (60) <strong>in</strong> which agonist-<strong>in</strong>duced<br />

Ca release plays a dom<strong>in</strong>ant role <strong>in</strong> modulat<strong>in</strong>g<br />

contraction (34, 86) and mur<strong>in</strong>e colonic myocytes. In<br />

rat ureteric myocytes, Ca puffs were observed dur<strong>in</strong>g<br />

release of low concentrations of IP 3 from a caged precursor,<br />

or by low concentrations of ACh. They were also<br />

observed spontaneously <strong>in</strong> Ca-overloaded myocytes (91).<br />

Spontaneous Ca puffs were observed <strong>in</strong> <strong>in</strong>tact rat ureteric<br />

preparations (60).<br />

Calcium puffs <strong>in</strong> tissues appear to have a wide<br />

range of amplitudes, time courses, and spatial spread,<br />

suggest<strong>in</strong>g that the IP 3Rs exist <strong>in</strong> clusters of variable<br />

numbers of channels and that with<strong>in</strong> these clusters a<br />

variable number of channels can be recruited (86).<br />

Calcium puffs <strong>in</strong> the ureteric myocytes were blocked<br />

selectively by <strong>in</strong>tracellular applications of hepar<strong>in</strong> and<br />

an antibody to IP 3R, but were unaffected by ryanod<strong>in</strong>e<br />

and <strong>in</strong>tracellular application of an antibody to RyR (60).<br />

When stimulated by agonists, propagated, ryanod<strong>in</strong>eresistant<br />

Ca waves appeared to result from the spatial<br />

recruitment of Ca-release sites by diffusion. This is<br />

consistent with data from multicellular preparations,<br />

where ryanod<strong>in</strong>e-resistant agonist-<strong>in</strong>duced Ca release<br />

was reported (34). Both Ca signals provide an <strong>in</strong>tegrated<br />

mechanism to regulate contractility <strong>in</strong> smooth<br />

muscle cells, where RyRs are absent, or poorly expressed.<br />

The possible role of Ca puffs <strong>in</strong> the control of excitability of<br />

rat ureteric smooth muscle cells has not been <strong>in</strong>vestigated,<br />

although we have observed spontaneous transient outward/<br />

<strong>in</strong>ward currents (STOICs) (Burdyga and Wray, unpublished<br />

data), which could result from activation of the BK and Cl Ca<br />

channels (91, 92).<br />

Localized Ca transients and puffs, resistant to ryanod<strong>in</strong>e<br />

and <strong>in</strong>hibited by xestospong<strong>in</strong>, U-73122 (an <strong>in</strong>hibitor<br />

of PLC), occurred spontaneously or dur<strong>in</strong>g P2Y receptor<br />

stimulation <strong>in</strong> mur<strong>in</strong>e colonic myocytes (34). The<br />

puffs were coupled to the activation of both BK and<br />

small-conductance Ca-activated K (SK) channels. Thus<br />

the release of Ca by G prote<strong>in</strong>-mediated activation of PLC<br />

can be l<strong>in</strong>ked to <strong>in</strong>hibitory responses via Ca puffs target<strong>in</strong>g<br />

SK channels. This is <strong>in</strong> marked contrast to the usual<br />

f<strong>in</strong>d<strong>in</strong>g that IP 3-dependent mechanisms are used by excitatory<br />

agonists <strong>in</strong> smooth muscles.


A role for IP 3Rs <strong>in</strong> the generation of Ca oscillations<br />

and waves seems to be a common feature of smooth<br />

muscles. The absence of local Ca signals, i.e., Ca puffs, is<br />

therefore surpris<strong>in</strong>g. The paucity of Ca puffs might be<br />

expla<strong>in</strong>ed by there be<strong>in</strong>g little or no cluster<strong>in</strong>g of the<br />

receptors <strong>in</strong> most smooth muscles. It may also be that Ca<br />

puffs have not been sought. Whether puffs are present <strong>in</strong><br />

other smooth muscles and whether they can contribute to<br />

the control of excitability seems to us to be an important<br />

question that should be urgently addressed.<br />

X. SARCOPLASMIC RETICULUM AND<br />

PLASMALEMMAL ION CHANNELS<br />

A. Overview of Ca-Activated Ion Channels<br />

In contrast to skeletal and cardiac muscle, smooth<br />

muscle cells express several types of Ca activated ion<br />

channels on the plasmalemma that can be differentially<br />

activated by SR Ca release events, such as Ca sparks, Ca<br />

puffs, and Ca waves. There are two major populations of<br />

Ca-activated channels that can be activated by SR Ca<br />

release <strong>in</strong> different types of smooth muscles: Ca-activated<br />

K (K Ca) and Ca-activated Cl (Cl Ca) channels. The K Ca<br />

channels are composed of at least three different members,<br />

which are separate molecular entities. Their different<br />

K conductances have led to their be<strong>in</strong>g known as big<br />

(BK), <strong>in</strong>termediate (IK), or small (SK) K channels. These<br />

channels also exhibit different <strong>in</strong>hibition profiles to various<br />

tox<strong>in</strong>s. (The BK channel is also referred to as the<br />

maxi K Ca.) Coupl<strong>in</strong>g between SR-mediated Ca events and<br />

these Ca activated ion channels can lead to changes <strong>in</strong><br />

membrane potential and/or the action potential and thus<br />

serve as powerful negative- or positive-feedback mechanisms,<br />

controll<strong>in</strong>g excitability and contractility of smooth<br />

muscles. Ca sparks can activate either or both of these<br />

Ca-activated channels produc<strong>in</strong>g STOCs (507), spontaneous<br />

transient <strong>in</strong>ward currents (STICs), or STOICs (811).<br />

Spontaneous transient membrane hyperpolarizations<br />

have also been reported <strong>in</strong> coronary artery (202) and<br />

gu<strong>in</strong>ea pig ureter (87) and spontaneous transient membrane<br />

depolarizations <strong>in</strong> urethral (249, 287) smooth muscles,<br />

confirm<strong>in</strong>g that STOCs and STICs can affect the<br />

membrane potential of smooth muscles. A Ca spark/<br />

STOCs coupl<strong>in</strong>g mechanism can also directly affect the<br />

parameters of the action potential by <strong>in</strong>creas<strong>in</strong>g the repolariz<strong>in</strong>g<br />

current and thus decreas<strong>in</strong>g the amplitude and<br />

duration of the action potential, as was shown for ur<strong>in</strong>ary<br />

bladder (247, 248, 254, 256, 288) and ureter (474). Calcium<br />

waves may also <strong>in</strong>directly <strong>in</strong>fluence excitability and contractility<br />

of smooth muscles through activation of Cadependent<br />

ion channels. In rat portal ve<strong>in</strong> myocytes, Ca<br />

sparks were demonstrated to activate STOCs, while Ca<br />

waves selectively activated Cl Ca channels (474). Interest-<br />

SMOOTH MUSCLE SR 153<br />

<strong>in</strong>gly, STICs but not STOCs were selectively blocked by<br />

<strong>in</strong>hibition of IP 3Rs (623), suggest<strong>in</strong>g that the variability <strong>in</strong><br />

Ca release events can also modulate the cell membrane<br />

potential through differential target<strong>in</strong>g of K Ca and Cl Ca<br />

channels.<br />

B. BK Channels<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

Transient outward K currents were first described <strong>in</strong><br />

smooth muscle cells by Benham and Bolton (1986), who<br />

used the term STOC to describe these events, which were<br />

later identified <strong>in</strong> a wide variety of smooth muscle cells<br />

(41, 70, 87, 150, 264, 277, 287, 308, 332, 382, 495, 507, 516,<br />

528, 568, 604, 617, 806, 810, 811). The native BK channels<br />

consist of four identical subunits that create a functional<br />

BK channel and four modulatory subunits which<br />

comb<strong>in</strong>e with the channel form<strong>in</strong>g subunits and affect<br />

their sensitivity to Ca (83, 93, 459, 514, 712). Immunocytochemical<br />

observations us<strong>in</strong>g antibodies aga<strong>in</strong>st the BK<br />

channel subunit, Ca channel 1C subunit and RyR,<br />

showed that the BK and RyR but not the Ca channel form<br />

clusters <strong>in</strong> myocytes (525). The BK channels have an<br />

estimated s<strong>in</strong>gle-channel conductance of 80 pS at 40<br />

mV with a physiological K concentration gradient and<br />

occur at a density of 1–4 channels/m 2 (41, 641, 689).<br />

Direct evidence for the causal relationship between Ca<br />

sparks and transient BK currents has been supplied by<br />

studies comb<strong>in</strong><strong>in</strong>g whole cell patch clamp<strong>in</strong>g with simultaneous<br />

confocal imag<strong>in</strong>g of Ca <strong>in</strong> voltage-clamped myocytes<br />

(83, 150, 302, 352, 474, 555, 749, 810). These experiments<br />

revealed good correlations between the temporal<br />

characteristics of Ca sparks and STOCs, although BK<br />

currents showed a faster decay relative to the Ca spark.<br />

These data are <strong>in</strong> good agreement with immunohistochemical<br />

observations that showed the close proximity of<br />

Ca spark-generat<strong>in</strong>g sites and BK channels (525).<br />

A Ca spark/STOC coupl<strong>in</strong>g mechanism is present <strong>in</strong><br />

many visceral and vascular smooth muscles and acts as a<br />

powerful negative-feedback mechanism, protect<strong>in</strong>g smooth<br />

muscle cells from overexcitability. Inhibition of Ca sparks<br />

with ryanod<strong>in</strong>e or SERCA pump <strong>in</strong>hibitors or their target BK<br />

channels (by low concentration of TEA or iberiotox<strong>in</strong>) results<br />

<strong>in</strong> augmentation of vascular tone (507) and an <strong>in</strong>crease<br />

<strong>in</strong> the excitability and term<strong>in</strong>ation of the refractory period <strong>in</strong><br />

phasic smooth muscles (87, 247). The importance of BK<br />

channels has been demonstrated with transgenic mice. Targeted<br />

deletion of regulatory 1-subunits of BK channels<br />

resulted <strong>in</strong> animals that developed an <strong>in</strong>crease <strong>in</strong> vascular<br />

tone and hypertension (618) as well as other pathologies<br />

such as ataxia (619), ur<strong>in</strong>ary <strong>in</strong>cont<strong>in</strong>ence (465), and erectile<br />

dysfunction (752). BK channels have a relatively low<br />

aff<strong>in</strong>ity for Ca. For example, at a physiological membrane<br />

potential of 40 mV, the K D of BK channels for Ca is 20<br />

M (554). Calcium reaches high levels (10–100 M) close


154 SUSAN WRAY AND THEODOR BURDYGA<br />

to the SR release site (505, 555), and the plasma membrane<br />

is <strong>in</strong> close proximity; thus BK channels are activated<br />

despite their low aff<strong>in</strong>ity for Ca. A s<strong>in</strong>gle Ca spark<br />

can produce 20 mV hyperpolarization <strong>in</strong> an isolated<br />

myocyte through activation of BK channels (202, 507).<br />

Vascular myocytes have a relatively high density of BK<br />

channels and do not require cluster<strong>in</strong>g of BK channels<br />

above a spark site. However, <strong>in</strong> visceral smooth muscle<br />

cells, where the density of BK channels is low, cluster<strong>in</strong>g<br />

of BK channels <strong>in</strong> the patches of the plasma membrane at<br />

sites frequently discharg<strong>in</strong>g sparks is required (525, 809).<br />

C. SK and IK Channels<br />

There are four different isoforms of the SK channel,<br />

SK1-4 (480), with SK4 also known as IK1. The clon<strong>in</strong>g of<br />

SK channels revealed the existence of at least three members<br />

of a family (SK1-3) (362), which were first identified<br />

<strong>in</strong> cultured rat skeletal muscle (55). These channels are<br />

highly sensitive to Ca (EC 50 0.5–0.7 M) and have a<br />

small s<strong>in</strong>gle-channel conductance (4–14 pS). In addition,<br />

they are selectively blocked by the bee venom tox<strong>in</strong><br />

apam<strong>in</strong> (135, 190, 247, 250, 254, 256). Of the three cloned<br />

SK channel subtypes (SK1-3), SK1 and SK3 channels are<br />

least sensitive to apam<strong>in</strong> (nanomolar apam<strong>in</strong> was required<br />

for their <strong>in</strong>hibition), and SK2 channels were more<br />

sensitive and <strong>in</strong>hibited <strong>in</strong> the subnanomolar range (67,<br />

362, 626, 727).<br />

S<strong>in</strong>ce their <strong>in</strong>itial discovery, SK channels have been<br />

described <strong>in</strong> a number of cell types, <strong>in</strong>clud<strong>in</strong>g rat pituitary<br />

cells (381), T lymphocytes (227), and adrenal chromaff<strong>in</strong><br />

cells (543). They have also been identified <strong>in</strong> some types<br />

of smooth muscles, e.g., ur<strong>in</strong>ary bladder (698), stomach<br />

(673), colon (364), uterus (360, 450), renal artery (206),<br />

portal ve<strong>in</strong> (293), and the pancreatic arterioles (666).<br />

Their distribution and functional importance are currently<br />

be<strong>in</strong>g established (247, 248, 256, 663), but there is<br />

evidence that SK3 is particularly relevant to contractility<br />

and excitability <strong>in</strong> smooth muscle. <strong>Function</strong>al studies performed<br />

on the <strong>in</strong>tact bladder revealed SK channels contribute<br />

to the afterhyperpolarization (AHP) follow<strong>in</strong>g<br />

tra<strong>in</strong>s of action potentials (255), as apam<strong>in</strong> blocked the<br />

AHP and <strong>in</strong>creased excitability and contractility without<br />

affect<strong>in</strong>g other parameters of the action potential.<br />

There is evidence that both SR Ca release and plasmalemmal<br />

Ca entry can activate SK channels. Hererra and<br />

Nelson (256) reported that SK channels <strong>in</strong> the bladder are<br />

activated by the rise <strong>in</strong> [Ca] which occurs upon Ca entry<br />

through L-type Ca channels dur<strong>in</strong>g the action potential<br />

but not by Ca sparks (256). The <strong>in</strong>ability of Ca sparks to<br />

activate SK channels was expla<strong>in</strong>ed by the small density<br />

of the SK channels near the discharg<strong>in</strong>g sites. Indeed, the<br />

density of SK channels <strong>in</strong> bladder myocytes was estimated<br />

to be 200 times lower than that of BK or L-type<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

Ca channels (257). The activation of SK channels occurs<br />

through the b<strong>in</strong>d<strong>in</strong>g of Ca to calmodul<strong>in</strong>, which is constitutively<br />

bound to the COOH term<strong>in</strong>us of the channel<br />

(503). Thus SK channels are well suited to respond to<br />

changes <strong>in</strong> global [Ca] (0.1–1 M), s<strong>in</strong>ce calmodul<strong>in</strong> is<br />

saturated by [Ca] below 1 M. However, Kong et al. (368)<br />

showed that IP 3-<strong>in</strong>duced Ca release <strong>in</strong> colonic myocytes<br />

activated SK. Their activation has been suggested to underlie<br />

the hyperpolarization and relaxation associated<br />

with <strong>in</strong>hibitory pur<strong>in</strong>ergic <strong>in</strong>put on these cells. The SK<br />

channels are also responsible for the hyperpolarization <strong>in</strong><br />

response to release of ATP from enteric <strong>in</strong>hibitory motorneurons<br />

(360). More studies are required before it is<br />

known if this is common to other smooth muscles.<br />

When mice were produced <strong>in</strong> which SK3 was absent,<br />

no aberrant phenotype was observed (68). However, overexpression<br />

of SK3 unexpectedly led to difficulties <strong>in</strong><br />

breath<strong>in</strong>g and parturition. Labor was prolonged and described<br />

as dystocic. These data suggest SK channel downregulation<br />

may be required for parturition and labor. Modzelewska<br />

et al. (485) subsequently showed that apam<strong>in</strong><br />

<strong>in</strong>hibits NO-<strong>in</strong>duced relaxation of the myometrium, but<br />

did not affect spontaneous contractions. In bladder, <strong>in</strong>hibition<br />

of SK3 channels has been associated with <strong>in</strong>stability<br />

(257).<br />

The IK channel, also known as KCNN4, K3.1, and<br />

formerly SK4 (527, 747), is the product of different genes<br />

from SK1-3 and exhibits only 40% am<strong>in</strong>o acid sequence<br />

identity (40). The IK have conductances of 50–70 pS<br />

(718). These channels are blocked by TRAM-34 and clotrimazole,<br />

are sensitive to charybdotox<strong>in</strong> but <strong>in</strong>sensitive<br />

to apam<strong>in</strong>, and are not time or voltage dependent (506). It<br />

is now considered that IK channels are the Gardos channels<br />

of erythrocytes, responsible for their high K permeability<br />

(269).<br />

In the endothelium, IK channels contribute to endothelial<br />

derived hyperpolariz<strong>in</strong>g factor (EDHF)-mediated<br />

responses. Deletion of IK channels attenuates ACh-<strong>in</strong>duced<br />

hyperpolarization of endothelial cells and vascular<br />

myocytes (637; see also Ref. 179). Blood pressure was<br />

elevated <strong>in</strong> the IK knockout mice <strong>in</strong> agreement with a role<br />

for EDHF <strong>in</strong> its regulation (81).<br />

The IK channel may be associated with proliferat<strong>in</strong>g<br />

arterial and airway smooth muscle (510, 628). The latter<br />

authors showed IK expression <strong>in</strong> human airway and suggest<br />

that their upregulation plays a role <strong>in</strong> disease. The<br />

mRNA for IK channels has also been found <strong>in</strong> human<br />

trachea (628). It is likely that IK channels will contribute<br />

to sett<strong>in</strong>g the membrane potential relatively negative <strong>in</strong><br />

the cells that express them. As mentioned already, IK<br />

channels <strong>in</strong> endothelial cells can lead to hyperpolarization<br />

of vascular myocytes. In human pulmonary artery, IK<br />

expression and current have been shown, but these are<br />

likely to come from endothelial cells not myoctyes (301,<br />

564). Thus, although there is a physiological importance


of IK channels to smooth muscle, most of this appears to<br />

be due to <strong>in</strong>direct effects.<br />

In summary, although there is now good evidence for<br />

SK channels play<strong>in</strong>g an important role <strong>in</strong> control of excitability<br />

and contraction <strong>in</strong> some types of smooth muscles,<br />

the functional role of these channels <strong>in</strong> other types of<br />

smooth muscles is still not clear. The Ca signals that<br />

activate SK and IK channels <strong>in</strong> different types of smooth<br />

muscles are not well established. The situation is even<br />

more complex for blood vessels, which conta<strong>in</strong> both endothelial<br />

and smooth muscle, both of which can express<br />

different sets of BK, SK, and IK channels. However, one<br />

can speculate that SK channels are expressed and functional<br />

<strong>in</strong> smooth muscle cells that are controlled by a<br />

burst of action potentials. Data obta<strong>in</strong>ed on <strong>in</strong>tact ur<strong>in</strong>ary<br />

bladder myocytes suggest that SK channels are <strong>in</strong>volved<br />

<strong>in</strong> control of the AHP which sets a “m<strong>in</strong>i refractory period”<br />

between action potentials. Dur<strong>in</strong>g this period, voltage<br />

<strong>in</strong>activation of Ca channels is prevented; thus the<br />

channels are available for generation of the next action<br />

potential. SK channels are well suited for this function,<br />

i.e., controll<strong>in</strong>g AHP as they are more sensitive to [Ca]<br />

than BK and thus can be activated by a relatively small<br />

rise <strong>in</strong> cytoplasmic [Ca] produced by a brief action potential.<br />

To achieve a graded rise of force <strong>in</strong> bladder, uterus,<br />

and <strong>in</strong>test<strong>in</strong>al muscles, a burst of action potentials is<br />

required which produces Ca oscillations. This <strong>in</strong> turn will<br />

result <strong>in</strong> a gradual summation of force and the generation<br />

of the large synchronized contractions needed for void<strong>in</strong>g<br />

or parturition. This mechanism of generation of muscle<br />

“tone” <strong>in</strong> phasic action potential-generat<strong>in</strong>g smooth muscles<br />

has many advantages over muscle tone produced by<br />

susta<strong>in</strong>ed membrane potential depolarization. One can<br />

also speculate that Ca oscillations are well suited to the<br />

generation of gradually ris<strong>in</strong>g force <strong>in</strong>duced by chang<strong>in</strong>g<br />

the frequency of action potentials. <strong>Smooth</strong> muscles controlled<br />

by s<strong>in</strong>gle action potentials, such as ureter, appear<br />

not to express SK channels. Instead, a Ca sparks/STOCs<br />

coupl<strong>in</strong>g mechanism operates, sett<strong>in</strong>g the refractory period<br />

to ensure that there is no summation of the action<br />

potential, which <strong>in</strong> the case of the ureter would impair the<br />

flow of ur<strong>in</strong>e and lead to kidney damage.<br />

D. Ca-Activated Cl Channels<br />

Chloride channels sensitive to Ca are a second population<br />

of Ca-sensitive plasma membrane ion channels<br />

that respond to both local (Ca sparks or puffs) or global<br />

(Ca spikes, waves, and oscillations) SR Ca release events.<br />

They are encoded by a family of genes and found <strong>in</strong> many<br />

smooth muscles (84, 163, 226). In the majority of smooth<br />

muscle cells, electrochemical driv<strong>in</strong>g force for Cl (E Cl) is<br />

<strong>in</strong> the range of 20 to 30 mV, i.e., more positive than the<br />

rest<strong>in</strong>g membrane potential, which is <strong>in</strong> the range of 50<br />

SMOOTH MUSCLE SR 155<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

to 60 mV. Thus the E Cl is outwardly directed, and activation<br />

of Cl currents will result <strong>in</strong> diffusion of Cl out of<br />

the cell, caus<strong>in</strong>g membrane depolarization, i.e., an <strong>in</strong>crease<br />

<strong>in</strong> the excitability of smooth muscle cells (321).<br />

Ca-activated Cl channel currents activated by agonist<strong>in</strong>duced<br />

global Ca signals from IP 3-sensitive Ca stores<br />

have been identified <strong>in</strong> a number of smooth muscles<br />

<strong>in</strong>clud<strong>in</strong>g coronary artery (355), anococcygeus muscle<br />

(97, 98), portal ve<strong>in</strong> (539), pulmonary artery (737), esophagus<br />

(4), trachea (310), <strong>in</strong>test<strong>in</strong>e (717), myometrium (17),<br />

and rat ureter (625). In some types of smooth muscle, Ca<br />

sparks were shown to activate Cl Ca to promote membrane<br />

depolarization and contraction (369, 810). STICs (810)<br />

were first observed <strong>in</strong> myocytes from trachea (309) and<br />

portal ve<strong>in</strong> (386, 736) and have subsequently been reported<br />

<strong>in</strong> mesenteric ve<strong>in</strong> (716), trachea (251, 308, 810),<br />

and pulmonary artery (270, 369), suggest<strong>in</strong>g that a Ca<br />

sparks/STIC coupl<strong>in</strong>g mechanism serves a specific<br />

functional role <strong>in</strong> these types of smooth muscles. The<br />

properties of STICs are consistent with Ca sparks caus<strong>in</strong>g<br />

the transient activation of Cl Ca <strong>in</strong> the plasma membrane,<br />

as they are abolished by chloride channel blockers<br />

and are <strong>in</strong>hibited by the depletion of SR Ca stores<br />

(251, 386, 736, 810).<br />

The coexistence of Cl Ca and BK channels <strong>in</strong> the sarcolemmal<br />

membrane adjacent to Ca spark sites suggests<br />

another level of f<strong>in</strong>e control of membrane excitability. In<br />

tracheal smooth muscle cells, STICs have been demonstrated<br />

to be activated by Ca sparks <strong>in</strong> conjunction with<br />

STOCs, lead<strong>in</strong>g to generation of biphasic STOICs (369,<br />

810). There are some notable differences <strong>in</strong> the properties<br />

of STICs and STOCs. The decay of STICs (t 1/2 65–182<br />

ms, Ref. 810) is similar to the decay of Ca sparks (810) and<br />

is much slower than the decay of STOCs (t 1/2 9–43 ms,<br />

Ref. 810). The amplitudes of STICs and STOCs are similar<br />

[with similar driv<strong>in</strong>g forces for Cl and K even though the<br />

unitary conductance of the BK channel (80 pS) is 30-fold<br />

greater than the Cl Ca channel (2.6 pS) (369)]. Therefore, a<br />

STIC appears to represent the activation by a Ca spark of<br />

at least 600 clustered Cl Ca channels. The activation characteristics<br />

and Ca sensitivities of BK and Cl Ca channels<br />

are also different. BK channels are both Ca and voltage<br />

sensitive, and depolarization <strong>in</strong>creases their sensitivity to<br />

[Ca] (107, 134, 136, 438, 508). The Cl Ca channels appear to<br />

have a higher aff<strong>in</strong>ity for Ca (half-maximal activation at<br />

370 nM, Ref. 538) than BK channels and are less voltage<br />

sensitive.<br />

The precise effect on membrane potential of the<br />

simultaneous activation of Cl Ca and BK channels by a Ca<br />

spark will depend on a number of factors. These <strong>in</strong>clude<br />

the poor voltage dependence of Cl Ca and high voltage<br />

dependence of BK channels whose P o changes about<br />

e-fold for a 12- to 14-mV change <strong>in</strong> potential (42, 383) and<br />

their conductances <strong>in</strong> the cell membrane. Thus, at negative<br />

potentials (70 to 60 mV), the predom<strong>in</strong>ant effect


156 SUSAN WRAY AND THEODOR BURDYGA<br />

of Ca spark discharge will be to trigger STICs (810), s<strong>in</strong>ce<br />

at these potentials the electrochemical driv<strong>in</strong>g force for Cl<br />

will be greater than that for K. The discharge of STICs will<br />

cause membrane depolarization and thus serve as a positive-feedback<br />

mechanism to trigger the activation of voltage-gated<br />

Ca channels and excitation of the myocytes. As<br />

depolarization <strong>in</strong>creases the frequency of Ca sparks and<br />

STOCs, sensitivity of BK channels to Ca and the electrochemical<br />

driv<strong>in</strong>g force for K will also be <strong>in</strong>creased. This<br />

will make BK more active and result <strong>in</strong> the generation of<br />

high-amplitude STOCs and hyperpolarization. This would<br />

lead to closure of voltage-gated Ca channels and relaxation<br />

of smooth muscle, as was reported for pressurized<br />

cerebral arteries (507). High sensitivity of Cl Ca to [Ca] and<br />

low sensitivity to voltage make them ideal ion channels to<br />

be targeted by agonists to elicit excitatory effects on<br />

smooth muscle cells. Unlike STICs, the whole cell Cl Ca<br />

currents decay much faster than global Ca transients as<br />

the <strong>in</strong>activation of Cl Ca current is determ<strong>in</strong>ed not by<br />

<strong>in</strong>tracellular [Ca], but by phosphorylation of the channel<br />

by CAM k<strong>in</strong>ase II (739). Therefore, the decay of a STIC<br />

appears to be determ<strong>in</strong>ed by the decl<strong>in</strong>e of Ca sparks,<br />

whereas the decay of the whole cell Cl Ca currents is<br />

controlled by CAM k<strong>in</strong>ase II activity.<br />

To summarize these data, we conclude that the expression<br />

of a range of different ion channels enables<br />

muscles to perform diverse functions <strong>in</strong> the human body.<br />

Recent data clearly <strong>in</strong>dicate a role of the SR <strong>in</strong> the Ca<br />

signal<strong>in</strong>g that controls smooth muscle function, via coupl<strong>in</strong>g<br />

Ca release from the SR to activation of ion channels.<br />

Discovery of Ca sparks, waves, and oscillations that can<br />

target several Ca-regulated channels (BK, SK, and IK) can<br />

produce positive or negative feedback controll<strong>in</strong>g contractility.<br />

For example, some tonic contractions, which<br />

can be achieved via susta<strong>in</strong>ed membrane depolarization<br />

or discharge of bursts of action potentials, can be effectively<br />

decreased via activation of a Ca sparks/STOCs coupl<strong>in</strong>g<br />

mechanism. BK channels coupled to Ca sparks may<br />

produce long refractory periods, whereas SK channels<br />

targeted by Ca entry may be associated with short refractory<br />

periods, as discussed above. Still other mechanisms<br />

may be present <strong>in</strong> different smooth muscles, and a better<br />

understand<strong>in</strong>g of them would aid understand<strong>in</strong>g of the<br />

control of their contractility under normal and pathological<br />

conditions.<br />

XI. SARCOPLASMIC RETICULUM AND<br />

DEVELOPMENT AND AGING<br />

In the nervous system and heart there is a relatively<br />

large amount of literature on the role of Ca homeostasis<br />

and the SR, and a grow<strong>in</strong>g understand<strong>in</strong>g of their importance<br />

to ag<strong>in</strong>g, development, and pathology. Less is<br />

known for smooth muscle, but there are some data spe-<br />

cific to fetal and/or neonatal smooth muscles and ag<strong>in</strong>g<br />

tissues and their SR, and this will be briefly presented.<br />

Perhaps given the diversity of expression <strong>in</strong> Ca release<br />

channels between different adult smooth muscles, it<br />

should not be expected that a uniform thesis can be<br />

presented. As some papers have exam<strong>in</strong>ed both development<br />

and ag<strong>in</strong>g, these data will be discussed together as<br />

appropriate. As changes <strong>in</strong> [Ca] are considered to be<br />

important for gene expression and pathological conditions<br />

<strong>in</strong> very many tissues, their role <strong>in</strong> development and<br />

ag<strong>in</strong>g <strong>in</strong> smooth muscles would seem to be worthy of<br />

study. It should also be acknowledged that changes<br />

<strong>in</strong>volv<strong>in</strong>g the SR may be direct, e.g., SERCA expression,<br />

or <strong>in</strong>direct, e.g., due to changes <strong>in</strong> Ca buffer<strong>in</strong>g or to<br />

prote<strong>in</strong>-coupled receptor sensitivity and IP 3 production.<br />

Other changes with development, e.g., development<br />

of force (502, 613), or Ca sensitivity (682), are not<br />

considered here.<br />

Although a complete data set is not available for any<br />

smooth muscle, it would seem reasonable to conclude<br />

that a change <strong>in</strong> the role of the SR occurs with the<br />

transition from fetal and neonatal life to adulthood. There<br />

is evidence for this at both structural and biochemical<br />

levels, as well as from physiological studies. Care has to<br />

be taken <strong>in</strong> the <strong>in</strong>terpretation of these data as it is not<br />

always possible to dist<strong>in</strong>guish SR from ER, and thus to<br />

establish if the greater ER prote<strong>in</strong> production <strong>in</strong> the neonates<br />

contributes to these differences. In addition, not all<br />

studies have been able to report directly on either<br />

<strong>in</strong>tracellular or lum<strong>in</strong>al [Ca]. Consequently, although<br />

changes <strong>in</strong> SR Ca uptake and release and Ca pool size<br />

can expla<strong>in</strong> the data, effects on processes such as Ca<br />

sensitization or excitability cannot always be excluded.<br />

Nevertheless, a greater contribution of SR to smooth<br />

muscle function <strong>in</strong> the neonate has been identified <strong>in</strong> a<br />

number of <strong>in</strong>stances, <strong>in</strong>clud<strong>in</strong>g uterus, bladder, and<br />

some blood vessels.<br />

Follow<strong>in</strong>g examples <strong>in</strong> other tissues, where ag<strong>in</strong>g has<br />

been l<strong>in</strong>ked to dysfunctions <strong>in</strong> Ca homeostasis and the SR<br />

(409, 570, 616), studies <strong>in</strong> smooth muscles have also<br />

po<strong>in</strong>ted to alterations <strong>in</strong> Ca signal<strong>in</strong>g (448, 593, 779). From<br />

this review of the data, it is apparent that cytoplasmic<br />

[Ca] can rise with ag<strong>in</strong>g <strong>in</strong> smooth muscles. This <strong>in</strong> turn<br />

may be attributed to a decl<strong>in</strong>e <strong>in</strong> SERCA activity, although<br />

more studies on this po<strong>in</strong>t are required. This rise <strong>in</strong> rest<strong>in</strong>g<br />

[Ca] will produce overcontraction and contribute to<br />

dysfunctions associated with ag<strong>in</strong>g such as hypertension<br />

and <strong>in</strong>cont<strong>in</strong>ence urge.<br />

XII. GENDER<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

The effects of female hormones on the vasculature<br />

and other smooth muscles are well recognized. While a<br />

discussion of these <strong>in</strong>terest<strong>in</strong>g differences is beyond the


scope of this review, a little can be said about the SR and<br />

gender.<br />

The earliest and still best documented effects of<br />

gender on SR function come from studies on cardiac<br />

muscle, <strong>in</strong>clud<strong>in</strong>g human myocardium (119, 144, 365,<br />

388, 547). SERCA levels are depressed <strong>in</strong> fail<strong>in</strong>g hearts,<br />

as is phospholamban phosphorylation. The latter characteristic,<br />

however, was only found <strong>in</strong> male hearts. Men<br />

have a greater <strong>in</strong>cidence of coronary heart disease and<br />

have augmented myocardial ischemic reperfusion <strong>in</strong>jury,<br />

which has been related to <strong>in</strong>creased SR Ca load<br />

(“overload”) (119). In smooth muscle, the few studies<br />

we have located have been on vascular smooth muscle.<br />

The f<strong>in</strong>d<strong>in</strong>gs of lower blood pressure <strong>in</strong> women than<br />

men, and the demonstration of male rats <strong>in</strong> hypertensive<br />

stra<strong>in</strong>s develop<strong>in</strong>g a greater elevation of blood<br />

pressure and with earlier onset than females, can be<br />

partly attributed to estrogen. There is an estrogendependent,<br />

protective enhanced release of NO <strong>in</strong> female<br />

endothelial cells (112). The cardiac studies mentioned<br />

above would also po<strong>in</strong>t to mechanisms directly<br />

present <strong>in</strong> the vascular myocytes and <strong>in</strong>volv<strong>in</strong>g the SR,<br />

as well as other aspects of Ca homeostasis. In addition,<br />

female hypertensive rats still exhibit an enhanced vasodilatory<br />

response <strong>in</strong> the absence of endothelium,<br />

compared with males (328).<br />

Aortic cells isolated from male hypertensive rats<br />

proliferated more rapidly than those from female rats<br />

(28). There were no differences <strong>in</strong> rest<strong>in</strong>g [Ca] between<br />

the cells, but a more pronounced <strong>in</strong>crease <strong>in</strong> [Ca] to<br />

angiotens<strong>in</strong> II occurred <strong>in</strong> males (411). As the authors<br />

noted, this could <strong>in</strong>dicate a greater ability of agonist to<br />

mobilize Ca from the SR or release threshold <strong>in</strong> males.<br />

In a follow up study, <strong>in</strong>tracellular [Ca] was measured<br />

and the augmented Ca response to angiotens<strong>in</strong> II <strong>in</strong><br />

males confirmed. However, SERCA <strong>in</strong>hibition with<br />

thapsigarg<strong>in</strong> produced a much larger [Ca] rise <strong>in</strong> females.<br />

Thus a role for <strong>in</strong>creased Ca entry rather than SR<br />

Ca release, <strong>in</strong> males, would appear more likely (412).<br />

Gender differences <strong>in</strong> Ca entry pathways have previously<br />

been demonstrated (496). This study also found<br />

no evidence for gender-based differences <strong>in</strong> SR Ca<br />

release. Effects of estrogens on SERCA expression <strong>in</strong><br />

smooth muscles have been suggested from a few studies<br />

(see sect. IIIE).<br />

It seems to us that there is little direct evidence thus<br />

far for effects of gender on Ca handl<strong>in</strong>g and SR function<br />

<strong>in</strong> smooth muscle cells. It is, however, premature to decide<br />

whether this is due to a lack of significant effects or<br />

the small amount of data currently available. Given the<br />

high <strong>in</strong>cidence and morbidity of cardiovascular disease, it<br />

would be fruitful to have more <strong>in</strong>formation from smooth<br />

muscles on these issues.<br />

SMOOTH MUSCLE SR 157<br />

XIII. pH, METABOLISM, AND SARCOPLASMIC<br />

RETICULUM FUNCTION<br />

A. Introduction<br />

That changes <strong>in</strong> pH can affect smooth muscle function<br />

has long been known (645). These changes can be<br />

<strong>in</strong>tracellular (pH i), occurr<strong>in</strong>g for example with changes of<br />

metabolism and activity, or extracellular (pH o), consequent<br />

to changes <strong>in</strong> perfusion or systemic pH disturbance,<br />

for example (768). The mechanisms by which a change <strong>in</strong><br />

pH i or pH o can affect E-C coupl<strong>in</strong>g <strong>in</strong> smooth muscle are<br />

numerous. Our focus is on SR Ca release processes and<br />

SERCA activity. For details on pH and smooth muscles,<br />

see the follow<strong>in</strong>g reviews (22, 732, 772). Changes <strong>in</strong> pH<br />

are closely associated with metabolic activity and accompanied<br />

by changes <strong>in</strong> ATP and P i concentrations, which as<br />

we discuss will also <strong>in</strong>fluence SR function.<br />

B. pH<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

Release through RyR has been shown to be pH sensitive,<br />

with alkal<strong>in</strong>ization <strong>in</strong>creas<strong>in</strong>g and acidification decreas<strong>in</strong>g<br />

the sensitivity of the release channels to Ca (151,<br />

513, 763). In a sk<strong>in</strong>ned portal ve<strong>in</strong> preparation (286),<br />

reduction <strong>in</strong> pH <strong>in</strong>creased CICR, suggest<strong>in</strong>g <strong>in</strong> this simplified<br />

system, where bath<strong>in</strong>g [Ca] is ma<strong>in</strong>ta<strong>in</strong>ed constant,<br />

that the RyR are either open for longer or have an <strong>in</strong>creased<br />

