Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

ORIGINAL ARTICLE

Dimerization of the Human Melanocortin 1


Receptor: Functional Consequences and
Dominant-Negative Effects
Berta L. Sánchez-Laorden1, Jesús Sánchez-Más1, Emma Martı́nez-Alonso2, José A. Martı́nez-Menárguez2,
José C. Garcı́a-Borrón1 and Celia Jiménez-Cervantes1

The melanocortin 1 receptor (MC1R), a GS-protein-coupled receptor (GPCR), is a key regulator of proliferation
and differentiation of epidermal melanocytes, and a determinant of human skin phototype and cancer risk.
Homodimerization has been demonstrated for several GPCRs, but little information is available for MC1R. SDS-
PAGE analysis of melanoma cells and heterologous cells expressing epitope-tagged MC1R revealed dimeric and
oligomeric species in detergent-solubilized extracts, confirmed by coimmunoprecipitation of differentially
tagged MC1R forms. Dimerization occurs early during MC1R biosynthesis, and is seen for mutants displaying
intracellular retention. These mutants exerted dominant-negative effects on wild-type (WT) MC1R. Conversely,
partial functional transcomplementation of selected loss-of-function mutants was observed. WT-MC1R lacks
cooperativity in agonist binding, yet coexpression of WT and a C-terminal deletion mutant yielded a form of
different pharmacological properties. The natural diminished function alleles R151C, R160W, and D294H,
associated with red hair, displayed dimerization and heterodimerization with WT. Coexpression of WT and
R151C or R160W reduced the density of binding sites on the plasma membrane of transfected cells, whereas
D294H mediated a dominant-negative effect on functional coupling to adenylyl cyclase. Therefore, subtle
changes of functional properties may be associated with different MC1R haplotypes, contributing to the
complexity of skin phenotype.
Journal of Investigative Dermatology (2006) 126, 172–181. doi:10.1038/sj.jid.5700036

INTRODUCTION pigments (Di Donato and Napolitano, 2003). Tyrosinase


Human hair and skin pigmentation results from the synthesis activity is controlled by a variety of mechanisms, including
of L-tyrosine-derived melanin pigments within epidermal hormonal stimulation by a melanocyte-stimulating hormone
melanocytes (Rees, 2003). By acting as effective sunscreens (aMSH) following activation of the melanocortin 1 receptor
able to absorb harmful ultraviolet radiation, melanin pig- (MC1R), a G-protein-coupled receptor (GPCR) positively
ments play a major role in photoprotection (Kadekaro et al., coupled to the cAMP pathway (Busca and Ballotti, 2000)
2003). The synthesis of melanin pigments is governed by expressed in epidermal melanocytes. In addition, MC1R
tyrosinase, an enzyme catalyzing the rate-limiting conversion activation increases the ratio of black and strongly photo-
of L-tyrosine into L-dopaquinone (DQ). In the absence of thiol protective eumelanins to yellowish and poorly photoprotec-
compounds, DQ evolves to black eumelanin pigments. tive pheomelanins (Abdel-Malek et al., 2000; Barsh et al.,
However, in the presence of cysteine or glutathione, addition 2000; Kadekaro et al., 2003). Accordingly, MC1R is a key
of sulfhydryl groups to DQ yields reddish pheomelanin regulator of mammalian pigmentation.
The human MC1R gene is highly polymorphic. Several
variants are associated with fair skin, red hair color (RHC)
1
Department of Biochemistry and Molecular Biology, University of Murcia, (Box et al., 1997; Smith et al., 1998; Schioth et al., 1999;
Espinardo, Spain and 2Department of Cell Biology, School of Medicine,
Sturm, 2002; Rees, 2003), a high number of freckles
University of Murcia, Espinardo, Spain
(Bastiaens et al., 2001a), and increased skin cancer risk
Correspondence: Dr Celia Jiménez-Cervantes, Department of Biochemistry
and Molecular Biology, School of Medicine, University of Murcia, 30100 (Valverde et al., 1996; Palmer et al., 2000; Bastiaens et al.,
Espinardo, Spain. E-mail: celiajim@um.es 2001b; Sturm, 2002; Sturm et al., 2003). The best character-
Abbreviations: DQ, L-dopaquinone; ER, endoplasmic reticulum; GPCR, ized variants are the RHC alleles R151C, R160W, and
G-protein-coupled receptor; HA, hyaluronate; HEK, human embryonic D294H that show diminished coupling to the cAMP signaling
kidney; LOF, loss of function; MC1R, melanocortin 1 receptor; aMSH, a
pathway (Schioth et al., 1999; Ringholm et al., 2004). The
melanocyte-stimulating hormone; NDP-MSH, norleucine4
7
D-phenylalanine -melanocyte-stimulating hormone; PBS, phosphate-buffered RHC alleles are very frequent, and in certain populations
saline; RHC, red hair color; WT, wild type about 40% of the individuals harbor at least one of them
Received 25 May 2005; revised 22 August 2005; accepted 6 September 2005 (Rees, 2003). Many other potentially relevant MC1R variants

172 Journal of Investigative Dermatology (2006), Volume 126 & 2006 The Society for Investigative Dermatology
BL Sánchez-Laorden et al.
Melanocortin 1 Receptor Homodimerization

have been described (Jimenez-Cervantes et al., 2001a, b; a MC1R


Sanchez-Más et al., 2002). 1.2 Flag-MC1R

pmol cAMP/g protein


HA-MC1R
Many GPCRs form dimers or oligomers (Milligan, 2001; 0.8 MC1R-TAG
Angers et al., 2002; Breitwieser, 2004). Dimerization pcDNA3
0.4
can modulate key properties of the receptors, including
ligand binding, coupling efficiency, desensitization, and 0.1
intracellular traffic from the endoplasmic reticulum (ER) to
the plasma membrane, and through endocytic pathways. A 0.0
Control –9 –7
recent study suggests that MC1R may dimerize in living Log[NDP-MSH] (M)
cells, but neither the mechanism nor the functional effect
of dimerization was reported (Mandrika et al., 2005). b 30 c 125

