You are on page 1of 28

1521-0081/66/3/570–597$25.00 http://dx.doi.org/10.1124/pr.113.

008425
PHARMACOLOGICAL REVIEWS Pharmacol Rev 66:570–597, July 2014
Copyright © 2014 by The American Society for Pharmacology and Experimental Therapeutics

ASSOCIATE EDITOR: FINN OLAV LEVY

Pharmacology and Signaling of MAS-Related


G Protein–Coupled Receptors
Hans Jürgen Solinski, Thomas Gudermann, and Andreas Breit
Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-Universität München, Munich, Germany

Abstract. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 571
I. Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 571
II. Genes Encoding MAS-Related G Protein–Coupled Receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 572
A. Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 572
B. Phylogeny . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 572
C. Expression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 574
D. Evolution. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 575
III. Pharmacology and Physiology of MAS-Related G Protein–Coupled Receptors . . . . . . . . . . . . . . . . . 577
A. Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 577
B. MAS-Related G Protein–Coupled Receptors A . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 577

Downloaded from by guest on March 16, 2019


1. Pharmacology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 577
2. Signaling Cascades and Physiologic Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 578
C. MAS-Related G Protein–Coupled Receptors B . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 579
D. MAS-Related G Protein–Coupled Receptors C . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 579
1. Pharmacology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 579
2. Signaling Cascades and Physiologic Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 581
a. Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 581
b. Pain-enhancing effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 581
c. Analgesic effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 583
d. Pruritogenic effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 583
E. MAS-Related G Protein–Coupled Receptors D . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 584
1. Pharmacology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 584
2. Signaling Cascades and Physiologic Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 584
F. MAS-Related G Protein–Coupled Receptors E to -H . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 585
G. MAS-Related G Protein–Coupled Receptors X . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 585
1. Pharmacology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 585
a. MAS-related G protein–coupled receptors X1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 585
b. MAS-related G protein–coupled receptors X2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 587
c. MAS-related G protein–coupled receptors X3 and -4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 587
2. Signaling Cascades and Physiologic Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 587
a. Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 587
b. Somatosensory functions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 588
c. Primary sensory neuron plasticity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 588
d. Mast cell biology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 591
e. Putative role of MAS-related G protein–coupled receptors X2 in sleep and
blood pressure regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 591
IV. Agonist-Promoted Internalization and Desensitization of MAS-Related
G Protein–Coupled Receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 591

This work was supported by a grant from the “Deutsche Forschungsgemeinschaft” [Grant BR 3346/3-1].
Address correspondence to: Dr. Andreas Breit, Walther-Straub-Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-
Universität München, Goethestrasse 33, 80336 Munich, Germany. E-mail: Andreas.Breit@lrz.uni-muenchen.de
T.G. and A.B. are co-senior authors.
dx.doi.org/10.1124/pr.113.008425.

570
MAS-Related G Protein–Coupled Receptors 571

V. Functional Interactions of MAS-Related G Protein–Coupled Receptors with


Other Receptor Families. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 592
A. Heteromultimerization among MAS-Related G Protein–Coupled
Receptors and Receptor Subtypes of Other GPCR Families . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 592
B. Inhibition of Tolerance to Morphine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 593
VI. Conclusions and Perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 594
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 594
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 594

Abstract——Signaling by heptahelical G protein– angiotensin-(1–7), alamandine, GABA, cortistatin-14,


coupled receptors (GPCR) regulates many vital body and cleavage products of proenkephalin, pro-
functions. Consequently, dysfunction of GPCR sig- opiomelanocortin, prodynorphin, or proneuropeptide-
naling leads to pathologic states, and approximately FF-A. The full spectrum of biologic roles of all MRGPR
30% of all modern clinical drugs target GPCR. One is still ill-defined, but there is evidence pointing to a role
decade ago, an entire new GPCR family was discov- of distinct MRGPR subtypes in nociception, pruritus,
ered, which was recently named MAS-related G sleep, cell proliferation, circulation, and mast cell
protein–coupled receptors (MRGPR) by the HUGO degranulation. This review article summarizes
Gene Nomenclature Committee. The MRGPR family findings published in the last 10 years on the
consists of ∼40 members that are grouped into nine phylogenetic relationships, pharmacology, sig-
distinct subfamilies (MRGPRA to -H and -X) and are naling, physiology, and agonist-promoted regula-
predominantly expressed in primary sensory neurons tion of all MRGPR subfamilies. Furthermore, we
and mast cells. All members are formally still con- highlight interactions between MRGPR and other
sidered "orphan" by the Committee on Receptor No- hormonal systems, paying particular attention
menclature and Drug Classification of the International to receptor multimerization and morphine toler-
Union of Basic and Clinical Pharmacology. However, ance. Finally, we discuss the challenges the field
several distinct peptides and amino acids are dis- faces presently and emphasize future directions of
cussed as potential ligands, including b-alanine, research.

I. Introduction identified for ;100 of the GPCR identified so far,


suggesting that many biologic functions of GPCR may
G protein–coupled receptors (GPCR), also known
not have been discovered yet (Davenport et al., 2013).
as seven-transmembrane domain receptors, constitute
It is reasonable to assume that these "orphan" GPCR
a large protein family that senses a plethora of distinct
have an immense therapeutic potential, because ;30%
physical and chemical stimuli (biogenic amines, amino
of all clinically relevant drugs already target GPCR
acids, ions, lipids, nucleotides, peptides, proteins, light, directly or modulate their cognate signaling pathways
odorants, and pheromones) outside the cell. Upon (Overington et al., 2006). Hence, in recent years, con-
GPCR activation, a host of intracellular signaling pro- siderable efforts have been undertaken to identify
teins (enzymes, transcription factors, ion channels) are endogenous ligands of "orphan" GPCR by high-
engaged, affecting vital cellular and organismal func- throughput screening strategies. As a result of one of
tions, such as cell proliferation, differentiation, de- these efforts, Lembo and coworkers (2002) reported a
velopment, survival, circulation, metabolism, neuronal group of human "orphan" receptors that are activated
signal transmission, and sensory perception. Approxi- by proenkephalin (PENK) cleavage products of the
mately 800 putative GPCR have been identified in bovine adrenal medulla (BAM) peptide family. The
humans and more than 300 of them are nonodorant authors called these receptors sensory neuron–specific
receptors (Fredriksson et al., 2003). Interestingly, en- G protein–coupled receptors (SNSR) because of their
dogenous or natural exogenous ligands have not been restricted expression pattern in primary sensory

ABBREVIATIONS: BAM, bovine adrenal medulla; CAP, capsaicin; CCR2, chemokine receptors 2; CFA, complete Freund’s adjuvant; CGRP,
calcitonin gene-related peptide; CHO, Chinese hamster ovary; D-APV, D-(2)-2-amino-5-phosphonopentanoic acid; DOP, d-OR; DRG, dorsal
root ganglia; ERK, extracellular signal-regulated kinase; GFP, green fluorescent protein; GPCR, G protein–coupled receptors; HC-030031,
1,2,3,6-tetrahydro-1,3-dimethyl-N-[4-(1-methylethyl)phenyl]-2,6-dioxo-7H-purine-7-acetamide, 2-(1,3-dimethyl-2,6-dioxo-1,2,3,6-tetrahydro-
7H-purin-7-yl)-N-(4-isopropylphenyl)acetamide; HEK, human embryonic kidney; MK-801, (5S,10R)-(+)-5-methyl-10,11-dihydro-5H-dibenzo
[a,d]cyclohepten-5,10-imine; MOP, m-OR; Mrg, MAS-related gene; MRGPR, MAS-related G protein–coupled receptors; MSH, melanocyte
stimulating hormone; NC-IUPHAR, Committee on Receptor Nomenclature and Drug Classification of the International Union of Basic and
Clinical Pharmacology; NFAT, nuclear factor of activated T cells; NMDA, N-methyl-D-aspartate; nNOS, neuronal nitric oxide synthetase;
NPAF, neuropeptide AF; NPFF, neuropeptide FF; OR, opioid receptors; PAR, protease-activated receptors; PENK, proenkephalin; PIP2,
phosphatidylinositol-3,4-bisphosphate; PKC, protein kinase C; PLC, phospholipase C; PTX, pertussis toxin; SNSR, sensory neuron–specific
receptors; TrkA, neurotrophic tyrosin kinase receptor 1; TRP, transient receptor potential cation channel; TRPA, TRP ankyrin; TRPM, TRP
melastatin 8; TRPV, TRP vanilloid 1; YM58483, N-[4-[3,5-bis(trifluoromethyl)-1H-pyrazol-1-yl]phenyl]-4-methyl-1,2,3-thiadiazole-5-
carboxamide.
572 Solinski et al.

neurons and postulated a role for SNSR in pain cascades, and physiologic effects. Significant progress
perception. Shortly before, Dong and colleagues has been made recently and, thus, several MRGPR
(2001) compared cDNA libraries from wild-type mice members emerged as most appealing pharmacological
and from mice lacking the transcription factor neuro- targets for analgesic, antipruritogenic, and antihyper-
genin 1. Mice without functional neurogenin 1 fail to tensive therapies. However, at the same time, advan-
develop the neurotrophic tyrosine kinase receptor 1 ces in our understanding of the physiology and
(TrkA)–positive subclass of dorsal root ganglia (DRG) pathophysiology of MRGPR are hampered by a complex
and trigeminal ganglia neurons that detect painful phylogeny, redundancy in ligand binding, and multi-
stimuli, so called TrkA+ nociceptors (Ma et al., 1999). faceted functions. This review is an attempt to summa-
Thus, the authors postulated that genes found in the rize our present knowledge about the MRGPR family
library of wild-type mice, but not in neurogenin 1– and to point out current caveats. We present an over-
deficient mice, should be specific for TrkA+ nociceptors. view of MRGPR genetics, including phylogeny and evo-
By this approach, the authors discovered an entire new lution, and of pharmacologic and physiologic details of
family of selectively expressed GPCR and named these each MRGPR subfamily. Finally, we will highlight
proteins MAS-related genes (Mrg), because of their agonist-induced MRGPR regulation and interactions
homology to the oncogenic GPCR MAS1. The Mrg with other GPCR families identified so far.
family in rodents and humans comprises ;40 members
that can be divided into several subfamilies. It soon
turned out that one of these subfamilies, the MrgX II. Genes Encoding MAS-Related G
family, is identical to the SNSR proteins (Lembo et al., Protein–Coupled Receptors
2002), such that the same receptor proteins were given A. Preface
two different names. Even more confusing, the rat
MrgC/SNSR was named after its proposed cognate MRGPR-encoding genes have been detected in
ligand BAM peptide-activated receptor with nonopioid mammals, including rodents, Canis lupus, Bos taurus,
activity, and MRG is also used as an abbreviation for and primates. No MRGPR genes have been identified
mortality factor on chromosome 4–related genes in lower vertebrates so far, probably because of less
[mortality factor on chromosome 4/MRG functions in advanced gene sequencing and annotation processes of
aging are reviewed elsewhere (Chen et al., 2010a)], lower vertebrate genomes. Only one MRGPR member
a group of proteins distinct from the GPCR superfam- was annotated in the genome of birds: Solely the
ily. Thus, inconsistencies in the nomenclature de- MRGPRH was found in Gallus gallus. The GPCR
veloped over the last decade may have contributed to superfamily is divided based on sequence homology
confusions in the field. To overcome this problem, the into class A to F. According to the existence of several
HUGO Gene Nomenclature Committee now refers to conserved motifs, for instance the NPxxY motif in
Mrg/SNSR proteins as MAS-related G protein–coupled transmembrane domain seven, the MRGPR family is
receptors (MRGPR) (www.genename.org). So far, a to- assigned to the rhodopsin-like class A (Dong et al.,
tal of 38 genes encoding MRGPR proteins has been 2001; Lembo et al., 2002; Fredriksson et al., 2003;
listed in National Center for Biotechnology Informa- Katritch et al., 2013). The rhodopsin-like GPCR class A
tion databases (orthologous proteins from distinct can be further subdivided into four groups designated
mammalian species are counted as one member, see a to d. MRGPR belong to the d-group of class A, which
Fig. 1 and Table 1). Therefore, to the best of our also comprises glycohormone, purinergic and the very
knowledge, MRGPR represent the nonodorant GPCR large family of olfactory receptors, with glycohormone
family with the largest number of members known so receptors being the receptor family most closely related
far. At present, MRGPR are arranged in nine sub- to the MRGPR family (Dong et al., 2001; Lembo et al.,
families, designated by capital letters, whereas individ- 2002; Fredriksson et al., 2003; Katritch et al., 2013).
ual subtypes are indicated by numbers. For instance,
the MRGPR subtype 1 of subfamily X, originally termed B. Phylogeny
MrgX1 (according to Dong et al., 2001) or SNSR4 The mammalian family of MRGPR can be subdivided
(according to Lembo et al., 2002), is now officially called into nine separate subfamilies (A–H and X) because of
MRGPRX1. This nomenclature is also recognized by sequence similarities (see Fig. 1 and Table 1). Sub-
Committee on Receptor Nomenclature and Drug Clas- families A, B, C, and H exist only in rodents, whereas
sification of the International Union of Basic and Clin- subfamily X is specific to primates including humans,
ical Pharmacology (NC-IUPHAR; Davenport et al., macaque, and rhesus monkey (Dong et al., 2001;
2013) and is recommended for use to improve scientific Lembo et al., 2002; Zylka et al., 2003; Zhang et al.,
communication. 2005; Burstein et al., 2006). In contrast, subfamilies D
Since their first description, considerable efforts to G are conserved in different mammalian species,
have been undertaken to characterize MRGPR in including rodents and primates (Dong et al., 2001;
terms of ligand binding profile, regulation, signaling Lembo et al., 2002; Zylka et al., 2003).
MAS-Related G Protein–Coupled Receptors 573

Fig. 1. Phylogeny of MAS-related G protein–coupled receptors. A phylogenetic tree of all 38 MRGPR members from the nine MRGPR subfamilies (A–
H, X) of mice (m), rat (r), human (h), and rhesus monkey (Rh) was computed using Geneious 7 (Biomatters, Auckland, New Zealand; Blosum62 cost
matrix, Jukes-Cantor genetic distance model, Neighbor-Joining tree build method). GeneID of MRGPR genes and their aliases are given in Table 1.
The scale bar indicates amino acid substitutions per site. Deeper insights into the phylogenetic relationship of the human MRGPR and other GPCR
families are provided within the excellent articles of Fredriksson or Katritch (Fredriksson et al., 2003; Katritch et al., 2013).

The MRGPRA subfamily composition differs consid- protein-encoding gene per species (Zylka et al., 2003). The
erably among rodents. Although only one gene is found MRGPRX subfamily was originally reported to consist of
in rats, mice harbor 18 protein-encoding MRGPRA four distinct genes that are all located on chromosome
genes as well as several pseudogenes (Dong et al., 11p15.1 in humans (Dong et al., 2001). Three of the four
2001; Zylka et al., 2003). The MRGPRB subfamily receptors discovered earlier were identified independently
comprises seven protein-encoding genes in rats, nine in by other researchers, and three additional receptors were
mice, and several pseudogenes in both species (Dong isolated from the same laboratory (Lembo et al., 2002),
et al., 2001; Zylka et al., 2003). According to Zylka et al. resulting in a total of seven distinct MRGPRX receptors.
(2003), a further subdivision of MRGPRB genes into However, three of the original putative MRGPRX
the subgroups B2, B4, and B8 is procurable and reflects members (SNSR2, -3, and -5) have not yet been listed
the basic set of MRGPRB in other rodent species, such by NC-IUPHAR, most likely because these receptors are
as gerbil. The MRGPRC subfamily consists of only one up to 98% identical to the listed MRGPRX and, thus,
gene in rats and one protein-encoding gene in addition probably represent polymorphisms but not distinct sub-
to 13 pseudogenes in mice (Han et al., 2002; Zylka et al., types. Hence, four distinct MRGPRX proteins (MRGPRX1
2003). The MRGPRD subfamily comprises only one to -4) are currently listed by NC-IUPHAR and will be
protein-encoding gene per species (Dong et al., 2001; reviewed herein. Table 2 provides an overview of the
Lembo et al., 2002; Zylka et al., 2003). Just like identities and relationships of all MRGPRX members
MRGPRD, MRGPRE to -H subtypes exhibit only one published and officially listed so far.
574 Solinski et al.

TABLE 1
Phylogeny of MAS-related G protein–coupled receptors
All 38 MRGPR from 4 different species are listed by their designated names, aliases, and GeneID. The ascribed RefSeq
records have been reviewed by National Center for Biotechnology Information (NCBI) staff and are annotated to the
reference genome (mouse: annotation release 103, rat: build 5.1, human: annotation release 105, rhesus monkey: build
1.2). A straight line below the GeneID indicates that the RefSeq record has not yet been subject to individual review by
NCBI staff. A GeneID in parentheses indicates that the underlying information was provided by an NCBI collaborator, an
NCBI RefSeq record is yet missing, and accordingly no annotation to the reference genome exists. Notice that if MRGPRA
and -B subfamily members are not numbered consecutively, numbers in between designate pseudogenes.

Subfamily Number Aliases GeneID

MRGPRA 1 MrgA1 (M) 233221*


— MrgA10, AdeninR, MRGPRX3 (R) 252960†
2a - (M) 668727*
2b MrgA2 (M) 235712*
3 MrgA3 (M) 233222*
4 MrgA4 (M) 235854*
5 MrgA5 (M) 404235*
6 MrgA6 (M) 381886*
7 MrgA7 (M) 404236*
8 MrgA8 (M) (404237)*
9 MrgA9 (M) 668725*
10 MrgA10 (M) (404243)*
11 MrgA11 (M) (404244)*
12 MrgA12 (M) (404245)*
13 MrgA13 (M) (404246)*
14 MrgA14 (M) (404247)*
15 MrgA15 (M) (404248)*
16 MrgA16, Gm486 (M) (404249)*
19 MrgA19 (M) (404250)*
MRGPRB 1 MrgB1 (M, R), MRGPRX2 (R) 233231*/404640†
2 MrgB2 (M, R), MRGPRX2-like (R) 243979*/404645†
3 MrgB3 (M, R) 404238*/(404656)†
4 MrgB4 (M, R) 233230*/404658†
5 MrgB5 (M, R) 404239*/404644†
6 MrgB6 (R) 502338†
8 MrgB8, MrgB13 (M, R) 404240*/404643†
10 MrgB10, MRGPRX2 (M) 243978*
11 MrgB11 (M) (404251)*
13 MrgB13 (M) 620137*
MRGPRC — Mrgprc11, MRGPRX1 (M, R), 404242*/282547†
MrgC11 (M), SNSR1 (R)
MRGPRD — MrgD, TGR7 (M, R, H, Rh) 211578*/293648†/
116512‡/709555x
MRGPRE — MrgE (M, R, H, Rh), GPR167 (H) 244238*/404660†/
116534‡/706822x
MRGPRF — RTA, MrgF, GPR140, GPR168 (M, R, H, Rh) 211577*/266762†/
116535‡/721546x
MRGPRG — MrgG (M, R, H, Rh), Gm1098 (M), GPR169 (H) 381974*/49929†
386746‡/721253x
MRGPRH — GPR90, MrgH (M, R) 80978*/404641†
MRGPRX 1 MrgX1 (H, Rh), SNSR4 (H) 259249‡/692101x
2 MrgX2 (H, Rh), MrgX2-2 (Rh) 117194‡/692078x
3 MrgX3 (H, Rh), SNSR1 (H) 117195‡/692077x
4 MrgX4 (H, Rh), SNSR6 (H) 117196‡/692123x
M, mouse; R, rat; H, human; Rh, rhesus monkey.
GeneIDs: *mouse; †rat; ‡human; xrhesus monkey.

