Download as pdf or txt
Download as pdf or txt
You are on page 1of 5

of a number of cell types (Post and

Brown 1996). Theoretically, all pro-


Constitutively Signaling G-Protein- teins that comprise the signal trans-
duction pathways of GPCRs are po-
Coupled Receptors and Human tential targets for mutational events
Disease that can lead to constitutive (agonist-
independent) signaling and conse-
Leandros Arvanitakis, Elizabeth Geras-Raaka and quently disease. Indeed, mutations of a
number of G proteins have been shown
Marvin C. Gershengorn to cause disease in humans (Spiegel
1995). For example, mutations that
cause constitutive activation of the ␣
Dysregulation of G-protein-coupled receptor (GPCR) function has subunit of the stimulatory G protein,
been shown to be associated with a growing number of human dis- Gs, lead to increased cAMP formation
eases. In some diseases, mutation of an endogenous GPCR causes associated with pituitary tumors in
patients with acromegaly, hyperfunc-
the receptor to lose the ability to bind agonist or signal (‘loss of func-
tional thyroid nodules and the
tion’ mutation), whereas another mutation causes the receptor to be McCune–Albright syndrome. Simi-
in an active state in the absence of agonist (‘gain of function’ mu- larly, activating mutations of the ras
tation), leading to ‘constitutive signaling activity’. A number of consti- protein dysregulate the cell cycle and
tutively active GPCRs are tumorigenic in vitro and in animal models, are involved in the pathogenesis of
and cause syndromes of hyperfunction and/or tumors in humans. many human cancers. In this review,
we will limit our discussion to consti-
The recent characterization of a constitutively active GPCR in the
tutively active GPCRs and highlight
genome of a disease-associated, human herpesvirus provides a poten- those that have been found to be as-
tial novel mechanism for viral tumorigenesis. sociated with human disease.

The seven-transmembrane-helix, gua- H2N


nine nucleotide-binding (G) protein-
coupled receptors (GPCRs) (Figure 1)
represent the largest family of signal- Extracellular
transducing molecules known (Strader
et al. 1994). There are three subfamilies
of GPCRs: rhodopsin/␤2-adrenergic
receptors, calcitonin/parathyroid hor-
mone/parathyroid hormone-related
peptide receptors and metabotropic Transmembrane
glutamate/calcium-sensing receptors.
GPCRs convey signals for extracellu-
lar regulatory molecules as diverse
as hormones, neurotransmitters and
growth factors, as well as for calcium Intracellular
ions and photons. By coupling to G
proteins and activating effectors such HOOC
as adenylyl cyclase, ion channels or
phospholipase C that lead to in- Figure 1. Putative two-dimensional topology of a GPCR. GPCRs are integral membrane
creases (or decreases) in intracellular proteins with extracellular, transmembrane and intracellular domains. The extracellular
mediator molecules such as cAMP, amino terminus and three extracellular loops are at the upper part of the figure. The
seven ␣ helices that span the cell-surface membrane are in the middle of the figure. The
calcium ions or inositol 1,4,5-trisphos-
three intracellular loops and the intracellular carboxyl terminus are at the lower part of
phate and 1,2-diacylglycerol, GPCRs the figure. In three dimensions (not shown), the seven helices form a helical bundle that
regulate the activity of every cell in approximates a cylinder, with the seventh helix close to and interacting with the first and
the body. GPCRs regulate proliferation second helices.

Leandros Arvanitakis is at the Molecular Virology Laboratory, Hellenic Pasteur Institute, 127 Vas. Sofias Avenue, Athens GR-115 21, Greece;
Elizabeth Geras-Raaka and Marvin C. Gershengorn are at the Division of Molecular Medicine, Department of Medicine, Cornell University
Medical College, 1300 York Avenue, New York, NY 10021, USA.