c.p.m. bound/dish
Dimer formation has been demonstrated for the related 100

Bx100/Bmax
(thousands)
MC4R and shown to result in a dominant-negative effect 20
75
of a mutant associated with severe early-onset obesity 50
10
(Biebermann et al., 2003). Flag-MC1R
25
We show here that MC1R forms dimers and oligomers WT-MC1R
0 0
(collectively termed dimers for simplicity) in heterologous WT FLAG HA TAG pcDNA3 –12 –11 –10 –9 –8 –7 –6 –5
Transfected plasmid
systems and melanoma cells. We also show that dimer Log[competing peptide] (M)
formation may result in dominant-negative effects of mutant Figure 1. Functional characterization of epitope-tagged MC1R. HEK 293T
alleles on wild type (WT)-MC1R, as well as some degree of cells transfected with the indicated variants were serum-deprived for 24 hours
functional complementation between mutants. Therefore, before the assays. (a) Functional coupling to cAMP generation. Cells were
MC1R dimerization may underpin the complexity of geno- treated for 30 minutes with various concentrations of NDP-MSH, or vehicle
(control), lysed, and cAMP was determined by radioimmunoassay. pcDNA3
type–phenotype associations in human pigmentation.
stands for empty vector. (b) Binding of [125I]NDP-MSH. Cells were incubated
with 1010 M [125I]NDP-MSH for 1 hour, washed, and the radioactivity
RESULTS associated was determined. (c) Competition binding analysis of WT- and Flag-
Functional analysis of epitope-tagged MC1R MC1R. Cells were incubated with 1010 M [125I]NDP-MSH and increasing
Three different epitope-tagged variants of the MC1R were concentrations of unlabeled ligand. The specifically bound radioactivity was
constructed: (1) a protein with a Flag epitope at the measured. Results are given as % maximal binding in the absence of
extracellular N-terminus immediately after the initial Met competitor.
residue (Flag-MC1R), (2) a fusion containing one hemagglu-
tinin (HA) epitope after the cytosolic C-terminal W317
residue (MC1R-HA), and (3) a form containing a longer C- were resistant to SDS, in that increasing its concentration
terminal tag with two consecutive HA epitopes followed by a from 0.5 to 3% did not change the aggregation status. No
hexahistidine tail (MC1R-Tag). When expressed in human immunoreactive bands were detected in cells transfected
embryonic kidney (HEK) 293T cells, Flag-MC1R and MC1R- with an MC1R construct lacking the Flag epitope.
HA performed essentially as WT in functional coupling to Under reducing conditions, the majority was a band of
adenylyl cyclase, as assessed by determination of agonist- Mr 30 kDa, consistent with dissociation of MC1R oligomers.
induced cAMP production. MC1R-Tag did not mediate a A 36-kDa band was also detected (Figure 2a). Upon
detectable cAMP response (Figure 1a). Cells transfected with deglycosylation with endoglycosidase H, the 36-kDa band
the WT, Flag-MC1R, or MC1R-HA forms bound similar disappeared, but the mobility of the 30-kDa protein did not
amounts of a radioactively labeled agonist (Figure 1b), but no change (Figure 2b). This suggests that the 30- and 36-kDa
specific binding was demonstrated for MC1R-Tag. For Flag- bands correspond to ‘‘de novo’’ and glycosylated MC1R,
MC1R, displacement experiments indicated an affinity for the respectively. In spite of the strong effect of mercaptoethanol
melanocortin ligand similar to WT (Figure 1c). on monomer:oligomer ratios, samples still contained bands
consistent with MC1R oligomers. In this case, the 58-kDa
Constitutive oligomerization of MC1R species appeared as a doublet, where only the lower mobility
We used the functional Flag-MC1R and MC1R-HA constructs band was sensitive to endoglycosidase H (Figure 2b).
in coimmunoprecipitation and Western blot experiments. To demonstrate that the 58 kDa bands were MC1R dimers,
Transfected HEK 293T cells were solubilized in the presence extracts from cells coexpressing Flag-MC1R and MC1R-HA were
of 10 mM iodoacetamide to prevent spurious disulfide bond immunoprecipitated with an anti-HA antibody and immuno-
formation. Samples were electrophoresed under reducing or blotted with an anti-Flag. Bands consistent with monomeric and
nonreducing conditions, with increasing concentrations of dimeric forms were detected (Figure 2c). These bands were not
SDS (Figure 2a). Samples run in the absence of b-mercap- seen in extracts from cells expressing either variant alone, or
toethanol displayed bands of high apparent molecular weight when these extracts were mixed prior to immunoprecipitation,
(Mr) that barely entered the gel, a prominent band of Mr excluding dimerization after sample solubilization.
58 kDa that may correspond to an MC1R dimer, and a minor We confirmed MC1R dimerization in a melanocytic
band of higher electrophoretic mobility, most likely corre- environment, at a lower expression level. We obtained
sponding to the monomeric species. The oligomeric forms clones of HBL melanoma cells stably expressing Flag-MC1R,

www.jidonline.org 173
BL Sánchez-Laorden et al.
Melanocortin 1 Receptor Homodimerization

a SB without -ME SB with -ME b


SDS (% ): 0.5 1.0 2.0 3.0 0.5 1.0 2.0 3.0 WT EndoH

MW (kD a) MW (kDa )

120
97 120
97
55.4
55.4
36.1

28.9 36.1

Erk- 2 28.9

c IP: -HA; WB: -Flag d HBL Clone 18 WT


MW (kDa)
Flag HA Mix HA+Flag
55.4
113
92

52.3 36.1

35.3 28.9

28.7

Erk- 2

Figure 2. Detection of MC1R dimers and oligomers in solubilized cell extracts. (a) Electrophoretic pattern of Flag-MC1R expressed in HEK 293T cells.
Detergent-solubilized extracts were treated (10 minutes, room temperature) with SDS from 0.5 to 3%, with or without b-mercaptoethanol. Samples were
analyzed by Western blot using an anti-Flag antibody. The mobility of marker proteins is shown on the left. Blots were reprobed with an anti-Erk2 antibody
as a control for comparable loading. (b) Effect of deglycosylation on the electrophoretic pattern of MC1R. Control extracts (WT) or extracts treated with
endoglycosidase H were immunoblotted with an anti-Flag antibody. Endoglycosidase H-sensitive bands in control extracts are highlighted by arrows.
(c) Coimmunoprecipitation of differentially epitope-tagged forms of MC1R. HEK 293T cells were transfected to express either Flag, HA, or a combination
(HA þ Flag) of both epitope-tagged forms of the WT-MC1R, and solubilized. Extracts from cells independently transfected with Flag-MC1R or MC1R-HA were
also mixed and incubated at room temperature for 30 minutes before immunoprecipitation (Mix). Samples were immunoprecipitated with an anti-HA
monoclonal antibody and the precipitates were immunoblotted with an anti-Flag antibody. Arrows highlight the monomer and dimer bands. (d) Detection of
MC1R dimers in human melanoma cells. Extracts from parental HBL, cells stably transfected with Flag-MC1R (clone 18), or HEK 293T cells (WT) transiently
expressing Flag-MC1R were immunoblotted with the anti-Flag antibody. Blots were reprobed with an anti-Erk2 antibody as control. Protein load was 15 (HBL
cells) or 8 mg (HEK cells).