C. Expression and McMahon, 1998). All MRGPRA, MRGPRC to -H,


The majority of MRGPR is expressed in isolectin B4- and MRGPRX members, as well as the MRGPRB4 and
positive small-diameter somatosensory afferents, which -5 subtype, can be subsumed to be expressed in isolectin
represent about half of the nociceptors per DRG (Snider B4-positive DRG neurons (Dong et al., 2001; Lembo

TABLE 2
The MAS-related G protein–coupled receptors subfamily X
An overview of MRGPRX subfamily members, their identity, and their interrelation according to different authors is
given.

Dong et al., 2001 Lembo et al., 2002 Burstein et al., 2006 NC-IUPHAR GeneID or Accession

MrgX1 SNSR4 MrgX1-1 (-2 exists additionally) MRGPRX1 259249


MrgX2 — MrgX2 MRGPRX2 117194
MrgX3 SNSR1 MrgX3-1 (-2 exists additionally) MRGPRX3 117195
MrgX4 SNSR6 MrgX4-1 MRGPRX4 117196
— SNSR5 MrgX4-2 — AF474991
— SNSR2 MrgX6 — AF474988
— SNSR3 MrgX7 — AF474989
MAS-Related G Protein–Coupled Receptors 575

et al., 2002; Robas et al., 2003; Zylka et al., 2003; Zhang to the assumption that neuronal subpopulations
et al., 2005; Tatemoto et al., 2006; Cox et al., 2008; characterized by their MRGPR expression pattern
Liu et al., 2009). None of the remaining MRGPRB might have distinct functions. In line with this hypoth-
members have been detected in primary sensory esis, MRGPRB4- or MRGPRD-positive primary sensory
neurons so far. However, high amounts of MRGPRB3 neurons in mice obtain distinct somatosensory input
and -8 transcripts, as well as lower copy numbers of the from different skin areas (Zylka et al., 2005; Liu et al.,
remaining three rat MRGPRB members, were mea- 2007), whereas MRGPRA3 expression specifies neurons
sured in rat peritoneal mast cells, whereas transcripts that induce itch without transducing nociceptive cues
of MRGPRA or -C were not present in these cells (Han et al., 2013).
(Tatemoto et al., 2006). Recently, MRGPRX1 and -2 In addition to the highly selective expression of
expression was also shown in human mast cells several MRGPR in small-diameter primary sensory
(Tatemoto et al., 2006; Subramanian et al., 2011b; neurons and the aforementioned expression of some
Solinski et al., 2013). Thus, human MRGPRX mirror MRGPR in mast cells, MRGPRD to -H and MRGPRX2
the expression pattern of rodent MRGPRB and -A/C, expression was also found in other tissues. Significant
lending support to the current theory of MRGPR evo- mRNA levels of MRGPRD were detected in urinary
lution, implying that MRGPRX1 and -2 genes inherited bladder, testis, uterus, and arteries (Shinohara et al.,
promoter elements from ancestral MRGPRB and -A/C 2004). MRGPRE transcripts were also monitored in
genes (for details see section II.D). medium- and large-diameter neurons of human DRG
The initial discovery of the MRGPR family pointed to sections (Zhang et al., 2005) and in other areas of the
a selective expression of MRGPR in primary sensory central nervous system, including cerebral cortex,
neurons derived from the TrkA+ population (Dong hippocampus, spinal cord, and cerebellum, as well as
et al., 2001). Perinatally, this results in a largely in human, mouse, and rat placenta (Zhang et al., 2005;
overlapping MRGPRA to -D expression in the same Milasta et al., 2006). MRGPRE and MRGPRF are also
population of neurons (Dong et al., 2001; Zylka et al., expressed in enteric neurons (Avula et al., 2011),
2003; Liu et al., 2008). After birth, TrkA expression belonging to one of the main divisions of the autonomic
ceases in roughly half of the formerly TrkA+ neurons nervous system that regulates gastrointestinal func-
(Molliver et al., 1997). Instead of TrkA, these cells start tions. In accordance with these findings, transcripts of
to express c-ret, the receptor of the glial-derived MRGPRF, formally known as rat thoracic aorta re-
neurotrophic factor (Molliver et al., 1997). In adult- ceptor, were detected in small and large intestine (Ross
hood, expression of all MRGPR is maintained in c-ret+, et al., 1990). More precisely, MRGPRE and MRGPRF
but not in the remaining TrkA+ neurons (Dong et al., proteins were detected in both myenteric and sub-
2001; Zylka et al., 2003; Liu et al., 2008). Interestingly, mucosal neurons, the latter showing coexpression of
despite the largely overlapping expression pattern of both receptors in 30–50% of neurons analyzed (Avula
all MRGPR in perinatal primary sensory neurons, in et al., 2011). The same study reported that intestinal
adulthood most MRGPR subtypes are not coexpressed schistosomiasis and trinitrobenzene sulfonic acid–
in the same c-ret+ population and exhibit a compart- induced ileitis resulted in decreased expression of both
mentalized expression pattern in distinct subpopula- receptors, whereas other neuronal marker proteins,
tions (Dong et al., 2001; Zylka et al., 2003; Liu et al., e.g., calretinin or neuronal nitric oxide synthetase
2008). Notably, these expression patterns are not (nNOS), were not affected by either inflammatory
identical in mice and rats, suggesting variations in insult. MRGPRH transcripts were also detectable in
the physiologic roles of distinct MRGPR in rodents. the heart (Wittenberger et al., 2001), and expression of
The analysis of transcriptional pathways responsible human MRGPRX2 was described in the adrenal glands
for expression of different MRGPR in distinct DRG and in several brain areas (Robas et al., 2003; Kamohara
populations in mice revealed that expression of et al., 2005).
MRGPRA to -D initially depends on the activity of
the runt-related transcription factor 1, explaining the D. Evolution
overlapping expression pattern in embryonic neurons The MRGPR subfamilies D to G constitute evolu-
(Liu et al., 2008). During postnatal development, runt- tionarily old genes that are conserved between rodents
related transcription factor 1 suppresses MRGPRA to –C, and primates. Accordingly, although all MRGPR are
but not MRGPRD expression, through its inhibitory encoded on only one chromosome in each species
C-terminal domain, leading to the segregation of analyzed, the subfamilies D to G lie clustered together
murine MRGPRD and MRGPRA to -C expression in a considerable distance apart from the chromosomal
adolescence. Furthermore, the authors found that region encoding subfamilies A to C in rodents or
the transcription factor Smad4 is indispensable for subfamily X in primates (Zylka et al., 2003). The
MRGPRB4 expression, but does not affect expression phylogenetic relationship between the latter MRGPR
of other MRGPR subtypes (Liu et al., 2008). The subfamilies poses the question of how and why the
distinct expression pattern of MRGPR in adulthood led entire MRGPR family developed in such a complex
576 Solinski et al.

manner during mammalian evolution. A current model also explain the highly specific expression of rodent
(see Fig. 2) rests on the assumption that an ancestral MRGPRA and -C and of human MRGPRX in primary
progenitor gene cluster of one MRGPRA/C/X gene and sensory neurons, assuming that those promoter ele-
one MRGPRB gene existed before rodent speciation ments crucial for the restrictive expression pattern
(Zylka et al., 2003). An unequal crossing over event led already existed in the progenitor gene and were pre-
to local MRGPRB gene duplication in the rodent served during gene duplication events (Dong et al.,
lineage, whereas MRGRPB genes were completely lost 2001; Lembo et al., 2002; Zylka et al., 2003; Zhang
in the primate lineage. In rodents, the MRGPRA/C/X et al., 2005). Likewise, additional expression of
progenitor developed into two separate gene subfami- MRGPRX1 and -2 in human mast cells could be the result
lies because of a further round of local gene duplica- of conserved ancestral promoter elements responsible for
tion. Specifically in mice, the de novo insertion of the MRGPRB expression in rat mast cells (Tatemoto et al.,
L1 retrotransposon initiated several additional local 2006). Thus, MRGPRX1 and -2 inherited their re-
gene duplication events of MRGPRA and -C genes. strictive expression in primary sensory neurons and mast
However, additional MRGPRC genes lost their tran- cells by a combination of gene expression regulatory
scriptional regulatory elements and are now pseudo- elements of rodent MRGPRA/C and certain rodent
genes. This assumption is supported by the observation MRGPRB genes.
that all MRGPR genes are found on only one chro- The development of single MRGPRX genes in
mosome, e.g., chromosome 1q in rats or chromosome primate evolution was also initiated by local gene
7B in mice. Notably, such an evolutionary model would duplication (note that all human MRGPRX genes are

Fig. 2. Schematic model displaying the putative divergent evolution of the MRGPRA/B/C/X gene cluster in primates and rodents. Based on the data
provided by Zylka et al. (2003), a theoretical model explaining the evolutionary events leading form a putative ancestral MRGPR gene cluster to the
primate and rodent lineage and further on to species-specific distinctions in the rodent lineage is given.
MAS-Related G Protein–Coupled Receptors 577

located on chromosome 11p15.1) (Dong et al., 2001; will summarize the available pharmacologic data of
Choi and Lahn, 2003; Zylka et al., 2003). Diversion of MRGPR members ordered by subfamily and review
distinct MRGPRX genes was driven by significant the current knowledge about signaling cascades and
positive selection pressure (Choi and Lahn, 2003; physiologic effects elicited by a given receptor-ligand
Fatakia et al., 2011), resulting in amino acid sub- pair.
stitutions in those parts of transmembrane domains
forming the ligand binding cavity, in extracellular B. MAS-Related G Protein–Coupled Receptors A
domains, and in the very C-terminal tail (Choi and 1. Pharmacology. Endogenous or exogenous ligands
Lahn, 2003; Fatakia et al., 2011). From a structural have been assigned to only 4 of 19 MRGPRA members,
point of view, the latter changes may have resulted in i.e., rat MRGPRA and three murine MRGPRA (see
diverging receptor proteins with regard to ligand Table 3). The murine MRGPRA members 1 and 4 were
specificity or affinity (extracellular domains, trans- the first MRGPR to be analyzed in a functional assay.
membrane domains) and the regulation of signaling After recombinant expression in human embryonic
(C-terminal tail). From an evolutionary point of view, kidney (HEK)293 cells, both receptors responded to
the strong positive selection pressure indicates that several peptides of the RFamide family (Dong et al.,
novel MRGPRX proteins are crucial for primate- 2001) first discovered in mollusks and soon shown
specific physiology and represent important dif- to modulate nociception in vertebrates (Price and
ferences between primates and rodents. Indeed, Greenberg, 1977; Yang et al., 1985). The mammalian
primate-specific genes are clearly over represented in neuropeptide FF (NPFF) increased intracellular
the fraction of human disease-causing genes (Hao et al., calcium concentrations via MRGPRA1 with a potency
2010). Interestingly, a continuing positive selection of ;200 nM, whereas the mammalian neuropeptide
pressure on MRGPRX genes may account for frequent AF (NPAF) likewise activated the MRGPRA4 sub-
polymorphisms of MRGPRX genes in the human pop- type with a potency of ;60 nM. These findings were
ulation and, thus, account for differences in published subsequently confirmed by others (Han et al., 2002;
MRGPRX/SNSR subtypes. Liu et al., 2009). Both peptides are also known to
activate NPFF receptor 1 or 2 with potencies and
III. Pharmacology and Physiology of affinities in the nanomolar range (Mollereau et al.,
MAS-Related G Protein–Coupled Receptors 2002). Thus, if these neuropeptides represent the
endogenous agonists of murine MRGPRA1 or -4,
A. Preface they would not be selective for the latter receptors.
To date, no MRGPR subtype has officially been In addition to endogenously occurring neuropep-
declared "deorphanized" by NC-IUPHAR (Davenport tides, murine MRGPRA1 was found to be a target of
et al., 2013). This recommendation would require at the human version of the b-salusin peptide, employ-
least two independent demonstrations of receptor- ing fluorescence imaging plate reader technology
ligand pairing published in refereed papers. To and MRGPRA1 overexpressing HEK293 or Chinese
"deorphanize" a receptor, NC-IUPHAR suggests using hamster ovary (CHO) cells (Wang et al., 2006b). As
radioligand binding and functional assays in in vitro the orthologous murine peptide failed to activate
systems and native tissues in conjunction with ana- MRGPRA1, it was concluded that b-salusin is only
tomic data indicating that the proposed ligand is a surrogate ligand of MRGPRA1.
present to activate the receptor in a given tissue. The antimalaria drug chloroquine was recently
Additionally, genetic approaches that alter expression shown to induce intracellular calcium transients in
levels of the receptor or the ligand might be beneficial. cells expressing MRGPRA3 (Liu et al., 2009). This
So far, at least one of the criteria mentioned is lacking response was characterized by an EC50 value of ;27 mM
for any given MRGPR. These criteria are particularly after heterologous expression in HEK293 cells and
hard to meet for human MRGPRX subtypes: because of was also noted in dissociated primary sensory neurons.
their specific expression in primates and their re- A cluster knockout mouse deficient of several MRGPR
stricted expression pattern in primary sensory neurons (including MRGPRA1-4, -A10, -A12, -A14, -A16, -A19,
and mast cells, it appears difficult to obtain data from -B4, -B5, and -C) failed to respond to chloroquine as
native tissues and to implement genetic approaches did wild-type neurons after small interfering RNA–
that would alter protein expression levels. However, mediated MRGPRA3 knockdown, indicating that
knowledge of MRGPR biology and pharmacology is MRGPRA3 are indeed targeted by chloroquine (Liu
expanding, a fact that is being appreciated by et al., 2009). Notably, chloroquine also induced calcium
NC-IUPHAR (Davenport et al., 2013). Interestingly, a signals after recombinant expression of rat MRGPRA in
certain degree of pharmacologic overlap of nonorthologous primary sensory neurons derived from the MRGPR
MRGPR members exists, although MRGPR phylogeny cluster knockout mouse (Liu et al., 2009).
is complex, including subfamilies that are specific to The purine adenine might serve as an endogenous
rodents and others that are primate specific. Thus, we agonist of rat MRGPRA, because in silico modeling
578 Solinski et al.

TABLE 3
Pharmacology of MAS-related G protein–coupled receptors A
MRGPRA subfamily members that have been assigned to a ligand are listed. All data were generated after
recombinant MRGPR expression.

Receptor Ligand EC50 Readout System Reference

nM
mMRGPRA1 FMRFamide 20 calcium HEK293-Ga15 Dong et al., 2001
NPFF 200 calcium HEK293-Ga15 Dong et al., 2001
hb-Salusin 300 calcium HEK293 Wang et al., 2006b
chloroquine 300,000 calcium HEK293 Liu et al., 2009
mMRGPRA4 NPAF 60 calcium HEK293-Ga15 Dong et al., 2001
mMRGPRA3 chloroquine 27,000 calcium HEK293 Liu et al., 2009
rMRGPRA chloroquine N.D. calcium mDRG neurons Liu et al., 2009
adenine 3 cAMP inhibition CHO Bender et al., 2002
60 GTPgS CHO Bender et al., 2002
GTPgS, guanosine 59-3-O-(thio)triphosphate; N.D., not determined; r, rat; m, mouse.

revealed a putative adenine binding pocket in the rat chloroquine-responsive neurons (Liu et al., 2009;
MRGPRA protein (Heo et al., 2007b). This assumption Wilson et al., 2011). In contrast to TRPA1, TRPV1 is
was corroborated by the observation that adenine not required for MRGPRA3-dependent neuronal exci-
bound to rat MRGPRA with a KD value of 24 nM, tation, and chloroquine-induced calcium signals were
when the receptor was overexpressed in CHO cells not detected in cells coexpressing MRGPRA3 and
(Bender et al., 2002). In this cellular model, adenine TRPV1 (Wilson et al., 2011). Functional coupling
inhibited forskolin-induced adenylyl cyclase activity between MRGPRA3 and TRPA1 was assigned to Gbg
and induced guanosine 59-O-(3-[35S]thio)triphosphate protein complexes because of the lack of calcium
binding to the plasma membrane with potencies of ;3 signals after incubation with the Gbg inhibitor gallein.
and ;60 nM, respectively. However, inhibition of The authors further showed that functional interac-
cAMP production by rat MRGPRA points to Gi/o and tions between TRPA1 and MRGPRA3 are required for
not to Gq/11 as downstream effectors in CHO cells. This the physiologic effects of the latter. Than and co-
observation contrasts with the functional properties of workers (2013) confirmed that direct activation of
murine MRGPRA1 that was found to exclusively MRGPRA3 by chloroquine excites murine primary
engage classic Gq/11 signaling cascades (Han et al., sensory neurons because of TRPA1 activation, because
2002; Wang et al., 2006b). the response of these neurons was abolished by the
2. Signaling Cascades and Physiologic Effects. TRPA1 inhibitor HC-030031 [1,2,3,6-tetrahydro-1,3-
Physiologic effects and detailed signaling cascades of dimethyl-N-[4-(1-methylethyl)phenyl]-2,6-dioxo-7H-
MRGPRA members have so far been described for purine-7-acetamide, 2-(1,3-dimethyl-2,6-dioxo-1,2,3,
the murine chloroquine-sensitive subfamily member 6-tetrahydro-7H-purin-7-yl)-N-(4-isopropylphenyl)
MRGPRA3 (see Fig. 3). Intradermal application of acetamide]. However, the authors revealed that
chloroquine induced a profound itching behavior at the MRGPRA3 was also able to induce calcium signals
injection site (Liu et al., 2009). This procedure did not in TRPA1-negative neurons. By use of YM58483 (N-
lead to alloknesis, an aberrant sensation of itch when [4-[3,5-bis(trifluoromethyl)-1H-pyrazol-1-yl]phenyl]-
nonitching skin near an itchy skin patch is lightly 4-methyl-1,2,3-thiadiazole-5-carboxamide) as a nonsubtype
touched (Akiyama et al., 2012). Chloroquine-induced selective canonical TRP and Pyr3 as a TRPC3 blocker,
calcium signals in primary sensory neurons were TRPCs and, in particular, the TRPC3 subtype were
completely inhibited by removal of extracellular cal- identified as the downstream effectors mediating
cium by EGTA and by ;90% after application of MRGPRA3-induced neuronal excitation in the ab-
ruthenium red, a blocker of transient receptor poten- sence of TRPA1 (Than et al., 2013). In addition, this
tial (TRP) cation channels (Liu et al., 2009). By use of work showed profound sensitization of TRPV1 chan-
calcium imaging of cultured primary sensory neurons nels via chloroquine-induced protein kinase C (PKC)
and MRGPRA3 transfected NG108 cells, intracellular activation and inhibition of the cold sensor TRP
signaling cascades were analyzed that might be re- melastatin 8 (TRPM8) via direct binding of the Gaq
sponsible for the sensation of itch (Wilson et al., 2011). subunit to the ion channel, because a Gaq chimera
Most interestingly, chloroquine induced an increase in unable to activate phospholipase C (PLC)b still
intracellular calcium through TRP ankyrin 1 (TRPA1) blocked TRPM8 activity in cells lacking functional
channels, because chloroquine-induced itching was not Gaq proteins (McKemy et al., 2002; Peier et al.,
observed in TRPA1-deficient mice (Wilson et al., 2011). 2002). Thus, although acute signaling of MRGPRA3
The TRP vanilloid 1 (TRPV1) channel, an established via TRPA1 solely leads to itch (Liu et al., 2009;
sensor of different painful stimuli such as heat and Wilson et al., 2011), chloroquine also activates cel-
protons (Caterina et al., 1997), is also expressed in lular signaling cascades that might affect other
MAS-Related G Protein–Coupled Receptors 579

