ArticlePDF Available

Epistatic connections between MITF and endothelin signaling in Waardenburg syndrome and other pigmentary disorders

Wiley
The FASEB Journal
Authors:

Abstract and Figures

Waardenburg syndrome (WS) is an inherited sensorineural deafness condition in humans caused by melanocyte deficiencies in the inner ear and forelock. Mutation of microphthalmia-associated transcription factor (MITF) is known to produce WS type IIA whereas mutations of either endothelin (EDN) or its receptor endothelin receptor B (EDNRB) produce WS type IV. However, a link between MITF haploinsufficiency and EDN signaling has not yet been established. Here we demonstrate mechanistic connections between EDN and MITF and their functional importance in melanocytes. Addition of EDN to cultured human melanocytes stimulated the phosphorylation of MITF in an EDNRB-dependent manner, which was completely abolished by mitogen-activated protein kinase kinase inhibition. The expression of melanocyte-specific MITF mRNA transcripts was markedly augmented after incubation with EDN1 and was followed by increased expression of MITF protein. Up-regulated expression of MITF was found to be mediated via both the mitogen-activated protein kinase-p90 ribosomal S6 kinase-cAMP response element-binding protein (CREB) and cAMP-protein kinase A-CREB pathways. In addition, EDNRB expression itself was seen to be dependent on MITF. The functional importance of these connections is illustrated by the ability of EDN to stimulate expression of melanocytic pigmentation and proliferation markers in an MITF-dependent fashion. Collectively these data provide mechanistic and epistatic links between MITF and EDN/EDNRB, critical melanocytic survival factors and WS genes.
Content may be subject to copyright.
The FASEB Journal Research Communication
Epistatic connections between microphthalmia-
associated transcription factor and endothelin
signaling in Waardenburg syndrome and other
pigmentary disorders
Kayo Sato-Jin,*
,1
Emi K. Nishimura
†,‡,1
, Eijiro Akasaka,* Wade Huber,
Hajime Nakano,* Arlo Miller,
Jinyan Du,
Min Wu,
Katsumi Hanada,*
Daisuke Sawamura,* David E. Fisher,
and Genji Imokawa*
,§,2
*Department of Dermatology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori,
Japan;
Department of Stem Cell Medicine, Cancer Research Institute, Kanazawa University,
Kanazawa, Ishikawa, Japan;
Dana-Farber Cancer Institute Melanoma Program and Children’s
Hospital Boston, Department of Pediatric Hematology/Oncology, Harvard Medical School, Boston,
Massachusetts, USA; and
§
Tokyo University of Technology, School of Bionics, Hachioji, Tokyo, Japan
ABSTRACT Waardenburg syndrome (WS) is an in-
herited sensorineural deafness condition in humans
caused by melanocyte deficiencies in the inner ear and
forelock. Mutation of microphthalmia-associated tran-
scription factor (MITF) is known to produce WS type
IIA whereas mutations of either endothelin (EDN) or
its receptor endothelin receptor B (EDNRB) produce
WS type IV. However, a link between MITF haploinsuf-
ficiency and EDN signaling has not yet been estab-
lished. Here we demonstrate mechanistic connections
between EDN and MITF and their functional impor-
tance in melanocytes. Addition of EDN to cultured
human melanocytes stimulated the phosphorylation of
MITF in an EDNRB-dependent manner, which was
completely abolished by mitogen-activated protein ki-
nase kinase inhibition. The expression of melanocyte-
specific MITF mRNA transcripts was markedly aug-
mented after incubation with EDN1 and was followed
by increased expression of MITF protein. Up-regulated
expression of MITF was found to be mediated via
both the mitogen-activated protein kinase-p90 ribo-
somal S6 kinase-cAMP response element-binding pro-
tein (CREB) and cAMP-protein kinase A-CREB path-
ways. In addition, EDNRB expression itself was seen to
be dependent on MITF. The functional importance of
these connections is illustrated by the ability of EDN to
stimulate expression of melanocytic pigmentation and
proliferation markers in an MITF-dependent fashion.
Collectively these data provide mechanistic and epi-
static links between MITF and EDN/EDNRB, critical
melanocytic survival factors and WS genes.—Sato-Jin,
K., Nishimura, E. K., Akasaka, E., Huber, W., Nakano,
H., Miller, A., Du, J., Wu, M., Hanada, K., Sawamura,
D., Fisher, D. E., Imokawa, G. Epistatic connections
between microphthalmia-associated transcription fac-
tor and endothelin signaling in Waardenburg syndrome
and other pigmentary disorders. FASEB J. 22, 1155–1168
(2008)
Key Words: cAMP response element-binding protein CREB cy-
clin-dependent kinase 2 melanocyte endothelin B receptor
Highly organized but complex interactions be-
tween environmental cues, signal transduction path-
ways, and transcription factors underlie the develop-
ment of a cell lineage. Mammalian pigmentation genes
provide an attractive system in which to analyze such
interactions in vivo because melanocyte distribution
and pigmentation are visually detectable. These prop-
erties have allowed for the identification of numerous
coat color mutations in mice, including a smaller
number of white spotting genes, which are essential for
melanocyte development or survival (as opposed to
pigmentation per se). In human skin, mutations or
hyperexpression of several pigmentation genes may be
associated with hypo- or hyperpigmentary disorders.
Melanocyte progenitors exit from the dorsal neural
tube and migrate toward the ventral midline through
the dorsolateral pathway to colonize the dermis, epi-
dermis, and hair follicles of the skin. They undergo
proliferation and differentiation dependent on envi-
ronmental cues during the colonization process. Once
in the skin, melanocytes may respond to exogenous
signals to stimulate their growth and melanogenesis,
thereby altering skin pigmentation levels. The molecu-
lar mechanisms whereby these cues are translated into
altered melanocyte cell fate, proliferation, or differen-
tiation are not well understood.
Mutations of the white spotting genes have been
similarly found in human hereditary deafness/depig-
1
These authors contributed equally to this work.
2
Correspondence: Tokyo University of Technology, School of
Bionics, Katayanagi Institute-W204, 1404-1 Katakura Hachioji,
Tokyo 192-0982 Japan. E-mail: imokawag@dream.ocn.ne.jp
doi: 10.1096/fj.07-9080com
11550892-6638/08/0022-1155 © FASEB
mentation syndromes. Recent studies have shown that
the transcription factor PAX3, which is responsible for
Waardenburg syndrome (WS) type I or III depending
on the precise mutation, modulates expression of the
central melanocytic transcriptional regulator microph-
thalmia-associated transcription factor (MITF) (1–3).
Similarly, the SOX10 transcription factor modulates
MITF expression and is mutated in WS type IV (1, 2, 4,
5). Additional studies have shown that KIT, which is
mutated in the piebaldism plus deafness condition
(Woolf’s syndrome), triggers a signaling pathway that
leads to MITF modulation via phosphorylation (6, 7).
Mutation of endothelin3 (EDN3) or its receptor
endothelin receptor B (EDNRB) causes loss of melano-
cytes as well as enteric ganglion cells (which are also
neural crest derivatives) both in humans with WS type
IV and in mutant mice (8 –10). EDNRB is required
between embryonic day 10 and 12.5 when melano-
blasts, which have just exited from the dorsal neural
tube, are proliferating and dispersing along the dorso-
lateral pathway (11). The significant increase in mela-
noblasts seen in wild-type embryos during this time
window is absent in Ednrb
sl/s-l
and Edn3
ls/ls
mutant
mice (12, 13), indicating that EDN3/EDNRB signaling
is critical for melanoblast proliferation (and subse-
quent survival) during development.
EDN peptide derived from endothelial cells is a
potent vasoconstrictor of vascular smooth muscle (14).
We demonstrated previously that human epidermal
keratinocytes secrete EDN1 and EDN2 in response to
ultraviolet (UV) irradiation (15, 16). Those EDNs can
act on melanocytes as melanogens in conjunction with
their mitogenic properties, playing an essential role in
stimulating epidermal pigmentation in several hyper-
pigmentary disorders including UVB melanosis (17). A
number of in vitro experiments have also demonstrated
that EDN1 as well as the related factor EDN3 (which
binds and activates the same receptor) promote mela-
nocyte proliferation and differentiation in cell culture
experiments (18). Characterization of the signaling
pathways involved in those responses demonstrated
that the EDN-induced signal transduction pathway
dominates the activation of protein kinase C (PKC)
through EDNRB, resulting in activation of the mitogen-
activated protein kinase (MAPK) pathway via a conver-
gent point of Raf-1 (19 –21). EDN signaling is also
thought to target activation of p90 ribosomal S6 kinase
(RSK) family kinases downstream of MAPK (22). In
contrast to the action of EDN1, ligands such as basic
fibroblast growth factor (bFGF) and stem cell factor
(SCF), which are associated with tyrosine kinases, stim-
ulate melanization of human melanocytes only weakly
despite their distinct actions as mitogens (23–25).
Therefore, the signaling mechanisms involved in the
dual and potent biological effects of EDN1 on prolifer-
ation and differentiation of human melanocytes re-
mains unclear, and molecular targets of EDNs down-
stream of MAPK that underlie EDN-induced
melanoblast or melanocyte proliferation/differentia-
tion are not well understood.
MITF, one of the WS genes, encodes a tissue-re-
stricted transcription factor of the basic helix-loop-helix
leucine-zipper type (26), which recognizes E-box-con-
taining promoter/enhancer elements (27, 28). It reg-
ulates melanocytic pigmentation as a transcription fac-
tor regulating the key melanogenic enzymes tyrosinase,
TRP1 and TRP2 (29 –32), and is also associated with
melanocyte survival (26, 28). Mutation of MITF results
in WS type IIA in humans (33–35), causing hypopig-
mentation of the skin and hair and corresponding
white coat color and deafness in mice. Interestingly,
germline mutations at loci encoding MITF, EDNRB, or
its ligand EDN3, lead to strikingly similar defects in
melanocytes (10, 36, 37). The phenotypic overlap be-
tween MITF and EDN/EDNRB mutations in humans and
mouse suggests the existence of a common functionally
important pathway for melanocyte development and its
cellular responses to environmental stimuli.
Recently, new roles for MITF in melanocytes have
been proposed, such as regulating the apoptotic inhib-
itor BclII (38), the cyclin-dependent kinase inhibitor
gene p21 (Cip1) (39), p16/Ink4a (40), and/or cyclin-
dependent kinase 2 (CDK2) (41) in addition to regu-
lating transcription of melanogenic enzymes. Thus,
characterizing the dynamics of MITF and its modula-
tion within melanocytes during EDN stimulation, which
reflects in vivo abnormal pigmentary events, would
provide insights into the regulatory roles of melano-
cyte-specific MITF (MITF-M) in melanocytes. Here, we
used EDN stimulation of intracellular signaling to
examine the dynamics of MITF-M in normal human
melanocytes. We demonstrate that EDN signals modu-
late the MITF via three kinetically distinct mechanisms,
thereby connecting an essential growth factor/receptor
to a master transcription factor MITF, which is pivotal
to melanocyte fate.
