Sie sind auf Seite 1von 10

Copyright ª Blackwell Munksgaard 2005 Review

doi: 10.1111/j.1600-0749.2005.00235.x

Adhesion, migration and communication in


melanocytes and melanoma

Nikolas K. Haass, Keiran S. M. Smalley, Ling Li anocyte and approximately 36 associated keratinocytes
and Meenhard Herlyn* (Fitzpatrick and Breathnach, 1963; Jimbow et al., 1976).
Melanocytes are located in the basal layer of the epider-
The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, mis, where they occur in a life-long stable ratio of 1:5
USA with basal keratinocytes. This balance is maintained
*Address correspondence to Dr Meenhard Herlyn, through regulated induction of melanocyte division. In
e-mail: herlynm@wistar.org order to proliferate, melanocytes need to decouple from
the basement membrane and from the keratinocytes,
Summary retract their dendrites, divide, and migrate along the
basement membrane before they finally re-couple to
Under normal conditions, homeostasis determines
the matrix and to keratinocytes to form another epider-
whether a cell remains quiescent, proliferates, differ-
mal melanin unit (reviewed in Haass and Herlyn, in
entiates, or undergoes apoptosis. In this state of
press). Melanocyte growth is controlled by the sur-
homeostasis, keratinocytes control melanocyte
rounding keratinocytes by (1) extracellular communica-
growth and behaviour through a complex system of
tion through paracrine growth factors, by (2) intracellular
paracrine growth factors and cell–cell adhesion mole-
communication through second messengers and signal
cules. Alteration of this delicate homeostatic balance
transduction and by (3) intercellular communication
and can lead to altered expression of cell–cell adhe-
through cell–cell adhesion molecules, cell–matrix adhe-
sion and cell communication molecules and to the
sion, and gap junctional intercellular communication
development of melanoma. Melanoma cells escape
(reviewed in Haass et al., 2004). Under normal condi-
from this control by keratinocytes through three
tions, homeostasis determines whether a cell remains
major mechanisms: (1) down-regulation of receptors
quiescent, proliferates, differentiates, or undergoes
important for communication with keratinocytes
apoptosis (reviewed in Bissell and Radisky, 2001). Dys-
such as E-cadherin, P-cadherin, desmoglein and con-
regulation of the homeostasis may disturb the balance
nexins, which is achieved through growth factors
of the epidermal melanin unit and trigger a continuous
produced by fibroblasts or keratinocytes; (2) up-regu-
proliferation of the melanocytes, which may lead to the
lation of receptors and signalling molecules not
development of melanoma. The hallmarks of solid
found on melanocytes but important for melanoma–
tumours are uncontrolled proliferation, derangement of
melanoma and melanoma–fibroblast interactions
cellular and morphological differentiation, invasion, and
such as N-cadherin, Mel-CAM, and zonula occludens
metastatic spread to distant organs. These characteris-
protein-1 (ZO-1); (3) loss of anchorage to the base-
tics can be in part attributed to alterations in adhesion
ment membrane because of an altered expression of
and communication between neoplastic cells and the
the extracellular-matrix binding integrin family. In
normal cells in their immediate microenvironment
the current review, we describe the alterations in
(reviewed in Hanahan and Weinberg, 2000; Park et al.,
cell–cell adhesion and communication associated
2000). The exact molecular mechanisms of this dysreg-
with melanoma development and progression, and
ulation in melanoma are yet unknown. However, it is
discuss how a greater understanding of these pro-
likely that melanoma cells escape from control through
cesses may aid the future therapy of this disease.
keratinocytes through (1) down-regulation of receptors
Key words: melanoma/metastasis/cadherins/cell adhe- important for communication with and adhesion to kera-
sion molecules/integrins tinocytes (e.g. E-cadherin); (2) up-regulation of receptors
and signalling molecules not found on melanocytes but
Received 18 March 2005, revised and accepted for pub-
important for melanoma–melanoma and melanoma–fi-
lication 5 April 2005
broblast interactions [e.g. N-cadherin, melanoma cell
adhesion molecule (Mel-CAM), zonula occludens pro-
tein-1 (ZO-1)]; and (3) loss of anchorage to the base-
Introduction
ment membrane because of an altered expression of
The epidermal melanin unit of the human epidermis the extracellular-matrix binding integrin family. In this
denotes the symbiotic relationship between one mel- review, we will focus on these three mechanisms.