selected on the basis of surface expression of the epitope as modifications taking place in this organelle. Brefeldin A
detected by flow-cytometric analysis. A positive clone was mediated a clear disorganization of the intense, compact, and
further analyzed, which displayed a moderate six-fold perinuclear staining of the cells with a Golgi-specific marker,
enrichment in MC1R (Bmax 1.8 vs 0.3 fmol/mg protein for confirming that the drug was active (not shown). Western
mock-transfected HBL cells). The electrophoretic pattern was blotting revealed a similar distribution of MC1R species in the
similar for this clone and transfected HEK cells (Figure 2d). presence or absence of the drug (Figure 3b), suggesting that
dimerization occurs in a pre-Golgi compartment. Finally, we
Oligomerization is an early event taking place in the ER took advantage of the observation that the LOF MC1R-Tag
To locate the subcellular compartment where MC1R dimer- variant is retained in the ER, as shown by immunolabeling
ization occurs, we first analyzed the electrophoretic pattern and electron microscopy. This variant was employed rather
of two MC1R loss-of-function (LOF) mutants, L93R (R93) and than the L93R or R162P alleles, because in our hands the
R162P (P162), that show impaired cell surface expression due anti-HA antibody used for its detection with protein A-gold
to retention in an intracellular compartment (Sánchez- particles performed better than the anti-Flag. In cells
Laorden et al., 2005). The electrophoretic pattern of these expressing MC1R-Tag, labeling was concentrated in endo-
mutants expressed in HEK 293T cells was similar to WT membranes with the morphology of ER cisternae, with very
(Figure 3a). This suggests that dimerization occurs before little labeling of plasma membranes (Figure 3c). Extracts of
reaching the plasma membrane. Second, we treated Flag- cells expressing MC1R-Tag showed bands corresponding to
MC1R-transfected cells with brefeldin A. This drug disrupts MC1R dimers (Figure 3f). Thus, the MC1R dimer is most likely
the Golgi apparatus and blocks the post-translational formed in the ER.

174 Journal of Investigative Dermatology (2006), Volume 126


BL Sánchez-Laorden et al.
Melanocortin 1 Receptor Homodimerization

a WT R93 P162 - b WT - B1 B 2 deletion of the five C-terminal amino acids, designated D5


MW (kDa) (Sánchez-Laorden et al., 2005; Sánchez-Más et al., 2005b).
MW (kDa)
120.0 HEK 293T cells transfected with these mutants did not show
97.0 120.0
97.0 significant specific binding (Figure 4a) and displayed very
55.4 weak cell surface expression of the receptor, as detected by
55.4 flow cytometric analysis of nonpermeabilized cells (not
36.1
28.9 36.1 shown), confirming previous results (Sánchez-Laorden
et al., 2005; Sánchez-Más et al., 2005b). Coexpression of
28.9
WT-MC1R with increasing amounts of plasmid encoding for
Erk-2 the mutants caused a dose-dependent reduction of
Erk-2
[125I]NDP-MSH binding to about 50% of controls (Figure
4a), consistent with a dominant-negative effect. Cotransfec-
tion of WT-MC1R and an unrelated thromboxane receptor
c f MC1R-Tag - MW (kDa) had no effect on [125I]NDP-MSH-specific binding, demon-
strating the specificity of the dominant-negative effect.
120.0
97.0 Coimmunoprecipitation of differentially epitope-tagged WT
and variants confirmed heterodimerization of the mutants
55.4 and WT (Figure 4b). This suggests that dimerization of WT-
MC1R and aberrantly processed variants may lead to
intracellular retention of the heterodimer, and further
demonstrate MC1R dimerization in living cells.
d 36.1

28.9 Functional characterization of oligomers


e GPCR dimerization often involves the swapping of helices
from each monomer within the dimer, so that two defective
monomers can form a correctly folded and partially
functional unit, provided that the mutations are located in
different domains (Breitwieser, 2004). Conversely, if the
mutations fall in the same protein domain, dimerization
Figure 3. Oligomerization of MC1R is an early event occurring in the ER.
should not restore function. We tested functional comple-
(a) Oligomerization of MC1R LOF variants with impaired plasma membrane
expression. Extracts of cells expressing Flag epitope-tagged WT, L93R and
mentation for the R93, P162, and D5 LOF forms. Although no
R162P, or empty vector (lane labeled ) were immunoblotted with anti-Flag. heterozygotes for these variants have been found, these forms
Protein load was 10 mg/lane, and an anti-Erk2 antibody was used as a control. provide excellent models for functional transcomplementa-
(b) Detection of MC1R oligomers in brefeldin A-treated cells. HEK 293T cells tion because the mutations are located in different protein
were transfected with WT Flag-MC1R or empty pcDNA (). The electro- regions (transmembrane fragments 2 and 4 for R93 and P162,
phoretic pattern of control (WT) and two independent brefeldin A-treated respectively, and C-terminus for D5), and the mutants are
samples (B1 and B2) was analyzed by immunoblotting. Comparable loading
complete LOF forms, thus yielding no background for
was ascertained with an anti-Erk2 antibody. (c–e) Intracellular localization of
MC1R-Tag. HEK 293T cells were transfected with MC1R-Tag, labeled with an
functional analysis. Two other deletion mutants missing the
anti-HA monoclonal antibody, followed by protein A-gold particles, and last 12 and 14 C-terminal amino acids, termed D12 and D14
visualized by electron microscopy. In the upper image, arrows highlight a (Sánchez-Más et al., 2005b), were also analyzed. For some
strong internal labeling, as opposed to the lack of immunoreactivity in the combinations, substantial binding and coupling activities
plasma membrane (pm). The middle image shows intense staining of internal were rescued (Figure 5). However, coexpression of the two
cisternae with the morphology of ER structures. The lower micrograph forms with the inactivating mutation in the same region, the
illustrates the residual labeling of the plasma membrane. (f) Oligomerization
C-terminal cytosolic tail, failed to restore ligand binding or
of MC1R-Tag. Extracts of HEK 293T cells transiently expressing MC1R-Tag
(lane labeled WT) or transfected with empty vector (lane labeled ) were coupling to adenylyl cyclase.
immunoblotted with anti-HA monoclonal antiserum. Arrows point to the We analyzed other functional effects of MC1R dimeriza-
monomer and dimer bands. Bar=200 nm. tion. Equilibrium binding data were analyzed for cooperativ-
ity with the Hill equation. The slope of Hill plots was not
significantly different from 1 (Hill number 1.0370.06, n ¼ 4),
LOF mutants displaying intracellular retention exert indicative of lack of allosteric effects on agonist binding.
a dominant-negative effect Moreover, neither agonist stimulation nor activation of
Since dimerization is probably an early event in MC1R adenylyl cyclase with forskolin modulated the monomer-
biosynthesis, aberrantly processed LOF mutants might exert a to-dimer ratio as estimated by Western blot (Figure 6a),
dominant-negative effect by trapping WT receptor in suggesting that dimerization is constitutive. However, we
intracellular compartments. This was tested by cotransfecting found evidence for a dimerization-dependent modulation of
HEK 293T cells with WT-MC1R and several mutants with MC1R pharmacology. Cotransfection of WT and the D5 LOF
impaired plasma membrane expression. These were the variant yielded a form of higher affinity for NDP-MSH than
natural mutants R93 and P162 and an artificial mutant with a WT, as assessed from left-shifted competition binding curves

www.jidonline.org 175
BL Sánchez-Laorden et al.
Melanocortin 1 Receptor Homodimerization