MATERIALS AND METHODS
Cell culture and treatment
Primary human melanocytes from neonatal foreskins (pro-
vided by Dr. Ruth Halaban, Yale University, New Haven, CT,
USA) were maintained between passages 1 and 3 in F10
medium (GIBCO-Life Technologies, Inc., Rockville, MD,
USA) supplemented with 7% fetal bovine serum (FBS),
penicillin/streptomycin/glutamine (GIBCO-Life Technolo-
gies, Inc.), 1 10
4
M 3-isobutyl-1-methylxanthine (Sigma-
Aldrich Corp., St. Louis, MO, USA), 50 ng/ml 12-O-tetra-
decanoyl phorbol-13-acetate (Sigma-Aldrich Corp.), 1 M
Na
3
VO
4
, and 1 10
3
M N
6
,2-O-dibutyryladenosine 3:5-
cyclic monophosphate (Sigma-Aldrich Corp.). Primary hu-
man melanocytes from another source (Cascade Biologics,
Portland, OR, USA) were maintained in medium 254 (Cas-
cade Biologics) supplemented with 3 ng/ml recombinant
bFGF, 5 g/ml insulin, 0.18 g/ml hydrocortisone, 5 g/ml
transferrin, 3 g/ml heparin, 10 ng/ml phorbol-12-myristate-
13-acetate (PMA), 0.2% (v/v) bovine pituitary extract (BPE),
and 0.5% (v/v) FBS (Cascade Biologics). The human mela-
noma cell line 501 MEL (gift of Dr. Ruth Halaban) was grown
in F10 medium with 10% FBS plus penicillin/streptomycin/
glutamine. In evaluation of signaling changes, human pri-
1156 Vol. 22 April 2008 SATO-JIN ET AL.The FASEB Journal
mary melanocytes were seeded in medium 254 or F10 me-
dium and at 24 h before the addition of ligands, the medium
was replaced with medium 254 depleted of FBS, PMA, bFGF,
and BPE or F10 medium supplemented only with penicillin/
streptomycin/glutamine. Human primary melanocytes were
stimulated with 10 nM EDN1 (Calbiochem, San Diego, CA,
USA or Sigma-Aldrich Corp.)/EDN3 (Calbiochem), 20
ng/ml recombinant human SCF (R&D Systems, Minneapolis,
MN, USA), and forskolin (FSK) (20 M) with or without 20
g/ml cycloheximide, 20 or 40 M mitogen-activated protein
kinase kinase (MEK) inhibitor PD98059 (New England Bio-
Labs, Ipswich, MA, USA or Sigma-Aldrich Corp.), 1 M PKC
inhibitor Go¨6983 (Calbiochem), 5 or 10 M protein kinase A
(PKA) inhibitor H89 (D. Western Therapeutics Institute, Inc,
Nagoya, Japan), 10 M p38 MAPK inhibitor SB203580 (Cal-
biochem), 100 nM phosphoinositol 3-kinase (PI3 kinase)
inhibitor wortmannin (Calbiochem), 1 M Akt inhibitor
SH-6 (Calbiochem), or 25 M proteasome inhibitor MG132
(Calbiochem) for the indicated times after 24 h of starvation
as described above. The 501 MEL human melanoma cells
were stimulated by EDN1/3 and FSK with or without the MEK
inhibitor PD98059 in F10 medium supplemented only with
penicillin/streptomycin/glutamine.
Adenovirus infection
Adenoviruses were used as described previously (42) and
were engineered to overexpress either wild-type human
MITF, R215del (dominant-negative MITF), or a green fluo-
rescence protein (GFP)/wee1-truncation hybrid (which tar-
gets GFP to the nucleus as vector control), all under the
control of the elongation factor promoter (42). Subconflu-
ent primary human melanocytes were incubated with concen-
trated adenoviruses in serum-free F10 medium supplemented
with 10 mM MgCl
2
for 30 min at a multiplicity of infection of
200 for each virus. After infection the medium was replaced
by fresh medium and cultured for indicated times until
stimulation.
Quantitative reverse transcriptase (RT) -polymerase chain
reaction (PCR)/TaqMan
RNA was isolated using the Ambion RNAqueous kit (Ambion,
Austin, TX, USA) and quantitated by spectrophotometry
(Beckman, Miami, FL, USA). TaqMan One-Step RT-PCR
Master Mix Reagents as well as GAPDH Control Reagents
(Applied Biosystems, Foster City, CA, USA) were used for
quantitative RT-PCR reactions, each containing 100 ng of
total sample RNA. Reactions were run for 40 cycles under the
following conditions: stage 1: 48°C, 30 min; stage 2: 95°C, 10
min; stage 3: 94°C, 20 s; and stage 4: 62°C, 1 min. The
message of human MITF, CDK2, and silver homolog (SILV)
was detected using the following primers (Integrated DNA
Technologies, Coralville, IA, USA) and TaqMan probe (Ap-
plied Biosystems): CDK2: forward 5-ATG GAG AAC TTC
CAA AAG GTG GA-3, reverse 5-CAG GCG GAT TTT CTT
AAG CG-3 primers and CDK2 probe 5-6-FAM-ATC GGA
GAG GGC ACG TAC GGA GTT GT-TAMRA-3; SILV: forward
5-TCT GGG CTG AGC ATT GGG-3, reverse 5-AGA CAG
TCA CTT CCA TGG TGT GTG-3 primers and SILV probe
5-6-FAM-CAG GCA GGG CAA TGC TGG GC-TAMRA-3; and
EDNRB: forward 5-TGA GTC TAT GTG CTC TGA GTA TTG
ACA-3, reverse 5-ACC TAT GGC TTC AGG GAC AGC-3
primers and EDNRB probe 5-6-FAM-TGT TTT GAT TTG
GGT GGT CTC TGT GGT TCT-TAMRA-3. All reactions were
run in triplicate on an ABI-PRISM 7700 instrument (Applied
Biosystems), and gene message levels were normalized to
GAPDH expression.
Quantitative RT-PCR/SYBR Green
RNA was isolated using an RNeasy mini-protocol kit (Qiagen,
Valencia, CA, USA) and quantitated by spectrophotometry
(Beckman). The first-strand cDNA was synthesized from total
RNA using Rever Tra Ace (TOYOBO, Nagoya, Japan). Reac-
tions were run under the following conditions: stage 1 30°C,
10 min; stage 2: 42°C, 20 min; and stage 3: 99°C, 5 min. iQ
SYBR Green Supermix Reagents (Bio-Rad Laboratories, Her-
cules, CA, USA) were used for quantitative RT-PCR reactions,
each containing 10 ng of total sample RNA. Reactions were
run for 40 cycles under the following conditions: stage 1:
95°C, 15 s; stage 2: 61°C for 30 s; and stage 3: 72°C, 30 s. The
message of human MITF-M and GAPDH was detected using
the following primers (FASMAC, Atsugi, Japan). MITF-M:
forward 5-TCC GTC TCT CAC TGG ATT GGT G-3, reverse
5-CGT GAA TGT GTG TTC ATG CCT GG-3; and GAPDH:
forward 5-GCC ATC AAT GAC CCC TTC ATT-3, reverse
5-TTG ACG GTG CCA TGG AAT TT-3. All reactions were
run in triplicate on a DNA Engine Opticon 2 Real-Time PCR
Detection System (Bio-Rad Laboratories), and gene message
levels were normalized to GAPDH expression.
Gel electrophoresis, immunoblotting,
and immunoprecipitation
For immunoblot analysis, cells were lysed in lysis buffer [50
mM Tris (pH 7.6), 150 mM NaCl, and 1% Triton X-100] plus
protease inhibitors (Complete mini-tablets; Boehringer
Mannheim, Mannheim, Germany) and phosphatase inhibi-
tors (20 mM NaPP, 10 mM NaF, and 1 mM Na
3
VO
4
, and 1
mM Na
3
VO
4
) and centrifuged at 13,000 rpm for 15 min. The
supernatant was harvested and lysed in 2 loading buffer
[125 mM Tris (pH 6.8), 4.6% sodium dodecyl sulfate (SDS),
20% glycerol, and 0.04% pyronin Y]. The mixture was then
boiled for 5 min. Samples were solubilized in SDS sample
buffer plus 50 mM dithiothreitol and boiled for 5 min. Total
protein from cell cultures of human melanocytes was sub-
jected to Western blotting with anti-MITF (C5; NeoMarkers,
Fremont, CA, USA), anti-phospho-extracellular signal-regu-
lated kinase (ERK) 1/2 (Cell Signaling Technologies, Dan-
vers, MA, USA or Santa Cruz Biotechnology, Santa Cruz, CA,
USA), anti-phospho-cAMP response element-binding protein
(CREB) (Cell Signaling Technologies), anti-phospho-
MEK1/2 (Cell Signaling Technologies), anti-CREB (Cell Sig-
naling Technologies), anti-ERK (Santa Cruz Biotechnolo-
gies), anti-MEK (Santa Cruz Biotechnologies), anti--actin
(Sigma-Aldrich Corp.), and anti--tubulin (Sigma-Aldrich
Corp.) antibody. Samples were run on SDS-PAGE gels, trans-
ferred onto nitrocellulose, blocked with 5% nonfat dry milk
in Tris-buffered saline (TBST) and probed with the respective
antibodies in TBST overnight at 4°C. Membranes were
washed 3 times for 15 min with TBST, probed with peroxi-
dase-conjugated secondary antibodies (Amersham Pharmacia
Biotech, Piscataway, NJ, USA or ICN Biomedicals Inc, Solon,
OH, USA) washed three times for 30 min in TBST and
developed by ECL (Amersham).
For immunoprecipitation, cells were lysed with lysis buffer
as above plus protease inhibitors. The soluble fraction was
incubated overnight at 4°C with anti-MITF antibody (D5) or
anti-phospho-S73 MITF (43), and subsequently protein G
agarose beads (GIBCO-Life Technologies, Inc.) were added
and the solution was incubated for an additional1hat4°C.
Beads were washed three times with cold PBS, resuspended in
SDS sample buffer, and boiled for 5 min. The eluted proteins
were resolved on SDS-PAGE and immunoblotted with anti-
MITF antibody (C5).
1157MOLECULAR INTERACTIONS BETWEEN MITF AND ENDOTHELIN
Transient in vitro reporter assay
For reporter assays, 501 MEL cells were plated in a 96-well
black and white tissue culture plate (Wallac, Waltham, MA,
USA) to a density of 1 10
4
cells/well. The following day,
cells were transfected with 40 ng of promoter [pGL2.basic,
pGL2.MITF cAMP response element (CRE) wild-type (CRE
wt), or pGL2.MITF (CRE) (CRE mut)] and 20 ng of
pRL/null (Promega, Southampton, UK) using FuGene6
transfection reagent (Roche Molecular Biochemicals,
Basel, Switzerland) as described previously (44). Cells were
allowed to incubate with the transfection mixture for 20 h.
The cells were then incubated with 20 M FSK, 10 nM
EDN1/3, or MEK inhibitor PD98059 for an additional 6 h.
Cells were washed once with PBS and lysed with 20 lof1
passive lysis buffer (Promega). The assay samples were then
analyzed on a 96-well plate luminometer (EG&G Berthold,
Bad Wildbad, Germany) using a Dual-Luciferase kit (Pro-
mega). Luciferase signals were normalized to correspond-
ing Renilla signals. Results are expressed as fold activation
over unstimulated vector control transfection for each
individual promoter set and are plated as the mean se
from at least three independent data points.
RESULTS
EDN signaling elicits phosphorylation of MITF
We first examined posttranslational modulation of
MITF via EDN signaling to assess whether treatment
with EDN1 stimulates the phosphorylation of MITF in
cultured human melanocytes. Western blotting of hu-
man melanocytes using antibody to MITF revealed that
there are two MITF species with relative mobilities
corresponding to molecular masses of 62 and 66 kDa
(Fig. 1A). The identities of the two MITF species with
different mobilities were suggested to be unphosphor-
ylated and phosphorylated MITFs because of their
changes with rapid kinetics and prior analyses by phos-
photryptic mapping (7). Activation of signaling by
EDN1 completely shifted the lower unphosphorylated
MITF band to the upper phosphorylated MITF band
with a peak at 10 min postincubation. This shift oc-
curred rapidly but transiently and the lower unphos-
phorylated band reappeared within 40 min. The mo-
bility shift was abrogated by BQ788, a selective EDNRB
Figure 1. A) EDN1 elicits a marked phosphorylation of MITF. Human primary melanocytes were stimulated with EDN1 (10 nM).