TEM Vol. 9, No. 1, 1998 © 1998, Elsevier Science Ltd, 1043-2760/98/$19.00. PII: S1043-2760(98)00007-1 27
• GPCR Signaling constitutive signaling activity (Table the use of these types of measure-
The changes in GPCR structure that 1). These include receptors from each ments constitutive activity has been
constitute receptor activation are not subfamily of GPCRs; for example, more readily demonstrated with
known. It is thought that changes in ␤2-adrenergic (Milano et al. 1994), GPCRs that couple to the adenylyl
the conformation of the receptor, in calcitonin (Cohen et al. 1997) and cyclase–cAMP signal transduction
particular, changes involving the rela- metabotropic glutamate (Prézeau pathway than to the phospholipase
tive positions of the seven putative ␣ et al. 1996) receptors. Moreover, in C–inositol 1,4,5-trisphosphate–1,2-
helices within the transmembrane some cases, for example mouse EP3 diacylglycerol cascade. Human calci-
bundle, lead to changes in the intra- prostaglandin receptors (Hasegawa tonin receptors, which couple to both
cellular loops, causing increased affin- et al. 1996), different isoforms pro- of these signal transduction path-
ity for coupling to a heterotrimeric duced by alternative RNA splicing ways (Nussenzveig et al. 1994), were
G protein and thence to G-protein exhibit different levels of agonist- shown to stimulate cyclic AMP for-
activation. Activation of G proteins, independent signaling activity. In the mation constitutively, but not inosi-
which are tethered to the inner surface majority of cases, constitutive activ- tol 1,4,5-trisphosphate formation
of the plasma membrane, is thought ity was shown with receptors studied (Cohen et al. 1997). Indeed, agonist-
to involve dissociation into two acti- in in vitro systems. It has become independent signaling activity of the
vated signaling molecules, namely, standard to show constitutive activity mouse thyrotropin-releasing hormone
the ␣ subunit bound to GTP and the by expressing GPCRs at various receptor could not be demonstrated
dimer of ␤␥ subunits, which are both levels (usually by transfection with by measuring second messenger for-
proximal mediators in signal trans- various amounts of plasmid encoding mation and relied on a sensitive re-
duction cascades. In most cases with cloned receptor complementary porter gene assay in which 1,2-diacyl-
native GPCRs, activation is initiated DNAs) in cells in culture and demon- glycerol activation of protein kinase
by agonist binding. Larger agonists strating that there is a direct relation- C leads to transcription of a firefly
bind primarily to the amino termini ship between the level of GPCR ex- luciferase reporter gene (Jinsi-Parimoo
and/or extracellular loops of GPCRs pression and basal signaling. Human and Gershengorn 1997). A clear
and smaller ligands bind within the ␤2-adrenergic receptors were found demonstration that constitutive sig-
GPCR transmembrane bundle. In to be constitutively active by showing naling by a native mammalian recep-
contrast, constitutively active GPCRs that basal adenylyl cyclase activity in tor may lead to changes in cell
achieve an active conformation with- membranes isolated from transfected function in vivo has been provided
out agonist binding. Cells have counter- cells correlated directly with the level for human ␤2-adrenergic receptors
regulatory mechanisms that attenuate of receptors (Samama et al. 1993) (Milano et al. 1994, Bond et al. 1995).
signaling by activated GPCRs. These and the basal level of cAMP in intact Transgenic mice overexpressing
mechanisms include acute desensit- cells (in the presence of cAMP phos- human ␤2-adrenergic receptors only
ization involving GPCR-specific pro- phodiesterase inhibitors) was found in their hearts exhibited enhanced
tein kinases and arrestins (homolo- to correlate directly with the level of myocardial function that was caused
gous desensitization) and second rat histamine H2 receptors (Smit et by the agonist-independent activity
messenger-activated protein kinases, al. 1996). It is noteworthy that with of these receptors.
such as protein kinases A and C
(heterologous desensitization). More
Table 1. Native mammalian GPCRs that exhibit constitutive activitya
chronic decreases in GPCR sensitiv-
ity may be caused by decreases in re-
Receptor Reference
ceptor number (downregulation) that
may be caused by decreased GPCR
Human ␤2-adrenergic Milano et al. (1994)
synthesis (decreased gene transcrip-
Rat B2 bradykinin Leeb-Lundberg et al. (1994)
tion or increased messenger RNA
Human calcitonin Cohen et al. (1997)
turnover) or enhanced GPCR degra-
Human CB1 cannabinoid Bouaboula et al. (1997)
dation secondary to increased recep-
Rat D1B dopamine Tiberi and Caron (1994)
tor internalization (by endocytosis
Rat metabotropic glutamate Prézeau et al. (1996)
or sequestration) and degradation
Rat histamine H2 Smit et al. (1996)
within lysosomes. These counter-
Rat 5-hydroxytryptamine2C Barker et al. (1994)
regulatory mechanisms, however, do
Mouse melanocortin Robbins et al. (1993)
not completely suppress constitutive
Rat ␦-opioid Costa et al. (1992)
signaling (Lefkowitz et al. 1993).
Mouse EP3 prostaglandin Hasegawa et al. (1996)
• Native Mammalian GPCRs Mouse thyrotropin-releasing hormone Jinsi-Parimoo and Gershengorn (1997)
A number of native mammalian aIf the same receptor from another species or another isoform of a listed receptor is constitutively active,