Fig. 3. Schematic presentation of signaling cascades, downstream targets, and physiologic effects proposed for murine MRGPRA3. Upon chloroquine-
induced activation of MRGPRA3 in primary sensory DRG/trigeminal ganglia neurons, the activity of several TRP channels is modulated via distinct
mechanisms. TRPC3 is activated via an unknown mechanism, TRPA1 activation is mediated by Gbg subunits, TRPV1 is sensitized via PKC-mediated
phosphorylation, and TRPM8 is inhibited via Gaq signaling independently from PLCb activation. MRGPRA3-mediated TRPA1 activation leads to itch,
whereas physiologic effects of TRPA1-independent signaling cascades have not yet been determined. Used with permission.

physiologic processes, such as nociception or tem- primary sensory neurons, but not activation of the
perature sensation. MRGPRB4 itself, by gentle stroking was demonstrated by
an elegant in vivo calcium imaging approach. Pharmaco-
C. MAS-Related G Protein–Coupled Receptors B genetic activation of MRGPRB4 expressing neurons
None of the rodent MRGPRB members has been in freely moving mice resulted in conditioned place
assigned to any ligand so far. However, in a reporter preference, suggesting that activation of these neurons
mouse that carries a gene for placental alkaline is positively reinforcing and might be induced by
phosphatase under the control of the MRGPRB4 interindividual social interactions such as allogroom-
promoter, a rare subset of primary sensory neurons ing (Vrontou et al., 2013).
that only innervates the hairy skin showed high
promoter activity (Liu et al., 2007). These neurons D. MAS-Related G Protein–Coupled Receptors C
were nonpeptidergic, TRPV1 negative, displayed 1. Pharmacology. The rodent MRGPRC subfamily
a broad arborization of their cutaneous termini, and consists of only one protein-encoding gene in rats and
are often associated with hair follicles. Most interest- mice, and both receptor subtypes have been suggested
ingly, MRGPRB4 positive neurons project to the spinal to bind several ligands (see Table 4). After heterologous
lamina IIo, a spinal pain processing center that is part expression in different cell systems, MRGPRC from rat
of the substantia gelatinosa (Rexed, 1952). These and mouse were activated to elicit calcium signals by
findings suggested that MRGPRB4 mark C-fiber tactile a set of different but structurally related peptides (see
afferents that are supposed to detect gentle touch Fig. 4A) with potencies in the low to medium nano-
and stroking. Indeed, activation of MRGPRB4-positive molar range (Grazzini et al., 2004; Han et al., 2005;
580 Solinski et al.

TABLE 4
Pharmacology of MAS-related G protein–coupled receptors C
MRGPRC subfamily members are listed by the ligands to which they have been assigned. Data were generated after
recombinant MRGPRC expression, despite when endogenous expression is indicated by “(e)”.

Receptor Ligand EC50 System Reference

nM
mMRGPRC 11 HEK293 (GFP-tag) Han et al., 2002
g 2-MSH 340 HEK293 Heo et al., 2007a
290 HEK293 Solinski et al., 2013
53 HEK293 (GFP-tag) Han et al., 2002
BAM8–22 292 HEK293 Heo et al., 2007a
330 HEK293 Solinski et al., 2013
Dynorphin-14 22 HEK293 (GFP-tag) Han et al., 2002
310 HEK293 Solinski et al., 2013
NPFF 54 HEK293 (GFP-tag) Han et al., 2002
358 HEK293 Heo et al., 2007a
NPAF 282 HEK293 (GFP-tag) Han et al., 2002
SLIGRLamide 10,000 CHO Liu et al., 2011
50,000 DRG neurons (e) Liu et al., 2011
rMRGPRC g 2-MSH 44 HEK293-Gaqi5 Grazzini et al., 2004
440 HEK293 Solinski et al., 2013
Tyr6-g 2-MSH6–12 15 HEK293-Gaqi5 Grazzini et al., 2004
BAM8–22 120 HEK293-Gaqi5 Grazzini et al., 2004
2600 HEK293 Solinski et al., 2013
m, mouse; r, rat.

Heo et al., 2007a; Solinski et al., 2010, 2013). Among some BAM peptides to opioid receptors (OR). Interest-
these peptides are g 2-melanocyte stimulating hor- ingly, ligand sharing with receptor subtypes of other
mone (g2-MSH), BAM peptides, dynorphin-14, and GPCR families is not restricted to MRGPRC, but
proneuropeptide-FF-A peptides. Notably, these pep- appears to be a common feature of many MRGPR as
tides originate from PENK (BAM), pro-opiomelanocortin summarized in Table 9 (Bowery et al., 1980; Quirion
(g 2-MSH), prodynorphin (dynorphin-14), and NPPFA and Weiss, 1983; Roselli-Rehfuss et al., 1993; de Lecea
(NPFF or NPAF) (see Fig. 4A). Such a wide range in et al., 1996; Fukusumi et al., 1997; Kaupmann et al.,
the binding profile of a distinct GPCR is quite unique 1997; Oosterom et al., 1999; Han et al., 2002; Lembo
and suggests that many different physiologic pathways et al., 2002; Mollereau et al., 2002; Robas et al., 2003;
rely on MRGPRC signaling. Activation of MRGPRC by Santos et al., 2003; Grazzini et al., 2004; Shinohara
BAM peptides was validated in primary sensory et al., 2004; Gembardt et al., 2008; Lautner et al.,
neurons of mice (Liu et al., 2009; Wilson et al., 2011) 2013). Although, ligand sharing between MRGPRC
and rats (Honan and McNaughton, 2007). However, and other GPCR subtypes is of physiologic interest, it
their similarities notwithstanding, several pharmaco- is also a major concern when considering the specificity
logical differences between murine and rat MRGPRC of distinct MRGPRC-activating peptides in vivo. In-
have also been observed (see Fig. 4B): NPFF peptides terestingly, after N-terminal truncation of g 2-MSH
displayed lower potency to activate the rat MRGPRC (g 2-MSH6–12) or of BAM1–22 (BAM8–22) activity toward
compared with the murine counterpart, and dynorphin- MRGPRC but not toward melanocortin 3 receptor or
14 did not activate rat MRGPRC at all (Han et al., OR is preserved (Lembo et al., 2002; Grazzini et al.,
2002; Grazzini et al., 2004; Solinski et al., 2013). 2004). Accordingly, g2-MSH6–12 or BAM8–22 are
Regarding efficacy, g2-MSH and BAM peptides are regarded as specific MRGPRC ligands and can be used
the most efficacious activators of both MRGPRC, with in in vivo studies to analyze the physiologic effects of
g2-MSH clearly surmounting BAM peptides (Solinski MRGPRC in mice or rats. When considering g2-MSH or
et al., 2013). Using in silico modeling and site-directed BAM derivatives as putative endogenous MRGPRC
mutagenesis, Heo and coworkers (2007a) revealed ligands, it is also important to investigate whether
that Tyr110, Asp161, and Asp179 of murine MRGPRC these peptides are actually released from precursors
are crucially involved in agonist binding by interact- under physiologic or pathophysiological conditions.
ing with a common C-terminal end of all peptides, g2-MSH and BAM1–22 have both been shown to be
which harbors the consensus sequence RF(Y)amide or present in several tissues, including spinal cord and
RF(Y)G. Notably, all three ligand-binding amino acids brain (Mizuno et al., 1980; Pelletier et al., 1981; Hollt
are conserved in rat MRGPRC. et al., 1982; DeBold et al., 1988a,b; Cai et al., 2007a).
Several peptides that are now recognized as Thus, peptides that bind other GPCRs and MRGPRC
MRGPRC agonists were previously shown to activate could serve as endogenous ligands of the latter. The
distinct receptors from other GPCR families, e.g., specific MRGPRC agonist BAM8–22 was also identi-
g2-MSH also binds to melanocortin 3 receptors and fied in vivo after microdialysis of exogenous BAM1–25
MAS-Related G Protein–Coupled Receptors 581

that peptides such as the "SLIGRL"-peptide are re-


leased from PAR through consecutive cleavage by two
or more proteases and in turn serve as ligand of
distinct GPCR. By use of CHO cells recombinantly
expressing MRGPRC, small interfering RNA–
mediated MRGPRC knockdown in wild-type primary
sensory neurons, and primary sensory neurons from
MRGPRC-deficient mice, "SLIGRL"-induced calcium
signals in the presence, but not in the absence of the
MRGPRC were identified, indicating that the "SLIGRL"-
peptide is indeed a ligand of this receptor. Analysis of
structure-activity relationships revealed that amida-
tion of the "SLIGRL" C terminus is important for its
function as a ligand, which fits well to the proposed
RF(Y)G/amide consensus sequence of other MRGPRC
agonists. Notably, "SLIGRL"-induced itch, which was
believed to be mediated by PAR2 (Shimada et al.,
2006), was not changed in PAR2-deficient mice (Liu
et al., 2011). In MRGPRC overexpressing CHO cells,
Fig. 4. Schematic presentation and comparison of the pharmacology of
BAM8–22-sensitive MRGPR. (A) Amino acid sequences and precursors
a high EC50 value of ;10 mM for "SLIGRL"-induced
of the classic peptidergic MRGPRC agonists are given. Dashed lines calcium signals was measured (Liu et al., 2011).
indicate that the synthesis of the peptide in vivo has not been detected However, because PAR2 and MRGPRC are both ex-
yet. The C-terminal consensus, which is proposed to be crucial for
MRGPRC binding, is highlighted in red. (B) The most potent agonists of pressed in primary sensory neurons, it is appealing to
murine (m) or rat (r) MRGRPC and human (h) MRGPRX1 are provided. speculate that the "SLIGRL" peptide might physiolog-
The ligands are drawn to scale by linearly modifying the font size in
accord with their published potencies (Han et al., 2002; Lembo et al., ically exist in concentrations high enough to stimulate
2002; Grazzini et al., 2004; Liu et al., 2009, 2011; Solinski et al., 2013). MRGPRC.
For simplicity, ligands that exhibit potencies more than 10 times lower 2. Signaling Cascades and Physiologic Effects.
than g 2-MSH (in the case of MRGPRC) or BAM8–22 (in the case of
MRGPRX1) are not shown. a. Preface. Rat and murine MRGPRC elicit their
effects primarily by the activation of G proteins. Both
receptors were shown to induce intracellular calcium
into the striatum of anesthetized rats, using a combi- release via activation of PLCb (Han et al., 2002;
nation of solid-phase preconcentration capillary elec- Grazzini et al., 2004). For murine MRGPRC coupling
trophoresis and imaging matrix-assisted laser desorption/ to Gq/11 proteins seems to be essential, because
ionization mass spectrometry (Zhang et al., 1999). MRGPRC-dependent calcium signaling was absent in
However, although the proteolytic machinery to generate murine embryonic fibroblasts derived from Gq/11-deficient
BAM8–22 in vivo is present in the brain, it has not yet embryos (Han et al., 2002). In accordance with their
been shown that BAM8–22 or g2-MSH6–12 are produced selective expression in small-diameter primary
endogenously in tissues adjacent to primary sensory sensory neurons, MRGPRC signaling in mice and
neurons. Thus, it remains unclear whether and under rats has been implicated in somatosensory detector
which circumstances specific MRGPRC ligands exist in functions in the skin. Indeed, published data on the
vivo. in vivo effects of MRGPRC agonists detected pain-
Very recently, it was proposed that the "SLIGRL"- enhancing, pruritogenic, but also analgesic effects
peptide activates murine MRGPRC (Liu et al., 2011). (summarized in Fig. 5).
"SLIGRL" is not a classic neuropeptide released from b. Pain-enhancing effects. Intraplantar injections
common precursor proteins but part of the tethered of the MRGPRC-specific peptides BAM8–22 or Tyr6-g 2-
ligand domain of murine protease-activated receptors 2 MSH6–12 into juvenile or adult rats dose dependently
(PAR2) [for further reading see these comprehensive resulted in acute nocifensive behavior, thermal hyper-
reviews (Steinhoff et al., 2005; Ramachandran and algesia, and mechanical allodynia (Grazzini et al.,
Hollenberg, 2008)]. PAR are GPCR that are activated 2004; Ndong et al., 2009). Likewise, intrathecal in-
by serine proteases such as thrombin or trypsin. To jection of MRGPRC agonists into juvenile or adult rats
activate PAR, proteases cleave N-terminal peptides and Kunming mice also induced acute pain-like be-
from the PAR protein, thereby exposing a “cryptic” havior and thermal hyperalgesia (Grazzini et al.,
PAR-activating sequence at the N terminus. Short 2004; Chang et al., 2009; Wei et al., 2010). MRGPRC
peptides that are derived from theses cryptic sequences may also be responsible for inflammatory pain,
have also been shown to activate PAR without previous because complete Freund’s adjuvant (CFA)–induced
cleavage, e.g., the "SLIGRL"-amide was established as thermal hyperalgesia was alleviated by RNAi-mediated
a PAR2-specific ligand. However, it may also be possible MRGPRC knockdown in rats (Ndong et al., 2009).
582 Solinski et al.

Fig. 5. Schematic presentation of signaling cascades, downstream targets, and physiologic effects proposed for rodent MRGPRC. Three physiologic
functions of MRGPRC in primary sensory DRG/trigeminal ganglia neurons have been identified: pruritogenic, pain-enhancing, and analgesic effects.
Itch is caused via PLCb-mediated activation of TRPA1. Acute pain, thermal hyperalgesia, and mechanical allodynia were found to depend on PKC-
mediated TRPV1 sensitization, peripheral CGRP release, and spinal nitric oxide (NO) production and NMDA receptor activity. Different pain-inducing
paradigms (marked with red bolts) lead to acute nociceptor activity as well as adaptive changes including CGRP induction, spinal NO production, and
windup of spinal dorsal horn neurons. These changes are occasionally accompanied by MRGPRC induction and increased abundance of BAM1–22 that
can lead to enhanced MRGPRC signaling. Analgesic effects of MRGPRC are partly triggered by engagement of MOP signaling that counteracts
adaptive changes in DRG and spinal neurons. Chronic application of morphine leads to tolerance, among others via induction of similar adaptive
changes as in pain paradigms. Rat MRGPRC have been shown to counteract these processes.

Notably, pain-enhancing effects of MRGPRC can only be 2009; Wilson et al., 2011). In rat primary sensory
assessed at low agonist doses (e.g., up to 20 nmol of Tyr6- neurons, BAM8–22 sensitized TRPV1 for its agonist
g2-MSH6–12) and in a rigid time window of a maximum capsaicin (CAP) via a PKC-dependent pathway (Honan
of 20 minutes postinjection (Wei et al., 2010). This low and McNaughton, 2007). The PKC isoforms d, «, and z
dose effect may be due to the activation of an analgesic were excluded to be part of this signaling cascade,
off-target at higher peptide concentrations, most likely because these isoforms did not translocate to the
the Kyotorphin receptor, which masked MRGPRC- plasma membrane after MRGPRC stimulation. TRPV1
induced pain, whereas rapid peptide degradation may sensitization was also made responsible for an in-
be responsible for the short-term response (Grazzini creased CAP- or heat-induced calcitonin gene-related
et al., 2004; Wei et al., 2010). peptide (CGRP) release from rat or mouse paw skin
Several publications identified heat-sensitive TRPV1 after preincubation with BAM1–22 in conjunction with
ion channels as major downstream targets of MRGPRC naloxone, an opioid receptor antagonist that blocks
responsible for its pain-enhancing effects (Honan and effects of BAM1–22 on opioid receptors but still allows
McNaughton, 2007; Hager et al., 2008; Ndong et al., BAM1–22 to act on MRGPRC (Hager et al., 2008).
MAS-Related G Protein–Coupled Receptors 583