IP: -HA; WB: -Flag


a WT/variant(1:1, 1:2, 1:3) b
WT-HA: + + + + +
70 Flag-variant: WT R93 – P162 ∆5
c.p.m. bound/dish (thousands)

60

50

40

30

0
W T R93 P162 ∆5 WT WT+pcDNA W T+R93 W T+P162 W T+∆5
+
TXR

Figure 4. LOF variants with compromised cell surface expression exert a dominant-negative effect on WT-MC1R. (a) Reduction of binding sites in cells
coexpressing variant and WT-MC1R. HEK 293T were transfected with variant and WT-MC1R, alone or in combination, as indicated, and binding of 1010 M
[125I]NDP-MSH was measured. For expression of a single MC1R form, 0.6 mg of plasmid DNA/well was employed. For cotransfections, the concentration of
WT-MC1R plasmid DNA was kept constant at 0.15 mg/well, and three variant plasmid concentrations were assayed (0.15, 0.3, and 0.45 mg/well), corresponding
to WT-to-variant ratios of 1:1, 1:2, and 1:3, respectively. Total concentration of plasmid DNA was kept constant at 0.6 mg/well with empty pcDNA. As a control
for specificity, WT-MC1R (0.15 mg/well) was cotransfected with the unrelated thromboxane a receptor (TxR) (0.45 mg/well), or with empty pcDNA.
(b) Heterodimerization of variant and WT-MC1R. HEK 293T cells were transfected to express WT-MC1R-HA and one of the following Flag-epitope-tagged
forms: WT, R93, P162, or D5, as indicated on the top of each lane. As a negative control, empty plasmid was used (lane labeled ). Detergent-solubilized
extracts were immunoprecipitated with an anti-HA monoclonal antibody and immunoblotted with anti-Flag.

a 3000
R93 P162 ∆5 ∆12

2000
c.p.m. bound

1000

0
V P162 ∆5 ∆12 ∆14 V R93 ∆5 ∆12 ∆14 V R93 P162 ∆12 ∆14 V R93 P162 ∆5 ∆14

b 0.10 0.10 0.10


pmoles cAMP/g protein

pmol cAMP/g protein


pmol cAMP/g protein

R93/pcDNA P162/pcDNA ∆5/pcDNA


R93/P162 P162/R93 ∆5/R93
R93/∆5 P162/∆5 ∆5/P162
0.05 R93/∆12 0.05
P162/∆12 ∆5/∆12
0.05
R93/∆14 P162/∆14 ∆5/∆14

0.00 0.00 0. 00
Control NDP-MSH Control NDP-MSH Control NDP-MSH

Figure 5. Functional transcomplementation of MC1R mutants. (a) The LOF MC1R mutants indicated on the top of each group of bars were cotransfected with
an identical amount of empty vector (V) or the mutants shown. Expression of plasma membrane-binding sites was estimated by incubation with 1010 M
[125I]NDP-MSH. Combinations that do not yield a partial recovery of binding are highlighted by arrows. (b) The L93R (left), R162P (middle), and D5 (right) forms
were cotransfected with empty pcDNA3 or the indicated variants. Functional coupling was estimated by measuring cAMP in cells challenged with vehicle
(control) or 107 M NDP-MSH.

(Figure 6b) and from the calculation of dissociation constants Functional behavior and dimerization of the RHC alleles
(1.8070.18 nM for WT vs 0.7370.08 nM in cotransfected The RHC alleles R151C, R160W, and D294H are frequent
cells, P ¼ 0.0017). Interestingly, maximal cAMP levels (Rees, 2003) and display reduced functional responses
achieved upon agonist stimulation were lower in cotrans- (Schioth et al., 1999; Ringholm et al., 2004). Therefore, it
fected samples than in cells transfected with WT alone was of interest to investigate whether these mutants form
(Figure 6c). homodimers and heterodimerize with WT-MC1R. We first

176 Journal of Investigative Dermatology (2006), Volume 126


BL Sánchez-Laorden et al.
Melanocortin 1 Receptor Homodimerization

a MSH Fsk b 100 a SB with -ME SB without -ME


C 30´ 60´ 30´ WT C151 W160 H294 C151W160H294

Bx100/Bmax
75
MW (KDa)
120 50 120
97 97
25 WT
55.4 ∆5+WT 55.4
0
–11 –10 –9 –8 –7
36.1 Log[competing peptide] (M) 36.1
28.9
28.9

Erk-2
Erk-2

c 1.25 W T/pcDNA
pmol cAMP/g protein

W T/∆ 5 b 2.25 Control


1.00

pmol cAMP/g protein


*** 2.00 10-11
0.75 10-7
1.75
0.50

0.25 1.0

0.00 ***
Control NDP-MSH 0.5

Figure 6. Functional characterization of MC1R dimers. (a) Lack of effect of 0.0


WT-F R151C R160W D294H
agonist and forskolin on WT-MC1R dimerization. HEK 293T cells expressing
Variant
WT-MC1R were challenged with 107 M NDP-MSH (30 or 60 minutes),
forskolin (105 M, 30 minutes), or vehicle (lane labeled C). Extracts were c 60
WT
immunoblotted with anti-Flag, and an anti-Erk2 antibody was used as a
c.p.m. bound/dish 50 R151C
loading control. (b) Competition binding analysis of HEK cells coexpressing
(thousands) R160W
WT-MC1R and D5. Cells were transfected with WT-MC1R and pcDNA3 (’) 40
D294H
or WT-MC1R and D5-MC1R (m) and incubated with 1010 M [125I]NDP-MSH 30
and increasing concentrations of unlabeled ligand. Results are expressed as %
20
maximal binding in the absence of competing ligand.
(c) Decreased stimulation of adenylyl cyclase in cells coexpressing WT and 10
D5. Cells cotransfected with WT-MC1R and pcDNA3 (empty bars) or D5
0
(closed bars) were challenged with 107 M NDP-MSH or vehicle (control). -10 -9 -8 -7 -6 -5
Their cAMP content was determined by radioimmunoassay (***Pp0.001). Log[competing peptide] (M)

d
70
analyzed the functional behavior of the RHC forms expressed
individually in HEK 293T cells. Western blot demonstrated 60