GPCRs have been shown to exhibit only one receptor is listed.

28 TEM Vol. 9, No. 1, 1998


An approach that is complemen- naturally or were constructed in the disorder (Kosugi et al. 1995). A
tary to that described above to show laboratory. Because the initial few constitutively activating mutation in
constitutive activity of receptors is constitutively active mutant GPCRs the first transmembrane domain of
the use of ligands that exhibit inverse were receptors with changes in the the receptor for parathyroid hor-
agonism (or negative antagonism) putative third intracellular loop or mone/parathyroid hormone-related
(Schutz and Freissmuth 1992). An sixth transmembrane helix, it was peptide has been described in a pa-
inverse agonist is a ligand that in- thought that these domains were es- tient with Jansen-type metaphyseal
hibits agonist-independent signaling pecially important sites for activating chondrodysplasia (Schipani et al.
activity (Costa et al. 1992). Inhibition mutations. It is now clear, however, 1995). A mutation in the second
of basal signaling by inverse agonists that mutations which affect many do- transmembrane domain of rhodopsin
provides evidence that the targeted mains in GPCRs can lead to agonist- leads to constitutive activation and
receptor is the specific cause of this independent activity. Several reviews causes congenital night blindness
activity. Ligands that display inverse have summarized many of the mu- (Rao et al. 1994), whereas a mutation
agonism have been identified for tations constructed in the laboratory, in the seventh transmembrane do-
a number of GPCRs including ␤2- and these will not be considered main of rhodopsin leads to persistent
adrenergic (Chidiac et al. 1994), further here [see, for example (Scheer activation of photoreceptor cells and
5-hydroxytryptamine2C (Labrecque and Cotecchia 1997)]. An important is the probable cause of retinal cell
et al. 1995), opioid (Costa et al. 1992), consequence of the expanding aware- degeneration in retinitis pigmentosa
parathyroid hormone/parathyroid ness of constitutive signaling activity (Robinson et al. 1992). The receptor
hormone-related peptide (Gardella et al. of GPCRs is the realization that a for thyroid-stimulating hormone
1996), B2 bradykinin (Leeb-Lundberg number of human diseases are caused (TSH) has been found to be mutated
et al. 1994) and thyrotropin-releasing by mutations in GPCRs that lead to at several sites that lead to constitu-
hormone (Heinflink et al. 1995) re- constitutive signaling (Table 2) (Spiegel tive activation (Van Sande et al.
ceptors; these ligands were used to 1995). Receptors from each subfamily 1995). When activating mutations
confirm the constitutive signaling of GPCRs have been found to cause of the TSH receptor occur in the
activity of these native receptors. human disease in this way. These germline, they cause familial non-
In conclusion, it is now evident mutations may arise in cells in the autoimmune hyperthyroidism, in
that a number of native mammalian germline or in somatic cells and are which there is symmetric enlarge-
receptors exhibit constitutive signal- dominant. A mutation that allows ment of the thyroid gland, whereas
ing activity. However, as these demon- activation at lower calcium levels was somatic mutations lead to hyperfunc-
strations have been made primarily identified in the extracellular amino- tioning (‘toxic’) thyroid adenomas
in in vitro systems in which there terminal domain of the calcium-sens- and the remainder of the thyroid
is marked overexpression of GPCRs, ing receptor that leads to familial gland is unaffected. Thus, a number
the physiologic role of agonist- hypoparathyroidism (autosomal domi- of mutant GPCRs have been found to
independent signaling of native nant hypocalcemia) (Chattopadhyay be constitutively active and cause dis-
GPCRs expressed at usual levels in et al. 1996). A mutation in the sixth ease in humans. The large number of
animals and humans is not clear. transmembrane helix of the receptor receptors in the GPCR family, along
for luteinizing hormone was the first with the diversity of signals mediated
• Endogenous GPCRs and Human mutation found to cause familial male by its members, makes it likely that
Disease precocious puberty (Shenker et al. additional examples of naturally oc-
Most GPCRs that have been found to 1993). A number of other activating curring activated GPCR mutants
be basally active are mutated recep- mutations of this receptor have been of pathogenic significance will be
tors in which mutations occurred found in other patients with this discovered.