Interestingly, BAM8–22 or g 2-MSH alone failed to increased abundance of its agonist BAM1–22 have
induce CGRP release, and, surprisingly, a combination been observed during the process of CFA-induced
of BAM1–22 and naloxone that sensitized TRPV1 for pain chronification (Cai et al., 2007a; Jiang et al.,
CAP still induced CGRP release from skin prepara- 2013). Consequently, acute intrathecal application of
tions derived from TRPV1-deficient mice. In rats, a BAM1–22-neutralizing antibody decreased CFA-
MRGPRC-mediated thermal hyperalgesia was abro- induced mechanical hyperalgesia, thus highlighting
gated by a specific TRPV1 inhibitor (Ndong et al., ongoing MRGPRC signaling during inflammatory
2009), and coexpression of TRPV1 with murine pain (Cai et al., 2007a). After CFA injection, enhanced
MRGPRC in NG108 cells resulted in augmented calcium MRGPRC signaling inhibited the induction of CGRP
signals by BAM8–22 (Wilson et al., 2011). Thus, TRPV1 in primary sensory and of nNOS in spinal projection
has been defined as a common target for cellular sig- neurons (Jiang et al., 2013), which is believed to exert
naling induced by MRGPRC, which might be of high long lasting effects on spinal sensitization [for details
physiologic significance because coexpression of TRPV1 see (Latremoliere and Woolf, 2009)]. This was attrib-
with MRGPRC in rodent primary sensory neurons was uted to the engagement of m-OR (MOP), as evidenced
frequently observed (Lembo et al., 2002; Hager et al., by the lack of inhibition after preincubation with the
2008; Liu et al., 2009). MOP inhibitor CTAP.
In addition to TRPV1, Chang and coworkers (2009) d. Pruritogenic effects. Concomitant to the obser-
identified the spinal N-methyl-D-aspartate (NMDA) vation that chloroquine-induced scratching behavior is
receptor and the nNOS as players in MRGPRC- mediated largely by murine MRGPRA3 signaling,
mediated thermal hyperalgesia, because the NMDA intradermal injections of BAM8–22 into the nape of
receptor antagonists D -APV [ D -(2 )-2-amino-5- the neck of mice also elicited a profound scratching
phosphonopentanoic acid] and MK-801 [(5S,10R)- behavior (Liu et al., 2009). This behavioral result was
(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5, confirmed by several other studies, the proposed
10-imine] and the nNOS inhibitor N G-nitro-L-arginine murine MRGPRC agonist "SLIGRL" also elicited itch,
methyl ester were found to dose-dependently inhibit and agonist-induced scratching was diminished to
the pronociceptive effects of Tyr6-g 2-MSH6–12 in ;25% in the MRGPR cluster knockout mouse (see
mice (Chang et al., 2009). Because nNOS inhibitors section III.B.1) that lacks MRGPRC (Liu et al., 2009,
also diminished NMDA-induced pain-like behavior 2011; Wilson et al., 2011; Akiyama et al., 2012; Han
(Kitto et al., 1992), it remained unclear whether both et al., 2013). However, intradermal injections into the
players are directly modulated by MRGPRC in the nape of the neck as a secure test for itch were
same cell. questioned because pain-inducing algogens and pruri-
c. Analgesic effects. In contrast to pain- togens elicit nearly indistinguishable reactions after
enhancing effects of MRGPRC, several studies have this application procedure (Shimada and LaMotte,
established analgesic effects of MRGPRC-specific ago- 2008). Instead the “cheek model of itch” was proposed,
nists (Hong et al., 2004; Zeng et al., 2004; Chen et al., which is based on intradermal cheek injections and
2006, 2008, 2012; Cai et al., 2007a; Guan et al., 2010; enables discrimination of itch and pain. Cheek injec-
Jiang et al., 2013). Acute nocifensive behavior after tions of pain-inducing algogens are followed by intense
formalin or NMDA injection was found to be diminished wiping, whereas pruritogens elicit a vigorous scratch-
dose-dependently by BAM1–22 plus naloxone and by ing response. Therefore it is important to note that
BAM8–22 or Tyr6-g 2-MSH6–12 applied alone (Hong BAM8–22 injected into the cheek of mice elicited only
et al., 2004; Zeng et al., 2004; Chen et al., 2006, 2008). In scratching without any wiping reaction (Wilson et al.,
parallel, spinal c-Fos immune reactivity, an established 2011). The notion that murine MRGPRC cause itch
marker of nociception (Bullitt, 1989), was reduced by without pain is further accompanied by the finding
MRGPRC agonists. Thermal hyperalgesia after intra- that ablation of MRGPRA3-positive primary sensory
plantar formalin or CFA injection, quantified either by neurons that almost all express MRGPRC (Zylka et al.,
tail flick or Hargreaves’s tests, were found to be 2003; Liu et al., 2009) completely abolished BAM8–22-
diminished by MRGPRC agonists (Hong et al., 2004; or "SLIGRL"-induced scratching (Han et al., 2013). The
Guan et al., 2010; Jiang et al., 2013). Interestingly, latter study is of particular interest because it showed
CFA-induced thermal hyperalgesia was augmented in that MRGPRA3/MRGPRC-positive neurons are involved
the MRGPR cluster knockout mice (see section III.B.1) in itch, but not pain perception in mice. Noteworthy,
that are deficient for MRGPRC (Guan et al., 2010). activation of MRGPRC by BAM8–22, but not by
Mechanical hyperalgesia, a hallmark of CFA-induced "SLIGRL" induced alloknesis in addition to acute
inflammatory and spinal nerve ligation– or chronic itch, which were both independent of histamine
constriction injury–induced neuropathic pain, was signaling (Akiyama et al., 2012).
also reduced by application of MRGPRC agonists Wilson and coworkers (2011) analyzed downstream
(Cai et al., 2007a; Guan et al., 2010; Chen et al., effectors involved in MRGPRC-induced itching. Al-
2012; Jiang et al., 2013). MRGPRC upregulation and though the percentage of primary sensory neurons that
584 Solinski et al.

elicited BAM8–22-induced calcium signals was dimin- calcium signals via human MRGPRD, although with
ished in TRPV1-deficient cells, action potentials after lower potency compared with b-alanine (Shinohara
BAM8–22 application were not altered. In contrast, et al., 2004; Ajit et al., 2010; Uno et al., 2012).
TRPA1 deficiency resulted in the reduction of both However, because b-alanine and GABA also bind to
responses, calcium signals and action potentials. Ac- other proteins, including GPCR and ion channels
cordingly, BAM8–22-induced scratching responses were (Bowery, 1993; Macdonald and Olsen, 1994; Tiedje
absent only in TRPA1-deficient mice, whereas TRPV1 et al., 2010), in vivo effects of both amino acids would
deficiency was dispensable. have to be interpreted with caution regarding the
molecular receptor entity.
E. MAS-Related G Protein–Coupled Receptors D In addition to amino acids, two peptides produced by
1. Pharmacology. The MRGPRD subfamily consists the renin-angiotensin system have also been identified
of only one receptor subtype per species, which is as MRGPRD ligands. Precisely, angiotensin-(1–7) and
conserved in rodents and primates, and several ligands alamandine [Ala1-angiotensin-(1–7)] were proposed to
have been implicated in MRGPRD pharmacology (see activate MRGPRD (Gembardt et al., 2008; Lautner
Table 5). b-[3H]Alanine is able to bind to the human et al., 2013). Angiotensin-(1–7) is also established to
MRGPRD after heterologous expression in CHO cells bind the MAS1 receptor in vivo (Santos et al., 2003)
(Shinohara et al., 2004). Measuring calcium signals in and is therefore not specific for MRGPRD. Further
these cells, an EC50 value of ;15 mM was determined screening approaches revealed novel exogenous com-
that is three to 4-fold below b-alanine concentrations pounds that specifically activate or inhibit MRGPRD
observed in rat sciatic nerve or cat brain samples activity (Zhang et al., 2007; Ajit et al., 2010; Uno et al.,
(Tallan et al., 1954; Marks et al., 1970). In line with 2012). These specific ligands will be very useful tools
this observation for human MRGPRD, rat or mouse for the analysis of the in vivo effects of MRGPRD in the
MRGPRD also responded to b-alanine after heterolo- future.
gous expression in CHO cells with calcium signals and 2. Signaling Cascades and Physiologic Effects.
EC50 values between 3 and 44 mM (Shinohara et al., Several cellular studies agree that MRGPRD couple to
2004; Zhang et al., 2007; Ajit et al., 2010). b-Alanine Gq/11 and pertussis toxin (PTX)–sensitive Gi/o proteins,
induced signaling in MRGPRD-enriched fractions of because MRGPRD induced PTX-insensitive calcium
primary sensory neurons from rat (Crozier et al., 2007)
releases and PTX-sensitive inhibition of forskolin-
and calcium signals or action potentials in murine
induced adenylyl cyclase activation (Shinohara et al.,
primary sensory neurons (Liu et al., 2012). Thus,
b-alanine has been established as a ligand of MRGPRD 2004; Ajit et al., 2010). Gi/o coupling links MRGPRD to
and could be responsible for their endogenous effects. KCNQ2/3 channels, and inhibition of KCNQ2/3 activity
Interestingly, two other amino acids, GABA and is mainly responsible for the inhibition of noninactivating
b-aminoisobutyric acid, have also been shown to induce potassium currents (M-currents) induced by MRGPRD in

TABLE 5
Pharmacology of MAS-related G protein–coupled receptors D
MRGPRD subfamily members are listed by the ligands to which they have been assigned. All data were generated
after recombinant MRGPRD expression.

Receptor Ligand EC50 Readout System Reference

nM
hMRGPRD b-Alanine 15 calcium CHO Shinohara et al., 2004
4 calcium CHO Ajit et al., 2010
23 calcium HEK293 Uno et al., 2012
220 GTPgS HEK293 Uno et al., 2012
350 IP HEK293 Uno et al., 2012
GABA 191 calcium CHO Shinohara et al., 2004
53 calcium CHO Ajit et al., 2010
4000 calcium HEK293 Uno et al., 2012
BABA 53 calcium HEK293 Uno et al., 2012
820 GTPgS HEK293 Uno et al., 2012
380 IP HEK293 Uno et al., 2012
Angiotensin-(1–7) N. D. arachidonic acid COS Gembardt et al., 2008
Alamandine N. D. nitric oxide CHO Lautner et al., 2013
mMRGPRD b-Alanine 44 calcium CHO Shinohara et al., 2004
rMRGPRD b-Alanine 14 calcium CHO Shinohara et al., 2004
4 calcium CHO Ajit et al., 2010
1 calcium CHO Zhang et al., 2007
GABA 191 calcium CHO Shinohara et al., 2004
50 calcium CHO Ajit et al., 2010
L-Carnosine 34 calcium CHO Shinohara et al., 2004
BABA, b-aminoisobutyric acid; GTPgS, guanosine 59-3-O-(thio)triphosphate; h, human; IP, phosphoinositol pro-
duction; m, mouse; N.D., not determined; r, rat.
MAS-Related G Protein–Coupled Receptors 585

primary sensory neurons (Crozier et al., 2007). This human blood and produces physiologic effects that
signaling cascade was implicated to contribute to resemble those produced by angiotensin-(1–7), includ-
MRGPRD-dependent enhanced neuronal activity ing relaxation of aortic rings. These actions of
(Crozier et al., 2007). Likewise, decreased neuronal alamandine were not mediated via MAS1 or angioten-
activity was detected in MRGPRD-deficient mice (Rau sin II type 2 receptors (AT2R) but required MRGPRD
et al., 2009). activation (Lautner et al., 2013). Surprisingly, the
Just like other MRGPR members, MRGPRD also abovementioned MRGPRD agonist b-alanine did not
specifically mark a discrete subset of primary sensory relax aortic rings, but blocked the effects of alaman-
neurons. Genetically engineered axonal tracers map dine, suggesting that both MRGPRD agonists direct
the stratum granulosum of the skin as the only pe- MRGPRD-induced signaling to different pathways.
ripheral target of MRGPRD-positive neurons (Zylka Effects of MRGPRD in primary sensory neurons and
et al., 2005). MRGPRD neurons form synapses in the the vasculature are summarized in Fig. 6.
spinal lamina II (Zylka et al., 2005) and convey their
signal monosynaptically to almost all known classes of F. MAS-Related G Protein–Coupled Receptors E to -H
spinal lamina II neurons, as evidenced by an optoge- The subfamilies MRGPRE to -H consist of only one
netical circuit mapping approach (Wang and Zylka, receptor per species and are conserved in rodents and
2009). MRGPRD-positive neurons were classified as primates, except for MRGPRH that only exists in
nonpeptidergic C-fiber nociceptors that coexpress the rodents. So far, no member of these MRGPR has ever
ionotropic ATP receptor P2X3, tetrodotoxin-insensitive been assigned to any ligand, which severely hampered
NaV channels, MOP, and partly the TRPV1 ion channel their functional characterization. A MRGPRE-deficient
(Shinohara et al., 2004; Zhang et al., 2005; Zylka et al., mouse strain, which exhibited normal acute pain
2005; Dussor et al., 2008). The functional analysis of responses in a hot plate assay but showed a trend
these cells for sensory transduction revealed a complex toward decreased nocifensive behavior in both phases
picture. MRGPRD-deficient mice exhibit deficits in of the formalin test as well as statistical significant
the detection of mechanical, heat, and cold stimuli, deficits in the induction, but not maintenance, of
whereas ablation of MRGPRD-positive neurons only mechanical allodynia after chronic constriction injury,
impacted acute mechanical pain and mechanical was also established (Cox et al., 2008).
hyperalgesia in a CFA model of inflammatory pain,
whereas heat and cold transduction was completely G. MAS-Related G Protein–Coupled Receptors X
preserved (Cavanaugh et al., 2009; Rau et al., 2009). 1. Pharmacology.
In accordance with the latter finding, ablation of a. MAS-related G protein–coupled receptors X1.
MRGPRD-positive primary sensory neurons reduced The human MRGPRX1 was the first primate-specific
the firing rate and abundance of spinal dorsal horn MRGPR to be assigned to a ligand, and several ad-
neurons responding to mechanical but not heat stimuli ditional agonistic and antagonistic compounds have
(Zhang et al., 2013). Finally, stimulation of MRGPRD- been identified thereafter (see Table 6). MRGPRX1
positive neurons with b-alanine only induced calcium have been shown to bind BAM peptides with high
signals and action potentials in ;40% of genetically affinity (KD value of BAM8–22 ;10 nM) after expres-
labeled MRGPRD-expressing cells (Liu et al., 2012). sion in HEK293 cells (Lembo et al., 2002). Likewise,
This response elicited itch in mice, because itch was BAM peptides elicited intracellular calcium release in
completely abrogated by MRGPRD deficiency. In vivo the same cells (Lembo et al., 2002). In accord with
electrophysiological recordings revealed that b-alanine– rodent MRGPRC, agonistic activity of BAM peptides
activated neurons are heat- and mechanosensitive, was completely preserved in an N-terminal truncated
whereas the remaining heat-insensitive subset of form of BAM1–22 (BAM8–22). EC50 values of either
MRGRPD neurons that did not respond to b-alanine BAM8–22 or BAM1–22 and MRGPRX1 in calcium
was solely mechanosensitive. Thus, the latter pop- assays varied between ;14 and ;800 nM (Lembo
ulation might be responsible for deficits in mechanical et al., 2002; Burstein et al., 2006; Tatemoto et al., 2006;
pain that was observed after ablation of MRGPRD- Shemesh et al., 2008; Malik et al., 2009; Solinski et al.,
positive neurons (Cavanaugh et al., 2009). Interestingly, 2013).
intradermal application of b-alanine during a pilot Despite sharing BAM peptides as agonists, human
human study that comprised 11 individuals also resulted MRGPRX1 and rodent MRGPRC also displayed phar-
in itch, pricking/stinging, and burning (Liu et al., 2012), macological differences (see Fig. 4B). For instance,
indicating that MRGPRD similarly affect itching in hu- proneuropeptide-FF-A cleavage products or dynorphin-
mans and rodents. 14, which are partial agonists of MRGPRC, did not
Transcripts of MRGPRD were also detected in other activate MRGPRX1 at all, and importantly, g 2-MSH,
tissues than in primary sensory neurons, including the most potent and efficacious agonist of rodent
arteries (Shinohara et al., 2004). Moreover, the angio- MRGPRC, only faintly activated MRGPRX1 when
tensinogen cleavage product alamandine circulates in chimeric G proteins were coexpressed (Lembo et al.,
586 Solinski et al.

Fig. 6. Schematic presentation of signaling cascades, downstream targets, and physiologic effects proposed for MRGPRD in primary sensory DRG/
trigeminal ganglia (TG) neurons and blood vessels. b-Alanine–induced activation of MRGPRD leads to activation of Gi/o and Gq/11 proteins. KCNQ2/3
channels that conduct the important background M-current are inhibited by MRGPRD via Gi/o and possibly Gq/11 proteins. MRGPRD have been shown
to elicit pruritogenic sensations and to contribute to normal mechanical and thermal pain thresholds. These distinct actions might be deployed by
MRGPRD signaling in two distinct populations of primary sensory neurons. In blood vessels, MRGPRD are activated by the angiotensinogen
metabolite alamandine. Signaling cascades that are engaged by alamandine include the endothelium-derived production of nitric oxide (NO) that leads
to vasorelaxation.

2002; Solinski et al., 2013). Moreover, cyclic dimers of In addition to potential endogenous agonists, exog-
the C-terminal part of g2-MSH were introduced to enous agonists of MRGPRX1, e.g., tetracyclic benzimi-
antagonize BAM8–22-induced MRGPRX1 activation, dazoles, have been proposed (Malik et al., 2009).
but do not bind rodent MRGPRC (Schmidt et al., 2009). Furthermore, 2,4-diaminopyrimidine derivates and 2,3-
Therefore, the common RF(Y)G/amide consensus of disubstituted azabicyclooctanes antagonize BAM8–22-
MRGPRC agonists does not apply to MRGPRX1. As induced signaling via MRGPRX1 (Kunapuli et al.,
a consequence MRGPRC exhibit a high promiscuity 2006; Bayrakdarian et al., 2011). Interestingly, the
toward many ligands, whereas MRGPRX1 are much MRGPRA3 ligand chloroquine also activated MRGPRX1,
more restrictive and solely bind BAM peptides, a although affinity was 1000-fold and efficacy ;2.5-fold
feature conserved to a certain degree in the MRGPRX1 reduced compared with BAM8–22 (Liu et al., 2009).
of rhesus monkeys (Burstein et al., 2006). Hence, in accordance with its evolutionary relation

TABLE 6
Pharmacology of human MAS-related G protein–coupled receptors X1
The potency of agonists is given as the EC50 value to induce calcium signaling in the given cellular system and the
potency of antagonists is given as the IC50 value to inhibit BAM8–22-induced calcium signaling in the given cellular
system. All data were generated after recombinant MRGPRX1 expression.