similar oligomerization characteristics and expression levels 50


Counts

for the three RHC forms and WT (Figure 7a). In functional 40


D294H
coupling experiments, the RHC variants showed a dramatic 30
WT
reduction of agonist-independent constitutive activity 20
R151C
(Sanchez-Mas et al., 2004). R151C and R160W displayed 10 R160W
significant agonist-induced signaling, and only the D294H
0
receptor was a nearly complete LOF protein (Figure 7b), 100 101 102 103 104
consistent with reports by others (Schioth et al., 1999; Figure 7. Dimerization of RHC variants. (a) Extracts of HEK 293T cells
Ringholm et al., 2004). Competition binding curves indicated expressing Flag-epitope-tagged WT-MC1R or RHC variants were electro-
a lower binding capacity in cells expressing the R151C and phoresed (8 mg/lane) with or without b-mercaptoethanol, and immunoblotted
R160W receptors (Figure 7c) compared with WT, but the with anti-Flag. Comparable loading was checked by staining for Erk2.
affinity of these forms for NDP-MSH was not impaired. (b) Functional coupling of the RHC variants. HEK 293T cells transfected with
Conversely, transfection with D294H yielded a higher WT-MC1R or the indicated variants (0.45 mg/dish) were challenged with
1011 or 107 M NDP-MSH, or vehicle (control). cAMP was determined by
density of binding sites (Bmax 6.270.7 fmol/mg protein for
radioimmunoassay. (c) Displacement curves for binding of [125I]NDP-MSH to
D294H and 3.870.6 fmol/mg protein for WT). The affinity of HEK 293T cells expressing the RHC variants. Cells transfected with the
D294H for NDP-MSH was nevertheless lower than WT, as indicated variants were incubated with 1010 M [125I]NDP-MSH and
demonstrated by a right-shifted displacement curve. increasing concentrations of unlabeled ligand, from 1010 to 106 M. The
Therefore, the density of binding sites was lower for radioactivity associated was measured. Note the two-fold maximal binding
R151C and R160W, intermediate for WT, and higher for obtained for WT or D294H compared with R151C and R160W. (d) Expression
of WT-MC1R and RHC variants on the plasma membrane of HEK 293T cells.
D294H. Since previous reports described very similar binding
Nonpermeabilized cells were stained for the Flag epitope and analyzed by
properties for RHC variants and WT (Schioth et al., 1999), we flow cytometry. A representative overlay histogram is shown. Blank (filled
wished to confirm this trend. Flow-cytometric analysis of curve) refers to cells transfected with WT-MC1R lacking the Flag epitope,
nonpermeabilized cells indicated higher plasma membrane treated in parallel to the samples.

www.jidonline.org 177
BL Sánchez-Laorden et al.
Melanocortin 1 Receptor Homodimerization

−Igepal + Igepal a WT/variant(1:3)


50

40 *** ***

c.p.m. bound/dish
(thousands)
30
WT
20

10

0
pCDNA R151C R160W D294H

b 2.0 WT
D294H
1.5 ***

pmol cAMP/g protein


WT + D294H
R151C 1.0
0.50

***
0.25 ***
***
***
0.00
Control 10–11 10–7

Figure 9. Functional analysis of coexpressed WT-MC1R and RHC variants.


(a) Expression of [125I]NDP-MSH binding sites. Cells were cotransfected with
R160W
WT (0.15 mg/dish) and empty pcDNA or the indicated variants (0.45 mg/dish)
and analyzed for binding of the iodinated ligand. (b) Inhibition of functional
coupling. Cells were transfected with WT-MC1R (0.15 mg/dish), D294H
(0.45 mg/dish), or cotransfected with both forms at a ratio of 1:3 (0.15 mg/dish
WT plus 0.45 mg/dish D294H). At 24 hours after transfection, cells were
challenged for 30 minutes with vehicle (control) or with NDP-MSH at a final
concentration of 1011 or 107 M, and their cAMP contents were determined
(***Pp0.001).

D294H
shown). Therefore, we analyzed the functional effects of
cotransfection with the WT and variant forms. Coexpression
of WT and R151C or R160W caused a modest but significant
decrease in binding, but D294H had no effect (Figure 9a).
Figure 8. Decreased expression of the R151C and R160W MC1R variants on This is consistent with the behavior of the variants expressed
the plasma membrane. HEK 293T cells grown on coverslips were transfected individually. For analysis of functional coupling to adenylyl
with the indicated variants or with WT-MC1R. Staining with an anti-Flag cyclase, we only used the nearly complete LOF D294H form,
monoclonal antibody followed by an Alexa 568-conjugated secondary
as it provides a simpler model system. Cotransfection of WT
antibody was performed without permeabilization, for visualization of
receptor molecules located on the plasma membrane (left column, labeled and D294H abolished completely the constitutive activity of
Igepal), or after permeabilization with 0.5% Igepal, 15 minutes at room MC1R, as well as the response to low concentrations of the
temperature (right column, labeled þ Igepal). agonist (1011 M). However, when cAMP accumulation was
measured following a challenge with a saturating concentra-
tion of NDP-MSH (107 M), no statistically significant
expression for D294H, and lower staining for R151C and differences were observed for cells expressing WT alone, or
R160W (Figure 7d). Moreover, fluorescence microscopy WT and the D294H form (Figure 9b).
showed a fainter labeling of nonpermeabilized cells trans-
fected with R151C and R160W, as compared with cells DISCUSSION
expressing WT or D294H (Figure 8). Conversely, staining of Human MC1R is a highly polymorphic gene accounting for
permeabilized cells was intense for cells expressing R151C or much of the normal variation of pigmentation traits (Rees,
R160W, suggesting that the decreased membrane staining 2003). By determining skin phototype and sun sensitivity,
was not due to lower expression levels but, rather, due to MC1R behaves as a modifier of skin cancer risk (Sturm,
partial intracellular retention (Figure 8). 2002). The MC1R gene encodes for the aMSH receptor, a
Coimmunoprecipitation of differentially epitope-tagged member of the GPCR superfamily. Many members of this
variants and WT followed by Western blot demonstrated family function as dimers or higher order oligomers (Milligan,
that the RHC mutants heterodimerized with WT-MC1R (not 2001; Angers et al., 2002; Breitwieser, 2004). Dimerization

