<<

Comprehensive Summaries of Uppsala Dissertations from the Faculty of Medicine 1105

______

Alpha-2 Adrenergic Receptors and

Effector Output in Relation to G- Coupling and Signalling Cross-Talk

BY

JOHNNY NÄSMAN

ACTA UNIVERSITATIS UPSALIENSIS UPPSALA 2001 Dissertation for the Degree of Doctor of Philosophy (Faculty of Medicine) in Physiology presented at Uppsala University in 2002

ABSTRACT

Näsman, J. 2001. Alpha-2 Adrenergic Receptors and Signal Transduction. Effector Output in Relation to G-Protein Coupling and Signalling Cross-Talk. Acta Universitatis Upsaliensis. Comprehensive Summaries of Uppsala Dissertations from the Faculty of Medicine 1105. 64 pp. Uppsala. ISBN 91-554-5195-0.

The alpha-2 adrenergic (α2-AR) subfamily includes three different subtypes, α2A-, α2B- and α2C-AR, all believed to exert their function through heterotrimeric Gi/o-. The present study was undertaken in order to investigate subtype differences in terms of cellular response and to explore other potential signalling pathways of α2-ARs. Evidence is provided for a strong Gs-protein coupling capability of the α2B-AR, leading to stimulation of (AC). The difference between the α2A- and α2B-AR subtypes, in this respect, was shown to be due to differences in the second intracellular loops of the receptor proteins. Substitution of the second loop in α2A-AR with the corresponding domain of α2B-AR enrolled the chimeric α2A/α2B receptor with functional α2B-AR properties. Dual Gi and Gs coupling can have different consequences for AC output. Using coexpression of receptors and G-proteins, it was shown that the ultimate cellular response of α2B-AR activation is largely dependent on the ratio of Gi- to Gs-protein amounts in the . Also Gi- and Go-proteins appear to have different regulatory influences on AC. Heterologous expression of AC2 together with Gi or Go and the α2A-AR resulted in receptor-mediated inhibition of C-stimulated AC2 activity through Go, whereas activation of Gi potentiated the activity. 2+ α2-ARs mobilize Ca in response to agonists in some cell types. This response was shown to depend on tonic purinergic receptor activity in transfected CHO cells. Elimination 2+ of the tonic receptor activity almost completely inhibited the Ca response of α2-ARs. In conclusion, α2-ARs can couple to multiple G-proteins, including Gi, Go and Gs. The cellular response to α2-AR activation depends on which receptor subtype is expressed, which cellular signalling constituents are engaged (G-proteins and effectors), and the signalling status of the effectors (dormant or primed).

Key words: Adrenergic, receptor, G-protein, adenylyl cyclase, cAMP, calcium, cross-talk.

Johnny Näsman, Department of Physiology, Uppsala University, Biomedical Center, P.O. Box 572, SE-751 23 Uppsala, Sweden

 Johnny Näsman 2001

ISSN 0282-7476 ISBN 91-554-5195-0

Printed in Sweden by Uppsala University, Tryck & Medier, Uppsala 2001 The thesis is based on the following papers, which are referred to in the text by their Roman numerals:

I Dual signalling by different octopamine receptors converges on adenylate cyclase in Sf9 cells. Näsman J., Kukkonen J.P., Åkerman K.E.O. Insect Biochem. Mol. Biol., In press 2001.

II The second intracellular loop of the α2-adrenergic receptors determines subtype-specific coupling to cAMP production. Näsman J., Jansson C., Åkerman K.E.O. J. Biol. Chem., 272, 9703-9708, 1997.

III Role of G-protein availability in differential signaling by alpha 2- adrenoceptors. Näsman J., Kukkonen J.P., Ammoun S., Åkerman K.E.O. Biochem. Pharmacol., 62, 913-922, 2001.

IV Endogenous extracellular purine redirect α2-adrenoceptor signaling. Åkerman K.E.O., Näsman J., Lund P.-E., Shariatmadari R., Kukkonen J.P. FEBS Lett., 430, 209-212, 1998.

V Modulation of adenylyl cyclase type 2 activity by Gi/o-protein coupled

receptors. Opposite regulation by Gi and Go. Näsman J., Kukkonen J.P., Holmqvist T., Åkerman K.E.O. Manuscript 2001.

Reprints were made with permission from the publishers. CONTENTS

Abbreviations...... 3 1 Introduction ...... 4 1.1 General background...... 4 1.2 G-protein coupled receptors and signal transduction...... 5 1.2.1 Receptors ...... 6 1.2.2 Transducers ...... 7 1.2.3 Effectors ...... 10 1.2.3.1 Adenylyl cyclase, cAMP, and PKA...... 10 1.2.3.2 C, Ca2+, and PKC ...... 13

1.3 α2-adrenergic receptors ...... 15 1.3.1 A subfamily of adrenergic receptors ...... 15 1.3.2 Physiological functions ...... 17 1.3.3 Cellular signalling ...... 19 1.4 Other receptors utilized in the study...... 21 1.4.1 Octopamine receptors...... 21 1.4.2 Purinergic receptors...... 22 1.4.3 Muscarinic receptors ...... 23 1.5 Why study heterologously expressed receptors? ...... 23 2 Aims of the study...... 25 3 Materials and methods...... 26 3.1 Cell cultures...... 26 3.2 BEVS and infection procedures ...... 26 3.3 Receptor quantification ...... 27 3.4 Receptor−G-protein interactions ...... 27 3.5 Measurement of cAMP ...... 28 3.6 Measurement of Ca2+...... 29 3.7 Mathematical modelling...... 29 4 Results ...... 30 4.1 Receptor signalling in Sf9 cells...... 30

4.2 Propensity of α2-ARs for Gs coupling...... 32 4.3 Signalling cross-talk...... 34 5 Discussion...... 36 5.1 Suitability of Sf9 cells for receptor characterization...... 36 5.2 Promiscuous G-protein coupling...... 39 5.3 Adenylyl cyclase regulation by the Ca2+ signalling pathway ...... 42 2+ 5.4 The α2-AR−Ca link ...... 43 6 Summary and conclusions...... 46 7 Acknowledgements ...... 48 8 References ...... 50 ABBREVIATIONS

AC adenylyl cyclase AR (as in α-AR and β-AR) BEVS Baculovirus Expression Vector System 2+ 2+ [Ca ]i intracellular concentration of free Ca CaM CaMK calmodulin kinase CHO Chinese hamster ovary (a cell line) Ctx toxin DAG diacylglycerol GPCR G-protein coupled receptor

Gx-protein heterotrimeric GTP-binding protein with x identity of α subunit

Gαx α subunit of heterotrimeric G-protein with x identity Gβγ βγ subunit complex of heterotrimeric G-protein i2/i3-loop second/third intracellular loop of receptors IBMX 3-isobutyl-1-methylxanthine

IP3 inositol 1,4,5-trisphosphate PDE PH pleckstrin homology PI phosphtidylinositol

PIP2 phosphtidylinositol 4,5-bisphosphate PK protein kinase (as in PKA and PKC)

PL phospholipase (as in PLC, PLA2, PLD) Ptx Sf9 Spodoptera frugiperda (a cell line) TM transmembrane domain of receptors TPA 12-O-tetradecanoyl phorbol-13-acetate

3 1 INTRODUCTION

1.1 General background All cells, whether single cell organisms or entities within multicellular organisms, need to sense the exterior to be able to adapt to changes. In the course of evolution, a multitude of such sensing molecules has evolved. A glimpse of this multitude comes from the realization that cells of our own bodies are regulated by a diversity of signals ranging from physical phenomena, such as light, through elementary ions, like calcium, to numerous and . The action of hormones and neurotransmitters is of prime importance and interest, which is reflected by the fact that the majority of drugs used in medical care are targeted toward the sites of action of these molecules. Some hormones are lipophilic and, thus, readily traverse the to exert their effects in the cell. Other hormones, and the neurotransmitters, bind to and activate plasma membrane receptors to convey the signal to the interior of the cell. In this way, a cell will be predisposed to the action of a particular only if the appropriate receptor is expressed, thereby ascertaining a level of selectivity

in hormone signalling. A majority of the cell surface receptors, including the α2-

adrenergic receptors (α2-ARs), belong to the superfamily of G-protein coupled receptors (GPCRs). Intracellular signalling by GPCRs is an area that has generated a large amount of scientific knowledge during the last decades. A major breakthrough in elucidation of hormone action via receptors came from work by Earl Sutherland in the 1950s and 1960s. Studying the effect of and on metabolism in the liver, he discovered that the hormone action on can be reconstituted in a cell-free system, as long as the particulate fraction of the cell material is included. This led to the discovery of cyclic 3’,5’-monophosphate (cAMP), a messenger molecule between hormone receptor and target , and the identification of the enzyme responsible for its synthesis, adenylyl cyclase (AC) (reviewed in Sutherland, 1972). Since then, cAMP has been implicated, not only in metabolic regulation, but in virtually all physiological processes studied. Most of the

4 actions of cAMP are mediated by (PKA). Another ubiquitous enzyme, involved in signal interpretation, is protein kinase C (PKC). A role for PKC in intracellular signalling was not recognized until it was demonstrated that the enzyme was activated by diacylglycerol (DAG) (reviewed in Nishizuka, 1984), a product of phosphatidylinositol (PI) breakdown resulting from receptor activation (Berridge, 1987). Activation of PKC is often implicated in physiological processes such as cellular proliferation and differentiation (see e.g. Watters & Parsons, 1999). The PI breakdown also has consequences for the of calcium ions (Ca2+) in cells. A physiological role for Ca2+ has been known for a long time (Ringer, 1883), but many aspects of Ca2+ signalling by receptors, and cellular consequences of Ca2+ transients, are still not clarified. cAMP, DAG, and Ca2+ are usually classified as second messenger molecules. The regulation of the cellular concentration of second messengers by GPCRs is fundamental to a precise regulation of the cellular machinery. One way to achieve an understanding of this precision is to elucidate the function of individual receptors in relation to the different signalling components with which they interact. The goal of the present study was to accomplish a step in this direction.

1.2 G-protein coupled receptors and signal transduction Over one percent of the human genome encodes for more than 1000 putative GPCRs (Flower, 1999). These have probably evolved as a consequence of the variety of signals a highly developed organism needs to respond to. GPCRs are intimately involved in most physiological processes that are under hormonal regulation. In addition, synaptic transmission, vision, , as well as perception of and smell, are also governed by GPCRs. It is therefore not surprising that the same signalling theme has a long evolutionary history; the yeast response pathway regulating mating behaviour also being relayed by GPCRs and G-proteins (Blumer & Thorner, 1991). GPCRs are, as the name implies, coupled to guanosine-5’-triphosphate (GTP)- binding proteins (G-proteins) of heterotrimeric α, β, γ composition. The G-proteins are

5 known as ’transducers’, transducing the signals to effector proteins. The signal transition from receptor to effector is thus dependent on protein−protein interactions and is delimited to the plasma membrane. The signal is further conveyed from the effector through diffusible second messengers, enabling a spreading of the intracellular signal. As indicated earlier, GPCR signalling is often a target for therapeutic intervention. Perturbation of normal signalling can sometimes have severe consequences. This is exemplified by the numerous activating mutations of GPCR signalling components found in different kinds of tumours (Marinissen & Gutkind, 2001). Other examples are the bacterial exotoxins of (pertussis toxin, Ptx) and Vibrio cholerae (, Ctx), both of which perturb G-protein function (Milligan, 1988).

1.2.1 Receptors The term ’receptive substance’, or receptor as we now call it, was coined by J. N. Langley in the early 20th century when studying the interaction of nicotine and curare on neuromuscular transmission (Langley, 1905). A long time passed before the ’receptive substances’ took the form of defined structural proteins. The first GPCR to be purified, sequenced (Ovchinnikov, 1982), and eventually cloned (Nathans & Hogness, 1983) was bovine . Based on the predicted structural topography, with seven hydrophobic amino acid stretches presumed to adopt membrane-spanning α helices, the rhodopsin protein showed similarities to bacteriorhodopsin, a bacterial proton pump for which the structure had been determined (Henderson & Unwin, 1975). However, it was not clear from the rhodopsin sequence that other receptors would have a similar structural arrangement. The purification of the β-ARs was accomplished in the early 1980s (Shorr et al., 1981, Shorr et al., 1982) and reconstitution experiments with receptor, G-protein, and AC confirmed the hypothesis of a three-component signalling system (May et al., 1985, Feder et al., 1986). In 1986,

two independent groups reported the cloning of β-ARs, the turkey β1-AR (Yarden et

al., 1986) and the hamster β2-AR (Dixon et al., 1986). The β-ARs show an overall

6 structural similarity to rhodopsin, with seven putative transmembrane helices (TMs) connected by hydrophilic intra- and extracellular loops, although the primary amino acid sequence homology to rhodopsin is weak. After the successful cloning of the β-ARs, it was soon noted that GPCRs constituted a large family of similarly structured proteins. The multitude of cloned GPCRs reported during the following years set the starting point for heterologous expression and thorough investigation of receptor function. All GPCRs are believed to function through a common molecular mechanism. The binding of an agonist causes conformational changes in the receptor protein. These changes then lead to activation of G-proteins by promoting GTP for GDP exchange of the α subunit. The agonist action on the receptor protein is a poorly understood process, although direct evidence has been obtained for conformational changes (Gether et al., 1995). How the activated receptor triggers G-protein activation is also rather poorly understood (Wess, 1998). On the other hand, molecular cloning of GPCRs provided the means for advanced structure−function studies and the mapping of receptor interaction sites. The first study on chimeric α2/β2-receptors, i.e. receptor hybrids with domains from both parental receptors, was reported in 1988 (Kobilka et al., 1988). It was concluded from the experiments that a particular section of the β-AR, including transmembrane helices (TM) five and six and the third intracellular loop (i3-loop) connecting the helices, is involved in the G-protein interaction. The chimeric receptor approach was soon realized to be a very efficient mean to map receptor−G-protein interactions, and a wealth of information has been gathered on this subject (see e.g. Wess, 1997).

1.2.2 Transducers G-proteins belong to a superfamily of . A protein that transduces the signal from receptor to effector was first anticipated by Dr. Rodbell and coworkers in 1971 in light of the guanine requirement for glucagon action on AC (Rodbell et al.,

1971). The AC stimulatory G-protein, Gs, was later purified by Dr. Gilman’s group and found to be heterotrimeric, composed of an α, a β, and a γ subunit (Northup et al.,

7 1980). Subsequently, the AC inhibitory G-protein, Gi, was identified (Katada & Ui, 1982, Codina et al., 1983, Bokoch et al., 1984). A few years earlier, it had been shown that catecholamine action on erythrocyte membranes stimulates GTPase activity (Cassel & Selinger, 1976). Analysis of the purified components revealed that the α subunits possess the GTPase activity. In the ground state a GDP molecule is bound and the α subunit is associated with the βγ complex. Receptor activation of the heterotrimer lowers the affinity for GDP, allowing GTP to bind. Based on in vitro studies, the effect can be explained by an increase in the affinity of G-proteins for Mg2+, which, in turn, promotes the nucleotide exchange (Birnbaumer & Birnbaumer, 1995). The α-GTP and βγ complex then dissociates from each other to associate with, and affect, effector proteins. The dissociation theory is widely used to explain G- protein action, although not universally accepted (see e.g. Rebois et al., 1997, Chidiac, 1998). The intrinsic GTPase activity of the α subunit then returns the G-protein to the ground state, accompanied by subunit association.

As for all other components in signal transduction, molecular cloning has revealed a diversity of G-proteins (reviewed in Gilman, 1987, Hepler & Gilman, 1992, Hildebrandt, 1997). Four major subfamilies can be outlined based on α subunit structure: Gs, Gi, Gq, and G12.

The Gs subfamily contains the short and long isoforms of αs, two splice variants

of a single gene, and. a closely related αs isoform, αolf. Golf is primarily found in olfactory neurons and mediates the effects of odorants to a specific isoform of AC

(Menco et al., 1992). Gs-proteins are ADP-ribosylated by the A (active) protomer of

Ctx, rendering the αs subunit constitutively active by stabilizing the GTP-bound form of the α subunit (Cassel & Selinger, 1977, Kahn & Gilman, 1984).

The Gi subfamily is the largest, containing three αi subunits, two αo subunits,

αgust, αz, and the αtr and αtc. All Gi subfamily proteins, except αz, are

substrates for Ptx-catalyzed ADP ribosylation, which uncouples the Gi subfamily

proteins from receptors (Ui, 1984, Milligan, 1988). It was by the use of Ptx that Gi was

8 identified as the inhibitory G-protein of AC (Katada & Ui, 1982). Gi subfamily proteins are also involved in intracellular Ca2+ homeostasis through inhibitory actions on voltage-gated Ca2+-channels and stimulation of PLCβ isoforms. These functions are presumably mediated by the Gβγ subunits and can thus also be ascribed to other

G-proteins. A clear-cut effector for Go-proteins has not been identified, despite its great abundance in the brain (Neer et al., 1984, Sternweis & Robishaw, 1984). Little is known about Gz, but it may be similar in action to Gi (Kozasa & Gilman, 1995).

The Gq subfamily of proteins include αq, α11, α14, α15, and α16, all of which

activate PLC-β (reviewed in Rhee, 2001). Expression of G16, and the mouse orthologue G15, is restricted to cells of hematopoietic origin, whereas the expression patterns of Gq and G11 are wide.

The function of G12 and G13 is largely unknown, but they appear to possess high transforming capacity when constitutively activated (for review, see Gudermann et al., 2000).