Lysates were harvested at the indicated times. EDN stimulation produced a mobility shift of MITF. Total protein blots were
probed for MITF and ERK1/2. Western blotting shows representative data. Densitometric data represent sem from three
independent experiments using melanocytes from the same donors. B) The EDNRB antagonist BQ788 abolishes the
EDN1/3-induced phosphorylation of ERK. Human primary melanocytes were treated with 10 nM EDN1/3 in the presence or
absence of BQ788 (10 M). Total protein blots were probed for MITF. C) MG132 prevents MITF degradation mediated through
proteasomal degradation. Human primary neonatal melanocytes were stimulated with EDN1 (10 nM) and cycloheximide
(CHX) (20 g/ml) in the presence or absence of MG132 protease inhibitor (25 ␮⌴). Lysates were harvested at the indicated
times and immunoblotted with MITF and -tubulin antibodies. D) S73 of MITF is phosphorylated by EDN3 stimulation. Human
501 MEL melanoma cells were stimulated with EDN3 (10 nM). Lysates were immunoprecipitated (IP) with anti-phospho-S73
(pS73) MITF antibodies or anti-MITF antibody (D5) followed by Western blotting for MITF (C5).
1158 Vol. 22 April 2008 SATO-JIN ET AL.The FASEB Journal
receptor antagonist (Fig. 1B). Proteasome inhibition
using MG132 (Fig. 1C) stabilized MITF in the upper
phosphorylated form induced by EDN1 treatment,
suggesting prevention of MITF degradation mediated
through proteasomal degradation. Immunoprecipita-
tion with anti-phospho-S73 MITF antibody showed that
serine 73 of MITF is phosphorylated by EDN stimula-
tion (Fig. 1D).
EDN signaling elicits the phosphorylation of ERK1/2
and MEK
EDN1 stimulation also increased the phosphorylation
of ERK1/2 with a peak at 10 min postincubation (Fig.
2A), which correlated temporally with the shift in MITF
protein. The increased phosphorylation occurred rap-
idly but transiently from 2 to 20 min after incubation
with EDN1; the phosphorylation of ERK1/2 was mark-
edly diminished within 40 min postincubation. Further,
the increased phosphorylation of ERK1/2 was accom-
panied by increased phosphorylation of MEK during a
comparable time; the increased phosphorylation oc-
curred rapidly but transiently from 2 to 20 min postin-
cubation and almost disappeared within 40 min (Fig.
2B).
Phosphorylation of MITF is specifically abolished by
inhibitors of MEK and PKC
To elucidate the signaling mechanisms involved in
phosphorylation of MITF, we compared time courses of
phosphorylation of MITF and ERK1/2 and used several
specific signaling inhibitors at 10 min postincubation
when the phosphorylation of MITF reaches a maxi-
mum. Western blot analysis revealed that phosphoryla-
tion of ERK1/2 occurred at a time course similar to the
mobility shift of MITF with a peak at 10 –20 min
postincubation with EDN3 in human melanocytes and
human melanoma cells (501 MEL) (Fig. 3). Treatment
with the MEK inhibitor PD98059 (20 M),60min
before addition of EDN1, diminished the increased
phosphorylation of MITF in the presence or absence of
cycloheximide, a translation inhibitor (which blocks de
novo protein synthesis) (Fig. 4A, B). Three independent
experiments showed that MEK inhibition elicits a sig-
nificant decrease in EDN-induced MITF phosphoryla-
tion (Fig. 5A). The MEK inhibitor PD98059 also signif-
icantly decreased the EDN-induced phosphorylation of
Figure 2. A) EDN1 elicits a marked phosphorylation of ERK1/2 followed by the phosphorylation of MITF. Human primary
melanocytes were stimulated with EDN1 (10 nM). Lysates were harvested at indicated times. Total protein blots were probed for
phospho-ERK1/2 (pERK1/2) and ERK1/2. Western blotting shows representative data. Densitometric data represent sem from
three independent experiments using melanocytes from the same donors. B) EDN1 elicits a marked phosphorylation of MEK
followed by phosphorylation of MITF or ERK1/2. Human primary melanocytes were stimulated with EDN1 (10 nM). Lysates
were harvested at indicated times. Total protein blots were probed for phospho-MEK (pMEK) and MEK. Western blotting shows
representative data. Densitometric data represent sem from three independent experiments using melanocytes from the same
donors.
Figure 3. EDN3 stimulation produced a mobility shift of MITF
accompanied by ERK phosphorylation. Human primary me-
lanocytes and human 501 MEL melanoma cells were stimu-
lated with EDN3 (10 nM) for the indicated times, and total
protein blots were probed for MITF (top), phospho-ERK1/2
(pERK1/2) and ERK1/2 (center), and -tubulin (bottom).
1159MOLECULAR INTERACTIONS BETWEEN MITF AND ENDOTHELIN
ERK1/2 (Fig. 5B). Although MEK inhibition was in-
complete, it also suppressed a corresponding degree of
MITF phosphorylation.
Similarly, treatment with the PKC inhibitor Go¨6983
(1 M), 60 min before addition of EDN1, significantly
abolished the increased phosphorylation of MITF (Fig.
6A), which was accompanied by marked inhibition in the
phosphorylation of ERK1/2 (Fig. 6B), consistent with the
possibility that suppression of MITF phosphorylation by
the PKC inhibitor was mediated via the inhibition of
ERK1/2 phosphorylation. In contrast, inhibitors of other
kinases such as PKA (H89, 5 M), p38 MAPK (SB203580,
10 M), PI3 kinase (wortmannin, 100 nM), and Akt
(SH-6, 1 M) did not significantly inhibit the phosphor-
ylation of MITF (Fig. 7A–D).
EDN signaling stimulates the expression of MITF
Because evidence suggests that MITF expression is
regulated by a signal transduction pathway using cAMP
as a second messenger (45, 46) and because we have
already shown that EDN signaling exerts a rapid and
marked increase in the level of cAMP in human mela-
nocytes (19), we determined whether EDN1 alters
MITF expression using quantitative RT-PCR analysis
and Western blotting. Within 40 min after EDN1 stim-
ulation there was an increase in the intensity of MITF-M
mRNA transcripts, relative to control GAPDH levels
(Fig. 8A). This increase occurred transiently with a
peak at 40 to 80 min postincubation and returned to
the control level within 3 h. Western blotting analysis
during 24 h after incubation with EDN1 revealed that
within2hoftreatment there was an increase in the
intensity of MITF protein (upper lower bands),
relative to control -actin levels (Fig. 8B). This increase
occurred transiently with a peak at 2 to 3 h postincu-
bation and returned to the control level within 24 h.
The increased production of MITF protein was signifi-
cantly diminished by the PKA inhibitor H89 (Fig. 8C),
suggesting that the stimulation was dependent on the
cAMP-PKA pathway.
Figure 4. PD98059 abolishes EDN1-induced phosphorylation
of MITF. Human primary melanocytes were stimulated with
EDN1 (10 nM) and without (A) or with (B) cycloheximide
(CHX) (20 g/ml) in the presence or absence of MEK
inhibitor PD98059 (20 M). Lysates were harvested at indi-
cated times and immunoblotted with MITF and -tubulin or
-actin antibodies.
Figure 5. PD98059 abolishes the EDN1-induced phosphorylation of MITF and ERK1/2. Human primary melanocytes were
stimulated with or without EDN1 (10 nM) in the presence or absence of PD98059 (20 M). Lysates were harvested at 10 min
and immunoblotted with MITF and ERK1/2 (A) or with phospho-ERK1/2 (pERK1/2) and ERK1/2 (B). Western blotting shows
representative data. Densitometric data represent mean sd from three independent experiments using melanocytes from the
same donors. *P 0.05.
1160 Vol. 22 April 2008 SATO-JIN ET AL.The FASEB Journal
EDN signaling activates CREB factors
To examine whether the transcription of the MITF
gene is dependent on binding of CREB to the MITF
promoter, luciferase reporter assays were performed
using plasmids containing the melanocyte-restricted
MITF promoter (46) driving luciferase in 501 MEL
melanoma cells (47). These assays showed that EDN (as
well as SCF) significantly induces MITF promoter activ-
ity (Fig. 9 and data not shown). The melanocytic MITF
promoter has a CRE consensus sequence where CREB/
ATF family members bind and transactivate (48). It has
been shown that whereas cAMP signaling stimulates the
MITF promoter via this conserved CRE (45, 46), ERK/
MAPK activation in melanocytes stimulates RSK, which
is known to phosphorylate and activate CREB factors in
other lineages (49, 50). We therefore examined the
possibility that activation of the MITF promoter is
mediated by the CRE site. As shown in Fig. 9 (black
bars), EDN-induced MITF promoter activation was
abrogated when the CRE site was mutated. Further-
more, the EDN responsiveness was also lost with MEK
inhibitor PD98059 treatment before EDN stimulation.
These data are consistent with the possibility that EDN
signaling induces MITF gene transcription using CREB
phosphorylation/activation significantly via the MAPK-
p90RSK pathway. Indeed, as seen in Fig. 10A, Western
blot analysis of EDN1/3 or SCF-treated melanocytes
using anti-phospho-ERK and anti-phospho-CREB
showed that both EDN and SCF stimulations induce
Figure 6. Go¨6983 diminishes the EDN1-induced phosphorylation of MITF and ERK1/2. Human primary neonatal melanocytes
were stimulated with or without EDN1 (10 nM) in the presence or absence of PKC inhibitor Go¨6983 (1 M). Lysates were
harvested at 10 min and immunoblotted with MITF and ERK1/2 (A) or with phospho-ERK1/2 (pERK1/2) and ERK1/2 (B).
Western blotting shows representative data. Densitometric data represent mean sd from three independent experiments
using melanocytes from the same donors. *P 0.05.
Figure 7. Other inhibitors do not elicit the EDN1 induced phosphorylation of
MITF or ERK1/2. Human primary melanocytes were stimulated with or without
EDN1 (10 nM) in the presence or absence of PKA inhibitor H89 (5 M) (A),
p38 MAPK inhibitor SB203580 (10 M) (B), PI3 kinase inhibitor wortmannin
(100 nM) (C) or Akt inhibitor SH-6 (1 M) (D). Lysates were harvested at 10
min and immunoblotted with MITF, phospho-ERK1/2 (pERK1/2), and
ERK1/2. Similar results were seen in three independent experiments using
melanocytes from the same donors.
1161MOLECULAR INTERACTIONS BETWEEN MITF AND ENDOTHELIN
significant MAPK and CREB phosphorylation, whereas
FSK treatment did not induce comparable ERK/MAPK
phosphorylation, although it did trigger strong CREB
phosphorylation that was not abrogated by MEK inhib-
itor (Fig. 10B).
The time course study of the phosphorylation of
CREB revealed that CREB was markedly phosphory-
lated by EDN stimulation with a broad peak from 3
through 30 min postincubation (Fig. 11A). The CREB
phosphorylation induced by EDN was abrogated to a
different extent depending on postincubation times
by the PKA inhibitor (H89) or the MEK inhibitor
(PD98059) (Fig. 11B–E). Although CREB phosphor-
ylation was significantly abolished at 5 min postincu-
bation by the MEK inhibitor but not by the PKA
inhibitor (Fig. 11B, C), the induced phosphoryla-
tions at 15 and 30 min postincubation were signifi-
cantly abrogated by the PKA inhibitor but not by the
MEK inhibitor (Fig. 11B, D, E), suggesting a biphasic
activation of CREB due to the differential time
course of signaling in the MAPK-RSK and cAMP-PKA
pathways. These findings, taken together, suggest
that EDN signaling up-regulates MITF expression
significantly through both MAPK-RSK and cAMP-
Figure 8. A) EDN1 increases transcription of MITF-M. Human
primary melanocytes were treated with 10 nM EDN1 and were
harvested and solubilized after the incubation times. Total
RNA extracts from treated or control human melanocytes
were reverse-transcribed and the cDNAs were PCR amplified
with specific primer sets. **P 0.01; *P 0.05. B) EDN1
stimulates production of MITF protein. Human primary
melanocytes were stimulated with EDN1 (10 nM). Lysates
were harvested at the indicated times. Total protein blots
were probed for MITF and -actin. Western blotting shows
representative data. Densitometric data represent sem from
three independent experiments using melanocytes from the
same donors. C) Human primary melanocytes were stimu-
lated with or without EDN1 (10 nM) in the presence or
absence of H89 (5 M). Lysates were harvested at3hand
immunoblotted with MITF and -actin. Western blotting
shows representative data. Densitometric data represent
mean sd from three independent experiments using
melanocytes from the same donors. *P 0.05.