150 Pigment Cell Res. 18; 150–159


Adhesion, migration and communication in melanocytes and melanoma

oma cells show an increase in expression of melanoma


Cell–cell adhesion of melanocytes and
cell adhesion molecule (MCAM, Mel-CAM, MUC18,
melanoma cells
CD146), L1 cell adhesion molecule (L1-CAM, CD171),
Cell–matrix interactions shift from collagen IV- and lami- activated leukocyte cell adhesion molecule (ALCAM,
nin-mediated adhesion in normal melanocytes to attach- CD166), vascular cell adhesion molecule 1 (VCAM-1,
ment through collagen I- and vitronectin-binding CD106), intercellular cell adhesion molecule 1 (ICAM-1,
receptors in melanoma cells. The changes in matrix CD54), and carcinoembryonic antigen-related cell adhe-
adhesion receptor composition often reflect aggressive sion molecule 1 (CEACAM1, CD66a). The adhesive
properties of melanoma cells as they invade the dermis functions of cellular adhesion molecules in homophilic
from the epidermis (reviewed in Johnson, 1999; Nesbit and heterophilic interactions are well documented, but
and Herlyn, 1994). Cell–cell adhesion receptors show their contributions to the malignant phenotype are less
similar changes during tumour progression (Figure 1). defined.
Among the classical cadherins, there is a shift from Mel-CAM mediates homologous and heterologous
E-cadherins found on normal melanocytes for attach- interactions between melanoma cells and endothelial
ment to keratinocytes to N-cadherin on melanoma cells, cells respectively via a heterophilic Ca2+-independent
which allows coupling to fibroblasts and endothelial cells adhesion to a currently unidentified ligand (Johnson
in the tumour stroma (Hsu et al., 2000). Even if melan- et al., 1997; Shih et al., 1997a,b). In melanocytic cells
oma cells express E-cadherin, it does not appear expression of Mel-CAM is first found in nevi, when the
functional. Whereas normal melanocytes express few cells have separated from the epidermal keratinocytes
cell–cell adhesion receptors of the immunoglobulin gene and have migrated into the dermis (Kraus et al., 1997;
superfamily of cell adhesion molecules (CAMs), melan- Shih et al., 1994). With progression to malignancy, Mel-

Figure 1. Cell–cell adhesion of


melanocytes and melanoma cells.
Melanocytes adhere to keratinocytes via
E-cadherin and desmoglein, which enables
them to communicate with each other
through gap junctions (A). In melanoma
cells E-cadherin is down-regulated. They
interact with each other through N-
cadherin, Mel-CAM/Mel-CAM ligand, avb3
integrin/L1-CAM, ALCAM and connexins,
with fibroblasts through N-cadherin and
connexins, and with endothelial cells
through N-cadherin, Mel-CAM/Mel-CAM
ligand, avb3 integrin/L1-CAM, a4b1 integrin/
VCAM-1 and connexins (B).

Pigment Cell Res. 18; 150–159 151


Haass et al.