P o as pH falls. In <strong>in</strong>tact preparations, this effect<br />

may be masked, as other processes <strong>in</strong>volved <strong>in</strong> Ca homeostasis<br />

are affected. In a recent study <strong>in</strong> <strong>in</strong>tact rat<br />

aortic preparations, it was concluded that acidic pH <strong>in</strong>duces<br />

Ca release from RyRs and contributes to the rate of<br />

rise of contraction (594). Studies on cardiac SR microsomes<br />

and lipid bilayers have shown that a fall <strong>in</strong> pH<br />

lowers the P o of the RyR (e.g., Refs. 602, 782). In isolated<br />

cells, Ca sparks are reduced, which is also consistent with<br />

a fall <strong>in</strong> RyR P o (30). In pressurized cerebral arteries,<br />

Heppner et al. (253) showed that a small alkal<strong>in</strong>ization,<br />

from pH 7.4 to 7.5, <strong>in</strong>creased the frequency of Ca sparks<br />

and, upon rais<strong>in</strong>g it further to pH 7.6, caused a shift from<br />

sparks to Ca waves <strong>in</strong> the cells and vasoconstriction.<br />

These effects were blocked by ryanod<strong>in</strong>e, but not <strong>in</strong>hibitors<br />

of IP 3R. The IP 3R, however, is also pH sensitive (71,<br />

146, 232, 709); alkal<strong>in</strong>ization <strong>in</strong>creases the sensitivity of<br />

IICR. These effects may be due to changes of pH affect<strong>in</strong>g<br />

IP 3 b<strong>in</strong>d<strong>in</strong>g to the receptor (497), or IP 3 production (8).<br />

Alkal<strong>in</strong>ization will also <strong>in</strong>crease the P o of IP 3R (324, 767).<br />

Changes of pH were also reported to be more potent<br />

when act<strong>in</strong>g at IP 3R3 rather than IP 3R1 (146). Further data<br />

are required to confirm isoform pH sensitivity.<br />

As noted <strong>in</strong> section III, protons are translocated dur<strong>in</strong>g<br />

SERCA activity from lumen to cytoplasm (395, 785)<br />

and may contribute to stabilization of SERCA when its Ca


158 SUSAN WRAY AND THEODOR BURDYGA<br />

b<strong>in</strong>d<strong>in</strong>g sites are vacant (700). The proton countertransport<br />

has been reported to stop around lum<strong>in</strong>al pH 8.0<br />

(551). Thus it may be anticipated that pH will affect<br />

SERCA Ca pump<strong>in</strong>g. Grover’s group have studied the<br />

effects of pH alteration on SERCA activity <strong>in</strong> a variety of<br />

smooth muscle preparations and concluded that alkal<strong>in</strong>ization<br />

decreased its activity and acidification <strong>in</strong>creased it<br />

(165, 166, 228). As a decrease <strong>in</strong> pH <strong>in</strong>hibits k<strong>in</strong>ase activity,<br />

then phosphorylation of phospholamban will be reduced,<br />

which <strong>in</strong> turn will produce a decrease <strong>in</strong> SERCA<br />

activity (606). Stimulation of SOCE, by SR Ca depletion,<br />

has also been reported to be pH sensitive and contribute<br />

to the effects of pH o on vascular tone (748).<br />

Thus, although sometimes overlooked as <strong>in</strong>vestigators<br />

focus on ion channels and changes <strong>in</strong> myofilament Ca<br />

sensitivity, effects of pH alterations on Ca release and<br />

SERCA activity <strong>in</strong> smooth muscles will be significant<br />

factors <strong>in</strong> functional changes.<br />

C. Metabolism<br />

Here we address how metabolic changes such as<br />

ATP, phosphocreat<strong>in</strong>e (PCr), and P i levels affect SERCA<br />

activity. Most methods of determ<strong>in</strong><strong>in</strong>g SERCA activity<br />

couple its cycl<strong>in</strong>g to ATP hydrolysis via measurements of<br />

NADH, with the rate of NADH disappearance be<strong>in</strong>g proportional<br />

to the rate of ATP hydrolysis by SERCA (414).<br />

Other approaches target the production of P i by SERCA<br />

activity <strong>in</strong> colorimetric-based assays (e.g., Ref. 109). In a<br />

recent paper, Williams et al. (762) described an HPLCbased<br />

method that also measured ATP, ADP, and PCr.<br />

The SERCA activity is calculated from the changes <strong>in</strong> ATP<br />

and ADP. The authors were also able to confirm that the<br />

PCr is used locally via creat<strong>in</strong>e k<strong>in</strong>ase to synthesize local<br />

ATP for SERCA (244, 366). Thus functional coupl<strong>in</strong>g and<br />

localization between PCr, creat<strong>in</strong>e k<strong>in</strong>ase, and SERCA<br />

occurred. This specific b<strong>in</strong>d<strong>in</strong>g of creat<strong>in</strong>e k<strong>in</strong>ase to<br />

SERCA had previously been reported (e.g., Refs. 367,<br />

598). As PCr levels are much lower <strong>in</strong> smooth compared<br />

with striated muscles (385, 659, 773), this position<strong>in</strong>g of<br />

creat<strong>in</strong>e k<strong>in</strong>ase by the SERCA membrane may be particularly<br />

useful <strong>in</strong> smooth muscles. Local metabolic<br />

control of SERCA may be advantageous dur<strong>in</strong>g conditions<br />

of metabolic stress, <strong>in</strong> ma<strong>in</strong>ta<strong>in</strong><strong>in</strong>g SERCA function<br />

and prevent<strong>in</strong>g cytoplasmic [Ca] from becom<strong>in</strong>g<br />

too high.<br />

Investigators have suggested, based on the location<br />

of glycolytic enzymes, that there is also a coupl<strong>in</strong>g between<br />

glycolysis and SERCA activity (168, 780). In smooth<br />

muscle, a large body of work, particularly from Lorenz<br />

and Paul (410), has led to the theory of metabolic compartmentation.<br />

Although beyond the scope of this review,<br />

there is good evidence that ATP, provided by oxidative<br />

phosphorylation, supports contraction, whereas that from<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

glycolysis supports membrane pumps <strong>in</strong> smooth muscle<br />

(246, 295, 420, 421). There is also a much greater lactate<br />

production and use of glycolysis under normoxic conditions<br />

<strong>in</strong> smooth muscle compared with many other cell<br />

types (422, 770). Dur<strong>in</strong>g hypoxia or ischemia, effects of<br />

pH and changes <strong>in</strong> metabolites on SERCA activity will be<br />

expected to contribute to contractile dysfunction (683).<br />

XIV. MATHEMATICAL MODELING<br />

Given the accepted complexities of SR Ca uptake and<br />

release events, and their reciprocal effect on cytoplasmic<br />

[Ca], plasmalemmal ion channels, and other organelles<br />

such as the mitochondria, there is merit <strong>in</strong> develop<strong>in</strong>g<br />

models of E-C coupl<strong>in</strong>g <strong>in</strong> smooth muscle cells. One<br />

considerable obstacle to the use of such models is the<br />

lack of quantitative data for many of the essential parameters,<br />

e.g., SR [Ca]. Even sett<strong>in</strong>g aside experimental differences<br />

such as temperature, species, and stimulation<br />

mode, for some smooth muscles, e.g., urethra and anococcygeus,<br />

many necessary measurements are simply not<br />

available; others such as vascular and uter<strong>in</strong>e smooth<br />

muscles have been better studied.<br />

There are several mathematical models of various<br />

aspects of E-C coupl<strong>in</strong>g <strong>in</strong> smooth muscle now available<br />

[e.g., airway, (376), mesenteric artery (335), cerebral artery<br />

(786)]. Other models have addressed more general<br />

questions, such as superficial buffer barrier (37), slow<br />

waves (289), prote<strong>in</strong> <strong>in</strong>teractions (171), and IP 3 changes<br />

(176). The use of such models could provide significant<br />

<strong>in</strong>sight <strong>in</strong>to the work<strong>in</strong>gs of the SR by tak<strong>in</strong>g an <strong>in</strong>tegrated<br />

approach. As such models become more detailed and SR<br />

focussed, and the “miss<strong>in</strong>g” parameters are experimentally<br />

determ<strong>in</strong>ed, then a better overall description and<br />

understand<strong>in</strong>g of the SR <strong>in</strong> different smooth muscles<br />

should emerge.<br />

XV. CONCLUSIONS AND FUTURE DIRECTIONS<br />

The SR is vital to the modulation and regulation of<br />

force <strong>in</strong> smooth muscles. Pathophysiologies are associated<br />

with perturbation of SERCA, Ca release mechanisms,<br />

and regulatory prote<strong>in</strong>s, but our appreciation of<br />

this lags beh<strong>in</strong>d studies on striated muscles. This can<br />

largely be attributed to the lack of clarity around the<br />

regulation of normal SR function <strong>in</strong> smooth muscle,<br />

which <strong>in</strong> turn is affected by genu<strong>in</strong>e differences between<br />

tissues. It is uncommon for <strong>in</strong>vestigators to address more<br />

than one smooth muscle <strong>in</strong> their studies, lead<strong>in</strong>g to pockets<br />

of detailed <strong>in</strong>formation <strong>in</strong> specific areas, but a lack of<br />

breadth. There are areas where real progress has been<br />

made, such as unravel<strong>in</strong>g the Ca spark-STOC mechanism<br />

of regulat<strong>in</strong>g excitability and function. Other areas rema<strong>in</strong><br />

controversial, such as the role of TRPCs <strong>in</strong> store-operated


Ca entry. A key question rema<strong>in</strong><strong>in</strong>g to be answered is<br />

whether <strong>in</strong> smooth muscle SR Ca depletion leads to I crac<br />

only through the Stim-Orai pathway. Another key question<br />

is how important SOCE is to those smooth muscles<br />

whose ma<strong>in</strong> source of Ca for contraction is L-type Ca<br />

entry; need such mechanisms be required if SERCA can<br />

refill from the plasmalemmal Ca entry? More measurements<br />

of SR lum<strong>in</strong>al Ca are also required, as they could<br />

provide important mechanistic <strong>in</strong>sight.<br />

Another area of unf<strong>in</strong>ished bus<strong>in</strong>ess is the role of the<br />

SR <strong>in</strong> shap<strong>in</strong>g Ca signals <strong>in</strong> smooth muscles. With Caactivated<br />

ion channels that can <strong>in</strong>crease or decrease<br />

membrane excitability be<strong>in</strong>g expressed <strong>in</strong> the same myocyte,<br />

elucidation of the role of local SR Ca release to<br />

global Ca signals is difficult. We have suggested that<br />

consideration of the pattern of excitation, i.e., s<strong>in</strong>gle<br />

short-last<strong>in</strong>g or plateau-type action potential or tra<strong>in</strong>s of<br />

action potentials, may dictate the <strong>in</strong>volvement or otherwise<br />

of the SR with BK, IK, and SK and Cl Ca channels<br />

play<strong>in</strong>g a role. Imag<strong>in</strong>g and molecular biology techniques<br />

have supported the views expressed over 30 years ago<br />

that there will be microdoma<strong>in</strong>s of Ca and signal<strong>in</strong>g moieties<br />

with<strong>in</strong> the myocyte. The SR Ca release channels and<br />

Ca-activated ion channels form part of the underly<strong>in</strong>g<br />

mechanism, with both the plasma membrane and SR<br />

membrane hav<strong>in</strong>g signal<strong>in</strong>g microdoma<strong>in</strong>s.<br />

The existence of different SR Ca stores, which may<br />

or may not completely <strong>in</strong>tercommunicate, also leads to<br />

specialization with<strong>in</strong> smooth muscles. Figure 5 illustrates<br />

some features of the store. This complexity of Ca signal<strong>in</strong>g<br />

<strong>in</strong> smooth muscle (and other tissues) is added to by<br />

the grow<strong>in</strong>g evidence that other organelles, <strong>in</strong>clud<strong>in</strong>g mitochondria,<br />

lysosomes, Golgi, and nucleus, <strong>in</strong>teract with<br />

the SR (ER). Furthermore, along with Ca and IP 3 as Ca<br />

release agents, newer messenger/modulators, <strong>in</strong>clud<strong>in</strong>g<br />

NAADP, cADPR, and possibly the L-type Ca channel, need<br />

to be brought <strong>in</strong>to the mix. Presumably the right mix of<br />

messenger(s) and Ca store(s), along with the right place<br />

and time, are how the myocyte decodes the Ca stimulus.<br />

With time, the SR will give up more of its secrets.<br />

This requires laboratories apply<strong>in</strong>g a range of techniques,<br />

<strong>in</strong>clud<strong>in</strong>g functional, electrophysiological, pharmacological,<br />

and molecular biological studies. We are encouraged<br />

that the <strong>in</strong>creased use of <strong>in</strong>tact tissues rather than cultured<br />

cells for molecular biology, along with the success<br />

of nonviral transfection methods, will result <strong>in</strong> much new<br />

data which are physiologically relevant and not dogged by<br />

concerns about the phenotypic changes occurr<strong>in</strong>g with<br />

cultur<strong>in</strong>g. Ultimately, an improved understand<strong>in</strong>g of the<br />

SR of smooth muscles will contribute to the development<br />

of new therapeutic approaches to tackle diseases. Given<br />

the pivotal role of smooth muscle to all the major systems<br />

of the body, this is a worthwhile goal.<br />

SMOOTH MUSCLE SR 159<br />

ACKNOWLEDGMENTS<br />

We are extremely grateful to M. Nelson for secretarial<br />

support and help and to Amanda Heath, Sarah Arrowsmith,<br />

Karen Noble, and Lydmylla Borysova for read<strong>in</strong>g drafts. We<br />

acknowledge and thank all our colleagues and group members<br />

who have contributed to the work <strong>in</strong> this review and engaged <strong>in</strong><br />

discussions with us over the years.<br />

Address for repr<strong>in</strong>t requests and other correspondence: S.<br />

Wray, Dept. of Physiology, Univ. of Liverpool, Crown Street,<br />

Liverpool, Merseyside L69 3BX, UK (e-mail: s.wray@liv.ac.uk).<br />

GRANTS<br />

We thank the British Heart Foundation, Action Medical<br />

Research, WellBe<strong>in</strong>g of Women, the Mothercare Trust, and the<br />

Wellcome Trust for support<strong>in</strong>g our research programs.<br />

REFERENCES<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

1. Abe F, Karaki H, Endoh M. Effects of cyclopiazonic acid and<br />

ryanod<strong>in</strong>e on cytosolic calcium and contraction <strong>in</strong> vascular smooth<br />

muscle. Br J Pharmacol 118: 1711–1716, 1996.<br />

2. Abrenica B, Gilchrist JS. Nucleoplasmic Ca 2 load<strong>in</strong>g is regulated<br />

by mobilization of per<strong>in</strong>uclear Ca 2 . Cell Calcium 28: 127–<br />

136, 2000.<br />

3. Abrenica B, Pierce GN, Gilchrist JS. Nucleoplasmic calcium<br />

regulation <strong>in</strong> rabbit aortic vascular smooth muscle cells. Can<br />

J Physiol Pharmacol 81: 301–310, 2003.<br />

4. Akbarali HI, Giles WR. Ca 2 and Ca 2 -activated Cl currents <strong>in</strong><br />

rabbit oesophageal smooth muscle. J Physiol 460: 117–133, 1993.<br />

5. Albert AP, Large WA. A Ca 2 -permeable non-selective cation<br />

channel activated by depletion of <strong>in</strong>ternal Ca 2 stores <strong>in</strong> s<strong>in</strong>gle<br />

rabbit portal ve<strong>in</strong> myocytes. J Physiol 538: 717–728, 2002.<br />

6. Albert AP, Large WA. Activation of store-operated channels by<br />

noradrenal<strong>in</strong>e via prote<strong>in</strong> k<strong>in</strong>ase C <strong>in</strong> rabbit portal ve<strong>in</strong> myocytes.<br />

J Physiol 544: 113–125, 2002.<br />

7. Albert AP, Saleh SN, Peppiatt-Wildman CM, Large WA. Multiple<br />

activation mechanisms of store-operated TRPC channels <strong>in</strong><br />

smooth muscle cells. J Physiol 583: 25–36, 2007.<br />

8. Albuquerque MLC, Leffler CW. pH o,pH i, and PCO 2 <strong>in</strong> stimulation<br />

of IP 3 and [Ca 2 ] <strong>in</strong> piglet cerebrovascular smooth muscle. Proc<br />

Soc Exp Biol Med 219: 326–334, 1998.<br />

9. Algenstaedt P, Antonetti DA, Yaffe MB, Kahn CR. Insul<strong>in</strong><br />

receptor substrate prote<strong>in</strong>s create a l<strong>in</strong>k between the tyros<strong>in</strong>e<br />

phosphorylation cascade and the Ca 2 -ATPases <strong>in</strong> muscle and<br />

heart. J Biol Chem 272: 23696–23702, 1997.<br />

10. Alicia S, Angelica Z, Carlos S, Alfonso S, Vaca L. STIM1 converts<br />

TRPC1 from a receptor-operated to a store-operated channel:<br />

mov<strong>in</strong>g TRPC1 <strong>in</strong> and out of lipid rafts. Cell Calcium 44: 479–491,<br />

2008.<br />

11. Allen DG, Eisner DA, Wray SC. Birthday present for digitalis.<br />

Nature 316: 674–675, 1985.<br />

12. Ambudkar IS, Ong HL, Liu X, Bandyopadhyay BC, Cheng KT.<br />

TRPC1: the l<strong>in</strong>k between functionally dist<strong>in</strong>ct store-operated calcium<br />

channels. Cell Calcium 42: 213–223, 2007.<br />

13. Amrani Y, Panettieri RA. Airway smooth muscle: contraction<br />

and beyond. Int J Biochem Cell Biol 35: 272–276, 2003.<br />

14. Amrani Y, Panettieri RA Jr, Frossard N, Bronner C. Activation<br />

of the TNF alpha-p55 receptor <strong>in</strong>duces myocyte proliferation and<br />

modulates agonist-evoked calcium transients <strong>in</strong> cultured human<br />

tracheal smooth muscle cells. Am J Respir Cell Mol Biol 15: 55–63,<br />

1996.<br />

15. Anger M, Samuel JL, Marotte F, Wuytack F, Rappaport L,<br />

Lompre AM. In situ mRNA distribution of sarco(endo)plasmic<br />

reticulum Ca 2 -ATPase isoforms dur<strong>in</strong>g ontogeny <strong>in</strong> the rat. J Mol<br />

Cell Cardiol 26: 539–550, 1994.<br />

16. Arnaudeau S, Kelley WL, Walsh JV Jr, Demaurex N. Mitochondria<br />

recycle Ca 2 to the endoplasmic reticulum and prevent the<br />

depletion of neighbor<strong>in</strong>g endoplasmic reticulum regions. J Biol<br />

Chem 276: 29430–29439, 2001.


160 SUSAN WRAY AND THEODOR BURDYGA<br />

17. Arnaudeau S, Lepretre N, Mironneau J. Chloride and monovalent<br />

ion-selective cation currents activated by oxytoc<strong>in</strong> <strong>in</strong> pregnant<br />

rat myometrial cells. Am J Obstet Gynecol 171: 491–501, 1994.<br />

18. Arnaudeau S, Macrez-Lepretre N, Mironneau J. Activation of<br />

calcium sparks by angiotens<strong>in</strong> II <strong>in</strong> vascular myocytes. Biochem<br />

Biophys Res Commun 222: 809–815, 1996.<br />

19. Asahi M, Green NM, Kurzydlowski K, Tada M, MacLennan<br />

DH. Phospholamban doma<strong>in</strong> IB forms an <strong>in</strong>teraction site with the<br />

loop between transmembrane helices M6 and M7 of sarco(endo)<br />

plasmic reticulum Ca 2 ATPases. Proc Natl Acad Sci USA 98:<br />

10061–10066, 2001.<br />

20. Asahi M, Kimura Y, Kurzydlowski K, Tada M, MacLennan DH.<br />

Transmembrane helix M6 <strong>in</strong> sarco(endo)plasmic reticulum Ca 2 -<br />

ATPase forms a functional <strong>in</strong>teraction site with phospholamban.<br />

Evidence for physical <strong>in</strong>teractions at other sites. J Biol Chem 274:<br />

32855–32862, 1999.<br />

21. Asahi M, Nakayama H, Tada M, Otsu K. Regulation of sarco(endo)<br />

plasmic reticulum Ca 2 adenos<strong>in</strong>e triphosphatase by phospholamban<br />

and sarcolip<strong>in</strong>: implication for cardiac hypertrophy and failure.<br />

Trends Cardiovasc Med 13: 152–157, 2003.<br />

22. Aust<strong>in</strong> C, Wray S. Interactions between Ca 2 and H and functional<br />

consequences <strong>in</strong> vascular smooth muscle. Circ Res 86: 353–<br />

363, 2000.<br />

24. Awad SS, Lamb HK, Morgan JM, Dunlop W, Gillespie JI.<br />

Differential espression of ryanod<strong>in</strong>e receptor RyR2 mRNA <strong>in</strong> the<br />

non-pregnant and pregnant human myometrium. Biochem J 322:<br />

777–783, 1997.<br />

25. Baba-Aissa F, Raeymaekers L, Wuytack F, De GC, Missiaen L,<br />

Casteels R. Distribution of the organellar Ca 2 transport ATPase<br />

SERCA2 isoforms <strong>in</strong> the cat bra<strong>in</strong>. Bra<strong>in</strong> Res 743: 141–153, 1996.<br />

26. Babiychuk EB, Smith RD, Burdyga TV, Babiychuk VS, Wray S,<br />

Draeger A. Membrane cholesterol selectively regulates smooth<br />

muscle phasic contraction. J Membr Biol 198: 95–101, 2004.<br />

27. Babu GJ, Bhupathy P, Timofeyev V, Petrashevskaya NN, Reiser<br />

PJ, Chiamvimonvat N, Periasamy M. Ablation of sarcolip<strong>in</strong><br />

enhances sarcoplasmic reticulum calcium transport and atrial contractility.<br />

Proc Natl Acad Sci USA 104: 17867–17872, 2007.<br />

28. Bacakova L, Mares V. Cell k<strong>in</strong>etics of aortic smooth muscle cells<br />

<strong>in</strong> long-term cultures prepared from rats raised under conventional<br />

and SPF conditions. Physiol Res 44: 389–398, 1995.<br />

29. Bai Y, Sanderson MJ. Airway smooth muscle relaxation results<br />

from a reduction <strong>in</strong> the frequency of Ca 2 oscillations <strong>in</strong>duced by<br />

a cAMP-mediated <strong>in</strong>hibition of the IP 3 receptor. Respir Res 7: 34,<br />

2006.<br />

30. Balnave CD, Vaughan-Jones RD. Effect of <strong>in</strong>tracellular pH on<br />

spontaneous Ca 2 sparks <strong>in</strong> rat ventricular myocytes. J Physiol<br />

528: 25–37, 2000.<br />

31. Barlow CA, Rose P, Pulver-Kaste RA, Lounsbury KM. Excitation-transcription<br />

coupl<strong>in</strong>g <strong>in</strong> smooth muscle. J Physiol 570: 59–64,<br />

2006.<br />

32. Baro I, Eisner DA. The effects of thapsigarg<strong>in</strong> on [Ca 2 ] i <strong>in</strong><br />

isolated rat mesenteric artery vascular smooth muscle cells.<br />

Pflügers Arch 420: 115–117, 1992.<br />

33. Barrero MJ, Montero M, Alvarez J. Dynamics of [Ca 2 ]<strong>in</strong>the<br />

endoplasmic reticulum and cytoplasm of <strong>in</strong>tact HeLa cells. A comparative<br />

study. J Biol Chem 272: 27694–27699, 1997.<br />

34. Baygu<strong>in</strong>ov O, Hagen B, Bonev AD, Nelson MT, Sanders KM.<br />

Intracellular calcium events activated by ATP <strong>in</strong> mur<strong>in</strong>e colonic<br />

myocytes. Am J Physiol Cell Physiol 279: C126–C135, 2000.<br />

35. Baygu<strong>in</strong>ov O, Hagen B, Kenyon JL, Saunders KM. Coupl<strong>in</strong>g<br />

strength between localized Ca 2 transients and K channels is<br />

regulated by prote<strong>in</strong> k<strong>in</strong>ase C. Am J Physiol Cell Physiol 281:<br />

C1512–C1523, 2001.<br />

36. Bayle D, Weeks D, Sachs G. The membrane topology of the rat<br />

sarcoplasmic and endoplasmic reticulum calcium ATPases by<br />

<strong>in</strong> vitro translation scann<strong>in</strong>g. J Biol Chem 270: 25678–25684, 1995.<br />

37. Bazzazi H, Kargac<strong>in</strong> ME, Kargac<strong>in</strong> GJ. Ca 2 regulation <strong>in</strong> the<br />

near-membrane microenvironment <strong>in</strong> smooth muscle cells. Biophys<br />

J 85: 1754–1765, 2003.<br />

38. Beard NA, Laver DR, Dulhunty AF. Calsequestr<strong>in</strong> and the calcium<br />

release channel of skeletal and cardiac muscle. Prog Biophys<br />

Mol Biol 85: 33–69, 2004.<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

39. Beech DJ, Muraki K, Flemm<strong>in</strong>g R. Non-selective cationic channels<br />

of smooth muscle and the mammalian homologues of Drosophila<br />

TRP. J Physiol 559: 685–706, 2004.<br />

40. Begenisich T, Nakamoto T, Ovitt CE, Nehrke K, Brugnara C,<br />

Alper SL, Melv<strong>in</strong> JE. <strong>Physiological</strong> roles of the <strong>in</strong>termediate<br />

conductance, Ca 2 -activated potassium channel Kcnn4. J Biol<br />

Chem 279: 47681–47687, 2004.<br />

41. Benham CD, Bolton TB. Spontaneous transient outward currents<br />

<strong>in</strong> s<strong>in</strong>gle visceral and vascualr smooth muscle cells of the rabbit.<br />

J Physiol 381: 385–406, 1986.<br />

42. Benham CD, Bolton TB, Lang RJ, Takewaki T. Calcium-activated<br />

potassium channels <strong>in</strong> s<strong>in</strong>gle smooth muscle cells of rabbit<br />

jejunum and gu<strong>in</strong>ea-pig mesenteric artery. J Physiol 371: 45–67,<br />

1986.<br />

43. Berchtold MW, Br<strong>in</strong>kmeier H, Muntener M. Calcium ion <strong>in</strong><br />

skeletal muscle: its crucial role for muscle function, plasticity, and<br />

disease. Physiol Rev 80: 1215–1265, 2000.<br />

44. Bergdahl A, Gomez MF, Wihlborg AK, Erl<strong>in</strong>ge D, Eyjolfson A,<br />

Xu SZ, Beech DJ, Dreja K, Hellstrand P. Plasticity of TRPC<br />

expression <strong>in</strong> arterial smooth muscle: correlation with store-operated<br />

Ca 2 entry. Am J Physiol Cell Physiol 288: C872–C880, 2005.<br />

45. Bergner A, Sanderson MJ. Acetylchol<strong>in</strong>e-<strong>in</strong>duced calcium signal<strong>in</strong>g<br />

and contraction of airway smooth muscle cells <strong>in</strong> lung slices.<br />

J Gen Physiol 119: 187–198, 2002.<br />

46. Bernardi P. Mitochondrial transport of cations: channels, exchangers,<br />

and permeability transition. Physiol Rev 79: 1127–1155,<br />

1999.<br />

47. Berra-Romani R, Mazzocco-Spezzia A, Pul<strong>in</strong>a MV, Golov<strong>in</strong>a<br />

VA. Ca 2 handl<strong>in</strong>g is altered when arterial myocytes progress from<br />

a contractile to a proliferative phenotype <strong>in</strong> culture. Am J Physiol<br />

Cell Physiol 295: C779–C790, 2008.<br />

48. Berridge MJ, Irv<strong>in</strong>e RF. Inositol trisphosphate, a novel second<br />

messenger <strong>in</strong> cellular signal transduction. Nature 312: 315–321,<br />

1984.<br />

49. Berridge MJ, Lipp P, Bootman MD. The versatility and universality<br />

of calcium signall<strong>in</strong>g. Nat Rev Mol Cell Biol 1: 11–21, 2000.<br />

50. Bers DM. Cardiac excitation-contraction coupl<strong>in</strong>g. Nature 415:<br />

198–205, 2002.<br />

51. Bers DM. Macromolecular complexes regulat<strong>in</strong>g cardiac ryanod<strong>in</strong>e<br />

receptor function. J Mol Cell Cardiol 37: 417–429, 2004.<br />

52. Bezprozvanny I, Watras J, Ehrlich BE. Bell-shaped calciumresponse<br />

curves of Ins(1,4,5)P 3- and calcium-gated channels from<br />

endoplasmic reticulum of cerebellum. Nature 351: 751–754, 1991.<br />

53. Bianchi K, Rimessi A, Prand<strong>in</strong>i A, Szabadkai G, Rizzuto R.<br />

Calcium and mitochondria: mechanisms and functions of a troubled<br />

relationship. Biochim Biophys Acta 1742: 119–131, 2004.<br />

54. Blatter LA. Depletion and fill<strong>in</strong>g of <strong>in</strong>tracellular calcium stores <strong>in</strong><br />

vascular smooth muscle. Cell Physiol 37: 503–512, 1995.<br />

55. Blatz AL, Magleby KL. S<strong>in</strong>gle apam<strong>in</strong>-blocked Ca-activated K <br />

channels of small conductance <strong>in</strong> cultured rat skeletal muscle.<br />

Nature 323: 718–720, 1996.<br />

56. Blauste<strong>in</strong> MP. Endogenous ouaba<strong>in</strong>: role <strong>in</strong> the pathogenesis of<br />

hypertension. Kidney Int 49: 1748–1753, 1996.<br />

57. Blauste<strong>in</strong> MP, Juhaszova M, Golov<strong>in</strong>a VA, Church PJ, Stanley<br />

EF. Na/Ca exchanger and PMCA localization <strong>in</strong> neurons and astrocytes:<br />

functional implications. Ann NY Acad Sci 976: 356–366,<br />

2002.<br />

58. Bobe R, Bredoux R, Corvazier E, Andersen JP, Clausen JD,<br />

Dode L, Kovacs T, Enouf J. Identification, expression, function,<br />

and localization of a novel (sixth) isoform of the human sarco/<br />

endoplasmic reticulum Ca 2 ATPase 3 gene. J Biol Chem 279:<br />

24297–24306, 2004.<br />

59. Bobe R, Bredoux R, Corvazier E, Lacabaratz-Porret C, Mart<strong>in</strong><br />

V, Kovacs T, Enouf J. How many Ca 2 ATPase isoforms are<br />

expressed <strong>in</strong> a cell type? A grow<strong>in</strong>g family of membrane prote<strong>in</strong>s<br />

illustrated by studies <strong>in</strong> platelets. Platelets 16: 133–150, 2005.<br />

60. Boitt<strong>in</strong> FX, Couss<strong>in</strong> F, Morel JL, Halet G, Macrez N, Mironneau<br />

J. Ca 2 signals mediated by Ins(1,4,5)P 3-gated channels <strong>in</strong> rat<br />

ureteric myocytes. Biochem J 349: 323–332, 2000.<br />

61. Boitt<strong>in</strong> FX, Galione A, Evans AM. Nicot<strong>in</strong>ic acid aden<strong>in</strong>e d<strong>in</strong>ucleotide<br />

phosphate mediates Ca 2 signals and contraction <strong>in</strong> arterial<br />

smooth muscle via a two-pool mechanism. Circ Res 91: 1168–<br />

1175, 2002.


62. Boitt<strong>in</strong> FX, Macrez N, Halet G, Mironneau J. Norep<strong>in</strong>ephr<strong>in</strong>e<strong>in</strong>duced<br />

Ca 2 waves depend on InsP 3 and ryanod<strong>in</strong>e receptor<br />

activation <strong>in</strong> vascular myocytes. Am J Physiol Cell Physiol 277:<br />

C139–C151, 1999.<br />

63. Bolot<strong>in</strong>a VM. Orai, STIM1 and iPLA2beta: a view from a different<br />

perspective. J Physiol 586: 3035–3042, 2008.<br />

64. Bolton TB. Mechanism of action of transmitters and other substances<br />

on smooth muscle. Physiol Rev 59: 606–718, 1979.<br />

65. Bolton TB, Gordienko DV, Povstyan OV, Harhun MI, Pucovsky<br />

V. <strong>Smooth</strong> muscle cells and <strong>in</strong>terstitial cells of blood vessels.<br />

Cell Calcium 35: 643–657, 2004.<br />

66. Bond M, Shuman H, Somlyo AP, Somlyo AV. Total cytoplasmic<br />

calcium <strong>in</strong> relaxed and maximally contracted rabbit portal ve<strong>in</strong><br />

smooth muscle. J Physiol 357: 185–201, 1984.<br />

67. Bond CT, Maylie J, Adelman JP. Small-conductance calciumactivated<br />

potassium channels. Ann NY Acad Sci 868: 370–378,<br />

1999.<br />

68. Bond CT, Sprengel R, Bissonnette JM, Kaufmann WA, Pribnow<br />

D, Neelands T, Storck T, Baetscher M, Jerecic J, Maylie<br />

J, Knaus HG, Seeburg PH, Adelman JP. Respiration and parturition<br />

affected by conditional overexpression of the Ca 2 -activated<br />

K channel subunit SK3. Science 289: 1942–1946, 2000.<br />

69. Bond M, Kitazawa T, Somlyo AP, Somlyo AV. Release and<br />

recycl<strong>in</strong>g of calcium by the sarcoplasmic reticulum <strong>in</strong> gu<strong>in</strong>ea-pig<br />

portal ve<strong>in</strong> smooth muscle. J Physiol 355: 677–695, 1984.<br />

70. Bonev AD, Jagger JH, Rubart M, Nelson MT. Activators of<br />

prote<strong>in</strong> k<strong>in</strong>ase C decreases Ca 2 spark frequency <strong>in</strong> smooth muscle<br />

cells from cerebral arteries. Am J Physiol Cell Physiol 273: C2090–<br />

C2095, 1997.<br />

71. Bootman MD, Missiaen L, Parys JB, De Smedt H, Casteels R.<br />

Control of <strong>in</strong>ositol 1,4,5-trisphosphate-<strong>in</strong>duced Ca 2 release by<br />

cytosolic Ca 2 . Biochem J 306: 445–451, 1995.<br />

72. Bootman MD, Niggli E, Berridge MJ, Lipp P. Imag<strong>in</strong>g the hieratchial<br />

Ca 2 signall<strong>in</strong>g system <strong>in</strong> HeLa cells. J Physiol 499: 307–314,<br />

1997.<br />

73. Borchman D, Simon R, Bicknell-Brown E. Variation <strong>in</strong> the lipid<br />

composition of rabbit muscle sarcoplasmic reticulum membrane<br />

with muscle type. J Biol Chem 257: 14136–14139, 1982.<br />

74. Borisova L, Wray S, Eisner DA, Burdyga T. How structure, Ca<br />

signals and cellular communications underlie function <strong>in</strong> precapillary<br />

vessels. Circ Res 105: 803–810, 2009.<br />

75. Borisova L, Wray S, Burdyga T. Insights <strong>in</strong>to the mechanisms<br />

controll<strong>in</strong>g Ca signall<strong>in</strong>g <strong>in</strong> myocytes of urter and ureteric microvessels.<br />

Ma<strong>in</strong> Meet<strong>in</strong>g of the <strong>Physiological</strong> Society, Glasgow,<br />

Scotland 2007, p. 239.<br />

76. Borisova L, Shmygol A, Wray S, Burdyga T. Evidence that a<br />

Ca 2 sparks/STOCs coupl<strong>in</strong>g mechanism is responsible for the<br />

<strong>in</strong>hibitory effect of caffe<strong>in</strong>e on electro-mechanical coupl<strong>in</strong>g <strong>in</strong><br />

gu<strong>in</strong>ea pig ureteric smooth muscle. Cell Calcium 42: 303–311, 2007.<br />

77. Bradley KN, Craig JW, Muir TC, McCarron JG. The sarcoplasmic<br />

reticulum and sarcolemma together form a passive Ca 2 trap <strong>in</strong><br />

colonic smooth muscle. Cell Calcium 36: 29–41, 2004.<br />

78. Bradley KN, Currie S, MacMillan D, Muir TC, McCarron JG.<br />

Cyclic ADP-ribose <strong>in</strong>creases Ca 2 removal <strong>in</strong> smooth muscle. J Cell<br />

Sci 116: 4291–4306, 2003.<br />

79. Bradley KN, Flynn ER, Muir TC, McCarron JG. Ca 2 regulation<br />

<strong>in</strong> gu<strong>in</strong>ea-pig colonic smooth muscle: the role of the Na -Ca 2<br />

exchanger and the sarcoplasmic reticulum. J Physiol 538: 465–482,<br />

2002.<br />

80. Bra<strong>in</strong>ard AM, Miller AJ, Martens JR, England SK. Maxi-K<br />

channels localize to caveolae <strong>in</strong> human myometrium: a role for an<br />

act<strong>in</strong>-channel-caveol<strong>in</strong> complex <strong>in</strong> the regulation of myometrial<br />

smooth muscle K current. Am J Physiol Cell Physiol 289: C49–<br />

C57, 2005.<br />

81. Brandes RP, Schmitz-W<strong>in</strong>nenthal FH, Feletou M, Godecke A,<br />

Huang PL, Vanhoutte PM, Flem<strong>in</strong>g I, Busse R. An endotheliumderived<br />

hyperpolariz<strong>in</strong>g factor dist<strong>in</strong>ct from NO and prostacycl<strong>in</strong> is<br />

a major endothelium-dependent vasodilator <strong>in</strong> resistance vessels of<br />

wild-type and endothelial NO synthase knockout mice. Proc Natl<br />

Acad Sci USA 97: 9747–9752, 2000.<br />

82. Brandman O, Liou J, Park WS, Meyer T. STIM2 is a feedback<br />

regulator that stabilizes basal cytosolic and endoplasmic reticulum<br />

Ca 2 levels. Cell 131: 1327–1339, 2007.<br />

SMOOTH MUSCLE SR 161<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

83. Brenner R, Perez GJ, Bonev AD, Eckman DM, Kosek JC,<br />

Wiler SW, Patterson AJ, Nelson MT, Aldrich RW. Vasoregulation<br />

by the B1 subunit of the calcium-activated potassium channel.<br />

Nature 407: 870–876, 2000.<br />

84. Britton FC, Ohya S, Horowitz B, Greenwood IA. Comparison<br />

of the properties of CLCA1 generated currents and I Cl(Ca) <strong>in</strong> mur<strong>in</strong>e<br />

portal ve<strong>in</strong> smooth muscle cells. J Physiol 539: 107–117, 2002.<br />