Table 2. Constitutively active GPCRs that cause disease in humans

Receptor Disease Reference

Calcium sensor Familial hypoparathyroidism Chattopadhyay et al. (1996)


LH Familial male precocious puberty Themmen et al. (1997)
PTH/PTHrP Jansen metaphyseal chondroplasia Schipani et al. (1995)
Rhodopsin Congenital night blindness Rao et al. (1994)
Rhodopsin Retinitis pigmentosa Robinson et al. (1992)
TSH Hyperfunctional thyroid nodules Van Sande et al. (1995)
TSH Familial non-autoimmune hyperthyroidism Van Sande et al. (1995)

LH, luteinizing hormone; PTH/PTHrP, parathyroid hormone/parathyroid hormone-related peptide; TSH, thyroid-stimulating hormone or thyrotropin.

TEM Vol. 9, No. 1, 1998 29


• Viral GPCRs and Disease is a recently identified virus that sarcoma lesions. Thus, because of its
Throughout their evolution, viruses is strongly associated with all forms tumorigenic and angiogenic poten-
have acquired host-cell genes that, of Kaposi’s sarcoma, primary effu- tial, KSHV-GPCR is likely, along with
most likely, enhance their propa- sion lymphomas and multicentric several other genes encoded by KSHV
gation. Presumably, over time, natural Castleman’s disease (Chang et al. (Russo et al. 1996), to play a role in
selection would favor those viruses 1994), and perhaps with multiple the pathogenesis of diseases associ-
that have recruited and subverted myeloma (Rettig et al. 1997). KSHV ated with KSHV infection.
genes critical to their survival within encodes a GPCR (ORF 74, KSHV-
host cells. It is thought that viruses GPCR) that is also homologous • Conclusions
propagate more readily in metaboli- to human chemokine receptors It is now evident that a number of
cally active cells and that activation of (Cesarman et al. 1996). KSHV-GPCR native GPCRs exhibit constitutive sig-
cellular metabolism is a consequence is expressed at the messenger RNA naling activity but the role of agonist-
of viral infection. There are three level in tissues from patients with independent activity in normal physi-
viruses known to encode homologs of Kaposi’s sarcoma and also in B-cell ology is not known. On the other
mammalian GPCRs. It is possible that lymphomas (Cesarman et al. 1996). hand, a number of constitutively ac-
virally encoded GPCRs are expressed For GPCRs encoded within viral tive GPCRs are involved in the patho-
in infected cells and enhance viral genomes, KSHV-GPCR is novel in genesis of human disease. Given the
survival/propagation by activating that it exhibits constitutive signaling large number of GPCRs encoded
cellular metabolism. Genes encoding activity (Arvanitakis et al. 1997). within the human genome, estimated
three GPCR homologs were found KSHV-GPCR signals via the phospho- to be more than 800, additional
in the genome of the human cyto- lipase C–inositol 1,4,5-trisphosphate– examples of this pathogenetic mecha-
megalovirus (Chee et al. 1990), a 1,2-diacylglycerol pathway (Arvanitakis nism are likely to be uncovered.
human herpesvirus involved in mono- et al. 1997) and activates the Jun Furthermore, the spectrum of dis-
nucleosis syndromes in normal hosts kinase/stress-activated protein kinase eases caused by constitutively active
and severe inflammatory disorders in and p38-mitogen-activated protein GPCRs is expanding to include dis-