CAM expression gradually increases and is highest in mary cutaneous malignant melanoma is significantly
metastatic melanoma cells (Johnson et al., 1996; Leh- associated with metastatic spread (Thies et al., 2002b).
mann et al., 1989; Shih et al., 1994; Xie et al., 1997). L1-CAM mediates adhesion both via homophilic (L1-
Furthermore, Mel-CAM expression correlates with the CAM-L1-CAM) and heterophilic (L1-CAM-avb3 integrin)
tumour thickness (Lehmann et al., 1989; Shih et al., mechanisms (Hortsch, 1996). In melanoma cell/melan-
1994). oma cell and in melanoma cell/endothelial cell interac-
Inhibition of Mel-CAM expression in metastatic mel- tions L1-CAM binds to avb3 integrin (Montgomery
anoma cells using genetic suppressor elements of et al., 1996). The interaction of L1-CAM and avb3 inte-
Mel-CAM cDNA leads to inhibition of adhesion grin has been shown to play an important role in tran-
between melanoma cells and to down-regulation of the sendothelial migration of melanoma cells (Voura et al.,
tumorigenic phenotype (Satyamoorthy et al., 2001). In 2001).
melanoma–melanoma interactions, Mel-CAM and its ALCAM is involved in homophilic (ALCAM–ALCAM)
ligand appear to play a role as co-receptor for N-cadh- (Degen et al., 1998) and heterophilic (ALCAM–CD6)
erin, because its down-modulation disrupts gap junc- (Patel et al., 1995) cell–cell–adhesion interactions. AL-
tional intercellular communication (Satyamoorthy et al., CAM is expressed in metastatic human melanoma cells,
2001). Furthermore, neutralizing antibodies to Mel-CAM whereas it is absent in non-metastatic cells (Degen
block melanoma metastasis in vivo and block the et al., 1998). Immunohistochemistry on a series of
homologous interaction between melanoma cells and human melanocytic lesions from common nevus to mel-
endothelial cells as well as the promoter and collage- anoma metastasis revealed that ALCAM expression cor-
nase activity of matrix metalloproteinase-2 (MMP-2) relates with melanoma progression (van Kempen et al.,
(McGary et al., 2002). Forced Mel-CAM expression of 2000). ALCAM is, therefore, proposed to be molecular
human melanoma cells has an influence on later sta- melanoma progression marker besides the b3 integrin
ges of the metastatic process only, namely, extravasa- subunit and ICAM-1 (see below). Using the N-terminally
tion and establishment of new foci of growth, but is truncated mutant DN-ALCAM to attenuate the function
not sufficient for this process per se (Schlagbauer-Wadl of wild type ALCAM in metastatic cells and to assess
et al., 1999). In contrast, transfection of Mel-CAM into the overall effect of reducing homotypic tumour cell
non-tumorigenic cells increases collagenase gene adhesion on tumorigenicity it has been shown that the
expression, leading to increased tumorigenicity (Xie intact cell adhesion function of ALCAM favours primary
et al., 1997). In three-dimensional skin reconstructs, tumour growth and represented a rate-limiting step for
the overexpression of Mel-CAM in melanoma cells tissue invasion, which supported the view that dynamic
allowed them to separate from the epidermis and control of ALCAM plays an important role in progression
invade into the dermis through the basement mem- (van Kempen et al., 2004). A broader scope on the func-
brane, whereas melanoma cells with little or no Mel- tions of ALCAM can be found in another review (Swart
CAM were poorly invasive (Satyamoorthy et al., 2001). et al., in press).
Mel-CAM expression in melanocytes, nevus cells, and VCAM-1 is a cytokine inducible cell adhesion molecule
radial growth phase melanoma cells is environmentally primarily found on vascular endothelial cells. As a recep-
regulated through direct cell–cell contact with keratino- tor for a4b1 integrin, expressed by malignant melanoma,
cytes (Li and Herlyn, 2000), but the mechanisms of it facilitates melanoma cell attachment to the vascular
this regulation are not known. Even advanced primary endothelium prior to extravasation (reviewed in Holz-
and metastatic melanoma cells down-regulate Mel- mann et al., 1998).
CAM when intimate attachment to keratinocytes is ICAM-1 in melanoma also correlates with melanoma
re-established by over-expression of E-cadherin (Hsu progression and increased risk of metastasis (Johnson
et al., 2000). Loss of several tumour-associated adhe- et al., 1989). Its expression in melanoma is stronger
sion receptors in E-cadherin expressing melanoma cells than in common nevi and increases with the Breslow
is associated with lack of invasion and increased apop- index in primary melanomas (Natali et al., 1990, 1997;
tosis of single cells in the dermal environment (Li and Schadendorf et al., 1993, 1995). The observation that
Herlyn, 2000). stage I patients with ICAM-1 positive melanomas had a
Although originally described as a neuronal cell adhe- significantly shorter disease-free interval and overall sur-
sion molecule restricted to nervous tissues, L1-CAM vival than those with ICAM-1 negative tumours (Natali
has also been detected in various other cell types as et al., 1997) and that the suppression of ICAM-1 in an
well as a number of tumours, including osteogenic sar- animal model reduced the metastatic capacity (Miele
coma, squamous cell carcinoma of the lung, rhabdomy- et al., 1994), supported the role of ICAM-1 in melanoma
osarcoma and retinoblastoma (Nolte et al., 1999; Thies progression and metastasis. ICAM-1 can be induced in
et al., 2002b). There is an increase in L1-CAM immuno- a cell-specific manner by several cytokines, e.g. tumour
reactivity in melanomas and metastases of melanoma necrosis factor-alpha (TNF-a), interleukin-1 (IL-1), and
compared with acquired melanocytic naevi (Fogel interferon-gamma (IFN-c). The ligands of ICAM-1 are
et al., 2003). The expression of L1-CAM in human pri- aLb2 (lymphocyte function-associated antigen 1, LFA-1)