85. Brown DA, London E. <strong>Function</strong>s of lipid rafts <strong>in</strong> biological membranes.<br />

Annu Rev Cell Dev Biol 14: 111–136, 1998.<br />

86. Burdyga T, Wray S. <strong>Sarcoplasmic</strong> reticulum function and contractile<br />

consequences <strong>in</strong> ureteric smooth muscles. Novartis Found<br />

Symp 246: 208–217, 2002.<br />

87. Burdyga T, Wray S. Action potential refractory period <strong>in</strong> ureter<br />

smooth muscle is set by Ca sparks and BK channels. Nature 436:<br />

559–562, 2005.<br />

88. Burdyga T, Wray S, Noble K. In situ calcium signal<strong>in</strong>g: no calcium<br />

sparks detected <strong>in</strong> rat myometrium. Ann NY Acad Sci 1101:<br />

85–96, 2007.<br />

89. Burdyga TV, Magura IS. Effects of caffe<strong>in</strong>e on the electrical and<br />

mechanical activity of gu<strong>in</strong>ea-pig ureter smooth muscle. Gen<br />

Physiol Biophys 5: 581–591, 1986.<br />

90. Burdyga TV, Shmigol A, Eisner DA, Wray S. A new technique<br />

for simultaneous and <strong>in</strong> situ measurements of Ca signals <strong>in</strong> arteriolar<br />

smooth muscle and endothelial cells. Cell Calcium 34: 27–33,<br />

2003.<br />

91. Burdyga TV, Taggart MJ, Crichton C, Smith Gl, Wray S. The<br />

mechanism of Ca 2 release from the SR of permeabilised gu<strong>in</strong>eapig<br />

and rat ureteric smooth muscle. Biochim Biophys Acta 1402:<br />

109–114, 1998.<br />

92. Burdyga TV, Taggart MJ, Wray S. Major difference between rat<br />

and gu<strong>in</strong>ea-pig ureter <strong>in</strong> the ability of agonists and caffe<strong>in</strong>e to<br />

release Ca 2 and <strong>in</strong>fluence force. J Physiol 489: 327–335, 1995.<br />

93. Burdyga TV, Wray S. The effect of cyclopiazonic acid on excitation-contraction<br />

coupl<strong>in</strong>g <strong>in</strong> gu<strong>in</strong>ea-pig ureteric smooth muscle:<br />

role of the SR. J Physiol 517: 855–865, 1999.<br />

94. Burk SE, Lytton J, MacLennan DH, Shull GE. cDNA clon<strong>in</strong>g,<br />

functional expression, and mRNA tissue distribution of a third<br />

organellar Ca 2 pump. J Biol Chem 264: 18561–18568, 1989.<br />

95. Butler TM, Davies RE. High-energy phosphates <strong>in</strong> smooth muscle.<br />

In: Handbook of Physiology. The Cardiovascular System. Vascular<br />

<strong>Smooth</strong> <strong>Muscle</strong>. Bethesda, MD: Am. Physiol. Soc., 1980, sect.<br />

2, vol. II, chapt. 10, p. 237–252.<br />

96. Bygrave FL, Benedetti A. What is the concentration of calcium<br />

ions <strong>in</strong> the endoplasmic reticulum? Cell Calcium 19: 547–551, 1996.<br />

97. Byrne NG, Large WA. Action of noradrenal<strong>in</strong>e on s<strong>in</strong>gle smooth<br />

muscle cells freshly dispersed from the rat anococcygeus muscle.<br />

J Physiol 389: 513–525, 1987.<br />

98. Byrne NG, Large WA. Membrane mechanism associated with<br />

muscar<strong>in</strong>ic receptor activation <strong>in</strong> s<strong>in</strong>gle cells freshly dispersed<br />

from the rat anococcygeus muscle. Br J Pharmacol 92: 371–379,<br />

1987.<br />

99. Cahalan MD, Zhang SL, Yerom<strong>in</strong> AV, Ohlsen K, Roos J, Stauderman<br />

KA. Molecular basis of the CRAC channel. Cell Calcium<br />

42: 133–144, 2007.<br />

100. Calcraft PJ, Ruas M, Pan Z, Cheng X, Arredouani A, Hao X,<br />

Tang J, Rietdorf K, Teboul L, Chuang KT, L<strong>in</strong> P, Xiao R, Wang<br />

C, Zhu Y, L<strong>in</strong> Y, Wyatt CN, Parr<strong>in</strong>gton J, Ma J, Evans AM,<br />

Galione A, Zhu MX. NAADP mobilizes calcium from acidic organelles<br />

through two-pore channels. Nature 459: 596–600, 2009.<br />

101. Camello C, Lomax R, Petersen OH, Tepik<strong>in</strong> AV. Calcium leak<br />

from <strong>in</strong>tracellular stores–the enigma of calcium signall<strong>in</strong>g. Cell<br />

Calcium 32: 355–361, 2002.<br />

102. Campbell AM, Kessler PD, Fambrough DM. The alternative<br />

carboxyl term<strong>in</strong>i of avian cardiac and bra<strong>in</strong> sarcoplasmic reticulum/endoplasmic<br />

reticulum Ca 2 -ATPases are on opposite sides of<br />

the membrane. J Biol Chem 267: 9321–9325, 1992.<br />

103. Campbell KP, MacLennan DH, Jorgensen AO, M<strong>in</strong>tzer MC.<br />

Purification and characterization of calsequestr<strong>in</strong> from can<strong>in</strong>e cardiac<br />

sarcoplasmic reticulum and identification of the 53,000 dalton<br />

glycoprote<strong>in</strong>. J Biol Chem 258: 1197–1204, 1983.<br />

104. Cannell MB, Cheng H, Lederer WJ. The control of calcium<br />

release <strong>in</strong> heart muscle. Science 268: 1045–1049, 1995.


162 SUSAN WRAY AND THEODOR BURDYGA<br />

105. Carafoli E. Intracellular calcium homeostasis. Annu Rev Biochem<br />

56: 395–433, 1987.<br />

106. Carafoli E, Br<strong>in</strong>i M. Calcium pumps: structural basis for and<br />

mechanism of calcium transmembrane transport. Curr Op<strong>in</strong> Chem<br />

Biol 4: 152–161, 2000.<br />

107. Carl C, Lee HK, Sanders KM. Regulation of ion channels <strong>in</strong><br />

smooth muscles by calcium. Am J Physiol Cell Physiol 261: C9–<br />

C34, 1996.<br />

108. Caroppo R, Colella M, Colasuonno A, DeLuisi A, Debellis L,<br />

Curci S, Hofer AM. A reassessment of the effects of lum<strong>in</strong>al<br />

[Ca 2 ] on <strong>in</strong>ositol 1,4,5-trisphosphate-<strong>in</strong>duced Ca 2 release from<br />

<strong>in</strong>ternal stores. J Biol Chem 278: 39503–39508, 2003.<br />

109. Carter SG, Karl DW. Inorganic phosphate assay with malachite<br />

green: an improvement and evaluation. J Biochem Biophys Methods<br />

7: 7–13, 1982.<br />

110. Cart<strong>in</strong> L, Lounsbury KM, Nelson MT. Coupl<strong>in</strong>g of Ca 2 to CREB<br />

activation and gene expression <strong>in</strong> <strong>in</strong>tact cerebral arteries from<br />

mouse: roles of ryanod<strong>in</strong>e receptors and voltage-dependent Ca 2<br />

channels. Circ Res 86: 760–767, 2000.<br />

111. Casteels R, Droogmans G. Exchange characteristics of the noradrenal<strong>in</strong>e-sensitive<br />

calcium store <strong>in</strong> vascular smooth muscle of<br />

rabbit ear artery. J Physiol 317: 263–279, 1981.<br />

112. Caul<strong>in</strong>-Glaser T, Garcia-Cardena G, Sarrel P, Sessa WC,<br />

Bender JR. 17-Estradiol regulation of human endothelial cell<br />

basal nitric oxide release, <strong>in</strong>dependent of cytosolic Ca 2 mobilization.<br />

Circ Res 81: 885–892, 1997.<br />

113. Chadwick CC, Saito A, Fleischer S. Isolation and characterization<br />

of the <strong>in</strong>ositol trisphosphate receptor from smooth muscle.<br />

Proc Natl Acad Sci USA 87: 2132–2136, 1990.<br />

114. Chalmers S, McCarron JG. The mitochondrial membrane potential<br />

and Ca 2 oscillations <strong>in</strong> smooth muscle. J Cell Sci 121: 75–85,<br />

2008.<br />

115. Chambers P, Neal E, Gillespie JI. Ryanod<strong>in</strong>e receptors <strong>in</strong> human<br />

bladder smooth muscle. Exp Physiol 84: 41–46, 1999.<br />

116. Chamley-Campbell J, Campbell GR, Ross R. The smooth muscle<br />

cell <strong>in</strong> culture. Physiol Rev 59: 1–61, 1979.<br />

117. Chandra A, Angle N. VEGF <strong>in</strong>hibits PDGF-stimulated calcium<br />

signal<strong>in</strong>g <strong>in</strong>dependent of phospholipase C and prote<strong>in</strong> k<strong>in</strong>ase C.<br />

J Surg Res 131: 302–309, 2006.<br />

118. Chatton JY, Liu H, Stucki JW. Simultaneous measurements of<br />

Ca 2 <strong>in</strong> the <strong>in</strong>tracellular stores and the cytosol of hepatocytes<br />

dur<strong>in</strong>g hormone-<strong>in</strong>duced Ca 2 oscillations. FEBS Lett 368: 165–168,<br />

1995.<br />

119. Chen J, Petranka J, Yamamura K, London RE, Steenbergen C,<br />

Murphy E. Gender differences <strong>in</strong> sarcoplasmic reticulum calcium<br />

load<strong>in</strong>g after isoproterenol. Am J Physiol Heart Circ Physiol 285:<br />

H2657–H2662, 2003.<br />

120. Chen Q, van Breemen C. The superficial buffer barrier <strong>in</strong> venous<br />

smooth muscle: sarcoplasmic reticulum refill<strong>in</strong>g and unload<strong>in</strong>g.<br />

Br J Pharmacol 109: 336–343, 1993.<br />

121. Chen W, Steenbergen C, Levy LA, Vance J, London RE, Murphy<br />

E. Measurement of free Ca 2 <strong>in</strong> sarcoplasmic reticulum <strong>in</strong><br />

perfused rabbit heart loaded with 1,2-bis(2-am<strong>in</strong>o-5,6-difluorophenoxy)ethane-N,N,N,N-tetraacetic<br />

acid by 19 F NMR. J Biol Chem<br />

271: 7398–7403, 1996.<br />

122. Cheng H, Lederer WJ, Cannell MB. Calcium sparks: elementary<br />

events underly<strong>in</strong>g excitation-contraction coupl<strong>in</strong>g <strong>in</strong> heart muscle.<br />

Science 262: 740–744, 1993.<br />

123. Cheranov SY, Jaggar JH. <strong>Sarcoplasmic</strong> reticulum calcium load<br />

regulates rat arterial smooth muscle calcium sparks and transient<br />

K Ca currents. J Physiol 544: 71–84, 2002.<br />

124. Cheranov SY, Jaggar JH. Mitochondrial modulation of Ca 2<br />

sparks and transient KCa currents <strong>in</strong> smooth muscle cells of rat<br />

cerebral arteries. J Physiol 556: 755–771, 2004.<br />

125. Ch<strong>in</strong>i EN, Beers KW, Dousa TP. Nicot<strong>in</strong>ate aden<strong>in</strong>e d<strong>in</strong>ucleotide<br />

phosphate (NAADP) triggers a specific calcium release system <strong>in</strong><br />

sea urch<strong>in</strong> eggs. J Biol Chem 270: 3216–3223, 1995.<br />

126. Churchill GC, Okada Y, Thomas JM, Genazzani AA, Patel S,<br />

Galione A. NAADP mobilizes Ca 2 from reserve granules, lysosome-related<br />

organelles, and <strong>in</strong> sea urch<strong>in</strong> eggs. Cell 111: 703–708,<br />

2002.<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

127. Collier ML, Ji G, Wang Y, Kotlikoff MI. Calcium-<strong>in</strong>duced calcium<br />

release <strong>in</strong> smooth muscle: loose coupl<strong>in</strong>g between the action<br />

potential and calcium release. J Gen Physiol 115: 653–662, 2000.<br />

128. Coll<strong>in</strong>s TJ, Lipp P, Berridge MJ, Li W, Bootman MD. Inositol<br />

1,4,5-trisphosphate-<strong>in</strong>duced Ca 2 release is <strong>in</strong>hibited by mitochondrial<br />

depolarization. Biochem J 347: 593–600, 2000.<br />

129. Cornwell TL, Pryzwansky KB, Wyatt TA, L<strong>in</strong>coln TM. Regulation<br />

of sarcoplasmic reticulum prote<strong>in</strong> phosphorylation by localized<br />

cyclic GMP-dependent prote<strong>in</strong> k<strong>in</strong>ase <strong>in</strong> vascular smooth muscle<br />

cells. Mol Pharmacol 40: 923–931, 1991.<br />

130. Coronado R, Morrissette J, Sukhareva M, Vaughan DM. Structure<br />

and function of ryanod<strong>in</strong>e receptors. Am J Physiol Cell<br />

Physiol 266: C1485–C1504, 1994.<br />

131. Cortel<strong>in</strong>g RL, Brett SE, Y<strong>in</strong> H, Zheng XL, Walsh MP, Welsh<br />

DG. The functional consequence of RhoA knockdown by RNA<br />

<strong>in</strong>terference <strong>in</strong> rat cerebral arteries. Am J Physiol Heart Circ<br />

Physiol 293: H440–H447, 2007.<br />

132. Cortes SF, Lemos VS, Stoclet JC. Alterations <strong>in</strong> calcium stores<br />

<strong>in</strong> aortic myocytes from spontaneously hypertensive rats. Hypertension<br />

29: 1322–1328, 1997.<br />

133. Couss<strong>in</strong> F, Macrez N, Morel JL, Mironneau J. Requirement of<br />

ryanod<strong>in</strong>e receptor subtypes 1 and 2 for Ca 2 -<strong>in</strong>duced Ca 2 release<br />

<strong>in</strong> vascular myocytes. J Biol Chem 275: 9596–9603, 2000.<br />

134. Cox DH, Cui J, Aldrich RW. Allosteric gat<strong>in</strong>g of a large conductance<br />

Ca-activated K channel. J Gen Physiol 110: 257–281, 1997.<br />

135. Creed KE, Ishikawa S, Ito Y. Electrical and mechanical activity<br />

recorded from rabbit ur<strong>in</strong>ary bladder <strong>in</strong> response to nerve stimulation.<br />

J Physiol 338: 149–164, 1983.<br />

136. Cui J, Cox DH, Aldrich RW. Intr<strong>in</strong>sic voltage dependence and<br />

Ca 2 regulation of mslo large conductance Ca-activated K channels.<br />

J Gen Physiol 109: 647–673, 1997.<br />

137. Curtis TM, Tumelty J, Stewart MT, Arora AR, Lai FA, Mc-<br />

Gahon MK, Scholfield CN, McGeown JG. Modification of<br />

smooth muscle Ca 2 -sparks by tetraca<strong>in</strong>e: evidence for sequential<br />

RyR activation. Cell Calcium 43: 142–154, 2008.<br />

138. Dabertrand F, Morel JL, Sorrent<strong>in</strong>o V, Mironneau J, Mironneau<br />

C, Macrez N. Modulation of calcium signall<strong>in</strong>g by dom<strong>in</strong>ant<br />

negative splice variant of ryanod<strong>in</strong>e receptor subtype 3 <strong>in</strong> native<br />

smooth muscle cells. Cell Calcium 40: 11–21, 2006.<br />

139. Dabertrand F, Fritz N, Mironneau J, Macrez N, Morel JL. Role<br />

of RYR3 splice variants <strong>in</strong> calcium signall<strong>in</strong>g <strong>in</strong> mouse non-pregnant<br />

and pregnant myometrium. Am J Physiol Cell Physiol 293:<br />

C848–C854, 2007.<br />

140. Dai J, Kuo KH, Leo JM, van BC, Lee CH. Rearrangement of the<br />

close contact between the mitochondria and the sarcoplasmic<br />

reticulum <strong>in</strong> airway smooth muscle. Cell Calcium 37: 333–340,<br />

2005.<br />

141. Dally S, Bredoux R, Corvazier E, Andersen JP, Clausen JD,<br />

Dode L, Fanchaouy M, Gelebart P, Monceau V, Del MF,<br />

Gwathmey JK, Hajjar R, Chaabane C, Bobe R, Raies A, Enouf<br />

J. Ca 2 -ATPases <strong>in</strong> non-fail<strong>in</strong>g and fail<strong>in</strong>g heart: evidence for a<br />

novel cardiac sarco/endoplasmic reticulum Ca 2 -ATPase 2 isoform<br />

(SERCA2c). Biochem J 395: 249–258, 2006.<br />

142. Daniel EE, Jury J, Wang YF. nNOS <strong>in</strong> can<strong>in</strong>e lower esophageal<br />

sph<strong>in</strong>cter: colocalized with Cav-1 and Ca 2 -handl<strong>in</strong>g prote<strong>in</strong>s?<br />

Am J Physiol Gastro<strong>in</strong>test Liver Physiol 281: G1101–G1114, 2001.<br />

143. Darby PJ, Kwan CY, Daniel EE. Caveolae from can<strong>in</strong>e airway<br />

smooth muscle conta<strong>in</strong> the necessary components for a role <strong>in</strong><br />

Ca 2 handl<strong>in</strong>g. Am J Physiol Lung Cell Mol Physiol 279: L1226–<br />

L1235, 2000.<br />

144. Dash R, Frank KF, Carr AN, Moravec CS, Kranias EG. Gender<br />

<strong>in</strong>fluences on sarcoplasmic reticulum Ca 2 -handl<strong>in</strong>g <strong>in</strong> fail<strong>in</strong>g human<br />

myocardium. J Mol Cell Cardiol 33: 1345–1353, 2001.<br />

145. Daub B, Ganitkevich VY. An estimate of rapid cytoplasmic calcium<br />

buffer<strong>in</strong>g <strong>in</strong> a s<strong>in</strong>gle smooth muscle cell. Cell Calcium 27:<br />

3–13, 2000.<br />

146. De Smet P, Parys JB, Vanl<strong>in</strong>gen S, Bultynck G, Callewaert G,<br />

Galione A, De Smedt H, Missiaen L. The relative order of IP 3<br />

sensitivity of types 1 and 3 IP 3 receptors is pH dependent. Pflügers<br />

Arch 438: 154–158, 1999.<br />

147. Debbas G, Hoffman L, Landon EJ, Hurwitz L. Electron microscopic<br />

localization of calcium <strong>in</strong> vascular smooth muscle. Anat Rec<br />

182: 447–471, 1975.


148. Dehaven WI, Jones BF, Petranka JG, Smyth JT, Tomita T,<br />

Bird GS, Putney JW Jr. TRPC channels function <strong>in</strong>dependently of<br />

STIM1 and Orai1. J Physiol 587: 2275–2298, 2009.<br />

149. Del Valle-Rodriguez A, Calderon E, Ruiz M, Ordonez A,<br />

Lopez-Barneo J, Urena J. Metabotropic Ca 2 channel-<strong>in</strong>duced<br />

Ca 2 release and ATP-dependent facilitation of arterial myocyte<br />

contraction. Proc Natl Acad Sci USA 103: 4316–4321, 2006.<br />

150. Desilets M, Driska SP, Baumgarten CM. Current fluctuations<br />

and oscillations <strong>in</strong> smooth muscle cells from hog carotid artery.<br />

Role of the sarcoplasmic reticulum. Circ Res 65: 708–722, 1989.<br />

151. Dettbarn C, Palade P. Effects of alkal<strong>in</strong>e pH on sarcoplasmic<br />

reticulum Ca 2 release and Ca 2 uptake. J Biol Chem 266: 8993–<br />

9001, 1991.<br />

152. Dev<strong>in</strong>e CE, Somlyo AV, Somlyo AP. <strong>Sarcoplasmic</strong> reticulum and<br />

excitation-contraction coupl<strong>in</strong>g <strong>in</strong> mammalian smooth muscles.<br />

J Cell Biol 52: 690–718, 1972.<br />

153. Dietrich A, Kalwa H, Storch U, Schnitzler M, Salanova B,<br />

P<strong>in</strong>kenburg O, Dubrovska G, Ess<strong>in</strong> K, Gollasch M, Birnbaumer<br />

L, Gudermann T. Pressure-<strong>in</strong>duced and store-operated<br />

cation <strong>in</strong>flux <strong>in</strong> vascular smooth muscle cells is <strong>in</strong>dependent of<br />

TRPC1. Pflügers Arch 455: 465–477, 2007.<br />

154. Dreja K, Nordstrom I, Hellstrand P. Rat arterial smooth muscle<br />

devoid of ryanod<strong>in</strong>e receptor function: effects on cellular Ca 2<br />

handl<strong>in</strong>g. Br J Pharmacol 132: 1957–1966, 2001.<br />

155. Dreja K, Voldstedlund M, V<strong>in</strong>ten J, Tranum-Jensen J, Hellstrand<br />

P, Sward K. Cholesterol depletion disrupts caveolae and<br />

differentially impairs agonist-<strong>in</strong>duced arterial contraction. Arterioscler<br />

Thromb Vasc Biol 22: 1267–1272, 2002.<br />

156. Drummond RM, Fay FS. Mitochondria contribute to Ca 2 removal<br />

<strong>in</strong> smooth muscle cells. Pflügers Arch 431: 473–482, 1996.<br />

157. Drummond RM, Tuft RA. Release of Ca 2 from the sarcoplasmic<br />

reticulum <strong>in</strong>creases mitochondrial [Ca 2 ] <strong>in</strong> rat pulmonary artery<br />

smooth muscle cells. J Physiol 516: 139–148, 1999.<br />

158. Du W, Stiber JA, Rosenberg PB, Meissner G, Eu JP. Ryanod<strong>in</strong>e<br />

receptors <strong>in</strong> muscar<strong>in</strong>ic receptor-mediated bronchoconstriction.<br />

J Biol Chem 280: 26287–26294, 2005.<br />

159. Duchen MR, Leyssens A, Crompton M. Transient mitochondrial<br />

depolarizations reflect focal sarcoplasmic reticular calcium release<br />

<strong>in</strong> s<strong>in</strong>gle rat cardiomyocites. J Cell Biol 142: 975–988, 2000.<br />

160. Duquette RA, Shmygol A, Vaillant C, Mobasheri A, Pope M,<br />

Burdyga T, Wray S. Viment<strong>in</strong>-positive, c-kit-negative <strong>in</strong>terstitial<br />

cells <strong>in</strong> human and rat uterus: a role <strong>in</strong> pacemak<strong>in</strong>g? Biol Reprod<br />

72: 276–283, 2005.<br />

161. Ebashi F, Ebashi S. Removal of calcium and relaxation <strong>in</strong> actomyos<strong>in</strong><br />

systems. Nature 194: 378–379, 1962.<br />

162. Eggermont JA, Wuytack F, Verbist J, Casteels R. Expression<br />

of endoplasmic-reticulum Ca 2 -pump isoforms and of phospholamban<br />

<strong>in</strong> pig smooth-muscle tissues. Biochem J 271: 649–653, 1990.<br />

163. Elble RC, Ju G, Nehrke K, DeBiasio J, K<strong>in</strong>gsley PD, Kotlikoff<br />

MI, Pauli BU. Molecular and functional characterization of a<br />

mur<strong>in</strong>e calcium-activated chloride channel expressed <strong>in</strong> smooth<br />

muscle. J Biol Chem 277: 18586–18591, 2002.<br />

164. Ellgaard L, Helenius A. Quality control <strong>in</strong> the endoplasmic reticulum.<br />

Nat Rev Mol Cell Biol 4: 181–191, 2003.<br />

165. Elmoselhi AB, Blennerhassett M, Samson SE, Grover AK.<br />

Properties of the sarcoplasmic reticulum Ca 2 -pump <strong>in</strong> coronary<br />

artery sk<strong>in</strong>ned smooth muscle. Mol Cell Biochem 151: 149–155,<br />

1995.<br />

166. Elmoselhi AB, Samson SE, Grover AK. SR Ca 2 pump heterogeneity<br />

<strong>in</strong> coronary artery: free radicals and IP 3-sensitive and -<strong>in</strong>sensitive<br />

pools. Am J Physiol Cell Physiol 271: C1652–C1659, 1996.<br />

167. Endo M. Calcium ion as a second messenger with special reference<br />

to excitation-contraction coupl<strong>in</strong>g. J Pharmacol Sci 100: 519–<br />

524, 2006.<br />

168. Entman ML, Keslensky SS, Chu A, Van W<strong>in</strong>kle WB. The sarcoplasmic<br />

reticulum-glycogenolytic complex <strong>in</strong> mammalian fast<br />

twitch skeletal muscle. Proposed <strong>in</strong> vitro counterpart of the contraction-activated<br />

glycogenolytic pool. J Biol Chem 255: 6245–<br />

6252, 1980.<br />

169. Ethier MF, Madison JM. LY294002, but not wortmann<strong>in</strong>, <strong>in</strong>creases<br />

<strong>in</strong>tracellular calcium and <strong>in</strong>hibits calcium transients <strong>in</strong><br />

bov<strong>in</strong>e and human airway smooth muscle cells. Cell Calcium 32:<br />

31–38, 2002.<br />

SMOOTH MUSCLE SR 163<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

170. Etter EF, M<strong>in</strong>ta A, Poenie M, Fay FS. Near-membrane [Ca 2 ]<br />

transients resolved us<strong>in</strong>g the Ca <strong>in</strong>dicator FFP18. Proc Natl Acad<br />

Sci USA 93: 5368–5373, 1996.<br />

171. Fajmut A, Brumen M, Schuster S. Theoretical model of the<br />

<strong>in</strong>teractions between Ca 2 , calmodul<strong>in</strong> and myos<strong>in</strong> light cha<strong>in</strong><br />

k<strong>in</strong>ase. FEBS Lett 579: 4361–4366, 2005.<br />

172. Ferguson DG, Young EF, Raeymaekers L, Kranias EG. Localization<br />

of phospholamban <strong>in</strong> smooth muscle us<strong>in</strong>g immunogold<br />

electron microscopy. J Cell Biol 107: 555–562, 1988.<br />

173. Feske S. Calcium signall<strong>in</strong>g <strong>in</strong> lymphocyte activation and disease.<br />

Nat Rev Immunol 7: 690–702, 2007.<br />

174. Feske S, Gwack Y, Prakriya M, Srikanth S, Puppel SH, Tanasa<br />

B, Hogan PG, Lewis RS, Daly M, Rao A. A mutation <strong>in</strong> Orai1<br />

causes immune deficiency by abrogat<strong>in</strong>g CRAC channel function.<br />

Nature 441: 179–185, 2006.<br />

175. Filipp<strong>in</strong> L, Magalhaes PJ, Di BG, Colella M, Pozzan T. Stable<br />

<strong>in</strong>teractions between mitochondria and endoplasmic reticulum allow<br />

rapid accumulation of calcium <strong>in</strong> a subpopulation of mitochondria.<br />

J Biol Chem 278: 39224–39234, 2003.<br />

176. F<strong>in</strong>k CC, Slepchenko B, Loew LM. Determ<strong>in</strong>ation of time-dependent<br />

<strong>in</strong>ositol-1,4,5-trisphosphate concentrations dur<strong>in</strong>g calcium release<br />

<strong>in</strong> a smooth muscle cell. Biophys J 77: 617–628, 1999.<br />

177. Fischer H, Fischer J, Boknik P, Gergs U, Schmitz W, Domschke<br />

W, Konturek JW, Neumann J. Reduced expression of<br />

Ca 2 -regulat<strong>in</strong>g prote<strong>in</strong>s <strong>in</strong> the upper gastro<strong>in</strong>test<strong>in</strong>al tract of patients<br />

with achalasia. World J Gastroenterol 12: 6002–6007, 2006.<br />

178. Fleischmann BK, Wang YX, Pr<strong>in</strong>g M, Kotlikoff MI. Voltagedependent<br />

calcium currents and cytosolic calcium <strong>in</strong> equ<strong>in</strong>e airway<br />

myocytes. J Physiol 492: 347–358, 1996.<br />

179. Flem<strong>in</strong>g I. Realiz<strong>in</strong>g its potential: the <strong>in</strong>termediate conductance<br />

Ca 2 -activated K channel (KCa3.1) and the regulation of blood<br />

pressure. Circ Res 99: 462–464, 2006.<br />

180. Fliegel L, Burns K, MacLennan DH, Reithmeier RA, Michalak<br />

M. Molecular clon<strong>in</strong>g of the high aff<strong>in</strong>ity calcium-b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong><br />

(calreticul<strong>in</strong>) of skeletal muscle sarcoplasmic reticulum. J Biol<br />

Chem 264: 21522–21528, 1989.<br />

181. Fliegel L, Leberer E, Green NM, MacLennan DH. The fasttwitch<br />

muscle calsequestr<strong>in</strong> isoform predom<strong>in</strong>ates <strong>in</strong> rabbit slowtwitch<br />

soleus muscle. FEBS Lett 242: 297–300, 1989.<br />

182. Fliegel L, Ohnishi M, Carpenter MR, Khanna VK, Reithmeier<br />

RA, MacLennan DH. Am<strong>in</strong>o acid sequence of rabbit fast-twitch<br />

skeletal muscle calsequestr<strong>in</strong> deduced from cDNA and peptide<br />

sequenc<strong>in</strong>g. Proc Natl Acad Sci USA 84: 1167–1171, 1987.<br />

183. Floyd R, Mobasheri A, Mart<strong>in</strong>-Vasallo P, Wray S. Na,K-ATPase<br />

isoforms <strong>in</strong> pregnant and nonpregnant rat uterus. Ann NY Acad Sci<br />

986: 614–616, 2003.<br />

184. Floyd R, Wray S. Calcium transporters and signall<strong>in</strong>g <strong>in</strong> smooth<br />

muscles. Cell Calcium 42: 467–476, 2007.<br />

185. Flynn ERM, Bradleyt KN, Muir TC, McCarron JG. <strong>Function</strong>ally<br />

separate <strong>in</strong>tracellular Ca 2 stores <strong>in</strong> smooth muscle. J Biol Chem<br />

276: 36411–36418, 2001.<br />

186. Frank PG, Hassan GS, Rodriguez-Feo JA, Lisanti MP. Caveolae<br />

and caveol<strong>in</strong>-1: novel potential targets for the treatment of<br />

cardiovascular disease. Curr Pharm Des 13: 1761–1769, 2007.<br />

187. Franz<strong>in</strong>i-Armstrong C, Kenney LJ, Varriano-Marston E. The<br />

structure of calsequestr<strong>in</strong> <strong>in</strong> triads of vertebrate skeletal muscle: a<br />

deep-etch study. J Cell Biol 105: 49–56, 1987.<br />

188. Freichel M, Vennekens R, Olausson J, Hoffmann M, Muller C,<br />

Stolz S, Scheunemann J, Weissgerber P, Flockerzi V. <strong>Function</strong>al<br />

role of TRPC prote<strong>in</strong>s <strong>in</strong> vivo: lessons from TRPC-deficient<br />

mouse models. Biochem Biophys Res Commun 322: 1352–1358,<br />

2004.<br />

189. Frischauf I, Sch<strong>in</strong>dl R, Derler I, Bergsmann J, Fahrner M,<br />

Roman<strong>in</strong> C. The STIM/Orai coupl<strong>in</strong>g mach<strong>in</strong>ery. Channels 2: 261–<br />

268, 2008.<br />

190. Fujii K, Foster CD, Brad<strong>in</strong>g AF, Parekh AB. Potassium channel<br />

blockers and the effects of cromakalim on the smooth muscle of<br />

the gu<strong>in</strong>ea-pig bladder. Br J Pharmacol 99: 779–785, 1990.<br />

191. Fujimoto T, Nakade S, Miyawaki K, Ogawa K. Localization of<br />

<strong>in</strong>ositol 1,4,5-trisphosphate receptor-like prote<strong>in</strong> <strong>in</strong> plasmalemmal<br />

caveolae. J Cell Biol 119: 1507–1513, 1992.