EC50 IC50 System Reference

nM
Agonist
BAM8–22 14 HEK293-Gaqi5 Lembo et al., 2002
25 HEK293 Burstein et al., 2006
40 HEK293 Tatemoto et al., 2006
8–50 CHO Kunapuli et al., 2006
150 HEK293 Solinski et al., 2013
BAM1–22 16 HEK293-Gaqi5 Lembo et al., 2002
800 HEK293 Burstein et al., 2006
80 CHO-Ga16 Shemesh et al., 2008
800 HEK293 Malik et al., 2009
Chloroquine 300,000 HEK293 Liu et al., 2009
Benzimidazole derivates 320 HEK293 Malik et al., 2009
Antagonist
Cyclic g 2-MSH6–12 derivates 12 HEK293-Gaqi5 Schmidt et al., 2009
Diaminopyrimidine derivates 14 HEK293-Gaqi5 Bayrakdarian et al., 2011
Azabicyclooctane derivates 100 CHO Kunapuli et al., 2006
MAS-Related G Protein–Coupled Receptors 587

MRGPRX1 share some pharmacological character- also shown to be involved in mast cell activation by
istics with rodent MRGPRC and MRGPRA subfamily a set of structurally similar, endogenous (Tatemoto
members but also display unique pharmacological et al., 2006; Subramanian et al., 2011a, 2013) or
features. exogenous (Kashem et al., 2011; Subramanian et al.,
b. MAS-related G protein–coupled receptors X2. 2011b) basic secretagogues (see section III.G.2.d).
The MRGPRX2 subtype is clearly distinguishable from Finally, in concordance with murine MRGPRC, the
MRGPRX1 regarding its pharmacology (see Table 7), human PAR2-derived "SLIGKV"-peptide was shown to
because MRGPRX2 do not bind BAM peptides activate MRGPRX2 after expression in CHO cells at
(Burstein et al., 2006). However, several other pep- a concentration of 20 mM (Liu et al., 2011).
tidergic ligands have been proposed in different heter- c. MAS-related G protein–coupled receptors X3
ologous or endogenous expression systems (Robas and -4. So far no study reported activation of MRGPRX3
et al., 2003; Kamohara et al., 2005; Burstein et al., and -4 by a given ligand. Therefore, data about signaling
2006; Tatemoto et al., 2006; Shemesh et al., 2008; induced by MRGPRX3 and -4 or their biologic role are
Malik et al., 2009; Kashem et al., 2011; Liu et al., 2011; not available at present. For MRGPRX3 it has been
Subramanian et al., 2011a,b, 2013). The best charac- shown that expression of this receptor subtype under
terized MRGPRX2 ligand is the cortistatin-14 peptide control of the b-actin promoter in rats resulted in
that activated MRGPRX2 with potencies in the increased proliferation of lens fiber cells and keratino-
medium to high nanomolar range (Robas et al., 2003; cytes in basal and suprabasal layers of the skin (Kaisho
Kamohara et al., 2005; Burstein et al., 2006; Shemesh et al., 2005). With regard to the MRGPRX4, a screening
et al., 2008; Malik et al., 2009). Notably, similar to approach using human colorectal cancer cells revealed
MRGPRX1, the ligand binding profile of MRGPRX2 is that the MRGPRX4 protein is one of 15 mutational hot
conserved in rhesus monkeys, because cortistatin-14 spots in these cancer cells (Gylfe et al., 2013). Thus,
also activates MRGPRX2 from this species (Burstein MRGPRX3 and -4 may behave as oncogenes in certain
et al., 2006; Malik et al., 2009). cancers.
MRGPRX2 show a broader expression pattern than 2. Signaling Cascades and Physiologic Effects.
other MRGPRX, because MRGPRX2 expression was a. Preface. MRGPRX1 and -2 function via acti-
detected in primary sensory neurons, several brain vation of G proteins, as indicated by their ability to
areas, mast cells, and the adrenal medulla (Kamohara induce guanosine 59-O-(3-[35S]thio)triphosphate incor-
et al., 2005). In line with this observation, proadreno- poration into plasma membrane fractions (Kamohara
medullin peptides that endogenously occur as side et al., 2005; Burstein et al., 2006; Tatemoto et al.,
products during adrenomedullin synthesis in the 2006). Both receptors further induced the release of
adrenal medulla have also been shown to activate calcium from internal stores via PLCb-mediated pro-
MRGPRX2 (Kamohara et al., 2005). MRGPRX2 were duction of inositol-1,4,5-trisphosphate (Lembo et al.,

TABLE 7
Pharmacology of human MAS-related G protein–coupled receptors X2
Potency of the ligand is given as the EC50 value. The lowest concentration (LC) used to elicit a significant effect when measuring the respective readout is given. Data were
generated after recombinant MRGPRX2 expression, despite when endogenous expression is indicated by “(e)”.

Ligand EC50 LC Readout System Reference

nM
Cortistatin-14 25 calcium HEK293-Ga15 Robas et al., 2003
50 calcium CHO Kamohara et al., 2005
65 cAMP inhibition CHO Kamohara et al., 2005
30 GTPgS CHO Kamohara et al., 2005
2000 calcium HEK293 Burstein et al., 2006
610 calcium CHO-Ga16 Shemesh et al., 2008
500 calcium HEK293 Malik et al., 2009
PAMP1-12 60 calcium CHO Kamohara et al., 2005
40 cAMP inhibition CHO Kamohara et al., 2005
20 GTPgS CHO Kamohara et al., 2005
Substance P 8000 calcium HEK293 Tatemoto et al., 2006
Benzimidazole derivates 400 calcium HEK293 Malik et al., 2009
SLIGKVamide N.D. calcium CHO Liu et al., 2011
PMX-53 100 calcium LAD2 (e), RBL-2H3, human Subramanian et al., 2011b
10 degranulation mast cells (e)
E7 1000 calcium HMC-1 (e), LAD2 (e), human Kashem et al., 2011
10 degranulation mast cells (e)
Substance P 100 degranulation human mast cells (e) Tatemoto et al., 2006
LL-37 100 calcium LAD2 (e), HMC-1, RBL-2H3, Subramanian et al., 2011a
1000 degranulation human mast cells (e)
b-Defensins 100 calcium LAD2 (e), RBL-2H3, HEK293, murine Subramanian et al., 2013
100 degranulation mast cells, human mast cells (e)
GTPgS, guanosine 59-3-O-(thio)triphosphate; N.D., not determined.
588 Solinski et al.

2002; Robas et al., 2003; Breit et al., 2006; Solinski opposing effects. In this context it should be mentioned
et al., 2012). Different groups agree that activation of that distinct effects of MRGPRX1 on neuronal activity
Gq/11 proteins is involved in this process (Lembo et al., were observed in different cell types and that in-
2002; Robas et al., 2003; Kamohara et al., 2005; Breit hibition of neuronal activity apparently depends on Gi/o
et al., 2006; Burstein et al., 2006). However, apart from and increased activity on Gq/11 signaling. Because G
its Gq/11 coupling, additional coupling to PTX-sensitive protein coupling of a given GPCR might differ in
Gi/o proteins was described for MRGPRX1 (Chen and distinct cell types (Hermans, 2003; Kukkonen, 2004),
Ikeda, 2004; Gales et al., 2005; Burstein et al., 2006) or distinct G protein coupling of the MRGPRX1 in dif-
-2 (Kamohara et al., 2005; Subramanian et al., 2013). ferent cells might be the most straightforward expla-
According to their expression pattern, MRGPRX1 nation for its opposing effects on neuronal activity.
and -2 may be involved in somatosensory detector Because of the lack of endogenous model systems,
functions and plasticity of primary sensory neurons effects of MRGPRX1 on neuronal activity have so far
(see sections III.G.2.b and III.G.2.c) and in mast cell only been analyzed after overexpression of the
biology (see section III.G.2.d). However, despite specific MRGPRX1 protein also affecting its G protein coupling
expression, data highlighting MRGPRX2-mediated profile (Kenakin, 1997, 2006). Thus, at this point it is
functions in primary sensory neurons are currently not clear whether MRGPRX1 can act analgetically by
elusive. Moreover, MRGPRX2 may also induce hypo- decreasing neuronal activity under certain conditions
tension and slow-wave sleep (see section III.G.2.e). and algetically by increasing neuronal activity in
Figures 7 and 8 give an overview of effects induced by different circumstances. Alternatively, the receptors
MRGPRX1 or MRGPRX2, respectively. may even induce both effects at the same time, such
b. Somatosensory functions. To shape an organ- that the net effect in a given cell would depend on
ism’s somatosensory performance, a GPCR needs to which of the two signaling pathways dominates the
transfer stimuli detected in the skin into enhanced other. It will be an important task for the future to
or reduced primary sensory neuron activity. Indeed, analyze whether and how MRGPRX1 regulate the
MRGPRX1 have been shown to modulate the activity activity of primary sensory neurons of primates and,
of several distinct ion channels, thereby shaping thus, nociception in vivo. These experiments are dif-
neuronal activity. After heterologous expression in rat ficult to perform, because functional primary sensory
DRG, hippocampal, and superior cervical ganglion neurons from humans are not available and rodents do
neurons, MRGPRX1 inhibited voltage-gated calcium not harbor MRGPRX1-encoding genes. However, ro-
channels, at least partly via PTX-sensitive Gi/o proteins, dent MRGPRC and human MRGPRX1 both bind
leading to decreased synaptic transmission (Chen and BAM8–22, which has been shown to induce pain-
Ikeda, 2004). On the other hand, MRGPRX1 also enhancing, analgesic, and pruritogenic effects in mice
inhibited M-type potassium channels via Gq/11 proteins or rats (see section III.D.2). Interestingly, in a seminal
in the same cells, thereby relieving the neuron from study in which BAM8–22 was applied to the skin of
a background potassium conductance that limits excit- healthy human volunteers using cowhage spicules,
ability (Chen and Ikeda, 2004). In line with this notion, acute itching accompanied by nociceptive sensations
human MRGPRX1 induced action potentials after (pricking, stinging, and burning) was reported (Sikand
heterologous expression in murine DRG neurons (Liu et al., 2011), thus showing that BAM8–22 acts
et al., 2009). The pain-enhancing TRPV1 might be similarly in rodents and humans and that MRGPRX1
involved in MRGPRX1-induced sensory neuron excita- function as somatosensory receptors with nociceptive
tion, inasmuch as MRGPRX1 were shown to sensitize and pruriceptive actions. However, because application
and directly activate this pain sensor (Solinski et al., by cowhage spicules is more likely to elicit itch
2012). Interestingly, MRGPRX1 engaged TRPV1 via two compared with pain (Sikand et al., 2009), more work
distinct signaling pathways: sensitization took place via is needed to determine unequivocally if MRGPRX1 ex-
PKC-mediated phosphorylation of serine residues 502 ert acute functions in pruriceptive and/or nociceptive
and 800 of the channel, whereas direct TRPV1 activa- primary sensory neurons in vivo.
tion relied on diacylglycerol binding to channel regions c. Primary sensory neuron plasticity.
encompassing tyrosine 511 and phosphatidylinositol- Small-diameter primary sensory neurons respond to
3,4-bisphosphate (PIP2) degradation. This multifaceted various acute or chronic stimuli, thereby showing high
modulation of TRPV1 activity is unique among the plastic changes in connectivity, morphology, or re-
GPCR superfamily (Chuang et al., 2001; Prescott and ceptor expression pattern [see Woolf and Ma (2007) for
Julius, 2003; Woo et al., 2008; Kim et al., 2009; Solinski further reading]. Interestingly, pathways that were
et al., 2012) and, thus, points to an important role of the implicated in adaptive responses of primary sensory
TRPV1-MRGPRX1 regulatory axis in somatosensation. neurons are well established players of proliferative
The fact that MRGPRX1 were shown to either signaling in dividing cells. One example is the extra-
dampen or increase neuronal activity raises the cellular signal-regulated kinase (ERK)-1/2 pathway
question about the mechanisms responsible for these that was implicated in adaptive responses of the
MAS-Related G Protein–Coupled Receptors 589

Fig. 7. Schematic presentation of signaling cascades, downstream targets, and physiologic effects proposed for human MRGPRX1 in primary sensory
DRG/trigeminal ganglia (TG) neurons and mast cells. (A) Application of BAM8–22 into human skin elicits histamine-independent pruriceptive and
nociceptive sensations. Recombinant human MRGPRX1 inhibit voltage-dependent calcium channels via Gi/o proteins and M-current-conducting
KCNQ2/3 channels via Gq/11 proteins. TRPV1 activity is enhanced by recombinant MRGPRX1 through PKC, diacylglycerol (DAG), and PIP2. (B) Effects
of sustained MRGPRX1 activation in recombinant cell systems include ERK-1/2 and NFAT activation, which induces c-Fos, early growth response
protein 1 (EGR-1), and CCR2 expression. In a human mast cell line endogenously expressing MRGPRX1, BAM8–22 enhanced the release of the CCR2
agonist chemokine ligand 2 (CCL2).

nociceptive system to chronic inflammation (Ko et al., induced activation of ERK-1/2 and serum response
2005; Wang et al., 2006a; Utreras et al., 2009; Su et al., element–dependent transcription as well as ERK-1/2–
2010; Romero et al., 2012). dependent expression of the immediate early genes
Several indications point to the engagement of c-Fos and early growth response protein 1 (Solinski
nuclear signaling pathways by MRGPRX, leading to et al., 2013). Because of the published pathophysiologic
transcriptional modulation. Overexpression of MRGPRX1 importance of this pathway in rodents and humans (Ko
or -4 increased proliferation of 3T3 fibroblasts, et al., 2005; Wang et al., 2006a; Utreras et al., 2009; Su
highlighting considerable constitutive activity of these et al., 2010; Romero et al., 2012), MRGPRX1 may
MRGPRX after recombinant expression (Burstein et al., represent interesting targets in chronic inflammatory
2006). We recently found that after expression in pain if this pathway is also activated by MRGPRX1 in
primary sensory neuron–derived F11 cells, MRGPRX1 vivo as well.
590 Solinski et al.

Fig. 8. Schematic presentation of signaling cascades, down-stream targets, and physiologic effects proposed for human MRGPRX2 in mast cells and
potential effects in chromaffin cells and hippocampal neurons. (A) Several agonists have been implicated in mast cell degranulation via activation of
MRGPRX2 including the C5a receptor antagonist PMX-53, the C3a receptor super-agonist E7, substance P (SP), the cathelicidin LL-37, and
b-defensins. PTX-sensitive Gi/o proteins via PKC and PLCb-activating G proteins, presumably Gq/11, elicit IgE-independent degranulation of connective
tissue type mast cells. (B) MRGPRX2 have been proposed to cause hypotensive and slow-wave sleep inducing effects. Hypotension might be mediated
via adrenomedullary PAMP1–20-induced inhibition of (nor)epinephrine release. Sleep phase modulation is conveyed by cortistatin-14 (COR-14) and
potentially engages MRGPRX2-induced reduction of hippocampus excitability.

It is interesting to mention that MRGPRX1 not only BAM8–22-induced NFAT activation led to increased
signal to the nucleus via classic proliferative pathways expression of chemokine receptors 2 (CCR2) in primary
but also induced transcriptional alterations in primary sensory neurons after recombinant MRGPRX1 expres-
sensory neurons via the calcineurin-dependent tran- sion (Solinski et al., 2013). CCR2 were consistently
scription factor nuclear factor of activated T cells found to be upregulated in primary sensory neurons
(NFAT) (Solinski et al., 2013). Activation of NFAT under neuropathic conditions and to cause pain chron-
was already implicated in several plastic changes of ification (Abbadie et al., 2003; Sun et al., 2006; Bhangoo
primary sensory neurons, for instance axonal growth et al., 2007, 2009; White et al., 2007; Jung et al., 2009;
(Graef et al., 2003; Nguyen and Di Giovanni, 2008) and Fu et al., 2010; Serrano et al., 2010; Wang et al., 2010).
activity-dependent transcription (Groth et al., 2007; Thus, MRGPRX1 may also be interesting targets in
Jackson et al., 2007; Jung and Miller, 2008). Moreover, chronic neuropathic pain.
MAS-Related G Protein–Coupled Receptors 591

d. Mast cell biology. Mast cells are long-lived neuroimmune interactions. In congruence with this
mononuclear cells that reside in tissues near external concept, MRGPRX1 were implicated in the secretion of
surfaces, e.g., in skin or mucosa, and thereby are chemokine ligand 2 from LAD2 mast cells after
among the first cells of the immune system to respond stimulation with BAM8–22 (Solinski et al., 2013).
to pathogens or allergens (Galli et al., 2011). Mast cells Given that MRGPRX1 induced the expression of the
fulfill their tasks in innate and adaptive immune chemokine ligand 2 receptor CCR2 in primary sensory
responses by secretion of a plethora of mediators neurons (Solinski et al., 2013), it is appealing to
(degranulation), invaluable for host defense (Galli hypothesize that MRGPRX1 could additionally en-
et al., 2011). Mast cell degranulation is caused by hance mast-cell-to-neuron signaling. Because mast
IgE-dependent and -independent processes (Metcalfe cells contribute to chronic pain (Zuo et al., 2003),
et al., 1997; Ferry et al., 2002). A very heterogeneous inhibition of such a paracrine signaling circuit
group of agents, either released endogenously by would be an interesting approach for future analgesic
immune cells, neurons, and epithelial cells, or applied therapy.
exogenously, mediates IgE-independent mast cell de- e. Putative role of MAS-related G protein–coupled
granulation and is referred to as secretagogues (Sharma receptors X2 in sleep and blood pressure regulation.
et al., 2002). However, underlying signaling pathways PAMP1-20 is a potent hypotensive peptide, functioning
are diverse and a matter of debate. mainly via inhibition of norepinephrine and epineph-
Endogenously expressed MRGPRX2 were proposed rine release from sympathetic neurons or adrenal
as receptors for different secretagogues in cultured chromaffin cells, respectively (Shimosawa et al., 1995;
human primary mast cells and in the differentiated Kobayashi et al., 2001). On the protein level, PAMP1-
human mast cell line LAD2 (Tatemoto et al., 2006; 20 is mainly distributed in adrenal medulla and atrium
Kashem et al., 2011; Subramanian et al., 2011a,b, (Washimine et al., 1994). Because MRGPRX2 proteins
2013). Early during infection, a first line of mast cell have been detected in adrenal chromaffin cells and
activation is thought to arise from complement- because PAMP1-20 activated MRGPRX2 in heterolo-
mediated anaphylatoxin production (e.g., C3a or C5a) gous expression systems, Kamohara and coworkers
(Kashem et al., 2011; Subramanian et al., 2011b).
(2005) proposed that epinephrine-dependent hypoten-
Although MRGPRX2 are not involved in mast cell
sive functions of PAMP1-20 might be mediated by
activation provoked by endogenous anaphylatoxins,
MRGPRX2 signaling.
mast cell degranulation induced by a synthetic C5a
Cortistatin-14 activates all five somatostatin receptor
receptor antagonist (PMX-53) or a C3a receptor super-
(SSTR1-5) subtypes, but some effects of cortistatin-14
agonist (E7) required MRGPRX2 expression (Kashem
appear to be clearly independent of these receptors and
et al., 2011; Subramanian et al., 2011b). Later on
are not caused by somatostatin (de Lecea, 2008). Among
during infection, epithelial cells can secrete b-defensins
these effects is the induction of slow-wave sleep that
or cathelicidins, both inducing mast cell degranulation
might be partly mediated by inhibition of hippocampal
via MRGPRX2 (Subramanian et al., 2011a, 2013).
neurons. Because MRGPRX2 are expressed in hippo-
Hence, MRGPRX2 can integrate paracrine input from
campal neurons of layer CA2-4 and because MRGPRX2
various cell types by detecting alterations of the local
responded specifically to cortistatin-14, Robas and co-
milieu and inducing mast cell degranulation.
workers (2003) put forward the proposal that MRGPRX2
Degranulation of mast cells after MRGPRX2 activa-
may be involved in cortistatin-14–mediated slow-wave
tion depends on PTX-sensitive G proteins, irrespective
sleep induction.
of the MRGPRX2 ligand under investigation (Tatemoto
et al., 2006; Kashem et al., 2011; Subramanian et al.,
2011a,b, 2013). However, MRGPRX2 activation by
IV. Agonist-Promoted Internalization and
LL-37 or human b-defensin-3 involves two different
Desensitization of MAS-Related
signaling cascades that act synergistically on degran-
G Protein–Coupled Receptors
ulation, but only one of which depends on Gi/o-induced
PKC activation (Subramanian et al., 2011a, 2013). The GPCR signaling is commonly regulated by agonist-
second PTX-insensitive pathway involved calcium in- induced receptor phosphorylation that leads to uncou-
flux and release, as suggested by the inhibitory actions pling from cognate G proteins (desensitization) and
of the calcium release–activated calcium channel subsequent receptor internalization (endocytosis), mostly
blocker, lathanium (La 3+), and the inositol 1,4,5- via association with b-arrestins (Ferguson, 2001). In
trisphosphate receptor and canonical TRP blocker, addition, b-arrestins have been shown to induce
2-aminoethoxydiphenyl borate (Subramanian et al., G protein-independent signaling pathways, further
2011a, 2013). increasing the repertoire of GPCR-mediated signaling
Because some basic secretagogues can be secreted by (Shenoy and Lefkowitz, 2003). Only a few GPCR have
primary sensory neurons, MRGPRX2 may also en- been shown to resist agonist-promoted processes that
hance neuron-to-mast cell signaling, thereby shaping negatively regulate receptor activity, including k-OR,
592 Solinski et al.