152 Pigment Cell Res. 18; 150–159


Adhesion, migration and communication in melanocytes and melanoma

and Mac1 on lymphocytes (van de Stolpe and van der Under two-dimensional culture conditions in vitro,
Saag, 1996). However, the specific role of ICAM-1 in melanoma cells overexpressing either the b3 integrin
melanoma progression remains to be determined. subunit or Mel-CAM did not show a significant differ-
Expression of ICAM-1 may promote aggregate forma- ence in growth in comparison with the control cells
tion with leucocytes, which can enhance survival in the transfected with the corresponding vector alone
vascular system and encourage extravasation (Aeed (Figure 2A, D). In contrast, both melanoma cells overex-
et al., 1988). On the other hand ICAM-1 is shed from pressing the b3 integrin subunit or Mel-CAM showed
melanoma cells (Giavazzi et al., 1992) – possibly in a striking phenotypes under three-dimensional conditions
form that inhibits lymphocyte-tumour cell interaction (Hsu et al., 1998; Satyamoorthy et al., 2001). Melanoma
and thus contribute to tumour survival (Becker et al., cells infected with the adenovirus b3/Ad5 or Mel-CAM/
1993). Ad5 invaded deep into the dermis and formed cell nests
CEACAM1 is involved in intercellular adhesion and resembling a VGP melanoma (Figure 2B, E), whereas
subsequent signal transduction events in a number of the lacZ/Ad5-infected control cells spread only horizon-
epithelia. In epithelial cells, CEACAM1 is believed to act tally in an RGP melanoma-like manner (Figure 2C, F).
as a growth suppressor, since its expression was Moreover, lacZ/Ad5-infected cells showed the clear
shown to be lost or significantly down- or dysregulated morphological signs of apoptosis, including nuclear con-
in carcinomas of liver (Hixson and McEntire, 1989), densation, membrane blebbing, and apoptotic bodies
prostate (Kleinerman et al., 1995), endometrium (Bam- (Figure 2C, F). However, the mechanism leading to this
berger et al., 1998), breast (Riethdorf et al., 1997) and phenomenon is yet unknown.
colon (Nollau et al., 1997). On the other hand, CEA-
CAM1 is up-regulated in non-small cell lung cancer (Sie-
Keratinocytes are the masters over
nel et al., 2003). CEACAM1 interacts with the b3
melanocytes and E-cadherin
integrin subunit via the CEACAM1 cytoplasmic domain.
overexpressing melanoma cells
CEACAM1 and the b3 integrin subunit co-localize at the
tumour–stroma interface of invading melanoma masses, Keratinocytes down-regulate the cell surface expression
suggesting that CEACAM1-integrin b3 interaction plays of the b3 integrin subunit and of Mel-CAM in melanoma
a role in melanoma cell migration and invasion (Brum- cells overexpressing E-cadherin (Hsu et al., 2000). In
mer et al., 2001). Expression of CEACAM1 in primary contrast to lacZ/Ad5-infected control cells (Figure 3A, B),
melanomas is associated with the subsequent develop- melanoma cells transduced with E-cad/Ad5 grown in co-
ment of metastatic disease (Thies et al., 2002a). Fur- culture with normal human keratinocytes showed a
thermore, the overexpression of CEACAM1 in complete down-regulation of the b3 integrin subunit and
CEACAM1-negative melanocytic cells and melanoma of Mel-CAM (Figure 3D, E).
cell lines increases the migratory and invasive growth Moreover, keratinocytes inhibit invasion of E-cadherin-
potentials in vitro (Ebrahimnejad et al., 2004) supporting overexpressing melanoma cells into the dermis in
the role of CEACAM1 in melanoma progression and human skin reconstructs (Figure 3C, F). Metastatic mel-
metastasis. anoma cells were transduced with either lacZ/Ad5 or
E-cad/Ad5 and incorporated into the epidermal compart-
ments of skin reconstructs. E-cadherin-overexpressing
Growth of cells is tissue context
melanoma cells grew exclusively in the epidermis, at
dependent
the epidermal/dermal junction, and in the upper dermis,
The function of cell adhesion molecules is profoundly displaying typical signs of apoptosis (Figure 3F). In con-
influenced by the extracellular milieu. Therefore, melan- trast, lacZ-transduced cells formed strands of cell nests
oma invasion should be examined under physiological that invaded deep into the dermis and were not apop-
conditions, e.g. by incorporating melanoma cells into totic (Figure 3C).
three-dimensional skin reconstructs. Skin reconstructs
consist of artificial skin rebuilt from isolated cell popula-
Melanocyte matrix adhesion and
tions and composed of a stratified, terminally differenti-
melanocyte migration
ated epidermal compartment of keratinocytes and
melanocytes, a dermal compartment consisting of fibro- The ability of a cell to adhere to its stromal microenvi-
blasts embedded in collagen, and a well established base- ronment is an integral feature of tissue organization.
ment membrane deposited by skin cells (Meier et al., The integrins constitute a major family of cell–cell and
2000). In human skin reconstructs, melanoma cells from cell–extracellular matrix (ECM) adhesion proteins. In
different stages of progression have the same properties addition to adhesive function, integrins are also involved
as cells in situ, i.e. non-tumorigenic melanomas are in signalling, and transmembrane cytoskeletal attach-
unable to invade the dermis from the epidermis, whereas ments (reviewed in Hynes, 2002). Each integrin consists
tumorigenic (advanced primary and metastatic) melan- of a non-covalently linked a and b subunit, which both
oma cells readily invade the dermis (Meier et al., 2000). are composed of an extracellular domain, a single mem-