164 SUSAN WRAY AND THEODOR BURDYGA<br />

192. Furstenau M, Lohn M, Reid C, Luft FC, Haller H, Gollasch M.<br />

Calcium sparks <strong>in</strong> human coronary artery smooth muscle cells<br />

resolved by confocal imag<strong>in</strong>g. J Hypertens 18: 1215–1222, 2000.<br />

193. Furuichi T, Kohda K, Miyawaki A, Mikoshiba K. Intracellular<br />

channels. Curr Op<strong>in</strong> Neurobiol 4: 294–303, 1994.<br />

194. Gabella G. Caveolae <strong>in</strong>tracellulaires and sarcoplasmic reiculum <strong>in</strong><br />

smooth muscle. J Cell Sci 8: 601–609, 1971.<br />

195. Gabella G. Structure of smooth muscle. In: <strong>Smooth</strong> <strong>Muscle</strong>: An<br />

Assessment of Current Knowledge, edited by Bulbr<strong>in</strong>g E, Brad<strong>in</strong>g<br />

AF, Jones AW and Tomita T. London: Arnold, 1981.<br />

196. Gabella G. Structural apparatus for force transmission <strong>in</strong> smooth<br />

muscles. Physiol Rev 64: 455–477, 1984.<br />

197. Gabella G. Structure of smooth muscles. In: Pharmacology of<br />

<strong>Smooth</strong> <strong>Muscle</strong>, edited by Szekeres L and Papp LG. Berl<strong>in</strong>: Spr<strong>in</strong>ger-<br />

Verlag, 1994.<br />

198. Gafni A, Yuh KC. A comparative study of the Ca 2 -Mg 2 dependent<br />

ATPase from skeletal muscles of young, adult and old rats.<br />

Mech Age<strong>in</strong>g Dev 49: 105–117, 1989.<br />

199. Gafni J, Munsch JA, Lam TH, Catl<strong>in</strong> MC, Costa LG, Mol<strong>in</strong>ski<br />

TF, Pessah IN. Xestospong<strong>in</strong>s: potent membrane permeable<br />

blockers of the <strong>in</strong>ositol 1,4,5-trisphosphate receptor. Neuron 19:<br />

723–733, 1997.<br />

200. Gambara G, Bill<strong>in</strong>gton RA, Debidda M, D’Alessio A, Palombi<br />

F, Ziparo E, Genazzani AA, Filipp<strong>in</strong>i A. NAADP-<strong>in</strong>duced Ca 2<br />

signal<strong>in</strong>g <strong>in</strong> response to endothel<strong>in</strong> is via the receptor subtype B<br />

and requires the <strong>in</strong>tegrity of lipid rafts/caveolae. J Cell Physiol 216:<br />

396–404, 2008.<br />

201. Ganitkevich V, Hasse V, Pfitzer G. Ca 2 -dependent and Ca 2 -<br />

<strong>in</strong>dependent regulation of smooth muscle contraction. J <strong>Muscle</strong><br />

Res Cell Motil 23: 47–52, 2002.<br />

202. Ganitkevich V, Isenberg G. Isolated gu<strong>in</strong>ea pig coronary smooth<br />

muscle cells. Acetylchol<strong>in</strong>e <strong>in</strong>duces hyperpolarization due to sarcoplasmic<br />

reticulum calcium release activat<strong>in</strong>g potassium channels.<br />

Circ Res 67: 525–528, 1990.<br />

203. Ganitkevich VY, Isenberg G. Contribution of calcium <strong>in</strong>duced<br />

calcium release to [Ca 2 ] i transients <strong>in</strong> myocytes from gu<strong>in</strong>ea-pig<br />

ur<strong>in</strong>ary bladder. J Physiol 458: 119–137, 1992.<br />

204. Ganitkevich VY. The amount of acetylchol<strong>in</strong>e mobilisable Ca 2 <strong>in</strong><br />

s<strong>in</strong>gle smooth muscle cells measured with the exogenous cytoplasmic<br />

Ca 2 buffer, Indo-1. Cell Calcium 20: 483–492, 1996.<br />

205. Gayan-Ramirez G, Vanzeir L, Wuytack F, Decramer M. Corticosteroids<br />

decrease mRNA levels of SERCA pumps, whereas they<br />

<strong>in</strong>crease sarcolip<strong>in</strong> mRNA <strong>in</strong> the rat diaphragm. J Physiol 524:<br />

387–397, 2000.<br />

206. Gebremedh<strong>in</strong> D, Kaldunski M, Jacobs ER, Harder DR, Roman<br />

RJ. Coexistence of two types of Ca 2 -activated K channels <strong>in</strong> rat<br />

renal arterioles. Am J Physiol Renal Fluid Electrolyte Physiol 270:<br />

F69–F81, 1996.<br />

207. Gelebart P, Mart<strong>in</strong> V, Enouf J, Papp B. Identification of a new<br />

SERCA2 splice variant regulated dur<strong>in</strong>g monocytic differentiation.<br />

Biochem Biophys Res Commun 303: 676–684, 2003.<br />

208. Gerasimenko OV, Gerasimenko JV, Tepik<strong>in</strong> AV, Petersen OH.<br />

ATP-dependent accumualtion and <strong>in</strong>ositol trisphosphate- or cyclic<br />

ADP-ribose mediated release of Ca 2 from the nuclear envelope.<br />

Cell 80: 1–20, 1995.<br />

209. Ghisdal P, Vandenberg G, Morel N. Rho-dependent k<strong>in</strong>ase is<br />

<strong>in</strong>volved <strong>in</strong> agonist-activated calcium entry <strong>in</strong> rat arteries. J Physiol<br />

551: 855–867, 2003.<br />

210. Giach<strong>in</strong>i FR, Chiao CW, Carneiro FS, Lima VV, Carneiro ZN,<br />

Dorrance AM, Tostes RC, Webb RC. Increased activation of<br />

stromal <strong>in</strong>teraction molecule-1/Orai-1 <strong>in</strong> aorta from hypertensive<br />

rats. A novel <strong>in</strong>sight <strong>in</strong>to vascular dysfunction. Hypertension 53:<br />

409–416, 2009.<br />

211. Giann<strong>in</strong>i G, Conti A, Mammarella S, Scrobogna M, Sorrent<strong>in</strong>o<br />

V. The ryanod<strong>in</strong>e receptor/calcium channel genes are widely and<br />

differentially expressed <strong>in</strong> mur<strong>in</strong>e bra<strong>in</strong> and peripheral tissues.<br />

J Cell Biol 128: 893–904, 1995.<br />

212. Goldbeter A, Dupont G, Berridge MJ. M<strong>in</strong>imal model for signal<strong>in</strong>duced<br />

Ca 2 oscillations and for their frequency encod<strong>in</strong>g through<br />

prote<strong>in</strong> phosphorylation. Proc Natl Acad Sci USA 87: 1461–1465,<br />

1990.<br />

213. Gollasch M, Wellman GC, Knot HJ, Jaggar JH, Damon DH,<br />

Bonev AD, Nelson MT. Ontogeny of local sarcoplasmic reticulum<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

Ca 2 signals <strong>in</strong> cerebral arteries: Ca 2 sparks as elementary physiological<br />

events. Circ Res 83: 1104–1114, 1998.<br />

214. Golov<strong>in</strong>a VA, Blauste<strong>in</strong> MP. Spatially and functionally dist<strong>in</strong>ct<br />

Ca 2 stores <strong>in</strong> sarcoplasmic and endoplasmic reticulum. Science<br />

275: 1643–1648, 1997.<br />

215. Golov<strong>in</strong>a VA, Blauste<strong>in</strong> MP. Unload<strong>in</strong>g and refill<strong>in</strong>g of two<br />

classes of spatially resolved endoplasmic reticulum Ca 2 stores <strong>in</strong><br />

astrocytes. Glia 31: 15–28, 2000.<br />

216. Golov<strong>in</strong>a VA, Platoshyn O, Bailey CL, Wang J, Limsuwan A,<br />

Sweeney M, Rub<strong>in</strong> LJ, Yuan JX. Upregulated TRP and enhanced<br />

capacitative Ca 2 entry <strong>in</strong> human pulmonary artery myocytes dur<strong>in</strong>g<br />

proliferation. Am J Physiol Heart Circ Physiol 280: H746–<br />

H755, 2001.<br />

217. Gomez MF, Stevenson AS, Bonev AD, Hill-Eubanks DC, Nelson<br />

MT. Oppos<strong>in</strong>g actions of <strong>in</strong>ositol 1,4,5-trisphosphate and ryanod<strong>in</strong>e<br />

receptors on nuclear factor of activated T-cells regulation<br />

<strong>in</strong> smooth muscle. J Biol Chem 277: 37756–37764, 2002.<br />

218. Gomez-Viquez L, Guerrero-Serna G, Garcia U, Guerrero-Hernandez<br />

A. SERCA pump optimizes Ca 2 release by a mechanism<br />

<strong>in</strong>dependent of store fill<strong>in</strong>g <strong>in</strong> smooth muscle cells. Biophys J 85:<br />

370–380, 2003.<br />

219. Gordienko DV, Bolton TB. Crosstalk between ryanod<strong>in</strong>e receptors<br />

and IP 3 receptors as a factor shap<strong>in</strong>g spontaneous Ca 2 -<br />

release events <strong>in</strong> rabbit portal ve<strong>in</strong> myocytes. J Physiol 542: 743–<br />

762, 2002.<br />

220. Gordienko DV, Bolton TB, Cannell MB. Variability <strong>in</strong> spontaneous<br />

subcellular calcium release <strong>in</strong> gu<strong>in</strong>ea-pig ileum smooth muscle<br />

cells. J Physiol 507: 707–720, 1998.<br />

221. Gordienko DV, Greenwood IA, Bolton TB. Direct visualization<br />

of sarcoplasmic reticulum regions discharg<strong>in</strong>g Ca 2 sparks <strong>in</strong> vascular<br />

myocytes. Cell Calcium 29: 13–28, 2001.<br />

222. Gordienko DV, Harhun MI, Kustov MV, Pucovsky V, Bolton<br />

TB. Sub-plasmalemmal [Ca 2 ] i upstroke <strong>in</strong> myocytes of the gu<strong>in</strong>eapig<br />

small <strong>in</strong>test<strong>in</strong>e evoked by muscar<strong>in</strong>ic stimulation: IP 3R-mediated<br />

Ca 2 release <strong>in</strong>duced by voltage-gated Ca 2 entry. Cell Calcium<br />

43: 122–141, 2008.<br />

223. Gordienko DV, Zholos AV, Bolton TB. Membrane ion channels<br />

as physiological targets for local Ca 2 signall<strong>in</strong>g. J Microsc 196:<br />

305–316, 1999.<br />

224. Grayson TH, Haddock RE, Murray TP, Wojcikiewicz RJ, Hill<br />

CE. Inositol 1,4,5-trisphosphate receptor subtypes are differentially<br />

distributed between smooth muscle and endothelial layers of<br />

rat arteries. Cell Calcium 36: 447–458, 2004.<br />

225. Greenwood IA, Helliwell RM, Large WA. Modulation of Ca 2 -<br />

activated Cl currents <strong>in</strong> rabbit portal ve<strong>in</strong> smooth muscle by an<br />

<strong>in</strong>hibitor of mitochondrial Ca 2 uptake. J Physiol 505: 53–64, 1997.<br />

226. Greenwood IA, Miller LJ, Ohya S, Horowitz B. The large conductance<br />

potassium channel beta-subunit can <strong>in</strong>teract with and<br />

modulate the functional properties of a calcium-activated chloride<br />

channel, CLCA1. J Biol Chem 277: 22119–22122, 2002.<br />

227. Grissmer S, Lewis RS, Cahalan MD. Ca 2 -activated K channels<br />

<strong>in</strong> human leukemic T cells. J Gen Physiol 99: 63–84, 1992.<br />

228. Grover AK, Samson SE. Coronary artery acidosis: pH and calcium<br />

pump stability. Am J Physiol Heart Circ Physiol 265: H1486–<br />

H1492, 1993.<br />

229. Grover AK, Xu A, Samson SE, Narayanan N. <strong>Sarcoplasmic</strong><br />

reticulum Ca 2 pump <strong>in</strong> pig coronary artery smooth muscle is<br />

regulated by a novel pathway. Am J Physiol Cell Physiol 271:<br />

C181–C187, 1996.<br />

230. Guerrero A, S<strong>in</strong>ger JJ, Fay FS. Simultaneous measurement of<br />

Ca 2 release and <strong>in</strong>flux <strong>in</strong>to smooth muscle cells <strong>in</strong> response to<br />

caffe<strong>in</strong>e. J Gen Physiol 104: 395–422, 1994.<br />

231. Guerrero-Hernandez A, Gomez-Viquez L, Guerrero-Serna G,<br />

Rueda A. Ryanod<strong>in</strong>e receptors <strong>in</strong> smooth muscle. Front Biosci 7:<br />

d1676–d1688, 2002.<br />

232. Guillemette G, Segui JA. Effects of pH, reduc<strong>in</strong>g and alkylat<strong>in</strong>g<br />

reagents on the b<strong>in</strong>d<strong>in</strong>g and Ca 2 release activities of <strong>in</strong>ositol<br />

1,4,5-triphosphate <strong>in</strong> the bov<strong>in</strong>e adrenal cortex. Mol Endocr<strong>in</strong>ol 2:<br />

1249–1255, 1988.<br />

233. Guo W, Campbell KP. Association of triad<strong>in</strong> with the ryanod<strong>in</strong>e<br />

receptor and calsequestr<strong>in</strong> <strong>in</strong> the lumen of the sarcoplasmic reticulum.<br />

J Biol Chem 270: 9027–9030, 1995.


234. Gurney AM, Hunter E. The use of small <strong>in</strong>terfer<strong>in</strong>g RNA to<br />

elucidate the activity and function of ion channel genes <strong>in</strong> an <strong>in</strong>tact<br />

tissue. J Pharmacol Toxicol Methods 51: 253–262, 2005.<br />

235. Guse AH. Biochemistry, biology, and pharmacology of cyclic adenos<strong>in</strong>e<br />

diphosphoribose (cADPR). Curr Med Chem 11: 847–855,<br />

2004.<br />

236. Gyorke S, Lukyanenko V, Gyorke I. Dual effects of tetraca<strong>in</strong>e on<br />

spontaneous calcium release <strong>in</strong> rat ventricular myocytes. J Physiol<br />

500: 297–309, 1997.<br />

237. Gyorke S, Terentyev D. Modulation of ryanod<strong>in</strong>e receptor by<br />

lum<strong>in</strong>al calcium and accessory prote<strong>in</strong>s <strong>in</strong> health and cardiac disease.<br />

Cardiovasc Res 77: 245–255, 2008.<br />

238. Gyorke S, Velez P, Suarez-Isla B, Fill M. Activation of s<strong>in</strong>gle<br />

cardiac and skeletal ryanod<strong>in</strong>e receptor channels by flash photolysis<br />

of caged Ca 2 . Biophys J 66: 1879–1886, 1994.<br />

239. Haddock RE, Hill CE. Differential activation of ion channels by<br />

<strong>in</strong>ositol 1,4,5-trisphosphate (IP 3)- and ryanod<strong>in</strong>e-sensitive calcium<br />

stores <strong>in</strong> rat basilar artery vasomotion. J Physiol 545: 615–627,<br />

2002.<br />

240. Hagar RE, Burgstahler AD, Nathanson MH, Ehrlich BE. Type<br />

III InsP 3 receptor channel stays open <strong>in</strong> the presence of <strong>in</strong>creased<br />

calcium. Nature 396: 81–84, 1998.<br />

241. Hajnoczky G, Csordas G, Madesh M, Pacher P. The mach<strong>in</strong>ery<br />

of local Ca 2 signall<strong>in</strong>g between sarco-endoplasmic reticulum and<br />

mitochondria. J Physiol 529: 69–81, 2000.<br />

242. Hajnoczky G, Hager R, Thomas AP. Mitochondria suppress local<br />

feedback activation of <strong>in</strong>ositol 1,4,5-trisphosphate receptors by<br />

Ca 2 . J Biol Chem 274: 14157–14162, 1999.<br />

243. Hakamata Y, Nakai J, Takeshima H, Imoto K. Primary structure<br />

and distribution of a novel ryanod<strong>in</strong>e receptor/calcium release<br />

channel from rabbit bra<strong>in</strong>. FEBS Lett 312: 229–235, 1992.<br />

244. Han JW, Thieleczek R, Vasanyi M, Heilmeyers LMG. Compartmentalized<br />

ATP synthesis <strong>in</strong> skeletal muscle triads. Biochemistry<br />

31: 377–384, 1992.<br />

245. Hardie RC, M<strong>in</strong>ke B. The trp gene is essential for a light-activated<br />

Ca 2 channel <strong>in</strong> Drosophila photoreceptors. Neuron 8: 643–651,<br />

1992.<br />

246. Hard<strong>in</strong> CD, Raeymaekers L, Paul RJ. Comparison of endogenous<br />

and exogenous sources of ATP <strong>in</strong> fuel<strong>in</strong>g Ca 2 uptake <strong>in</strong><br />

smooth muscle plasma membrane vesicles. J Gen Physiol 99: 21–<br />

40, 1992.<br />

247. Hashitani H, Brad<strong>in</strong>g AF. Ionic basis for the regulation of spontaneous<br />

excitation <strong>in</strong> detrusor smooth muscle cells of the gu<strong>in</strong>eapig<br />

ur<strong>in</strong>ary bladder. Br J Pharmacol 140: 159–169, 2003.<br />

248. Hashitani H, Brad<strong>in</strong>g AF, Suzuki H. Correlation between spontaneous<br />

electrical, calcium and mechanical activity <strong>in</strong> detrusor<br />

smooth muscle of the gu<strong>in</strong>ea-pig bladder. Br J Pharmacol 141:<br />

183–193, 2004.<br />

249. Hashitani H, Van Helden DF, Suzuki H. Properties of spontaneous<br />

depolarizations <strong>in</strong> circular smooth muscle cells of rabbit urethra.<br />

Br J Pharmacol 1627–1632, 1996.<br />

250. Hashitani H, Yanai Y, Suzuki H. Role of <strong>in</strong>terstitial cells and gap<br />

junctions <strong>in</strong> the transmission of spontaneous Ca 2 signals <strong>in</strong> detrusor<br />

smooth muscles of the gu<strong>in</strong>ea-pig ur<strong>in</strong>ary bladder. J Physiol<br />

559: 567–581, 2004.<br />

251. Henmi S, Imaizumi Y, Muraki K, Watanabe M. Characteristics<br />

of caffe<strong>in</strong>e-<strong>in</strong>duced and spontaneous <strong>in</strong>ward currents and related<br />

<strong>in</strong>tracellular Ca 2 storage sites <strong>in</strong> gu<strong>in</strong>ea-pig tracheal smooth muscle<br />

cells. Eur J Pharmacol 282: 219–228, 1995.<br />

252. Heppner TJ, Bonev AD, Nelson MT. Elementary pur<strong>in</strong>ergic Ca 2<br />

transients evoked by nerve stimulation <strong>in</strong> rat ur<strong>in</strong>ary bladder<br />

smooth muscle. J Physiol 564: 201–212, 2005.<br />

253. Heppner TJ, Bonev AD, Santana LF, Nelson MT. Alkal<strong>in</strong>e pH<br />

shifts Ca 2 sparks to Ca 2 waves <strong>in</strong> smooth muscle cells of pressurized<br />

cerebral arteries. Am J Physiol Heart Circ Physiol 283:<br />

H2169–H2176, 2002.<br />

254. Herrera GM, Heppner TJ, Nelson MT. Regulation of ur<strong>in</strong>ary<br />

bladder smooth muscle contractions by ryanod<strong>in</strong>e receptors and<br />

BK and SK channels. Am J Physiol Regul Integr Comp Physiol 279:<br />

R60–R68, 2000.<br />

255. Herrera GM, Heppner TJ, Nelson MT. Voltage dependence of<br />

the coupl<strong>in</strong>g of Ca 2 sparks to BK(Ca) channels <strong>in</strong> ur<strong>in</strong>ary bladder<br />

smooth muscle. Am J Physiol Cell Physiol 280: C481–C490, 2001.<br />

SMOOTH MUSCLE SR 165<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

256. Herrera GM, Nelson MT. Differential regulation of SK and BK<br />

channels by Ca 2 signals from Ca 2 channels and ryanod<strong>in</strong>e receptors<br />

<strong>in</strong> gu<strong>in</strong>ea-pig ur<strong>in</strong>ary bladder myocytes. J Physiol 541: 483–<br />

492, 2002.<br />

257. Herrera GM, Pozo MJ, Zvara P, Petkov GV, Bond CT, Adelman<br />

JP, Nelson MT. Ur<strong>in</strong>ary bladder <strong>in</strong>stability <strong>in</strong>duced by selective<br />

suppression of the mur<strong>in</strong>e small conductance calcium-activated<br />

potassium (SK3) channel. J Physiol 551: 893–903, 2003.<br />

258. Herrmann-Frank A, Darl<strong>in</strong>g E, Meissner G. <strong>Function</strong>al characterization<br />

of the Ca 2 -gated Ca 2 release channel of vascular<br />

smooth muscle sarcoplasmic reticulum. Pflügers Arch 418: 353–<br />

359, 1991.<br />

259. Heumann HG. The subcellular localization of calcium <strong>in</strong> vertebrate<br />

smooth muscle: calcium-conta<strong>in</strong><strong>in</strong>g and calcium-accumulat<strong>in</strong>g<br />

structures <strong>in</strong> muscle cells of mouse <strong>in</strong>test<strong>in</strong>e. Cell Tissue Res<br />

169: 221–231, 1976.<br />

260. Hewavitharana T, Deng X, Soboloff J, Gill DL. Role of STIM<br />

and Orai prote<strong>in</strong>s <strong>in</strong> the store-operated calcium signal<strong>in</strong>g pathway.<br />

Cell Calcium 42: 173–182, 2007.<br />

261. Hidalgo C, de la FM, Gonzalez ME. Role of lipids <strong>in</strong> sarcoplasmic<br />

reticulum: a higher lipid content is required to susta<strong>in</strong> phosphoenzyme<br />

decomposition than phosphoenzyme formation. Arch<br />

Biochem Biophys 247: 365–371, 1986.<br />

262. Higashida H, Egorova A, Higashida C, Zhong ZG, Yokoyama S,<br />

Noda M, Zhang JS. Sympathetic potentiation of cyclic ADP-ribose<br />

formation <strong>in</strong> rat cardiac myocytes. J Biol Chem 274: 33348–33354,<br />

1999.<br />

263. Hirota S, Helli PB, Catalli A, Chew A, Janssen LJ. Airway<br />

smooth muscle excitation-contraction coupl<strong>in</strong>g and airway hyperresponsiveness.<br />

Can J Physiol Pharmacol 83: 725–732, 2005.<br />

264. Hisada T, Kurachi Y, Sugimoto T. Properties of membrane currents<br />

<strong>in</strong> isolated smooth muscle cells from gu<strong>in</strong>ea-pig trachea.<br />

Pflügers Arch 416: 151–161, 1990.<br />

265. Hisayama T, Takayanagi I, Okamoto Y. Ryanod<strong>in</strong>e reveals multiple<br />

contractile and relaxant mechanisms <strong>in</strong> vascular smooth muscle:<br />

simultaneous measurements of mechanical activity and of<br />

cytoplasmic free Ca 2 level with fura-2. Br J Pharmacol 100:<br />

677–684, 1990.<br />

266. Hofer AM, Curci S, Machen TE, Schulz I. ATP regulates calcium<br />

leak from agonist-sensitive <strong>in</strong>ternal calcium stores. FASEB J 10:<br />

302–308, 1996.<br />

267. Hofer AM, Fasolato C, Pozzan T. Capacitative Ca 2 entry is<br />

closely l<strong>in</strong>ked to the fill<strong>in</strong>g state of <strong>in</strong>ternal Ca 2 stores: a study<br />

us<strong>in</strong>g simultaneous measurements of I CRAC and <strong>in</strong>tralum<strong>in</strong>al<br />

[Ca 2 ]. J Cell Biol 140: 325–334, 1998.<br />

268. Hofer AM, Schlue WR, Curci S, Machen TE. Spatial distribution<br />

and quantitation of free lum<strong>in</strong>al [Ca] with<strong>in</strong> the InsP 3-sensitive<br />

<strong>in</strong>ternal store of <strong>in</strong>dividual BHK-21 cells: ion dependence of InsP 3<strong>in</strong>duced<br />

Ca release and reload<strong>in</strong>g. FASEB J 9: 788–798, 1995.<br />

269. Hoffman JF, Jo<strong>in</strong>er W, Nehrke K, Potapova O, Foye K, Wickrema<br />

A. The hSK4 (KCNN4) isoform is the Ca 2 -activated K <br />

channel (Gardos channel) <strong>in</strong> human red blood cells. Proc Natl Acad<br />

Sci USA 100: 7366–7371, 2003.<br />

270. Hogg RC, Wang Q, Helliwell RM, Large WA. Properties of<br />

spontaneous <strong>in</strong>ward currents <strong>in</strong> rabbit pulmonary artery smooth<br />

muscle cells. Pflügers Arch 425: 233–240, 1993.<br />

271. Horowitz A, Menice CB, Laporte R, Morgan KG. Mechanisms<br />

of smooth muscle contraction. Physiol Rev 76: 967–1003, 1996.<br />

272. Hoth M, Penner R. Depletion of <strong>in</strong>tracellular calcium stores activates<br />

a calcium current <strong>in</strong> mast cells. Nature 355: 353–356, 1992.<br />

273. Hotta S, Yamamura H, Ohya S, Imaizumi Y. Methyl-beta-cyclodextr<strong>in</strong><br />

prevents Ca 2 -<strong>in</strong>duced Ca 2 release <strong>in</strong> smooth muscle cells<br />

of mouse ur<strong>in</strong>ary bladder. J Pharmacol Sci 103: 121–126, 2007.<br />

274. House SJ, Potier M, Bisaillon J, S<strong>in</strong>ger HA, Trebak M. The<br />

non-excitable smooth muscle: calcium signal<strong>in</strong>g and phenotypic<br />

switch<strong>in</strong>g dur<strong>in</strong>g vascular disease. Pflügers Arch 456: 769–785,<br />

2008.<br />

275. Huddart H, Hunt S. Visceral <strong>Muscle</strong>: Its Structure and <strong>Function</strong>.<br />

Glasgow, Scotland: Blackie, 1975.<br />

276. Humbert JP, Matter N, Artault JC, Koppler P, Malviya AN.<br />

Inositol 1,4,5-trisphosphate receptor is located to the <strong>in</strong>ner nuclear<br />

membrane v<strong>in</strong>dicat<strong>in</strong>g regulation of nuclear calcium signal<strong>in</strong>g by<br />

<strong>in</strong>ositol 1,4,5-trisphosphate. Discrete distribution of <strong>in</strong>ositol phos-


166 SUSAN WRAY AND THEODOR BURDYGA<br />

phate receptors to <strong>in</strong>ner and outer nuclear membranes. J Biol<br />

Chem 271: 478–485, 1996.<br />

277. Hume JR, Leblanc N. Macroscopic K currents <strong>in</strong> s<strong>in</strong>gle smooth<br />

muscle cells of the rabbit portal ve<strong>in</strong>. J Physiol 413: 49–73, 1989.<br />

278. Hurwitz L, Jo<strong>in</strong>er PD, Von HS. Calcium pools utilized for contraction<br />

<strong>in</strong> smooth muscle. Am J Physiol 213: 1299–1304, 1967.<br />

279. Hyvel<strong>in</strong> JM, Guibert C, Marthan R, Sav<strong>in</strong>eau JP. Cellular<br />

mechanisms and role of endothel<strong>in</strong>-1-<strong>in</strong>duced calcium oscillations<br />

<strong>in</strong> pulmonary arterial myocytes. Am J Physiol Lung Cell Mol<br />

Physiol 275: L269–L282, 1998.<br />

280. Ianoul A, Grant DD, Rouleau Y, Bani-Yaghoub M, Johnston<br />

LJ, Pezacki JP. Imag<strong>in</strong>g nanometer doma<strong>in</strong>s of beta-adrenergic<br />

receptor complexes on the surface of cardiac myocytes. Nat Chem<br />

Biol 1: 196–202, 2005.<br />

281. I<strong>in</strong>o M. Calcium-<strong>in</strong>duced calcium release mechanism <strong>in</strong> gu<strong>in</strong>ea pig<br />

taenia caeca. J Gen Physiol 94: 363–383, 1989.<br />

282. I<strong>in</strong>o M. Biphasic Ca 2 dependence of <strong>in</strong>ositol 1,4,5-triphosphate<strong>in</strong>duced<br />

calcium release <strong>in</strong> smooth muscle cells of the gu<strong>in</strong>ea-pig<br />

taenia caeci. J Gen Physiol 95: 1103–1122, 1990.<br />

283. I<strong>in</strong>o M. Molecular basis of spatio-temporal dynamics <strong>in</strong> <strong>in</strong>ositol<br />

1,4,5-trisphosphate-mediated Ca 2 signall<strong>in</strong>g. Jpn J Pharmacol 82:<br />

15–20, 2000.<br />

284. I<strong>in</strong>o M, Kasai H, Yamazawa T. Visualization of neural control of<br />

<strong>in</strong>tracellular Ca 2 concentration <strong>in</strong> s<strong>in</strong>gle vascular smooth muscle<br />

cells <strong>in</strong> situ. EMBO J 13: 5026–5031, 1994.<br />

285. I<strong>in</strong>o M, Kobayashi T, Endo M. Use of ryanod<strong>in</strong>e for functional<br />

removal of the calcium store <strong>in</strong> smooth muscle cells of the gu<strong>in</strong>eapig.<br />

Biochem Biophys Res Commun 152: 417–422, 1988.<br />

286. I<strong>in</strong>o S, Hayashi H, Saito H, Tokuno H, Tomita T. Effects of<br />

<strong>in</strong>tracellular pH on calcium currents and <strong>in</strong>tracellular calcium ions<br />

<strong>in</strong> the smooth muscle of rabbit portal ve<strong>in</strong>. Exp Physiol 79: 669–<br />

680, 1994.<br />

287. Imaizumi Y, Muraki K, Watanabe M. Ionic currents <strong>in</strong> s<strong>in</strong>gle<br />

smooth muscle cells from the ureter of the gu<strong>in</strong>ea-pig. J Physiol<br />

411: 131–159, 1989.<br />

288. Imaizumi Y, Torii Y, Ohi Y, Nagano N, Atsuki K, Yamamura H,<br />

Muraki K, Watanabe M, Bolton TB. Ca 2 images and K current<br />

dur<strong>in</strong>g depolarization <strong>in</strong> smooth muscle cells of the gu<strong>in</strong>ea-pig vas<br />

deferens and ur<strong>in</strong>ary bladder. J Physiol 510: 705–719, 1998.<br />

289. Imtiaz MS, Smith DW, Van Helden DF. A theoretical model of<br />

slow wave regulation us<strong>in</strong>g voltage-dependent synthesis of <strong>in</strong>ositol<br />

1,4,5-trisphosphate. Biophys J 83: 1877–1890, 2002.<br />

290. Inesi G. Sequential mechanism of calcium b<strong>in</strong>d<strong>in</strong>g and translocation<br />

<strong>in</strong> sarcoplasmic reticulum adenos<strong>in</strong>e triphosphatase. J Biol<br />

Chem 262: 16338–16342, 1987.<br />

291. Inesi G, Lewis D, Murphy AJ. Interdependence of H ,Ca 2 , and<br />

P i (or vanadate) sites <strong>in</strong> sarcoplasmic reticulum ATPase. J Biol<br />

Chem 259: 996–1003, 1984.<br />

292. Inoue M, l<strong>in</strong> H, Imanaga I, Ogawa K, Warash<strong>in</strong>a A. InsP 3<br />

receptor type 2 and oscillatory and monophasic Ca 2 transients <strong>in</strong><br />

rat adrenal chromaff<strong>in</strong> cells. Cell Calcium 35: 59–70, 2004.<br />

293. Inoue R, Kitamura K, Kuriyama H. Two Ca-dependent K-channels<br />

classified by the application of tetraethylammonium distribute<br />

to smooth muscle membranes of the rabbit portal ve<strong>in</strong>. Pflügers<br />

Arch 405: 173–179, 1985.<br />

294. Ishida Y, Paul RJ. Ca 2 clearance <strong>in</strong> smooth muscle: lessons from<br />

gene-altered mice. J <strong>Smooth</strong> <strong>Muscle</strong> Res 41: 235–245, 2005.<br />

295. Ishida Y, Ries<strong>in</strong>ger I, Wallimann T, Paul RJ. Compartmentation<br />

of ATP synthesis and utilization <strong>in</strong> smooth muscle: roles of aerobic<br />

glycolysis and creat<strong>in</strong>e k<strong>in</strong>ase. Mol Cell Biochem 133–134: 39–50,<br />

1994.<br />

296. Ishii K, Hirose K, I<strong>in</strong>o M. Ca 2 shuttl<strong>in</strong>g between endoplasmic<br />

reticulum and mitochondria underly<strong>in</strong>g Ca 2 oscillations. EMBO<br />

Rep 7: 390–396, 2006.<br />

297. Islam MO, Yoshida Y, Koga T, Kojima M, Kangawa K, Imai S.<br />

Isolation and characterization of vascular smooth muscle <strong>in</strong>ositol<br />

1,4,5-trisphosphate receptor. Biochem J 316: 295–302, 1996.<br />

298. Isshiki M, Anderson RG. Calcium signal transduction from<br />

caveolae. Cell Calcium 26: 201–208, 1999.<br />

299. Itoh T, Kajikuri J, Kuriyama H. Characteristic features of noradrenal<strong>in</strong>e-<strong>in</strong>duced<br />

Ca 2 mobilization and tension <strong>in</strong> arterial smooth<br />

muscle of the rabbit. J Physiol 457: 297–341, 1992.<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

300. Jabr RI, Toland H, Gelband CH, Wang XX, Hume JR. Prom<strong>in</strong>ent<br />

role of <strong>in</strong>tracellular Ca 2 release <strong>in</strong> hypoxic vasoconstriction<br />

of can<strong>in</strong>e pulmonary artery. Br J Pharmacol 122: 21–30, 1997.<br />

301. Jackson WF. Potassium channels and proliferation of vascular<br />

smooth muscle cells. Circ Res 97: 1211–1212, 2005.<br />

302. Jaggar JH, Leffler CW, Cheranov SY, Tcheranova D, Cheng X.<br />

Carbon monoxide dilates cerebral arterioles by enhanc<strong>in</strong>g the coupl<strong>in</strong>g<br />

of Ca 2 sparks to Ca 2 -activated K channels. Circ Res 91:<br />

610–617, 2002.<br />

303. Jaggar JH, Nelson MT. Differential regulation of Ca 2 sparks and<br />

Ca 2 waves by UTP <strong>in</strong> rat cerebral artery smooth muscle cells.<br />

Am J Physiol Cell Physiol 279: C1528–C1539, 2000.<br />

304. Jaggar JH, Porter VA, Lederer WJ, Nelson MT. Calcium sparks<br />

<strong>in</strong> smooth muscle. Am J Physiol Cell Physiol 278: C235–C256, 2000.<br />

305. Jaggar JH, Stevenson AS, Nelson MT. Voltage dependence of<br />

Ca 2 sparks <strong>in</strong> <strong>in</strong>tact cerebral arteries. Am J Physiol Cell Physiol<br />

274: C1755–C1761, 1998.<br />

306. Jaggar JH, Wellman GC, Heppner TJ, Porter VA, Perez GJ,<br />

Gollasch M, Kleppisch T, Rubart M, Stevenson AS, Lederer<br />

WJ, Knot HJ, Bonev AD, Nelson MT. Ca 2 channels, ryanod<strong>in</strong>e<br />

receptors and Ca 2 -activated K channels: a functional unit for<br />

regulat<strong>in</strong>g arterial tone. Acta Physiol Scand 164: 577–587, 1998.<br />

307. Janiak R, Wilson SM, Montague S, Hume JR. Heterogeneity of<br />

calcium stores and elementary release events <strong>in</strong> can<strong>in</strong>e pulmonary<br />

arterial smooth muscle cells. Am J Physiol Cell Physiol 280: C22–<br />

C33, 2001.<br />

308. Janssen LJ, Simms SM. Spontaneous transient <strong>in</strong>ward currents <strong>in</strong><br />

rhythmicity <strong>in</strong> can<strong>in</strong>e and gu<strong>in</strong>ea-pig tracheal smooth muscle cells.<br />

Pflügers Arch 427: 473–480, 1994.<br />

309. Janssen LJ, Sims SM. Acetylchol<strong>in</strong>e activates non-selective cation<br />

and chloride conductances <strong>in</strong> can<strong>in</strong>e and gu<strong>in</strong>ea-pig tracheal<br />

myocytes. J Physiol 453: 197–218, 1992.<br />

310. Janssen LJ, Sims SM. Empty<strong>in</strong>g and refill<strong>in</strong>g of Ca 2 store <strong>in</strong><br />

tracheal myocytes as <strong>in</strong>dicated by ACh-evoked currents and contraction.<br />

Am J Physiol Cell Physiol 265: C877–C886, 1993.<br />

311. Je HD, Gangopadhyay SS, Ashworth TD, Morgan KG. Calpon<strong>in</strong><br />

is required for agonist-<strong>in</strong>duced signal transduction–evidence from<br />

an antisense approach <strong>in</strong> ferret smooth muscle. J Physiol 537:<br />

567–577, 2001.<br />

312. Ji G, Barsotti RJ, Feldman ME, Kotlikoff MI. Stretch-<strong>in</strong>duced<br />

calcium release <strong>in</strong> smooth muscle. J Gen Physiol 119: 533–544,<br />

2002.<br />

313. Ji G, Feldman M, Doran R, Zipfel W, Kotlikoff MI. Ca 2 -<br />

<strong>in</strong>duced Ca 2 release through localized Ca 2 uncag<strong>in</strong>g <strong>in</strong> smooth<br />

muscle. J Gen Physiol 127: 225–235, 2006.<br />

314. Ji G, Feldman ME, Greene KS, Sorrent<strong>in</strong>o V, X<strong>in</strong> HB, Kotlikoff<br />

MI. RYR2 prote<strong>in</strong>s contribute to the formation of Ca 2<br />

sparks <strong>in</strong> smooth muscle. J Gen Physiol 123: 377–386, 2004.<br />

315. Jiang D, Xiao B, Li X, Chen SR. <strong>Smooth</strong> muscle tissues express<br />

a major dom<strong>in</strong>ant negative splice variant of the type 3 Ca 2 release<br />

channel (ryanod<strong>in</strong>e receptor). J Biol Chem 278: 4763–4769, 2003.<br />

316. Jiang HH, Song B, Lu GS, Wen QJ, J<strong>in</strong> XY. Loss of ryanod<strong>in</strong>e<br />

receptor calcium-release channel expression associated with overactive<br />

ur<strong>in</strong>ary bladder smooth muscle contractions <strong>in</strong> a detrusor<br />

<strong>in</strong>stability model. BJU Int 96: 428–433, 2005.<br />

317. Jiang QX, Thrower EC, Chester DW, Ehrlich BE, Sigworth FJ.<br />

Three-dimensional structure of the type 1 <strong>in</strong>ositol 1,4,5-trisphosphate<br />

receptor at 24 A resolution. EMBO J 21: 3575–3581, 2002.<br />

318. John LM, Lechleiter JD, Camacho P. Differential modulation of<br />

SERCA2 isoforms by calreticul<strong>in</strong>. J Cell Biol 142: 963–973, 1998.<br />

319. Johnson JD, Snyder CH, Walsh MP, Flynn M. Effects of myos<strong>in</strong><br />

light cha<strong>in</strong> k<strong>in</strong>ase and peptides on Ca exchange with the N- and<br />

C-term<strong>in</strong>al Ca b<strong>in</strong>d<strong>in</strong>g sites of calmodul<strong>in</strong>. J Biol Chem 271: 761–<br />

767, 1996.<br />

320. Johnson S, Michalak M, Opas M, Eggleton P. The <strong>in</strong>s and outs<br />

of calreticul<strong>in</strong>: from the ER lumen to the extracellular space.<br />

Trends Cell Biol 11: 122–129, 2001.<br />

321. Jones K, Shmygol A, Kupittayanant S, Wray S. Electrophysiological<br />

characterization and functional importance of calcium-activated<br />

chloride channel <strong>in</strong> rat uter<strong>in</strong>e myocytes. Pflügers Arch 448:<br />

36–43, 2004.