Figure 9. EDN1/3 Significantly induces MITF promoter ac-
tivity. 501 MEL melanoma cells were transfected with the
human MITF promoter (387 to 97) or CREB binding site
(CRE) mutant promoter driving luciferase. Transfected cells
were treated with FSK or EDN1/3. Further, in one case,
EDN3 was added after PD98059 (20 M) treatment. Lucif-
erase activity was corrected for transfection efficiency using
constitutive Renilla (sea pansy) luciferase activity. Luciferase
activities were normalized with no promoter control
(pGL2.basic). The value of no stimulation control was nor-
malized to 1. Data represent sem from three independent
experiments using melanocytes from the same donors.
1162 Vol. 22 April 2008 SATO-JIN ET AL.The FASEB Journal
PKA pathways, which converge on CREB and exhibit
differential (complementary) kinetics.
EDN induces MITF-dependent transactivation of the
cell cycle regulator CDK2 and the melanosomal
protein SILV
The above results place MITF downstream of EDN
signals and suggest that significant aspects of EDN-
induced melanocyte proliferation or differentiation
may be mediated by MITF. We have recently found that
SILV and CDK2 are both MITF target genes (41, 51).
Quantitative RT-PCR revealed that EDN stimulation
significantly increased the expression levels of SILV and
CDK2 in primary melanocytes (Fig. 12). We therefore
asked whether up-regulation of these targets by EDN
was mediated by MITF. To test this hypothesis, EDN3
stimulation was repeated in the presence of control or
dominant-negative MITF adenoviruses (52) in primary
melanocytes (Fig. 12). The increased expression levels
of SILV and CDK2 induced by EDN administration were
both selectively blocked by dominant-negative MITF.
The dominant-negative MITF virus itself did not affect
MAPK activation (data not shown). These results dem-
onstrate that EDN stimulates expression of SILV and
CDK2, two genes that are involved in melanocyte dif-
ferentiation and proliferation, respectively, and this
stimulated expression is dependent on endogenous
MITF.
Expression of EDN receptor EDNRB is dependent
on MITF
Analysis of MITF expression in human melanocytes
stimulated with EDN1/3 suggested that the EDN-MITF
signaling pathways are regulated by multiple feedback
pathways. Our previous microarray analysis suggested
that expression of EDNRB is regulated via MITF in
human melanocytes (38). Thus, we examined EDNRB
expression by human melanocytes infected with adeno-
virus expressing the wild-type and dominant-negative
form of MITF. As shown in Fig. 13, EDNRB mRNA
expression was significantly induced with wild-type
MITF and inhibited by dominant-negative MITF, indi-
cating that EDNRB expression is dependent on MITF.
DISCUSSION
Despite increasing knowledge of the functions of MITF
in melanoblasts/melanocytes during their develop-
ment and of the roles of EDNs in several pigmentary
disorders (10, 33–37), the effects of EDN-activated
intracellular signaling on MITF dynamics, including its
phosphorylation, have received little attention. Thus, it
remains to be clarified as to how the activation of the
PKC, MAPK, and cAMP pathways initiated by EDN
receptor binding (19 –21) are linked to survival as well
as to the stimulation of mitogenesis and melanogenesis
of melanocytes in association with the dynamics of
MITF. Although MAPK is known to link MITF to KIT
signaling in melanocytes via MITF phosphorylation (7,
53), it had not been established whether stimulation of
human melanocytes with EDN affects the phosphoryla-
tion of MITF due to the activation of the MAPK
pathway. Here we demonstrate that EDN1 signaling
elicits a marked phosphorylation of MITF with a peak at
10 min postincubation with EDN1. This phosphoryla-
tion of MITF occurs at Ser
73
in an EDNRB-sensitive
manner and is accompanied by or preceded by the
phosphorylation of ERK1/2 and MEK, which indicates
activation of the MAPK pathway. Treatment with a MEK
inhibitor markedly suppresses the phosphorylation of
MITF. The inhibition of MITF phosphorylation is ac-
companied by a significant suppression of the phos-
phorylation of ERK1/2 at a comparable time. Because
ERK2 is the kinase responsible for this MITF phosphor-
ylation (7) and because the increased phosphorylation
of ERK1 and 2 occurs with response kinetics similar to
those of EDN1, these findings strongly suggest that
activation of the MAPK pathway by EDN1 directly
Figure 10. A) EDN1/3 and SCF elicit a marked phosphoryla-
tion of CREB and ERK1/2. Human primary melanocytes were
stimulated with EDN1 (10 nM), EDN3 (10 nM), and SCF (20
ng/ml). Total protein blots were probed for MITF, phospho-
CREB (pCREB), phospho-ERK1/2 (pERK1/2), and -tubu-
lin. B) FSK induces a marked CREB phosphorylation that is
not abrogated by PD98059. Human primary melanocytes
were stimulated with FSK (20 M) in the presence or absence
of 20 M PD98059 for 15 min. Total protein blots were
probed for MITF, phospho-CREB, phospho-ERK1/2, and
-tubulin.
1163MOLECULAR INTERACTIONS BETWEEN MITF AND ENDOTHELIN
triggers MITF phosphorylation in human melanocytes
similar to the stimulation by SCF.
In the signaling pathway initiated by EDN1 activation
of the EDNRB in human melanocytes, we found that
the activation of PKC is linked probably at a convergent
point of Raf-1 to the phosphorylation of ERK1/2, which
indicates activation of the MAPK pathway (21). In
addition, we demonstrated that EDN1 signaling elicits a
marked increase in intracellular cAMP levels in a
PKC-dependent manner, indicating cross-talk between
PKC and the cAMP pathway (19). Based on the above
observations regarding EDN signaling, we used several
specific signaling inhibitors in this study to show that
MITF phosphorylation is significantly abolished by a
Figure 11. A) EDN1 elicits a
marked phosphorylation of CREB
with a peak at 3–15 min. Human
primary melanocytes were stimu-
lated with EDN1 (10 nM). Lysates
were harvested at indicated times.
EDN stimulation produced phos-
phorylation of CREB. Total pro-
tein blots were probed for phos-
pho-CREB (p-CREB), phospho-
ERK1/2 (pERK1/2), ERK1/2, and
-actin. Western blotting shows
representative data. Densitometric
data represent sem from three in-
dependent experiments using melanocytes from the same donors. B) PD98059 or H89 diminishes the EDN1-induced
phosphorylation of CREB. Human primary neonatal melanocytes were stimulated with EDN1 (10 nM) in the presence or
absence of PD98059 (40 M) and H89 (10 M). Lysates were harvested at indicated times and immunoblotted with
phospho-CREB, CREB, phospho-ERK1/2, and -actin. C–E) PD98059 and H89 diminishes the EDN1-induced phosphorylation
of CREB with a different time course. Human primary neonatal melanocytes were stimulated with or without EDN1 (10 nM)
in the presence or absence of PD98059 (40 M) and H89 (10 M). Lysates were harvested at 5 min (C), 15 min (D), or 30 min
(E) and immunoblotted with phospho-CREB, CREB, and -actin. Western blotting shows representative data. Densitometric
data represent mean sd from three independent experiments using melanocytes from the same donors. **P 0.01; *P
0.05; n.s., not significant.
Figure 12. EDN induces MITF-dependent trans-
activation of cell cycle regulator CDK2 and
melanosomal protein SILV. CDK2 (A) and
SILV (B) mRNA expressions were examined in
human primary melanocytes infected with ade-
novirus (ad.) expressing the dominant-negative
(dn) form of MITF and stimulated with EDN3.
Data represent sem from three experiments
using melanocytes from the same donors.
1164 Vol. 22 April 2008 SATO-JIN ET AL.The FASEB Journal
specific PKC inhibitor, but not by p38 MAPK, PI3
kinase, Akt, or PKA inhibitors. The fact that the inhi-
bition of MITF phosphorylation by the PKC inhibitor is
accompanied by a significant suppression of ERK1/2
phosphorylation suggests that the MITF phosphoryla-
tion induced by EDN signaling is not directly triggered
by the activation of PKC but is evoked by the activation
of ERK1/2, which occurs subsequently as a result of the
activation of PKC.
In contrast to MITF Ser
73
phosphorylation, which is
associated with the signal transduction pathway involv-
ing MAPK, MITF expression is probably regulated by a
signal transduction pathway using cAMP as a second
messenger as the MITF promoter contains a CRE (48).
In fact, elevated levels of intracellular cAMP, triggered
either by -melanocyte-stimulating hormone (-MSH)
or by FSK, lead to rapid and potent induction of the
MITF promoter (45, 46). Because EDN1 signaling
results in a marked increase in cAMP levels in human
melanocytes in a PKC-dependent manner (19), we
asked whether EDN1 stimulation alters MITF expres-
sion via CREB phosphorylation. In this connection, our
study demonstrates that EDN1 treatment elicits a rapid
increase in MITF-M mRNA transcripts with a peak at
4080 min postincubation. This is followed by a rapid
and transient increase in MITF protein with a peak at
2–3 h after EDN1 stimulation. As for signaling mecha-
nisms underlying stimulated MITF expression, we
found that 1) the increased production of MITF pro-
tein was significantly diminished by the PKA inhibitor
H89, 2) EDN-induced MITF promoter activation was
abrogated when the CRE site was mutated, and 3) EDN
stimulation induces a significant CREB phosphoryla-
tion in concert with MAPK phosphorylation. These
findings strongly suggest that the increased expression
of MITF after EDN stimulation is mediated via CREB
activation. As for the signaling pathway leading to
CREB activation after EDN stimulation, we demon-
strated that both the potent induction of the MITF
promoter and CREB phosphorylation by EDN stimula-
tion are abrogated by the MEK inhibitor (PD98059).
Because CREB phosphorylation after FSK stimulation
was not significantly affected by the MEK inhibitor, our
results suggest the possibility that the MAPK-RSK path-
way is also implicated in EDN-induced MITF gene
expression through CREB phosphorylation/activation.
This finding is consistent with our results (see Fig. 10A)
that SCF stimulation induces CREB phosphorylation
concomitant with ERK phosphorylation despite the fact
that SCF signaling does not up-regulate intracellular
cAMP levels (19). It is known that CREB is activated by
cAMP-dependent protein kinase as well as by all three
members of the RSK family (RSK1–3) in cells stimu-
lated by activators of the Ras-MEK-ERK1/2 cascade
(49). Therefore, as depicted in Fig. 14, it is likely that
EDN signaling triggers MITF expression via CREB
phosphorylation/activation due to both cAMP-PKA
and MAPK-p90RSK cascades, which occur with differ-
ent (biphasic) kinetics, PKA being much later than
tMAPK.
-MSH, KIT, and EDN signaling pathways are impli-
cated in the modulation and the expression of MITF,
but they do so in very different ways. -MSH stimulation
of melanocytes up-regulates cAMP, which increases the
transcription of MITF through a CRE in the MITF
promoter (45, 46). In contrast, KIT stimulation elicits a
very rapid MAPK-mediated phosphorylation of MITF,
which induces the enhanced recruitment of p300/
CREB-binding protein (54), the coactivator family that
interacts with and modulates the transcriptional activity
of MITF. All of that occurs over the course of minutes.
On the other hand, EDN stimulation not only elicits
very rapid MAPK-mediated phosphorylation of MITF
within minutes but also profoundly increases MITF
protein expression within hours. These dual actions
Figure 13. EDNRB expression is dependent on MITF func-
tion. EDNRB mRNA expression were examined in human
primary melanocytes infected with adenovirus expressing the
wild-type (WT) or dominant-negative (DN) form of MITF.