Pigment Cell Res. 18; 150–159 153


Haass et al.

A
B C

D
E F

Figure 2. Growth of cells is tissue context dependent. In vitro growth of melanoma cells SBcl2 after b3 integrin overexpression (A). Two
days after viral infection, 2 · 105 cells were seeded into six-well tissue culture plates. Cell growth was monitored daily. Average cell number
from triplicate wells was plotted for SBcl2 cells. Despite the striking phenotype in three-dimensional models (see B, C) there is no significant
difference in the growth in a monolayer (A). Effect of b3 integrin subunit overexpression on melanoma invasion and survival in three-
dimensional skin reconstructs (B, C). Virus-infected SBcl2 melanoma cells were incorporated into the epidermis of skin reconstructs as
previously described (Hsu et al., 1998). b3/Ad5-infected SBcl2 cells grew in an invasive pattern reminiscent of VGP primary melanoma (B);
whereas lacZ/Ad5-infected cells spread horizontally, resembling RGP primary melanoma (C). Control virus-infected cells at the dermal/
epidermal junction displayed apoptotic features, including nucleus condensation, membrane blebbing, and presence of apoptotic bodies (C).
Mel-CAM-dependent invasion, survival, and growth of melanoma cells in organotypic skin reconstructs (Satyamoorthy et al., 2001). SBcl-2
cells after transduction with Mel-CAM using an adenoviral vector invaded into the dermis (E), whereas control adenovector LacZ/Ad5
transduced, non-tumorigenic, Mel-CAM-negative SBcl2 melanoma cells grew in the epidermis but did not invade into the dermis (F). In vitro
growth of melanoma cells after Mel-CAM overexpression (D). Again, there is no significant difference in the growth in a monolayer as
opposed to the three-dimensional model (E, F).

brane-spanning domain, and a short cytoplasmic tail. In are able to move forwards through the ECM (Raucher
total there are 18 a and 8 b subunits, giving rise to 25 and Sheetz, 2000). Interestingly, although cells require
distinct integrin permutations; all with different sub- some adhesion to the ECM to generate the traction
strate selectivities (reviewed in Hynes, 2002). They can needed for invasion, maximal rates of cell movement
be grouped according to their evolutionary relationships are found only at intermediate levels of adhesion. Thus,
and ligand specificity. Knockout experiments have at low levels of cell adhesion, insufficient traction is
revealed the different roles and ligand specificities of generated to facilitate movement, whereas strong adhe-
the integrins (reviewed in Hynes, 1996, 2002). During sion to the ECM effectively blocks any motility (Palecek
migration, cells project lamellipodia that attach to the et al., 1998, 1999). The optimal situation for cell invasion
ECM. At the same time cell–ECM contacts are being is, therefore, somewhere in-between the two states,
broken at the trailing edge of the cell, and thus the cells allowing some adhesion at the leading edge of the cell