322. Jorgensen AO, Campbell KP. Evidence for the presence of calsequestr<strong>in</strong><br />

<strong>in</strong> two structurally different regions of myocardial sarcoplasmic<br />

reticulum. J Cell Biol 98: 1597–1602, 1984.<br />

323. Jorgensen AO, Shen AC, Campbell KP, MacLennan DH. Ultrastructural<br />

localization of calsequestr<strong>in</strong> <strong>in</strong> rat skeletal muscle by<br />

immunoferrit<strong>in</strong> label<strong>in</strong>g of ultrath<strong>in</strong> frozen sections. J Cell Biol 97:<br />

1573–1581, 1983.<br />

324. Joseph SK, Rice HL, Williamson JR. The effect of external<br />

calcium and pH on <strong>in</strong>ositol trisphosphate-mediated calcium release<br />

from cerebellum microsomal fractions. Biochem J 258: 261–265,<br />

1989.<br />

325. Juan YS, Onal B, Broadaway S, Cosgrove J, Leggett RE, Whitbeck<br />

C, De E, Sokol R, Lev<strong>in</strong> RM. Effect of castration on male<br />

rabbit lower ur<strong>in</strong>ary tract tissue enzymes. Mol Cell Biochem 301:<br />

227–233, 2007.<br />

326. Juhaszova M, Blauste<strong>in</strong> MP. Na pump low and high ouaba<strong>in</strong><br />

aff<strong>in</strong>ity subunit isoforms are differently distributed <strong>in</strong> cells. Proc<br />

Natl Acad Sci USA 94: 1800–1805, 1997.<br />

327. Jung C, Zima AV, Szentesi P, Jona I, Blatter LA, Niggli E. Ca 2<br />

release from the sarcoplasmic reticulum activated by the low aff<strong>in</strong>ity<br />

Ca 2 chelator TPEN <strong>in</strong> ventricular myocytes. Cell Calcium<br />

41: 187–194, 2007.<br />

328. Kahonen M, Tolvanen JP, Sall<strong>in</strong>en K, Wu X, Porsti I. Influence<br />

of gender on control of arterial tone <strong>in</strong> experimental hypertension.<br />

Am J Physiol Heart Circ Physiol 275: H15–H22, 1998.<br />

329. Kamishima T, McCarron JG. Depolarization-evoked <strong>in</strong>creases <strong>in</strong><br />

cytosolic calcium concentration <strong>in</strong> isolated smooth muscle cells <strong>in</strong><br />

rat portal ve<strong>in</strong>. J Physiol 492: 61–74, 1996.<br />

330. Kamishima T, McCarron JG. Regulation of cytosolic Ca 2 concentration<br />

by Ca 2 stores <strong>in</strong> s<strong>in</strong>gle smooth muscle cells from rat<br />

cerebral arteries. J Physiol 501: 497–508, 1997.<br />

331. Kang TM, Rhee PL, Rhee JC, So I, Kim KW. Intracellular Ca 2<br />

oscillations <strong>in</strong> vascular smooth muscle cells. J <strong>Smooth</strong> <strong>Muscle</strong> Res<br />

31: 390–394, 1995.<br />

332. Kang TM, So I, Kim KW. Caffe<strong>in</strong>e- and histam<strong>in</strong>e-<strong>in</strong>duced oscillations<br />

of K(Ca) current <strong>in</strong> s<strong>in</strong>gle smooth muscle cells of rabbit<br />

cerebral artery. Pflügers Arch 431: 91–100, 1995.<br />

333. Kannan MS, Prakash YS, Brenner T, Mickelson JR, Sieck GC.<br />

Role of ryanod<strong>in</strong>e receptor channels <strong>in</strong> Ca 2 oscillations of porc<strong>in</strong>e<br />

tracheal smooth muscle. Am J Physiol Lung Cell Mol Physiol 272:<br />

L659–L664, 1997.<br />

334. Kao J, Fortner CN, Liu LH, Shull GE, Paul RJ. Ablation of the<br />

SERCA3 gene alters epithelium-dependent relaxation <strong>in</strong> mouse<br />

tracheal smooth muscle. Am J Physiol Lung Cell Mol Physiol 277:<br />

L264–L270, 1999.<br />

335. Kapela A, Bezerianos A, Tsoukias NM. A mathematical model<br />

of Ca 2 dynamics <strong>in</strong> rat mesenteric smooth muscle cell: agonist<br />

and NO stimulation. J Theor Biol 2008.<br />

336. Karaki H, Ozaki H, Hori M, Mitsui-Saito M, Amano KI, Harada<br />

KI, Miyamoto S, Nakazawa H, Won KJ, Sato K. Calcium movements,<br />

distribution and functions <strong>in</strong> smooth muscle. Pharmacol<br />

Rev 49: 157–230, 1997.<br />

337. Karczewski P, Hendrischke T, Wolf WP, Morano I, Bartel S,<br />

Schrader J. Phosphorylation of phospholamban correlates with<br />

relaxation of coronary artery <strong>in</strong>duced by nitric oxide, adenos<strong>in</strong>e,<br />

and prostacycl<strong>in</strong> <strong>in</strong> the pig. J Cell Biochem 70: 49–59, 1998.<br />

338. Karczewski P, Kelm M, Hartmann M, Schrader J. Role of<br />

phospholamban <strong>in</strong> NO/EDRF-<strong>in</strong>duced relaxation <strong>in</strong> rat aorta. Life<br />

Sci 51: 1205–1210, 1992.<br />

339. Kargac<strong>in</strong> GJ. Calcium signal<strong>in</strong>g <strong>in</strong> restricted diffusion spaces.<br />

Biophys J 67: 262–272, 1994.<br />

340. Kasai M, Kawasaki T, Yamaguchi N. Regulation of the ryanod<strong>in</strong>e<br />

receptor calcium release channel: a molecular complex system.<br />

Biophys Chem 82: 173–181, 1999.<br />

341. Kato K, Furuya K, Tsutsui I, Ozaki T, Yamagishi S. Cyclic<br />

AMP-mediated <strong>in</strong>hibition of noradrenal<strong>in</strong>e-<strong>in</strong>duced contraction and<br />

Ca 2 <strong>in</strong>flux <strong>in</strong> gu<strong>in</strong>ea-pig vas deferens. Exp Physiol 85: 387–398,<br />

2000.<br />

342. Katsuyama H, Ito S, Itoh T, Kuriyama H. Effects of ryanod<strong>in</strong>e<br />

on acetylchol<strong>in</strong>e-<strong>in</strong>duced Ca 2 mobilization <strong>in</strong> s<strong>in</strong>gle smooth muscle<br />

cells of the porc<strong>in</strong>e coronary artery. Pflügers Arch 419: 460–<br />

466, 1991.<br />

SMOOTH MUSCLE SR 167<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

343. Kawasaki T, Kasai M. Regulation of calcium channel <strong>in</strong> sarcoplasmic<br />

reticulum by calsequestr<strong>in</strong>. Biochem Biophys Res Commun<br />

199: 1120–1127, 1994.<br />

344. Khan I, Tabb T, Garfield RE, Jones LR, Fom<strong>in</strong> VP, Samson SE,<br />

Grover AK. Expression of the <strong>in</strong>ternal calcium pump <strong>in</strong> pregnant<br />

rat uterus. Cell Calcium 14: 111–117, 1993.<br />

345. Kim BJ, Jun JY, So I, Kim KW. Involvement of mitochondrial<br />

Na -Ca 2 exchange <strong>in</strong> <strong>in</strong>test<strong>in</strong>al pacemak<strong>in</strong>g activity. World J Gastroenterol<br />

12: 796–799, 2006.<br />

346. Kim M, Han IS, Koh SD, Perr<strong>in</strong>o BA. Roles of CaM k<strong>in</strong>ase II and<br />

phospholamban <strong>in</strong> SNP-<strong>in</strong>duced relaxation of mur<strong>in</strong>e gastric fundus<br />

smooth muscles. Am J Physiol Cell Physiol 291: C337–C347,<br />

2006.<br />

347. Kim M, Hennig GW, Smith TK, Perr<strong>in</strong>o BA. Phospholamban<br />

knockout <strong>in</strong>creases CaM k<strong>in</strong>ase II activity and <strong>in</strong>tracellular Ca 2<br />

wave activity and alters contractile responses of mur<strong>in</strong>e gastric<br />

antrum. Am J Physiol Cell Physiol 294: C432–C441, 2008.<br />

348. Kim M, Perr<strong>in</strong>o BA. CaM k<strong>in</strong>ase II activation and phospholamban<br />

phosphorylation by SNP <strong>in</strong> mur<strong>in</strong>e gastric antrum smooth muscles.<br />

Am J Physiol Gastro<strong>in</strong>test Liver Physiol 292: G1045–G1054, 2007.<br />

349. Kim SJ, Ahn SC, Kim JK, Kim YC, So I, Kim KW. Changes <strong>in</strong><br />

<strong>in</strong>tracellular Ca 2 concentration <strong>in</strong>duced by L-type Ca 2 channel<br />

current <strong>in</strong> gu<strong>in</strong>ea pig gastric myocytes. Am J Physiol Cell Physiol<br />

273: C1947–C1956, 1997.<br />

350. K<strong>in</strong>near NP, Wyatt CN, Clark JH, Calcraft PJ, Fleischer S,<br />

Jeyakumar LH, Nixon GF, Evans AM. Lysosomes co-localize<br />

with ryanod<strong>in</strong>e receptor subtype 3 to form a trigger zone for<br />

calcium signall<strong>in</strong>g by NAADP <strong>in</strong> rat pulmonary arterial smooth<br />

muscle. Cell Calcium 44: 190–201, 2008.<br />

351. Kirber MT, Etter EF, Bellve KA, Lifshitz LM, Tuft RA, Fay FS,<br />

Walsh JV, Fogarty KE. Relationship of Ca 2 sparks to STOCs<br />

studied with 2D and 3D imag<strong>in</strong>g <strong>in</strong> fel<strong>in</strong>e oesophageal smooth<br />

muscle cells. J Physiol 531: 315–327, 2001.<br />

352. Kirber MT, Guerrero-Hernandez A, Bowman DS, Fogarty KE,<br />

Tuft RA, S<strong>in</strong>ger JJ, Fay FS. Multiple pathways responsible for<br />

the stretch-<strong>in</strong>duced <strong>in</strong>crease <strong>in</strong> Ca 2 concentration <strong>in</strong> toad smooth<br />

muscle cells. J Physiol 524: 3–17, 2000.<br />

353. Kle<strong>in</strong> C, Malviya AN. Mechanism of nuclear calcium signal<strong>in</strong>g by<br />

<strong>in</strong>ositol 1,4,5-trisphosphate produced <strong>in</strong> the nucleus, nuclear located<br />

prote<strong>in</strong> k<strong>in</strong>ase C and cyclic AMP-dependent prote<strong>in</strong> k<strong>in</strong>ase.<br />

Front Biosci 13: 1206–1226, 2008.<br />

354. Kle<strong>in</strong> MG, Cheng H, Santana LF, Jiang YH, Lederer WJ,<br />

Schneider MF. Two mechanisms of quantized calcium release <strong>in</strong><br />

skeletal muscle. Nature 379: 455–458, 1996.<br />

355. Klockner U, Isenberg G. Endothel<strong>in</strong> depolarizes myocytes from<br />

porc<strong>in</strong>e coronary and human mesenteric arteries through a Caactivated<br />

chloride current. Pflügers Arch 418: 168–175, 1991.<br />

356. Knot HJ, Nelson MT. Regulation of arterial diameter and wall<br />

[Ca 2 ] <strong>in</strong> cerebral arteries of rat by membrane potential and <strong>in</strong>travascular<br />

pressure. J Physiol 508: 199–209, 1998.<br />

357. Knot HJ, Standen NB, Nelson MT. Ryanod<strong>in</strong>e receptors regulate<br />

arterial diamter and wall [Ca 2 ] <strong>in</strong> cerebral arteries of rat via<br />

Ca 2 -dependent K channels. J Physiol 508: 211–221, 1998.<br />

358. Kobayashi S, Kitizawa T, Somlyo AV, Somlyo AP. Cytosolic<br />

hepar<strong>in</strong> <strong>in</strong>hibits muscar<strong>in</strong>ic and -adrenergic Ca 2 release <strong>in</strong><br />

smooth muscle. J Biol Chem 264: 17997–18004, 1989.<br />

359. Kobayashi YM, Jones LR. Identification of triad<strong>in</strong> 1 as the predom<strong>in</strong>ant<br />

triad<strong>in</strong> isoform expressed <strong>in</strong> mammalian myocardium.<br />

J Biol Chem 274: 28660–28668, 1999.<br />

360. Koh SD, Dick GM, Sanders KM. Small-conductance Ca 2 -dependent<br />

K channels activated by ATP <strong>in</strong> mur<strong>in</strong>e colonic smooth<br />

muscle. Am J Physiol Cell Physiol 273: C2010–C2021, 1997.<br />

361. Kohda M, Komori S, Unno T, Ohashi H. Characterization of<br />

action potential-triggered [Ca 2 ] i transients <strong>in</strong> s<strong>in</strong>gle smooth muscle<br />

cells of gu<strong>in</strong>ea-pig ileum. Br J Pharmacol 122: 477–486, 1997.<br />

362. Kohler M, Hirschberg B, Bond CT, K<strong>in</strong>zie JM, Marrion NV,<br />

Maylie J, Adelman JP. Small-conductance, calcium-activated potassium<br />

channels from mammalian bra<strong>in</strong>. Science 273: 1709–1714,<br />

1996.<br />

363. Koller A, Schlossmann J, Ashman K, Uttenweiler-Joseph S,<br />

Ruth P, Hofmann F. Association of phospholamban with a cGMP<br />

k<strong>in</strong>ase signal<strong>in</strong>g complex. Biochem Biophys Res Commun 300:<br />

155–160, 2003.


168 SUSAN WRAY AND THEODOR BURDYGA<br />

364. Kong ID, Koh SD, Baygu<strong>in</strong>ov O, Sanders KM. Small conductance<br />

Ca 2 -activated K channels are regulated by Ca 2 -calmodul<strong>in</strong>-dependent<br />

prote<strong>in</strong> k<strong>in</strong>ase II <strong>in</strong> mur<strong>in</strong>e colonic myocytes.<br />

J Physiol 524: 331–337, 2000.<br />

365. Konhilas JP, Le<strong>in</strong>wand LA. The effects of biological sex and diet<br />

on the development of heart failure. Circulation 116: 2747–2759,<br />

2007.<br />

366. Korge P, Byrd SK, Campbell KB. <strong>Function</strong>al coupl<strong>in</strong>g between<br />

sarcoplasmic-reticulum-bound creat<strong>in</strong>e k<strong>in</strong>ase and Ca 2 -ATPase.<br />

Eur J Biochem 213: 973–980, 1993.<br />

367. Korge P, Campbell KB. Local ATP regeneration is important for<br />

sarcoplasmic reticulum Ca 2 pump function. Am J Physiol Cell<br />

Physiol 267: C357–C366, 1994.<br />

368. Kotlikoff MI. Calcium-<strong>in</strong>duced calcium release <strong>in</strong> smooth muscle:<br />

the case for loose coupl<strong>in</strong>g. Prog Biophys Mol Biol 83: 171–191,<br />

2003.<br />

369. Kotlikoff MI, Wang YX. Calcium release and calcium-activated<br />

chloride channels <strong>in</strong> airway smooth muscle cells. Am J Respir Crit<br />

Care Med 158: S109–S114, 1998.<br />

370. Kovac JR, Chrones T, Sims SM. Temporal and spatial dynamics<br />

underly<strong>in</strong>g capacitative calcium entry <strong>in</strong> human colonic smooth<br />

muscle. Am J Physiol Gastro<strong>in</strong>test Liver Physiol 294: G88–G98,<br />

2008.<br />

371. Kowarski D, Shutman H, Somlyo AP, Somlyo AV. Calcium<br />

release by noradrenal<strong>in</strong>e from central sarcoplasmic reticulum <strong>in</strong><br />

rabbit pulmonary artery smooth muscle. J Physiol 366: 153–175,<br />

1985.<br />

372. Kraan J, Koeter GH, vd Mark TW, Sluiter HJ, De VK. Changes<br />

<strong>in</strong> bronchial hyperreactivity <strong>in</strong>duced by 4 weeks of treatment with<br />

antiasthmatic drugs <strong>in</strong> patients with allergic asthma: a comparison<br />

between budesonide and terbutal<strong>in</strong>e. J Allergy Cl<strong>in</strong> Immunol 76:<br />

628–636, 1985.<br />

373. Kra<strong>in</strong>ev AG, Ferr<strong>in</strong>gton DA, Williams TD, Squier TC, Bigelow<br />

DJ. Adaptive changes <strong>in</strong> lipid composition of skeletal sarcoplasmic<br />

reticulum membranes associated with ag<strong>in</strong>g. Biochim Biophys<br />

Acta 1235: 406–418, 1995.<br />

374. Kubota Y, Hashitani H, Fukuta H, Kubota H, Kohri K, Suzuki<br />

H. Role of mitochondria <strong>in</strong> the generation of spontaneous activity<br />

<strong>in</strong> detrusor smooth muscles of the gu<strong>in</strong>ea pig bladder. J Urol 170:<br />

628–633, 2003.<br />

375. Kuo KH, Herrera AM, Seow CY. Ultrastructure of airway smooth<br />

muscle. Respir Physiol Neurobiol 137: 197–208, 2003.<br />

376. Kuo TH, Liu BF, Yu Y, Wuytack F, Raeymaekers L, Tsang W.<br />

Co-ord<strong>in</strong>ated regulation of the plasma membrane calcium pump<br />

and the sarco(endo)plasmic reticular calcium pump gene expression<br />

by Ca 2 . Cell Calcium 21: 399–408, 1997.<br />

377. Kupittayanant S, Luckas MJM, Wray S. Effects of <strong>in</strong>hibit<strong>in</strong>g the<br />

sarcoplasmic reticulum on spontaneous and oxytoc<strong>in</strong>-<strong>in</strong>duced contractions<br />

of human myometrium. Br J Obstet Gynaecol 109: 289–<br />

296, 2002.<br />

378. Lalli J, Harrer JM, Luo W, Kranias EG, Paul RJ. Targeted<br />

ablation of the phospholamban gene is associated with a marked<br />

decrease <strong>in</strong> sensitivity <strong>in</strong> aortic smooth muscle. Circ Res 80: 506–<br />

513, 1997.<br />

379. Lalli MJ, Shimizu S, Sutliff RL, Kranias EG, Paul RJ. [Ca 2 ] i<br />

homeostasis and cyclic nucleotide relaxation <strong>in</strong> aorta of phospholamban-deficient<br />

mice. Am J Physiol Heart Circ Physiol 277:<br />

H963–H970, 1999.<br />

380. Lamont C, Wier WG. Different roles of ryanod<strong>in</strong>e receptors and<br />

<strong>in</strong>ositol (1,4,5)-trisphosphate receptors <strong>in</strong> adrenergically stimulated<br />

contractions of small arteries. Am J Physiol Heart Circ<br />

Physiol 287: H617–H625, 2004.<br />

381. Lang DG, Ritchie AK. Large and small conductance calciumactivated<br />

potassium channels <strong>in</strong> the GH3 anterior pituitary cell l<strong>in</strong>e.<br />

Pflügers Arch 410: 614–622, 1987.<br />

382. Lang RJ. Identification of the major membrane currents <strong>in</strong> freshly<br />

dispersed s<strong>in</strong>gle smooth muscle cells of gu<strong>in</strong>ea-pig ureter. J Pharmacol<br />

412: 375–395, 1989.<br />

383. Langton PD, Nelson MT, Huang Y, Standen NB. Block of calcium-activated<br />

potassium channels <strong>in</strong> mammalian arterial myocytes<br />

by tetraethylammonium ions. Am J Physiol Heart Circ<br />

Physiol 2600: H927–H934, 1991.<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

384. Laporte R, Hui A, Laher I. Pharmacological modulation of sarcoplasmic<br />

reticulum function <strong>in</strong> smooth muscle. Pharmacol Rev<br />

56: 439–513, 2004.<br />

385. Larcombe-McDouall JB, Buttell N, Harrison N, Wray S. In vivo<br />

pH and metabolite changes dur<strong>in</strong>g a s<strong>in</strong>gle contraction <strong>in</strong> rat uter<strong>in</strong>e<br />

smooth muscle. J Physiol 518: 783–790, 1999.<br />

386. Large WA, Wang Q. Characteristics and physiological role of the<br />

Ca 2 -activated Cl conductance <strong>in</strong> smooth muscle. Am J Physiol<br />

Cell Physiol 271: C435–C454, 1996.<br />

387. Le Jemtel TH, Lambert F, Levitsky DO, Clergue M, Anger M,<br />

Gabbiani G, Lompre AM. Age-related changes <strong>in</strong> sarcoplasmic<br />

reticulum Ca 2 -ATPase and alpha-smooth muscle act<strong>in</strong> gene expression<br />

<strong>in</strong> aortas of normotensive and spontaneously hypertensive<br />

rats. Circ Res 72: 341–348, 1993.<br />

388. Leblanc N, Chartier D, Gossel<strong>in</strong> H, Rouleau JL. Age and gender<br />

differences <strong>in</strong> excitation-contraction coupl<strong>in</strong>g of the rat ventricle.<br />

J Physiol 511: 533–548, 1998.<br />

389. Ledbetter MW, Pre<strong>in</strong>er JK, Louis CF, Mickelson JR. Tissue<br />

distribution of ryanod<strong>in</strong>e receptor isoforms and alleles determ<strong>in</strong>ed<br />

by reverse transcription polymerase cha<strong>in</strong> reaction. J Biol Chem<br />

269: 31544–31551, 1994.<br />

390. Lee AG. How lipids affect the activities of <strong>in</strong>tegral membrane<br />

prote<strong>in</strong>s. Biochim Biophys Acta 1666: 62–87, 2004.<br />

391. Lee HC, Aarhus R. A derivative of NADP mobilizes calcium stores<br />

<strong>in</strong>sensitive to <strong>in</strong>ositol trisphosphate and cyclic ADP-ribose. J Biol<br />

Chem 270: 2152–2157, 1995.<br />

392. Lee HC, Walseth TF, Bratt GT, Hayes RN, Clapper DL. Structural<br />

determ<strong>in</strong>ation of a cyclic metabolite of NAD with <strong>in</strong>tracellular<br />

Ca 2 -mobiliz<strong>in</strong>g activity. J Biol Chem 264: 1608–1615, 1989.<br />

393. Leijten PA, Van BC. The effects of caffe<strong>in</strong>e on the noradrenal<strong>in</strong>esensitive<br />

calcium store <strong>in</strong> rabbit aorta. J Physiol 357: 327–339, 1984.<br />

394. Lesh RE, Nixon GF, Fleisher S, Airey JA, Somlyo AP, Somlyo<br />

AV. Localisation of ryanod<strong>in</strong>e receptors <strong>in</strong> smooth muscle. Circ<br />

Res 82: 175–185, 1998.<br />

395. Levy D, Seigneuret M, Bluzat A, Rigaud JL. Evidence for proton<br />

countertransport by the sarcoplasmic reticulum Ca 2 -ATPase dur<strong>in</strong>g<br />

calcium transport <strong>in</strong> reconstituted proteoliposomes with low<br />

ionic permeability. J Biol Chem 265: 19524–19534, 1990.<br />

396. Li J, Sukumar P, Milligan CJ, Kumar B, Ma ZY, Munsch CM,<br />

Jiang LH, Porter KE, Beech DJ. Interactions, functions, and<br />

<strong>in</strong>dependence of plasma membrane STIM1 and TRPC1 <strong>in</strong> vascular<br />

smooth muscle cells. Circ Res 103: e97–104, 2008.<br />

397. Li Y, Soos TJ, Li X, Wu J, Degennaro M, Sun X, Littman DR,<br />

Birnbaum MJ, Polakiewicz RD. Prote<strong>in</strong> k<strong>in</strong>ase C Theta <strong>in</strong>hibits<br />

<strong>in</strong>sul<strong>in</strong> signal<strong>in</strong>g by phosphorylat<strong>in</strong>g IRS1 at Ser(1101). J Biol Chem<br />

279: 45304–45307, 2004.<br />

398. Liao Y, Erxleben C, Abramowitz J, Flockerzi V, Zhu MX,<br />

Armstrong DL, Birnbaumer L. <strong>Function</strong>al <strong>in</strong>teractions among<br />

Orai1, TRPCs, and STIM1 suggest a STIM-regulated heteromeric<br />

Orai/TRPC model for SOCE/Icrac channels. Proc Natl Acad Sci<br />

USA 105: 2895–2900, 2008.<br />

399. L<strong>in</strong>grel J, Moseley A, Dostanic I, Cougnon M, He S, James P,<br />

Woo A, O’Connor K, Neumann J. <strong>Function</strong>al roles of the alpha<br />

isoforms of the Na,K-ATPase. Ann NY Acad Sci 986: 354–359, 2003.<br />

400. Liou J, Kim ML, Heo WD, Jones JT, Myers JW, Ferrell JE Jr,<br />

Meyer T. STIM is a Ca 2 sensor essential for Ca 2 -store-depletiontriggered<br />

Ca 2 <strong>in</strong>flux. Curr Biol 15: 1235–1241, 2005.<br />

401. Liu GGXK, Xu X, Huang JJ, Xiao JC, Zhang JP, Song HP.<br />

17-Oestradiol regulates the expression of Na /K -ATPase 1subunit,<br />

sarcoplasmic reticulum Ca 2 -ATPase and carbonic anhydrase<br />

IV <strong>in</strong> H9C2 cells. Cl<strong>in</strong> Exp Pharmacol Physiol 34: 998–1004,<br />

2007.<br />

402. Liu LH, Paul RJ, Sutliff RL, Miller ML, Lorenz JN, Pun RYK,<br />

Duffy JJ, Doetschman T, Kimura Y, MacLennan DH, Hoy<strong>in</strong>g<br />

JB, Shull GE. Defective endothelium-dependent relaxation of vascular<br />

smooth muscle and endothelial cell Ca 2 signall<strong>in</strong>g <strong>in</strong> mice<br />

lack<strong>in</strong>g sacro(endo)plasmic reticulum Ca 2 -ATPase isoform 3.<br />

J Biol Chem 272: 30538–30545, 1997.<br />

403. Liu M, Albert AP, Large WA. Facilitatory effect of Ins(1,4,5)P 3 on<br />

store-operated Ca 2 -permeable cation channels <strong>in</strong> rabbit portal<br />

ve<strong>in</strong> myocytes. J Physiol 566: 161–171, 2005.


404. Liu W, Meissner G. Structure-activity relationship of xanth<strong>in</strong>es<br />

and skeletal muscle ryanod<strong>in</strong>e receptor/Ca 2 release channel.<br />

Pharmacology 54: 135–143, 1997.<br />

405. Loew LM, Tuft RA, Carr<strong>in</strong>gton W, Fay FS. Imag<strong>in</strong>g <strong>in</strong> five<br />

dimensions: time-dependent membrane potentials <strong>in</strong> <strong>in</strong>dividual mitochondria.<br />

Biophys J 65: 2396–2407, 1993.<br />

406. Lohn M, Furstenau M, Sagach V, Elger M, Schulze W, Luft FC,<br />

Haller H, Gollasch M. Ignition of calcium sparks <strong>in</strong> arterial and<br />

cardiac muscle through caveolae. Circ Res 87: 1034–1039, 2000.<br />

407. Lohn M, Jessner W, Furstenau M, Wellner M, Sorrent<strong>in</strong>o V,<br />

Haller H, Luft FC, Gollasch M. Regulation of calcium sparks and<br />

spontaneous transient outward currents by RyR3 <strong>in</strong> arterial vascular<br />

smooth muscle cells. Circ Res 89: 1051–1057, 2001.<br />

408. Lomax RB, Camello C, Van CF, Petersen OH, Tepik<strong>in</strong> AV.<br />

Basal and physiological Ca 2 leak from the endoplasmic reticulum<br />

of pancreatic ac<strong>in</strong>ar cells. Second messenger-activated channels<br />

and translocons. J Biol Chem 277: 26479–26485, 2002.<br />

409. Lopes GS, Ferreira AT, Oshiro ME, Vladimirova I, Jurkiewicz<br />

NH, Jurkiewicz A, Smaili SS. Ag<strong>in</strong>g-related changes of <strong>in</strong>tracellular<br />

Ca 2 stores and contractile response of <strong>in</strong>test<strong>in</strong>al smooth<br />

muscle. Exp Gerontol 41: 55–62, 2006.<br />

410. Lorenz JN, Paul RJ. Dependence of Ca 2 channel currents on<br />

endogenous and exogenous sources of ATP <strong>in</strong> portal ve<strong>in</strong> smooth<br />

muscle. Am J Physiol Heart Circ Physiol 272: H987–H994, 1997.<br />

411. Loukotova J, Bacakova L, Zicha J, Kunes J. The <strong>in</strong>fluence of<br />

angiotens<strong>in</strong> II on sex-dependent proliferation of aortic VSMC isolated<br />

from SHR. Physiol Res 47: 501–505, 1998.<br />

412. Loukotova J, Kunes J, Zicha J. Gender-dependent difference <strong>in</strong><br />

cell calcium handl<strong>in</strong>g <strong>in</strong> VSMC isolated from SHR: the effect of<br />

angiotens<strong>in</strong> II. J Hypertens 20: 2213–2219, 2002.<br />

413. Lu W, Wang J, Shimoda LA, Sylvester JT. Differences <strong>in</strong> STIM1<br />

and TRPC expression <strong>in</strong> proximal and distal pulmonary arterial<br />

smooth muscle are associated with differences <strong>in</strong> Ca 2 responses<br />

to hypoxia. Am J Physiol Lung Cell Mol Physiol 295: L104–L113,<br />

2008.<br />

414. Luck<strong>in</strong> KA, Favero TG, Klug GA. Prolonged exercise <strong>in</strong>duces<br />

structural changes <strong>in</strong> SR Ca 2 -ATPase of rat muscle. Biochem Med<br />

Metab Biol 46: 391–405, 1991.<br />

415. Lui PP, Kong SK, Tsang D, Lee CY. The nuclear envelope of<br />

rest<strong>in</strong>g C6 glioma cells is able to release and uptake Ca 2 <strong>in</strong> the<br />

absence of chemical stimulation. Pflügers Arch 435: 357–361, 1998.<br />

416. Luik RM, Wu MM, Buchanan J, Lewis RS. The elementary unit<br />

of store-operated Ca 2 entry: local activation of CRAC channels by<br />

STIM1 at ER-plasma membrane junctions. J Cell Biol 174: 815–825,<br />

2006.<br />

417. Luo D, Nakazawa M, Yoshida Y, Cai J, Imai S. Effects of three<br />

different Ca 2 pump ATPase <strong>in</strong>hibitors on evoked contractions <strong>in</strong><br />

rabbit aorta and activities of Ca 2 pump ATPases <strong>in</strong> porc<strong>in</strong>e aorta.<br />

Gen Pharmacol 34: 211–220, 2000.<br />

418. Luo W, Grupp IL, Harrer J, Ponniah S, Grupp G, Duffy JJ,<br />

Doetschman T, Kranias EG. Targeted ablation of the phospholamban<br />

gene is associated with markedly enhanced myocardial<br />

contractility and loss of beta-agonist stimulation. Circ Res 75:<br />

401–409, 1994.<br />

419. Lynch JM, Chilibeck K, Qui Y, Michalak M. Assembl<strong>in</strong>g pieces<br />

of the cardiac puzzle; calreticul<strong>in</strong> and calcium-dependent pathways<br />

<strong>in</strong> cardiac development, health, and disease. Trends Cardiovasc<br />

Med 16: 65–69, 2006.<br />

420. Lynch RM, Kuettner CP, Paul RJ. Glycogen metabolism dur<strong>in</strong>g<br />

tension generation and ma<strong>in</strong>tenance <strong>in</strong> vascular smooth muscle.<br />

Am J Physiol Cell Physiol 257: C763–C742, 1989.<br />

421. Lynch RM, Paul RJ. Compartmentation of glycolysis and glycogenolysis<br />

<strong>in</strong> vascular smooth muscle. Science 222: 1344–1346, 1983.<br />

422. Lynch RM, Paul RJ. Compartmentation of carbohydrate metabolism<br />

<strong>in</strong> vascular smooth muscle. Am J Physiol Cell Physiol 252:<br />

C328–C334, 1987.<br />

423. Lyssand JS, Def<strong>in</strong>o MC, Tang XB, Hertz AL, Feller DB,<br />

Wacker JL, Adams ME, Hague C. Blood pressure is regulated by<br />

an alpha 1D-adrenergic receptor/dystroph<strong>in</strong> signalosome. J Biol<br />

Chem 283: 18792–18800, 2008.<br />

424. Lytton J, Nigam SK. Intracellular calcium: molecules and pools.<br />

Curr Op<strong>in</strong> Cell Biol 4: 220–226, 1992.<br />

SMOOTH MUSCLE SR 169<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

425. Lytton J, Westl<strong>in</strong> M, Burk SE, Shull GE, MacLennan DH.<br />

<strong>Function</strong>al comparisons between isoforms of the sarcoplasmic or<br />

endoplasmic reticulum family of calcium pumps. J Biol Chem 267:<br />

14483–14489, 1992.<br />

426. Lytton J, Westl<strong>in</strong> M, Hanley MR. Thapsigarg<strong>in</strong> <strong>in</strong>hibits the sarcoplasmic<br />

or endoplasmic reticulum Ca-ATPase family of calcium<br />

pumps. J Biol Chem 266: 17067–17071, 1991.<br />

427. MacKrill JJ. Prote<strong>in</strong>-prote<strong>in</strong> <strong>in</strong>teractions <strong>in</strong> <strong>in</strong>tracellular Ca 2 -<br />

release channel function. Biochem J 337: 345–361, 1999.<br />

428. MacLennan DH. Purification and properties of an adenos<strong>in</strong>e<br />

triphosphatase from sarcoplasmic reticulum. J Biol Chem 245:<br />

4508–4518, 1970.<br />

429. MacLennan DH. Isolation of a second form of calsequestr<strong>in</strong>. J Biol<br />

Chem 249: 980–984, 1974.<br />

430. MacLennan DH, bu-Abed M, Kang C. Structure-function relationships<br />

<strong>in</strong> Ca 2 cycl<strong>in</strong>g prote<strong>in</strong>s. J Mol Cell Cardiol 34: 897–918,<br />

2002.<br />

431. MacLennan DH, Kranias EG. Phospholamban: a crucial regulator<br />

of cardiac contractility. Nat Rev Mol Cell Biol 4: 566–577, 2003.<br />

432. MacLennan DH, Wong PT. Isolation of a calcium-sequester<strong>in</strong>g<br />

prote<strong>in</strong> from sarcoplasmic reticulum. Proc Natl Acad Sci USA 68:<br />

1231–1235, 1971.<br />

433. MacMillan D, Chalmers S, Muir TC, McCarron JG. IP 3-mediated<br />

Ca 2 <strong>in</strong>creases do not <strong>in</strong>volve the ryanod<strong>in</strong>e receptor, but<br />

ryanod<strong>in</strong>e receptor antagonists reduce IP 3-mediated Ca 2 <strong>in</strong>creases<br />

<strong>in</strong> gu<strong>in</strong>ea-pig colonic smooth muscle cells. J Physiol 569:<br />

533–544, 2005.<br />

434. Magnier-Gaubil C, Herbert JM, Quarck R, Papp B, Corvazier<br />

E, Wuytack F, Levy-Toledano S, Enouf J. <strong>Smooth</strong> muscle cell<br />

cycle and proliferation. Relationship between calcium <strong>in</strong>flux and<br />

sarco-endoplasmic reticulum Ca 2 ATPase regulation. J Biol Chem<br />

271: 27788–27794, 1996.<br />

435. Mall S, Broadbridge R, Harrison SL, Gore MG, Lee AG, East<br />

JM. The presence of sarcolip<strong>in</strong> results <strong>in</strong> <strong>in</strong>creased heat production<br />

by Ca 2 -ATPase. J Biol Chem 281: 36597–36602, 2006.<br />

436. Malviya AN, Kle<strong>in</strong> C. Mechanism regulat<strong>in</strong>g nuclear calcium signal<strong>in</strong>g.<br />

Can J Physiol Pharmacol 84: 403–422, 2006.<br />

437. Marks AR, Tempst P, Chadwick CC, Riviere L, Fleischer S,<br />

Nadal-G<strong>in</strong>ard B. <strong>Smooth</strong> muscle and bra<strong>in</strong> <strong>in</strong>ositol 1,4,5-trisphosphate<br />

receptors are structurally and functionally similar. J Biol<br />

Chem 265: 20719–20722, 1990.<br />

438. Markwardt F, Isenberg G. Gat<strong>in</strong>g of maxi K channels studied by<br />

Ca 2 concentration jumps <strong>in</strong> excised <strong>in</strong>side-out multi-channel<br />

patches (myocytes from gu<strong>in</strong>ea pig ur<strong>in</strong>ary bladder). J Gen Physiol<br />

99: 841–862, 1992.<br />

439. Mart<strong>in</strong> C, Chapman KE, Thornton S, Ashley RH. Changes <strong>in</strong> the<br />

expression of myometrial ryanod<strong>in</strong>e receptor mRNAs dur<strong>in</strong>g human<br />

pregnancy. Biochim Biophys Acta 1451: 343–352, 1999.<br />

440. Mart<strong>in</strong> C, Hyvel<strong>in</strong> JM, Chapman KE, Marthan R, Ashley RH,<br />

Sav<strong>in</strong>eau JP. Pregnant rat myometrial cells show heterogeneous<br />

ryanod<strong>in</strong>e- and caffe<strong>in</strong>e-sensitive calcium stores. Am J Physiol Cell<br />

Physiol 277: C243–C252, 1999.<br />

441. Mart<strong>in</strong> V, Bredoux R, Corvazier E, van GR, Kovacs T, Gelebart<br />

P, Enouf J. Three novel sarco/endoplasmic reticulum Ca 2 -<br />

ATPase (SERCA) 3 isoforms. Expression, regulation, and function<br />

of the membranes of the SERCA3 family. J Biol Chem 277: 24442–<br />

24452, 2002.<br />

442. Martonosi AN, Pikula S. The structure of the Ca 2 -ATPase of<br />

sarcoplasmic reticulum. Acta Biochim Pol 50: 337–365, 2003.<br />

443. Marty I. Triad<strong>in</strong>: a multi-prote<strong>in</strong> family for which purpose? Cell<br />

Mol Life Sci 61: 1850–1853, 2004.<br />

444. Masuo M, Toyo oka T, Sh<strong>in</strong> WS, Sugimoto T. Growth-dependent<br />

alterations of <strong>in</strong>tracellular Ca 2 -handl<strong>in</strong>g mechanisms of vascular<br />

smooth muscle cells. PDGF negatively regulates functional expression<br />

of voltage-dependent, IP 3-mediated, Ca 2 -<strong>in</strong>duced Ca 2 release<br />

channels. Circ Res 69: 1327–1339, 1991.<br />

445. Matthew A, Shmygol A, Wray S. Ca 2 entry, efflux and release <strong>in</strong><br />

smooth muscle. Biol Res 37: 617–624, 2004.<br />

446. Matthew AJG, Kupittayanant S, Burdyga TV, Wray S. Characterization<br />

of contractile activity and <strong>in</strong>tracellular Ca 2 signall<strong>in</strong>g <strong>in</strong><br />

mouse myometrium. J Soc Gynecol Invest 11: 207–212, 2004.<br />

447. Mattiazzi A, Mund<strong>in</strong>a-Weilenmann C, Guoxiang C, Vittone L,<br />

Kranias E. Role of phospholamban phosphorylation on Thr17 <strong>in</strong>


170 SUSAN WRAY AND THEODOR BURDYGA<br />

cardiac physiological and pathological conditions. Cardiovasc Res<br />

68: 366–375, 2005.<br />

448. Matz RL, Varez de SM, Schott C, Andriantsitoha<strong>in</strong>a R. Preservation<br />

of vascular contraction dur<strong>in</strong>g age<strong>in</strong>g: dual effect on calcium<br />

handl<strong>in</strong>g and sensitization. Br J Pharmacol 138: 745–750,<br />

2003.<br />

449. Mauban JRH, Lamont C, Balke CW, Wier WG. Adrenergic stimulation<br />

of rat resistance arteries affects Ca 2 sparks, Ca 2 waves,<br />

and Ca 2 oscillations. Am J Physiol Heart Circ Physiol 280:<br />

H2399–H2405, 2001.<br />

450. Mazzone J, Buxton IL. Changes <strong>in</strong> small conductance potassium<br />

channel expression <strong>in</strong> human myometrium dur<strong>in</strong>g pregnancy measured<br />

by RT-PCR. Proc West Pharmacol Soc 46: 74–77, 2003.<br />

451. McCarron JG, Chalmers S, Bradley KN, MacMillan D, Muir<br />

TC. Ca 2 microdoma<strong>in</strong>s <strong>in</strong> smooth muscle. Cell Calcium 40: 461–<br />