immunocompromised patients. One kinase pathways (Bais et al. 1998). eases caused by infectious agents. We
of these genes, US28, was found to KSHV-GPCR is also more promiscu- think that a more complete eluci-
encode a GPCR that is a functional ous than most chemokine receptors dation of the roles of constitutively
homolog of human C-C chemokine in that it binds C-C and C-X-C chemo- active GPCRs (which may be pro-
receptors (Gao and Murphy 1994). kines. Because constitutive activation duced by mutation of native GPCR
[Chemokines are peptides that cause of the signaling pathways activated genes or by acquisition of novel
chemotaxis. Chemokines may be di- by KSHV-GPCR induces cell prolifer- GPCR genes during infection) in
vided into several classes, including ation and transformation (Post and human disease and an understanding
C-C chemokines (␤ chemokines), in Brown 1996) and constitutively ac- at the molecular level of how these
which two highly conserved Cys tive GPCRs cause tumors in humans pathogenic GPCRs could be inacti-
residues at the peptide amino termini (see above), the potential role of vated may allow rational develop-
are proximate to one another, and KSHV-GPCR in the pathogenesis of ment of specific inverse agonists as
C-X-C chemokines (␣ chemokines), the tumors associated with KSHV in- therapeutic agents.
in which these two highly conserved fection was studied. We showed that
Cys residues are separated by any expression of KSHV-GPCR in rat References
amino acid.] The genome of Herpes- NRK fibroblasts stimulates cell pro-
Ahuja SK, Murphy PM: 1993. Molecular
virus saimiri, a T-lymphotropic liferation (Arvanitakis et al. 1997), piracy of mammalian interleukin-8 recep-
gammaherpesvirus which causes that KSHV-GPCR can transform tor type B by Herpesvirus saimiri. J Biol
lymphoproliferative disorders in sev- mouse NIH 3T3 fibroblasts in vitro Chem 268:20691–20694.
eral non-human primates, contains a and that KSHV-GPCR-expressing Arvanitakis L, Geras-Raaka E, Varma A, et
gene, ECRF3, which encodes a GPCR NIH 3T3 cells form tumors in mice al.: 1997. Human herpesvirus KSHV en-
homolog (Nicholas et al. 1992) that is (Bais et al. 1998). Thus, KSHV-GPCR codes a constitutively active G-protein-
a functional homolog of human C-X-C displays activities of human onco- coupled receptor linked to cell prolifer-
ation. Nature 385:347–350.
receptors (Ahuja and Murphy 1993). genes. Moreover, we found that in
US28 and ECRF3 are activated by NIH 3T3 cells, KSHV-GPCR induces Bais C, Santomasso B, Coso O, et al.: 1998.
certain C-C or C-X-C chemokines, re- the expression of vascular endo- Kaposi’s sarcoma associated G protein-
coupled receptor is a viral oncogene and
spectively, but do not exhibit consti- thelial growth factor, a potent and
angiogenesis activator. Nature 391:86–89.
tutive signaling activity. The role(s) efficacious stimulator of angiogen-
Barker EL, Westphal RS, Schmidt D, et al.:
of US28 or ECRF3 in disease patho- esis. This latter finding, which must
1994. Constitutively active 5-hydroxytrypt-
genesis is unclear. be shown in relevant cell types, is amine2C receptors reveal novel inverse ag-
Kaposi’s sarcoma-associated herpes- noteworthy because exuberant angio- onist activity of receptor ligands. J Biol
virus (KSHV, human herpesvirus 8) genesis is characteristic of Kaposi’s Chem 269:11687–11690.