Data represent sem from three experiments using melano-
cytes from the same donors.
Figure 14. Schematic diagram of the EDN-MITF signaling
pathway. EDN signals profoundly regulate the central mela-
nocyte transcription factor MITF in two ways: 1) up-regula-
tion of the MITF gene and 2) direct MITF phosphorylation
through MAPK. This pathway forms a feedback loop with
MITF-dependent up-regulation of EDNRB gene expression.
1165MOLECULAR INTERACTIONS BETWEEN MITF AND ENDOTHELIN
triggered by EDN stimulation are likely to be associated
with the dual potent effects of EDN on both the
proliferation and the differentiation of human melano-
cytes (15).
It is well known that KIT-mediated MAPK phosphor-
ylation triggers a short-lived MITF activation as well as a
net degradation of MITF mediated by proteasomes (7,
53). Our data also showed that a rapid disappearance of
phosphorylated MITF during EDN stimulation occurs
via its rapid degradation through proteasomes. EDN1
signaling elicits the increased production of MITF
protein and sustains MITF protein at more than control
levels over 24 h after the addition of EDN1. The
increased production of MITF protein is significantly
diminished by the PKA inhibitor H89, suggesting that
the stimulation occurs at least via the cAMP pathway.
Thus, it is likely that the cAMP-mediated new produc-
tion of MITF protein may compensate to some extent
for MITF degradation, which is stimulated by MAPK-
mediated MITF phosphorylation and subsequent ubiq-
uitination.
In this study, we demonstrate a molecular link be-
tween MITF and EDN/EDNRB, mutations, which pro-
duce WS type II and IV, respectively. Our findings show
that EDN signaling modulates the MITF by up-regula-
tion of MITF gene expression and MITF phosphoryla-
tion, which is associated with altered half-life. These
effects are both mediated by the ERK/MAPK pathway
but follow distinct kinetics. Furthermore, our transfec-
tion studies using dominant-negative MITF demon-
strate that expression of EDNRB is dependent on MITF
function, suggesting that EDNRB is a downstream
target of MITF. These findings connect several central
regulators of melanocyte development/proliferation/
differentiation into a signaling pathway that is dis-
rupted in WS types II and IV. The deafness and white
forelock (spotting) seen in patients with WS types I–IV
and Woolf syndrome thus probably results from MITF
dosage insufficiency due to disruption of 1) signaling
pathways that modulate MITF expression (EDN3,
EDNRB, and KIT), 2) transcription factors that regu-
late MITF expression (PAX3 and SOX10), or 3) muta-
tions within MITF itself. The finding of WS type II MITF
null mutant alleles (34) is consistent with the conclu-
sion that MITF dosage (haploinsufficiency) is the crit-
ical determinant of this autosomal dominant (heterozy-
gous) clinical syndrome. In addition, the discovery of
signaling pathways that modulate MITF promoter activ-
ity might theoretically permit therapeutic up-regulation
of the remaining wild-type MITF allele in these patients.
This possibility may also exist through the MSH path-
way, which signals via cAMP to CREB and similarly
stimulates MITF expression in melanocytes (45, 46).
As for signaling mechanisms underlying EDN-in-
duced proliferation and differentiation (melanin syn-
thesis) in association with MITF function, the present
study demonstrated that EDN induces MITF-depen-
dent transactivation of the CDK2 and SILV genes. CDK2
is a major cell cycle regulator important for S phase
progression (54). Therefore, a direct link of MITF with
CDK2 gene transactivation during EDN signaling may
contribute to the EDN-induced stimulation of melano-
cyte proliferation (21). SILV (PMEL17) is a melanoso-
mal protein (55) whose mutation in mice (si/si) results
in reduced melanosome number and the silver pig-
ment color defect (56). Details of the transcriptional
regulation of these genes by MITF have been described
previously (41, 51). The induction of SILV gene trans-
activation during EDN signaling is also consistent with
EDN-induced stimulation of melanin synthesis (includ-
ing melanosome formation) in human melanocytes
(21) as MITF can act as a transcription factor for
multiple components in the pigmentation pathway
(29–32).
In conclusion, as depicted in Fig. 14, EDN signals
profoundly regulate the central melanocyte transcrip-
tion factor MITF in two ways: 1) direct MITF phosphor-
ylation through MAPK and 2) up-regulation of MITF
gene expression. This pathway is also definitively asso-
ciated with up-regulation of EDNRB gene expression.
Thus, our findings associate EDN/EDNR signaling with
MITF modulation as a central regulator of melanocyte
fate, leading to the formation of a feedback loop with
strong epistatic connections. The dynamics of MITF
phosphorylation and production initiated by the acti-
vation of EDN/EDNR provides a deep insight into the
pathogenesis of WS types II and IV as well as the
biological mechanisms underlying other EDN-related
hyper- or hypopigmentary disorders.
REFERENCES
1. Bondurand, N., Pingault, V., Goerich, D. E., Lemort, N., Sock,
E., Caignec, C. L., Wegner, M., and Goossens, M. (2000)
Interaction among SOX10, PAX3 and MITF, three genes altered
in Waardenburg syndrome. Hum. Mol. Genet. 9, 1907–1917
2. Potterf, S. B., Furumura, M., Dunn, K. J., Arnheiter, H., and
Pavan, W. J. (2000) Transcription factor hierarchy in Waarden-
burg syndrome: regulation of MITF expression by SOX10 and
PAX3. Hum. Genet. 107, 1– 6
3. Watanabe, A., Takeda, K., Ploplis, B., and Tachibana, M. (1998)
Epistatic relationship between Waardenburg syndrome genes
MITF and PAX3. Nat. Genet. 18, 283–286
4. Lee, M., Goodall, J., Verastegui, C., Ballotti, R., and Goding,
C. R. (2000) Direct regulation of the Microphthalmia promoter
by Sox10 links Waardenburg-Shah syndrome (WS4)-associated
hypopigmentation and deafness to WS2. J. Biol. Chem. 275,
37978–37983
5. Verastegui, C., Bille, K., Ortonne, J. P., and Ballotti, R. (2000)
Regulation of the microphthalmia-associated transcription fac-
tor gene by the Waardenburg syndrome type 4 gene, SOX10.
J. Biol. Chem. 275, 30757–30760
6. Halaban, R., Hebert, D. N., and Fisher, D. E. (2003) Biology of
melanocytes: biology and development of skin. In Fitzpatrick’s
Dermatology in General Medicine, 6th ed. (Freedberg, I. M., Eisen,
A. Z., Wolff, K., Austen, K. F., Goldsmith L. A., and Katz, S. I.
eds) pp. 127–148, McGraw-Hill, New York
7. Hemesath, T. J., Price, E. R., Takemoto, C., Badalian, T., and
Fisher, D. E. (1998) MAP kinase links the transcription factor
Microphthalmia to c-Kit signalling in melanocytes. Nature 391,
298–301
8. Baynash, A. G., Hosoda, K., Giaid, A., Richardson, J. A., Emoto,
N., Hammer, R. E., and Yanagisawa, M. (1994) Interaction of
endothelin-3 with endothelin-B receptor is essential for devel-
1166 Vol. 22 April 2008 SATO-JIN ET AL.The FASEB Journal
opment of epidermal melanocytes and enteric neurons. Cell 79,
1277–1285
9. Hosoda, K., Hammer, R. E., Richardson, J. A., Baynash, A. G.,
Cheung, J. C., Giaid, A., and Yanagisawa, M. (1994) Targeted
and natural (piebald-lethal) mutations of endothelin-B receptor
gene produce megacolon associated with spotted coat color in
mice. Cell 79, 1267–1276
10. Puffenberger, E. G., Hosoda, K., Washington, S. S., Nakao, K.,
deWit, D., Yanagisawa, M., and Chakravart, A. (1994) A missense
mutation of the endothelin-B receptor gene in multigenic
Hirschsprung’s disease. Cell 79, 1257–1266
11. Shin, M. K., Levorse, J. M., Ingram, R. S., and Tilghman, S. M.
(1999) The temporal requirement for endothelin receptor-B
signalling during neural crest development. Nature 402, 496
501
12. Pavan, W. J., and Tilghman, S. M. (1994) Piebald lethal (s
l
) acts
early to disrupt the development of neural crest-derived mela-
nocytes. Proc. Natl. Acad. Sci. U. S. A. 91, 7159–7163
13. Yoshida, H., Kunisada, T., Kusakabe, M., Nishikawa, S., and
Nishikawa, S. I. (1996) Distinct stages of melanocyte differenti-
ation revealed by analysis of nonuniform pigmentation patterns.
Development 122, 1207–1214
14. Yanagisawa, M., Kurihara, H., Kimura, S., Tomobe, Y., Koba-
yashi, M., Mitsui, Y., Yazaki, Y., Goto, K., and Masaki, T. (1988)
A novel potent vasoconstrictor peptide produced by vascular
endothelial cells. Nature 332, 411– 415
15. Yada, Y., Higuchi, K., and Imokawa, G. (1991) Effects of
endothelins on signal transduction and proliferation in human
melanocytes. J. Biol. Chem. 266, 18352–18357
16. Imokawa, G., Yada, Y., and Miyagishi, M. (1992) Endothelins
secreted from human keratinocytes are intrinsic mitogens for
human melanocytes. J. Biol. Chem. 267, 24675–24680
17. Imokawa, G., Miyagishi, M., and Yada, Y. (1995) Endothelin-1 as
a new melanogen: coordinated expression of its gene and the
tyrosinase gene in UVB-exposed human epidermis. J. Invest.