b3-adrenoceptors, and SSTR4 (Nantel et al., 1993; Chu more than one endocytosis resistant member. Whether
et al., 1997; Csaba and Dournaud, 2001). MRGPRX3 and -4 also resist agonist-mediated de-
Detailed analysis of the subcellular distribution of sensitization still needs to be confirmed in the future.
human MRGPRD fused to green fluorescent protein Notably, serine residues in the C-terminal tail of a given
(GFP) in CHO cells after a 30-minute period of GPCR are profoundly involved in agonist-induced re-
stimulation with saturating concentrations of b-alanine ceptor regulation (Ferguson, 2001) and, as already
(Shinohara et al., 2004) revealed translocation of the mentioned, C-terminal tails of MRGPRX developed
receptor protein from the plasma membrane to punctate under strong positive selection pressure during evo-
intracellular vesicles, indicative of ligand-induced lution. It is tempting to speculate that considerable
internalization. Dose-dependent internalization of physiologic benefits have been earned by enriching
c-myc–tagged rat MRGPRD by b-alanine after recombi- the lack of receptor desensitization and endocytosis
nant expression in HEK293 cells was also identified, among MRGPRX. A summary of the available litera-
using either a qualitative immunofluorescence or a ture analyzing ligand-induced regulation of MRGPR
quantitative enzyme-linked immunosorbent assay ap- is given in Table 8.
proach (Milasta et al., 2006). Thus, MRGPRD, the only
MRGPR conserved in rodents and primates and as-
signed to an agonist, apparently belongs to the group of V. Functional Interactions of MAS-Related
endocytosis-prone GPCR. G Protein–Coupled Receptors with Other
After recombinant expression of murine MRGPRA1 Receptor Families
or MRGPRA4 in HEK293 cells, Dong and coworkers
(2001) observed a prominent decline in ligand-induced A. Heteromultimerization among MAS-Related
calcium signals after repetitive short term stimulation G Protein–Coupled Receptors and Receptor Subtypes
with FLRFamide or NPAF, indicating agonist- of Other GPCR Families
promoted desensitization. Desensitization was revers- Heteromultimerization among GPCR has been pro-
ible by ligand wash-out over ;20 minutes and most posed to alter ligand binding, G protein coupling,
likely occurred because of phosphorylation and endo- protein trafficking, and agonist-promoted desensitiza-
cytosis of the respective receptor protein. In fact, in tion of the receptors involved (Bulenger et al., 2005).
HEK293 cells murine MRGPRA1 fused to GFP was Thus, direct protein-protein interactions due to re-
internalized to punctate intracellular vesicles after 30 ceptor multimerization represent an important mech-
minutes of agonist stimulation (Han et al., 2002). anism by which distinct hormone systems interact with
Likewise, MRGPRC-GFP fusion proteins also in- each other (Jordan et al., 2001; Torvinen et al., 2005;
ternalized after 30 minutes of stimulation by saturat- Parenty et al., 2008). Because MRGPRX1 and OR
ing concentrations of g2-MSH in HEK293 cells (Han share a common ligand, the BAM1–22 peptide, are
et al., 2002). This work was corroborated by the finding coexpressed in primary sensory neurons, and both
that 1) other full agonists of murine MRGPRC in- affect pain modulation, functional interactions be-
cluding BAM8–22 are able to induce the same extent of tween MRGPRX1 and the d-OR (DOP) due to multi-
MRGPRC endocytosis; 2) endocytosis is not confined to merization have been analyzed. Bioluminescence
HEK293 cells, but also detectable in several DRG resonance energy transfer experiments revealed that
neuron-derived cell lines; 3) b-arrestins are indispens- MRGPRX1 interacted with human DOP in living
able for proper ligand-induced MRGPRC endocytosis in HEK293 cells stably overexpressing each receptor
Cos7 cells (Solinski et al., 2010). Notably, rat MRGPRC (Breit et al., 2006). Individual activation of either
was also found to be sensitive to BAM8–22- or DOP (with Leu-Enkephalin) or MRGPRX1 (with
g2-MSH–induced endocytosis in the same cell models BAM8–22) allowed both receptors to modulate their
(Solinski et al., 2010). In sharp contrast to rodent respective signaling pathways. In contrast, the DOP/
BAM8–22-sensitive MRGPRC, human MRGPRX1 MRGPRX1 bivalent agonist BAM1–22, which activated
resisted BAM8–22-induced desensitization and endo- each receptor expressed individually, fully activated
cytosis after recombinant expression in different the MRGPRX1 but did not promote DOP-mediated
cellular systems, including primary sensory neuron- signaling within the heteromultimer. Similarly, concom-
derived F11 and ND-C cells (Solinski et al., 2010). itant activation of the DOP/MRGPRX1 heteromultimer
Thus, identical ligands regulate MRGPRC and MRGPRX1 by selective DOP and MRGPRX1 agonists (Leu-
endocytosis in diametrically opposed ways. Moreover, Enkephalin and BAM8–22) promoted MRGPRX1 but
similar to MRGPRX1, ligand-induced phosphorylation not DOP signaling. Furthermore, coexpression of the
and internalization of the MRGPRX2 was not detectable endocytosis resistant MRGPRX1 inhibited agonist-
(Subramanian et al., 2011a). Thus, absence of agonist- promoted internalization of the endocytosis-prone
promoted receptor desensitization appears as a common DOP. Overall, these data suggest that MRGPRX1
feature among MRGPRX, defining the MRGPRX sub- signaling within the DOP/MRGPRX1 heteromultimer
family as the first among all GPCR families to harbor acts in a dominant-negative fashion on DOP signaling
MAS-Related G Protein–Coupled Receptors 593
TABLE 8
Ligand-promoted desensitization and endocytosis of MAS-related G protein–coupled receptors
MRGPR that have been analyzed for their ligand-induced regulation are listed in conjunction with the respective ligand and expression system
used.

Receptor Ligand Desensitization Endocytosis System Reference

mMRGPRA1 FLRFamide YES YES HEK293 Dong et al., 2001


Han et al., 2002
mMRGPRA4 NPAF YES N.D. HEK293 Dong et al., 2001
mMRGPRC g2-MSH YES YES HEK293, F11, ND-C, Cos7 Han et al., 2002
BAM8–22 Solinski et al., 2010
Dyn-14
rMRGPRC g2-MSH BAM8–22 N.D. YES HEK293, F11, ND-C Solinski et al., 2010
hMRGPRD b-Alanine N.D. YES CHO Shinohara et al., 2004
rMRGPRD b-Alanine N.D. YES HEK293 Milasta et al., 2006
hMRGPRX1 BAM8–-22 NO NO HEK293, F11, ND-C, Cos7 Solinski et al., 2010
hMRGPRX2 Cortistatin-14 NO NO HEK293 Subramanian et al., 2011a
LL-37 LAD2, HMC-1
h, human; m, mouse; N.D., not determined; r, rat.

and thus, could affect the fine tuning of pain sensation BAM-sensitive MRGPRC and opioid receptors have
regulated by DOP. also been described in vivo. A major problem of chronic
MRGPR have not only been shown to interact with pain treatment with morphine is the development of
members of other GPCR families, but also among each tolerance characterized by declining analgesic effects
other. The rat MRGPRD and -E subtypes form multi- after repeated dosing. The mechanisms that cause the
meric complexes when expressed in HEK293 cells or underlying adaptation processes are manifold and
peripheral nociceptive neurons, as previously indicated not completely understood [for further reading see
by coimmunoprecipitation and time-resolved fluores- Williams et al. (2013)]. In rats, tolerance to morphine can
cence resonance energy transfer (Milasta et al., 2006). be generated by its daily application over six to seven
These interactions increased the potency of b-alanine days. Interestingly, after intrathecal application of
to phosphorylate ERK-1/2, maintained the capacity of MRGPRC-specific agonists in rats, morphine regained
b-alanine to elevate intracellular calcium concentra- ;50% of its analgesic actions in formerly morphine-
tions, and inhibited b-alanine–induced internalization tolerant animals on the consecutive day (Jiang et al.,
of MRGPRD. Given the large number of MRGPR 2006; Cai et al., 2007b; Chen et al., 2010b). MRGPRC
family members and assuming that heteromultimeric agonists were not only able to restore analgesic efficacy
interactions also exist between other subtypes, one after tolerance to morphine had already been estab-
would predict an immense variety of putative binding lished, intrathecally applied BAM8–22 or Tyr6-g 2-
sites for MRGPR-selective ligands, expanding the MSH6–12 on every second day during induction of
impact of this receptor family on many physiologic morphine tolerance dose-dependently inhibited the de-
pathways even further. velopment of morphine tolerance (Cai et al., 2007b; Chen
et al., 2010b). Compensatory upregulation of pain-
B. Inhibition of Tolerance to Morphine enhancing molecular players in the DRG and spinal
As mentioned above, BAM-sensitive, human cord including PKCg-dependent upregulation of nNOS
MRGPRX1 functionally interact with the opioid system or CGRP is involved in the development of morphine
based on receptor multimerization. Interestingly, tolerance (Menard et al., 1996; Kolesnikov et al., 1997;
multimerization-independent interactions between Granados-Soto et al., 2000). Because MRGPRC agonists

TABLE 9
Ligand sharing between MAS-related G protein–coupled receptors and members of other GPCR families
Ligands that have been implicated to bind and activate both, MRGPR subtypes and other GPCR, are given.

Ligand MRGPR Reference Other GPCR Reference

BAM1–22 r/mMRGPRC hMRGPRX1 Lembo et al., 2002 DOP, MOP, KOP Quirion and Weiss,1983
Han et al., 2002
Grazzini et al., 2004
NPFF mMRGPRC Han et al., 2002 NPFF1R, NPFF2R Mollereau et al., 2002
g 2-MSH r/mMRGPRC Han et al., 2002 MC3R, MC4R Roselli-Rehfuss et al., 1993
Grazzini et al., 2004 Oosterom et al., 1999
GABA h/rMRGPRD Shinohara et al., 2004 GABAB Bowery et al., 1980
Kaupmann et al., 1997
Angiotensin-(1–7) hMRGPRD Gembardt, 2008 MAS1, AT2R Santos et al., 2003
Walters, 2005
Cortistatin-14 hMRGPRX2 Robas, 2003 SSTR1-5 de Lecea et al., 1996
Fukusumi et al., 1997
h, human; KOP, k-OR; m, mouse; MC3R, melanocortin 3 receptor; r, rat.
594 Solinski et al.