30 TEM Vol. 9, No. 1, 1998


Bond RA, Leff P, Johnson TD, et al.: 1995. Heinflink M, Nussenzveig DR, Grimberg H, Rettig MB, Ma HJ, Vescio RA, et al.: 1997.
Physiological effects of inverse agonists in et al.: 1995. A constitutively active mutant Kaposi’s sarcoma-associated herpesvirus
transgenic mice with myocardial over- thyrotropin-releasing hormone receptor is infection of bone marrow dendritic cells
expression of the ␤2-adrenoceptor. Nature chronically down-regulated in pituitary from multiple myeloma patients. Science
374:272–276. cells: evidence using chlordiazepoxide as 276:1851–1854.
a negative antagonist. Mol Endocrinol
Bouaboula M, Perrachon S, Milligan L, et 9:1455–1460. Robbins LS, Nadeau JH, Johnson KR, et al.:
al.: 1997. A selective inverse agonist for 1993. Pigmentation phenotypes of variant
central cannabinoid receptor inhibits Jinsi-Parimoo A, Gershengorn MC: 1997. extension locus alleles result from point
mitogen-activated protein kinase acti- Constitutive activity of native thyrotropin- mutations that alter MSH receptor func-
vation stimulated by insulin or insulin- releasing hormone receptors revealed tion. Cell 72:827–834.
like growth factor 1 – evidence for a new using a protein kinase C-responsive re-
model of receptor/ligand interactions. J porter gene. Endocrinology 138:1471–1475. Robinson PR, Cohen GB, Zhukovsky EA,
Biol Chem 272:22330–22339. et al.: 1992. Constitutively active mutants
Kosugi S, Van Dop C, Geffner ME, et al.: of rhodopsin. Neuron 9:719–725.
Cesarman E, Nador RG, Bai F, et al.: 1996. 1995. Characterization of heterogeneous
Kaposi’s sarcoma associated herpesvirus mutations causing constitutive activation Russo JJ, Bohenzky RA, Chien MC, et al.:
contains G protein-coupled receptor and of the luteinizing hormone receptor in fa- 1996. Nucleotide sequence of the Kaposi
cyclin D homologs which are expressed milial male precocious puberty. Hum Mol sarcoma-associated herpesvirus (HHV8).
in Kaposi’s sarcoma and malignant Genet 4:183–188. Proc Natl Acad Sci USA 93:14862–14867.
lymphoma. J Virol 70:8218–8223. Samama P, Cotecchia S, Costa T, et al.:
Labrecque J, Fargin A, Bouvier M, et al.:
Chang Y, Cesarman E, Pessin MS, et al.: 1995. Serotonergic antagonists differen- 1993. A mutation-induced activated state
1994. Identification of herpesvirus- tially inhibit spontaneous activity and of the ␤2-adrenergic receptor. Extending
like DNA sequences in AIDS-associated decrease ligand binding capacity of the rat the ternary complex model. J Biol Chem
Kaposi’s sarcoma. Science 266: 5-hydoxytryptamine type 2C receptor in 268:4625–4636.
1865–1869. Sf9 cells. Mol Pharmacol 48:150–159. Scheer A, Cotecchia S: 1997. Constitutively
Chattopadhyay N, Mithal A, Brown EM: Leeb-Lundberg LMF, Mathis SA, Herzig active G protein-coupled receptors: poten-
1996. The calcium-sensing receptor: a MCS: 1994. Antagonists of bradykinin tial mechanisms of receptor activation.
window into the physiology and patho- that stabilize a G-protein-uncoupled state J Recept Signal Transduct Res 17:57–73.
physiology of mineral ion metabolism. of the B2 receptor act as inverse agonists Schipani E, Kruse K, Jüppner H: 1995. A
Endocr Rev 17:289–307. in rat myometrial cells. J Biol Chem constitutively active mutant PTH-PTHrP
269:25970–25973. receptor in Jansen-type metaphyseal
Chee MS, Satchwell SC, Preddie E, et al.:
1990. Human cytomegalovirus encodes Lefkowitz RJ, Cotecchia S, Samama P, et al.: chondrodysplasia. Science 268:98–100.
three G protein-coupled receptor homo- 1993. Constitutive activity of receptors Schutz W, Freissmuth M: 1992. Reverse in-