Dermatol. 105, 32–37
18. Reid, K., Turnley, A. M., Maxwell, G. D., Kurihara, Y., Kurihara,
H., Bartlett, P. F., and Murphy, M. (1996) Multiple roles for
endothelin in melanocyte development: regulation of progeni-
tor number and stimulation of differentiation. Development 122,
3911–3919
19. Imokawa, G., Yada, Y., Morisaki, N., and Kimura, M. (1996)
Signalling mechanisms of endothelin-induced mitogenesis and
melanogenesis in human melanocytes. Biochem. J. 330, 1235–1239
20. Imokawa, G., Kobayashi, T., Miyagishi, M., Higashi, K., and
Yada, Y. (1997) The role of endothelin-1 in epidermal hyper-
pigmentation and signaling mechanisms of mitogenesis and
melanogenesis. Pigment Cell. Res. 10, 218–228
21. Imokawa, G., Kobayasi, T., and Miyagishi, M. (2000) Intracellu-
lar signaling mechanisms leading to synergistic effects of endo-
thelin-1 and stem cell factor on proliferation of cultured human
melanocytes. J. Biol. Chem. 275, 33321–33328
22. Bohm, M., Moellmann, G., Cheng, E., Alvarez-Franco, M.,
Wagner, S., Sassone-Corsi, P., and Halaban, R. (1995) Identifi-
cation of p90RSK as the probable CREB-Ser133 kinase in
human melanocytes. Cell Growth Differ. 6, 291–302
23. Halaban, R., Langdon, R., Birchall, N., Cuono, C., Baird, A.,
Scott, G., Moellmann, G., and McGuire, J. (1988) Basic fibro-
blast growth factor from human keratinocytes is a natural
mitogen for melanocytes. J. Cell Biol. 107, 1611–1619
24. Matsumoto, K., Tajima, H., and Nakamura, T. (1991) Hepato-
cyte growth factor is a potent stimulator of human melanocyte
DNA synthesis and growth. Biochem. Biophys. Res. Commun. 176,
45–51
25. Yaar, M., and Gilchrest, B. A. (1991) Human melanocyte growth
and differentiation: a decade of new data. J. Invest. Dermatol. 97,
611–617
26. Hodgkinson, C. A., Moore, K. J., Nakayama, A., Steingrimsson,
E., Copeland, N. G., Jenkins, N. A., and Arnheiter, H. (1993)
Mutations at the mouse microphthalmia locus are associated
with defects in a gene encoding a novel basic-helix-loop-helix-
zipper protein. Cell 74, 395– 404
27. Hemesath, T. J., Steingrimsson, E., McGill, G., Hansen, M. J.,
Vaught, J., Hodgkinson, C. A., Arnheiter, H., Copeland, N. G.,
Jenkins, N. A., and Fisher, D. E. (1994) Microphthalmia, a
critical factor in melanocyte development, defines a discrete
transcription factor family. Genes Dev. 8, 2770 –2780
28. Steingrimsson, E., Moore, K. J., Lamoreux, M. L., Ferre-
D’Amare, A. R., Burley, S. K., Zimring, D. C., Skow, L. C.,
Hodgkinson, C. A., Arnheiter, H., Copeland, N. G., and Jenkins,
N. A. (1994) Molecular basis of mouse microphthalmia (mi)
mutations helps explain their developmental and phenotypic
consequences. Nat. Genet. 8, 256 –263
29. Bentley, N. J., Eisen, T., and Goding, C. R. (1994) Melanocyte-
specific expression of the human tyrosinase promoter: activa-
tion by the microphthalmia gene product and role of the
initiator. Mol. Cell. Biol. 14, 79968006
30. Bertolotto, C., Busca, R., Abbe, P., Bille, K., Aberdam, E.,
Ortonne, J. P., and Ballotti, R. (1998) Different cis-acting
elements are involved in the regulation of TRP1 and TRP2
promoter activities by cyclic AMP: pivotal role of M boxes
(GTCATGTGCT) and of microphthalmia. Mol. Cell. Biol. 18,
694–702
31. Yasumoto, K., Mahalingam, H., Suzuki, H., Yoshizawa, M., and
Yokoyama, K. (1995) Transcriptional activation of the melano-
cyte-specific genes by the human homolog of the mouse Mi-
crophthalmia protein. J. Biochem. (Tokyo) 118, 874 881
32. Yavuzer, U., Keenan, E., Lowings, P., Vachtenheim, J., Currie,
G., and Goding, C. R. (1995) The Microphthalmia gene product
interacts with the retinoblastoma protein in vitro and is a target
for deregulation of melanocyte-specific transcription. Oncogene
10, 123–134
33. Hughes, A. E., Newton, V. E., Liu, X. Z., and Read, A. P. (1994)
Waardenburg syndrome type 2 maps close to the human
homologue of the microphthalmia gene at chromosome 3p12–
p14.1. Nat. Genet. 7, 509 –512
34. Tachibana, M., Takeda, K., Nobukuni, Y., Urabe, K., Long, J. E.,
Meyers, K. A., Aaronson, S. A., and Miki, T. (1996) Waarden-
burg syndrome type 2 maps close to the human homologue of
the microphthalmia gene at chromosome 3p12–p14.1. Nat.
Genet. 14, 50 –54
35. Tassabehji, M., Newton, V. E., and Read, A. P. (1994) Waarden-
burg syndrome type 2 caused by mutations in the human
microphthalmia (MITF) gene. Nat. Genet. 8, 251–255
36. Edery, P., Attie´, T., Amiel, J., Pelet, A., Eng, C., Hofstra, R.,
Martelli, H., Bidaud, C., Munnich, A., and Lyonnet, S. (1996)
Mutation of the endothelin-3 gene in the Waardenburg-Hirsch-
sprung phenotype (Shah-Waardenburg syndrome). Nat. Genet.
12, 442– 444
37. Hofstra, R. M., Osinga, J., Tan-Sindhunata, G., Wu, Y., Kam-
steeg, E. J., Stulp, R. P., van Ravenswaaij-Arts, C., Majoor-
Krakauer, D., Angrist, M., Chakravarti, A., Meijers, C., and Buys,
C. H. (1996) A homozygous mutation in the endothelin-3 gene
associated with a combined Waardenburg type 2 and Hirsch-
sprung phenotype (Shah-Waardenburg syndrome). Nat. Genet.
12, 445– 447
38. McGill, G. G., Horstmann, M., Widlund, H. R., Du, J.,
Motyckova, G., Nishimura, E. K., Lin, Y. L., Ramaswamy, S.,
Avery, W., Ding, H. F., Jordan, S. A., Jackson, I. J., Korsmeyer,
S. J., Golub, T. R., and Fisher, D. E. (2002) Bcl2 regulation
by the melanocyte master regulator Mitf modulates lin-
eage survival and melanoma cell viability. Cell 109,
707–718
39. Carreira, S., Goodall, J., Aksan, I., La Rocca, S. A., Galibert,
M. D., Denat, L., Larue, L., and Goding, C. R. (2005) Mitf
cooperates with Rb1 and activates p21
Cip1
expression to regulate
cell cycle progression. Nature 433, 764–769
40. Loercher, A. E., Tank, E. M., Delston, R. B., and Harbour, J. W.
(2005) MITF links differentiation with cell cycle arrest in
melanocytes by transcriptional activation of INK4A. J. Cell Biol.
168, 35– 40
41. Wu, M., Hemesath, T. J., Takemoto, C. M., Horstmann, M. A.,
Wells, A. G., Price, E. R., Fisher, D. Z., and Fisher, D. E. (2000)
c-Kit triggers dual phosphorylations, which couple activation
and degradation of the essential melanocyte factor Mi. Genes
Dev. 14, 301–312
42. Du, J., Widlund, H. R., Horstmann, M. A., Ramaswamy, S., Ross,
K., Huber, W. E., Nishimura, E. K., Golub, T. R., and Fisher,
D. E. (2004) Critical role of CDK2 for melanoma growth linked
to its melanocyte-specific transcriptional regulation by MITF.
Cancer Cell 6, 565–576
43. Weilbaecher, K. N., Motyckova, G., Huber, W. E., Takemoto,
C. M., Hemesath, T. J., Xu, Hershey, Y., C.L., Dowland, N. R.,
Wells, A. G., and Fisher, D. E. (2001) Linkage of M-CSF
1167MOLECULAR INTERACTIONS BETWEEN MITF AND ENDOTHELIN
signaling to Mitf, TFE3, and the osteoclast defect in MITF
mi/mi
mice. Mol. Cell 8, 749 –758
44. Huber, W. E., Price, E. R., Widlund, H. R., Du, J., Davis, I. J.,
Wegner, M., and Fisher, D. E. (2003) A tissue restricted cAMP
transcriptional response: SOX10 modulates -melanocyte-stim-
ulating hormone-triggered expression of microphthalmia-asso-
ciated transcription factor in melanocytes. J. Biol. Chem. 278,
45224 45230
45. Bertolotto, C., Abbe, P., Hemesath, T. J., Bille, K., and Fisher,
D. E. Ortonne, J. P., and Ballotti, R. (1998) Microphthalmia
gene product as a signal transducer in cAMP-induced differen-
tiation of melanocytes. J. Cell Biol. 142, 827–835
46. Price, E. R., Horstmann, M. A., Wells, A. G., Weilbaecher, K. N.,
Takemoto, C. M., Landis, M. W., and Fisher, D. E. (1998)
-Melanocyte-stimulating hormone signaling regulates expres-
sion of microphthalmia, a gene deficient in Waardenburg syn-
drome. J. Biol. Chem. 273, 33042–33047
47. Zakut, R., Perlis, R., Eliyahu, S., Yarden, Y., Givol, D., Lyman,
S. D., and Halaban, R. (1993) KIT ligand (mast cell growth
factor) inhibits the growth of KIT-expressing melanoma cells.
Oncogene 8, 2221–2229
48. Fuse, N., Yasumoto, K., Suzuki, H., Takahashi, K., and Shiba-
hara, S. (1996) Identification of a melanocyte-type promoter of
the microphthalmia-associated transcription factor gene. Bio-
chem. Biophys. Res. Commun. 219, 702–707
49. Xing, J., Ginty, D. D., and Greenberg, M. E. (1996) Coupling of
the RAS-MAPK pathway to gene activation by RSK2, a growth
factor-regulated CREB kinase. Science 273, 959–963
50. De Cesare, D., Jacquot, S., Hanauer, A., and Sassone-Corsi, P.
(1998) Rsk-2 activity is necessary for epidermal growth factor-
induced phosphorylation of CREB protein and transcription of
c-fos gene. Proc. Natl. Acad. Sci. U. S. A. 95, 12202–12207
51. Du, J., Miller, A. J., Widlund, H. R., Horstmann, M. A.,
Ramaswamy, S., and Fisher, D. E. (2003) MLANA/MART1
and SILV/PMEL17/GP100 are transcriptionally regulated by
MITF in melanocytes and melanoma. Am. J. Pathol. 163,
333–343
52. Widlund, H. R., Horstmann, M. A., Price, E. R., Cui, J., Lessnick,
S. L., Wu, M., He, X., and Fisher, D. E. (2002) -Catenin-
induced melanoma growth requires the downstream target
Microphthalmia-associated transcription factor. J. Cell Biol. 158,
1079–1087
53. Price, E. R., Ding, H. F., Badalian, T., Bhattacharya, S., Take-
moto, C., Yao, T. P., Hemesath, T. J., and Fisher, D. E. (1998)
Lineage-specific signaling in melanocytes: c-kit stimulation re-
cruits p300/CBP to microphthalmia. J. Biol. Chem. 273, 17983–
17986
54. Ekholm, S. V., and Reed, S. I. (2000) Regulation of G
1
cyclin-
dependent kinases in the mammalian cell cycle. Curr. Opin. Cell
Biol. 12, 676 684
55. Kobayashi, T., Urabe, K., Orlow, S. J., Higashi, K., Imokawa,
G., Kwon, B. S., Potterf, B., and Hearing, V. J. (1994) The
Pmel 17/silver locus protein: characterization and investiga-
tion of its melanogenic function. J. Biol. Chem. 269, 29198
29205
56. Orlow, S. J., Silvers, W. K., Zhou, B. K., and Mintz, B. (1998)
Comparative decreases in tyrosinase, TRP-1, TRP-2, and Pmel
17/silver antigenic proteins from melanotic to amelanotic
stages of syngeneic mouse cutaneous melanomas and metastases
Cancer Res. 58, 1521–1523
Received for publication August 7, 2007.
Accepted for publication October 18, 2007.
1168 Vol. 22 April 2008 SATO-JIN ET AL.The FASEB Journal
... While one melanogenesis pathway is known to be activated by αMSH, locally-released endothelin-1 (ET-1) activates the melanocyte endothelin B receptor (ET B R) to trigger a distinct pathway in response to UV exposure (Kawakami & Fisher, 2017). ET B R stimulation activates PLC to generate diacylglycerol (DAG) and IP 3 -mediated Ca 2+ store mobilisation, which concurrently stimulate the RAS/MAPK signalling pathway to drive MITF activation and melanogenesis (Imokawa et al., 1996;Sato-Jin et al., 2008) (Figure 1C). Stanisz et al. (2012) showed that Orai1 knockdown blunts in vitro primary melanocyte viability, and inhibition with 2-APB inhibits tyrosinase activity and UVor ET-1-induced melanin production. ...
Article
Full-text available
The skin is a complex organ that acts as a protective layer against the external environment. It protects the internal tissues from harmful agents, dehydration, ultraviolet radiation and physical injury as well as conferring thermoregulatory control, sensation, immunological surveillance and various biochemical functions. The diverse cell types that make up the skin include 1) keratinocytes, which form the bulk of the protective outer layer; 2) melanocytes, which protect the body from ultraviolet radiation by secreting the pigment melanin; and 3) cells that form the secretory appendages: eccrine and apocrine sweat glands, and the sebaceous gland. Emerging evidence suggests that store-operated Ca2+ entry (SOCE), whereby depletion of intracellular Ca2+ stores triggers Ca2+ influx across the plasma membrane, is central to the normal physiology of these cells and thus skin function. Numerous skin pathologies including dermatitis, anhidrotic ectodermal dysplasia, hyperhidrosis, hair loss and cancer are now linked to dysfunction in SOCE proteins. Principal amongst these are the stromal interaction molecules (STIMs) that sense Ca2+ depletion and Orai channels that mediate Ca2+ influx. In this review, the roles of STIM, Orai and other store-operated channels are discussed in the context of keratinocyte differentiation, melanogenesis, and eccrine sweat secretion. We explore not only STIM1-Orai1 as drivers of SOCE, but also independent actions of STIM, and emerging signal cascades stemming from their activities. Roles are discussed for the elusive transient receptor potential canonical channel (TRPC) complex in keratinocytes, Orai channels in Ca2+-cyclic AMP signal crosstalk in melanocytes, and Orai isoforms in eccrine sweat gland secretion.