493, 2006.<br />

452. McCarron JG, Craig JW, Bradley KN, Muir TC. Agonist-<strong>in</strong>duced<br />

phasic and tonic responses <strong>in</strong> smooth muscle are mediated<br />

by InsP 3. J Cell Sci 115: 2207–2218, 2002.<br />

453. McCarron JG, Flynn ERM, Bradley KN, Muir TC. Two Ca 2<br />

entry pathways mediate InsP 3-sensitive store refill<strong>in</strong>g <strong>in</strong> gu<strong>in</strong>ea-pig<br />

colonic smooth muscle. J Physiol 525: 113–124, 2000.<br />

454. McCarron JG, Muir TC. Mitochondrial regulation of the cytosolic<br />

Ca 2 concentration and the InsP 3-sensitive Ca 2 store <strong>in</strong> gu<strong>in</strong>ea-pig<br />

colonic smooth muscle. J Physiol 516: 149–162, 1999.<br />

455. McCarron JG, Olson ML. A s<strong>in</strong>gle lum<strong>in</strong>ally cont<strong>in</strong>uous sarcoplasmic<br />

reticulum with apparently separate Ca 2 stores <strong>in</strong> smooth<br />

muscle. J Biol Chem 283: 7206–7218, 2008.<br />

456. McCloskey KD, Hollywood MA, Thornbury KD, Ward SM,<br />

McHale NG. Kit-like immunopositive cells <strong>in</strong> sheep mesenteric<br />

lymphatic vessels. Cell Tissue Res 310: 77–84, 2002.<br />

457. McFadzean I, Gibson A. The develop<strong>in</strong>g relationship between<br />

receptor-operated and store-operated calcium channels <strong>in</strong> smooth<br />

muscle. Br J Pharmacol 135: 1–13, 2002.<br />

458. McGraw DW, Fogel KM, Kong S, Litonjua AA, Kranias EG,<br />

Aronow BJ, Liggett SB. Transcriptional response to persistent<br />

beta2-adrenergic receptor signal<strong>in</strong>g reveals regulation of phospholamban,<br />

which alters airway contractility. Physiol Gen 27: 171–177,<br />

2006.<br />

459. McManus OB, Helms LM, Pallanck L, Ganetzky B, Swanson R,<br />

Leonard RJ. <strong>Function</strong>al role of the beta subunit of high conductance<br />

calcium-activated potassium channels. Neuron 14: 645–650,<br />

1995.<br />

460. Meissner G. Ryanod<strong>in</strong>e receptor/Ca 2 release channels and their<br />

regulation by endogenous effectors. Annu Rev Physiol 56: 485–508,<br />

1994.<br />

461. Meissner G. Molecular regulation of cardiac ryanod<strong>in</strong>e receptor<br />

ion channel. Cell Calcium 35: 621–628, 2004.<br />

462. Mejia-Alvarez R, Kettlun C, Rios E, Stern M, Fill M. Unitary<br />

Ca 2 current through cardiac ryanod<strong>in</strong>e receptor channels under<br />

quasi-physiological ionic conditions. J Gen Physiol 113: 177–186,<br />

1999.<br />

463. Meldolesi J, Pozzan T. The endoplasmic reticulum Ca 2 store: a<br />

view from the lumen. Trends Biochem Sci 23: 10–14, 1998.<br />

464. Mercer JC, Dehaven WI, Smyth JT, Wedel B, Boyles RR, Bird<br />

GS, Putney JW Jr. Large store-operated calcium selective currents<br />

due to co-expression of Orai1 or Orai2 with the <strong>in</strong>tracellular<br />

calcium sensor, Stim1. J Biol Chem 281: 24979–24990, 2006.<br />

465. Meredith AL, Thorneloe KS, Werner ME, Nelson MT, Aldrich<br />

RW. Overactive bladder and <strong>in</strong>cont<strong>in</strong>ence <strong>in</strong> the absence of the BK<br />

large conductance Ca 2 -activated K channel. J Biol Chem 279:<br />

36746–36752, 2004.<br />

466. Mery L, Mesaeli N, Michalak M, Opas M, Lew DP, Krause KH.<br />

Overexpression of calreticul<strong>in</strong> <strong>in</strong>creases <strong>in</strong>tracellular Ca 2 storage<br />

and decreases store-operated Ca 2 <strong>in</strong>flux. J Biol Chem 271: 9332–<br />

9339, 1996.<br />

467. Mesaeli N, Nakamura K, Zvaritch E, Dickie P, Dziak E, Krause<br />

KH, Opas M, MacLennan DH, Michalak M. Calreticul<strong>in</strong> is essential<br />

for cardiac development. J Cell Biol 144: 857–868, 1999.<br />

468. Michalak M, Corbett EF, Mesaeli N, Nakamura K, Opas M.<br />

Calreticul<strong>in</strong>: one prote<strong>in</strong>, one gene, and many functions. Biochem J<br />

344: 281–292, 1999.<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

469. Michalak M, Groenendyk J, Szabo E, Gold LI, Opas M. Calreticul<strong>in</strong>,<br />

a multi-process calcium-buffer<strong>in</strong>g chaperone of the endoplasmic<br />

reticulum. Biochem J 417: 651–666, 2009.<br />

470. Michalak M, Robert Parker JM, Opas M. Ca 2 signal<strong>in</strong>g and<br />

calcium b<strong>in</strong>d<strong>in</strong>g chaperones of the endoplasmic reticulum. Cell<br />

Calcium 32: 269–278, 2002.<br />

471. Michikawa T, Miyawaki A, Furuichi T, Mikoshiba K. Inositol<br />

1,4,5-trisphosphate receptors and calcium signal<strong>in</strong>g. Crit Rev Neurobiol<br />

10: 39–55, 1996.<br />

472. Milner RE, Baksh S, Shemanko C, Carpenter MR, Smillie L,<br />

Vance JE. Calreticul<strong>in</strong>, not calsequestr<strong>in</strong>, is the major calcium<br />

b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong> of smooth muscle sarcoplasmic reticulum and liver<br />

endoplasmic reticulum. J Biol Chem 266: 7155–7165, 1991.<br />

473. Miriel VA, Mauban JRH, Blauste<strong>in</strong> MP, Wier WG. Local and<br />

cellular Ca 2 transients <strong>in</strong> smooth muscle of pressurized rat resistance<br />

arteries dur<strong>in</strong>g myogenic and agonist stimulation. J Physiol<br />

518: 815–824, 1999.<br />

474. Mironneau J, Arnaudeau S, rez-Lepretre N, Boitt<strong>in</strong> FX. Ca 2<br />

sparks and Ca 2 waves activate different Ca 2 -dependent ion channels<br />

<strong>in</strong> s<strong>in</strong>gle myocytes from rat portal ve<strong>in</strong>. Cell Calcium 20:<br />

153–160, 1996.<br />

475. Mironneau J, Couss<strong>in</strong> F, Jejakumar LH, Fleischer S, Mironneau<br />

C, Macrez N. Contribution of ryanod<strong>in</strong>e receptor sub-type 3<br />

to Ca 2 responses <strong>in</strong> Ca 2 -overloaded cultured rat portal ve<strong>in</strong><br />

myocytes. J Biol Chem 14: 11257–11264, 2001.<br />

476. Mironneau J, Macrez N, Morel JL, Sorrent<strong>in</strong>o V, Mironneau<br />

C. Identification and function of ryanod<strong>in</strong>e receptor subtype 3 <strong>in</strong><br />

non-pregnant mouse myometrial cells. J Physiol 538: 707–716,<br />

2002.<br />

477. Missiaen L, De Smedt H, Droogmans G, Casteels R. Ca release<br />

<strong>in</strong>duced by <strong>in</strong>ositol 1,4,5-trisphosphate is a steady-state phenomenon<br />

controlled by lum<strong>in</strong>al Ca <strong>in</strong> permeabilized cells. Nature 357:<br />

599–601, 1992.<br />

478. Missiaen L, De Smedt H, Parys JB, Raeymaekers L, Droogmans<br />

G, Van den Bosch L, Casteels R. K<strong>in</strong>etics of the nonspecific<br />

calcium leak from non-mitochondrial calcium stores <strong>in</strong><br />

permeabilized A7r5 cells. Biochem J 317: 849–853, 1996.<br />

479. Missiaen L, De Smedt H, Droogmans G, Declerck I, Plessers L,<br />

Casteels R. Uptake characteristics of the InsP 3-sensitive and -<strong>in</strong>sensitive<br />

Ca 2 pools <strong>in</strong> porc<strong>in</strong>e aortic smooth-muscle cells: different<br />

Ca 2 sensitivity of the Ca 2 -uptake mechanism. Biochem Biophys<br />

Res Commun 174: 1183–1188, 1991.<br />

480. Missiaen L, Parys JB, De Smedt H, Himpens B, Casteels R.<br />

Inhibition of <strong>in</strong>ositol trisphosphate-<strong>in</strong>duced calcium release by caffe<strong>in</strong>e<br />

is prevented by ATP. Biochem J 300: 81–84, 1994.<br />

481. Missiaen L, Taylor CW, Berridge MJ. Lum<strong>in</strong>al Ca 2 promot<strong>in</strong>g<br />

spontaneous Ca 2 release from <strong>in</strong>ositol trisphosphate-sensitive<br />

stores <strong>in</strong> rat hepatocytes. J Physiol 455: 623–640, 1992.<br />

482. Missiaen L, Vanoevelen J, Parys JB, Raeymaekers L, De SH,<br />

Callewaert G, Erneux C, Wuytack F. Ca 2 uptake and release<br />

properties of a thapsigarg<strong>in</strong>-<strong>in</strong>sensitive nonmitochondrial Ca 2<br />

store <strong>in</strong> A7r5 and 16HBE14o cells. J Biol Chem 277: 6898–6902,<br />

2002.<br />

483. Mitchell RW, Halayko AJ, Kahraman S, Solway J, Wylam ME.<br />

Selective restoration of calcium coupl<strong>in</strong>g to muscar<strong>in</strong>ic M(3) receptors<br />

<strong>in</strong> contractile cultured airway myocytes. Am J Physiol<br />

Lung Cell Mol Physiol 278: L1091–L1100, 2000.<br />

484. Miyakawa T, Mizushima A, Hirose K, Yamazawa T, Bezprozvanny<br />

I, Kurosaki T, I<strong>in</strong>o M. Ca 2 -sensor region of IP 3 receptor<br />

controls <strong>in</strong>tracellular Ca 2 signal<strong>in</strong>g. EMBO J 20: 1674–1680, 2001.<br />

485. Modzelewska B, Kostrzewska A, Sipowicz M, Kleszczewski T,<br />

Batra S. Apam<strong>in</strong> <strong>in</strong>hibits NO-<strong>in</strong>duced relaxation of the spontaneous<br />

contractile activity of the myometrium from non-pregnant<br />

women. Reprod Biol Endocr<strong>in</strong>ol 1: 8, 2003.<br />

486. Mogami H, Tepik<strong>in</strong> AV, Petersen OH. Term<strong>in</strong>ation of cytosolic<br />

Ca 2 signals: Ca 2 reuptake <strong>in</strong>to <strong>in</strong>tracellular stores is regulated by<br />

the free Ca 2 concentration <strong>in</strong> the store lumen. EMBO J 17: 435–<br />

442, 1998.<br />

487. Moller JV, Nissen P, Sorensen TL, Le Maire M. Transport<br />

mechanism of the sarcoplasmic reticulum Ca 2 -ATPase pump.<br />

Curr Op<strong>in</strong> Struct Biol 15: 387–393, 2005.<br />

488. Monteith GR, Blauste<strong>in</strong> MP. Heterogeneity of mitochondrial<br />

matrix free Ca 2 : resolution of Ca 2 dynamics <strong>in</strong> <strong>in</strong>dividual mito-


chondria <strong>in</strong> situ. Am J Physiol Cell Physiol 276: C1193–C1204,<br />

1999.<br />

489. Montisano DF, Cascarano J, Pickett CB, James TW. Association<br />

between mitochondria and rough endoplasmic reticulum <strong>in</strong> rat<br />

liver. Anat Rec 203: 441–450, 1982.<br />

490. Moore ED, Voigt T, Kobayashi YM, Isenberg G, Fay FS, Gallitelli<br />

MF, Franz<strong>in</strong>i-Armstrong C. Organization of Ca 2 release<br />

units <strong>in</strong> excitable smooth muscle of the gu<strong>in</strong>ea-pig ur<strong>in</strong>ary bladder.<br />

Biophys J 87: 1836–1847, 2004.<br />

491. Moore EDW, Etter EF, Philipson KD, Carr<strong>in</strong>gton WA, Fogarty<br />

KE, Lifshitz LM, Fay FS. Coupl<strong>in</strong>g of the Na /Ca 2 exchanger,<br />

Na /K pump and sarcoplasmic reticulum <strong>in</strong> smooth muscle. Nature<br />

365: 657–660, 1993.<br />

492. Morales S, Camello PJ, Alcon S, Salido GM, Mawe G, Pozo<br />

MJ. Coactivation of capacitative calcium entry and L-type calcium<br />

channels <strong>in</strong> gu<strong>in</strong>ea pig gallbladder. Am J Physiol Gastro<strong>in</strong>test<br />

Liver Physiol 286: G1090–G1100, 2004.<br />

493. Morel JL, Fritz N, Lavie JL, Mironneau J. Crucial role of type<br />

2 <strong>in</strong>ositol 1,4,5-trisphosphate receptors for acetylchol<strong>in</strong>e-<strong>in</strong>duced<br />

Ca 2 oscillations <strong>in</strong> vascular myocytes. Arterioscler Thromb Vasc<br />

Biol 23: 1567–1575, 2003.<br />

494. Morimura K, Ohi Y, Yamamura H, Ohya S, Muraki K, Imaizumi<br />

Y. Two-step Ca 2 <strong>in</strong>tracellular release underlies excitation-contraction<br />

coupl<strong>in</strong>g <strong>in</strong> mouse ur<strong>in</strong>ary bladder myocytes. Am J Physiol<br />

Cell Physiol 290: C388–C403, 2006.<br />

495. Muraki K, Imaizumi Y, Kojima T, Kawai T, Watanabe M.<br />

Effects of tetraethylammonium and 4-am<strong>in</strong>opyrid<strong>in</strong>e on outward<br />

currents and excitability <strong>in</strong> can<strong>in</strong>e tracheal smooth muscle cells.<br />

Br J Pharmacol 100: 507–515, 1990.<br />

496. Murphy JG, Khalil RA. Gender-specific reduction <strong>in</strong> contractility<br />

and [Ca 2 ] i <strong>in</strong> vascular smooth muscle cells of female rat. Am J<br />

Physiol Cell Physiol 278: C834–C844, 2000.<br />

497. Murphy TV, Broad LM, Garland CJ. Characterisation of <strong>in</strong>ositol<br />

1,4,5-triphosphate b<strong>in</strong>d<strong>in</strong>g sites <strong>in</strong> rabbit aortic smooth muscle. Eur<br />

J Pharmacol 290: 145–150, 1995.<br />

498. Nakade S, Rhee SK, Hamanaka H, Mikoshiba K. Cyclic AMPdependent<br />

phosphorylation of an immunoaff<strong>in</strong>ity-purified homotetrameric<br />

<strong>in</strong>ositol 1,4,5-trisphosphate receptor (type I) <strong>in</strong>creases<br />

Ca 2 flux <strong>in</strong> reconstituted lipid vesicles. J Biol Chem 269: 6735–<br />

6742, 1994.<br />

499. Nakamura H, Jilka RL, Boland R, Martonosi AN. Mechanism of<br />

ATP hydrolysis by sarcoplasmic reticulum and the role of phospholipids.<br />

J Biol Chem 251: 5414–5423, 1976.<br />

500. Nakamura K, Robertson M, Liu G, Dickie P, Nakamura K, Guo<br />

JQ, Duff HJ, Opas M, Kavanagh K, Michalak M. Complete heart<br />

block and sudden death <strong>in</strong> mice overexpress<strong>in</strong>g calreticul<strong>in</strong>. J Cl<strong>in</strong><br />

Invest 107: 1245–1253, 2001.<br />

501. Nakamura K, Zupp<strong>in</strong>i A, Arnaudeau S, Lynch J, Ahsan I,<br />

Krause R, Papp S, De SH, Parys JB, Muller-Esterl W, Lew DP,<br />

Krause KH, Demaurex N, Opas M, Michalak M. <strong>Function</strong>al<br />

specialization of calreticul<strong>in</strong> doma<strong>in</strong>s. J Cell Biol 154: 961–972,<br />

2001.<br />

502. Nakanishi T, Gu H, Momma K. Developmental changes <strong>in</strong> the<br />

effect of acidosis on contraction, <strong>in</strong>tracellular pH, and calcium <strong>in</strong><br />

the rabbit mesenteric small artery. Pediatr Res 42: 750–757, 1997.<br />

503. Nakayama S, Chihara. S, Clark JF, Huang SM, Horiuchi T,<br />

Tomita T. Consequences of metabolic <strong>in</strong>hibition <strong>in</strong> smooth muscle<br />

isolated from gu<strong>in</strong>ea-pig stomach. J Physiol 505: 229–240, 1997.<br />

504. Nazer MA, Van BC. A role for the sarcoplasmic reticulum <strong>in</strong> Ca 2<br />

extrusion from rabbit <strong>in</strong>ferior vena cava smooth muscle. Am J<br />

Physiol Heart Circ Physiol 274: H123–H131, 1998.<br />

505. Neher E. Vesicle pools and Ca 2 microdoma<strong>in</strong>s: new tools for<br />

understand<strong>in</strong>g their roles <strong>in</strong> neurotransmitter release. Neuron 20:<br />

389–399, 1998.<br />

506. Nehrke K, Qu<strong>in</strong>n CC, Begenisich T. Molecular identification of<br />

Ca 2 -activated K channels <strong>in</strong> parotid ac<strong>in</strong>ar cells. Am J Physiol<br />

Cell Physiol 284: C535–C546, 2003.<br />

507. Nelson MT, Cheng H, Rubart M, Santana LF, Bonev AD, Knot<br />

HJ, Lederer WJ. Relaxation of arterial smooth muscle by calcium<br />

sparks. Science 270: 633–637, 1995.<br />

508. Nelson MT, Quayle JM. <strong>Physiological</strong> roles and properties of<br />

potassium channels <strong>in</strong> arterial smooth muscle. Am J Physiol Cell<br />

Physiol 268: C799–C822, 1995.<br />

SMOOTH MUSCLE SR 171<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

509. Newton T, Black JP, Butler J, Lee AG, Chad J, East JM.<br />

Sarco/endoplasmic-reticulum calcium ATPase SERCA1 is ma<strong>in</strong>ta<strong>in</strong>ed<br />

<strong>in</strong> the endoplasmic reticulum by a retrieval signal located<br />

between residues 1 and 211. Biochem J 371: 775–782, 2003.<br />

510. Neylon CB. Vascular biology of endothel<strong>in</strong> signal transduction.<br />

Cl<strong>in</strong> Exp Pharmacol Physiol 26: 149–153, 1999.<br />

511. Neylon CB, Richards SM, Larsen MA, Agrotis A, Bobik A.<br />

Multiple types of ryanod<strong>in</strong>e receptor/Ca 2 release channels are<br />

expressed <strong>in</strong> vascular smooth muscle. Biochem Biophys Res Commun<br />

215: 814–821, 1995.<br />

512. Ng LC, Kyle BD, Lennox AR, Shen XM, Hatton WJ, Hume JR.<br />

Cell culture alters Ca 2 entry pathways activated by store-depletion<br />

or hypoxia <strong>in</strong> can<strong>in</strong>e pulmonary arterial smooth muscle cells.<br />

Am J Physiol Cell Physiol 294: C313–C323, 2008.<br />

513. Nielsen H, Aalkjaer C, Mulvany MJ. Differential contractile<br />

effects of changes <strong>in</strong> carbon dioxide tension on rat mesenteric<br />

resistance arteries precontracted with noradrenal<strong>in</strong>e. Pflügers<br />

Arch 419: 51–56, 1991.<br />

514. Nimigean CM, Magleby KL. The beta subunit <strong>in</strong>creases the Ca 2<br />

sensitivity of large conductance Ca 2 -activated potassium channels<br />

by reta<strong>in</strong><strong>in</strong>g the gat<strong>in</strong>g <strong>in</strong> the burst<strong>in</strong>g states. J Gen Physiol<br />

113: 425–440, 1999.<br />

515. Nixon GF, Migneri GA, Somlyo AV. Immunogold localisation of<br />

<strong>in</strong>ositol 1,4,5-triphosphate receptors and characterisation of ultrastructural<br />

features of the sarcoplasmic reticulum <strong>in</strong> phasic and<br />

tonic smooth muscle. J <strong>Muscle</strong> Res Cell Motil 15: 682–699, 1994.<br />

516. Noack T, Deitmer P, Lammel E. Characterization of membrane<br />

currents <strong>in</strong> s<strong>in</strong>gle smooth muscle cells from the gu<strong>in</strong>ea-pig gastric<br />

antrum. J Physiol 451: 387–417, 1992.<br />

517. Nobe K, Sutliff RL, Kranias EG, Paul RJ. Phospholamban regulation<br />

of bladder contractility: evidence from gene-altered mouse<br />

models. J Physiol 535: 867–878, 2001.<br />

518. Noble K, Matthew A, Burdyga T, Wray S. A review of recent<br />

<strong>in</strong>sights <strong>in</strong>to the role of the sarcoplasmic reticulum and Ca entry <strong>in</strong><br />

uter<strong>in</strong>e smooth muscle. Eur J Obstet Gynecol Reprod Biol Suppl<br />

144: S11–19, 2009.<br />

519. Noble K, Wray S. The role of the sarcoplasmic reticulum <strong>in</strong><br />

neonatal uter<strong>in</strong>e smooth muscle: enhanced role compared to adult<br />

rat. J Physiol 545: 557–566, 2002.<br />

520. Noble K, Zhang J, Wray S. Lipid rafts, the sarcoplasmic reticulum<br />

and uter<strong>in</strong>e calcium signall<strong>in</strong>g: an <strong>in</strong>tegrated approach. J Physiol<br />

570: 29–35, 2006.<br />

521. Norregaard A, Vilsen B, Andersen JP. Transmembrane segment<br />

M3 is essential to thapsigarg<strong>in</strong> sensitivity of the sarcoplasmic reticulum<br />

Ca 2 -ATPase. J Biol Chem 269: 26598–26601, 1994.<br />

522. Odermatt A, Becker S, Khanna VK, Kurzydlowski K, Leisner<br />

E, Pette D, MacLennan DH. Sarcolip<strong>in</strong> regulates the activity of<br />

SERCA1, the fast-twitch skeletal muscle sarcoplasmic reticulum<br />

Ca 2 -ATPase. J Biol Chem 273: 12360–12369, 1998.<br />

523. Odermatt A, Taschner PE, Scherer SW, Beatty B, Khanna VK,<br />

Cornblath DR, Chaudhry V, Yee WC, Schrank B, Karpati G,<br />

Breun<strong>in</strong>g MH, Knoers N, MacLennan DH. Characterization of<br />

the gene encod<strong>in</strong>g human sarcolip<strong>in</strong> (SLN), a proteolipid associated<br />

with SERCA1: absence of structural mutations <strong>in</strong> five patients<br />

with Brody disease. Genomics 45: 541–553, 1997.<br />

524. Oh-hora M, Rao A. Calcium signal<strong>in</strong>g <strong>in</strong> lymphocytes. Curr Op<strong>in</strong><br />

Immunol 20: 250–258, 2008.<br />

525. Ohi Y, Yamamura H, Nagano N, Ohya S, Muraki K, Watanabe M,<br />

Imaizumi Y. Local Ca 2 transients and distribution of BK channels<br />

and ryanod<strong>in</strong>e receptors <strong>in</strong> smooth muscle cells of gu<strong>in</strong>ea-pig vas<br />

deferens and ur<strong>in</strong>ary bladder. J Physiol 534: 313–326, 2001.<br />

526. Ohta T, Kawai K, Ito S, Nakazato Y. Ca 2 entry activated by<br />

empty<strong>in</strong>g of <strong>in</strong>tracellular Ca 2 stores <strong>in</strong> ileal smooth muscle of the<br />

rat. Br J Pharmacol 114: 1165–1170, 1995.<br />

527. Ohya S, Kimura S, Kitsukawa M, Muraki K, Watanabe M,<br />

Imaizumi Y. SK4 encodes <strong>in</strong>termediate conductance Ca 2 -activated<br />

K channels <strong>in</strong> mouse ur<strong>in</strong>ary bladder smooth muscle cells.<br />

Jpn J Pharmacol 84: 97–100, 2000.<br />

528. Ohya Y, Kitamura K, Kuriyama H. Cellular calcium regulates<br />

outward currents <strong>in</strong> rabbit <strong>in</strong>test<strong>in</strong>al smooth muscle cell. Am J<br />

Physiol Cell Physiol 252: C401–C410, 1987.<br />

529. Opazo SA, Zhang W, Wu Y, Turner CE, Tang DD, Gunst SJ.<br />

Tension development dur<strong>in</strong>g contractile stimulation of smooth


172 SUSAN WRAY AND THEODOR BURDYGA<br />

muscle requires recruitment of paxill<strong>in</strong> and v<strong>in</strong>cul<strong>in</strong> to the membrane.<br />

Am J Physiol Cell Physiol 286: C433–C447, 2004.<br />

530. Oritani K, K<strong>in</strong>cade PW. Identification of stromal cell products<br />

that <strong>in</strong>teract with pre-B cells. J Cell Biol 134: 771–782, 1996.<br />

531. Ostwald TJ, MacLennan DH. Isolation of a high aff<strong>in</strong>ity calciumb<strong>in</strong>d<strong>in</strong>g<br />

prote<strong>in</strong> from sarcoplasmic reticulum. J Biol Chem 249:<br />

974–979, 1974.<br />

532. Otsu K, Willard HF, Khanna VK, Zorzato F, Green NM, Mac-<br />

Lennan DH. Molecular clon<strong>in</strong>g of cDNA encod<strong>in</strong>g the Ca 2 release<br />

channel (ryanod<strong>in</strong>e receptor) of rabbit cardiac muscle sarcoplasmic<br />

reticulum. J Biol Chem 265: 13472–13483, 1990.<br />

533. Owens GK. Regulation of differentiation of vascular smooth muscle<br />

cells. Physiol Rev 75: 487–517, 1995.<br />

534. Ozog A, Pouzet B, Bobe R, Lompre AM. Characterization of the<br />

3 end of the mouse SERCA 3 gene and tissue distribution of mRNA<br />

spliced variants. FEBS Lett 427: 349–352, 1998.<br />

535. Pabelick CM, Prakash YS, Kannan MS, Jones KA, Warner DO,<br />

Sieck GC. Effect of halothane on <strong>in</strong>tracellular calcium oscillations<br />

<strong>in</strong> porc<strong>in</strong>e tracheal smooth muscle cells. Am J Physiol Lung Cell<br />

Mol Physiol 276: L81–L89, 1999.<br />

536. Pabelick CM, Sieck GC, Prakash AS. Significance of spatial and<br />

temporal heterogeneity of calcium transients <strong>in</strong> smooth muscle.<br />

J Appl Physiol 91: 488–496, 2001.<br />

537. Pacaud P, Loirand G. Release of Ca 2 by noradrenal<strong>in</strong>e and ATP<br />

from the same Ca 2 store sensitive to both InsP 3 and Ca 2 <strong>in</strong> rat<br />

portal ve<strong>in</strong> myocytes. J Physiol 484: 549–555, 1995.<br />

538. Pacaud P, Loirand G, Grégoire G, Mironneau C, Mironneau J.<br />

Calcium-dependence of the calcium-activated chloride current <strong>in</strong><br />

smooth muscle cells of rat portal ve<strong>in</strong>. Pflügers Arch 421: 125–130,<br />

1992.<br />

539. Pacaud P, Loirand G, Mironneau C, Mironneau J. Noradrenal<strong>in</strong>e<br />

activates a calcium-activated chloride conductance and <strong>in</strong>creases<br />

the voltage-dependent calcium current <strong>in</strong> cultured s<strong>in</strong>gle<br />

cells of rat portal ve<strong>in</strong>. Br J Pharmacol 97: 139–146, 1989.<br />

540. Pagano RE, Mart<strong>in</strong> OC, Kang HC, Haugland RP. A novel fluorescent<br />

ceramide analogue for study<strong>in</strong>g membrane traffic <strong>in</strong> animal<br />

cells: accumulation at the Golgi apparatus results <strong>in</strong> altered spectral<br />

properties of the sph<strong>in</strong>golipid precursor. J Cell Biol 113: 1267–<br />

1279, 1991.<br />

541. Pani B, Ong HL, Liu X, Rauser K, Ambudkar IS, S<strong>in</strong>gh BB.<br />

Lipid rafts determ<strong>in</strong>e cluster<strong>in</strong>g of STIM1 <strong>in</strong> endoplasmic reticulum-plasma<br />

membrane junctions and regulation of store-operated<br />

Ca 2 entry (SOCE). J Biol Chem 283: 17333–17340, 2008.<br />

542. Parekh AB, Putney JW Jr. Store-operated calcium channels.<br />

Physiol Rev 85: 757–810, 2005.<br />

543. Park YB. Ion selectivity and gat<strong>in</strong>g of small conductance Ca 2 -<br />

activated K channels <strong>in</strong> cultured rat adrenal chromaff<strong>in</strong> cells.<br />

J Physiol 481: 555–570, 1994.<br />

544. Parker I, Zang WJ, Wier WG. Ca 2 sparks <strong>in</strong>volv<strong>in</strong>g multiple<br />

Ca 2 release sites along Z-l<strong>in</strong>es <strong>in</strong> rat heart cells. J Physiol 497:<br />

31–38, 1996.<br />

545. Parod RJ, Putney JW Jr. The role of calcium <strong>in</strong> the receptor<br />

mediated control of potassium permeability <strong>in</strong> the rat lacrimal<br />

gland. J Physiol 281: 371–381, 1978.<br />

546. Patel S, Joseph SK, Thomas AP. Molecular properties of <strong>in</strong>ositol<br />

1,4,5-trisphosphate receptors. Cell Calcium 25: 247–264, 1999.<br />

547. Pavlovic M, Schaller A, Ste<strong>in</strong>er B, Berdat P, Carrel T, Pfammatter<br />

JP, Ammann RA, Gallati S. Gender modulates the expression<br />

of calcium-regulat<strong>in</strong>g prote<strong>in</strong>s <strong>in</strong> pediatric atrial myocardium.<br />

Exp Biol Med 230: 853–859, 2005.<br />

548. Peachey LD, Porter KR. Intracellular impulse conduction <strong>in</strong><br />

smooth muscle cells. Science 129: 721–722, 1959.<br />

549. Peel SE, Liu B, Hall IP. A key role for STIM1 <strong>in</strong> store operated<br />

calcium channel activation <strong>in</strong> airway smooth muscle. Respir Res 7:<br />

119, 2006.<br />

550. Peel SE, Liu B, Hall IP. ORAI and store-operated calcium <strong>in</strong>flux<br />

<strong>in</strong> human airway smooth muscle cells. Am J Respir Cell Mol Biol<br />

38: 744–749, 2008.<br />

551. Pe<strong>in</strong>elt C, Apell HJ. K<strong>in</strong>etics of the Ca 2 ,H , and Mg 2 <strong>in</strong>teraction<br />

with the ion-b<strong>in</strong>d<strong>in</strong>g sites of the SR Ca-ATPase. Biophys J 82:<br />

170–181, 2002.<br />

552. Pe<strong>in</strong>elt C, Vig M, Koomoa DL, Beck A, Nadler MJ, Koblan-<br />

Huberson M, Lis A, Fleig A, Penner R, K<strong>in</strong>et JP. Amplification<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

of CRAC current by STIM1 and CRACM1 (Orai1). Nat Cell Biol 8:<br />

771–773, 2006.<br />

553. Penna A, Demuro A, Yerom<strong>in</strong> AV, Zhang SL, Safr<strong>in</strong>a O, Parker<br />

I, Cahalan MD. The CRAC channel consists of a tetramer formed<br />

by Stim-<strong>in</strong>duced dimerization of Orai dimers. Nature 456: 116–120,<br />

2008.<br />

554. Perez GJ, Bonev AD, Nelson MT. Micromolar Ca 2 from sparks<br />

activates Ca 2 -sensitive K channels <strong>in</strong> rat cerebral artery smooth<br />

muscle. Am J Physiol Cell Physiol 281: C1769–C1775, 2001.<br />

555. Perez GJ, Bonev AD, Patlak JB, Nelson MT. <strong>Function</strong>al coupl<strong>in</strong>g<br />

of ryanod<strong>in</strong>e receptors to K Ca channels <strong>in</strong> smooth muscle<br />

cells from rat cerebral arteries. J Gen Physiol 113: 229–237, 1999.<br />

556. Perez JF, Sanderson MJ. The contraction of smooth muscle cells<br />

of <strong>in</strong>trapulmonary arterioles is determ<strong>in</strong>ed by the frequency of<br />

Ca 2 oscillations <strong>in</strong>duced by 5-HT and KCl. J Gen Physiol 125:<br />

555–567, 2005.<br />

557. Perez JF, Sanderson MJ. The frequency of calcium oscillations<br />

<strong>in</strong>duced by 5-HT, ACH, and KCl determ<strong>in</strong>e the contraction of<br />

smooth muscle cells of <strong>in</strong>trapulmonary bronchioles. J Gen Physiol<br />

125: 535–553, 2005.<br />

558. Periasamy M, Huke S. SERCA pump level is a critical determ<strong>in</strong>ant<br />

of Ca 2 homeostasis and cardiac contractility. J Mol Cell Cardiol<br />

33: 1053–1063, 2001.<br />

559. Periasamy M, Kalyanasundaram A. SERCA pump isoforms: their<br />

role <strong>in</strong> calcium transport and disease. <strong>Muscle</strong> Nerve 35: 430–442,<br />

2007.<br />

560. Periasamy M, Reed TD, Liu LH, Ji Y, Loukianov E, Paul RJ,<br />

Nieman ML, Riddle T, Duffy JJ, Doetschman T, Lorenz JN,<br />

Shull GE. Impaired cardiac performance <strong>in</strong> heterozygous mice<br />

with a null mutation <strong>in</strong> the sarco(endo)plasmic reticulum Ca 2 -<br />

ATPase isoform 2 (SERCA2) gene. J Biol Chem 274: 2556–2562,<br />

1999.<br />

561. Perk<strong>in</strong>s GA, Renken CW, Song JY, Frey TG, Young SJ, Lamont<br />

S, Martone ME, L<strong>in</strong>dsey S, Ellisman MH. Electron tomography<br />

of large, multicomponent biological structures. J Struct Biol<br />

120: 219–227, 1997.<br />

562. Petkov GV, Boev KK. The role of sarcoplasmic reticulum and<br />

sarcoplasmic reticulum Ca 2 -ATPase <strong>in</strong> the smooth muscle tone of<br />

the cat gastric fundus. Pflügers Arch 431: 928–935, 1996.<br />

563. Pfleiderer PJ, Lu KK, Crow MT, Keller RS, S<strong>in</strong>ger HA. Modulation<br />

of vascular smooth muscle cell migration by calcium/calmodul<strong>in</strong>-dependent<br />

prote<strong>in</strong> k<strong>in</strong>ase II-delta 2. Am J Physiol Cell<br />

Physiol 286: C1238–C1245, 2004.<br />

564. Platoshyn O, Remillard CV, Fantozzi I, Mandegar M, Sison<br />

TT, Zhang S, Burg E, Yuan JX. Diversity of voltage-dependent K <br />

channels <strong>in</strong> human pulmonary artery smooth muscle cells. Am J<br />

Physiol Lung Cell Mol Physiol 287: L226–L238, 2004.<br />

565. Poburko D, Kuo KH, Dai J, Lee CH, van Breemen C. Organellar<br />

junctions promote targeted Ca 2 signal<strong>in</strong>g <strong>in</strong> smooth muscle: why<br />

two membranes are better than one. Trends Pharmacol Sci 25:<br />

8–15, 2004.<br />

566. Poch E, Leach S, Snape S, Cacic T, MacLennan DH, Lytton J.<br />

<strong>Function</strong>al characterization of alternatively spliced human<br />

SERCA3 transcripts. Am J Physiol Cell Physiol 275: C1449–C1458,<br />

1998.<br />

567. Popescu LM, de Bruijn WC, Zelck U, Ionescu N. Intracellular<br />

distribution of calcium <strong>in</strong> smooth muscle: facts and artifacts. A<br />

correlation of cytochemical, biochemical and X-ray microanalytical<br />

f<strong>in</strong>d<strong>in</strong>gs. Morphol Embryol 26: 251–258, 1980.<br />

568. Porter VA, Bonev AD, Knot HJ, Heppner TJ, Stevenson AS,<br />

Kleppisch T, Lederer WJ, Nelson MT. Frequency modulation of<br />

Ca 2 sparks is <strong>in</strong>volved <strong>in</strong> regulation of arterial diameter by cyclic<br />

nucleotides. Am J Physiol Cell Physiol 274: C1346–C1355, 1998.<br />

569. Potier M, Trebak M. New developments <strong>in</strong> the signal<strong>in</strong>g mechanisms<br />

of the store-operated calcium entry pathway. Pflügers Arch<br />

457: 405–415, 2008.<br />

570. Pottorf WJ, Duckles SP, Buchholz JN. Adrenergic nerves compensate<br />

for a decl<strong>in</strong>e <strong>in</strong> calcium buffer<strong>in</strong>g dur<strong>in</strong>g age<strong>in</strong>g. J Auton<br />

Pharmacol 20: 1–13, 2000.<br />

571. Powis DA, Marley PD. Ions, channels, and receptors. Ann NY<br />

Acad Sci 971: 95–99, 2002.