The Gβγ subunit family is made up of five β subunit genes and eleven γ subunit genes (Hildebrandt, 1997). The different possible combinations of these, and their potential selectivity for specific Gα subunits, may contribute to specificity in receptor signalling. However, there does not seem to be much specificty in the association of either Gβγ dimers or Gαβγ trimers, except in a few cases (Fawzi et al., 1991, Schmidt et al., 1992, Yan et al., 1996). Initially, Gβγ subunits were considered inhibitory modules of Gα subunits by stabilizing the inactive GDP-bound form of Gα (Cerione et al., 1986b). Receptor activation of G-proteins reguires the heterotrimeric composi- tion (Fung, 1983), indicating a role in receptor recognition as well. In addition, Gβγ subunits are hydrophobic, partially due to a γ-associated prenyl group, having an important role in anchoring the Gα subunits to the membrane (Sternweis, 1986). The first demonstrated direct effector action of Gβγ was the activation of K+-channels in atrial myocytes (Logothetis et al., 1987). Subsequently, Gβγ subunits were shown to

2+ affect several AC isoforms, PLCs of the β family, PLA2, and voltage-gated Ca -

9 channels (reviewed in Sternweis, 1994). An interesting aspect of Gβγ signalling is that the β subunits contain WD40 repeats, about 40 amino acid long stretches with Trp(W)-Asp(D) dipeptide sequences (Wang et al., 1994). These structures are believed to specify protein−protein interactions by binding to pleckstrin homology (PH) domains found in a multitude of signalling molecules, including PLCs and β-AR kinases (Shaw, 1995).

1.2.3 Effectors Classically, GPCRs are linked to effectors by G-proteins. This is not true in all cases (Hall et al., 1999). Nevertheless, a multitude of G-protein−effector interactions have been demonstrated. The widely accepted effectors, directly affected by G-proteins, include adenylyl cyclase (AC), cGMP phosphodiesterase (cGMP PDE), (PLC), and Ca2+- and K+-channels. Here the focus will be on AC and PLC.

1.2.3.1 Adenylyl cyclase, cAMP, and PKA AC is a ubiquitous enzyme synthesizing cAMP from Mg2+-ATP. Before the molecular cloning era, the mammalian membrane-bound ACs were divided into a common Ca2+- insensitive isoform and a neuronal Ca2+/CaM-activated isoform. Pfeuffer and colleagues managed to purify these rare membrane protein species by the use of forskolin affinity matrix (Pfeuffer et al., 1985a, Pfeuffer et al., 1985b). In 1989, the cDNA for the type 1 neuronal isoform (AC1) was reported (Krupinski et al., 1989), and subsequently a whole family a related isoforms were identified (reviewed in Sunahara et al., 1996). The topographical structure of the ACs shows similarity to membrane transporters such as P glycoprotein and the cyctic fibrosis transmembrane conductance regulator (CFTR). The protein traverses the membrane 12 times in two cassetts, adopting a pseudo-dimeric structure. The cytoplasmic regions between the cassettes and distal to the second cassett make up the catalytic center, and are also the sites of regulation by G-proteins and protein kinases (Tang & Hurley, 1998). ACs are subject to many regulatory inputs, indicating that the principal control of cellular

10 cAMP concentration lies at the level of its synthesis, and that fine-tuning of the enzymes is central to the control. Molecular cloning of the bovine AC1 and the successful expression in Sf9 insect cells (Tang et al., 1991) were important pioneering work for establishing regulatory properties of ACs. Heterologous expression of different isoforms of ACs has since then greatly improved our understanding of AC regulation. The mammalian ACs can be largely grouped into three subfamilies based on regulatory properties (Table 1): the Ca2+/CaM-stimulated isoforms, the Gβγ-stimulated isoforms, and the Ca2+-inhibited isoforms. Several AC isoforms, homologous to mammalian ACs, are also found in invertebrates (Levin et al., 1992, Iourgenko et al., 1997, Korswagen et al., 1998). As can be seen from table 1, there is a large potential for cross-talk between traditional Ca2+ mobilizing receptors and cAMP signalling already at the stage of cAMP synthesis. Overall, the number of positive and negative regulators of ACs are well in balance to tightly regulate the cAMP synthesis through a variety of receptors. An enigma, in this regard, is the Gβγ-stimulated subgroup, and in particular the AC2 isoform that has been demonstrated to have high basal activity (Pieroni et al., 1995), but no inhibitory input from receptors.

11 Table 1. Regulators of mammalian AC isoforms. Physiological regulators Subfamily Isoform Stimulators Inhibitors 2+ a 2+ Ca /CaM- AC1 Gs , Ca /CaM Gi, Go, Gβγ, CaMKIV b 2+ stimulated AC3 Gs, Ca /CaM Gi, CaMKII 2+ AC8 Gs, Ca /CaM Gi

Gβγ-stimulated AC2 Gs, Gβγ, PKC none? c AC4 Gs, Gβγ PKC

AC7 Gs, Gβγ, PKC ? 2+ d 2+ e Ca -inhibited AC5 Gs, PKC Ca , Gi, PKA, Gβγ 2+ e AC6 Gs Ca , Gi, PKA, Gβγ f 2+ AC9 Gs Ca /, Gi? a AC1 may not be stimulated by Gs activation in vivo due to concomitant inhibition by Gβγ

(Wayman et al., 1994), but it is stimulated in vitro by Gαs (Tang et al., 1991). b Stimulation of this isoform by Ca2+/CaM is dubious, since stimulation in vitro reguires supraphysiological concentrations of Ca2+ (Choi et al., 1992). c activated PKC reduces Gs- but not forskolin-stimulated activity (Zimmermann & Taussig, 1996). d Inhibition of this isoform by Ca2+ is questionable. e Gβγ effect determined by cotransfection of ACs and Gβγ in COS-7 cells (Bayewitch et al., 1998). f AC9 is usually not included in this subfamily, but it is inhibited by Ca2+ via calcineurin (Antoni et al., 1998).

12 cAMP is chemically stable, but it is degraded in cells by PDEs to the inactive metabolite 5’-AMP. There are at least 15 gene products for PDEs, grouped into seven subfamilies (Spina et al., 1998). Some of these are specific for cAMP, some for cGMP, and some are unselective. Since the activity of most of the PDEs is regulated mainly by substrate availability, the inhibition by different natural or synthetic agents is in the focus of PDE research. Theophylline (1,3-dimethylxanthine) is the archetypal PDE inhibitor. IBMX (3-isobutyl-1-methylxanthine) is closely related to theophylline, show little subtype selectivity (Ukena et al., 1993), and often used in cell culture experiments to address AC or cAMP functions.

PKA is a multifunctional cAMP-dependent serine-threonine kinase that mediates most of the effects of cAMP (Walsh & van Patten, 1994). The PKA family of enzymes are assembled from the products of four Regulatory and, at least, two Catalytic subunit genes. The R subunits homodimerize, and each R binds one C subunit forming a tetrameric holoenzyme. Activation is triggered by cAMP binding to the R subunits and subsequent dissociation of the C subunits. Besides the tight control of the cellular cAMP concentration, which is crucial for PKA activation, the increased awareness of compartmentalization in signalling has also led to identification of multiple A Kinase Anchoring Proteins (AKAPs), providing a degree of subcellular specificity of PKA action (Dell’Acqua & Scott, 1997).

1.2.3.2 Phospholipase C, Ca2+, and PKC Phospholipase Cs are a group of enzymes catalyzing the hydrolysis of phosphatidyl- inositol 4,5-bisphosphate (PIP2) to inositol 1,4,5-trisphosphate (IP3) and diacylglycerol 2+ (DAG). IP3 mobilizes Ca from intracellular depots and DAG is an activator of PKC. In the 1970s, Ca2+ was being recognized as a messenger molecule of hormone action (reviewed in Rasmussen, 1970, Exton, 1981). At the same time, the involve- ment of phosphatidylinositol (PI) in signalling was subjected to investigation (Michell, 1975). Sir Michael Berridge pulled these findings together and showed, with the help 2+ of colleagues, that IP3 was the missing link between receptor activation and Ca

13 mobilization (Streb et al., 1983, reviewed in Berridge & Irvine, 1984). This discovery set the starting point for a whole new research branch trying to characterize PLCs, IP3 action, and Ca2+ signalling. Today at least 11 different isoforms of PLCs, grouped into four subfamilies, β, γ, δ, and ε, have been identified (Rhee, 2001). The PLC-β

isoforms are subject to GPCR signalling through Ptx-insensitive Gq subfamily of proteins as well as through Ptx-sensitive G-protein βγ subunits. Four different isoforms of PLC-βs, β1-4, are all activated by Gαq/11 proteins. PLC-β2 appears to be

restricted to hematopoietic cells, and is likely to be the target of Gα16, which also activates PLC-β1 and -β3 isoforms (Kozasa et al., 1993). Gβγ has been shown to interact with PLC-β1, -β2, and -β3, with the highest affinity for PLC-β2 (Runnels & Scarlata, 1999), by binding to the PH domain of the PLC-βs (Wang et al., 1999).

2+ The IP3 product of PLC action mobilizes Ca from intracellular stores, mainly the endoplasmatic reticulum. This, in turn, evokes a capacitative inflow of Ca2+ from extracellular space. Ca2+ acts as a universal messenger involved in crucial events of life cycle, such as fertilization, muscle contraction, , proliferation, metabo- lism, and gene expression (Berridge, 1997). It is therefore not surprising that a multitude of molecules are involved in regulating intracellular Ca2+ homeostasis and Ca2+ actions. The reviewing of these is beyond the scope of this thesis.

Dr. Nishizuka and colleagues discovered PKC in 1977 (Inoue et al., 1977). This enzyme was initially identified as a Ca2+-activated, phospholipid-dependent serine- threonine kinase, with no obvious relation to signal transduction. Around 1980, it was shown that DAG greatly increases the affinity of PKC for Ca2+ and thereby, in a sense, activates it (Takai et al., 1979, reviewed in Nishizuka, 1984). As DAG is one of

the products of PIP2 hydrolysis, it is obvious that PKC is a target for PLC-activating receptors. PKC has long been implicated in cellular transformation due to the tumour promoting activity of phorbol esters, which bind to and activate PKC (Castagna et al., 1982).

14 As can be anticipated, the PKC enzymes constitute a family of related proteins that differ in their tissue distribution and regulatory properties (reviewed in Hug & Sarre, 1993). They can be grouped into three subfamilies based on activation requirements: the classical PKC isoforms are activated by DAG and phorbol esters in a Ca2+- dependent way, the novel PKC isoforms are activated by DAG and phorbol esters in a Ca2+-independent way, and the atypical PKC isoforms are not activated by conventional GPCR signalling.

1.3 α2-adrenergic receptors

1.3.1 A subfamily of adrenergic receptors Noradrenaline and adrenaline exert their effects through a large family of GPCRs encoded by nine different genes in humans, three of each of β-ARs, α1-ARs and α2- ARs (Pepperl & Regan, 1994). The α-ARs were first divided from β-AR by Dr. Ahlqvist in the 1940s based on agonist potency differences of evoked physiological responses (Ahlquist, 1948). Later, the α-ARs were further subdivided into α1- and α2- ARs based on micro-anatomical location as pre- or postsynaptic receptors, respectively (Langer, 1974). Isotope labelling of ligands finally enabled a pharmacological 3 distinction between these subtypes using α1-selective [ H]prazosin and α2-selective [3H]yohimbine (Starke, 1981). The original classification by Dr. Langer did not hold for very long as α2-ARs were found both on postsynaptic sites and in nonsynaptic locations, such as platelets (Berthelsen & Pettinger, 1977).

The human platelet α2A-AR was cloned in 1987 (Kobilka et al., 1987). The predicted protein is highly homologous to the β-ARs in TM regions. The intracellular

domains differ from the β-ARs, the α2A-AR having a relatively long i3-loop and a short carboxyl terminus whereas the opposite is true for β-ARs. The expressed receptor shows similar binding properties as the pharmacologically defined

α2A-AR (Bylund, 1985), with high affinity for yohimbine and low affinity for

15 prazosin. Soon after cloning of the first α2-AR, the α2C-AR (Regan et al., 1988) and

the α2B-AR (Lomasney et al., 1990) subtypes were cloned. All three subtype homologues in rat and mouse have also been cloned and sequenced (see e.g. Pepperl

& Regan, 1994). The rodent α2A- and α2B-ARs are of some interest to the present

study. The mouse α2B-AR subtype (Chruscinski et al., 1992) is virtually identical to the human orthologue in the TM regions, but only 64% homologous in the i3-loop, suggesting that this region need not be highly conserved for preserved functions. The i2-loops are identical between mouse and human α2B-AR.

The rodent α2A-AR has different antagonist binding properties than the human

α2A-AR. This finding initially classified the rodent receptor as a new pharmacological subtype, α2D-AR (Lanier et al., 1991). The α2D-AR is now accepted as the orthologue

of human α2A-AR and will be referred to as α2A-AR throughout this study. The basis for the different affinities of human and mouse α2A-ARs for yohimbine has been shown to be due, at least partially, to a single amino acid difference in TM5 (Link et al., 1992).

In the original paper by Dr. Kobilka and collegues on chimeric α2/β2-receptors, the TM7 of both parent receptors was strongly implicated in agonist and antagonist binding specificity (Kobilka et al., 1988). However, TM7 has an important role in intramolecular interactions, stabilizing the three-dimensional conformation of the receptor (Suryanarayana et al., 1992), and may not be directly involved in ligand interactions of the α2-ARs. It has been shown that a conserved aspartic acid residue in 113 TM3 (Asp in α2A-AR) provides the counterion to the positively charged amino group of catecholamine ligands (Wang et al., 1991, for review, see also Savarese & 204 Fraser, 1992). A serine residue in TM5 (Ser in human α2A-AR) is believed to interact with the para-hydroxyl group of the catechol moiety of the natural ligands 200 (Wang et al., 1991). Another serine residue (Ser in human α2A-AR) has also been implicated in ligand binding affinity and signalling potency (Wang et al., 1991, Rudling et al., 1999).

16 1.3.2 Physiological functions

Central effects of α2-AR agonists include hypotension, bradycardia, sedation, sleep induction, analgesia, mydriasis, and modulation of growth hormone release from the pituitary (reviewed in Ruffolo et al., 1993, Aantaa et al., 1995). Clonidine has long been used as an antihypertensive drug in clinical practice. Clonidine stimulation of

central α2-ARs reduces the sympathetic outflow to the periphery, leading to a reduction in arterial blood pressure and bradycardia. The exact site of action is a matter of debate, but it appears to include several centers in the medulla including nucleus tractus solitarius, a center that receives baroreceptor input (Ruffolo et al., 1993, Aantaa et al., 1995, Nicholas et al., 1996, Singewald & Philippu, 1996).

Sedation is a common side effect of clonidine, and the α2-AR agonists xylasine and medetomidine are frequently used in veterinary medicine for sedation and also for analgesia. Locus coeruleus, the predominant noradrenergic nucleus in the mammalian

brain, is rich in α2-ARs and is probably the site of action for the sedative/hypnotic effect of α2-AR agonists (Correa-Sales et al., 1992). In the periphery, noradrenaline released from sympathetic nerve endings and adrenaline released from the exert the effects of sympathetic stimulation by activation of adrenergic receptors located presynaptically on nerve endings, or post- and/or extrasynaptically on target tissues. As Langer pointed out in his subdivision of adrenergic receptors, α2-ARs are predominantly located on presynaptic nerve terminals, and when activated inhibit release

(Langer, 1974). Presynaptic α2-ARs are found on all sympathetically innervated

tissues examined (Ruffolo et al., 1993). Post- and/or extrasynaptic α2-ARs are found in vascular beds of both arterial and venous origin. Activation of these receptors leads to contraction and vasoconstriction (reviewed in Langer & Hicks, 1984). It may also lead to vasodilation through increased production of nitric oxide in

the endothelia (Bockman et al., 1993). The arterial α2-ARs are believed to be mainly extrasynaptic, responding to circulating adrenaline, whereas the postsynaptic receptors

17 would be of α1-AR origin (Ruffolo et al., 1993). Another smooth muscle tissue with

α2-ARs is the uterus (Hoffman et al., 1981). An intersting α2−β2-AR cross-talk in relation to pregnancy appears to occur in this tissue (Mhaouty et al., 1995). At mid- pregnancy the myometrial α2A-AR predominates and has the ability to potentiate β- AR-stimulated cAMP synthesis. This could be a mechanism of keeping the muscles in

a relaxed state. At term, the α2B-AR is upregulated and attenuates the β-AR response to provide for a forced contraction at delivery.

In several target tissues, α2-AR activation is associated with inhibitory functions (Ruffolo et al., 1993). In the gastrointestinal tract, secretion and motility is decreased, and in pancreas, release is inhibited by beta cell α2-ARs. In adipose tissue, α2-ARs inhibit lipolysis, and in the , α2-ARs counteract action. A long known target for circulating catecholamines with undisputable extrajunctional α2-ARs are the platelets. Adrenaline induces aggregation of platelets and potentiates aggregation induced by other agents such as ADP and thrombin

(Thomas, 1967). However, the physiological benefit of α2-AR-mediated platelet aggregation is not clear, neither is the intracellular signalling cascade regulating it (Ruffolo et al., 1993, Keularts et al., 2000).

Most classical functions of α2-ARs are elucidated using clonidine, p-

aminoclonidine, and UK14,304 as selective α2-AR agonists (Berlan et al., 1992, Hieble et al., 1995). These ligands are unselective for the different subtypes, and most functions have been related to the α2A-AR subtype based on prazosin insensitivity (Hieble et al., 1995). Lately, the techniques of gene knock-out or knock-in in mice have been introduced in an effort to elucidate the in vivo function of different α2-AR subtypes (reviewed in Kable et al., 2000). Using knock-out mice, it has been shown

that the long-lasting antihypertensive effect of agonists is mediated by the α2A subtype. Upon administration of the drugs to animals, there is an initial hypertensive pressor

response. This pressor response is absent in α2B-AR knock-out mice, suggesting that

18 the peripheral pressor response is mediated by α2B-AR action on the vasculature. Other subtype-related effects demonstrated in knock-out mice are the salt-induced

hypertension, which is abolished in α2B-AR knock-outs (heterozygotes), and the presynaptic inhibition of noradrenaline release, which appears to lack an α2B-AR component, at least in sympathetic nerves innervating the heart.

1.3.3 Cellular signalling

A diminution of cAMP synthesis through Gi-proteins is the most frequently observed cellular signalling function of α2-AR activation (Fain & Garcia-Sainz, 1980). The white adipocytes provides a good example of functional consequences of traditional inhibition of AC activity. A hormone-sensitive lipase (HSL) in these cells is responsible for triglyceride breakdown and lipolysis. This HSL is stimulated by PKA- mediated , following that α2-AR activation decreases the activity of the AC−PKA−HSL pathway and counteracts β-AR stimulation of the same (Lafontan & Berlan, 1993).