178 Journal of Investigative Dermatology (2006), Volume 126


BL Sánchez-Laorden et al.
Melanocortin 1 Receptor Homodimerization

has been shown to modulate pharmacological properties inhibitory effects were not due to reduced MC1R plasma
such as agonist-binding affinity and coupling efficacy. For membrane expression, as shown by binding studies, and
some GPCRs, dimerization may be a necessary processing therefore most likely reflect changes in the coupling efficacy
step required for efficient cell surface expression (Salahpour of the heterodimer.
et al., 2004). Moreover, dimerization seems to account for In summary, we have shown that MC1R dimerization
the disease-related dominant-negative effects of certain takes place in living cells and has a wide range of functional
GPCR mutants, including MC4R, a member of the melano- consequences, including partial functional complementation
cortin receptor subfamily with high sequence similarity with of inactive forms, dominant-negative effects decreasing WT
MC1R (Biebermann et al., 2003). expression on the cell surface, and, for certain heterodimeric
Our results prove that MC1R undergoes constitutive combinations, a modulation of pharmacological properties,
dimerization in heterologous cells overexpressing the recep- such as affinity for agonists and coupling efficacy. Changes in
tor. This was shown by Western blot and coimmunoprecipi- other functional properties, such as desensitization (Sánchez-
tation, and by the finding of dominant-negative effects and of Más et al., 2005a) or internalization, are also possible and are
a modulation of the pharmacological properties of the being investigated. Therefore, the presence of mutant MC1R
receptor in cells coexpressing different variants. The resis- alleles may have consequences beyond those based on
tance to SDS of oligomeric species, with similar monomer:- dosage effects and haploinsufficiency, thus yielding a
dimer ratios for SDS concentrations ranging from 0.5 to 3.0%, complex spectrum of graded responses to the melanocortins.
as well as the strong dissociating effect of the reducing agent Given the high number of MC1R natural variants and the high
2-mercaptoethanol, suggests a role of disulfide bonding in frequency of variant alleles in some populations, our results
dimerization. Concerns have been raised about the use of highlight an unexpected complexity of the MC1R–melano-
heterologous systems as a model to study GPCR dimeriza- cortin system. This complexity is fully consistent with the
tion, as a strong overexpression may lead to irrelevant diversity of hair color shades, skin phototypes, and sun
oligomerization. Thus, we also demonstrated dimerization in sensitivities (Rees, 2003; Bohm et al., 2005), and may extend
melanoma cells stably expressing near-physiological MC1R to other aspects of skin biology, since MC1R has been shown
levels. Therefore, dimerization of MC1R is not an artifact of to regulate specific functions of skin cells other than
protein overexpression or cell solubilization, and most likely melanocytes (Bohm et al., 2004; Elliott et al., 2004).
reflects the situation in melanocytes.
MC1R dimerization occurs early in receptor ontogeny, MATERIALS AND METHODS
probably in the ER. This is shown by (i) the finding of dimers Expression constructs
of the deglycosylated ‘‘de novo’’ form of MC1R, (ii) the All expression constructs were prepared with the pcDNA3 vector
dimerization behavior of LOF mutants displaying intracellu- (Invitrogen, Carlsbad, CA). The following constructs have been
lar retention, (iii) presence of dimers in cells with a disrupted described: WT-MC1R (Jimenez-Cervantes et al., 2001b), MC1R-Tag
Golgi apparatus following brefeldin A treatment, and (iv) (Sanchez-Más et al., 2002), Flag-MC1R, the C-terminus-deleted
efficient dimerization of an MC1R artificial variant retained in mutants D5-, D12-, and D14-MC1R (Sánchez-Más et al., 2005), and
the ER. Therefore, MC1R dimerization may be required for Flag epitope-labeled L93R and R162P variants (Sánchez-Laorden
export to the plasma membrane, as shown for other GPCRs et al., 2005). The RHC variants R151C, R160W, and D294H were
(Angers et al., 2002; Salahpour et al., 2004). Moreover, the obtained by mutagenesis, using the QuickChange kit (Stratagene, La
dominant-negative effect exerted by LOF mutants with Jolla, CA) and WT Flag-MC1R as template. An MC1R-HA construct
intracellular retention is best explained by trapping WT units bearing a single copy of the hemagglutinin (HA) epitope immedi-
in internal compartments. A similar mechanism has been ately after the C-terminal W317 residue was obtained by PCR
postulated for dominant-negative mutants of other GPCRs amplification of WT-MC1R, using the same forward primer as for
(Angers et al., 2002; Salahpour et al., 2004). Interestingly, a cloning of the WT gene and a reverse primer containing the HA
heterozygous human melanoma cell line bearing the L93R epitope sequence. Sequences of mutagenic primers are available on
allele does not respond to agonists by activating cAMP request. All constructs were verified by automated sequencing.
production, in spite of expressing an active adenylyl cyclase
(Sanchez-Más et al., 2002), consistent with a dominant- Cell culture and transfection
negative effect of L93R. HEK 293T cells were cultured in RPMI 1640 with 10% fetal calf
We considered the possible functional consequences of serum (FCS), 100 U/ml penicillin, and 100 mg/ml streptomycin. Cells
MC1R dimerization. Although no cooperativity in agonist grown to approximately 50% confluence were transfected with
binding was demonstrated by Hill analysis, cotransfection of Lipofectamine 2000 (Invitrogen). HBL human melanoma cells were
WT and the D5 mutant yielded a form of different affinity and grown in DMEM supplemented with antibiotics and 10% FCS. Stable
lower efficacy in activating cAMP synthesis as compared with transfectants were obtained with Lipofectamine. Clones were
WT. This demonstrates that MC1R dimerization can yield selected with geneticin (Mas et al., 2003).
functional units with slightly different pharmacological
properties. This was further shown by studies with the Functional assays
frequent RHC forms. The D294H variant abolished constitu- All experiments were performed in independent duplicate or
tive signaling as well as signaling at low agonist concentra- triplicate dishes of 12-well plates and repeated at least twice, so
tions in HEK cells coexpressing WT and variant MC1R. These that each experimental point is the mean7standard error of mean for

www.jidonline.org 179
BL Sánchez-Laorden et al.
Melanocortin 1 Receptor Homodimerization