154 Pigment Cell Res. 18; 150–159


Adhesion, migration and communication in melanocytes and melanoma

A B C

D E F

Figure 3. Keratinocytes down-regulate cell surface expression of Mel-CAM by E-cadherin-expressing melanoma cells. Melanoma cells were
transduced with either E-cad/Ad5 or lacZ/Ad5. After 24 h, cells were mixed with normal human keratinocytes at a 1:5 ratio and the co-cultures
were stained 7 d later. (A) Identification of lacZ-transduced WM115 melanoma cells in co-cultures using mAb Mel-5, a melanosomal marker,
followed by Cy3-conjugated secondary antibody. (B) lacZ-transduced melanoma cells double-stained with mAb A32 defining Mel-CAM.
(D) E-cadherin-transduced melanoma cells identified in the co-cultures using mAb Mel-5. (E) E-cadherin-transduced melanoma cells
double-stained with anti-Mel-CAM mAb A32. Similar down-regulation was observed using SAP mAb against b3 subunit of the vitronectin
receptor (not shown). Scale bars: 40 lm. Keratinocytes inhibit invasion of E-cadherin-expressing melanoma cells into the dermis in human
skin reconstructs (C, F). 1205Lu metastatic melanoma cells were transduced with either lacZ/Ad5 (C) or E-cad/Ad5 (F) and incorporated into
the epidermal compartments of skin reconstructs. At maturation, reconstructs were harvested, fixed, and embedded in paraffin for
haematoxylin and eosin staining. E-cadherin-expressing melanoma cells grew exclusively in the epidermis, at the epidermal/dermal junction,
and in the upper dermis, displaying typical signs of apoptosis (F). In contrast, lacZ-transduced cells formed strands of cell nests that invaded
deep into the dermis and were not apoptotic (C). Magnification: ·20.

to generate traction and the release of contacts at the binds fibronectin (reviewed in van der Flier and Sonnen-
back of the cell to allow forward motion. In this situ- berg, 2001). An important factor in metastatic spread of
ation, integrins are critical at controlling the adhesion to melanoma is its interaction with the various compo-
and release from the ECM. This process is termed nents of the ECM. Only transformed melanocytes can
inside-out signalling, and provides a way for the inte- survive in the altered environment of the dermis, and
grins to respond to cues generated intracellularly to alter this survival seems to be dependent upon expression of
their environmental interaction. These changes of adhe- the cell–ECM adhesion molecules such as the integrins.
siveness are regulated by protein–protein interactions at Increased and altered expression profiles of the inte-
the cytoplasmic tail of the integrin, altering its affinity grins are known to be indicative of melanoma progres-
for its ligands. In melanoma, both aVb3 and b1 integrins sion and melanocytes and early stage primary
are highly expressed at the leading edge of invasive melanoma do not express b3 integrin (Van Belle et al.,
explants (Brooks et al., 1996; Hegerfeldt et al., 2002). 1999). Thus a number of studies have reported that
The movement of primary melanoma through 3D collagen expression of aVb3 and aVb1 integrins are prognostic for
matrix is particularly dependent upon the expression of melanoma progression (Albelda et al., 1990; Felding-
b1-integrin, which is expressed at the leading edge of Habermann et al., 1992; Hsu et al., 1998; Li et al., 1998;
the explant (Hegerfeldt et al., 2002). It further seems Melchiori et al., 1995; Natali et al., 1993; Schumacher
that this integrin is playing an organizing role in the and Schaumburg-Lever, 1999). Of the two, aVb3 integrin
migratory process, as inhibition of b1 integrin activity, is important for adhesion of melanoma cells to dermal
using blocking antibodies, disrupts the polarity of the collagen and the suppression of apoptosis (Montgomery
explant and leads to the development of several leading et al., 1994; Petitclerc et al., 1999). A direct role for the
edges (Hegerfeldt et al., 2002). integrins in melanoma progression is illustrated by
The most relevant integrin combinations to melanoma experiments showing that the forced expression of b3
include - aVb3: which binds to fibronectin, fibrinogen, integrin in an early phase melanoma (radial growth
von Willebrand factor, vitronectin, certain types of colla- phase or RGP) leads to functional expression of the
gen and laminin, and integrin a5b1: which selectively aVb3 integrin and an altered malignant phenotype, which

Pigment Cell Res. 18; 150–159 155


Haass et al.