In general, though, a noticeable direct physiological consequence of α2-ARs mediated via inhibition of cAMP synthesis seems unusual. Events like inhibition of insulin release (Ullrich & Wollheim, 1984), inhibition of neurotransmitter release (Lipscombe et al., 1989), platelet aggregation (Haslam et al., 1978), and mitogenic signalling through MAPK (Kribben et al., 1997) all appear independent of the cAMP pathway. Cellular activation of PLC (Dorn et al., 1997), PLA2 (Sweatt et al., 1986), and PLD (Jinsi et al., 1996) by α2-ARs is even more distant to a cAMP decrease. Nevertheless, cAMP is easily measured and AC regulation serves as the most reliable functional indicator for α2-AR activation.

The inhibition of cAMP synthesis by α2-ARs has been studied in numerous clonal cell lines, expressing endogenous or ectopic α2-ARs, as well as in primary cell

cultures. In some cell types, the cAMP synthesis is increased upon α2-AR activation or exhibit U-shaped inhibitory and stimulatory regulation depending on agonist conc-

19 entration. Table 2 lists a number of such "anomalous" behaviours of defined receptor subtypes expressed in different cell types.

Table 2. Stimulatory cAMP responses via α2-ARs. Receptor Cell type Effect on cAMPa References

α2A CHO U (Fraser et al., 1989, Eason et al., 1992) ↑(Ptx) (Jones et al., 1991, Wang et al., 1991, Eason et al., 1992, Airriess et al., 1997, Rudling et al., 1999) CHW ↑(Ptx) (Cotecchia et al., 1990) COS-7 ↑ (Federman et al., 1992, Eason & Liggett, 1995) PC-12 U (Duzic & Lanier, 1992) JEG-3 Ub (Pepperl & Regan, 1993) HEK 293 ↑c (Chabre et al., 1994) Rat-1 U,↑(Ptx) (Sautel & Milligan, 1998)

α2B CHO ↑(Ptx) (Eason et al., 1992) ↑ (Pohjanoksa et al., 1997, Rudling et al., 2000) PC-12 ↑ (Duzic & Lanier, 1992) JEG-3 ↑b (Pepperl & Regan, 1993) S115 ↑(Ptx) (Jansson et al., 1994) Sf9 ↑ (Jansson et al., 1995)

α2C CHO ↑(Ptx) (Eason et al., 1992, Rudling et al., 2000) b JEG-3 ↑(Ptx) (Pepperl & Regan, 1993) a ↑ means stimulation, ↑(Ptx) means stimulation revealed after Ptx treatment, and U means U-shaped responses. b indirect assay via CREB-mediated . c cells overexpressing Gs-proteins.

20 In many of the studies referred to in table 2, a comparison of signalling between receptor subtypes has been made in the same cell type. With this background information, it is evident that the α2C-AR is very resticted in stimulatory cAMP effects. Comparison between α2A-AR and α2B-AR suggests that the α2B-AR is more

disposed for stimulation, although comparable results for α2A-AR are obtained in some cell types. Whereas a stimulation of cAMP synthesis is usually not seen with endogenous 2+ α2-ARs, an increase in [Ca ]i in response to agonists is observed in tissue preparations of smooth muscle (Aburto et al., 1993), in primary cultures of astrocytes (Salm & McCarthy, 1990), and in clonal cell lines expressing endogenous receptors, e.g. HEL human erythroleukemia cells (Michel et al., 1989) and NG108-15 rat neuroblastoma×glioma cells (Holmberg et al., 1998). Ca2+ signalling is also evident in several transfected cell lines (Åkerman et al., 1996, Dorn et al., 1997, Kukkonen et al., 1998).

Modulation of activity is crucial for α2-AR-mediated inhibition of neurotransmitter release. Both K+- and Ca2+-channels have been implicated in this function (Isom & Limbird, 1988). K+-channel activation and Ca2+-channel inhibition are presumed to be dependent on protein−protein interaction and, thus, independent of cAMP (Lipscombe et al., 1989, Surprenant et al., 1992).

1.4 Other receptors utilized in the study

1.4.1 Octopamine receptors Octopus salivary glands contain enormous amounts of octopamine (Erspamer & Boretti, 1951, Saavedra, 1974). Octopamine is also found in neuronal as well as non- neuronal tissues of most other invertebrates (reviewed in Roeder, 1999). The invertebrate octopaminergic system is thought to be homologous to the vertebrate adrenergic system. This is not only due to the structural analogy of octopamine and

21 noradrenaline, but also for the restricted localization of octopaminergic neurons, similar to adrenergic neurons, in CNS, release of octopamine in stress situations, and the functional consequences of the release (Roeder, 1999). The first receptor to be characterized for octopamine was found in the CNS of the cockroach Periplaneta americana (Nathanson & Greengard, 1973). A classification of octopamine receptors was introduced by Dr. Evans in the early 1980s. Using the locust extensor tibiae muscle as a model, three different subtypes could be recognized on functional and pharmacological criteria, class 1, 2A and 2B (Evans, 1981). Later a pharmacologically distinct receptor subtype, class 3, was characterized in neuronal tissue of the locust (Roeder, 1992). Many of the characterized octopamine receptors seem to be coupled to AC activation (class 2 and 3 receptors), but receptors coupled to Ca2+ mobilization (class 1 receptors) are also found (Roeder, 1999). The Sf9 cell line is one of the few clonal cell lines expressing endogenous octopamine receptors, making it useful for studies of octopamine receptor signalling (Orr et al., 1992).

1.4.2 Purinergic receptors Adenine nucleotides are released from cells either from vesicles or in response to stress or hypoxia. Cellular effects of extracellular adenine nucleotides are mediated by purinergic receptors, which are expressed, in some form, by most cell types (Ralevic & Burnstock, 1998). A physiological effect of administered adenosine was shown more than 70 years ago (Drury & Szent-Györgyi, 1929). Much later, adenosine was demonstrated to either stimulate (Sattin & Rall, 1970) or inhibit (van Calker et al., 1978) cAMP

production in brain tissue, thereby setting a need for classification into adenosine A1 and A2 receptors (van Calker et al., 1979). In the 1970s, evidence were accumulating

for specific ATP receptors as well, and the purinergic receptors were classified into P1

(adenosine) and P2 (ATP) receptors (Burnstock, 1980). Now, it is generally accepted

that the P1 receptors encompass four subtypes of adenosine receptors linked to

stimulation (A2A, A2B) or inhibition (A1, A3) of AC (Fredholm et al., 1994). The P2

22 receptor family contains numerous ionotropic (P2X) and metabotropic (P2Y) receptors

(Fredholm et al., 1994). The P2Y receptors are mostly linked to PLC activation through

Gq subfamily proteins (O’Connor et al., 1991).

1.4.3 Muscarinic receptors The sympathetic action of noradrenaline is in most tissues counteracted by the parasympathetic action of on muscarinic receptors. The name for this subfamily of acetylcholine receptors comes from the plant alkaloid muscarine, a potent agonist at these receptors noticed by Sir Henry Dale some 90 years ago. Muscarinic receptors, in contrast to adrenergic receptors, are also found in invertebrates (Shapiro et al., 1989). Molecular cloning of the muscarinic receptors, in humans as well as in

rodents, has revealed a family of five distinct subtypes, M1-M5 (Ashkenazi & Peralta, 1994). Expression studies of these receptors have coined a consensus view of functional coupling to the Gq subfamily (M1, M3, M5) or the Gi subfamily (M2, M4) of proteins.

1.5 Why study heterologously expressed receptors? Examination of distinct receptor subtypes in vivo is extremely difficult for several reasons. First, in the case of adrenergic receptors, the sympathetic innervation of tissues makes it difficult to distinguish responses of pre- and postsynaptic receptors (Berlan et al., 1992). Second, the available quantities of tissues are restricted, and the ethical aspects are of high weight in experimental research involving animals. Third, single cells may contain multiple receptor subtypes with similar ligand binding properties, e.g. adipocytes may contain up to five different adrenergic receptors (Lafontan & Berlan, 1993). Fourth, ligands with sufficient subtype selectivity are not available for distinction between α2-AR subtypes. Fifth, the amount of receptors in native tissues is usually very low, reflected by the fact that an overall 80,000 -

23 100,000-fold purification is necessary to obtain a homogenous α2-AR population from human platelets (Regan et al., 1982). Another way to study receptors is by the use of immortalized cell lines. However, the establishment of cell lines with endogenous distinct receptor subtypes is not an easy task. For α2-ARs, this has been a secondary profit of already established cell lines in some fortunate cases. These include human HT-29 cells, expressing α2A-

AR (Bylund et al., 1988), rat NG108-15 cells, expressing α2B-AR (Bylund et al.,

1988), opossum OK cells and human HepG2 cells, expressing α2C-AR (Murphy &

Bylund, 1988, Schaak et al., 1997). A human cell line expressing only the α2B-AR subtype has not been found. Heterologous expression of receptors is a way of getting unlimited amounts of single receptor subtypes in a defined setting. Most receptors appear to behave naturally also in an unnatural environment, i.e. human receptors can be expressed in non-human cells and vice versa, and retain their capacity to bind ligands and relay signals. Heterologous expression systems also have their limitations. One such important limitation is that the signalling machinery is defined by the cells and made up of fixed tranducers and effectors. Endogenous effector proteins, such as ACs, are often heterogenous in composition (Marjamaki et al., 1997, Varga et al., 1998), and lack of selective blockers for these makes it impossible to hint particular isoforms as targets of receptor action. A way to overcome this obstacle is by genetic engineering of cells to either impair endogenous expression of isoforms or overexpress exogenous isoforms. Heterologous expression of receptors and signalling molecules has led to the discovery of a vast diversity of receptor signalling pathways. Some of these may be physiologically redundant, or even non-existing, in vivo. Nevertheless, given the difficulties of examining receptors in native tissues, our current understanding of receptor structure and function would not have been possible without heterologous expression.

24 2 AIMS OF THE STUDY

The aims of this study were:

- to characterize receptor signalling cascades in Sf9 insect cells and to optimize the Baculovirus Expression Vector System (BEVS) for functional studies of GPCRs.

- to determine what factor(s) is responsible for the opposite coupling of α2A- and α2B- ARs to cAMP production in Sf9 cells.

- to characterize α2-AR signalling to specific AC isoforms.

2+ - to characterize the Ca signalling mechanism of α2-ARs.

- to highlight the signalling differences between α2-AR subtypes.

25 3 MATERIALS AND METHODS

This section is a brief summary of the material and methods used in the different studies and mostly a complement to these. For more details the reader is referred to individual papers.

3.1 Cell cultures The Sf9 cell line is a clonal cell line isolated from Sf21 cell culture, which originates from Spodoptera frugiperda pupal ovarian tissue (Vaughn et al., 1977). The Sf9 cell line was maintained as suspension culture in glass spinner bottles with constant stirring at 25-27°C. The cells also grow as adherent cultures and were usually seeded on tissue culture dishes for the purpose of infection with recombinant baculovirus (I, II, III, V). The buffer used during experimental conditions was Na+-based. As K+ is the most abundant cation in the culture medium, and in the hemolymph of lepidopteran insects, a K+-based medium (Vachon et al., 1995) was tested in functional assays with + α2-ARs. No significant differences in the outcome compared to the Na -based buffer were detected (data not shown).

Stable α2-AR transfectants of Chinese hamster ovary (CHO) cells (Pohjanoksa et al., 1997) were obtained through Dr. Mika Scheinin (University of Turku, Turku, Finland), and grown in MEM alpha supplemented with antibiotics and 5% foetal calf serum at 37°C (IV). The clones expressed comparable levels of the different human

α2-AR subtypes (Pohjanoksa et al., 1997, Kukkonen et al., 1998).

3.2 BEVS and infection procedures The Baculovirus Expression Vector System (BEVS) uses the Autographa californica Nuclear Polyhedrosis Virus (AcNPV, family Baculoviridae) to deliver genes into insect cells for efficient eukaryotic expression. There are several commercially available systems for generation of recombinant baculovirus. The basic for all of them is to replace a non-essential gene of the baculovirus genome with the gene of interest. All recombinant virus constructs used in this study are replacing the polyhedrin gene,

26 and use the polyhedrin gene promoter to drive the expression of the recombinant genes. Infection of cells can be done simply by adding a small amount of virus to cell cultures and allow spreading of infection by diffusion. This method is inefficient for coexpression studies involving multiple viruses. To obtain efficient coexpression, and a synchronized infection of all cells, the infection was done by sequential overlay of cells with high-titer virus stocks (III, V). The efficiency of coexpression was tested by sequential infection with wild-type virus, producing occlusion bodies visible under light microscope, and a recombinant virus expressing Green Fluorescent Protein (GFP), which can be detected with fluorescense microscopy. Examination of cells 48 h post infection indicated coexpression in virtually all cells (> 90%).

3.3 Receptor quantification 3 3 α2-AR expression was monitored by specific binding of [ H]RX821002 ([ H]2- methoxy-idazoxan) to cell homogenates (II) or membrane fractions of homogenates 3 (III). [ H]RX821002 is a selective α2-AR antagonist, that exhibits low non-specific binding, and binds with high affinity also to the rodent α2A orthologue, in contrast to [3H]rauwolscine and [3H]yohimbine (Erdbrugger et al., 1995, Deupree et al., 1996).

Saturation binding studies of the human α2-AR subtypes expressed in Sf9 cells give

affinity konstant (Kd) values slightly under 1 nM for the α2A-AR and around 3.5 nM

for the α2B-AR (Uhlen et al., 1998). These values are similar to the values obtained for the human subtypes in CHO cells (Pohjanoksa et al., 1997), corresponding rat subtypes in COS-1 cells, and corresponding porcine subtypes in tissue preparations (Uhlen et al., 1998).

3.4 Receptor−G-protein interactions

Pertussis toxin (Ptx) was used to ablate receptor−Gi/o-protein interactions (II, V). There has been a debate regarding Ptx sensitivity of Sf9 cell G-proteins. Some investigators have failed to abolish responses with Ptx (Vasudevan et al., 1992,

27 Mulheron et al., 1994), while others have not encountered problems (Richardson &

Hosey, 1992, Ng et al., 1993). In the paper first describing the expression of α2A-AR in Sf9 cells (Jansson et al., 1995), Ptx significantly inhibited the receptor response, but did not abolish it. We have also noticed that the inhibition of AC2 (V) is not completely abolished by Ptx treatment when using high concentrations of UK14,304

(> 3 µM) at the human α2A-AR. Such problems have not been met with the M4 receptor. The nonhydrolyzable GTP analogue GTPγS can be used to activate G-proteins independently of receptors in membranes or permeabilized cells. When applied at low concentrations, it can also be used to monitor receptor-catalyzed G-protein activation

(III). An increase in GTPγS binding to Gi/o-proteins is readily observed upon receptor

stimulation. Receptor-catalyzed binding to Gs-proteins is more difficult to obtain, and usually has a low signal-to-noise ratio (Wieland & Jakobs, 1994). A similar problem is encountered in GTPase assay, as a result of the extremely low rate of GTP hydrolysis by Gs-proteins (Milligan, 1988).

3.5 Measurement of cAMP

Gs-protein coupled receptors increase AC activity, which is easily measured as an increase in the cAMP content of cells. To measure Gi-protein coupled receptor action, a stimulant needs to be included. Forskolin, a diterpene isolated from the plant Coleus forskohlii, is known to activate AC independently of receptor activation (Seamon et al., 1981). Agonist-dependent inhibition of forskolin-stimulated cAMP production is therefore often used as a measure of Gi-protein activation by receptors. To measure cAMP accumulation (I, II, III, IV, V), cells were incubated with [3H]adenine to label the ATP pool, from which cAMP is formed. Cells were then stimulated, whereafter the cAMP was extracted, separated from other labelled contaminants by ion exchange chromatography and adsorption to insoluble aluminium oxide salt, and quantified by liquid scintillation counting. The method lacks spatial resolution of cAMP concentrations, but gives highly reproducible results of total cellular cAMP accumulation. The absolute concentration of cAMP cannot be obtained

28 by this method, since the radiolabelled cAMP (and ATP) makes up only part of the total cellular cAMP content. The cAMP level is instead related to the labelled ATP and ADP pools, eluted from the ion exchange step (Krishna et al., 1968), and gives a relative measure of the amount of conversion of ATP to cAMP.

3.6 Measurement of Ca2+ Fura-2, a fluorescent probe highly selective for Ca2+, is delivered into cells as an acetoxymethyl ester conjugate (Fura-2 AM) (Grynkiewicz et al., 1985). The fura-2 molecule is trapped inside the cells after the removal of the AM group. Fura-2 shows a spectrum shift upon binding Ca2+ ions, 380 nm being the wavelength for maximal excitation in the absence and 340 nm in the presence of Ca2+, providing the basis for measurements. The affinity constant for the fura-2−Ca2+ complex at 37°C, and physiological salt concentrations, is ∼220 nM (∼280 nM at 22°C), setting the sensitivity and the limitations of the probe for Ca2+ changes. Most cell types can be assayed for Ca2+ signalling in suspension using fura-2 (I, IV). However, suspension measurements do not provide information about cell heterogeneity nor spatial resolution of signalling. In addition, perfusion of cells is not technically possible. For the above purposes, the fura-2-loaded cells, on cover slips, were excited under microscopic view (IV).

3.7 Mathematical modelling A mathematical model of receptor-regulated AC activity was developed to account for promiscuous Gi/Gs-protein coupling (III). The model includes three different steps. The first step describes the agonist activation of the receptor, in which agonist concentration and efficacy can be varied to give different concentrations of activated receptor. The second step describes the activation of two different G-proteins, Gi and

Gs, by the activated receptor from the first step. In the second step, the outcome, i.e. relative concentrations of activated Gi and Gs, can be modified by varying the receptor’s affinity for the different G-proteins as well as by varying mutual competitiveness between the G-proteins for activation by the receptor. The third step

29 describes how Gi and Gs allosterically modulate AC to either decrease (Gi) or increase

(Gs) the catalytic activity. Although sufficiently sophisticated to enable the modelling work performed in paper III, the model suffers from some limitations and a refined model has been described after the publication of paper III (Kukkonen et al., 2001).