572. Pozo MJ, Perez GJ, Nelson MT, Mawe GM. Ca 2 sparks and BK<br />

currents <strong>in</strong> gallbladder myocytes: role <strong>in</strong> CCK-<strong>in</strong>duced response.<br />

Am J Physiol Gastro<strong>in</strong>test Liver Physiol 282: G165–G174, 2002.<br />

573. Pozzan T, Rizzuto R, Volpe P, Meldolesi J. Molecular and<br />

cellular physiology of <strong>in</strong>tracellular calcium stores. Physiol Rev 74:<br />

595–636, 1994.<br />

574. Prakash YS, Kannan MS, Sieck GC. Regulation of <strong>in</strong>tracellular<br />

calcium oscillations <strong>in</strong> porc<strong>in</strong>e tracheal smooth muscle cells. Am J<br />

Physiol Cell Physiol 272: C966–C975, 1997.<br />

575. Prakriya M, Feske S, Gwack Y, Srikanth S, Rao A, Hogan PG.<br />

Orai1 is an essential pore subunit of the CRAC channel. Nature 443:<br />

230–233, 2006.<br />

576. Prestwich SA, Bolton TB. Inhibition of muscar<strong>in</strong>ic receptor<strong>in</strong>duced<br />

<strong>in</strong>ositol phospholipid hydrolysis by caffe<strong>in</strong>e -adrenoceotors<br />

and prote<strong>in</strong> k<strong>in</strong>ase C <strong>in</strong> <strong>in</strong>test<strong>in</strong>al smooth muscle. Br J<br />

Pharmacol 114: 602–611, 1995.<br />

577. Prosser CL, Burnstock G, Kahn J. Conduction <strong>in</strong> smooth muscle:<br />

comparative structural properties. Am J Physiol 199: 545–552,<br />

1960.<br />

578. Putney JW Jr. A model for receptor-regulated calcium entry. Cell<br />

Calcium 7: 1–12, 1986.<br />

579. Putney JW Jr. Capacitative Calcium Entry. Aust<strong>in</strong>, TX: Landes,<br />

1997.<br />

580. Putney JW Jr. Capacitative calcium entry <strong>in</strong> the nervous system.<br />

Cell Calcium 34: 339–344, 2003.<br />

581. Putney JW Jr. Recent breakthroughs <strong>in</strong> the molecular mechanism<br />

of capacitative calcium entry (with thoughts on how we got here).<br />

Cell Calcium 42: 103–110, 2007.<br />

582. Raeymaekers L, Eggermont JA, Wuytack F, Casteels R. Effects<br />

of cyclic nucleotide dependent prote<strong>in</strong> k<strong>in</strong>ases on the endoplasmic<br />

reticulum Ca 2 pump of bov<strong>in</strong>e pulmonary artery. Cell<br />

Calcium 11: 261–268, 1990.<br />

583. Raeymaekers L, Hofmann F, Casteels R. Cyclic GMP-dependent<br />

prote<strong>in</strong> k<strong>in</strong>ase phosphorylates phospholamban <strong>in</strong> isolated sarcoplasmic<br />

reticulum from cardiac and smooth muscle. Biochem J<br />

252: 269–273, 1988.<br />

584. Raeymaekers L, Jones LR. Evidence for the presence of phospholamban<br />

<strong>in</strong> the endoplasmic reticulum of smooth muscle. Biochim<br />

Biophys Acta 882: 258–265, 1986.<br />

585. Raeymaekers L, Verbist J, Wuytack F, Plessers L, Casteels R.<br />

Expression of Ca 2 b<strong>in</strong>d<strong>in</strong>g prote<strong>in</strong>s of the sarcoplasmic reticulum<br />

of striated muscle <strong>in</strong> the endoplasmic reticulum of pig smooth<br />

muscles. Cell Calcium 14: 581–589, 1993.<br />

586. Ramos-Franco J, Bare D, Caenepeel S, Nani A, Fill M, Mignery<br />

G. S<strong>in</strong>gle-channel function of recomb<strong>in</strong>ant type 2 <strong>in</strong>ositol<br />

1,4,5-trisphosphate receptor. Biophys J 79: 1388–1399, 2000.<br />

587. Rembold CM, Chen XL. The buffer barrier hypothesis, [Ca 2 ] i<br />

homogeneity, and sarcoplasmic reticulum function <strong>in</strong> sw<strong>in</strong>e carotid<br />

artery. J Physiol 513: 477–492, 1998.<br />

588. Rembold CM, Kendall JM, Campbell AK. Measurement of<br />

changes <strong>in</strong> sarcoplasmic reticulum [Ca 2 ] <strong>in</strong> rat tail artery with<br />

targeted apoaequor<strong>in</strong> delivered by an adenoviral vector. Cell Calcium<br />

21: 69–79, 1997.<br />

589. Rest<strong>in</strong>i CA, Moreira JE, Bendhack LM. Cross-talk between the<br />

sarcoplasmic reticulum and the mitochondrial calcium handl<strong>in</strong>g<br />

systems may play an important role <strong>in</strong> the regulation of contraction<br />

<strong>in</strong> anococcygeus smooth muscle. Mitochondrion 6: 71–81, 2006.<br />

590. Rizzuto R, Br<strong>in</strong>i M, Pozzan T. Target<strong>in</strong>g recomb<strong>in</strong>ant aequor<strong>in</strong> to<br />

specific <strong>in</strong>tracellular organelles. Methods Cell Biol 40: 339–358,<br />

1994.<br />

591. Rizzuto R, P<strong>in</strong>ton P, Carr<strong>in</strong>gton WA, Fay FS, Fogarty KE,<br />

Lifshitz LM, Tuft RA, Pozzan T. Close contacts with the endoplasmic<br />

reticulum as determ<strong>in</strong>ants of mitochondrial Ca 2 responses.<br />

Science 280: 1763–1766, 1998.<br />

592. Rizzuto R, Pozzan T. Microdoma<strong>in</strong>s of <strong>in</strong>tracellular Ca 2 : molecular<br />

determ<strong>in</strong>ants and functional consequences. Physiol Rev 86:<br />

369–408, 2006.<br />

593. Roberts D, Gelper<strong>in</strong> D, Wiley JW. Evidence for age-associated<br />

reduction <strong>in</strong> acetylchol<strong>in</strong>e release and smooth muscle response <strong>in</strong><br />

the rat colon. Am J Physiol Gastro<strong>in</strong>test Liver Physiol 267: G515–<br />

G522, 1994.<br />

SMOOTH MUSCLE SR 173<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

594. Rohra DK, Saito SY, Ohizumi Y. <strong>Function</strong>al role of ryanod<strong>in</strong>esensitive<br />

Ca 2 stores <strong>in</strong> acidic pH-<strong>in</strong>duced contraction <strong>in</strong> Wistar<br />

Kyoto rat aorta. Life Sci 72: 1259–1269, 2003.<br />

595. Roos J, DiGregorio PJ, Yerom<strong>in</strong> AV, Ohlsen K, Lioudyno M,<br />

Zhang S, Safr<strong>in</strong>a O, Kozak JA, Wagner SL, Cahalan MD, Velicelebi<br />

G, Stauderman KA. STIM1, an essential and conserved<br />

component of store-operated Ca 2 channel function. J Cell Biol<br />

169: 435–445, 2005.<br />

596. Ross R, Klebanoff SJ. F<strong>in</strong>e structural changes <strong>in</strong> uter<strong>in</strong>e smooth<br />

muscle and fibroblasts <strong>in</strong> response to estrogen. J Cell Biol 32:<br />

155–167, 1967.<br />

597. Rossi AE, Dirksen RT. <strong>Sarcoplasmic</strong> reticulum: the dynamic<br />

calcium governor of muscle. <strong>Muscle</strong> Nerve 33: 715–731, 2006.<br />

598. Rossi AM, Eppenberger HM, Volpe P, Cotrufo R, Wallimann T.<br />

<strong>Muscle</strong>-type MM creat<strong>in</strong>e k<strong>in</strong>ase is specifically bound to sarcoplasmic<br />

reticulum and can support Ca 2 uptake and regulate local<br />

ATP/ADP ratios. J Biol Chem 265: 5258–5266, 1990.<br />

599. Rossi D, Sorrent<strong>in</strong>o V. Molecular genetics of ryanod<strong>in</strong>e receptors<br />

Ca 2 -release channels. Cell Calcium 32: 307–319, 2002.<br />

600. Rousseau E, Lad<strong>in</strong>e J, Liu QY, Meissner G. Activation of the<br />

Ca 2 release channel of skeletal muscle sarcoplasmic reticulum by<br />

caffe<strong>in</strong>e and related compounds. Arch Biochem Biophys 267: 75–<br />

86, 1988.<br />

601. Rousseau E, Meissner G. S<strong>in</strong>gle cardiac sarcoplasmic reticulum<br />

Ca 2 -release channel: activation by caffe<strong>in</strong>e. Am J Physiol Heart<br />

Circ Physiol 256: H328–H333, 1989.<br />

602. Rousseau E, P<strong>in</strong>kos J. pH modulates conduct<strong>in</strong>g and gat<strong>in</strong>g<br />

behaviour of s<strong>in</strong>gle calcium release channels. Pflügers Arch 415:<br />

645–647, 1990.<br />

603. Roux E, Marhl M. Role of sarcoplasmic reticulum and mitochondria<br />

<strong>in</strong> Ca 2 removal <strong>in</strong> airway myocytes. Biophys J 86: 2583–2595,<br />

2004.<br />

604. Rusko J, Bolton TB, Aaronson P, Bauer V. Effects of phenylephr<strong>in</strong>e<br />

<strong>in</strong> s<strong>in</strong>gle isolated smooth muscle cells of rabbit and gu<strong>in</strong>ea<br />

pig taenia caeci. Eur J Pharmacol 184: 325–328, 1990.<br />

605. Sabnis RW, Deligeorgiev TG, Jachak MN, Dalvi TS. DiOC6(3):<br />

a useful dye for sta<strong>in</strong><strong>in</strong>g the endoplasmic reticulum. Biotech Histochem<br />

72: 253–258, 1997.<br />

606. Sag CM, Dybkova N, Neef S, Maier LS. Effects on recovery<br />

dur<strong>in</strong>g acidosis <strong>in</strong> cardiac myocytes overexpress<strong>in</strong>g CaMKII. J Mol<br />

Cell Cardiol 43: 696–709, 2007.<br />

607. Saito A, Seiler S, Chu A, Fleischer S. Preparation and morphology<br />

of sarcoplasmic reticulum term<strong>in</strong>al cisternae from rabbit skeletal<br />

muscle. J Cell Biol 99: 875–885, 1984.<br />

608. Sakuntabhai A, Ruiz-Perez V, Carter S, Jacobsen N, Burge S,<br />

Monk S, Smith M, Munro CS, O’Donovan M, Craddock N,<br />

Kucherlapati R, Rees JL, Owen M, Lathrop GM, Monaco AP,<br />

Strachan T, Hovnanian A. Mutations <strong>in</strong> ATP2A2, encod<strong>in</strong>g a Ca 2<br />

pump, cause Darier disease. Nat Genet 21: 271–277, 1999.<br />

609. Saleh SN, Albert AP, Peppiatt CM, Large WA. Angiotens<strong>in</strong> II<br />

activates two cation conductances with dist<strong>in</strong>ct TRPC1 and TRPC6<br />

channel properties <strong>in</strong> rabbit mesenteric artery myocytes. J Physiol<br />

577: 479–495, 2006.<br />

610. Samso M, Wagenknecht T. Contributions of electron microscopy<br />

and s<strong>in</strong>gle-particle techniques to the determ<strong>in</strong>ation of the ryanod<strong>in</strong>e<br />

receptor three-dimensional structure. J Struct Biol 121: 172–<br />

180, 1998.<br />

611. Sanborn BM. Hormonal signal<strong>in</strong>g and signal pathway crosstalk <strong>in</strong><br />

the control of myometrial calcium dynamics. Sem<strong>in</strong> Cell Dev Biol<br />

18: 305–314, 2007.<br />

612. Sanders KM. Regulation of smooth muscle excitation and contraction.<br />

Neurogastroenterol Motil 20 Suppl 1: 39–53, 2008.<br />

613. Sandoval RJ, Injeti ER, Gerthoffer WT, Pearce WJ. Postnatal<br />

maturation modulates relationships among cytosolic Ca 2 , myos<strong>in</strong><br />

light cha<strong>in</strong> phosphorylation, and contractile tone <strong>in</strong> ov<strong>in</strong>e cerebral<br />

arteries. Am J Physiol Heart Circ Physiol 293: H2183–H2192, 2007.<br />

614. Santana LF, Kranias EG, Lederer WJ. Calcium sparks and<br />

excitation-contraction coupl<strong>in</strong>g <strong>in</strong> phospholamban-deficient mouse<br />

ventricular myocytes. J Physiol 503: 21–29, 1997.<br />

615. Sarcevic B, Brookes V, Mart<strong>in</strong> TJ, Kemp BE, Rob<strong>in</strong>son PJ.<br />

Atrial natriuretic peptide-dependent phosphorylation of smooth<br />

muscle cell particulate fraction prote<strong>in</strong>s is mediated by cGMPdependent<br />

prote<strong>in</strong> k<strong>in</strong>ase. J Biol Chem 264: 20648–20654, 1989.


174 SUSAN WRAY AND THEODOR BURDYGA<br />

616. Satrustegui J, Villalba M, Pereira R, Bogonez E, Mart<strong>in</strong>ez-<br />

Serrano A. Cytosolic and mitochondrial calcium <strong>in</strong> synaptosomes<br />

dur<strong>in</strong>g ag<strong>in</strong>g. Life Sci 59: 429–434, 1996.<br />

617. Saunders HM, Farley JM. Spontaneous transient outward currents<br />

and Ca 2 -activated K channels <strong>in</strong> sw<strong>in</strong>e tracheal smooth<br />

muscle cells. J Pharmacol Exp Ther 257: 1114–1120, 1991.<br />

618. Sausbier M, Arntz C, Bucurenciu I, Zhao H, Zhou XB, Sausbier<br />

U, Feil S, Kamm S, Ess<strong>in</strong> K, Sailer CA, Abdullah U,<br />

Krippeit-Drews P, Feil R, Hofmann F, Knaus HG, Kenyon C,<br />

Shipston MJ, Storm JF, Neuhuber W, Korth M, Schubert R,<br />

Gollasch M, Ruth P. Elevated blood pressure l<strong>in</strong>ked to primary<br />

hyperaldosteronism and impaired vasodilation <strong>in</strong> BK channel-deficient<br />

mice. Circulation 112: 60–68, 2005.<br />

619. Sausbier M, Hu H, Arntz C, Feil S, Kamm S, Adelsberger H,<br />

Sausbier U, Sailer CA, Feil R, Hofmann F, Korth M, Shipston<br />

MJ, Knaus HG, Wolfer DP, Pedroarena CM, Storm JF, Ruth P.<br />

Cerebellar ataxia and Purk<strong>in</strong>je cell dysfunction caused by Ca 2 -<br />

activated K channel deficiency. Proc Natl Acad Sci USA 101:<br />

9474–9478, 2004.<br />

620. Sav<strong>in</strong>eau JP, Mironneau J, Mironneau C. Contractile properties<br />

of chemically sk<strong>in</strong>ned fibers from pregnant rat myometrium: existence<br />

of an <strong>in</strong>ternal Ca-store. Pflügers Arch 411: 296–303, 1988.<br />

621. Schmitt JP, Kamisago M, Asahi M, Li GH, Ahmad F, Mende U,<br />

Kranias EG, MacLennan DH, Seidman JG, Seidman CE. Dilated<br />

cardiomyopathy and heart failure caused by a mutation <strong>in</strong><br />

phospholamban. Science 299: 1410–1413, 2003.<br />

622. Scott BT, Simmerman HK, Coll<strong>in</strong>s JH, Nadal-G<strong>in</strong>ard B, Jones<br />

LR. Complete am<strong>in</strong>o acid sequence of can<strong>in</strong>e cardiac calsequestr<strong>in</strong><br />

deduced by cDNA clon<strong>in</strong>g. J Biol Chem 263: 8958–8964, 1988.<br />

623. Sergeant GP, Hollywood MA, McCloskey KD, McHale NG,<br />

Thornbury KD. Role of IP 3 <strong>in</strong> modulation of spontaneous activity<br />

<strong>in</strong> pacemaker cells of rabbit urethra. Am J Physiol Cell Physiol 280:<br />

C1349–C1356, 2001.<br />

624. Serysheva II, Schatz M, Van HM, Chiu W, Hamilton SL. Structure<br />

of the skeletal muscle calcium release channel activated with<br />

Ca 2 and AMP-PCP. Biophys J 77: 1936–1944, 1999.<br />

625. Shabir S, Borisova L, Wray S, Burdyga T. Rho-k<strong>in</strong>ase <strong>in</strong>hibition<br />

and electromechanical coupl<strong>in</strong>g <strong>in</strong> phasic smooth muscle; Ca 2 -<br />

dependent and <strong>in</strong>dependent mechanisms. J Physiol 560: 839–855,<br />

2004.<br />

626. Shah M, Haylett DG. The pharmacology of hSK1 Ca 2 -activated<br />

K channels expressed <strong>in</strong> mammalian cell l<strong>in</strong>es. Br J Pharmacol<br />

129: 627–630, 2000.<br />

627. Shannon TR, G<strong>in</strong>sburg KS, Bers DM. Reverse mode of the<br />

sarcoplasmic reticulum calcium pump and load-dependent cytosolic<br />

calcium decl<strong>in</strong>e <strong>in</strong> voltage-clamped cardiac ventricular myocytes.<br />

Biophys J 78: 322–333, 2000.<br />

628. Shepherd MC, Duffy SM, Harris T, Cruse G, Schuliga M,<br />

Brightl<strong>in</strong>g CE, Neylon CB, Bradd<strong>in</strong>g P, Stewart AG. KCa3.1<br />

Ca 2 activated K channels regulate human airway smooth muscle<br />

proliferation. Am J Respir Cell Mol Biol 37: 525–531, 2007.<br />

629. Shima H, Blauste<strong>in</strong> MP. Modulation of evoked contractions <strong>in</strong> rat<br />

arteries by ryanod<strong>in</strong>e, thapsigarg<strong>in</strong>, and cyclopiazonic acid. Circ<br />

Res 70: 968–977, 1992.<br />

630. Shmigol A, Eisner DA, Wray S. Properties of voltage-activated<br />

[Ca 2 ] transients <strong>in</strong> s<strong>in</strong>gle smooth muscle cells isolated from pregnant<br />

rat uterus. J Physiol 511: 803–811, 1998.<br />

631. Shmigol AV, Eisner DA, Wray S. The role of the sarcoplasmic<br />

reticulum as a calcium s<strong>in</strong>k <strong>in</strong> uter<strong>in</strong>e smooth muscle cells.<br />

J Physiol 520: 153–163, 1999.<br />

633. Shmigol AV, Eisner DA, Wray S. Simultaneous measurements of<br />

changes <strong>in</strong> sarcoplasmic reticulum and cytosolic [Ca 2 ] <strong>in</strong> rat<br />

uter<strong>in</strong>e smooth muscle cells. J Physiol 531: 707–713, 2001.<br />

634. Shmygol A, Noble K, Wray S. Depletion of membrane cholesterol<br />

elim<strong>in</strong>ates the Ca 2 -activated component of outward potassium<br />

current and decreases membrane capacitance <strong>in</strong> rat uter<strong>in</strong>e myocytes.<br />

J Physiol 445–456, 2007.<br />

635. Shmygol A, Wray S. <strong>Function</strong>al architecture of the SR calcium<br />

store <strong>in</strong> uter<strong>in</strong>e smooth muscle. Cell Calcium 35: 501–508, 2004.<br />

636. Shmygol A, Wray S. Modulation of agonist-<strong>in</strong>duced Ca 2 release<br />

by SR Ca 2 load: direct SR and cytosolic Ca 2 measurements <strong>in</strong> rat<br />

uter<strong>in</strong>e myocytes. Cell Calcium 37: 215–223, 2005.<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

637. Si H, Heyken WT, Wolfle SE, Tysiac M, Schubert R, Grgic I,<br />

Vilianovich L, Gieb<strong>in</strong>g G, Maier T, Gross V, Bader M, De WC,<br />

Hoyer J, Kohler R. Impaired endothelium-derived hyperpolariz<strong>in</strong>g<br />

factor-mediated dilations and <strong>in</strong>creased blood pressure <strong>in</strong> mice<br />

deficient of the <strong>in</strong>termediate-conductance Ca 2 -activated K channel.<br />

Circ Res 99: 537–544, 2006.<br />

638. Sieck GC, Kannan MS, Prakash YS. Heterogeneity <strong>in</strong> dynamic<br />

regulation of <strong>in</strong>tracellular calcium <strong>in</strong> airway smooth muscle cells.<br />

Can J Physiol Pharmacol 75: 878–888, 1997.<br />

639. Simmerman HK, Jones LR. Phospholamban: prote<strong>in</strong> structure,<br />

mechanism of action, and role <strong>in</strong> cardiac function. Physiol Rev 78:<br />

921–947, 1998.<br />

640. Sims SM, Jiao Y, Zheng ZG. Intracellular calcium stores <strong>in</strong> isolated<br />

tracheal smooth muscle cells. Am J Physiol Lung Cell Mol<br />

Physiol 271: L300–L309, 1996.<br />

641. S<strong>in</strong>ger JJ, Walsh JV. Characterization of calcium-activated potassium<br />

channels <strong>in</strong> s<strong>in</strong>gle smooth muscle cells us<strong>in</strong>g the patch-clamp<br />

technique. Pflügers Arch 408: 98–111, 1987.<br />

642. Sitsapesan R, Williams AJ. Regulation of the gat<strong>in</strong>g of the sheep<br />

cardiac sarcoplasmic reticulum Ca 2 -release channel by lum<strong>in</strong>al<br />

Ca 2 . J Membr Biol 137: 215–226, 1994.<br />

643. Slack JP, Grupp IL, Luo W, Kranias EG. Phospholamban ablation<br />

enhances relaxation <strong>in</strong> the mur<strong>in</strong>e soleus. Am J Physiol Cell<br />

Physiol 273: C1–C6, 1997.<br />

644. Smani T, Zakharov SI, Csutora P, Leno E, Trepakova ES,<br />

Bolot<strong>in</strong>a VM. A novel mechanism for the store-operated calcium<br />

<strong>in</strong>flux pathway. Nat Cell Biol 6: 113–120, 2004.<br />

645. Smith GL, Aust<strong>in</strong> C, Crichton C, Wray S. A review of the actions<br />

and control of <strong>in</strong>tracellular pH <strong>in</strong> vascular smooth muscle. Cardiovasc<br />

Res 38: 316–331, 1998.<br />

646. Smith GL, Duncan AM, Neary P, Bruce L, Burton FL. P i <strong>in</strong>hibits<br />

the SR Ca 2 pump and stimulates pump-mediated Ca 2 leak <strong>in</strong><br />

rabbit cardiac myocytes. Am J Physiol Heart Circ Physiol 279:<br />

H577–H585, 2000.<br />

647. Smith MJ, Koch GL. Multiple zones <strong>in</strong> the sequence of calreticul<strong>in</strong><br />

(CRP55, calregul<strong>in</strong>, HACBP), a major calcium b<strong>in</strong>d<strong>in</strong>g ER/SR prote<strong>in</strong>.<br />

EMBO J 8: 3581–3586, 1989.<br />

648. Smith RD, Babiychuk EB, Noble K, Draeger A, Wray S. Increased<br />

cholesterol decreases uter<strong>in</strong>e activity: functional effects of<br />

cholesterol <strong>in</strong> pregnant rat myometrium. Am J Physiol Cell Physiol<br />

288: C982–C988, 2005.<br />

649. Smyth JT, Dehaven WI, Jones BF, Mercer JC, Trebak M,<br />

Vazquez G, Putney JW Jr. Emerg<strong>in</strong>g perspectives <strong>in</strong> store-operated<br />

Ca 2 entry: roles of Orai, Stim and TRP. Biochim Biophys<br />

Acta 1763: 1147–1160, 2006.<br />

650. Soares S, Thompson M, White T, Isbell A, Yamasaki M,<br />

Prakash Y, Lund FE, Galione A, Ch<strong>in</strong>i EN. NAADP as a second<br />

messenger: neither CD38 nor base-exchange reaction are necessary<br />

for <strong>in</strong> vivo generation of NAADP <strong>in</strong> myometrial cells. Am J Physiol<br />

Cell Physiol 292: C227–C239, 2007.<br />

651. Soboloff J, Spassova MA, Dziadek MA, Gill DL. Calcium signals<br />

mediated by STIM and Orai prote<strong>in</strong>s–a new paradigm <strong>in</strong> <strong>in</strong>terorganelle<br />

communication. Biochim Biophys Acta 1763: 1161–1168,<br />

2006.<br />

652. Somlyo AP. Excitation-contraction coupl<strong>in</strong>g and the ultrastructure<br />

of smooth muscle. Circ Res 57: 497–507, 1985.<br />

653. Somlyo AP, Somlyo AV. Signal transduction and regulation <strong>in</strong><br />

smooth muscle. Nature 372: 231–236, 1994.<br />

654. Somlyo AP, Somlyo AV. Ca 2 sensitivity of smooth muscle and<br />

nonmuscle myos<strong>in</strong> II: modulated by G prote<strong>in</strong>s, k<strong>in</strong>ases, and myos<strong>in</strong><br />

phosphatase. Physiol Rev 83: 1325–1358, 2003.<br />

655. Somlyo AV, Gonzalez-Serratos HG, Shuman H, McClellan G,<br />

Somlyo AP. Calcium release and ionic changes <strong>in</strong> the sarcoplasmic<br />

reticulum of tetanized muscle: an electron-probe study. J Cell Biol<br />

90: 577–594, 1981.<br />

656. Somlyo AV, Somlyo AP. Strontium accumulation by sarcoplasmic<br />

reticulum and mitochondria <strong>in</strong> vascular smooth muscle. Science<br />

174: 955–958, 1971.<br />

657. Sorrent<strong>in</strong>o V, Rizzuto R. Molecular genetics of Ca 2 stores and<br />

<strong>in</strong>tracellular Ca 2 signall<strong>in</strong>g. Trends Pharmacol Sci 22: 459–464,<br />

2001.<br />

658. Spassova MA, Hewavitharana T, Xu W, Soboloff J, Gill DL. A<br />

common mechanism underlies stretch activation and receptor ac-


tivation of TRPC6 channels. Proc Natl Acad Sci USA 103: 16586–<br />

16591, 2006.<br />

659. Spurway NC, Wray S. A phosphorus nuclear magnetic resonance<br />

study of metabolites and <strong>in</strong>tracellular pH <strong>in</strong> rabbit vascular smooth<br />

muscle. J Physiol 393: 57–71, 1987.<br />

660. Stathopulos PB, Zheng L, Li GY, Plev<strong>in</strong> MJ, Ikura M. Structural<br />

and mechanistic <strong>in</strong>sights <strong>in</strong>to STIM1-mediated <strong>in</strong>itiation of storeoperated<br />

calcium entry. Cell 135: 110–122, 2008.<br />

661. Steenbergen JM, Fay FS. The quantal nature of calcium release<br />

to caffe<strong>in</strong>e <strong>in</strong> s<strong>in</strong>gle smooth muscle cells results from activation of<br />

the sarcoplasmic reticulum Ca 2 -ATPase. J Biol Chem 271: 1821–<br />

1824, 1996.<br />

662. Stevenson AS, Gomez MF, Hill-Eubanks DC, Nelson MT.<br />

NFAT4 movement <strong>in</strong> native smooth muscle. A role for differential<br />

Ca 2 signal<strong>in</strong>g. J Biol Chem 276: 15018–15024, 2001.<br />

663. Stocker M. Ca 2 -activated K channels: molecular determ<strong>in</strong>ants<br />

and function of the SK family. Nat Rev Neurosci 5: 758–770, 2004.<br />

664. Strang CJ, Henson E, Okamoto Y, Paz MA, Gallop PM. Separation<br />

and determ<strong>in</strong>ation of alpha-am<strong>in</strong>o acids by boroxazolidone<br />

formation. Anal Biochem 178: 276–286, 1989.<br />

665. Strehler EE, Treiman M. Calcium pumps of plasma membrane<br />

and cell <strong>in</strong>terior. Curr Mol Med 4: 323–335, 2004.<br />

666. Stuenkel EL. S<strong>in</strong>gle potassium channels recorded from vascular<br />

smooth muscle cells. Am J Physiol Heart Circ Physiol 257: H760–<br />

H769, 1989.<br />

667. Sugiyama T, Goldman WF. Measurement of SR free Ca 2 and<br />

Mg 2 <strong>in</strong> permeabilized smooth muscle cells with use of furaptra.<br />

Am J Physiol Cell Physiol 269: C698–C705, 1995.<br />

668. Sugiyama T, Matsuda Y, Mikoshiba K. Inositol 1,4,5-trisphosphate<br />

receptor associated with focal contact cytoskeletal prote<strong>in</strong>s.<br />

FEBS Lett 466: 29–34, 2000.<br />

669. Sutko JL, Airey JA. Ryanod<strong>in</strong>e receptor Ca 2 release channels:<br />

does diversity <strong>in</strong> form equal diversity <strong>in</strong> function? Physiol Rev 76:<br />

1027–1071, 1996.<br />

670. Sutko JL, Airey JA, Welch W, Ruest L. The pharmacology of<br />

ryanod<strong>in</strong>e and related compounds. Pharmacol Rev 49: 53–98, 1997.<br />

671. Sutliff RL, Conforti L, Weber CS, Kranias EG, Paul RJ. Regulation<br />

of the spontaneous contractile activity of the portal ve<strong>in</strong> by<br />

the sarcoplasmic reticulum: evidence from the phospholamban<br />

gene-ablated mouse. Vascul Pharmacol 41: 197–204, 2004.<br />

672. Sutliff RL, Hoy<strong>in</strong>g JB, Kadambi VJ, Kranias EG, Paul RJ.<br />

Phospholamban is present <strong>in</strong> endothelial cells and modulates endothelium-dependent<br />

relaxation. Evidence from phospholamban<br />

gene-ablated mice. Circ Res 84: 360–364, 1999.<br />

673. Suzuki K, Ito KM, M<strong>in</strong>ayoshi Y, Suzuki H, Asano M, Ito K.<br />

Modification by charybdotox<strong>in</strong> and apam<strong>in</strong> of spontaneous electrical<br />

and mechanical activity of the circular smooth muscle of the<br />

gu<strong>in</strong>ea-pig stomach. Br J Pharmacol 109: 661–666, 1993.<br />

674. Swanson JE, Lokesh BR, K<strong>in</strong>sella JE. Ca 2 -Mg 2 ATPase of<br />

mouse cardiac sarcoplasmic reticulum is affected by membrane n-6<br />

and n-3 polyunsaturated fatty acid content. J Nutr 119: 364–372,<br />

1989.<br />

675. Sward K, Dreja K, L<strong>in</strong>dqvist A, Persson E, Hellstrand P.<br />

Influence of mitochondrial <strong>in</strong>hibition on global and local [Ca 2 ] i <strong>in</strong><br />

rat tail artery. Circ Res 90: 792–799, 2002.<br />

676. Sweadner KJ, Donnet C. Structural similarities of Na,K-ATPase<br />

and SERCA, the Ca 2 -ATPase of the sarcoplasmic reticulum. Biochem<br />

J 356: 685–704, 2001.<br />

677. Sweeney M, Jones CJ, Greenwood SL, Baker PN, Taggart MJ.<br />

Ultrastructural features of smooth muscle and endothelial cells of<br />

isolated isobaric human placental and maternal arteries. Placenta<br />

27: 635–647, 2006.<br />

678. Szado T, Kuo KH, Bernard-Helary K, Poburko D, Lee CH,<br />

Seow C, Ruegg UT, van Breemen C. Agonist-<strong>in</strong>duced mitochondrial<br />

Ca 2 transients <strong>in</strong> smooth muscle. FASEB J 17: 28–37, 2003.<br />

679. Tada M, Katz AM. Phosphorylation of the sarcoplasmic reticulum<br />

and sarcolemma. Annu Rev Physiol 44: 401–423, 1982.<br />

680. Tada M, Kirchberger MA, Katz AM. Phosphorylation of a 22,000dalton<br />

component of the cardiac sarcoplasmic reticulum by adenos<strong>in</strong>e<br />

3:5-monophosphate-dependent prote<strong>in</strong> k<strong>in</strong>ase. J Biol Chem<br />

250: 2640–2647, 1975.<br />

SMOOTH MUSCLE SR 175<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

681. Tada M, Yamamoto T, Tonomura Y. Molecular mechanism of<br />

active calcium transport by sarcoplasmic reticulum. Physiol Rev<br />

58: 1–79, 1978.<br />

682. Taggart MJ, Wray S. Contribution of sarcoplasmic reticular calcium<br />

to smooth muscle contractile activation: gestational dependence<br />

<strong>in</strong> isolated rat uterus. J Physiol 511: 133–144, 1998.<br />

683. Taggart MJ, Wray S. Hypoxia and smooth muscle function: key<br />

regulatory events dur<strong>in</strong>g metabolic stress. J Physiol 509: 315–325,<br />

1998.<br />

684. Takahashi A, Camacho P, Lechleiter JD, Herman B. Measurement<br />

of <strong>in</strong>tracellular calcium. Physiol Rev 79: 1089–1125, 1999.<br />

685. Takahashi Y, Watanabe H, Murakami M, Ono K, Munehisa Y,<br />

Koyama T, Nobori K, Iijima T, Ito H. <strong>Function</strong>al role of stromal<br />

<strong>in</strong>teraction molecule 1 (STIM1) <strong>in</strong> vascular smooth muscle cells.<br />

Biochem Biophys Res Commun 361: 934–940, 2007.<br />

686. Takemura H, Hughes AR, Thastrup O, Putney JW Jr. Activation<br />

of calcium entry by the tumor promoter thapsigarg<strong>in</strong> <strong>in</strong> parotid<br />

ac<strong>in</strong>ar cells. Evidence that an <strong>in</strong>tracellular calcium pool and not an<br />

<strong>in</strong>ositol phosphate regulates calcium fluxes at the plasma membrane.<br />

J Biol Chem 264: 12266–12271, 1989.<br />

687. Takeshima H, Ikemoto T, Nishi M, Nishiyama N, Shimuta M,<br />

Sugitani Y, Kuno J, Saito I, Saito H, Endo M, I<strong>in</strong>o M, Noda T.<br />

Generation and characterization of mutant mice lack<strong>in</strong>g ryanod<strong>in</strong>e<br />

receptor type 3. J Biol Chem 271: 19649–19652, 1996.<br />

688. Takeshima H, Komazaki S, Nishi M, I<strong>in</strong>o M, Kangawa K.<br />

Junctophil<strong>in</strong>s: a novel family of junctional membrane complex<br />

prote<strong>in</strong>s. Mol Cell 6: 11–22, 2000.<br />

689. Tanaka Y, Meera P, Song M, Knaus H, Toro L. Molecular<br />

constituents of maxi K Ca channels <strong>in</strong> human coronary smooth<br />

muscle: predom<strong>in</strong>ant alpha and beta subunit complexes. J Physiol<br />

502: 545–557, 1997.<br />

690. Tang DD, Gunst SJ. Depletion of focal adhesion k<strong>in</strong>ase by antisense<br />

depresses contractile activation of smooth muscle. Am J<br />

Physiol Cell Physiol 280: C874–C883, 2001.<br />

691. Tang DD, Tan J. Downregulation of profil<strong>in</strong> with antisense oligodeoxynucleotides<br />

<strong>in</strong>hibits force development dur<strong>in</strong>g stimulation<br />

of smooth muscle. Am J Physiol Heart Circ Physiol 285: H1528–<br />

H1536, 2003.<br />

692. Tansey MG, Luby-Phelps K, Kamm KE, Stull JT. Ca 2 -dependent<br />

phosphorylation of myos<strong>in</strong> light cha<strong>in</strong> k<strong>in</strong>ase decreases the<br />

Ca 2 sensitivity of light cha<strong>in</strong> phosphorylation with<strong>in</strong> smooth muscle<br />

cells. J Biol Chem 269: 9912–9920, 1994.<br />

693. Tasker PN, Michelangeli F, Nixon GF. Expression and distribution<br />

of the type 1 and type 3 <strong>in</strong>ositol 1,4,5-trisphosphate receptor <strong>in</strong><br />

develop<strong>in</strong>g vascular smooth muscle. Circ Res 84: 536–542, 1999.<br />

694. Tasker PN, Taylor CW, Nixon GF. Expression and distribution<br />

of InsP 3 receptor subtypes <strong>in</strong> proliferat<strong>in</strong>g vascular smooth muscle<br />

cells. Biochem Biophys Res Commun 273: 907–912, 2000.<br />

695. Taylor CW. Inositol trisphosphate receptors: Ca 2 -modulated <strong>in</strong>tracellular<br />

Ca 2 channels. Biochim Biophys Acta 1436: 19–33,<br />

1998.<br />

696. Terasaki M, Reese TS. Characterization of endoplasmic reticulum<br />

by co-localization of BiP and dicarbocyan<strong>in</strong>e dyes. J Cell Sci<br />

101: 315–322, 1992.<br />

697. Thar<strong>in</strong> S, Dziak E, Michalak M, Opas M. Widespread tissue<br />

distribution of rabbit calreticul<strong>in</strong>, a non-muscle functional analogue<br />

of calsequestr<strong>in</strong>. Cell Tissue Res 269: 29–37, 1992.<br />

698. Thorneloe KS, Knorn AM, Doetsch PE, Lash<strong>in</strong>ger ES, Liu AX,<br />

Bond CT, Adelman JP, Nelson MT. Small-conductance, Ca 2 -<br />

activated K channel 2 is the key functional component of SK<br />

channels <strong>in</strong> mouse ur<strong>in</strong>ary bladder. Am J Physiol Regul Integr<br />

Comp Physiol 294: R1737–R1743, 2008.<br />

699. Totary-Ja<strong>in</strong> H, Naveh-Many T, Riahi Y, Kaiser N, Eckel J,<br />

Sasson S. Calreticul<strong>in</strong> destabilizes glucose transporter-1 mRNA <strong>in</strong><br />

vascular endothelial and smooth muscle cells under high-glucose<br />

conditions. Circ Res 97: 1001–1008, 2005.<br />

700. Toyoshima C, Inesi G. Structural basis of ion pump<strong>in</strong>g by Ca 2 -<br />

ATPase of the sarcoplasmic reticulum. Annu Rev Biochem 73:<br />

269–292, 2004.<br />

701. Toyoshima C, Nakasako M, Nomura H, Ogawa H. Crystal structure<br />

of the calcium pump of sarcoplasmic reticulum at 2.6 A<br />

resolution. Nature 405: 647–655, 2000.