... The interaction of EDN3 with EDNRB is essential for vasoconstriction, proliferative activities, and the development of NC-derived cell lineages, such as melanocytes and enteric neurons (Bondurand et al., 2018). In a cultured human melanocytes cell line, EDNRB signaling was shown to influence the expression and posttranslational modifications of the MITF gene (Sato-Jin et al., 2008). Mice lacking the EDN3/EDNRB receptor-mediated signaling showed defects in enteric neurons and melanocytes derived from a trunk/vagal NC, resulting in megacolons and coat color spotting (Baynash et al., 1994). ...
... MITF is regulated via several pathways, some of which include: (1) the MC1R (melanocortin 1 receptor) pathway, (2) the wingless/integrated (WNT) pathway, (3) the c-KIT pathway, and (4) the endothelin B receptor (ETBR) pathway. Together, these conserved, well-studied signaling pathways regulate the development of melanin-producing cells in both mammalian and zebrafish skin [48][49][50][51][52][53][54][55][56][57][58][59][60][61][62][63][64][65]. ...
Article
Full-text available
Simple Summary Zebrafish (Danio rerio) is an emerging model for studying many diseases, including disorders originating in black pigment cells, melanocytes. In this review of the melanocyte literature, we discuss the current knowledge of melanocyte biology relevant to understanding different forms of albinism and the potential of the zebrafish model system for finding novel mechanisms and treatments. Abstract Melanin is the pigment that protects DNA from ultraviolet (UV) damage by absorbing excess energy. Melanin is produced in a process called melanogenesis. When melanogenesis is altered, diseases such as albinism result. Albinism can result in an increased skin cancer risk. Conversely, black pigment cell (melanocyte) development pathways can be misregulated, causing excessive melanocyte growth that leads to melanoma (cancer of melanocytes). Zebrafish is an emerging model organism used to study pigment disorders due to their high fecundity, visible melanin development in melanophores (melanocytes in mammals) from 24 h post-fertilization, and conserved melanogenesis pathways. Here, we reviewed the conserved developmental pathways in zebrafish melanophores and mammalian melanocytes. Additionally, we summarized the progress made in understanding pigment cell disease and evidence supporting the strong potential for using zebrafish to find novel treatment options for albinism.
... Thus, increased expression of mitfa in our melanoma samples is likely associated with survival and proliferation of tumor cells as the samples were at the same time significantly enriched for the cell cycle and proliferation-related signatures. The key role of human MITF in melanocyte differentiation occurs through direct transcriptional control of several genes including TYR, TYRP1, DCT, KIT, MC1R, PMEL17 and SLC45A2 as well as indirect ones such as EDNRB (Sato-Jin, Nishimura et al. 2008, Cheli, Ohanna et al. 2010. We have found an array of pigmentation genes including kita, dct, tyrp1a, tyrp1b, ednrba, ednrbb, mc1r, pmela and slc45a2 to be significantly upregulated during late melanocyte regeneration and melanoma, strongly suggesting that their expression is transcriptionally controlled by mitfa. ...
Preprint
Full-text available
Tissue regeneration and cancer share remarkable features including activation of cell proliferation and migration. Yet, tumors considerably differ from the regenerating tissue with respect to abnormal proliferation, invasive growth and metastasis. Thus, it is likely that cancer resembles early stages of regeneration with increased proliferation, but separates from the later stages with reduced proliferation and enhanced differentiation. Here, by exploiting the zebrafish melanocytes that can efficiently regenerate and be induced to undergo malignant melanoma, we unravel the transcriptome profiles of the regenerating melanocytes during early and late regeneration, and the melanocytic nevi and malignant melanoma. Our global comparison of the gene expression profiles of melanocyte regeneration and nevi/melanoma uncovers opposite regulation of Wnt and BMP signaling pathways between regeneration and cancer. Functional inhibition of canonical Wnt or BMP pathways during melanocyte regeneration impeded the regenerative capacity of the melanocytes. In contrast, activation of either pathway significantly promoted melanocyte regeneration and potently suppressed migration of human melanoma cells. Therefore, mechanisms of regeneration can be exploited to correct dysregulated molecular pathways and malignant transformation in cancer.
... Also, MITF interacts with various cofactors to regulate gene expression. Thus, MITF was shown to associate with chromatin modifiers and remodellers such as CBP/p300 (Sato-Jin et al., 2008), NURF and SWI/SNF complexes containing BRG1 , and with essential transcription factors including β-catenin (A. . ...
Thesis
The Nucleosome Remodelling and Deacetylation (NuRD) complex is an epigenetic regulator of gene expression that includes two mutually exclusive ATPase subunits CHD3 and CHD4. Our results show that NuRD associates with essential melanoma cell transcription factors namely MITF and SOX10. However, despite their physical association and genomic co-localization, CHD4-NuRD does not appear to act as a cofactor for MITF or SOX10 regulated gene expression. Nevertheless, CHD4 silencing leads to a slow growth phenotype and de-represses the expression of PADI1 (Protein Arginine DeIminase 1) and PADI3, two enzymes involved in converting arginines to citrullines in melanoma and multiple types of cancer cells. Increased expression of PADI1 and PADI3 enhances citrullination of arginines within the key glycolytic regulatory enzyme PKM2 then promoting excessive glycolysis, lowering ATP levels and slowing down proliferation. PKM2 citrullination lowers its sensitivity to allosteric inhibitors thus shifting equilibrium towards allosteric activators thereby bypassing the normal physiological regulation of glycolysis. Overall, our results lead to describe a novel pathway linking, epigenetic regulation of PADI1 and PADI3 expression by CHD4/NuRD and reprogramming of PKM2 allosteric regulation through arginines citrullination, to glycolytic flux and cancer cell proliferation.
... While MITF promotes a melanocytic cell state, Axl is associated with a less differentiated, stem-like cell state. ETBR is a MITF target gene [21,38] and it could account for MAPK inhibitor resistance-associated with elevated MITF expression in melanoma cells. In the cellular systems studied here, ETBR was the only functional endothelin receptor that was expressed. ...
Article
Full-text available
The clinical benefit of MAPK pathway inhibition in melanoma patients carrying BRAF mutations is temporal. After the initial response to treatment, the majority of tumors will develop resistance and patients will relapse. Here we demonstrate that the endothelin-endothelin receptor B (ETBR) signaling pathway confers resistance to MAPK pathway inhibitors in BRAF mutated melanoma. MAPK blockade, in addition to being anti-proliferative, induces a phenotypic change which is characterized by increased expression of melanocyte-specific genes including ETBR. In the presence of MAPK inhibitors, activation of ETBR by endothelin enables the sustained proliferation of melanoma cells. In mouse models of melanoma, including patient-derived xenograft models, concurrent inhibition of the MAPK pathway and ETBR signaling resulted in a more effective anti-tumor response compared to MAPK pathway inhibition alone. The combination treatment significantly reduced tumor growth and prolonged survival compared to therapies with MAPK pathway inhibitors alone. The phosphoproteomic analysis revealed that ETBR signaling did not induce resistance towards MAPK pathway inhibitors by restoring MAPK activity, but instead via multiple alternative signaling pathways downstream of the small G proteins GNAq/11. Together these data indicate that a combination of MAPK pathway inhibitors with ETBR antagonists could have a synergistically beneficial effect in melanoma patients with hyperactivated MAPK signaling pathways.
Article
Full-text available
Melanoma is the deadliest form of skin cancer and develops from the melanocytes that are responsible for the pigmentation of the skin. The skin is also a highly regenerative organ, harboring a pool of undifferentiated melanocyte stem cells that proliferate and differentiate into mature melanocytes during regenerative processes in the adult. Melanoma and melanocyte regeneration share remarkable cellular features, including activation of cell proliferation and migration. Yet, melanoma considerably differs from the regenerating melanocytes with respect to abnormal proliferation, invasive growth, and metastasis. Thus, it is likely that at the cellular level, melanoma resembles early stages of melanocyte regeneration with increased proliferation but separates from the later melanocyte regeneration stages due to reduced proliferation and enhanced differentiation. Here, by exploiting the zebrafish melanocytes that can efficiently regenerate and be induced to undergo malignant melanoma, we unravel the transcriptome profiles of the regenerating melanocytes during early and late regeneration and the melanocytic nevi and malignant melanoma. Our global comparison of the gene expression profiles of melanocyte regeneration and nevi/melanoma uncovers the opposite regulation of a substantial number of genes related to Wnt signaling and transforming growth factor beta (TGF-β)/(bone morphogenetic protein) BMP signaling pathways between regeneration and cancer. Functional activation of canonical Wnt or TGF-β/BMP pathways during melanocyte regeneration promoted melanocyte regeneration but potently suppressed the invasiveness, migration, and proliferation of human melanoma cells in vitro and in vivo. Therefore, the opposite regulation of signaling mechanisms between melanocyte regeneration and melanoma can be exploited to stop tumor growth and develop new anti-cancer therapies.
Article
Full-text available
Hair graying is a common and visible sign of aging resulting from decreased or absence of melanogenesis. Although it has been established that gray hair greatly impacts people's mental health and social life, there is no effective countermeasure other than hair dyes. It has long been thought that reversal of gray hair on a large scale is rare. However, a recent study reported that individual gray hair darkening is a common phenomenon, suggesting the possibility of large-scale reversal of gray hair. In this article, we summarize the regulation mechanism of melanogenesis and review existing cases of hair repigmentation caused by several factors, including monoclonal antibodies drugs, tyrosine kinase inhibitors (TKIs), immunomodulators, other drugs, micro-injury, and tumors, and speculate on the mechanisms behind them. This review offers some insights for further research into the modulation of melanogenesis and presents a novel perspective on the development of clinical therapies, with emphasis on topical treatments.
Article
Full-text available
The natural colour of hair shafts is formed at the bulb of hair follicles, and it is coupled to the hair growth cycle. Three critical processes must happen for efficient pigmentation: (1) melanosome biogenesis in neural crest-derived melanocytes, (2) the biochemical synthesis of melanins (melanogenesis) inside melanosomes, and (3) the transfer of melanin granules to surrounding pre-cortical keratinocytes for their incorporation into nascent hair fibres. All these steps are under complex genetic control. The array of natural hair colour shades are ascribed to polymorphisms in several pigmentary genes. A myriad of factors acting via autocrine, paracrine, and endocrine mechanisms also contributes for hair colour diversity. Given the enormous social and cosmetic importance attributed to hair colour, hair dyeing is today a common practice. Nonetheless, the adverse effects of the long-term usage of such cosmetic procedures demand the development of new methods for colour change. In this context, case reports of hair lightening, darkening and repigmentation as a side-effect of the therapeutic usage of many drugs substantiate the possibility to tune hair colour by interfering with the biology of follicular pigmentary units. By scrutinizing mammalian pigmentation, this review pinpoints key targetable processes for the development of innovative cosmetics that can safely change the hair colour from the inside out.