corresponds to that of the later invasive form of melan-


oma (vertical growth phase or VGP). Other studies have
also shown that expression of aV or b3 integrin
increased metastatic potential of melanoma cells (Fel-
ding-Habermann et al., 1992; Filardo et al., 1996; Li
et al., 1998, 2001). Plating of aVb3 overexpressing mel-
anoma cells onto vitronectin led to increased activation
of FAK and Src, demonstrating a role for these kinases
in integrin-mediated invasion. The aVb3 integrin combina-
tion enhances melanoma cell survival on collagen by
altering the Bcl2:Bax ratio (Petitclerc et al., 1999).
Whereas overexpression of aVb3 integrin enhances mel-
anoma progression and survival, it has also been shown
A that antagonists of this integrin complex can block mel-
anoma growth by inducing apoptosis (Petitclerc et al.,
1999).
In melanocytes in situ a3b1, a6b1 (Figure 4) and avb1
are expressed, while the a1, a2, a4, a7, b2, b4, b5 and b6
subunits as well as a5b1 and avb3 are not expressed
(reviewed in Kuphal et al., in press). However, there is a
strong difference in the integrin expression pattern of in
situ melanocytes compared with cultured melanocytes.
From all above-mentioned integrins, a selective subset
of melanoma associated heterodimers namely a3b1,a6b1
and avb3 are expressed in normal cultured melanocytes,
whereas a1b1 and a4b1 are not expressed.
In situ integrin aV, a2, a3, and a4 levels are increased
in primary and metastatic melanomas (Hartstein et al.,
B
1997; Moretti et al., 1993; Nikkola et al., 2004) whereas
a6 levels are decreased (Danen et al., 1994; Natali et al.,
1993; Yoshinaga et al., 1993). However, whereas in cul-
tured cells with low metastatic potential a1, a2 and a6
levels are undetectable, highly metastatic cells – in con-
trast to tissue sections from melanoma patients – show
up-regulated a6 levels, moderate a1, and no a7 expres-
sion (Ziober et al., 1999). Integrin a3 has also been
shown to be up-regulated in cultured cells and to
enhance motility of melanoma cells (Yoshinaga et al.,
1993; Zhu et al., 2002). A broader scope on the integrin
signalling in malignant melanoma can be found in
another review (Kuphal et al., in press).

C
Conclusions
The study of cell–cell and cell–matrix adhesion in nor-
mal skin has helped us to understand which genes are

Figure 4. Keratinocytes and fibroblasts in human skin reconstructs


produce a basement membrane. Skin reconstructs were stained
with monoclonal antibodies against Collagen IV (A) and against pan-
Laminin (B). In both stainings there is a clear demarcation of the
basement membrane as indicated by the positivity of Collagen IV
and Laminin, respectively. Human foetal foreskin was stained with
the mAb GoH3 against the a6 integrin subunit, showing the basal
layer of the epidermis (C). A counter-staining of the a6 integrin
subunit (green in D) and HMB45 (red in D) shows that the a6
D integrin subunit is not only expressed in the basal keratinocytes,
but also in the melanocytes (co-localization, yellow in D).