logues. Nature 344:774–777. coupled to guanine nucleotide regulatory trinsic activity of antagonists on G pro-
proteins. Trends Pharmacol Sci tein-coupled receptors. Trends Pharmacol
Chidiac P, Hebert TE, Valiquette M, et al.: 14:303–307.
1994. Inverse agonist activity of ␤-adren- Sci 13:376–380.
ergic antagonists. Mol Pharmacol Milano CA, Allen LF, Rockman HA, et al.: Shenker A, Laue L, Kosugi S, et al.: 1993. A
45:490–499. 1994. Enhanced myocardial function in constitutively activating mutation of the
transgenic mice overexpressing the ␤2- luteinizing hormone receptor in familial
Cohen DP, Thaw CN, Varma A, et al.: 1997. adrenergic receptor. Science 264:582–586.
Human calcitonin receptors exhibit male precocious puberty. Nature
agonist-independent (constitutive) signal- Nicholas J, Cameron KR, Honess RW: 1992. 365:652–654.
ing activity. Endocrinology 138:1400–1405. Herpesvirus saimiri encodes homologues Smit MJ, Leurs R, Alewijnse AE, et al.: 1996.
of G protein-coupled receptors and cyclins. Inverse agonism of histamine H2 antag-
Costa T, Ogino Y, Munson PJ, et al.: 1992. Nature 355:362–365.
Drug efficacy at guanine nucleotide- onists accounts for upregulation of spon-
binding regulatory protein-linked recep- Nussenzveig DR, Thaw CN, Gershengorn taneously active histamine H2 receptors.
tors: thermodynamic interpretation of MC: 1994. Inhibition of inositol phos- Proc Natl Acad Sci USA 93:6802–6807.
negative antagonism and of receptor phate second messenger formation by Spiegel AM: 1995. Defects in G protein-
activity in the absence of ligand. Mol intracellular loop one of a human calci- coupled signal transduction in human
Pharmacol 41:549–560. tonin receptor. Expression and mutational disease. Annu Rev Physiol 58:143–170.
analysis of synthetic receptor genes. J Biol
Gao J-L, Murphy PM: 1994. Human cyto- Chem 269:28123–28129. Strader CD, Fong TM, Tota MR, et al.: 1994.
megalovirus open reading frame US28 Structure and function of G protein-
encodes a functional ␤ chemokine recep- Post GR, Brown JH: 1996. G protein- coupled receptors. Annu Rev Biochem
tor. J Biol Chem 269:28539–28542. coupled receptors and signaling pathways 63:101–132.
regulating growth responses. FASEB J
Gardella TJ, Luck MD, Jensen GS, et al.: 10:741–749. Themmen APN, Martens JWM, Brunner
1996. Inverse agonism of amino-termi- HG: 1997. Gonadotropin receptor mu-
nally truncated parathyroid hormone Prézeau L, Gomeza J, Ahern S, et al.: 1996. tations. J Endocrinol 153:179–183.
(PTH) and PTH-related peptide (PTHrP) Changes in the carboxyl-terminal domain
analogs revealed with constitutively ac- of metabotropic glutamate receptor 1 by Tiberi M, Caron MG: 1994. High agonist-
tive mutant PTH/PTHrP receptors. alternative splicing generate receptors independent activity is a distinguishing
Endocrinology 137:3936–3941. with differing agonist-independent activ- feature of the dopamine D1B receptor
ity. Mol Pharmacol 49:422–429. subtype. J Biol Chem 269:27925–27931.
Hasegawa H, Negishi M, Ichikawa A: 1996.
Two isoforms of the prostaglandin E re- Rao VR, Cohen GB, Oprian DD: 1994. Van Sande J, Parma J, Tonacchera M, et al.:
ceptor EP3 subtype different in agonist- Rhodopsin mutation G90D and a mol- 1995. Somatic and germline mutations of
independent constitutive activity. J Biol ecular mechanism for congenital night the TSH receptor gene in thyroid diseases.
Chem 271:1857–1860. blindness. Nature 367:639–642. J Clin Endocrinol Metab 80:2577–2585.

TEM Vol. 9, No. 1, 1998 31

You might also like