Article
Full-text available
Background Human skin is constantly exposed to solar ultraviolet radiation (UVR). Skin has a unique ability to respond to UVR by increasing its pigmentation, a protective process driven by the melanogenesis of human epidermal melanocytes (HEMs). However, the molecular mechanisms used by HEMs to detect and respond to UVR remain unclear. Objectives We investigated the function and potential mechanism of opsin 5 (OPN5) in melanogenesis in HEMs. Methods We used NaOH and L‐DOPA methods to determine the melanin content and tyrosinase activity in HEMs, respectively. The expression of OPN5 in HEMs and explant tissues was detected by Western blotting (WB), reverse transcription PCR (RT‐PCR) and immunofluorescence assays. RT‐PCR and WB detected OPN5 expression levels in treated and untreated melanocytes. Subsequently, WB and RT‐PCR were used to detect changes in the expression of tyrosinase (TYR) and tyrosine‐related proteases (TRP1, TRP2) caused by changes in the OPN5 expression level. Moreover, changes in signalling pathway protein molecules were also tested. Results Here, we report that OPN5 is the key sensor in HEMs responsible for melanogenesis induced by UVR. OPN5‐induced melanogenesis requires calcium‐dependent G protein‐coupled receptor and PKC signal transduction, thus contributing to the UVR‐induced MITF response to mediate downstream cellular effects and providing evidence of OPN5 function in mammalian phototransduction. Remarkably, OPN5 activation is necessary for UVR‐induced increase in cellular melanin and has an inherent function in melanocyte melanogenesis. Conclusion Our results provide insight into the molecular mechanisms of UVR in melanocytes and may reveal molecular targets for preventing pigmentation or pigment diseases.
Article
Full-text available
We demonstrate here that human melanocytes could be regulated by endothelin (ET) derivatives, potent vasoconstrictive peptides synthesized by endothelial cells, to stimulate their proliferation and melanization via a receptor-mediated signal transduction pathway. Receptor-binding assay using [125I]ET indicated that unlabeled ET-1 or ET-2 competitively inhibited each binding of labeled ETs to melanocytes with a concentration for half-maximal inhibition (IC50) of 0.7 or 0.9 nM, respectively. The dissociation constant (Kd) and the number of sites of the specific bindings of ET-1 and those of ET-2 were almost the same (Kd: 1.81 nM, binding sites: 7.0-8.0 x 10(4) per cell). Upon incubation with cultured cells, the mass contents of inositol 1,4,5-trisphosphate and intracellular calcium level were substantially increased by 10 nM ET-1, ET-2, and ET-3, but not by big-ET with maximal response at 80-130-s postincubation. The addition of ET-1 and ET-2 at 1-50 nM concentrations caused human melanocytes to significantly stimulate DNA [( 3H]thymidine incorporation) and melanin synthesis (3H2O release and [14C] thiouracil incorporation). Furthermore, ETs exhibited an additive stimulatory effect on basic fibroblast growth factor-stimulated DNA synthesis. In a long-term serum-free culture system, the strongest stimulation of growth by 10 nM ET-1 or ET-2 was observed in the presence of 10 nM cholera toxin and 0.2% bovine pituitary extract, resulting in a 4.5-fold increase in cell number for 12 culture days. These findings strongly suggest involvement of ET in the mechanism regulating proliferation and melanization of human melanocytes.
Article
Full-text available
We recently demonstrated that human melanocyte proliferation and differentiation could be stimulated by endothelin (ET) derivatives via a receptor-mediated signal transduction pathway (Yada, Y., Higuchi, K., and Imokawa, G. (1991) J. Biol. Chem. 266, 18352-18357). We show here that the growth factors for human melanocytes are produced and secreted by the surrounding cells, namely human keratinocytes for ET-1 and Big-ET-1. Northern blots have revealed the presence of ET-1 gene transcripts in proliferating human keratinocytes. The ET-1 production by human keratinocytes increased after irradiation with ultraviolet B (UVB) in a dose-dependent manner, accompanied by the significant secretion of interleukin 1alpha (IL-1alpha). Among the cytokines related to UVB-induced cellular reactions and keratinocyte growth, only IL-1alpha and -1beta stimulated the secretion of ET-1 and Big-ET-1 but not of ET-3 and Big-ET-3 in a time-dependent manner. Northern blots for IL-1alpha-stimulated or UVB-exposed human keratinocytes revealed that production of ET-1 gene transcripts markedly increased (by about 300 or 1,200%) with constant levels of beta-actin gene transcripts. In a parallel study, the medium conditioned by UVB-exposed human keratinocytes elicited a significant anti-ET-1 antibody-suppressible increase in DNA synthesis by cultured human melanocytes in a UV dose-dependent manner, which was associated with a marked and rapid (80 s) increase in the intracellular calcium level upon incubation with human melanocytes. These studies suggest that ETs produced and secreted by keratinocytes play an essential role in the maintenance of melanocyte proliferation and UV hyperpigmentation in the epidermis.
Article
Full-text available
In the human skin, melanocytes are present in the epidermis and hair follicles. The basic features of these cells are the ability to melanin production and the origin from neural crest cells. This last element is important because there are other cells able to produce melanin but of different embryonic origin (pigmented epithelium of retina, some neurons, adipocytes). The life cycle of melanocyte consists of several steps including differentiation of melanocyte lineage/s from neural crest, migration and proliferation of melanoblasts, differentiation of melanoblasts into melanocytes, proliferation and maturation of melanocytes at the target places (activity of melanogenic enzymes, melanosome formation and transport to keratinocytes) and eventual cell death (hair melanocytes). Melanocytes of the epidermis and hair are cells sharing some common features but in general they form biologically different populations living in unique niches of the skin.
Article
The injection of an antagonistic anti-murine c-kit mono-clonal antibody ACK2 during mouse embryonic development produced three distinctive pigmentation patterns on the coat of the offspring. Pattern 1 consisted of pigmentation in craniofacial and caudal regions and was induced by an ACK2 injection between 9.5 and 11.5 days post coitum (dpc). In pattern 2, the entire coat was unpigmented and was induced by the injection at around 13.0 dpc. Pattern 3 consisted of pigmented patches spreading ventrolaterally from the dorsoanterior trunk regions towards the anterior and posterior directions and it was induced by ACK2 administered at 14.5-15.0 dpc. We investigated the embryological basis of these nonuniform pigmentation patterns to elucidate the process of melanoblast differentiation between lineage commitment and colonization into devel-oping hair follicles. The results showed the following. (1) Melanocyte differentiation at the embryonic stage from 10.5 to 12.5 dpc progresses in a spatially nonuniform fashion, being faster in the craniofacial and caudal regions than in the trunk; pattern 1 reflects this. (2) Melanoblasts are activated to proliferate synchronously upon entering into the epidermis; pattern 2 correlates with this process. (3) c-kit functions as a survival signal for proliferating melanoblasts in the epidermis. (4) The melanoblasts that enter develop-ing hair follicles can survive without a c-kit signal; pattern 3 essentially represents the hair follicles colonized by these cells. Analysis of the melanoblast distribution of ls/ls embryos that bear a loss-of-function mutation in the endothelin 3 gene suggested that endothelin 3 is required for early melanoblast differentiation before entering into the epidermis, whereas proliferation in the epidermis takes place without this molecule. Based on these data, we propose 4 distinct steps of embryonic melanocyte differentiation: (1) migration in the dermis, which requires both c-kit and endothelin 3; (2) a stage before epidermal entry that is resistant to anti-c-kit mAb; (3) cell proliferation after entering the epidermal layer, which requires c-kit and endothelin receptor B but not endothelin 3 and (4) integration into developing hair follicles, which renders melanoblasts resistant to anti-c-kit mAb. Thus, melanoblast differentiation proceeds by alternately repeating c-kit-dependent and c-kit-independent stages and c-kit functions as a survival factor for the proliferating melanoblasts.
Article
Microphthalmia (Mi) is a bHLHZip transcription factor that is essential for melanocyte development and postnatal function. It is thought to regulate both differentiated features of melanocytes such as pigmentation as well as proliferation/survival, based on phenotypes of mutant mouse alleles. Mi activity is controlled by at least two signaling pathways. Melanocyte-stimulating hormone (MSH) promotes transcription of the Mi gene through cAMP elevation, resulting in sustained Mi up-regulation over many hours. c-Kit signaling up-regulates Mi function through MAP kinase phosphorylation of Mi, thereby recruiting the p300 transcriptional coactivator. The current study reveals that c-Kit signaling triggers two phosphorylation events on Mi, which up-regulate transactivation potential yet simultaneously target Mi for ubiquitin-dependent proteolysis. The specific activation/degradation signals derive from MAPK/ERK targeting of serine 73, whereas serine 409 serves as a substrate for p90 Rsk-1. An unphosphorylatable double mutant at these two residues is at once profoundly stable and transcriptionally inert. These c-Kit-induced phosphorylations couple transactivation to proteasome-mediated degradation. c-Kit signaling thus triggers short-lived Mi activation and net Mi degradation, in contrast to the profoundly increased Mi expression after MSH signaling, potentially explaining the functional diversity of this transcription factor in regulating proliferation, survival, and differentiation in melanocytes.
Article
The silver mutation in mice causes progressive graying of hair due to the loss of functional follicular melanocytes. Recently the silver locus gene (called Pmel 17) has been cloned; its encoded product shares homology with a chick melanosomal matrix protein and a bovine retinal pigment epithelial protein. Although the sequence of the silver gene and the correlation of its expression with pigment production have been reported, its function in melanogenesis is still unknown. In an effort to characterize that function, we have synthesized the predicted carboxyl-terminal peptide of the mouse Pmel 17 protein and generated a rabbit polyclonal antibody (alpha PEP13) to it; that antibody recognized the silver protein specifically. The immunoaffinity-purified silver protein lacked all of the known melanogenic catalytic activities which other tyrosinase-related proteins (TRP) have, nor did it appear to modulate any of those TRP activities. Metabolic labeling experiments demonstrated that the silver protein disappears in vivo within a few hours, indicating that it is rapidly degraded, or quickly processed to lose its carboxyl terminus. Crossreactivity experiments showed that a recently reported anti-melanosomal matrix protein antibody (alpha MX) also recognizes the silver protein, although at a different epitope from that of alpha PEP13. Using Western immunoblotting, we analyzed subcellular fractions isolated from B16 F10 melanoma cells and found that the silver protein was rich in the melanosome fraction but was absent from coated vesicles which deliver TRPs to melanosomes. These results suggest that the silver locus product is a melanosomal matrix protein which may contribute to melanogenesis as a structural protein, although the possibility remains that it also has a novel catalytic function in melanogenesis.
Article
To survive and proliferate in pure culture, human melanocytes require basic fibroblast growth factor (bFGF) and cAMP. Without these factors, even in the presence of serum, the cells die. Melanocytes cultured in the presence of keratinocytes, however, survive for weeks without added bFGF and cAMP. We show here that the growth factor for melanocytes produced by human keratinocytes is bFGF because its activity can be abolished by neutralizing antibodies to bFGF and by a bFGF synthetic peptide that inhibits the binding of the growth factor to its receptor. The melanocyte mitogen in keratinocytes is cell associated and increases after irradiation with ultraviolet B. Northern blots reveal bFGF gene transcripts in keratinocytes but not melanocytes. These studies demonstrate that bFGF elaborated by keratinocytes in vitro sustains melanocyte growth and survival, and they suggest that keratinocyte-derived bFGF is the natural growth factor for normal human melanocytes in vivo.
Article
Waardenburg syndrome (WS) is associated with neural crest-derived melanocyte deficiency caused by mutations in either one of three transcription factors: MITF, PAX3, and SOX10. However, the hierarchical relationship of these transcription factors is largely unknown. We show that SOX10 is capable of transactivating the MITF promoter 100-fold, and that this transactivation is further stimulated by PAX3. Promoter deletion and mutational analyses indicate that SOX10 can activate MITF expression through binding to a region that is evolutionarily conserved between the mouse and human MITF promoters. A SOX10 mutant that models C-terminal truncations in WS can reduce wild-type SOX10 induction of MITF, suggesting these mutations may act in a dominant-negative fashion. Our data support a model in which the hypopigmentation in WS, of which these factors have been implicated, results from a disruption in function of the central melanocyte transcription factor MITF.