4 RESULTS

4.1 Receptor signalling in Sf9 cells 2+ Stimulation of Sf9 cells with octopamine led to an elevation of [Ca ]i and cAMP 2+ production (I). The potency of octopamine was similar for both events. The [Ca ]i increase originated from intracellular stores as well as from the extracellular space.

Activation of the expressed human , linked to Gs-proteins, resulted in cAMP elevation with no concomitant increase in Ca2+, excluding the role of cAMP in Ca2+ mobilization. When Ca2+ was chelated with EGTA, there was an attenuation of the cAMP increase in response to octopamine, suggesting a role for 2+ Ca in AC activation. However, activation of the muscarinic M3 receptor, linked to 2+ Gq-proteins, increased [Ca ]i, but had no effect on basal cAMP production. Using a panel of octopamine receptor/α2-AR antagonists, we found that the dual octopamine responses are mediated by different receptor populations in Sf9 cells.

It was not tested whether octopamine increases the IP3 production, indicative of

PLC activation. The muscarinic Gq-protein coupled receptors do increase the IP3 concentration in Sf9 cells (Kukkonen et al., 1996). It is also interesting in this context to note that the heterologously expressed human α2B-AR is coupled to IP3 production 2+ and [Ca ]i increase in Sf9 cells (Holmberg et al., 1998), and apparently uses the same dual coupling mechanism as the octopamine receptors to increase cAMP production (Figure 1).

30 5 Ca2+ EGTA 4 Iono

3

2 cAMP accumulation cAMP 1

0 _ NA NA NA

2+ Figure 1. Effects of Ca chelation on α2B-AR-mediated cAMP production. Sf9 cells expressing recombinant human α2B-AR were assayed for noradrenaline (NA)- stimulated cAMP production in the presence of 1 mM Ca2+ or in the presence of 1 mM EGTA. Some cells were pretreated with ionomycin (Iono) in the presence of EGTA to deplete cells of Ca2+. Experiments were performed in the presence of 30 µM forskolin. The concentration of NA was 100 µM. Data are means ± SEM from three different experiments performed in triplicate.

2+ The endogenous AC(s) in Sf9 cells integrated two signals, Gs and Ca , to increase the total cAMP synthesis. How this is mechanistically regulated is at present unknown. Comparison to the mammalian AC1, which is directly activated by the Ca2+/CaM complex (Tang et al., 1991), indicated that the Sf9-AC is regulated in a different manner, requiring a primary stimulant for Ca2+ effects. In paper V we investigated receptor regulation of the mammalian AC2,

heterologously expressed in Sf9 cells. The PLC-linked M3 receptor was able to stimulate this AC isoform via PKC. The PKC isozyme(s) involved probably belong to

31 the classical subfamily based on phorbol ester (TPA) sensitivity (V) and a Ca2+- dependence (data not shown).

All α2-AR subtypes can inhibit forskolin-stimulated Sf9-AC activity (Oker-

Blom et al., 1993, Jansson et al., 1995, II, III, V). In the case of human α2B-AR, the inhibition was increased when coexpressed with mammalian Gi-proteins (III). The

complex regulation of AC2 by Go-proteins, with both inhibitory and stimulatory input depending on primary stimulus, was also obtained with the endogenous G-proteins (V).

4.2 Propensity of α2-ARs for Gs coupling In paper II we investigated if the opposite coupling of the heterologously expressed

α2A- and α2B-ARs to cAMP production (Jansson et al., 1995) resided in the receptor proteins as sequence differences. The mouse orthologues of these subtypes were expressed in Sf9 cells at similar densities. The α2B-AR action on cAMP production was U-shaped, with inhibition at low and potentiation at higher noradrenaline

concentrations. The α2A-AR inhibited the AC activity in the equivalent concentration

range. The potentiating effect of the α2B-AR was hypothesized to result from promiscuous coupling to Gs-proteins and, if so, ought to involve cytoplasmic domains of the receptor. An exchange of the i3-loops between the subtypes did not alter the responses of the parent receptors. The carboxyl terminal domain neither seemed to be

involved. Of the remaining cytoplasmic domains, the i2-loop from α2B-AR conferred

the stimulatory effect to α2A-AR. Only a few amino acid residues of the i2-loops differ between the subtypes. Site-directed mutagenesis of single amino acid residues in the

i2-loop of α2A-AR was not enough to convincingly demonstrate an α2B-AR functionality, but a double mutant (S134→A, L143→S) showed an enhanced stimulatory capacity, suggesting that several amino acid residues are involved in the coupling specificity.

In paper III we wanted to provide evidence for true Gs-protein coupling of α2B- AR, and get insight into the molecular mechanisms that define the U-shaped

32 regulation of cAMP production. For these purposes, we coexpressed Gi- or Gs-proteins together with the human α2B-AR in Sf9 cells. An interaction between Gs and α2B-AR

was demonstrated by enhanced GTPγS binding to Gs-proteins upon receptor activation. An increase in the potency of noradrenaline to stimulate cAMP production,

and an increased maximal response, were also evident with Gs coexpression, indicative of an interaction in the cellular milieu as well.

The α2-ARs are coupled to an inhibition of cAMP synthesis in many cell types.

An increase in the Gi-protein level should thus, in the case of α2B-AR, if not displace, then at least counteract the effect of Gs on AC. Gi coexpression did attenuate the forskolin stimulation via α2B-AR, but part of the stimulatory component remained. However, the potentiating effect with noradrenaline could be overcome with longer

infection times, when the concentration of Gi-proteins was prominently increased (Figure 2).

There was an obvious difference in the activation mode of α2B-AR by different agonists. Noradrenaline produced U-shaped dose-response curves, while UK14,304 elicited the stimulatory phase only weakly in the same concentration range (II, III). The phenomenon, whereby different agonists can induce different responses through the same receptor, has been termed agonist-specific coupling (Robb et al., 1994). As can be seen from figure 2, this can have a major impact on AC regulation via

promiscuous Gs/Gi-protein coupling. In this context, it is worth pointing out that the partial agonist clonidine, with apparent low intrinsic efficacy for Gs coupling, induces inhibitory responses that are "fuller" than those of certain "full agonists".

33 200 α2B (25 h p.i.) α + G (25 h p.i.) 150 2B i α2B + Gi (38 h p.i.) 100

50

0

cAMP (percent change of forskolin) of change (percent cAMP -50 NA Oxy UK Clon

Figure 2. Ligand activity at α2B-AR coexpressed with Gi-proteins. Sf9 cells expressing recombinant human α2B-AR alone or together with Gi-proteins were assayed for agonist-mediated changes in forskolin-stimulated cAMP production at different times post infection (p.i.). The agonists were noradrenaline (NA), oxymetazoline (Oxy), UK14,304 (UK) and clonidine (Clon). The concentrations used were 100 µM for all agonists. Part of these data are presented in paper III, table 1. Data are means ± SEM (means ± SD, 38 h p.i.) from three different experiments (one experiment, 38 h p.i.) performed in triplicate.

4.3 Signalling cross-talk The AC activity in Sf9 cells gave an example of cross-talk between different receptors at the effector level. Two distinct signal pathways, elicited by different octopamine receptor populations, activating different G-proteins, converge on the AC activity to amplify the cAMP increase (I). In this system, Gs-protein activation, by one type of receptor, has an obligatory role for the potentiating effect of Ca2+ on the AC activity.

In paper IV it was shown that activation of purinergic Gq-coupled receptors was, in a 2+ similar way, obligatory for α2-AR-mediated increase in [Ca ]i in CHO cells. 2+ Stimulation of α2-ARs resulted in an increase in [Ca ]i of about 200 nM. Treatment of cells with to hydrolyze extracellular purine nucleotides significantly 2+ reduced the α2-AR-mediated [Ca ]i increase, as did the block of P2Y receptors with suramin. Neither apyrase nor suramin unspecifically influenced the G-protein coupling

34 of α2-ARs, as judged from the AC inhibition. With continuous perfusion of cells, to 2+ remove released purine nucleotides, the Ca response to α2-AR stimulation was

virtually absent, indicating that activation of Gq subfamily proteins is a prerequisite for

PLC activation through α2-ARs. 2+ Among the α2-ARs, only the α2B-AR subtype mediates a Ca response in Sf9 cells (Holmberg et al., 1998). Continuous perfusion of these cells did not attenuate the 2+ α2B-AR-mediated Ca response (Holmberg et al., 1998). In an effort to elucidate the 2+ mechanism for the Ca response in this cell type, we coexpressed the α2B-AR with Gi- 2+ or Gs-proteins. The Ca response was attenuated about 70% with Gi coexpression,

whereas Gs had no major influence on the response (Figure 3).

700

600

500 i ]

2+ 400

[Ca 300 ∆ 200

100

0 α -AR 2B + Gi + Gs

2+ Figure 3. Effect of G-protein expression on α2B-AR-mediated Ca signalling. Sf9 cells expressing recombinant human α2B-AR alone or together with Gi- or Gs-proteins 2+ were assayed for changes in [Ca ]i in response to noradrenaline (1 µM). Data are means ± SD from two experiments.

35 In paper V another example of cross-talk between receptors was demonstrated. PKC activation, by TPA or M3 receptor activation, elevated the cAMP content of Sf9 cells expressing AC2, but not of wild-type or AC1 expressing cells. The stimulation was

almost totally inhibited by activation of the α2A-AR or the M4 receptor expressed in the same cells. Since AC2 has been considered refractory to inhibitory G-proteins, this led us to further investigate this apparent discrepancy. We could show that concomitant activation of Gs-proteins switches the response of α2A-AR to stimulatory, presumably an effect via Gβγ, as it was sensitive to Gβγ scavenging by Gα11. To identify the G-protein species mediating the inhibition, we developed a method that take advantage of the viral turn-off of host protein synthesis. This enabled us to eliminate most of the receptor coupling to endogenous G-proteins by what we call "time-restricted pertussis toxin pretreatment", i.e. Ptx-sensitive endogenous G-proteins were uncoupled by Ptx pretreatment prior to infection with viruses coding for signal- ling components, including G-proteins. Since Ptx was removed soon after infection, the newly synthesized G-proteins could escape ADP ribosylation. Using this approach, it was shown that Go-proteins could inhibit TPA-stimulated activity and potentiate Gs-

stimulated activity of AC2. Expression of Gi, on the other hand, potentiated both of these primary stimulus events. The identity of Gβγ subunits were the same for both G- protein species, indicating that the identity of Gα subunit determines the outcome.

5 DISCUSSION

5.1 Suitability of Sf9 cells for receptor characterization Early reports on GPCRs in Sf9 cells described the successful expression of different receptors, and purification of functional receptors from these cells (George et al., 1989, Parker et al., 1991, Reilander et al., 1991, Vasudevan et al., 1991). Expression of GPCRs in Sf9 cells ususally results in high receptor densities, well suited for studying pharmacological binding characteristics (Parker et al., 1991, Mouillac et al., 1992, Quehenberger et al., 1992, Mills et al., 1993, Oker-Blom et al., 1993, Hartman

36 IV & Northup, 1996). The use of Sf9 cells for functional studies became somewhat dubious by a report stating an absence of Gi-proteins in these cells (Quehenberger et al., 1992). Nevertheless, soon afterwards, inhibition of Sf9-AC activity via the 5-HT1B receptor (Ng et al., 1993) and via the α2C-AR (Oker-Blom et al., 1993) was reported.

We have later shown that all three α2-AR subtypes can inhibit forskolin-stimulated AC activity in Sf9 cells (Jansson et al., 1995, II, III). In the case of the human α2B-AR, the inhibition was increased when coexpressed with mammalian Gi-proteins, suggesting Sf9-AC sensitivity toward this G-protein as well (III). The inhibitory G-proteins in Sf9 cells have not been characterized in detail, but at least a Go-like G-protein appears to be expressed (Ng et al., 1993, Mulheron et al.,

1994). Go-proteins are expressed predominantly in neural tissues, but are also found in

other places. The Gαo subunit identified in is also expressed in oocytes and ovarian nurse cells, in addition to its neuronal localization (Schmidt et al., 1989). Having in mind that Sf9 cells originate from ovarian tissue, it is not unlikely

that a Spodoptera Go is expressed in these cells. The endogenous inhibitory G-protein

can obviously substitute for the mammalian Go-protein for the inhibition of AC2 (V), supporting this hypothesis. Sf9 cells have often been used to address G-protein selectivity of various receptors by expression of selected receptor−G-protein pairs (Butkerait et al., 1995, Grunewald et al., 1996, Hartman IV & Northup, 1996, Barr et al., 1997, III). This has, on some occasions, led to confirmation of presumptive G-protein coupling of receptors (Butkerait et al., 1995, Barr et al., 1997, III). One reason for utilizing Sf9 cells for establishing receptor−G-protein coupling preferences is that relatively high levels of expression of desired proteins are obtained, improving the signal-to-noise ratio. An- other feature of Sf9 cells is the low level of endogenous GPCRs interfering with the ectopic receptors (Hu et al., 1994, Butkerait et al., 1995, Barr et al., 1997), as well as low levels of endogenous G-proteins functionally coupled to expressed receptors, as determined by lack of GTP-sensitive high-affinity agonist binding sites (Quehenberger et al., 1992, Butkerait et al., 1995, Grunewald et al., 1996).

37 Assessment of signalling pathways in whole cells could have some advantages of the above notions. For example, purinergic receptor signalling is obviously "disturbing" the cellular signalling status when working with isolated cell cultures (IV, Florio et al., 1999, Ostrom et al., 2000). We have not detected changes in cAMP production, or Ca2+ homeostasis, with the purinergic receptor agonists ATP and adenosine in uninfected Sf9 cells, or in cells infected with wild-type virus. This suggests that Sf9 cells could be a cell line of choice when such tonic signalling is not to interfere with the experimental outcome. Potentially interfering receptors expressed by the Sf9 cells are the octopamine receptors (Orr et al., 1992, Hu et al., 1994, I). This is especially relevant in adrenergic receptor research due to the structural similarity of ligands acting on receptors from these families. It has also been shown that octopamine activates α2-ARs (Airriess et al., 1997, Rudling et al., 2000). When tested on 2+ uninfected Sf9 cells, noradrenaline had an effect on the [Ca ]i and cAMP production at high concentrations (> 3 uM) (Figure 4). The effect of clonidine was prominent on 2+ the [Ca ]i level, but modest on cAMP accumulation. UK14,304 did not seem to activate the endogenous octopamine receptors (data not shown), and could be the agonist of choice for studying α2-ARs. Unfortunately, UK14,304 appears not to

activate the α2-ARs in the same manner as noradrenaline (see Figure 2).

38 1.0 300

0.8 250 i

200 ] 0.6 2+ 150

0.4 [Ca 100 ∆ 0.2 cAMP accumulation 50 ∆ 0.0 0 NA Oxy Clon Oct NA Oxy Clon Oct

Figure 4. Ligand activity at octopamine receptors in Sf9 cells. Uninfected Sf9 cells were assayed for α2-AR agonist-mediated changes of cAMP 2+ accumulation or [Ca ]i. The agonists were: noradrenaline (NA), oxymetazoline (Oxy), clonidine (Clon) and p-octopamine (Oct). The concentrations used were 100 µM for all agonists. The data for cAMP accumulation are given as changes from basal level and are 2+ means ± SD, N=3. Data for [Ca ]i are given as changes from basal level and are means ± SD, N=2.

A drawback of the BEVS is that the viral infection is lethal to cells. This renders the Sf9 cells unable to maintain ionic gradients and metabolic equilibrium status late in infection. A primary reason for this deterioration is the virally mediated turn-off of host protein synthesis. However, this can be turned to an advantage in receptor research by Ptx pretreatment prior to infection to abolish receptor coupling to endo- genous G-proteins, and subsequent recovery of signal pathways by heterologous expression of desired G-proteins (V).

5.2 Promiscuous G-protein coupling A receptor is primarily coupled to a specific subset of G-proteins and activation of these by the receptor is efficacious and occurs with high agonist potency. For example,

pioneering work with purified platelet α2-AR and G-proteins, reconstituted in phospholipid vesicles, revealed that this receptor activates Gi and Go very efficiently,

whereas activation of Gt and Gs is negligible (Cerione et al., 1986a). It has later been shown that many receptors are not that strict in selecting G-protein partners, but

39 instead they exhibit additional coupling to G-proteins from distantly related subfamilies (Gudermann et al., 1996). It is this phenomenon that is usually referred to as promiscuous G-protein coupling.

Activation of α2-ARs results in a seemingly controversial stimulation of cAMP synthesis in some cell types. This phenomenon was initially shown to be mediated by

PLA2 activation (Jones et al., 1991), but subsequent studies also revealed a probable

Gs-protein interaction of the α2A-AR in transfected CHO cells (Eason et al., 1992). In

Sf9 cells, the α2B-AR is linked to an increase in cAMP production, whereas the α2A- AR subtype decrease forskolin-stimulated AC activity (Jansson et al., 1995). A

productive Gs-protein coupling of the α2B-AR in these cells was demonstrated in paper III. Work from Dr. Liggett’s lab has suggested that there are subtle differences in the

propensities of the different α2-AR subtypes for Gs activation with the endogenous catecholamine ligands (Eason et al., 1994). Our initial studies pointed to an all-or-none difference between α2B-AR, on the one hand, and α2A-AR and α2C-AR on the other

(Oker-Blom et al., 1993, Jansson et al., 1995). A true difference between the α2A- and

α2B-AR subtypes, although not as dramatic, was demonstrated in paper II, in which the coupling differences cannot be ascribed to differences in receptor densities. It now seems clear that the α2B-AR subtype is more propensive for Gs coupling than the other subtypes (Eason & Liggett, 1993, Pepperl & Regan, 1993, II, Rudling et al., 2000,

III). The weak capability of the α2A-AR to stimulate cAMP production in the presence

of surplus Gs-proteins also supports this notion (III). The structure of the i2-loop appears to be a determinant for this subdivision

(II). The whole selection structure for Gs coupling is probably not confined to this area, since potent Gs activation is also obtained with α2A in different cell types (see Table 2). The first study on this issue showed that deletion of the amino terminus of

the i3-loop of α2A-AR eliminates stimulation of AC activity, indicating a Gs coupling domain in this area (Eason & Liggett, 1995). However, substitution of this domain with corresponding domain from β2-AR, the prototype Gs coupled receptor, attenuated the Gs coupling when compared with the intact α2A-AR (Eason & Liggett, 1995). On

40 the other hand, substitution of the i2-loop with β2-AR sequence significantly improved the Gs coupling (Jewell-Motz et al., 1997), supporting our hypothesis about the importance of i2-loop structure for Gs coupling (II).