blocked enhanced PKCg activity in the spinal cord and simplify the development of drugs tested in established
diminished nNOS and CGRP immune reactivity in DRG animal models but intended to target human
and spinal cord slices, this signaling cascade might MRGPRX1. In parallel, it would be of interest to
inhibit morphine tolerance by MRGPRC (Chen et al., develop primate- or even human-based DRG-derived
2010b). cell models that endogenously express MRGPRX1.
Considering mast cells, a step forward has been made
at this point, because the human mast cell line LAD2
VI. Conclusions and Perspectives
has been shown to endogenously express MRGPRX1
Over the last decade, our knowledge about MRGPR and -2. Furthermore, the problem of ligand-sharing
has been significantly extended: 1) the murine between MRGPR and subtypes of other GPCR families,
MRGPRA3 subtype has been identified as the itch- observed for many endogenous ligands (see Table 9), is
mediating chloroquine receptor; 2) rodent MRGPRC a major drawback in the field. Thus, screening efforts
have also been shown to be involved in itching and yielding MRGPR subtype–specific agonists and antag-
MRGPRC have been made responsible for analgesic onists would significantly help attain a better un-
and pain-enhancing effects; 3) MRGPRD enhance derstanding of the physiologic pathways engaged by
neuronal excitability and induce vasorelaxation; and MRGPR in vivo.
4) human MRGPRX also modulate excitability of
primary sensory neurons and gene expression in these Acknowledgments
cells. However, because of the lack of suitable model The authors thank Dr. Alexander Dietrich (Walther-Straub-
systems, these data are solely based on experiments Institut für Pharmakologie und Toxikologie, Ludwig-Maximilians-
Universität München, Munich, Germany) for critical reading of the
obtained after heterologous expression of the MRGPRX1
manuscript.
protein. In mast cells, endogenously expressing the
MRGPRX1 and -2 subtype effects on degranulation and
Authorship Contributions
mediator release have been found. Adaptive evolu-
Wrote or contributed to the writing of the manuscript: Solinski,
tionary development and resistance to agonist- Gudermann, Breit.
promoted desensitization/endocytosis further highlight
the extraordinary features of the primate-specific References
MRGPRX subfamily. Notably, MRGPRX can be consid- Abbadie C, Lindia JA, Cumiskey AM, Peterson LB, Mudgett JS, Bayne EK,
ered as valuable drug targets with low or even no off- DeMartino JA, MacIntyre DE, and Forrest MJ (2003) Impaired neuropathic pain
responses in mice lacking the chemokine receptor CCR2. Proc Natl Acad Sci USA
target effects because of their restricted expression in 100:7947–7952.
primary sensory neurons and mast cells. Ajit SK, Pausch MH, Kennedy JD, and Kaftan EJ (2010) Development of a FLIPR
assay for the simultaneous identification of MrgD agonists and antagonists from
However, our knowledge about the MRGPR family is a single screen. J Biomed Biotechnol 2010:2010.
Akiyama T, Carstens MI, Ikoma A, Cevikbas F, Steinhoff M, and Carstens E (2012)
still superficial and central questions need to be Mouse model of touch-evoked itch (alloknesis). J Invest Dermatol 132:1886–1891.
addressed: 1) to what extent do MRGPR serve as Avula LR, Buckinx R, Alpaerts K, Costagliola A, Adriaensen D, Van Nassauw L,
and Timmermans JP (2011) The effect of inflammation on the expression and
markers for primary sensory neurons that execute distribution of the MAS-related gene receptors MrgE and MrgF in the murine
specific functions; 2) what is the difference between ileum. Histochem Cell Biol 136:569–585.
Bayrakdarian M, Butterworth J, Hu YJ, Santhakumar V, and Tomaszewski MJ
distinct rodent species in this regard; 3) what is the (2011) Development of 2,4-diaminopyrimidine derivatives as novel SNSR4 antag-
onists. Bioorg Med Chem Lett 21:2102–2105.
physiologic role of distinct MRGPR in these specialized Bender E, Buist A, Jurzak M, Langlois X, Baggerman G, Verhasselt P, Ercken M,
neurons? Consider, in particular, that some subtypes of Guo HQ, Wintmolders C, and Van den Wyngaert I, et al. (2002) Characterization of
an orphan G protein-coupled receptor localized in the dorsal root ganglia reveals
the MRGPRA and -X subfamilies are not yet analyzed, adenine as a signaling molecule. Proc Natl Acad Sci USA 99:8573–8578.
mainly because not a single ligand for these receptors Bhangoo S, Ren D, Miller RJ, Henry KJ, Lineswala J, Hamdouchi C, Li B, Monahan
PE, Chan DM, and Ripsch MS, et al. (2007) Delayed functional expression of
is available. Similarly, progress in the work with neuronal chemokine receptors following focal nerve demyelination in the rat:
a mechanism for the development of chronic sensitization of peripheral nociceptors.
MRGPRE to -H receptors is significantly hindered by Mol Pain 3:38.
the lack of agonists. Moreover, it will be essential to Bhangoo SK, Ripsch MS, Buchanan DJ, Miller RJ, and White FA (2009) Increased
chemokine signaling in a model of HIV1-associated peripheral neuropathy. Mol
define the signaling events responsible for the effects of Pain 5:48.
MRGPRC agonists on nociception. Interestingly, hu- Bowery NG (1993) GABAB receptor pharmacology. Annu Rev Pharmacol Toxicol 33:
109–147.
man MRGPRX1 have been shown to enhance or inhibit Bowery NG, Hill DR, Hudson AL, Doble A, Middlemiss DN, Shaw J, and Turnbull M
(1980) (-)Baclofen decreases neurotransmitter release in the mammalian CNS by
signaling pathways that supposedly modify neuronal an action at a novel GABA receptor. Nature 283:92–94.
activity, suggesting that MRGPRX1 might also exert Breit A, Gagnidze K, Devi LA, Lagacé M, and Bouvier M (2006) Simultaneous acti-
vation of the delta opioid receptor (deltaOR)/sensory neuron-specific receptor-4
proalgetic and analgesic effects in humans. In this (SNSR-4) hetero-oligomer by the mixed bivalent agonist bovine adrenal medulla
context it will be important to analyze to which extent peptide 22 activates SNSR-4 but inhibits deltaOR signaling. Mol Pharmacol 70:
686–696.
data obtained from BAM8–22-sensitive rodent MRGPRC Bulenger S, Marullo S, and Bouvier M (2005) Emerging role of homo- and hetero-
dimerization in G-protein-coupled receptor biosynthesis and maturation. Trends
can be extended to BAM8–22-sensitive human MRGPRX1. Pharmacol Sci 26:131–137.
Convincing evidence that rodents can in fact be used Bullitt E (1989) Induction of c-fos-like protein within the lumbar spinal cord and
thalamus of the rat following peripheral stimulation. Brain Res 493:391–397.
as a native model systems for human BAM8–22- Burstein ES, Ott TR, Feddock M, Ma JN, Fuhs S, Wong S, Schiffer HH, Brann MR,
sensitive MRGPRX1 would propel the "deorphaniza- and Nash NR (2006) Characterization of the Mas-related gene family: structural
and functional conservation of human and rhesus MrgX receptors. Br J Pharmacol
tion" process of these receptors forward and would 147:73–82.
MAS-Related G Protein–Coupled Receptors 595
Cai M, Chen T, Quirion R, and Hong Y (2007a) The involvement of spinal bovine Galli SJ, Borregaard N, and Wynn TA (2011) Phenotypic and functional plasticity of
adrenal medulla 22-like peptide, the proenkephalin derivative, in modulation of cells of innate immunity: macrophages, mast cells and neutrophils. Nat Immunol
nociceptive processing. Eur J Neurosci 26:1128–1138. 12:1035–1044.
Cai Q, Jiang J, Chen T, and Hong Y (2007b) Sensory neuron-specific receptor agonist Gembardt F, Grajewski S, Vahl M, Schultheiss HP, and Walther T (2008) Angio-
BAM8-22 inhibits the development and expression of tolerance to morphine in rats. tensin metabolites can stimulate receptors of the Mas-related genes family. Mol
Behav Brain Res 178:154–159. Cell Biochem 319:115–123.
Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, and Julius D Graef IA, Wang F, Charron F, Chen L, Neilson J, Tessier-Lavigne M, and Crabtree
(1997) The capsaicin receptor: a heat-activated ion channel in the pain pathway. GR (2003) Neurotrophins and netrins require calcineurin/NFAT signaling to
Nature 389:816–824. stimulate outgrowth of embryonic axons. Cell 113:657–670.
Cavanaugh DJ, Lee H, Lo L, Shields SD, Zylka MJ, Basbaum AI, and Anderson DJ Granados-Soto V, Kalcheva I, Hua X, Newton A, and Yaksh TL (2000) Spinal PKC
(2009) Distinct subsets of unmyelinated primary sensory fibers mediate behavioral activity and expression: role in tolerance produced by continuous spinal morphine
responses to noxious thermal and mechanical stimuli. Proc Natl Acad Sci USA 106: infusion. Pain 85:395–404.
9075–9080. Grazzini E, Puma C, Roy MO, Yu XH, O’Donnell D, Schmidt R, Dautrey S, Ducharme
Chang M, Li W, Peng YL, Gao YH, Yao J, Han RW, and Wang R (2009) Involvement J, Perkins M, and Panetta R, et al. (2004) Sensory neuron-specific receptor acti-
of NMDA receptor in nociceptive effects elicited by intrathecal [Tyr6] gamma2- vation elicits central and peripheral nociceptive effects in rats. Proc Natl Acad Sci
MSH(6-12), and the interaction with nociceptin/orphanin FQ in pain modulation in USA 101:7175–7180.
mice. Brain Res 1271:36–48. Groth RD, Coicou LG, Mermelstein PG, and Seybold VS (2007) Neurotrophin acti-
Chen H and Ikeda SR (2004) Modulation of ion channels and synaptic transmission vation of NFAT-dependent transcription contributes to the regulation of pro-
by a human sensory neuron-specific G-protein-coupled receptor, SNSR4/mrgX1, nociceptive genes. J Neurochem 102:1162–1174.
heterologously expressed in cultured rat neurons. J Neurosci 24:5044–5053. Guan Y, Liu Q, Tang Z, Raja SN, Anderson DJ, and Dong X (2010) Mas-related
Chen M, Tominaga K, and Pereira-Smith OM (2010a) Emerging role of the MORF/ G-protein-coupled receptors inhibit pathological pain in mice. Proc Natl Acad Sci
MRG gene family in various biological processes, including aging. Ann N Y Acad USA 107:15933–15938.
Sci 1197:134–141. Gylfe AE, Kondelin J, Turunen M, Ristolainen H, Katainen R, Pitkänen E, Kaasinen
Chen P, Wang D, Li M, Zhang Y, Quirion R, and Hong Y (2010b) Modulation of E, Rantanen V, Tanskanen T, and Varjosalo M, et al. (2013) Identification of
sensory neuron-specific receptors in the development of morphine tolerance and its candidate oncogenes in human colorectal cancers with microsatellite instability.
neurochemical mechanisms. J Neurosci Res 88:2952–2963. Gastroenterology 145:540–543.
Chen T, Cai Q, and Hong Y (2006) Intrathecal sensory neuron-specific receptor Hager UA, Hein A, Lennerz JK, Zimmermann K, Neuhuber WL, and Reeh PW (2008)
agonists bovine adrenal medulla 8-22 and (Tyr6)-gamma2-MSH-6-12 inhibit Morphological characterization of rat Mas-related G-protein-coupled receptor C
formalin-evoked nociception and neuronal Fos-like immunoreactivity in the spinal and functional analysis of agonists. Neuroscience 151:242–254.
cord of the rat. Neuroscience 141:965–975. Han KH, Ryu J, Hong KH, Ko J, Pak YK, Kim JB, Park SW, and Kim JJ (2005) HMG-
Chen T, Hu Z, Quirion R, and Hong Y (2008) Modulation of NMDA receptors by CoA reductase inhibition reduces monocyte CC chemokine receptor 2 expression
intrathecal administration of the sensory neuron-specific receptor agonist BAM8- and monocyte chemoattractant protein-1-mediated monocyte recruitment in vivo.
22. Neuropharmacology 54:796–803. Circulation 111:1439–1447.
Chen T, Jiang J, Huang H, Wang D, Liu Y, and Hong Y (2012) Role of bovine adrenal Han L, Ma C, Liu Q, Weng HJ, Cui Y, Tang Z, Kim Y, Nie H, Qu L, and Patel KN,
medulla 22 (BAM22) in the pathogenesis of neuropathic pain in rats with spinal et al. (2013) A subpopulation of nociceptors specifically linked to itch. Nat Neurosci
nerve ligation. Eur J Pharmacol 685:24–29. 16:174–182.
Choi SS and Lahn BT (2003) Adaptive evolution of MRG, a neuron-specific gene Han SK, Dong X, Hwang JI, Zylka MJ, Anderson DJ, and Simon MI (2002) Orphan G
family implicated in nociception. Genome Res 13:2252–2259. protein-coupled receptors MrgA1 and MrgC11 are distinctively activated by RF-
Chu P, Murray S, Lissin D, and von Zastrow M (1997) Delta and kappa opioid amide-related peptides through the Galpha q/11 pathway. Proc Natl Acad Sci USA
receptors are differentially regulated by dynamin-dependent endocytosis when 99:14740–14745.
activated by the same alkaloid agonist. J Biol Chem 272:27124–27130. Hao L, Ge X, Wan H, Hu S, Lercher MJ, Yu J, and Chen WH (2010) Human func-
Chuang HH, Prescott ED, Kong H, Shields S, Jordt SE, Basbaum AI, Chao MV, tional genetic studies are biased against the medically most relevant primate-
and Julius D (2001) Bradykinin and nerve growth factor release the capsaicin specific genes. BMC Evol Biol 10:316–323.
receptor from PtdIns(4,5)P2-mediated inhibition. Nature 411:957–962. Heo J, Han SK, Vaidehi N, Wendel J, Kekenes-Huskey P, and Goddard WA 3rd
Cox PJ, Pitcher T, Trim SA, Bell CH, Qin W, and Kinloch RA (2008) The effect of (2007a) Prediction of the 3D structure of FMRF-amide neuropeptides bound to the
deletion of the orphan G - protein coupled receptor (GPCR) gene MrgE on pain-like mouse MrgC11 GPCR and experimental validation. ChemBioChem 8:1527–1539.
behaviours in mice. Mol Pain 4:2. Heo J, Vaidehi N, Wendel J, and Goddard WA 3rd (2007b) Prediction of the 3-D
Crozier RA, Ajit SK, Kaftan EJ, and Pausch MH (2007) MrgD activation inhibits structure of rat MrgA G protein-coupled receptor and identification of its binding
KCNQ/M-currents and contributes to enhanced neuronal excitability. J Neurosci site. J Mol Graph Model 26:800–812.
27:4492–4496. Hermans E (2003) Biochemical and pharmacological control of the multiplicity of
Csaba Z and Dournaud P (2001) Cellular biology of somatostatin receptors. Neuro- coupling at G-protein-coupled receptors. Pharmacol Ther 99:25–44.
peptides 35:1–23. Höllt V, Haarmann I, Grimm C, Herz A, Tulunay FC, and Loh HH (1982) Pro-
Davenport AP, Alexander SP, Sharman JL, Pawson AJ, Benson HE, Monaghan AE, enkephalin intermediates in bovine brain and adrenal medulla: characterization of
Liew WC, Mpamhanga CP, Bonner TI, and Neubig RR, et al. (2013) International immunoreactive peptides related to BAM-22P and peptide F. Life Sci 31:
Union of Basic and Clinical Pharmacology. LXXXVIII. G protein-coupled receptor list: 1883–1886.
recommendations for new pairings with cognate ligands. Pharmacol Rev 65:967–986. Honan SA and McNaughton PA (2007) Sensitisation of TRPV1 in rat sensory neu-
de Lecea L (2008) Cortistatin—functions in the central nervous system. Mol Cell rones by activation of SNSRs. Neurosci Lett 422:1–6.
Endocrinol 286:88–95. Hong Y, Dai P, Jiang J, and Zeng X (2004) Dual effects of intrathecal BAM22 on
de Lecea L, Criado JR, Prospero-Garcia O, Gautvik KM, Schweitzer P, Danielson PE, nociceptive responses in acute and persistent pain—potential function of a novel
Dunlop CL, Siggins GR, Henriksen SJ, and Sutcliffe JG (1996) A cortical neuropep- receptor. Br J Pharmacol 141:423–430.
tide with neuronal depressant and sleep-modulating properties. Nature 381:242–245. Jackson JG, Usachev YM, and Thayer SA (2007) Bradykinin-induced nuclear factor
DeBold CR, Menefee JK, Nicholson WE, and Orth DN (1988a) Proopiomelanocortin of activated T-cells-dependent transcription in rat dorsal root ganglion neurons.
gene is expressed in many normal human tissues and in tumors not associated Mol Pharmacol 72:303–310.
with ectopic adrenocorticotropin syndrome. Mol Endocrinol 2:862–870. Jiang J, Huang J, and Hong Y (2006) Bovine adrenal medulla 22 reverses anti-
DeBold CR, Nicholson WE, and Orth DN (1988b) Immunoreactive proopiomelano- nociceptive morphine tolerance in the rat. Behav Brain Res 168:167–171.
cortin (POMC) peptides and POMC-like messenger ribonucleic acid are present in Jiang J, Wang D, Zhou X, Huo Y, Chen T, Hu F, Quirion R, and Hong Y (2013) Effect
many rat nonpituitary tissues. Endocrinology 122:2648–2657. of Mas-related gene (Mrg) receptors on hyperalgesia in rats with CFA-induced
Dong X, Han S, Zylka MJ, Simon MI, and Anderson DJ (2001) A diverse family of inflammation via direct and indirect mechanisms. Br J Pharmacol 170:1027–1040.
GPCRs expressed in specific subsets of nociceptive sensory neurons. Cell 106:619–632. Jordan BA, Trapaidze N, Gomes I, Nivarthi R, and Devi LA (2001) Oligomerization of
Dussor G, Zylka MJ, Anderson DJ, and McCleskey EW (2008) Cutaneous sensory opioid receptors with beta 2-adrenergic receptors: a role in trafficking and mitogen-
neurons expressing the Mrgprd receptor sense extracellular ATP and are putative activated protein kinase activation. Proc Natl Acad Sci USA 98:343–348.
nociceptors. J Neurophysiol 99:1581–1589. Jung H, Bhangoo S, Banisadr G, Freitag C, Ren D, White FA, and Miller RJ (2009)
Fatakia SN, Costanzi S, and Chow CC (2011) Molecular evolution of the trans- Visualization of chemokine receptor activation in transgenic mice reveals periph-
membrane domains of G protein-coupled receptors. PLoS ONE 6:e27813. eral activation of CCR2 receptors in states of neuropathic pain. J Neurosci 29:
Ferguson SS (2001) Evolving concepts in G protein-coupled receptor endocytosis: the 8051–8062.
role in receptor desensitization and signaling. Pharmacol Rev 53:1–24. Jung H and Miller RJ (2008) Activation of the nuclear factor of activated T-cells
Ferry X, Brehin S, Kamel R, and Landry Y (2002) G protein-dependent activation of (NFAT) mediates upregulation of CCR2 chemokine receptors in dorsal root
mast cell by peptides and basic secretagogues. Peptides 23:1507–1515. ganglion (DRG) neurons: a possible mechanism for activity-dependent tran-
Fredriksson R, Lagerström MC, Lundin LG, and Schiöth HB (2003) The G-protein- scription in DRG neurons in association with neuropathic pain. Mol Cell Neu-
coupled receptors in the human genome form five main families. Phylogenetic rosci 37:170–177.
analysis, paralogon groups, and fingerprints. Mol Pharmacol 63:1256–1272. Kaisho Y, Watanabe T, Nakata M, Yano T, Yasuhara Y, Shimakawa K, Mori I,
Fu ES, Zhang YP, Sagen J, Candiotti KA, Morton PD, Liebl DJ, Bethea JR, Sakura Y, Terao Y, and Matsui H, et al. (2005) Transgenic rats overexpressing the
and Brambilla R (2010) Transgenic inhibition of glial NF-kappa B reduces pain human MrgX3 gene show cataracts and an abnormal skin phenotype. Biochem
behavior and inflammation after peripheral nerve injury. Pain 148:509–518. Biophys Res Commun 330:653–657.
Fukusumi S, Kitada C, Takekawa S, Kizawa H, Sakamoto J, Miyamoto M, Hinuma Kamohara M, Matsuo A, Takasaki J, Kohda M, Matsumoto M, Matsumoto S, Soga T,
S, Kitano K, and Fujino M (1997) Identification and characterization of a novel Hiyama H, Kobori M, and Katou M (2005) Identification of MrgX2 as a human
human cortistatin-like peptide. Biochem Biophys Res Commun 232:157–163. G-protein-coupled receptor for proadrenomedullin N-terminal peptides. Biochem
Galés C, Rebois RV, Hogue M, Trieu P, Breit A, Hébert TE, and Bouvier M (2005) Biophys Res Commun 330:1146–1152.
Real-time monitoring of receptor and G-protein interactions in living cells. Nat Kashem SW, Subramanian H, Collington SJ, Magotti P, Lambris JD, and Ali H
Methods 2:177–184. (2011) G protein coupled receptor specificity for C3a and compound 48/80-induced
596 Solinski et al.