176 SUSAN WRAY AND THEODOR BURDYGA<br />

702. Toyoshima C, Nomura H. Structural changes <strong>in</strong> the calcium pump<br />

accompany<strong>in</strong>g the dissociation of calcium. Nature 418: 605–611,<br />

2002.<br />

703. Trafford AW, Diaz ME, Eisner DA. A novel, rapid and reversible<br />

method to measure Ca buffer<strong>in</strong>g and time-course of total sarcoplasmic<br />

reticulum Ca content <strong>in</strong> cardiac ventricular myocytes.<br />

Pflügers Arch 437: 501–503, 1999.<br />

704. Trafford AW, Diaz ME, O’Neill SC, Eisner DA. Integrative<br />

analysis of calcium signall<strong>in</strong>g <strong>in</strong> cardiac muscle. Front Biosci 7:<br />

d843–d852, 2002.<br />

705. Trepakova ES, Gericke M, Hirakawa Y, Weisbrod RM, Cohen<br />

RA, Bolot<strong>in</strong>a VM. Properties of a native cation channel activated<br />

by Ca 2 store depletion <strong>in</strong> vascular smooth muscle cells. J Biol<br />

Chem 276: 7782–7790, 2001.<br />

706. Tribe RM, Bor<strong>in</strong> ML, Blauste<strong>in</strong> MP. <strong>Function</strong>ally and spatially<br />

dist<strong>in</strong>ct Ca 2 stores are revealed <strong>in</strong> cultured vascular smooth muscle<br />

cells. Proc Natl Acad Sci USA 91: 5908–5912, 1994.<br />

707. Tribe RM, Moriarty P, Poston L. Calcium homeostatic pathways<br />

change with gestation <strong>in</strong> human myometrium. Biol Reprod 63:<br />

748–755, 2000.<br />

708. Tsugorka A, Rios E, Blatter LA. Imag<strong>in</strong>g elementary events of<br />

calcium release <strong>in</strong> skeletal muscle cells. Science 269: 1723–1726,<br />

1995.<br />

709. Tsukioka M, I<strong>in</strong>o M, Endo M. pH dependence of <strong>in</strong>ositol 1,4,5trisphosphate-<strong>in</strong>duced<br />

Ca 2 release <strong>in</strong> permeabilized smooth muscle<br />

cells of the gu<strong>in</strong>ea-pig. J Physiol 475.3: 369–375, 1994.<br />

710. Uyama Y, Imaizumi Y, Watanabe M. Cyclopiazonic acid, an<br />

<strong>in</strong>hibitor of Ca 2 -ATPase <strong>in</strong> sarcoplasmic reticulum, <strong>in</strong>creases excitability<br />

<strong>in</strong> ileal smooth muscle. Br J Pharmacol 110: 565–572,<br />

1993.<br />

711. Vallot O, Combettes L, Jourdon P, Inamo J, Marty I, Claret M,<br />

Lompre A. Intracellular Ca 2 handl<strong>in</strong>g <strong>in</strong> vascular smooth muscle<br />

cells is affected by proliferation. Arterioscler Thromb Vasc Biol 20:<br />

1225–1235, 2000.<br />

712. Valverde MA, Rojas P, Amigo J, Cosmelli D, Orio P, Bahamonde<br />

MI, Mann GE, Vergara C, Latorre R. Acute activation of<br />

Maxi-K channels (hSlo) by estradiol b<strong>in</strong>d<strong>in</strong>g to the beta subunit.<br />

Science 285: 1929–1931, 1999.<br />

713. Van Breemen C. Calcium requirement for activation of <strong>in</strong>tact<br />

aortic smooth muscle. J Physiol 272: 317–329, 1977.<br />

714. Van Breemen C, Chen Q, Laher I. Superficial buffer barrier<br />

function of smooth muscle sarcoplasmic reticulum. Trends Pharmacol<br />

Sci 16: 98–105, 1995.<br />

715. Van Breemen C, Lukeman S, Leijten P, Yamamoto H, Loutzenhiser<br />

R. The role of superficial SR <strong>in</strong> modulat<strong>in</strong>g force development<br />

<strong>in</strong>duced by Ca entry <strong>in</strong>to arterial smooth muscle. J Cardiovasc<br />

Pharmacol 8 Suppl: S111–S116, 1986.<br />

716. Van Helden DF. Spontaneous and noradrenal<strong>in</strong>e-<strong>in</strong>duced transient<br />

depolarizations <strong>in</strong> the smooth muscle of gu<strong>in</strong>ea-pig mesenteric<br />

ve<strong>in</strong>. J Physiol 437: 511–541, 1991.<br />

717. Van RC, Lazdunski M. Endothel<strong>in</strong> and vasopress<strong>in</strong> activate low<br />

conductance chloride channels <strong>in</strong> aortic smooth muscle cells.<br />

Pflügers Arch 425: 156–163, 1993.<br />

718. Vandorpe DH, Shmukler BE, Jiang L, Lim B, Maylie J, Adelman<br />

JP, de FL, Cappell<strong>in</strong>i MD, Brugnara C, Alper SL. cDNA<br />

clon<strong>in</strong>g and functional characterization of the mouse Ca 2 -gated<br />

K channel, mIK1. Roles <strong>in</strong> regulatory volume decrease and erythroid<br />

differentiation. J Biol Chem 273: 21542–21553, 1998.<br />

719. Vangheluwe P, Raeymaekers L, Dode L, Wuytack F. Modulat<strong>in</strong>g<br />

sarco(endo)plasmic reticulum Ca 2 ATPase 2 (SERCA2) activity:<br />

cell biological implications. Cell Calcium 38: 291–302, 2005.<br />

720. Vangheluwe P, Schuermans M, Zador E, Waelkens E, Raeymaekers<br />

L, Wuytack F. Sarcolip<strong>in</strong> and phospholamban mRNA<br />

and prote<strong>in</strong> expression <strong>in</strong> cardiac and skeletal muscle of different<br />

species. Biochem J 389: 151–159, 2005.<br />

721. Vassilopoulos S, Thevenon D, Rezgui SS, Brocard J, Chapel A,<br />

Lacampagne A, Lunardi J, Dewaard M, Marty I. Triad<strong>in</strong>s are<br />

not triad-specific prote<strong>in</strong>s: two new skeletal muscle triad<strong>in</strong>s possibly<br />

<strong>in</strong>volved <strong>in</strong> the architecture of sarcoplasmic reticulum. J Biol<br />

Chem 280: 28601–28609, 2005.<br />

722. Venkatachalam K, van Rossum DB, Patterson RL, Ma HT, Gill<br />

DL. The cellular and molecular basis of store-operated calcium<br />

entry. Nat Cell Biol 4: E263–E272, 2002.<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

723. Ver HM, Heymans S, Antoons G, Reed T, Periasamy M, Awede<br />

B, Lebacq J, Vangheluwe P, Dewerch<strong>in</strong> M, Collen D, Sipido K,<br />

Carmeliet P, Wuytack F. Replacement of the muscle-specific<br />

sarcoplasmic reticulum Ca 2 -ATPase isoform SERCA2a by the<br />

nonmuscle SERCA2b homologue causes mild concentric hypertrophy<br />

and impairs contraction-relaxation of the heart. Circ Res 89:<br />

838–846, 2001.<br />

724. Verboomen H, Wuytack F, De SH, Himpens B, Casteels R.<br />

<strong>Function</strong>al difference between SERCA2a and SERCA2b Ca 2<br />

pumps and their modulation by phospholamban. Biochem J 286:<br />

591–595, 1992.<br />

725. Verboomen H, Wuytack F, Eggermont JA, De JS, Missiaen L,<br />

Raeymaekers L, Casteels R. cDNA clon<strong>in</strong>g and sequenc<strong>in</strong>g of<br />

phospholamban from pig stomach smooth muscle. Biochem J 262:<br />

353–356, 1989.<br />

726. Verboomen H, Wuytack F, Van Den BL, Mertens L, Casteels R.<br />

The functional importance of the extreme C-term<strong>in</strong>al tail <strong>in</strong> the<br />

gene 2 organellar Ca 2 -transport ATPase (SERCA2a/b). Biochem J<br />

303: 979–984, 1994.<br />

727. Vergara C, Latorre R, Marrion NV, Adelman JP. Calcium-activated<br />

potassium channels. Curr Op<strong>in</strong> Neurobiol 8: 321–329, 1998.<br />

728. Verkhratsky A. Physiology and pathophysiology of the calcium<br />

store <strong>in</strong> the endoplasmic reticulum of neurons. Physiol Rev 85:<br />

201–279, 2005.<br />

729. Villa A, Pod<strong>in</strong>i P, Panzeri MC, Sol<strong>in</strong>g HD, Volpe P, Meldolesi<br />

J. The endoplasmic-sarcoplasmic reticulum of smooth muscle:<br />

immunocytochemistry of vas deferens fibres reveals specialized<br />

subcompartments differently equipped for the control of Ca 2<br />

homeostasis. J Cell Biol 121: 1041–1051, 1993.<br />

730. Volpe P, Mart<strong>in</strong>i A, Furlan S, Mendolesi J. Calsequestr<strong>in</strong> is a<br />

component of smooth muscles: the skeletal- and cardiac-muscle<br />

isoforms are both present, although <strong>in</strong> highly variable amounts and<br />

ratios. Biochem J 301: 465–469, 1994.<br />

731. Wagner-Mann C, Hu Q, Sturek M. Multiple effects of ryanod<strong>in</strong>e<br />

on <strong>in</strong>tracellular free Ca 2 <strong>in</strong> smooth muscle cells from bov<strong>in</strong>e and<br />

porc<strong>in</strong>e coronary artery: modulation of sarcoplasmic reticulum<br />

function. Br J Pharmacol 105: 903–911, 1992.<br />

732. Wakabayashi I, Poteser M, Groschner K. Intracellular pH as a<br />

determ<strong>in</strong>ant of vascular smooth muscle function. J Vasc Res 43:<br />

238–250, 2006.<br />

733. Wamhoff BR, Bowles DK, Owens GK. Excitation-transcription<br />

coupl<strong>in</strong>g <strong>in</strong> arterial smooth muscle. Circ Res 98: 868–878, 2006.<br />

734. Wang S, Trumble WR, Liao H, Wesson CR, Dunker AK, Kang<br />

CH. Crystal structure of calsequestr<strong>in</strong> from rabbit skeletal muscle<br />

sarcoplasmic reticulum. Nat Struct Biol 5: 476–483, 1998.<br />

735. Wang HJ, Guay G, Pogan L, Sauve R, Nabi IR. Calcium regulates<br />

the association between mitochondria and a smooth subdoma<strong>in</strong> of<br />

the endoplasmic reticulum. J Cell Biol 150: 1489–1498, 2000.<br />

736. Wang Q, Hogg RC, Large WA. Properties of spontaneous <strong>in</strong>ward<br />

currents recorded <strong>in</strong> smooth muscle cells isolated from the rabbit<br />

portal ve<strong>in</strong>. J Physiol 451: 525–537, 1992.<br />

737. Wang Q, Large WA. Action of histam<strong>in</strong>e on s<strong>in</strong>gle smooth muscle<br />

cells dispersed from the rabbit pulmonary artery. J Physiol 468:<br />

125–139, 1993.<br />

738. Wang Y, Deng X, Hewavitharana T, Soboloff J, Gill DL. Stim,<br />

orai and trpc channels <strong>in</strong> the control of calcium entry signals <strong>in</strong><br />

smooth muscle. Cl<strong>in</strong> Exp Pharmacol Physiol 35: 1127–1133, 2008.<br />

739. Wang Y, Kotlikoff MI. Inactivation of calcium-activated chloride<br />

channels <strong>in</strong> smooth muscle by calcium/calmodul<strong>in</strong>-dependent prote<strong>in</strong><br />

k<strong>in</strong>ase. Proc Natl Acad Sci USA 94: 14918–14923, 1997.<br />

740. Wang YX, Zheng YM, Abdullaev I, Kotlikoff MI. Metabolic<br />

<strong>in</strong>hibition with cyanide <strong>in</strong>duces calcium release <strong>in</strong> pulmonary artery<br />

myocytes and Xenopus oocytes. Am J Physiol Cell Physiol<br />

284: C378–C388, 2003.<br />

741. Ward SM, Baker SA, De FA, Sanders KM. Propagation of slow<br />

waves requires IP 3 receptors and mitochondrial Ca 2 uptake <strong>in</strong><br />

can<strong>in</strong>e colonic muscles. J Physiol 549: 207–218, 2003.<br />

742. Ward SM, Ordog T, Koh SD, Baker SA, Jun JY, Amberg G,<br />

Monaghan K, Sanders KM. Pacemak<strong>in</strong>g <strong>in</strong> <strong>in</strong>terstitial cells of<br />

Cajal depends upon calcium handl<strong>in</strong>g by endoplasmic reticulum<br />

and mitochondria. J Physiol 525: 355–361, 2000.


743. Warren GB, Houslay MD, Metcalfe JC, Birdsall NJ. Cholesterol<br />

is excluded from the phospholipid annulus surround<strong>in</strong>g an active<br />

calcium transport prote<strong>in</strong>. Nature 255: 684–687, 1975.<br />

744. Waser M, Mesaeli N, Spencer C, Michalak M. Regulation of<br />

calreticul<strong>in</strong> gene expression by calcium. J Cell Biol 138: 547–557,<br />

1997.<br />

745. Watras J. Regulation of calcium uptake <strong>in</strong> bov<strong>in</strong>e aortic sarcoplasmic<br />

reticulum by cyclic AMP-dependent prote<strong>in</strong> k<strong>in</strong>ase. J Mol Cell<br />

Cardiol 20: 711–723, 1988.<br />

746. Wayman CP, McFadzean I, Gibson A, Tucker JF. Two dist<strong>in</strong>ct<br />

membrane currents activated by cyclopiazonic acid-<strong>in</strong>duced calcium<br />

store depletion <strong>in</strong> s<strong>in</strong>gle smooth muscle cells of the mouse<br />

anococcygeus. Br J Pharmacol 117: 566–572, 1996.<br />

747. Wei AD, Gutman GA, Aldrich R, Chandy KG, Grissmer S,<br />

Wulff H. International Union of Pharmacology. LII. Nomenclature<br />

and molecular relationships of calcium-activated potassium channels.<br />

Pharmacol Rev 57: 463–472, 2005.<br />

748. Weirich J, Dumont L, Fleckenste<strong>in</strong>-Grun G. Contribution of<br />

store-operated Ca 2 entry to pH o-dependent changes <strong>in</strong> vascular<br />

tone of porc<strong>in</strong>e coronary smooth muscle. Cell Calcium 35: 9–20,<br />

2004.<br />

749. Wellman GC, Nathan DJ, Saundry CM, Perez G, Bonev AD,<br />

Penar PL, Tranmer BI, Nelson MT. Ca 2 sparks and their function<br />

<strong>in</strong> human cerebral arteries. Stroke 33: 802–808, 2002.<br />

750. Wellman GC, Nelson MT. Signal<strong>in</strong>g between SR and plasmalemma<br />

<strong>in</strong> smooth muscle: sparks and the activation of Ca 2 -sensitive<br />

ion channels. Cell Calcium 34: 211–229, 2003.<br />

751. Wellman GC, Santana LF, Bonev AD, Nelson MT. Role of the<br />

phospholamban <strong>in</strong> the modulation of arterial Ca 2 sparks and<br />

Ca 2 -activated K channels by cAMP. Am J Physiol Cell Physiol<br />

281: C1029–C1037, 2001.<br />

752. Werner ME, Zvara P, Meredith AL, Aldrich RW, Nelson MT.<br />

Erectile dysfunction <strong>in</strong> mice lack<strong>in</strong>g the large-conductance calciumactivated<br />

potassium (BK) channel. J Physiol 567: 545–556, 2005.<br />

753. White C, McGeown G. Imag<strong>in</strong>g of changes <strong>in</strong> sarcoplasmic reticulum<br />

[Ca 2 ] us<strong>in</strong>g Oregon Green BAPTA 5N and confocal laser<br />

scann<strong>in</strong>g microscopy. Cell Calcium 31: 151–159, 2002.<br />

754. White C, McGeown JG. Ca 2 uptake by the sarcoplasmic reticulum<br />

decreases the amplitude of depolarization-dependent [Ca 2 ] i<br />

transients <strong>in</strong> rat gastric myocytes. Pflügers Arch 440: 488–495,<br />

2000.<br />

755. White C, McGeown JG. Carbachol triggers RyR-dependent Ca 2<br />

release via activation of IP 3 receptors <strong>in</strong> isolated rat gastric myocytes.<br />

J Physiol 542: 725–733, 2002.<br />

756. White C, McGeown JG. Inositol 1,4,5-trisphosphate receptors<br />

modulate Ca 2 sparks and Ca 2 store content <strong>in</strong> vas deferens<br />

myocytes. Am J Physiol Cell Physiol 285: C195–C204, 2003.<br />

757. White TA, Kannan MS, Walseth TF. Intracellular calcium signal<strong>in</strong>g<br />

through the cADPR pathway is agonist specific <strong>in</strong> porc<strong>in</strong>e<br />

airway smooth muscle. FASEB J 17: 482–484, 2003.<br />

758. Wibo M, Godfra<strong>in</strong>d T. Comparative localization of <strong>in</strong>ositol 1,4,5trisphosphate<br />

and ryanod<strong>in</strong>e receptors <strong>in</strong> <strong>in</strong>test<strong>in</strong>al smooth muscle:<br />

an analytical subfractionation study. Biochem J 297: 415–423, 1994.<br />

759. Wier WG, Yue DT, Marban E. Effects of ryanod<strong>in</strong>e on <strong>in</strong>tracellular<br />

Ca 2 transients <strong>in</strong> mammalian cardiac muscle. Federation<br />

Proc 44: 2989–2993, 1985.<br />

760. Wilcox RA, Challiss RA, Liu C, Potter BV, Nahorski SR. Inositol-1,3,4,5-tetrakisphosphate<br />

<strong>in</strong>duces calcium mobilization via the<br />

<strong>in</strong>ositol-1,4,5-trisphosphate receptor <strong>in</strong> SH-SY5Y neuroblastoma<br />

cells. Mol Pharmacol 44: 810–817, 1993.<br />

761. Williams DB. Beyond lect<strong>in</strong>s: the calnex<strong>in</strong>/calreticul<strong>in</strong> chaperone<br />

system of the endoplasmic reticulum. J Cell Sci 119: 615–623, 2006.<br />

762. Williams JH, Vidt SE, R<strong>in</strong>ehart J. Measurement of sarcoplasmic<br />

reticulum Ca 2 ATPase activity us<strong>in</strong>g high-performance liquid<br />

chromatography. Anal Biochem 372: 135–139, 2008.<br />

763. Williams JH, Ward CW. Reduced Ca 2 -<strong>in</strong>duced Ca 2 release from<br />

skeletal muscle sarcoplasmic reticulum at low pH. Can J Physiol<br />

Pharmacol 70: 926–930, 1992.<br />

764. Wilson DP, Sutherland C, Walsh MP. Ca 2 activation of smooth<br />

muscle contraction. Evidence for the <strong>in</strong>volvement of calmodul<strong>in</strong><br />

that is bound to the Triton-<strong>in</strong>soluble fraction even <strong>in</strong> the absence of<br />

Ca 2 . J Biol Chem 277: 2186–2192, 2002.<br />

SMOOTH MUSCLE SR 177<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

765. Wojcikiewicz RJ. Type I, II, and III <strong>in</strong>ositol 1,4,5-trisphosphate<br />

receptors are unequally susceptible to down-regulation and are<br />

expressed <strong>in</strong> markedly different proportions <strong>in</strong> different cell types.<br />

J Biol Chem 270: 11678–11683, 1995.<br />

766. Wojcikiewicz RJH, Luo SG. Differences among type I, II, and III<br />

<strong>in</strong>ositol-1,4,5-trisphosphate receptors <strong>in</strong> ligand-b<strong>in</strong>d<strong>in</strong>g aff<strong>in</strong>ity <strong>in</strong>fluence<br />

the sensitivity of calcium stores to <strong>in</strong>ositol-1,4,5-trisphosphate.<br />

Mol Pharmacol 53: 656–662, 1998.<br />

767. Worley PF, Baraban JM, Supattapone S, Wilson V, Snyder SH.<br />

Characterization of <strong>in</strong>ositol trisphosphate receptor b<strong>in</strong>d<strong>in</strong>g <strong>in</strong><br />

bra<strong>in</strong>. Regulation by pH and calcium. J Biol Chem 262: 12132–<br />

12136, 1987.<br />

768. Wray S. <strong>Smooth</strong> muscle <strong>in</strong>tracellular pH: measurement, regulation,<br />

and function. Am J Physiol Cell Physiol 254: C213–C225, 1988.<br />

769. Wray S, Burdyga T, Noble K. Calcium signall<strong>in</strong>g <strong>in</strong> smooth muscle.<br />

Cell Calcium 397–407, 2005.<br />

770. Wray S, Dugg<strong>in</strong>s K, Iles R, Nyman L, Osman VA. The effects of<br />

metabolic <strong>in</strong>hibition and <strong>in</strong>tracellular pH on rat uter<strong>in</strong>e force production.<br />

Exp Physiol 77: 307–319, 1992.<br />

771. Wray S, Jones K, Kupittayanant S, Matthew AJG, Monir-<br />

Bishty E, Noble K, Pierce SJ, Quenby S, Shmygol AV. Calcium<br />

signall<strong>in</strong>g and uter<strong>in</strong>e contractility. J Soc Gynecol Invest 10: 252–<br />

264, 2003.<br />

772. Wray S, Smith RD. Mechanisms of action of pH-<strong>in</strong>duced effects on<br />

vascular smooth muscle. Mol Cell Biochem 263: 163–172, 2004.<br />

773. Wray S, Tofts P. Direct <strong>in</strong> vivo measurement of absolute metabolite<br />

concentrations us<strong>in</strong>g 31 P nuclear magnetic resonance spectroscopy.<br />

Biochim Biophys Acta 886: 399–405, 1986.<br />

774. Wu KD, Bungard D, Lytton J. Regulation of SERCA Ca 2 pump<br />

expression by cytoplasmic [Ca 2 ] <strong>in</strong> vascular smooth muscle cells.<br />

Am J Physiol Cell Physiol 280: C843–C851, 2001.<br />

775. Wu MM, Buchanan J, Luik RM, Lewis RS. Ca 2 store depletion<br />

causes STIM1 to accumulate <strong>in</strong> ER regions closely associated with<br />

the plasma membrane. J Cell Biol 174: 803–813, 2006.<br />

776. Wu Y, Kuzma J, Marechal E, Graeff R, Lee HC, Foster R, Chua<br />

NH. Abscisic acid signal<strong>in</strong>g through cyclic ADP-ribose <strong>in</strong> plants.<br />

Science 278: 2126–2130, 1997.<br />

777. Wuytack F, Raeymaekers L, Missiaen L. Molecular physiology<br />

of the SERCA and SPCA pumps. Cell Calcium 32: 279–305, 2002.<br />

778. Wuytack F, Raeymaekers L, Verbist J, nes LR, Casteels R.<br />

<strong>Smooth</strong>-muscle endoplasmic reticulum conta<strong>in</strong>s a cardiac-like<br />

form of calsequestr<strong>in</strong>. Biochim Biophys Acta 899: 151–158, 1987.<br />

779. Xiong Z, Sperelakis N, Noffs<strong>in</strong>ger A, Fenoglio-Preiser C.<br />

Changes <strong>in</strong> calcium channel current densities <strong>in</strong> rat colonic smooth<br />

muscle cells dur<strong>in</strong>g development and ag<strong>in</strong>g. Am J Physiol Cell<br />

Physiol 265: C617–C625, 1993.<br />

780. Xu KY, Zweier JL, Becker LC. <strong>Function</strong>al coupl<strong>in</strong>g between<br />

glycolysis and sarcoplasmic reticulum Ca 2 transport. Circ Res 77:<br />

88–97, 1995.<br />

781. Xu L, Lai A, Cohn A, Etter E, Guerrero A, Fay FS, Meissner G.<br />

Evidence for a Ca 2 -gated ryanod<strong>in</strong>e-sensitive Ca 2 release channel<br />

<strong>in</strong> visceral smooth muscle. Proc Natl Acad Sci USA 91: 3294–<br />

3298, 1994.<br />

782. Xu L, Mann G, Meissner G. Regulation of cardiac Ca 2 release<br />

channel (ryanod<strong>in</strong>e receptor) by Ca 2 , H , Mg 2 , and aden<strong>in</strong>e<br />

nucleotides under normal and simulated ischemic conditions. Circ<br />

Res 79: 1100–1109, 1996.<br />

783. Xu L, Tripathy A, Pasek DA, Meissner G. Potential for pharmacology<br />

of ryanod<strong>in</strong>e receptor/calcium release channels. Ann NY<br />

Acad Sci 853: 130–148, 1998.<br />

784. Xuan YT, Wang OL, Whorton AR. Thapsigarg<strong>in</strong> stimulates Ca 2<br />

entry <strong>in</strong> vascular smooth muscle cells: nicardip<strong>in</strong>e-sensitive and -<strong>in</strong>sensitive<br />

pathways. Am J Physiol Cell Physiol 262: C1258–C1265,<br />

1992.<br />

785. Yamaguchi M, Kanazawa T. Co<strong>in</strong>cidence of H b<strong>in</strong>d<strong>in</strong>g and Ca 2<br />

dissociation <strong>in</strong> the sarcoplasmic reticulum Ca-ATPase dur<strong>in</strong>g ATP<br />

hydrolysis. J Biol Chem 260: 4896–4900, 1985.<br />

786. Yang J, Clark JW Jr, Bryan RM, Robertson CS. The myogenic<br />

response <strong>in</strong> isolated rat cerebrovascular arteries: vessel model.<br />

Med Eng Phys 25: 711–717, 2003.<br />

787. Yang XR, L<strong>in</strong> MJ, Yip KP, Jeyakumar LH, Fleischer S, Leung<br />

GP, Sham JS. Multiple ryanod<strong>in</strong>e receptor subtypes and heterogeneous<br />

ryanod<strong>in</strong>e receptor-gated Ca 2 stores <strong>in</strong> pulmonary arte-


178 SUSAN WRAY AND THEODOR BURDYGA<br />

rial smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 289:<br />

L338–L348, 2005.<br />

788. Yerom<strong>in</strong> AV, Zhang SL, Jiang W, Yu Y, Safr<strong>in</strong>a O, Cahalan MD.<br />

Molecular identification of the CRAC channel by altered ion selectivity<br />

<strong>in</strong> a mutant of Orai. Nature 443: 226–229, 2006.<br />

789. Yoshikawa A, van Breemen C, Isenberg G. Buffer<strong>in</strong>g of plasmalemmal<br />

Ca 2 current by sarcoplasmic reticulum of gu<strong>in</strong>ea pig<br />

ur<strong>in</strong>ary bladder myocytes. Am J Physiol Cell Physiol 271: C833–<br />

C841, 1996.<br />

790. Young HS, Jones LR, Stokes DL. Locat<strong>in</strong>g phospholamban <strong>in</strong><br />

co-crystals with Ca 2 -ATPase by cryoelectron microscopy. Biophys<br />

J 81: 884–894, 2001.<br />

791. Young HS, Stokes DL. The mechanics of calcium transport. J<br />

Membr Biol 198: 55–63, 2004.<br />

792. Young RC, Mathur SP. Focal sarcoplasmic reticulum calcium<br />

stores and diffuse <strong>in</strong>ositol 1,4,5-trisphosphate and ryanod<strong>in</strong>e receptors<br />

<strong>in</strong> human myometrium. Cell Calcium 26: 69–75, 1999.<br />

793. Young RC, Schumann R, Zhang P. Intracellular calcium gradients<br />

<strong>in</strong> cultured human uter<strong>in</strong>e smooth muscle: a functionally important<br />

subplasmalemmal space. Cell Calcium 29: 183–189, 2001.<br />

794. Yule DI. Subtype-specific regulation of <strong>in</strong>ositol 1,4,5-trisphosphate<br />

receptors: controll<strong>in</strong>g calcium signals <strong>in</strong> time and space. J Gen<br />

Physiol 117: 431–434, 2001.<br />

795. Yusufi AN, Cheng J, Thompson MA, Burnett JC, Grande JP.<br />

Differential mechanisms of Ca 2 release from vascular smooth<br />

muscle cell microsomes. Exp Biol Med 227: 36–44, 2002.<br />

796. Yusufi AN, Cheng J, Thompson MA, Ch<strong>in</strong>i EN, Grande JP.<br />

Nicot<strong>in</strong>ic acid-aden<strong>in</strong>e d<strong>in</strong>ucleotide phosphate (NAADP) elicits<br />

specific microsomal Ca 2 release from mammalian cells. Biochem<br />

J 353: 531–536, 2001.<br />

797. Zhang J, Bricker L, Wray S, Quenby S. Poor uter<strong>in</strong>e contractility<br />

<strong>in</strong> obese women. Br J Obstet Gynaecol 114: 343–348, 2007.<br />

798. Zhang J, Kendrick A, Quenby S, Wray S. Contractility and<br />

calcium signall<strong>in</strong>g of human myometrium are profoundly affected<br />

by cholesterol manipulation: implications for labour? Reprod Sci<br />

14: 456–466, 2007.<br />

799. Zhang F, Li PL. Reconstitution and characterization of a nicot<strong>in</strong>ic<br />

acid aden<strong>in</strong>e d<strong>in</strong>ucleotide phosphate (NAADP)-sensitive Ca 2 release<br />

channel from liver lysosomes of rats. J Biol Chem 282:<br />

25259–25269, 2007.<br />

800. Zhang F, Zhang G, Zhang AY, Koeberl MJ, Wallander E, Li PL.<br />

Production of NAADP and its role <strong>in</strong> Ca 2 mobilization associated<br />

with lysosomes <strong>in</strong> coronary arterial myocytes. Am J Physiol Heart<br />

Circ Physiol 291: H274–H282, 2006.<br />

801. Zhang J, Lee MY, Cavalli M, Chen L, Berra-Romani R, Balke<br />

CW, Bianchi G, Ferrari P, Hamlyn JM, Iwamoto T, L<strong>in</strong>grel JB,<br />

Matteson DR, Wier WG, Blauste<strong>in</strong> MP. Sodium pump alpha2<br />

subunits control myogenic tone and blood pressure <strong>in</strong> mice.<br />

J Physiol 569: 243–256, 2005.<br />

Physiol Rev VOL 90 JANUARY 2010 www.prv.org<br />

802. Zhang L, Bradley ME, Buxton IL. Inositolpolyphosphate b<strong>in</strong>d<strong>in</strong>g<br />

sites and their likely role <strong>in</strong> calcium regulation <strong>in</strong> smooth muscle.<br />

Int J Biochem Cell Biol 27: 1231–1248, 1995.<br />

803. Zhang L, Kelley J, Schmeisser G, Kobayashi YM, Jones LR.<br />

Complex formation between junct<strong>in</strong>, triad<strong>in</strong>, calsequestr<strong>in</strong>, and the<br />

ryanod<strong>in</strong>e receptor. Prote<strong>in</strong>s of the cardiac junctional sarcoplasmic<br />

reticulum membrane. J Biol Chem 272: 23389–23397, 1997.<br />

804. Zhang SL, Yerom<strong>in</strong> AV, Zhang XH, Yu Y, Safr<strong>in</strong>a O, Penna A,<br />

Roos J, Stauderman KA, Cahalan MD. Genome-wide RNAi<br />

screen of Ca 2 <strong>in</strong>flux identifies genes that regulate Ca 2 releaseactivated<br />

Ca 2 channel activity. Proc Natl Acad Sci USA 103:<br />

9357–9362, 2006.<br />

805. Zheng YM, Wang QS, Rathore R, Zhang Josep WH, Mazurkiewicz<br />

E, Sorrent<strong>in</strong>o V, S<strong>in</strong>ger HA, Kotlikoff MI, Wang YX.<br />

Neurotransmitter-<strong>in</strong>duced calcium release and contraction <strong>in</strong> pulmonary<br />

artery smooth muscle cells. Am J Physiol Lung Cell Mol<br />

Physiol 289: L338–L348, 2005.<br />

806. Zholos AV, Baidan LV, Shuba MF. Properties of the late transient<br />

outward current <strong>in</strong> isolated <strong>in</strong>test<strong>in</strong>al smooth muscle cells of the<br />

gu<strong>in</strong>ea-pig. J Physiol 443: 555–574, 1991.<br />

807. ZhuGe R, Fogarty KE, Baker SP, McCarron JG, Tuft RA,<br />

Lifshitz LM, Walsh JV Jr. Ca 2 spark sites <strong>in</strong> smooth muscle cells<br />

are numerous and differ <strong>in</strong> number of ryanod<strong>in</strong>e receptors, largeconductance<br />

K channels, and coupl<strong>in</strong>g ratio between them. Am J<br />

Physiol Cell Physiol 287: C1577–C1588, 2004.<br />

808. ZhuGe R, Fogarty KE, Tuft RA, Lifshitz LM, Sayar K, Walsh<br />

JV. Dynamics of signal<strong>in</strong>g between Ca 2 sparks and Ca 2 -activated<br />

K channels studied wih a novel image-based method for direct<br />

<strong>in</strong>tracellular measurement of ryanod<strong>in</strong>e receptor Ca 2 current.<br />

J Gen Physiol 116: 845–864, 2000.<br />

809. ZhuGe R, Fogarty KE, Tuft RA, Walsh JV Jr. Spontaneous<br />

transient outward currents arise from microdoma<strong>in</strong>s where BK<br />

channels are exposed to a mean Ca 2 concentration on the order of<br />

10 M dur<strong>in</strong>g a Ca 2 spark. J Gen Physiol 120: 15–27, 2002.<br />

810. ZhuGe R, Sims SM, Tuft RA, Fogarty KE, Walsh JV. Ca 2<br />

sparks activate K and Cl channels, result<strong>in</strong>g <strong>in</strong> spontaneous<br />

transient currents <strong>in</strong> gu<strong>in</strong>ea-pig tracheal myocytes. J Physiol 513:<br />

711–718, 1998.<br />

811. ZhuGe R, Tuft RA, Fogarty KE, Bellve K, Fay FS, Walsh JV.<br />

The <strong>in</strong>fluence of sarcoplasmic reticulum Ca 2 concentration on<br />

Ca 2 sparks and spontaneous transient outward currents <strong>in</strong> s<strong>in</strong>gle<br />

smooth muscle cells. J Gen Physiol 113: 215–228, 1999.<br />

812. Zorzato F, Fujii J, Otsu K, Phillips M, Green NM, Lai FA,<br />

Meissner G, MacLennan DH. Molecular clon<strong>in</strong>g of cDNA encod<strong>in</strong>g<br />

human and rabbit forms of the Ca 2 release channel (ryanod<strong>in</strong>e<br />

receptor) of skeletal muscle sarcoplasmic reticulum. J Biol Chem<br />

265: 2244–2256, 1990.<br />

813. Zou J, Hofer AM, Lurtz MM, Gadda G, Ellis AL, Chen N,<br />

Huang Y, Holder A, Ye Y, Louis CF, Welshhans K, Rehder V,<br />

Yang JJ. Develop<strong>in</strong>g sensors for real-time measurement of high<br />

Ca 2 concentrations. Biochemistry 46: 12275–12288, 2007.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!