The dose-response curve forms for α2-AR action on AC activity varies a lot from cell type to cell type and from receptor subtype to receptor subtype, including U- shaped relations in addition to monophasic stimulatory and inhibitory curves. Even more intriguing, all these different forms of dose-response curves can be obtained in a single cell type with a single receptor subtype (III). To be able to explain these

behaviours as a simple consequence of dual Gi and Gs coupling, we developed a mathematical model that monitor AC activity in relation to Gi and Gs activation (III). Using this model, it was possible to reproduce the different curves obtained in experiments, suggesting that relative affinities of the receptor for Gi and Gs activation can alone explain the different behaviours. The results from the modelling also suggested that different agonists, like noradrenaline and UK14,304, differentially activate the receptor and thereby dictate relative affinity differences of the receptor for the two G-protein species.

It is difficult to correlate the promiscuous G-protein coupling of α2-ARs to a specific physiological effect. It is clear though, that a decrease in cAMP synthesis

cannot explain many effects of α2-AR activation observed in vivo (Isom & Limbird, 1988). A comment on recent progress in β-AR research on this topic may be valid here. Prior to cloning of the β-AR, purification enabled reconstitution of this receptor with G-proteins. From these experiments it was concluded that the β-AR cross-reacted

with Gi (Cerione et al., 1985). A couple of years ago, it was shown that β-ARs evoke a Ca2+ signal in submandibular gland cells through a mechanism involving activation of Gi-proteins (Luo et al., 1999). Very recently, Dr. Kobilka’s group provided evidence for promiscuous coupling of β2-AR to Ptx-sensitive G-proteins in myocyte cultures from β1-AR knock-out mice, the result of which was recorded as a decrease in contraction rate (Devic et al., 2001). These studies implicates that promiscuous coupling may be a general mode of diversifying receptor signalling and not simply a

41 laboratory phenomenon due to high expression levels of receptors, or abnormal signalling environments of used cell types.

5.3 Adenylyl cyclase regulation by the Ca2+ signalling pathway AC regulation by Ca2+/CaM is perhaps the most well conserved regulatory theme of cAMP synthesis. Pathogenicity of Bacillus anthracis and Bordetella pertussis is partially due to secreted, soluble Ca2+/CaM-stimulated AC enzymes (Danchin, 1993). In higher organisms, Ca2+-regulated AC activity is particularly associated with neural functions (Dudai & Zvi, 1984, Xia & Storm, 1997, Chern, 2000). In Sf9 cells, the AC 2+ 2+ activity was not stimulated by an increase in [Ca ]i, but the primary, Ca -

independent, cAMP elevation with octopamine, and with α2B-AR activation, was 2+ highly potentiated by a concomitant increase in [Ca ]i. The potency of forskolin for 2+ AC stimulation was similar in the absence and presence of elevated [Ca ]i, suggesting that Ca2+ does not alter the affinity for the primary stimulant. A likely explanation is that Ca2+ increases the activity of the enzyme, integrating the Ca2+ signal with the direct activator. Signalling molecules with the property of integrating multiple signals are called coincidence detectors (Iyengar, 1996). Coincidence detection of ACs is thought to play important roles in transcription, synaptic plasticity and endocrine function.

AC2 is a coincidence detector for Gαs and PKC as well as Gβγ subunits (reviewed in Sunahara et al., 1996). A cross-talk between PKC and AC was suggested in 1987 in a paper showing phorbol ester-mediated phosphorylation of AC in erythrocytes (Yoshimasa et al., 1987). However, it is still unclear what physiological role PKC play in cAMP signalling. AC2 has been suggested to play a role in synaptic plasticity (Defer et al., 2000), although this has never been demonstrated. The AC2 isoform has in several instances been implicated in growth and development. In NIH- 3T3 cells, ectopic expression of AC2 is sufficient to inhibit proliferation in response to serum (Smit et al., 1998). Neuronal, as well as mesodermal, differentiation of the teratocarcinoma P19 cells is accompanied by an upregulation of AC2 mRNA (Lipskaia et al., 1997, Lipskaia et al., 1998). A related isoform of the mammalian AC2

42 is expressed in Caenorhabditis elegans. Loss of function of this isoform results in early larval lethality, indicating an important role in development (Korswagen et al., 1998). In mammals, AC2 is widely expressed in the brain with no specific area preference (see e.g. Mons & Cooper, 1995). This suggests a role in common neuronal action or development. In light of the results presented in paper V, it is tempting to speculate, as follows, about such a common action. Neurotransmitter release is a 2+ 2+ complicated process triggered by [Ca ]i increases. The amount of Ca entering the nerve terminal is strictly regulated by voltage-gated Ca2+-channels. Some of these channels are targets for PKC-mediated upregulation of activity (Zamponi et al., 1997),

whereas attenuation is transmitted from GPCRs, such as α2-ARs, via activation of Go-

or Gi-proteins (for review, see e.g. Dolphin, 1998). Recent studies have implicated a role also for cAMP in the neurotransmitter release process (Trudeau et al., 1996, Chavis et al., 1998, Chang et al., 2000). Thus, if AC2 and Ca2+-channels were co- localized in the nerve terminals, both a coordinated stimulation of AC2 and Ca2+-

channels via PKC, or a coordinated inhibition of the same molecules via Go-proteins, could account for synergistic effects on transmitter release.

2+ 5.4 The α2-AR−Ca link 2+ There are now compelling evidence that α2-ARs can alter the [Ca ]i by other mechanisms than through ion channel modulation. Activation of α2-ARs in a variety of cell types mobilizes Ca2+ from intracellular stores (Michel et al., 1989, Salm & McCarthy, 1990, Åkerman et al., 1996, Dorn et al., 1997). In light of the multitude of 2+ Gi/o coupled receptors that have been shown to mobilize Ca , it is no wonder that the

α2-ARs behave similarly. It was early reported that, for some receptors, the link to Ca2+ mobilization and PLC activation is sensitive to Ptx (see e.g. Ui et al., 1988). The finding that PLC-βs are stimulated by Gβγ subunits gave a satisfactory explanation to

2+ this phenomenon. For the α2-ARs, a Gβγ-dependent Ca mobilization has been de- monstrated in COS-7 cells (Dorn et al., 1997).

43 Characterization of PLC-β isoforms in vitro suggested a direct activation by

Gβγ, without conditioning by Gq subfamily proteins (Camps et al., 1992, Katz et al., 2+ 1992). In CHO cells, expressing either subtype of α2-ARs, the α2-AR-mediated Ca mobilization was highly dependent on tonic stimulation of Gq coupled receptors (IV). This led us to hypothesize that Gβγ-mediated PLC signalling requires concomitant

αq/11/14/16 activation. The mechanistic explanation has been elaborated in a study by

Dr. Wong and colleagues, showing that Gi-protein coupled opioid receptors could not

activate PLC, unless coexpressed with Gα16, in COS-7 cells (Chan et al., 2000). On the same theme, they showed that the opioid receptors acquire the ability to stimulate

PLC in cells cotransfected with constitutively activated Gαq or Gα14. Further support for the relevance of cross-talk or promiscuous G-protein coupling in Ca2+ signalling has appeared from two papers describing signalling in knock-out mice deficient in Gq family proteins. First, PLC activation by thrombin, a Gq and Gi coupled receptor, is

abolished in platelets from Gαq-deficient mice, indicating that Gi activation alone is insufficient for PLC activation (Offermanns et al., 1997). Second, the C5a receptor, which is strongly coupled to Ca2+ mobilization through Ptx-sensitive G-proteins, 2+ looses the Ca -mobilizing ability in macrophages from Gα15 knock-out mice, while MAPK activation is normal (Davignon et al., 2000).

Most Gi-protein coupled receptors in hematopoietic cells, such as the C5a 2+ receptor described above, usually respond to agonists with robust [Ca ]i increases.

Hematopoietic cells express the Gα15/16 subtype of G-proteins (Amatruda et al., 1991).

The G15/16-protein has been found rather unselective for receptor interactions (reviewed in Milligan et al., 1996). Thus, if there is promiscuous receptor coupling to 2+ G15/16, the Ca response should largely benefit from this. In HEL cells, α2-ARs give prominent Ca2+ responses (Michel et al., 1989, Kukkonen et al., 1997, Jansson et al., 1998), compared to for example primary astrocytes (Enkvist et al., 1996) and NG108-

15 cells (Holmberg et al., 1998). A reason for this may be promiscuous receptor−G16- protein coupling in HEL cells, which has been shown for purinergic receptors in this cell line (Baltensperger & Porzig, 1997).

44 Apyrase treatment of CHO cells did not totally abolish, especially in the case of the 2+ 2+ α2B-AR, the Ca response of α2-ARs (IV). In some instances, the Ca signalling mediated by α2-ARs is not abolished by Ptx treatment. In Ptx-treated NG108-15 cells, 2+ the Ca response of the endogenous α2B-AR is inhibited by about 65% (Holmberg et 2+ al., 1998). In CHO cells, the α2B-AR-mediated [Ca ]i increase is attenuated by about

80% in Ptx-treated cells, whereas α2A- and α2C-AR responses are completely blocked 2+ (Kukkonen et al., 1998). Of the α2-ARs, only the α2B-AR subtype mediates a Ca response in Sf9 cells, and the response in this cell type is insensitive to Ptx (Holmberg 2+ et al., 1998). Contrary to expectations, the Ca response of α2B-AR was attenuated by

Gi coexpression in these cells (Figure 3), suggesting a Gβγ-independent pathway. 2+ Although the α2B-AR is promiscuously coupled to Gs-proteins, the Ca response is probably not associated with this G-protein species (I). The hypothesis is that another

G-protein than Gi/o or Gs is involved. A Gq subfamily protein is a good candidate. 2+ However, we have not detected any change in the Ca signalling mode of α2B-AR in

Sf9 cells with coexpression of the mammalian G11-protein, which, if coupled to the receptor, would be expected to work in an analogous fashion to Gs-protein expression, increasing agonist potency and/or maximum response (III). This does not exclude the possibility of the insect Gq as the linker, but additional work will be needed to either 2+ exclude or include this, as well as other, potential pathways in α2-AR-mediated Ca signalling.

45 6 SUMMARY AND CONCLUSIONS

The use of BEVS appears well suited for functional characterization of receptor signalling. The signalling components of Sf9 cells are well recognized by mammalian receptors, and heterologous expression of complementary signalling molecules can be used to assess these in signal transduction of particular GPCRs. The lack of endogenous receptors for most mammalian hormones and neurotransmitters in Sf9 cells provides a good cellular background for heterologous receptor characterization. In addition, the predetermined fate of baculovirus-infected cells, with the concomitant inhibited host protein synthesis, can be used advantageously for examination of Ptx- sensitive G-protein function.

The α2B-AR was demonstrated to couple promiscuously to Gs-proteins both in membrane preparations and live cells. The ability of the mouse and the human α2B-AR subtype to induce stimulatory cAMP responses in Sf9 cells through endogenous G- proteins suggests a similarly productive Gs coupling, and an interspecies similarity in this regard. Using a chimeric receptor approach, it was demonstrated that the structure

of the i2-loop of α2B-AR is a determinant for Gs coupling, and that it is this loop that

differentiates the G-protein coupling selectivities of the α2A- and α2B-ARs. The influence of receptor density on the cellular response is also obvious from the studies.

The human α2B-AR, expressed at relatively high levels, induces mainly stimulatory responses. The chimeric mouse α2A/α2B-i2 loop receptor is also primarily coupled to stimulation at high receptor densities, but exhibits U-shaped inhibitory and stimulatory responses at lower densities. The mouse α2B-AR, expressed at relatively low density, exhibits a U-shaped cAMP response.

The AC stimulatory capacity of α2-ARs may not always be associated with a Gs- protein coupling, as shown in paper V. The results of paper V demonstrate that a

selective activation of Gi or Go has distinct consequences for AC2 regulation.

46 Coupling of the α2A-AR to Go resulted in inhibition of the PKC-stimulated AC2

activity. In contrast, Gi activation, by the same receptor, resulted in potentiation of the

PKC-stimulated AC2 activity. A Gβγ-mediated potentiation of Gs-stimulated AC2 activity was evident for both types of G-proteins, indicating that the α subunits of Go and Gi have different roles for this AC isoform.

2+ Ca mobilization by α2-ARs is sensitive to Ptx in some cell types and is likely to be a 2+ result of Gβγ-mediated stimulation of PLC. In CHO cells, the Ca response of α2- ARs was attenuated, almost to zero response in some cases, if extracellular purine nucleotides were removed. This implicated purinergic receptors in the signalling

cascade, which was confirmed by attenuation of α2-AR response in the presence of suramin, a purinergic . The results suggest a receptor cross-talk at the level of PLC enzymes. The tonic purinergic receptor activation will provide PLCs with GTP-loaded α subunits of the Gq subfamily proteins and thereby prime the PLC for Gβγ signalling. Thus, Gα subunit activation seems to be obligatory for Gβγ signalling in this model system.

A trend of the α2A-AR subtype being promiscuously coupled to Gs, whereas the α2B-

AR is nearly unproductive in Gs coupling, was inferred from the first paper on

promiscuous G-protein coupling of α2-ARs (Eason et al., 1992). In our hands, the α2B- 2+ AR is far more propensive for Gs coupling than the α2A-AR. Regarding the Ca signalling, all α2-AR subtypes have the ability to regulate PLC activity in CHO cells. 2+ In Sf9 cells, only the α2B-AR is coupled to a [Ca ]i increase, suggesting again a

fundamental difference between α2-AR subtypes on functional criteria.

47 7 ACKNOWLEDGEMENTS

This study was conducted at the Department of Physiology, Uppsala University under supervision of professor Karl Åkerman. The study was funded by The European Commission, The Medical Research Council of Sweden, The Research Fund of Sweden, The Clas Groschinsky Foundation, The Åke Wiberg Foundation and The Lars Hiertas Minnesfond

I would like to thank: Professor Karl Åkerman for providing an opportunity for a young biochemistry student to take part in up to date scientific research, for sharing his knowledge in cellular signalling, and for being a friend, and a solver of all kinds of problems.

Professor Gunnar Flemström, my co-supervisor, for his advice and guidance on a variety of matters.

Dr Jyrki Kukkonen, co-author and friend, for invaluable help with pharmacological issues and computer managing. Dr Kukkonen has always been open for discussions of all kinds of matters and is much appreciated for this. Dr Kukkonen also read this thesis and supplied valid criticism. Dr Per-Eric Lund, co-author and friend, for numerous discussions of ion channels, cars and other every day matter. Dr Staffan Uhlén for sharing his excellent knowledge of adrenoceptor pharmacology and for our discussions about child behaviour. Dr Mikael Jolkkonen for fruitful cooperation in my second project on muscarinic toxins. Dr Viktoria Göransson for helpful guidance of thesis writing.

48 All former and present PhD student friends at the department, and especially those of the Åkerman group: Ramin Shariatmadari, co-author, Tomas Holmqvist, co-author, Sylwia Ammoun, co-author, and Elena Krioukova, for all their help and encourage- ment. Tomas is also acknowledged for his appreciated humorous inputs in everyday misery and for critical reading of the thesis before printing.

Technical assistant Karin Nygren for taking care of all kinds of different matters required for on-going research and for the friendly welcome of the "Finns" to the department.

All other persons at the department and especially the technical personnel for help with all matters associated with being a PhD student.

My former colleagues at the Åbo Akademi University, Turku, Finland. Some of the persons deserve a mentioning here: Dr Christian Jansson, co-author, for teaching me the basic of cAMP and how to measure it, Dr Christian Oker-Blom for introducing the BEVS, Pekka Ojala and Heli Hämäläinen for their excellent supervision of molecular biology techniques.

My parents for their love and support.

My life partner, Pi, and our children, Max and My, for sharing their lives with me.

49 8 REFERENCES

Aantaa, R., Marjamaki, A., and Scheinin, M. (1995) Molecular pharmacology of alpha 2- adrenoceptor subtypes. Ann. Med. 27: 439-449. Aburto, T. K., Lajoie, C., and Morgan, K. G. (1993) Mechanisms of signal transduction during alpha 2-adrenergic receptor-mediated contraction of vascular smooth muscle. Circ. Res. 72: 778-785. Ahlquist, R. P. (1948) A study of adrenotropic receptors. Am. J. Physiol. 153: 586-600. Airriess, C. N., Rudling, J. E., Midgley, J. M., and Evans, P. D. (1997) Selective inhibition of adenylyl cyclase by octopamine via a human cloned alpha 2A-adrenoceptor. Br. J. Pharmacol. 122: 191-198. Amatruda, T. T., Steele, D. A., Slepak, V. Z., and Simon, M. I. (1991) G alpha 16, a alpha subunit specifically expressed in hematopoietic cells. Proc. Natl. Acad. Sci. U. S. A. 88: 5587-5591. Antoni, F. A., Palkovits, M., Simpson, J., Smith, S. M., Leitch, A. L., Rosie, R., Fink, G., and Paterson, J. M. (1998) Ca2+/calcineurin-inhibited adenylyl cyclase, highly abundant in forebrain regions, is important for learning and memory. J. Neurosci. 18: 9650-9661. Ashkenazi, A., and Peralta, E. G. (1994) Muscarinic Acetylcholine Receptors. In Handbook of Receptors and Channels (Peroutka, S. J., ed), pp. 1-27, CRC Press, Inc., Boca Raton. Baltensperger, K., and Porzig, H. (1997) The P2U purinoceptor obligatorily engages the G16 to mobilize intracellular Ca2+ in human erythroleukemia cells. J. Biol. Chem. 272: 10151-10159. Barr, A. J., Brass, L. F., and Manning, D. R. (1997) Reconstitution of receptors and GTP- binding regulatory proteins (G proteins) in Sf9 cells. A direct evaluation of selectivity in receptor.G protein coupling. J. Biol. Chem. 272: 2223-2229. Bayewitch, M. L., Avidor-Reiss, T., Levy, R., Pfeuffer, T., Nevo, I., Simonds, W. F., and Vogel, Z. (1998) Inhibition of adenylyl cyclase isoforms V and VI by various Gbetagamma subunits. FASEB J. 12: 1019-1025. Berlan, M., Montastruc, J. L., and Lafontan, M. (1992) Pharmacological prospects for alpha 2-adrenoceptor antagonist therapy. Trends Pharmacol. Sci. 13: 277-282. Berridge, M. J. (1987) and diacylglycerol: two interacting second messengers. Annu. Rev. Biochem. 56: 159-193. Berridge, M. J. (1997) Elementary and global aspects of calcium signalling. J Physiol 499: 291-306. Berridge, M. J., and Irvine, R. F. (1984) Inositol trisphosphate, a novel second messenger in cellular signal transduction. Nature 312: 315-321. Berthelsen, S., and Pettinger, W. A. (1977) A functional basis for classification of alpha- adrenergic receptors. Life Sci. 21: 595-606. Birnbaumer, L., and Birnbaumer, M. (1995) Signal transduction by G proteins: 1994 edition. J. Recept. Signal Transduct. Res. 15: 213-252. Blumer, K. J., and Thorner, J. (1991) Receptor-G protein signaling in yeast. Annu. Rev. Physiol. 53: 37-57. Bockman, C. S., Jeffries, W. B., and Abel, P. W. (1993) Binding and functional characterization of alpha-2 adrenergic receptor subtypes on pig vascular endothelium. J. Pharmacol. Exp. Ther. 267: 1126-1133.