degranulation in human mast cells: roles of Mas-related genes MrgX1 and MrgX2. Ndong C, Pradhan A, Puma C, Morello JP, Hoffert C, Groblewski T, O’Donnell D,
Eur J Pharmacol 668:299–304. and Laird JM (2009) Role of rat sensory neuron-specific receptor (rSNSR1) in
Katritch V, Cherezov V, and Stevens RC (2013) Structure-function of the G protein- inflammatory pain: contribution of TRPV1 to SNSR signaling in the pain pathway.
coupled receptor superfamily. Annu Rev Pharmacol Toxicol 53:531–556. Pain 143:130–137.
Kaupmann K, Huggel K, Heid J, Flor PJ, Bischoff S, Mickel SJ, McMaster G, Angst Nguyen T and Di Giovanni S (2008) NFAT signaling in neural development and axon
C, Bittiger H, and Froestl W, et al. (1997) Expression cloning of GABA(B) receptors growth. Int J Dev Neurosci 26:141–145.
uncovers similarity to metabotropic glutamate receptors. Nature 386:239–246. Oosterom J, Nijenhuis WA, Schaaper WM, Slootstra J, Meloen RH, Gispen WH,
Kenakin T (1997) Differences between natural and recombinant G protein-coupled Burbach JP, and Adan RA (1999) Conformation of the core sequence in melano-
receptor systems with varying receptor/G protein stoichiometry. Trends Pharmacol cortin peptides directs selectivity for the melanocortin MC3 and MC4 receptors.
Sci 18:456–464. J Biol Chem 274:16853–16860.
Kenakin T (2006) Collateral efficacy as a pharmacological problem applied to new Overington JP, Al-Lazikani B, and Hopkins AL (2006) How many drug targets are
drug discovery. Expert Opin Drug Discov 1:635–652. there? Nat Rev Drug Discov 5:993–996.
Kim YH, Park CK, Back SK, Lee CJ, Hwang SJ, Bae YC, Na HS, Kim JS, Jung SJ, Parenty G, Appelbe S, and Milligan G (2008) CXCR2 chemokine receptor antagonism
and Oh SB (2009) Membrane-delimited coupling of TRPV1 and mGluR5 on pre- enhances DOP opioid receptor function via allosteric regulation of the CXCR2-DOP
synaptic terminals of nociceptive neurons. J Neurosci 29:10000–10009. receptor heterodimer. Biochem J 412:245–256.
Kitto KF, Haley JE, and Wilcox GL (1992) Involvement of nitric oxide in spinally Peier AM, Moqrich A, Hergarden AC, Reeve AJ, Andersson DA, Story GM, Earley TJ,
mediated hyperalgesia in the mouse. Neurosci Lett 148:1–5. Dragoni I, McIntyre P, and Bevan S, et al. (2002) A TRP channel that senses cold
Ko SW, Vadakkan KI, Ao H, Gallitano-Mendel A, Wei F, Milbrandt J, and Zhuo M stimuli and menthol. Cell 108:705–715.
(2005) Selective contribution of Egr1 (zif/268) to persistent inflammatory pain. Pelletier G, Leclerc R, Benjannet S, Seidah NG, and Chrétien M (1981) Immuno-
J Pain 6:12–20. histochemical localization of gamma-melanocyte-stimulating hormone in the rat
Kobayashi H, Yamamoto R, Kitamura K, Kuwasako K, Minami S, Yanagita T, pituitary gland. Regul Pept 2:81–89.
Shiraishi S, Yokoo H, Eto T, and Wada A (2001) Selective inhibition of nicotinic Prescott ED and Julius D (2003) A modular PIP2 binding site as a determinant of
cholinergic receptors by proadrenomedullin N-terminal 12 peptide in bovine ad- capsaicin receptor sensitivity. Science 300:1284–1288.
renal chromaffin cells. Brain Res Mol Brain Res 87:175–183. Price DA and Greenberg MJ (1977) Structure of a molluscan cardioexcitatory neu-
Kolesnikov YA, Pan YX, Babey AM, Jain S, Wilson R, and Pasternak GW (1997) ropeptide. Science 197:670–671.
Functionally differentiating two neuronal nitric oxide synthase isoforms through Quirion R and Weiss AS (1983) Peptide E and other proenkephalin-derived peptides
antisense mapping: evidence for opposing NO actions on morphine analgesia and are potent kappa opiate receptor agonists. Peptides 4:445–449.
tolerance. Proc Natl Acad Sci USA 94:8220–8225. Ramachandran R and Hollenberg MD (2008) Proteinases and signalling: patho-
Kukkonen JP (2004) Regulation of receptor-coupling to (multiple) G proteins. A physiological and therapeutic implications via PARs and more. Br J Pharmacol
challenge for basic research and drug discovery. Receptors Channels 10:167–183. 153 (Suppl 1):S263–S282.
Kunapuli P, Lee S, Zheng W, Alberts M, Kornienko O, Mull R, Kreamer A, Hwang JI, Rau KK, McIlwrath SL, Wang H, Lawson JJ, Jankowski MP, Zylka MJ, Anderson
Simon MI, and Strulovici B (2006) Identification of small molecule antagonists of DJ, and Koerber HR (2009) Mrgprd enhances excitability in specific populations of
the human mas-related gene-X1 receptor. Anal Biochem 351:50–61. cutaneous murine polymodal nociceptors. J Neurosci 29:8612–8619.
Latremoliere A and Woolf CJ (2009) Central sensitization: a generator of pain hy- Rexed B (1952) The cytoarchitectonic organization of the spinal cord in the cat.
persensitivity by central neural plasticity. J Pain 10:895–926. J Comp Neurol 96:414–495.
Lautner RQ, Villela DC, Fraga-Silva RA, Silva N, Verano-Braga T, Costa-Fraga F, Robas N, Mead E, and Fidock M (2003) MrgX2 is a high potency cortistatin receptor
Jankowski J, Jankowski V, Sousa F, and Alzamora A, et al. (2013) Discovery and expressed in dorsal root ganglion. J Biol Chem 278:44400–44404.
characterization of alamandine: a novel component of the renin-angiotensin sys- Romero A, González-Cuello A, Laorden ML, Campillo A, Vasconcelos N, Romero-
tem. Circ Res 112:1104–1111. Alejo E, and Puig MM (2012) Effects of surgery and/or remifentanil administration
Lembo PM, Grazzini E, Groblewski T, O’Donnell D, Roy MO, Zhang J, Hoffert C, Cao on the expression of pERK1/2, c-Fos and dynorphin in the dorsal root ganglia in
J, Schmidt R, and Pelletier M, et al. (2002) Proenkephalin A gene products activate mice. Naunyn Schmiedebergs Arch Pharmacol 385:397–409.
a new family of sensory neuron—specific GPCRs. Nat Neurosci 5:201–209. Roselli-Rehfuss L, Mountjoy KG, Robbins LS, Mortrud MT, Low MJ, Tatro JB,
Liu Q, Sikand P, Ma C, Tang Z, Han L, Li Z, Sun S, LaMotte RH, and Dong X (2012) Entwistle ML, Simerly RB, and Cone RD (1993) Identification of a receptor for
Mechanisms of itch evoked by b-alanine. J Neurosci 32:14532–14537. gamma melanotropin and other proopiomelanocortin peptides in the hypothala-
Liu Q, Tang Z, Surdenikova L, Kim S, Patel KN, Kim A, Ru F, Guan Y, Weng HJ, mus and limbic system. Proc Natl Acad Sci USA 90:8856–8860.
and Geng Y, et al. (2009) Sensory neuron-specific GPCR Mrgprs are itch receptors Ross PC, Figler RA, Corjay MH, Barber CM, Adam N, Harcus DR, and Lynch KR
mediating chloroquine-induced pruritus. Cell 139:1353–1365. (1990) RTA, a candidate G protein-coupled receptor: cloning, sequencing, and tis-
Liu Q, Vrontou S, Rice FL, Zylka MJ, Dong X, and Anderson DJ (2007) Molecular sue distribution. Proc Natl Acad Sci USA 87:3052–3056.
genetic visualization of a rare subset of unmyelinated sensory neurons that may Santos RA, Simoes e Silva AC, Maric C, Silva DM, Machado RP, de Buhr I, Heringer-
detect gentle touch. Nat Neurosci 10:946–948. Walther S, Pinheiro SV, Lopes MT, and Bader M, et al. (2003) Angiotensin-(1–7) is
Liu Q, Weng HJ, Patel KN, Tang Z, Bai H, Steinhoff M, and Dong X (2011) The an endogenous ligand for the G protein-coupled receptor Mas. Proc Natl Acad Sci
distinct roles of two GPCRs, MrgprC11 and PAR2, in itch and hyperalgesia. Sci USA 100:8258–8263.
Signal 4:ra45. Schmidt R, Butterworth J, O’Donnell D, Santhakumar V, and Tomaszewski M (2009)
Liu Y, Yang FC, Okuda T, Dong X, Zylka MJ, Chen CL, Anderson DJ, Kuner R, Cyclic dimers of C-terminal gamma2-MSH analogs as selective antagonists of the
and Ma Q (2008) Mechanisms of compartmentalized expression of Mrg class human sensory nerve-specific receptor (SNSR-4). Adv Exp Med Biol 611:111–112.
G-protein-coupled sensory receptors. J Neurosci 28:125–132. Serrano A, Paré M, McIntosh F, Elmes SJ, Martino G, Jomphe C, Lessard E, Lembo
Ma Q, Fode C, Guillemot F, and Anderson DJ (1999) Neurogenin1 and neurogenin2 PM, Vaillancourt F, and Perkins MN, et al. (2010) Blocking spinal CCR2 with
control two distinct waves of neurogenesis in developing dorsal root ganglia. Genes AZ889 reversed hyperalgesia in a model of neuropathic pain. Mol Pain 6:90.
Dev 13:1717–1728. Sharma BB, Apgar JR, and Liu FT (2002) Mast cells. Receptors, secretagogues, and
Macdonald RL and Olsen RW (1994) GABAA receptor channels. Annu Rev Neurosci signaling. Clin Rev Allergy Immunol 22:119–148.
17:569–602. Shemesh R, Toporik A, Levine Z, Hecht I, Rotman G, Wool A, Dahary D, Gofer E,
Malik L, Kelly NM, Ma JN, Currier EA, Burstein ES, and Olsson R (2009) Discovery Kliger Y, and Soffer MA, et al. (2008) Discovery and validation of novel peptide
of non-peptidergic MrgX1 and MrgX2 receptor agonists and exploration of an ini- agonists for G-protein-coupled receptors. J Biol Chem 283:34643–34649.
tial SAR using solid-phase synthesis. Bioorg Med Chem Lett 19:1729–1732. Shenoy SK and Lefkowitz RJ (2003) Multifaceted roles of beta-arrestins in the reg-
Marks N, Datta RK, and Lajtha A (1970) Distribution of amino acids and of exo- and ulation of seven-membrane-spanning receptor trafficking and signalling. Biochem
endopeptidases along vertebrate and invertebrate nerves. J Neurochem 17:53–63. J 375:503–515.
McKemy DD, Neuhausser WM, and Julius D (2002) Identification of a cold receptor Shimada SG and LaMotte RH (2008) Behavioral differentiation between itch and
reveals a general role for TRP channels in thermosensation. Nature 416:52–58. pain in mouse. Pain 139:681–687.
Menard DP, van Rossum D, Kar S, St Pierre S, Sutak M, Jhamandas K, and Quirion Shimada SG, Shimada KA, and Collins JG (2006) Scratching behavior in mice in-
R (1996) A calcitonin gene-related peptide receptor antagonist prevents the de- duced by the proteinase-activated receptor-2 agonist, SLIGRL-NH2. Eur J Phar-
velopment of tolerance to spinal morphine analgesia. J Neurosci 16:2342–2351. macol 530:281–283.
Metcalfe DD, Baram D, and Mekori YA (1997) Mast cells. Physiol Rev 77: Shimosawa T, Ito Y, Ando K, Kitamura K, Kangawa K, and Fujita T (1995) Proa-
1033–1079. drenomedullin NH(2)-terminal 20 peptide, a new product of the adrenomedullin
Milasta S, Pediani J, Appelbe S, Trim S, Wyatt M, Cox P, Fidock M, and Milligan G gene, inhibits norepinephrine overflow from nerve endings. J Clin Invest 96:
(2006) Interactions between the Mas-related receptors MrgD and MrgE alter sig- 1672–1676.
nalling and trafficking of MrgD. Mol Pharmacol 69:479–491. Shinohara T, Harada M, Ogi K, Maruyama M, Fujii R, Tanaka H, Fukusumi S,
Mizuno K, Minamino N, Kangawa K, and Matsuo H (1980) A new family of endog- Komatsu H, Hosoya M, and Noguchi Y, et al. (2004) Identification of a G protein-
enous “big” Met-enkephalins from bovine adrenal medulla: purification and coupled receptor specifically responsive to beta-alanine. J Biol Chem 279:
structure of docosa- (BAM-22P) and eicosapeptide (BAM-20P) with very potent 23559–23564.
opiate activity. Biochem Biophys Res Commun 97:1283–1290. Sikand P, Dong X, and Lamotte RH (2011) BAM8-22 peptide produces itch and
Mollereau C, Mazarguil H, Marcus D, Quelven I, Kotani M, Lannoy V, Dumont Y, nociceptive sensations in humans independent of histamine release. J Neurosci 31:
Quirion R, Detheux M, and Parmentier M, et al. (2002) Pharmacological charac- 7563–7567.
terization of human NPFF(1) and NPFF(2) receptors expressed in CHO cells by Sikand P, Shimada SG, Green BG, and LaMotte RH (2009) Similar itch and noci-
using NPY Y(1) receptor antagonists. Eur J Pharmacol 451:245–256. ceptive sensations evoked by punctate cutaneous application of capsaicin, hista-
Molliver DC, Wright DE, Leitner ML, Parsadanian AS, Doster K, Wen D, Yan Q, mine and cowhage. Pain 144:66–75.
and Snider WD (1997) IB4-binding DRG neurons switch from NGF to GDNF de- Snider WD and McMahon SB (1998) Tackling pain at the source: new ideas about
pendence in early postnatal life. Neuron 19:849–861. nociceptors. Neuron 20:629–632.
Nantel F, Bonin H, Emorine LJ, Zilberfarb V, Strosberg AD, Bouvier M, and Marullo Solinski HJ, Boekhoff I, Bouvier M, Gudermann T, and Breit A (2010) Sensory
S (1993) The human beta 3-adrenergic receptor is resistant to short term agonist- neuron-specific MAS-related gene-X1 receptors resist agonist-promoted endocyto-
promoted desensitization. Mol Pharmacol 43:548–555. sis. Mol Pharmacol 78:249–259.
MAS-Related G Protein–Coupled Receptors 597
Solinski HJ, Petermann F, Rothe K, Boekhoff I, Gudermann T, and Breit A (2013) Wang H and Zylka MJ (2009) Mrgprd-expressing polymodal nociceptive neurons
Human Mas-related G protein-coupled receptors-X1 induce chemokine receptor 2 innervate most known classes of substantia gelatinosa neurons. J Neurosci 29:
expression in rat dorsal root ganglia neurons and release of chemokine ligand 2 13202–13209.
from the human LAD-2 mast cell line. PLoS ONE 8:e58756. Wang TT, Yuan WL, Ke Q, Song XB, Zhou X, Kang Y, Zhang HT, Lin Y, Hu YL,
Solinski HJ, Zierler S, Gudermann T, and Breit A (2012) Human sensory neuron- and Feng ZT, et al. (2006a) Effects of electro-acupuncture on the expression of c-jun
specific Mas-related G protein-coupled receptors-X1 sensitize and directly activate and c-fos in spared dorsal root ganglion and associated spinal laminae following
transient receptor potential cation channel V1 via distinct signaling pathways. removal of adjacent dorsal root ganglia in cats. Neuroscience 140:1169–1176.
J Biol Chem 287:40956–40971. Wang Z, Takahashi T, Saito Y, Nagasaki H, Ly NK, Nothacker HP, Reinscheid RK,
Steinhoff M, Buddenkotte J, Shpacovitch V, Rattenholl A, Moormann C, Vergnolle N, Yang J, Chang JK, and Shichiri M, et al. (2006b) Salusin beta is a surrogate ligand
Luger TA, and Hollenberg MD (2005) Proteinase-activated receptors: transducers of the mas-like G protein-coupled receptor MrgA1. Eur J Pharmacol 539:145–150.
of proteinase-mediated signaling in inflammation and immune response. Endocr Washimine H, Kitamura K, Ichiki Y, Yamamoto Y, Kangawa K, Matsuo H, and Eto T
Rev 26:1–43. (1994) Immunoreactive proadrenomedullin N-terminal 20 peptide in human tis-
Su JH, Chen YF, Tang JR, Wu L, Zhang P, Yu LB, Niu Q, and Xiao H (2010) sue, plasma and urine. Biochem Biophys Res Commun 202:1081–1087.
Protective effects of the calcium-channel blocker flunarizine on crush injury of Wei C, Huang W, Xing X, and Dong S (2010) Dual effects of [Tyr(6)]-gamma2-MSH(6-
sciatic nerves in a rat model. Neurol India 58:530–536. 12) on pain perception and in vivo hyperalgesic activity of its analogues. J Pept Sci
Subramanian H, Gupta K, Guo Q, Price R, and Ali H (2011a) Mas-related gene X2 16:451–455.
(MrgX2) is a novel G protein-coupled receptor for the antimicrobial peptide LL-37 White FA, Jung H, and Miller RJ (2007) Chemokines and the pathophysiology of
in human mast cells: resistance to receptor phosphorylation, desensitization, and neuropathic pain. Proc Natl Acad Sci USA 104:20151–20158.
internalization. J Biol Chem 286:44739–44749. Williams JT, Ingram SL, Henderson G, Chavkin C, von Zastrow M, Schulz S, Koch T,
Subramanian H, Gupta K, Lee D, Bayir AK, Ahn H, and Ali H (2013) b-Defensins Evans CJ, and Christie MJ (2013) Regulation of m-opioid receptors: desensitization,
activate human mast cells via Mas-related gene X2. J Immunol 191:345–352. phosphorylation, internalization, and tolerance. Pharmacol Rev 65:223–254.
Subramanian H, Kashem SW, Collington SJ, Qu H, Lambris JD, and Ali H (2011b) Wilson SR, Gerhold KA, Bifolck-Fisher A, Liu Q, Patel KN, Dong X, and Bautista DM
PMX-53 as a dual CD88 antagonist and an agonist for Mas-related gene 2 (MrgX2) (2011) TRPA1 is required for histamine-independent, Mas-related G protein-
in human mast cells. Mol Pharmacol 79:1005–1013. coupled receptor-mediated itch. Nat Neurosci 14:595–602.
Sun JH, Yang B, Donnelly DF, Ma C, and LaMotte RH (2006) MCP-1 enhances Wittenberger T, Schaller HC, and Hellebrand S (2001) An expressed sequence tag
excitability of nociceptive neurons in chronically compressed dorsal root ganglia. (EST) data mining strategy succeeding in the discovery of new G-protein coupled
J Neurophysiol 96:2189–2199. receptors. J Mol Biol 307:799–813.
Tallan HH, Moore S, and Stein WH (1954) Studies on the free amino acids and Woo DH, Jung SJ, Zhu MH, Park CK, Kim YH, Oh SB, and Lee CJ (2008) Direct
related compounds in the tissues of the cat. J Biol Chem 211:927–939. activation of transient receptor potential vanilloid 1(TRPV1) by diacylglycerol
Tatemoto K, Nozaki Y, Tsuda R, Konno S, Tomura K, Furuno M, Ogasawara H, (DAG). Mol Pain 4:42–56.
Edamura K, Takagi H, and Iwamura H, et al. (2006) Immunoglobulin E- Woolf CJ and Ma Q (2007) Nociceptors—noxious stimulus detectors. Neuron 55:
independent activation of mast cell is mediated by Mrg receptors. Biochem Bio- 353–364.
phys Res Commun 349:1322–1328. Yang HY, Fratta W, Majane EA, and Costa E (1985) Isolation, sequencing, synthesis,
Than JY, Li L, Hasan R, and Zhang X (2013) Excitation and modulation of TRPA1, and pharmacological characterization of two brain neuropeptides that modulate
TRPV1, and TRPM8 channel-expressing sensory neurons by the pruritogen chlo- the action of morphine. Proc Natl Acad Sci USA 82:7757–7761.
roquine. J Biol Chem 288:12818–12827. Zeng X, Huang H, and Hong Y (2004) Effects of intrathecal BAM22 on noxious stimulus-
Tiedje KE, Stevens K, Barnes S, and Weaver DF (2010) Beta-alanine as a small evoked c-fos expression in the rat spinal dorsal horn. Brain Res 1028:170–179.
molecule neurotransmitter. Neurochem Int 57:177–188. Zhang H, Stoeckli M, Andren PE, and Caprioli RM (1999) Combining solid-phase
Torvinen M, Marcellino D, Canals M, Agnati LF, Lluis C, Franco R, and Fuxe K preconcentration, capillary electrophoresis and off-line matrix-assisted laser
(2005) Adenosine A2A receptor and dopamine D3 receptor interactions: evidence of desorption/ionization mass spectrometry: intracerebral metabolic processing of
functional A2A/D3 heteromeric complexes. Mol Pharmacol 67:400–407. peptide E in vivo. J Mass Spectrom 34:377–383.
Uno M, Nishimura S, Fukuchi K, Kaneta Y, Oda Y, Komori H, Takeda S, Haga T, Zhang J, Cavanaugh DJ, Nemenov MI, and Basbaum AI (2013) The modality-specific
Agatsuma T, and Nara F (2012) Identification of physiologically active substances contribution of peptidergic and non-peptidergic nociceptors is manifest at the level
as novel ligands for MRGPRD. J Biomed Biotechnol 2012:816159. of dorsal horn nociresponsive neurons. J Physiol 591:1097–1110.
Utreras E, Futatsugi A, Rudrabhatla P, Keller J, Iadarola MJ, Pant HC, Zhang L, Taylor N, Xie Y, Ford R, Johnson J, Paulsen JE, and Bates B (2005) Cloning
and Kulkarni AB (2009) Tumor necrosis factor-alpha regulates cyclin-dependent and expression of MRG receptors in macaque, mouse, and human. Brain Res Mol
kinase 5 activity during pain signaling through transcriptional activation of p35. Brain Res 133:187–197.
J Biol Chem 284:2275–2284. Zhang R, Yan PK, Zhou CH, Liao JY, and Wang MW (2007) Development of a ho-
Vrontou S, Wong AM, Rau KK, Koerber HR, and Anderson DJ (2013) Genetic mogeneous calcium mobilization assay for high throughput screening of mas-
identification of C fibres that detect massage-like stroking of hairy skin in vivo. related gene receptor agonists. Acta Pharmacol Sin 28:125–131.
Nature 493:669–673. Zuo Y, Perkins NM, Tracey DJ, and Geczy CL (2003) Inflammation and hyperalgesia
Walters PE, Gaspari TA, and Widdop RE (2005) Angiotensin-(1–7) acts as a vaso- induced by nerve injury in the rat: a key role of mast cells. Pain 105:467–479.
depressor agent via angiotensin II type 2 receptors in conscious rats. Hypertension Zylka MJ, Dong X, Southwell AL, and Anderson DJ (2003) Atypical expansion in mice
45:960–966. of the sensory neuron-specific Mrg G protein-coupled receptor family. Proc Natl
Wang CH, Zou LJ, Zhang YL, Jiao YF, and Sun JH (2010) The excitatory effects of Acad Sci USA 100:10043–10048.
the chemokine CCL2 on DRG somata are greater after an injury of the ganglion Zylka MJ, Rice FL, and Anderson DJ (2005) Topographically distinct epidermal no-
than after an injury of the spinal or peripheral nerve. Neurosci Lett 475:48–52. ciceptive circuits revealed by axonal tracers targeted to Mrgprd. Neuron 45:17–25.

You might also like