nX4. Radioligand-binding assays were carried out with [125I]NDP- secondary antibody. Samples were examined with a Zeiss Axiophot
MSH, specific activity 2000 Ci/mmol (Amersham Pharmacia Bio- fluorescence microscope.
tech, Little Chalfont, Buckinghamshire, UK), and unlabeled NDP-
MSH up to 106 M when required. The experimental procedure has Immunoelectron microscopy
been described (Sanchez-Más et al., 2002). Functional coupling was HEK 293T cells expressing MC1R-Tag were fixed with 2%
analyzed in cells serum-deprived for 24 hours prior to a 30-minute paraformaldehyde and 0.2% glutaraldehyde in 0.1 M sodium
stimulation with 1013–107 M NDP-MSH. Cells were lysed in phosphate buffer, pH 7.4, washed, embedded in 10% gelatin,
preheated 0.1 N HCl (701C) and cAMP was measured with a cooled on ice, and cut into 1 mm3 blocks. Blocks were infused with
radioimmunoassay kit (Amersham) as described (Jimenez-Cervantes 2.3 M sucrose at 41C overnight, frozen in liquid nitrogen, and stored
et al., 2001a; Sanchez-Más et al., 2002). until cryoultramicrotomy. Sections (B50 nm thick) were cut at
1201C in a Leica Ultracut T/FCS. Ultrathin sections were picked
Immunoprecipitation and Western blot up in a mix of 1.8% methylcellulose and 2.3 M sucrose (1:1).
HEK 293T cells were transfected with MC1R-HA, alone or in Cryosections were collected on carbon- and formvar-coated copper
combination with Flag epitope-labeled MC1R variants. Approxi- grids and incubated with an anti-HA monoclonal antibody,
mately 2  106 cells were washed twice with phosphate-buffered followed by a rabbit anti-mouse antiserum and 10 nm protein
saline and solubilized in 200 ml of iodoacetamide-containing A-gold (Martinez-Alonso et al., 2005). Sections were treated with 1%
solubilization buffer (50 mM Tris-HCl, pH 8, 1% Igepal CA-640, glutaraldehyde, counterstained with uranyl acetate, pH 7.0, and
1 mM EDTA, 0.1 mM phenylmethylsulfonyl fluoride, and 10 mM iodo- embedded in methyl cellulose-uranyl acetate, pH 4.0 (9:1). Grids
acetamide). The sample was centrifuged (105,000  g, 30 minutes) were examined with a Philips Tecnai 12 electron microscope.
and the supernatant was precleared with 20 ml of protein G-agarose
slurry, for 1 hour, followed by centrifugation. Monoclonal anti-HA Other procedures
antibody (2 mg) (clone HA-7, from Sigma, St Louis, MO) and protein For data normalization, cells were solubilized in 10 mM phosphate
G-agarose slurry (20 ml) were added. After a 3-hour incubation, the buffer, pH 7, and 1% Igepal CA-640, containing 0.1 mM EDTA and
sample was centrifuged and the pellet was washed five times 0.1 mM phenylmethylsulfonyl fluoride. The extract was centrifuged
with 10 mM Tris-HCl, pH 7.5, 500 mM KCl, 300 mM NaCl, and and the supernatant used for protein determination by the
0.05% Triton X-100. Elution was performed in 20 ml of 2% SDS for bicinchoninic acid method. For deglycosylation studies, extracts
30 minutes at room temperature. Eluted supernatants were were incubated at 371C for 4 hours with endoglycosidase H (5 U) in
mixed in a ratio of 2:1 with an electrophoresis sample buffer 50 mM phosphate buffer, pH 7.0, 10 mM EDTA, and 0.1% SDS, as
(180 mM Tris-HCl, pH 6.8, 15% glycerol, 9% SDS, 0.075% described (Olivares et al., 2003).
bromophenol blue, and 7.5% b-mercaptoethanol). Electrophoresis Differences between experimental data were assessed for
and Western blotting were performed as described (Sanchez-Más significance using the Student’s t-test. Curve fitting and statistical
et al., 2002; Sánchez-Laorden et al., 2005). Direct Western blot was analysis were performed with the GraphPad Prism package
performed with extracts of cells expressing Flag-MC1R variants. (GraphPad Software, San Diego, CA).
Blots were probed with anti-Flag M2 monoclonal antibody- The medical ethical committee of the University of Murcia
peroxidase conjugate (Sigma) and stained with a chemiluminescent approved all described studies.
substrate (Amersham).
CONFLICT OF INTEREST
Flow-cytometric analysis The author states no conflict of interest.
Approximately 250,000 HEK 293T cells expressing the relevant
constructs were incubated for 30 minutes in a volume of 100 ml with ACKNOWLEDGMENTS
anti-Flag M2 monoclonal antibody (Sigma) at a dilution of 1:25. J.S.M. and B.L.S.-L. contributed equally to this work. This work was supported
Cells were washed twice with 2% FCS and 0.01% NaN3 in by Grant SAF2003-03411 (to J.C.G.B.) from the CICYT, Spain, and FEDER
funds (European Community). J.S.M. is a fellow of the Fundación Séneca
phosphate-buffered saline, and incubated with a phycoerythrin- (Comunidad Autónoma de la Región de Murcia). B.L.S.-L. and E.M.-A. hold
labeled anti-mouse IgG (1:50) for 30 minutes at 41C. Cells were fellowships from the Ministry of Education (Spain). Work by E.M.-A. and
washed, resuspended in 500 ml of 0.4% paraformaldehyde in J.A.M.-M. was supported by Grant BFU2004-05568 (CICYT, Spain). We thank
Professor G. Ghanem (Free University of Brussels, Belgium) for HBL cells and
phosphate-buffered saline, and analyzed in a Becton Dickinson
Dr C. Martı́nez (Centro de Hemodonación, Hospital Reina Sofı́a, Murcia,
FACScan. Spain) for thromboxane a receptor cDNA.

Immunofluorescence microscopy
Cells grown on coverslips were fixed with paraformaldehyde (4%) in REFERENCES
phosphate-buffered saline. To assess the effects of brefeldin A, Abdel-Malek Z, Scott MC, Suzuki I, Tada A, Im S, Lamoreux L et al. (2000)
control and brefeldin A-treated (1 mg/ml, 30 minutes) cells were The melanocortin-1 receptor is a key regulator of human cutaneous
permeabilized (0.1% saponine) and immunostained for the Golgi pigmentation. Pigment Cell Res 13(Suppl 8):156–62
matrix protein GM130 with a monoclonal antibody and an Alexa Angers S, Salahpour A, Bouvier M (2002) Dimerization: an emerging concept
for G protein-coupled receptor ontogeny and function. Annu Rev
488-conjugated secondary antibody (Martinez-Alonso et al.,
Pharmacol Toxicol 42:409–35
2005). For detection of WT and variant MC1R, cells expressing
Barsh G, Gunn T, He L, Schlossman S, Duke-Cohan J (2000) Biochemical and
the relevant forms were labeled with the anti-Flag M2 mono- genetic studies of pigment-type switching. Pigment Cell Res 13(Suppl
clonal antibody (Sigma), followed by an Alexa 568-conjugated 8):48–53