50 Bokoch, G. M., Katada, T., Northup, J. K., Ui, M., and Gilman, A. G. (1984) Purification and properties of the inhibitory guanine nucleotide-binding regulatory component of adenylate cyclase. J. Biol. Chem. 259: 3560-3567. Burnstock, G. (1980) Purinergic nerves and receptors. Prog. Biochem. Pharmacol. 16: 141- 154. Butkerait, P., Zheng, Y., Hallak, H., Graham, T. E., Miller, H. A., Burris, K. D., Molinoff, P. B., and Manning, D. R. (1995) Expression of the human 5-hydroxytryptamine1A receptor in Sf9 cells. Reconstitution of a coupled phenotype by co-expression of mammalian G protein subunits. J. Biol. Chem. 270: 18691-18699. Bylund, D. B. (1985) Heterogeneity of alpha-2 adrenergic receptors. Pharmacol. Biochem. Behav. 22: 835-843. Bylund, D. B., Ray-Prenger, C., and Murphy, T. J. (1988) Alpha-2A and alpha-2B adrenergic receptor subtypes: antagonist binding in tissues and cell lines containing only one subtype. J. Pharmacol. Exp. Ther. 245: 600-607. Camps, M., Carozzi, A., Schnabel, P., Scheer, A., Parker, P. J., and Gierschik, P. (1992) Isozyme-selective stimulation of phospholipase C-beta 2 by G protein beta gamma- subunits. Nature 360: 684-686. Cassel, D., and Selinger, Z. (1976) Catecholamine-stimulated GTPase activity in turkey erythrocyte membranes. Biochim. Biophys. Acta 452: 538-551. Cassel, D., and Selinger, Z. (1977) Mechanism of adenylate cyclase activation by cholera toxin: inhibition of GTP hydrolysis at the regulatory site. Proc. Natl. Acad. Sci. U. S. A. 74: 3307-3311. Castagna, M., Takai, Y., Kaibuchi, K., Sano, K., Kikkawa, U., and Nishizuka, Y. (1982) Direct activation of calcium-activated, phospholipid-dependent protein kinase by tumor- promoting phorbol esters. J. Biol. Chem. 257: 7847-7851. Cerione, R. A., Regan, J. W., Nakata, H., Codina, J., Benovic, J. L., Gierschik, P., Somers, R. L., Spiegel, A. M., Birnbaumer, L., Lefkowitz, R. J., and et al. (1986a) Functional reconstitution of the alpha 2-adrenergic receptor with guanine nucleotide regulatory proteins in phospholipid vesicles. J. Biol. Chem. 261: 3901-3909. Cerione, R. A., Staniszewski, C., Benovic, J. L., Lefkowitz, R. J., Caron, M. G., Gierschik, P., Somers, R., Spiegel, A. M., Codina, J., and Birnbaumer, L. (1985) Specificity of the functional interactions of the beta-adrenergic receptor and rhodopsin with guanine nucleotide regulatory proteins reconstituted in phospholipid vesicles. J. Biol. Chem. 260: 1493-1500. Cerione, R. A., Staniszewski, C., Gierschik, P., Codina, J., Somers, R. L., Birnbaumer, L., Spiegel, A. M., Caron, M. G., and Lefkowitz, R. J. (1986b) Mechanism of guanine nucleotide regulatory protein-mediated inhibition of adenylate cyclase. Studies with isolated subunits of in a reconstituted system. J. Biol. Chem. 261: 9514-9520. Chabre, O., Conklin, B. R., Brandon, S., Bourne, H. R., and Limbird, L. E. (1994) Coupling of the alpha 2A-adrenergic receptor to multiple G-proteins. A simple approach for estimating receptor-G-protein coupling efficiency in a transient expression system. J. Biol. Chem. 269: 5730-5734. Chan, J. S., Lee, J. W., Ho, M. K., and Wong, Y. H. (2000) Preactivation permits subsequent stimulation of phospholipase C by G(i)-coupled receptors. Mol. Pharmacol. 57: 700-708.

51 Chang, D. J., Li, X. C., Lee, Y. S., Kim, H. K., Kim, U. S., Cho, N. J., Lo, X., Weiss, K. R., Kandel, E. R., and Kaang, B. K. (2000) Activation of a heterologously expressed octopamine receptor coupled only to adenylyl cyclase produces all the features of presynaptic facilitation in aplysia sensory neurons. Proc. Natl. Acad. Sci. U. S. A. 97: 1829-1834. Chavis, P., Mollard, P., Bockaert, J., and Manzoni, O. (1998) Visualization of cyclic AMP- regulated presynaptic activity at cerebellar granule cells. Neuron 20: 773-781. Chern, Y. (2000) Regulation of adenylyl cyclase in the central nervous system. Cell. Signal. 12: 195-204. Chidiac, P. (1998) Rethinking receptor-G protein-effector interactions. Biochem. Pharmacol. 55: 549-556. Choi, E. J., Xia, Z., and Storm, D. R. (1992) Stimulation of the type III olfactory adenylyl cyclase by calcium and calmodulin. Biochemistry 31: 6492-6498. Chruscinski, A. J., Link, R. E., Daunt, D. A., Barsh, G. S., and Kobilka, B. K. (1992) Cloning and expression of the mouse homolog of the human alpha 2-C2 adrenergic receptor. Biochem. Biophys. Res. Commun. 186: 1280-1287. Codina, J., Hildebrandt, J., Iyengar, R., Birnbaumer, L., Sekura, R. D., and Manclark, C. R. (1983) Pertussis toxin substrate, the putative Ni component of adenylyl cyclases, is an alpha beta heterodimer regulated by guanine nucleotide and magnesium. Proc. Natl. Acad. Sci. U. S. A. 80: 4276-4280. Correa-Sales, C., Rabin, B. C., and Maze, M. (1992) A hypnotic response to dexmedetomidine, an alpha 2 agonist, is mediated in the locus coeruleus in rats. Anesthesiology 76: 948-952. Cotecchia, S., Kobilka, B. K., Daniel, K. W., Nolan, R. D., Lapetina, E. Y., Caron, M. G., Lefkowitz, R. J., and Regan, J. W. (1990) Multiple second messenger pathways of alpha- adrenergic receptor subtypes expressed in eukaryotic cells. J. Biol. Chem. 265: 63-69. Danchin, A. (1993) Phylogeny of adenylyl cyclases. Adv. Second Messenger Phosphoprotein Res. 27: 109-162. Davignon, I., Catalina, M. D., Smith, D., Montgomery, J., Swantek, J., Croy, J., Siegelman, M., and Wilkie, T. M. (2000) Normal hematopoiesis and inflammatory responses despite discrete signaling defects in Galpha15 knockout mice. Mol. Cell. Biol. 20: 797-804. Defer, N., Best-Belpomme, M., and Hanoune, J. (2000) Tissue specificity and physiological relevance of various isoforms of adenylyl cyclase. Am. J. Physiol. 279: F400-F416. Dell’Acqua, M. L., and Scott, J. D. (1997) Protein kinase A anchoring. J. Biol. Chem. 272: 12881-12884. Deupree, J. D., Hinton, K. A., Cerutis, D. R., and Bylund, D. B. (1996) Buffers differentially alter the binding of [3H]rauwolscine and [3H]RX821002 to the alpha-2 adrenergic receptor subtypes. J. Pharmacol. Exp. Ther. 278: 1215-1227. Devic, E., Xiang, Y., Gould, D., and Kobilka, B. (2001) Beta-adrenergic receptor subtype- specific signaling in cardiac myocytes from beta(1) and beta(2) adrenoceptor knockout mice. Mol. Pharmacol. 60: 577-583. Dixon, R. A., Kobilka, B. K., Strader, D. J., Benovic, J. L., Dohlman, H. G., Frielle, T., Bolanowski, M. A., Bennett, C. D., Rands, E., Diehl, R. E., and et al. (1986) Cloning of the gene and cDNA for mammalian beta-adrenergic receptor and homology with rhodopsin. Nature 321: 75-79. Dolphin, A. C. (1998) Mechanisms of modulation of voltage-dependent calcium channels by G proteins. J Physiol 506: 3-11.

52 Dorn, G. W., Oswald, K. J., McCluskey, T. S., Kuhel, D. G., and Liggett, S. B. (1997) Alpha 2A-adrenergic receptor stimulated calcium release is transduced by Gi-associated G(beta gamma)-mediated activation of phospholipase C. Biochemistry 36: 6415-6423. Drury, A. N., and Szent-Györgyi, A. (1929) The physiological activity of adenine compounds with special reference to their action upon the mammalian heart. J Physiol (Lond) 68: 213-237. Dudai, Y., and Zvi, S. (1984) Adenylate cyclase in the Drosophila memory mutant rutabaga displays an altered Ca2+ sensitivity. Neurosci. Lett. 47: 119-124. Duzic, E., and Lanier, S. M. (1992) Factors determining the specificity of signal transduction by guanine nucleotide-binding protein-coupled receptors. III. Coupling of alpha 2- adrenergic receptor subtypes in a cell type-specific manner. J. Biol. Chem. 267: 24045- 24052. Eason, M. G., Jacinto, M. T., and Liggett, S. B. (1994) Contribution of ligand structure to activation of alpha 2-adrenergic receptor subtype coupling to Gs. Mol. Pharmacol. 45: 696-702. Eason, M. G., Kurose, H., Holt, B. D., Raymond, J. R., and Liggett, S. B. (1992) Simultaneous coupling of alpha 2-adrenergic receptors to two G-proteins with opposing effects. Subtype-selective coupling of alpha 2C10, alpha 2C4, and alpha 2C2 adrenergic receptors to Gi and Gs. J. Biol. Chem. 267: 15795-15801. Eason, M. G., and Liggett, S. B. (1993) Functional alpha 2-adrenergic receptor-Gs coupling undergoes agonist-promoted desensitization in a subtype-selective manner. Biochem. Biophys. Res. Commun. 193: 318-323. Eason, M. G., and Liggett, S. B. (1995) Identification of a Gs coupling domain in the amino terminus of the third intracellular loop of the alpha 2A-adrenergic receptor. Evidence for distinct structural determinants that confer Gs versus Gi coupling. J. Biol. Chem. 270: 24753-24760. Enkvist, M. O., Hamalainen, H., Jansson, C. C., Kukkonen, J. P., Hautala, R., Courtney, M. J., and Akerman, K. E. (1996) Coupling of astroglial alpha 2-adrenoreceptors to second messenger pathways. J. Neurochem. 66: 2394-2401. Erdbrugger, W., Raulf, M., Otto, T., and Michel, M. C. (1995) Does [3H]2-methoxy- idazoxan (RX 821002) detect more alpha-2-adrenoceptor agonist high-affinity sites than [3H]rauwolscine? A comparison of nine tissues and cell lines. J. Pharmacol. Exp. Ther. 273: 1287-1294. Erspamer, V., and Boretti, G. (1951) Identification and characterization by paper chromatography of enteramine, octopamine, , , and allied substances in extracts of posterior salivary glands of Octopoda and in other tissue extracts of vertebrates and invertebrates. Arch. Int. Pharmacodyn. 88: 296-332. Evans, P. D. (1981) Multiple receptor types for octopamine in the locust. J. Physiol. (Lond.) 318: 99-122. Exton, J. H. (1981) Molecular mechanisms involved in alpha-adrenergic responses. Mol. Cell. Endocrinol. 23: 233-264. Fain, J. N., and Garcia-Sainz, J. A. (1980) Role of phosphatidylinositol turnover in alpha 1 and of adenylate cyclase inhibition in alpha 2 effects of catecholamines. Life Sci. 26: 1183-1194. Fawzi, A. B., Fay, D. S., Murphy, E. A., Tamir, H., Erdos, J. J., and Northup, J. K. (1991) Rhodopsin and the retinal G-protein distinguish among G-protein beta gamma subunit forms. J. Biol. Chem. 266: 12194-12200.

53 Feder, D., Im, M. J., Klein, H. W., Hekman, M., Holzhofer, A., Dees, C., Levitzki, A., Helmreich, E. J., and Pfeuffer, T. (1986) Reconstitution of beta 1-adrenoceptor-dependent adenylate cyclase from purified components. EMBO J. 5: 1509-1514. Federman, A. D., Conklin, B. R., Schrader, K. A., Reed, R. R., and Bourne, H. R. (1992) Hormonal stimulation of adenylyl cyclase through Gi-protein beta gamma subunits. Nature 356: 159-161. Florio, C., Frausin, F., Vertua, R., and Gaion, R. M. (1999) Amplification of the cyclic AMP response to forskolin in pheochromocytoma PC12 cells through adenosine A(2A) purinoceptors. J. Pharmacol. Exp. Ther. 290: 817-824. Flower, D. R. (1999) Modelling G-protein-coupled receptors for drug design. Biochim. Biophys. Acta 1422: 207-234. Fraser, C. M., Arakawa, S., McCombie, W. R., and Venter, J. C. (1989) Cloning, sequence analysis, and permanent expression of a human alpha 2-adrenergic receptor in Chinese hamster ovary cells. Evidence for independent pathways of receptor coupling to adenylate cyclase attenuation and activation. J. Biol. Chem. 264: 11754-11761. Fredholm, B. B., Abbracchio, M. P., Burnstock, G., Daly, J. W., Harden, T. K., Jacobson, K. A., Leff, P., and Williams, M. (1994) Nomenclature and classification of purinoceptors. Pharmacol. Rev. 46: 143-156. Fung, B. K. (1983) Characterization of transducin from bovine retinal rod outer segments. I. Separation and reconstitution of the subunits. J. Biol. Chem. 258: 10495-10502. George, S. T., Arbabian, M. A., Ruoho, A. E., Kiely, J., and Malbon, C. C. (1989) High- efficiency expression of mammalian beta-adrenergic receptors in baculovirus-infected insect cells. Biochem. Biophys. Res. Commun. 163: 1265-1269. Gether, U., Lin, S., and Kobilka, B. K. (1995) Fluorescent labeling of purified beta 2 adrenergic receptor. Evidence for ligand-specific conformational changes. J. Biol. Chem. 270: 28268-28275. Gilman, A. G. (1987) G proteins: transducers of receptor-generated signals. Annu. Rev. Biochem. 56: 615-649. Grunewald, S., Reilander, H., and Michel, H. (1996) In vivo reconstitution of D2S receptor-mediated G protein activation in baculovirus-infected insect cells: preferred coupling to Gi1 versus Gi2. Biochemistry 35: 15162-15173. Grynkiewicz, G., Poenie, M., and Tsien, R. Y. (1985) A new generation of Ca2+ indicators with greatly improved fluorescence properties. J. Biol. Chem. 260: 3440-3450. Gudermann, T., Grosse, R., and Schultz, G. (2000) Contribution of receptor/G protein signaling to cell growth and transformation. Naunyn Schmiedebergs Arch. Pharmacol. 361: 345-362. Gudermann, T., Kalkbrenner, F., and Schultz, G. (1996) Diversity and selectivity of receptor- G protein interaction. Annu. Rev. Pharmacol. Toxicol. 36: 429-459. Hall, R. A., Premont, R. T., and Lefkowitz, R. J. (1999) Heptahelical receptor signaling: beyond the G protein paradigm. J Cell Biol 145: 927-932. Hartman IV, J. L., and Northup, J. K. (1996) Functional reconstitution in situ of 5- hydroxytryptamine2c (5HT2c) receptors with alphaq and inverse agonism of 5HT2c receptor antagonists. J. Biol. Chem. 271: 22591-22597. Haslam, R. J., Davidson, M. M., and Desjardins, J. V. (1978) Inhibition of adenylate cyclase by adenosine analogues in preparations of broken and intact human platelets. Evidence for the unidirectional control of platelet function by cyclic AMP. Biochem. J. 176: 83-95. Henderson, R., and Unwin, P. N. (1975) Three-dimensional model of purple membrane obtained by electron microscopy. Nature 257: 28-32.

54 Hepler, J. R., and Gilman, A. G. (1992) G proteins. Trends Biochem. Sci. 17: 383-387. Hieble, J. P., Bondinell, W. E., and Ruffolo, R. R., Jr. (1995) Alpha- and beta-adrenoceptors: from the gene to the clinic. 1. Molecular biology and adrenoceptor subclassification. J. Med. Chem. 38: 3415-3444. Hildebrandt, J. D. (1997) Role of subunit diversity in signaling by heterotrimeric G proteins. Biochem. Pharmacol. 54: 325-339. Hoffman, B. B., Lavin, T. N., Lefkowitz, R. J., and Ruffolo, R. R. (1981) Alpha adrenergic receptor subtypes in rabbit uterus: mediation of myometrial contraction and regulation by estrogens. J. Pharmacol. Exp. Ther. 219: 290-295. Holmberg, C. I., Kukkonen, J. P., Bischoff, A., Näsman, J., Courtney, M. J., Michel, M. C., and Åkerman, K. E. (1998) Alpha2B-adrenoceptors couple to Ca2+ increase in both endogenous and recombinant expression systems. Eur. J. Pharmacol. 363: 65-74. Hu, Y., Rajan, L., and Schilling, W. P. (1994) Ca2+ signaling in Sf9 insect cells and the functional expression of a rat brain M5 muscarinic receptor. Am. J. Physiol. 266: C1736- 1743. Hug, H., and Sarre, T. F. (1993) Protein kinase C isoenzymes: divergence in signal transduction? Biochem. J. 291: 329-343. Inoue, M., Kishimoto, A., Takai, Y., and Nishizuka, Y. (1977) Studies on a cyclic nucleotide- independent protein kinase and its proenzyme in mammalian tissues. II. Proenzyme and its activation by calcium-dependent protease from rat brain. J. Biol. Chem. 252: 7610- 7616. Iourgenko, V., Kliot, B., Cann, M. J., and Levin, L. R. (1997) Cloning and characterization of a Drosophila adenylyl cyclase homologous to mammalian type IX. FEBS Lett. 413: 104- 108. Isom, L. L., and Limbird, L. E. (1988) What happens next? A hypothesis linking the biochemical and electrophysiological sequelae of alpha-2 adrenergic receptor occupancy with the diverse receptor-mediated phsiological effects. In The Alpha-2 Adrenergic Receptors (Limbird, L. E., ed), pp. 323-363, Humana press, Clifton, NJ. Iyengar, R. (1996) Gating by cyclic AMP: expanded role for an old signaling pathway. Science 271: 461-463. Jansson, C. C., Karp, M., Oker-Blom, C., Näsman, J., Savola, J. M., and Åkerman, K. E. (1995) Two human alpha 2-adrenoceptor subtypes alpha 2A-C10 and alpha 2B-C2 expressed in Sf9 cells couple to transduction pathway resulting in opposite effects on cAMP production. Eur. J. Pharmacol. 290: 75-83. Jansson, C. C., Kukkonen, J. P., Nasman, J., Huifang, G., Wurster, S., Virtanen, R., Savola, J. M., Cockcroft, V., and Akerman, K. E. (1998) Protean agonism at alpha2A- adrenoceptors. Mol. Pharmacol. 53: 963-968. Jansson, C. C., Marjamäki, A., Luomala, K., Savola, J. M., Scheinin, M., and Åkerman, K. E. (1994) Coupling of human alpha 2-adrenoceptor subtypes to regulation of cAMP production in transfected S115 cells. Eur. J. Pharmacol. 266: 165-174. Jewell-Motz, E. A., Donnelly, E. T., Eason, M. G., and Liggett, S. B. (1997) Role of the amino terminus of the third intracellular loop in agonist-promoted downregulation of the alpha2A-adrenergic receptor. Biochemistry 36: 8858-8863. Jinsi, A., Paradise, J., and Deth, R. C. (1996) A tyrosine kinase regulates alpha-adrenoceptor- stimulated contraction and activation in the rat aorta. Eur. J. Pharmacol. 302: 183-190.

55 Jones, S. B., Halenda, S. P., and Bylund, D. B. (1991) Alpha 2-adrenergic receptor stimulation of phospholipase A2 and of adenylate cyclase in transfected Chinese hamster ovary cells is mediated by different mechanisms. Mol. Pharmacol. 39: 239-245. Kable, J. W., Murrin, L. C., and Bylund, D. B. (2000) In vivo gene modification elucidates subtype-specific functions of alpha(2)-adrenergic receptors. J. Pharmacol. Exp. Ther. 293: 1-7. Kahn, R. A., and Gilman, A. G. (1984) Purification of a protein required for ADP- ribosylation of the stimulatory regulatory component of adenylate cyclase by cholera toxin. J. Biol. Chem. 259: 6228-6234. Katada, T., and Ui, M. (1982) ADP ribosylation of the specific membrane protein of C6 cells by islet-activating protein associated with modification of adenylate cyclase activity. J. Biol. Chem. 257: 7210-7216. Katz, A., Wu, D., and Simon, M. I. (1992) Subunits beta gamma of heterotrimeric G protein activate beta 2 isoform of phospholipase C. Nature 360: 686-689. Keularts, I. M., van Gorp, R. M., Feijge, M. A., Vuist, W. M., and Heemskerk, J. W. (2000) alpha(2a)-adrenergic receptor stimulation potentiates calcium release in platelets by modulating cAmp levels. J. Biol. Chem. 275: 1763-1772. Kobilka, B. K., Kobilka, T. S., Daniel, K., Regan, J. W., Caron, M. G., and Lefkowitz, R. J. (1988) Chimeric alpha 2-,beta 2-adrenergic receptors: delineation of domains involved in effector coupling and ligand binding specificity. Science 240: 1310-1316. Kobilka, B. K., Matsui, H., Kobilka, T. S., Yang-Feng, T. L., Francke, U., Caron, M. G., Lefkowitz, R. J., and Regan, J. W. (1987) Cloning, sequencing, and expression of the gene coding for the human platelet alpha 2-adrenergic receptor. Science 238: 650-656. Korswagen, H. C., van der Linden, A. M., and Plasterk, R. H. (1998) G protein hyperactivation of the Caenorhabditis elegans adenylyl cyclase SGS-1 induces neuronal degeneration. EMBO J. 17: 5059-5065. Kozasa, T., and Gilman, A. G. (1995) Purification of recombinant G proteins from Sf9 cells by hexahistidine tagging of associated subunits. Characterization of alpha 12 and inhibition of adenylyl cyclase by alpha z. J. Biol. Chem. 270: 1734-1741. Kozasa, T., Hepler, J. R., Smrcka, A. V., Simon, M. I., Rhee, S. G., Sternweis, P. C., and Gilman, A. G. (1993) Purification and characterization of recombinant G16 alpha from Sf9 cells: activation of purified phospholipase C isozymes by G-protein alpha subunits. Proc. Natl. Acad. Sci. U. S. A. 90: 9176-9180. Kribben, A., Herget-Rosenthal, S., Lange, B., Erdbrugger, W., Philipp, T., and Michel, M. C. (1997) Alpha2-adrenoceptors in opossum kidney cells couple to stimulation of mitogen- activated protein kinase independently of adenylyl cyclase inhibition. Naunyn Schmiedebergs Arch. Pharmacol. 356: 225-232. Krishna, G., Weiss, B., and Brodie, B. B. (1968) A simple, sensitive method for the assay of adenyl cyclase. J. Pharmacol. Exp. Ther. 163: 379-385. Krupinski, J., Coussen, F., Bakalyar, H. A., Tang, W. J., Feinstein, P. G., Orth, K., Slaughter, C., Reed, R. R., and Gilman, A. G. (1989) Adenylyl cyclase amino acid sequence: possible channel- or transporter-like structure. Science 244: 1558-1564. Kukkonen, J. P., Huifang, G., Jansson, C. C., Wurster, S., Cockcroft, V., Savola, J. M., and Åkerman, K. E. (1997) Different apparent modes of inhibition of alpha2A-adrenoceptor by alpha2-adrenoceptor antagonists. Eur. J. Pharmacol. 335: 99-105.

56 Kukkonen, J. P., Näsman, J., Ojala, P., Oker-Blom, C., and Åkerman, K. E. (1996) Functional properties of muscarinic receptor subtypes Hm1, Hm3 and Hm5 expressed in Sf9 cells using the baculovirus expression system. J. Pharmacol. Exp. Ther. 279: 593- 601. Kukkonen, J. P., Näsman, J., and Åkerman, K. E. O. (2001) Modelling of promiscuous receptor-Gi/Gs-protein coupling and effector response. Trends Pharmacol. Sci. 22: 617- 623. Kukkonen, J. P., Renvaktar, A., Shariatmadari, R., and Åkerman, K. E. (1998) Ligand- and subtype-selective coupling of human alpha-2 adrenoceptors to Ca++ elevation in Chinese hamster ovary cells. J. Pharmacol. Exp. Ther. 287: 667-671. Lafontan, M., and Berlan, M. (1993) Fat cell adrenergic receptors and the control of white and brown fat cell function. J. Lipid Res. 34: 1057-1091. Langer, S. Z. (1974) Presynaptic regulation of catecholamine release. Biochem. Pharmacol. 23: 1793-1800. Langer, S. Z., and Hicks, P. E. (1984) Alpha-adrenoreceptor subtypes in blood vessels: physiology and pharmacology. J. Cardiovasc. Pharmacol. 6: S547-558. Langley, J. N. (1905) On the reaction of cells and of nerv-endings to certain poisons, chiefly as regards the reaction of striated muscle to nicotine and curari. J. Physiol. (Lond.) 33: 374-413. Lanier, S. M., Downing, S., Duzic, E., and Homcy, C. J. (1991) Isolation of rat genomic clones encoding subtypes of the alpha 2-adrenergic receptor. Identification of a unique receptor subtype. J. Biol. Chem. 266: 10470-10478. Levin, L. R., Han, P. L., Hwang, P. M., Feinstein, P. G., Davis, R. L., and Reed, R. R. (1992) The Drosophila learning and memory gene rutabaga encodes a Ca2+/Calmodulin- responsive adenylyl cyclase. Cell 68: 479-489. Link, R., Daunt, D., Barsh, G., Chruscinski, A., and Kobilka, B. (1992) Cloning of two mouse genes encoding alpha 2-adrenergic receptor subtypes and identification of a single amino acid in the mouse alpha 2-C10 homolog responsible for an interspecies variation in antagonist binding. Mol. Pharmacol. 42: 16-27. Lipscombe, D., Kongsamut, S., and Tsien, R. W. (1989) Alpha-adrenergic inhibition of sympathetic neurotransmitter release mediated by modulation of N-type calcium-channel gating. Nature 340: 639-642. Lipskaia, L., Djiane, A., Defer, N., and Hanoune, J. (1997) Different expression of adenylyl cyclase isoforms after retinoic acid induction of P19 teratocarcinoma cells. FEBS Lett. 415: 275-280. Lipskaia, L., Grepin, C., Defer, N., and Hanoune, J. (1998) Adenylyl cyclase activity and gene expression during mesodermal differentiation of the P19 embryonal carcinoma cells. J. Cell. Physiol. 176: 50-56. Logothetis, D. E., Kurachi, Y., Galper, J., Neer, E. J., and Clapham, D. E. (1987) The beta gamma subunits of GTP-binding proteins activate the muscarinic K+ channel in heart. Nature 325: 321-326. Lomasney, J. W., Lorenz, W., Allen, L. F., King, K., Regan, J. W., Yang-Feng, T. L., Caron, M. G., and Lefkowitz, R. J. (1990) Expansion of the alpha 2-adrenergic receptor family: cloning and characterization of a human alpha 2-adrenergic receptor subtype, the gene for which is located on chromosome 2. Proc. Natl. Acad. Sci. U. S. A. 87: 5094-5098. Luo, X., Zeng, W., Xu, X., Popov, S., Davignon, I., Wilkie, T. M., Mumby, S. M., and Muallem, S. (1999) Alternate coupling of receptors to Gs and Gi in pancreatic and submandibular gland cells. J. Biol. Chem. 274: 17684-17690.

57 Marinissen, M. J., and Gutkind, J. S. (2001) G-protein-coupled receptors and signaling networks: emerging paradigms. Trends Pharmacol. Sci. 22: 368-376. Marjamaki, A., Sato, M., Bouet-Alard, R., Yang, Q., Limon-Boulez, I., Legrand, C., and Lanier, S. M. (1997) Factors determining the specificity of signal transduction by guanine nucleotide-binding protein-coupled receptors. Integration of stimulatory and inhibitory input to the effector adenylyl cyclase. J. Biol. Chem. 272: 16466-16473. May, D. C., Ross, E. M., Gilman, A. G., and Smigel, M. D. (1985) Reconstitution of catecholamine-stimulated adenylate cyclase activity using three purified proteins. J. Biol. Chem. 260: 15829-15833. Menco, B. P., Bruch, R. C., Dau, B., and Danho, W. (1992) Ultrastructural localization of olfactory transduction components: the G Golf alpha and type III adenylyl cyclase. Neuron 8: 441-453. Mhaouty, S., Cohen-Tannoudji, J., Bouet-Alard, R., Limon-Boulez, I., Maltier, J. P., and Legrand, C. (1995) Characteristics of the alpha 2/beta 2-adrenergic receptor-coupled adenylyl cyclase system in rat myometrium during pregnancy. J. Biol. Chem. 270: 11012- 11016. Michel, M. C., Brass, L. F., Williams, A., Bokoch, G. M., LaMorte, V. J., and Motulsky, H. J. (1989) Alpha 2-adrenergic receptor stimulation mobilizes intracellular Ca2+ in human erythroleukemia cells. J. Biol. Chem. 264: 4986-4991. Michell, R. H. (1975) Inositol phospholipids and function. Biochim. Biophys. Acta 415: 81-147. Milligan, G. (1988) Techniques used in the identification and analysis of function of pertussis toxin-sensitive guanine nucleotide binding proteins. Biochem. J. 255: 1-13. Milligan, G., Marshall, F., and Rees, S. (1996) G16 as a universal G protein adapter: implications for agonist screening strategies. Trends Pharmacol. Sci. 17: 235-237. Mills, A., Allet, B., Bernard, A., Chabert, C., Brandt, E., Cavegn, C., Chollet, A., and Kawashima, E. (1993) Expression and characterization of human D4 dopamine receptors in baculovirus-infected insect cells. FEBS Lett. 320: 130-134. Mons, N., and Cooper, D. M. (1995) Adenylate cyclases: critical foci in neuronal signaling. Trends Neurosci. 18: 536-542. Mouillac, B., Caron, M., Bonin, H., Dennis, M., and Bouvier, M. (1992) Agonist-modulated of beta 2-adrenergic receptor in Sf9 cells. J. Biol. Chem. 267: 21733- 21737. Mulheron, J. G., Casanas, S. J., Arthur, J. M., Garnovskaya, M. N., Gettys, T. W., and Raymond, J. R. (1994) Human 5-HT1A receptor expressed in insect cells activates endogenous G(o)-like G protein(s). J. Biol. Chem. 269: 12954-12962. Murphy, T. J., and Bylund, D. B. (1988) Characterization of alpha-2 adrenergic receptors in the OK cell, an opossum kidney cell line. J. Pharmacol. Exp. Ther. 244: 571-578. Nathans, J., and Hogness, D. S. (1983) Isolation, sequence analysis, and intron-exon arrangement of the gene encoding bovine rhodopsin. Cell 34: 807-814. Nathanson, J. A., and Greengard, P. (1973) Octopamine-sensitive adenylate cyclase. Evidence for a biological role of octopamine in nervous tissue. Science 180: 308-310. Neer, E. J., Lok, J. M., and Wolf, L. G. (1984) Purification and properties of the inhibitory guanine nucleotide regulatory unit of brain adenylate cyclase. J. Biol. Chem. 259: 14222- 14229. Ng, G. Y., George, S. R., Zastawny, R. L., Caron, M., Bouvier, M., Dennis, M., and O’Dowd, B. F. (1993) Human serotonin1B receptor expression in Sf9 cells: phosphorylation, palmitoylation, and adenylyl cyclase inhibition. Biochemistry 32: 11727-11733.

58 Nicholas, A. P., Hokfelt, T., and Pieribone, V. A. (1996) The distribution and significance of CNS adrenoceptors examined with in situ hybridization. Trends Pharmacol. Sci. 17: 245- 255. Nishizuka, Y. (1984) The role of protein kinase C in cell surface signal transduction and tumour promotion. Nature 308: 693-698. Northup, J. K., Sternweis, P. C., Smigel, M. D., Schleifer, L. S., Ross, E. M., and Gilman, A. G. (1980) Purification of the regulatory component of adenylate cyclase. Proc. Natl. Acad. Sci. U. S. A. 77: 6516-6520. O’Connor, S. E., Dainty, I. A., and Leff, P. (1991) Further subclassification of ATP receptors based on agonist studies. Trends Pharmacol. Sci. 12: 137-141. Offermanns, S., Toombs, C. F., Hu, Y. H., and Simon, M. I. (1997) Defective platelet activation in G alpha(q)-deficient mice. Nature 389: 183-186. Oker-Blom, C., Jansson, C., Karp, M., Lindqvist, C., Savola, J. M., Vlak, J., and Åkerman, K. (1993) Functional analysis of the human alpha 2C-C4 adrenergic receptor in insect cells expressed by a luciferase-based baculovirus vector. Biochim. Biophys. Acta 1176: 269- 275. Orr, N., Orr, G. L., and Hollingworth, R. M. (1992) The Sf9 cell line as a model for studying insect octopamine-receptors. Insect Biochem. Mol. Biol. 22: 591-597. Ostrom, R. S., Gregorian, C., and Insel, P. A. (2000) Cellular release of and response to ATP as key determinants of the set-point of signal transduction pathways. J. Biol. Chem. 275: 11735-11739. Ovchinnikov, Y. (1982) Rhodopsin and bacteriorhodopsin: structure-function relationships. FEBS Lett. 148: 179-191. Parker, E. M., Kameyama, K., Higashijima, T., and Ross, E. M. (1991) Reconstitutively active G protein-coupled receptors purified from baculovirus-infected insect cells. J. Biol. Chem. 266: 519-527. Pepperl, D. J., and Regan, J. W. (1993) Selective coupling of alpha 2-adrenergic receptor subtypes to cyclic AMP-dependent reporter gene expression in transiently transfected JEG-3 cells. Mol. Pharmacol. 44: 802-809. Pepperl, D. J., and Regan, J. W. (1994) Adrenergic Receptors. In Handbook of Receptors and Channels (Peroutka, S. J., ed), pp. 45-78, CRC Press, Inc., Boca Raton. Pfeuffer, E., Dreher, R. M., Metzger, H., and Pfeuffer, T. (1985a) Catalytic unit of adenlyate cyclase: purification and identification by affinity crosslinking. Proc. Natl. Acad. Sci. U. S. A. 82: 3086-3090. Pfeuffer, E., Mollner, S., and Pfeuffer, T. (1985b) Adenylate cyclase from bovine brain cortex: purification and characterization of the catalytic unit. EMBO J. 4: 3675-3679. Pieroni, J. P., Harry, A., Chen, J., Jacobowitz, O., Magnusson, R. P., and Iyengar, R. (1995) Distinct characteristics of the basal activities of adenylyl cyclases 2 and 6. J. Biol. Chem. 270: 21368-21373. Pohjanoksa, K., Jansson, C. C., Luomala, K., Marjamaki, A., Savola, J. M., and Scheinin, M. (1997) Alpha2-adrenoceptor regulation of adenylyl cyclase in CHO cells: dependence on receptor density, receptor subtype and current activity of adenylyl cyclase. Eur. J. Pharmacol. 335: 53-63. Quehenberger, O., Prossnitz, E. R., Cochrane, C. G., and Ye, R. D. (1992) Absence of G(i) proteins in the Sf9 insect cell. Characterization of the uncoupled recombinant N-. J. Biol. Chem. 267: 19757-19760. Ralevic, V., and Burnstock, G. (1998) Receptors for purines and pyrimidines. Pharmacol. Rev. 50: 413-492.