180 Journal of Investigative Dermatology (2006), Volume 126


BL Sánchez-Laorden et al.
Melanocortin 1 Receptor Homodimerization

Bastiaens M, ter Huurne J, Gruis N, Bergman W, Westendorp R, Vermeer BJ Milligan G (2001) Oligomerisation of G-protein-coupled receptors. J Cell Sci
et al. (2001a) The melanocortin-1-receptor gene is the major freckle 114:1265–71
gene. Hum Mol Genet 10:1701–8 Olivares C, Solano F, Garcia-Borron JC (2003) Conformation-dependent post-
Bastiaens MT, ter Huurne JA, Kielich C, Gruis NA, Westendorp RG, Vermeer translational glycosylation of tyrosinase. Requirement of a specific
BJ et al. (2001b) Melanocortin-1 receptor gene variants determine the interaction involving the CuB metal binding site. J Biol Chem
risk of nonmelanoma skin cancer independently of fair skin and red hair. 278:15735–43
Am J Hum Genet 68:884–94 Palmer JS, Duffy DL, Box NF, Aitken JF, O’Gorman LE, Green AC et al. (2000)
Biebermann H, Krude H, Elsner A, Chubanov V, Gudermann T, Gruters A Melanocortin-1 receptor polymorphisms and risk of melanoma: is the
(2003) Autosomal-dominant mode of inheritance of a melanocortin-4 association explained solely by pigmentation phenotype? Am J Hum
receptor mutation in a patient with severe early-onset obesity is due to a Genet 66:176–86
dominant-negative effect caused by receptor dimerization. Diabetes Rees JL (2003) Genetics of hair and skin color. Annu Rev Genet 37:67–90
52:2984–8
Ringholm A, Klovins J, Rudzish R, Phillips S, Rees JL, Schioth HB (2004)
Bohm M, Raghunath M, Sunderkotter C, Schiller M, Stander S, Brzoska T et al. Pharmacological characterization of loss of function mutations of the
(2004) Collagen metabolism is a novel target of the neuropeptide alpha- human melanocortin 1 receptor that are associated with red hair. J Invest
melanocyte-stimulating hormone. J Biol Chem 279:6959–66 Dermatol 123:917–23
Bohm M, Wolff I, Scholzen TE, Robinson SJ, Healy E, Luger TA et al. (2005) Salahpour A, Angers S, Mercier JF, Lagace M, Marullo S, Bouvier M (2004)
alpha-Melanocyte-stimulating hormone protects from ultraviolet radia- Homodimerization of the beta2-adrenergic receptor as a prerequisite for
tion-induced apoptosis and DNA damage. J Biol Chem 280:5795–802 cell surface targeting. J Biol Chem 279:33390–7
Box NF, Wyeth JR, O’Gorman LE, Martin NG, Sturm RA (1997) Character- Sánchez-Laorden B, Sanchez-Mas J, Turpin MC, Garcia-Borron JC, Jimenez-
ization of melanocyte stimulating hormone receptor variant alleles in Cervantes C (2005) Variant aminoacids in different domains of the
twins with red hair. Hum Mol Genet 6:1891–7 human melanocortin 1 receptor impair cell surface expression. Cell Mol
Breitwieser GE (2004) G protein-coupled receptor oligomerization: implica- Biol (in press)
tions for G protein activation and cell signaling. Circ Res 94:17–27 Sánchez-Más J, Guillo LA, Zanna P, Jimenez-Cervantes C, Garcia-Borron JC
Busca R, Ballotti R (2000) Cyclic AMP a key messenger in the regulation of (2005a) Role of G protein-coupled receptor kinases in the homologous
skin pigmentation. Pigment Cell Res 13:60–9 desensitization of the human and mouse melanocortin 1 receptors. Mol
Di Donato P, Napolitano A (2003) 1,4-Benzothiazines as key intermediates in Endocrinol 19:1035–48
the biosynthesis of red hair pigment pheomelanins. Pigment Cell Res Sánchez-Más J, Hahmann C, Gerritsen I, Garcia-Borron JC, Jimenez-
16:532–9 Cervantes C (2004) Agonist-independent, high constitutive activity of
Elliott RJ, Szabo M, Wagner MJ, Kemp EH, MacNeil S, Haycock JW (2004) the human melanocortin 1 receptor. Pigment Cell Res 17:386–95
alpha-Melanocyte-stimulating hormone, MSH 11–13 KPV and adreno- Sánchez-Más J, Sanchez-Laorden BL, Andreu Guillo L, Jimenez-Cervantes C,
corticotropic hormone signalling in human keratinocyte cells. J Invest Garcia-Borron JC (2005b) The melanocortin 1 receptor carboxyl
Dermatol 122:1010–9 terminal pentapeptide is essential for MC1R expression. Peptides
Jiménez-Cervantes C, Germer S, Gonzalez P, Sanchez J, Sanchez CO, Garcia- 26:1848–57
Borron JC (2001a) Thr40 and Met122 are new partial loss-of-function natural Sanchez-Más J, Olivares SC, Ghanem G, Haycock J, Lozano Teruel JA,
mutations of the human melanocortin 1 receptor. FEBS Lett 508:44–8 Garcia-Borron JC et al. (2002) Loss-of-function variants of the human
Jiménez-Cervantes C, Olivares C, Gonzalez P, Morandini R, Ghanem G, melanocortin-1 receptor gene in melanoma cells define structural
Garcia-Borron JC (2001b) The Pro162 variant is a loss-of-function determinants of receptor function. Eur J Biochem 269:6133–41
mutation of the human melanocortin 1 receptor gene. J Invest Dermatol Schioth HB, Phillips SR, Rudzish R, Birch-Machin MA, Wikberg JE, Rees JL
117:156–8 (1999) Loss of function mutations of the human melanocortin 1 receptor
Kadekaro AL, Kavanagh RJ, Wakamatsu K, Ito S, Pipitone MA, Abdel-Malek are common and are associated with red hair. Biochem Biophys Res
ZA (2003) Cutaneous photobiology. The melanocyte vs. the sun: who Commun 260:488–91
will win the final round? Pigment Cell Res 16:434–47 Smith R, Healy E, Siddiqui S, Flanagan N, Steijlen PM, Rosdahl I et al. (1998)
Mandrika I, Petrovska R, Wikberg J (2005) Melanocortin receptors form Melanocortin 1 receptor variants in an Irish population. J Invest Dermatol
constitutive homo- and heterodimers. Biochem Biophys Res Commun 111:119–22
326:349–54 Sturm RA (2002) Skin colour and skin cancer – MC1R, the genetic link.
Martinez-Alonso E, Egea G, Ballesta J, Martinez-Menarguez JA (2005) Melanoma Res 12:405–16
Structure and dynamics of the Golgi complex at 15 degrees C: low Sturm RA, Duffy DL, Box NF, Chen W, Smit DJ, Brown DL et al. (2003) The
temperature induces the formation of Golgi-derived tubules. Traffic role of melanocortin-1 receptor polymorphism in skin cancer risk
6:32–44 phenotypes. Pigment Cell Res 16:266–72
Mas JS, Gerritsen I, Hahmann C, Jimenez-Cervantes C, Garcia-Borron JC Valverde P, Healy E, Sikkink S, Haldane F, Thody AJ, Carothers A et al. (1996)
(2003) Rate limiting factors in melanocortin 1 receptor signalling through The Asp84Glu variant of the melanocortin 1 receptor (MC1R) is
the cAMP pathway. Pigment Cell Res 16:540–7 associated with melanoma. Hum Mol Genet 5:1663–6

www.jidonline.org 181

You might also like