59 Rasmussen, H. (1970) Cell communication, calcium ion, and cyclic . Science 170: 404-412. Rebois, R. V., Warner, D. R., and Basi, N. S. (1997) Does subunit dissociation necessarily accompany the activation of all heterotrimeric G proteins? Cell. Signal. 9: 141-151. Regan, J. W., Barden, N., Lefkowitz, R. J., Caron, M. G., DeMarinis, R. M., Krog, A. J., Holden, K. G., Matthews, W. D., and Hieble, J. P. (1982) Affinity chromatography of human platelet alpha 2-adrenergic receptors. Proc. Natl. Acad. Sci. U. S. A. 79: 7223- 7227. Regan, J. W., Kobilka, T. S., Yang-Feng, T. L., Caron, M. G., Lefkowitz, R. J., and Kobilka, B. K. (1988) Cloning and expression of a human kidney cDNA for an alpha 2-adrenergic receptor subtype. Proc. Natl. Acad. Sci. U. S. A. 85: 6301-6305. Reilander, H., Boege, F., Vasudevan, S., Maul, G., Hekman, M., Dees, C., Hampe, W., Helmreich, E. J., and Michel, H. (1991) Purification and functional characterization of the human beta 2-adrenergic receptor produced in baculovirus-infected insect cells. FEBS Lett. 282: 441-444. Rhee, S. G. (2001) Regulation of phosphoinositide-specific phospholipase C. Annu. Rev. Biochem. 70: 281-312. Richardson, R. M., and Hosey, M. M. (1992) Agonist-induced phosphorylation and desensitization of human m2 muscarinic cholinergic receptors in Sf9 insect cells. J. Biol. Chem. 267: 22249-22255. Ringer, S. (1883) A further contribution regarding the influence of the different constitutents of the blood on the contraction of the heart. J. Physiol. (Lond.) 4: 29-42. Robb, S., Cheek, T. R., Hannan, F. L., Hall, L. M., Midgley, J. M., and Evans, P. D. (1994) Agonist-specific coupling of a cloned Drosophila octopamine/tyramine receptor to multiple second messenger systems. EMBO J. 13: 1325-1330. Rodbell, M., Birnbaumer, L., Pohl, S. L., and Krans, H. M. (1971) The glucagon-sensitive adenyl cyclase system in plasma membranes of rat liver. V. An obligatory role of guanylnucleotides in glucagon action. J. Biol. Chem. 246: 1877-1882. Roeder, T. (1992) A new octopamine receptor class in locust nervous tissue, the octopamine 3 (OA3) receptor. Life Sci. 50: 21-28. Roeder, T. (1999) Octopamine in invertebrates. Prog. Neurobiol. 59: 533-561. Rudling, J. E., Kennedy, K., and Evans, P. D. (1999) The effect of site-directed mutagenesis of two transmembrane serine residues on agonist-specific coupling of a cloned human alpha2A-adrenoceptor to adenylyl cyclase. Br. J. Pharmacol. 127: 877-886. Rudling, J. E., Richardson, J., and Evans, P. D. (2000) A comparison of agonist-specific coupling of cloned human alpha(2)-adrenoceptor subtypes. Br J Pharmacol 131: 933- 941. Ruffolo, R. R., Nichols, A. J., Stadel, J. M., and Hieble, J. P. (1993) Pharmacologic and therapeutic applications of alpha 2-adrenoceptor subtypes. Annu. Rev. Pharmacol. Toxicol. 33243-33279. Runnels, L. W., and Scarlata, S. F. (1999) Determination of the affinities between heterotrimeric G protein subunits and their phospholipase C-beta effectors. Biochemistry 38: 1488-1496. Saavedra, J. M. (1974) Enzymatic-isotopic method for octopamine at the picogram level. Anal. Biochem. 59: 628-633. Salm, A. K., and McCarthy, K. D. (1990) Norepinephrine-evoked calcium transients in cultured cerebral type 1 astroglia. Glia 3: 529-538.

60 Sattin, A., and Rall, T. W. (1970) The effect of adenosine and adenine nucleotides on the cyclic adenosine 3’, 5’- content of guinea pig cerebral cortex slices. Mol. Pharmacol. 6: 13-23. Sautel, M., and Milligan, G. (1998) Loss of activation of Gs but not Gi following expression of an alpha2A-adrenoceptor-Gi1alpha fusion protein. FEBS Lett. 436: 46-50. Savarese, T. M., and Fraser, C. M. (1992) In vitro mutagenesis and the search for structure- function relationships among G protein-coupled receptors. Biochem. J. 283: 1-19. Schaak, S., Cayla, C., Blaise, R., Quinchon, F., and Paris, H. (1997) HepG2 and SK-N-MC: two human models to study alpha-2 adrenergic receptors of the alpha-2C subtype. J. Pharmacol. Exp. Ther. 281: 983-991. Schmidt, C. J., Garen-Fazio, S., Chow, Y. K., and Neer, E. J. (1989) Neuronal expression of a newly identified Drosophila melanogaster G protein alpha 0 subunit. Cell. Regul. 1: 125- 134. Schmidt, C. J., Thomas, T. C., Levine, M. A., and Neer, E. J. (1992) Specificity of G protein beta and gamma subunit interactions. J. Biol. Chem. 267: 13807-13810. Seamon, K. B., Padgett, W., and Daly, J. W. (1981) Forskolin: unique diterpene activator of adenylate cyclase in membranes and in intact cells. Proc. Natl. Acad. Sci. U. S. A. 78: 3363-3367. Shapiro, R. A., Wakimoto, B. T., Subers, E. M., and Nathanson, N. M. (1989) Characterization and functional expression in mammalian cells of genomic and cDNA clones encoding a Drosophila muscarinic acetylcholine receptor. Proc. Natl. Acad. Sci. U. S. A. 86: 9039-9043. Shaw, G. (1995) The pleckstrin homology domain: an intriguing multifunctional protein module. BioEssays 18: 35-46. Shorr, R. G., Lefkowitz, R. J., and Caron, M. G. (1981) Purification of the beta-adrenergic receptor. Identification of the hormone binding subunit. J. Biol. Chem. 256: 5820-5826. Shorr, R. G., Strohsacker, M. W., Lavin, T. N., Lefkowitz, R. J., and Caron, M. G. (1982) The beta 1-adrenergic receptor of the turkey erythrocyte. Molecular heterogeneity revealed by purification and photoaffinity labeling. J. Biol. Chem. 257: 12341-12350. Singewald, N., and Philippu, A. (1996) Involvement of biogenic amines and amino acids in the central regulation of cardiovascular homeostasis. Trends Pharmacol. Sci. 17: 356-363. Smit, M. J., Verzijl, D., and Iyengar, R. (1998) Identity of adenylyl cyclase isoform determines the rate of cell cycle progression in NIH 3T3 cells. Proc. Natl. Acad. Sci. U. S. A. 95: 15084-15089. Spina, D., Landells, L. J., and Page, C. P. (1998) The role of phosphodiesterase enzymes in allergy and asthma. Adv. Pharmacol. 44: 33-89. Starke, K. (1981) Alpha-adrenoceptor subclassification. Rev. Physiol. Biochem. Pharmacol. 88: 199-236. Sternweis, P. C. (1986) The purified alpha subunits of Go and Gi from bovine brain require beta gamma for association with phospholipid vesicles. J. Biol. Chem. 261: 631-637. Sternweis, P. C. (1994) The active role of beta gamma in signal transduction. Curr. Opin. Cell Biol. 6: 198-203. Sternweis, P. C., and Robishaw, J. D. (1984) Isolation of two proteins with high affinity for guanine nucleotides from membranes of bovine brain. J. Biol. Chem. 259: 13806-13813. Streb, H., Irvine, R. F., Berridge, M. J., and Schulz, I. (1983) Release of Ca2+ from a nonmitochondrial intracellular store in pancreatic acinar cells by inositol-1,4,5- trisphosphate. Nature 306: 67-69.

61 Sunahara, R. K., Dessauer, C. W., and Gilman, A. G. (1996) Complexity and diversity of mammalian adenylyl cyclases. Annu. Rev. Pharmacol. Toxicol. 36: 461-480. Surprenant, A., Horstman, D. A., Akbarali, H., and Limbird, L. E. (1992) A point mutation of the alpha 2-adrenoceptor that blocks coupling to potassium but not calcium currents. Science 257: 977-980. Suryanarayana, S., von Zastrow, M., and Kobilka, B. K. (1992) Identification of intramolecular interactions in adrenergic receptors. J. Biol. Chem. 267: 21991-21994. Sutherland, E. W. (1972) Studies on the mechanism of hormone action. Science 177: 401- 408. Sweatt, J. D., Connolly, T. M., Cragoe, E. J., and Limbird, L. E. (1986) Evidence that Na+/H+ exchange regulates receptor-mediated phospholipase A2 activation in human platelets. J. Biol. Chem. 261: 8667-8673. Takai, Y., Kishimoto, A., Kikkawa, U., Mori, T., and Nishizuka, Y. (1979) Unsaturated diacylglycerol as a possible messenger for the activation of calcium-activated, phospholipid-dependent protein kinase system. Biochem. Biophys. Res. Commun. 91: 1218-1224. Tang, W. J., and Hurley, J. H. (1998) Catalytic mechanism and regulation of mammalian adenylyl cyclases. Mol. Pharmacol. 54: 231-240. Tang, W. J., Krupinski, J., and Gilman, A. G. (1991) Expression and characterization of calmodulin-activated (type I) adenylylcyclase. J. Biol. Chem. 266: 8595-8603. Thomas, D. P. (1967) Effect of catecholamines on platelet aggregation caused by thrombin. Nature 215: 298-299. Trudeau, L. E., Emery, D. G., and Haydon, P. G. (1996) Direct modulation of the secretory machinery underlies PKA-dependent synaptic facilitation in hippocampal neurons. Neuron 17: 789-797. Uhlen, S., Dambrova, M., Näsman, J., Schioth, H. B., Gu, Y., Wikberg-Matsson, A., and Wikberg, J. E. (1998) [3H]RS79948-197 binding to human, rat, guinea pig and pig alpha2A-, alpha2B- and alpha2C-adrenoceptors. Comparison with MK912, RX821002, rauwolscine and yohimbine. Eur. J. Pharmacol. 343: 93-101. Ui, M. (1984) Islet-activating protein, pertussis toxin: a probe for function of the inhibitory guanine nucleotide regulatory component of adenylate cyclase. Trends Pharmacol. Sci. 5: 277-279. Ui, M., Okajima, F., Katada, T., and Murayama, T. (1988) Roles of GTP regulatory proteins, the substrates of islet-activating protein, in receptor-mediated adenylate cyclase inhibition, phospholipase C activation, and cell proliferation. Adv. Second Messenger Phosphoprotein Res. 21: 39-45. Ukena, D., Schudt, C., and Sybrecht, G. W. (1993) Adenosine receptor-blocking xanthines as inhibitors of phosphodiesterase isozymes. Biochem. Pharmacol. 45: 847-851. Ullrich, S., and Wollheim, C. B. (1984) Islet cyclic AMP levels are not lowered during alpha 2-adrenergic inhibition of insulin release. J. Biol. Chem. 259: 4111-4115. Vachon, V., Paradis, M. J., Marsolais, M., Schwartz, J. L., and Laprade, R. (1995) Endogenous K+/H+ exchange activity in the Sf9 insect cell line. Biochemistry 34: 15157- 15164. Walsh, D. A., and van Patten, S. M. (1994) Multiple pathway signal transduction by the cAMP-dependent protein kinase. FASEB J. 8: 1227-1236. van Calker, D., Muller, M., and Hamprecht, B. (1978) Adenosine inhibits the accumulation of cyclic AMP in cultured brain cells. Nature 276: 839-841.

62 van Calker, D., Muller, M., and Hamprecht, B. (1979) Adenosine regulates via two different types of receptors, the accumulation of cyclic AMP in cultured brain cells. J. Neurochem. 33: 999-1005. Wang, C. D., Buck, M. A., and Fraser, C. M. (1991) Site-directed mutagenesis of alpha 2A- adrenergic receptors: identification of amino acids involved in ligand binding and receptor activation by agonists. Mol. Pharmacol. 40: 168-179. Wang, D. S., Shaw, R., Winkelmann, J. C., and Shaw, G. (1994) Binding of PH domains of beta-adrenergic receptor kinase and beta-spectrin to WD40/beta-transducin repeat containing regions of the beta-subunit of trimeric G-proteins. Biochem. Biophys. Res. Commun. 203: 29-35. Wang, T., Pentyala, S., Rebecchi, M. J., and Scarlata, S. (1999) Differential association of the pleckstrin homology domains of C-beta 1, C-beta 2, and C-delta 1 with lipid bilayers and the beta gamma subunits of heterotrimeric G proteins. Biochemistry 38: 1517-1524. Varga, E. V., Stropova, D., Rubenzik, M., Wang, M., Landsman, R. S., Roeske, W. R., and Yamamura, H. I. (1998) Identification of adenylyl cyclase isoenzymes in CHO and B82 cells. Eur. J. Pharmacol. 348: R1-2. Vasudevan, S., Premkumar, L., Stowe, S., Gage, P. W., Reilander, H., and Chung, S. H. (1992) Muscarinic acetylcholine receptor produced in recombinant baculovirus infected Sf9 insect cells couples with endogenous G-proteins to activate ion channels. FEBS Lett. 311: 7-11. Vasudevan, S., Reilander, H., Maul, G., and Michel, H. (1991) Expression and cell membrane localization of rat M3 muscarinic acetylcholine receptor produced in Sf9 insect cells using the baculovirus system. FEBS Lett. 283: 52-56. Watters, D. J., and Parsons, P. G. (1999) Critical targets of protein kinase C in differentiation of tumour cells. Biochem. Pharmacol. 58: 383-388. Vaughn, J. L., Goodwin, R. H., Tompkins, G. J., and McCawley, P. (1977) The establishment of two cell lines from the insect Spodoptera frugiperda (Lepidoptera; Noctuidae). In Vitro 13: 213-217. Wayman, G. A., Impey, S., Wu, Z., Kindsvogel, W., Prichard, L., and Storm, D. R. (1994) Synergistic activation of the type I adenylyl cyclase by Ca2+ and Gs-coupled receptors in vivo. J. Biol. Chem. 269: 25400-25405. Wess, J. (1997) G-protein-coupled receptors: molecular mechanisms involved in receptor activation and selectivity of G-protein recognition. FASEB J. 11: 346-354. Wess, J. (1998) Molecular basis of receptor/G-protein-coupling selectivity. Pharmacol. Ther. 80: 231-264. Wieland, T., and Jakobs, K. H. (1994) Measurement of receptor-stimulated guanosine 5’-O- (gamma-thio)triphosphate binding by G proteins. Methods Enzymol. 237: 3-13. Xia, Z., and Storm, D. R. (1997) Calmodulin-regulated adenylyl cyclases and neuromodulation. Curr. Opin. Neurobiol. 7: 391-396. Yan, K., Kalyanaraman, V., and Gautam, N. (1996) Differential ability to form the G protein betagamma complex among members of the beta and gamma subunit families. J. Biol. Chem. 271: 7141-7146. Yarden, Y., Rodriguez, H., Wong, S. K., Brandt, D. R., May, D. C., Burnier, J., Harkins, R. N., Chen, E. Y., Ramachandran, J., Ullrich, A., and et al. (1986) The avian beta- adrenergic receptor: primary structure and membrane topology. Proc. Natl. Acad. Sci. U. S. A. 83: 6795-6799.

63 Yoshimasa, T., Sibley, D. R., Bouvier, M., Lefkowitz, R. J., and Caron, M. G. (1987) Cross- talk between cellular signalling pathways suggested by phorbol-ester-induced adenylate cyclase phosphorylation. Nature 327: 67-70. Zamponi, G. W., Bourinet, E., Nelson, D., Nargeot, J., and Snutch, T. P. (1997) Crosstalk between G proteins and protein kinase C mediated by the calcium channel alpha1 subunit. Nature 385: 442-446. Zimmermann, G., and Taussig, R. (1996) Protein kinase C alters the responsiveness of adenylyl cyclases to G protein alpha and betagamma subunits. J. Biol. Chem. 271: 27161- 27166. Åkerman, K. E. O., Holmberg, C. I., Gee, H.-F., Renvaktar, A., Soini, S., Kukkonen, J. P., Näsman, J., Enkvist, K., Courtney, M. J., and Jansson, C. (1996) Alpha2 adrenergic receptor activation and intracellular Ca2+. In Alpha2-adrenergic receptors: Structure, Function and Therapeutic Implications (Lanier, S., and Limbird, L., eds), pp. 85-93, Harwood Academic Publishers, Amsterdam.

644