156 Pigment Cell Res. 18; 150–159


Adhesion, migration and communication in melanocytes and melanoma

down-regulated or functionally dysregulated in melan- tor expression in melanocytic tumour progression. Histopatholo-
oma. We have learned that E-cadherin is a critical gy 24, 249–256.
Degen, W.G., van Kempen, L.C., Gijzen, E.G., van Groningen, J.J.,
adhesion receptor between melanocytes and keratino-
van Kooyk, Y., Bloemers, H.P., and Swart, G.W. (1998). MEMD,
cytes and we have defined how growth of melano- a new cell adhesion molecule in metastasizing human melanoma
cytes can be stimulated by growth factors. We are cell lines, is identical to ALCAM (activated leukocyte cell adhe-
beginning to understand how melanocytes migrate sion molecule). Am. J. Pathol. 152, 805–813.
over the basement membrane to separate from each Ebrahimnejad, A., Streichert, T., Nollau, P., Horst, A.K., Wagener,
other. Least is known about the mechanisms of C., Bamberger, A.M., and Brummer, J. (2004). CEACAM1 enhan-
growth control that stop the melanocytes from prolifer- ces invasion and migration of melanocytic and melanoma cells.
Am. J. Pathol. 165, 1781–1787.
ating and migrating and that make them extend their
Felding-Habermann, B., Mueller, B.M., Romerdahl, C.A., and Cher-
dendrites far into the epidermis. We need to under- esh, D.A. (1992). Involvement of integrin alpha V gene expres-
stand the contributing factors that control growth, mor- sion in human melanoma tumorigenicity. J. Clin. Invest. 89,
phology and positioning of melanocytes. We expect 2018–2022.
each of the contributing regulatory mechanisms to be Filardo, E.J., Deming, S.L., and Cheresh, D.A. (1996). Regulation of
deregulated in melanoma cells. In turn, melanoma cells cell migration by the integrin beta subunit ectodomain. J. Cell
may be forced to revert to growth control by keratino- Sci. 109 (Pt 6), 1615–1622.
Fitzpatrick, T.B., and Breathnach, A.S. (1963). [The Epidermal Mel-
cytes, even with pharmacological approaches. Poten-
anin Unit System]. Dermatol. Wochenschr. 147, 481–489.
tially a static balance can be obtained if melanoma van der Flier, A., and Sonnenberg, A. (2001). Function and interac-
cells respond to the same signals as melanocytes in tions of integrins. Cell Tissue Res. 305, 285–298.
the epidermis, albeit in a distant organ where they Fogel, M., Mechtersheimer, S., Huszar, M., Smirnov, A., Abu-Dahi,
have metastasized. A., Tilgen, W., Reichrath, J., Georg, T., Altevogt, P., and Gutw-
ein, P. (2003). L1 adhesion molecule (CD 171) in development
and progression of human malignant melanoma. Cancer Lett.
Acknowledgements 189, 237–247.
Giavazzi, R., Chirivi, R.G., Garofalo, A., Rambaldi, A., Hemingway, I.,
We thank all members of Meenhard Herlyn’s lab for intellectual
Pigott, R., and Gearing, A.J. (1992). Soluble intercellular adhesion
support. We apologize to all those colleagues whose important
molecule 1 is released by human melanoma cells and is associated
work we could not cite due to space limitations.
with tumor growth in nude mice. Cancer Res. 52, 2628–2630.
Haass, N.K., and Herlyn, M. (in press) Normal human melanocyte
homeostasis as a paradigm for understanding melanoma.
References
J. Invest. Dermatol. Symp. Proc.
Aeed, P.A., Nakajima, M., and Welch, D.R. (1988). The role of poly- Haass, N.K., Smalley, K.S.M., and Herlyn, M. (2004). The role of
morphonuclear leukocytes (PMN) on the growth and metastatic altered cell–cell communication in melanoma progression.
potential of 13762NF mammary adenocarcinoma cells. Int. J. J. Mol. Histol. 35, 309–318.
Cancer 42, 748–759. Hanahan, D., and Weinberg, R.A. (2000). The hallmarks of cancer.
Albelda, S.M., Mette, S.A., Elder, D.E., Stewart, R., Damjanovich, Cell 100, 57–70.
L., Herlyn, M., and Buck, C.A. (1990). Integrin distribution in Hartstein, M.E., Grove, A.S., Jr, and Woog, J.J. (1997). The role of
malignant melanoma: association of the beta 3 subunit with the integrin family of adhesion molecules in the development of
tumor progression. Cancer Res. 50, 6757–6764. tumors metastatic to the orbit. Ophthal. Plast. Reconstr. Surg.
Bamberger, A.M., Riethdorf, L., Nollau, P., Naumann, M., Erdmann, 13, 227–238.
I., Gotze, J., Brummer, J., Schulte, H.M., Wagener, C., and Lon- Hegerfeldt, Y., Tusch, M., Brocker, E.B., and Friedl, P. (2002). Col-
ing, T. (1998). Dysregulated expression of CD66a (BGP, C-CAM), lective cell movement in primary melanoma explants: plasticity
an adhesion molecule of the CEA family, in endometrial cancer. of cell–cell interaction, beta1-integrin function, and migration
Am. J. Pathol. 152, 1401–1406. strategies. Cancer Res. 62, 2125–2130.
Becker, J.C., Termeer, C., Schmidt, R.E., and Brocker, E.B. (1993). Hixson, D.C., and McEntire, K.D. (1989). On rat transplantable and
Soluble intercellular adhesion molecule-1 inhibits MHC-restricted primary hepatocellular carcinomas. Cancer Res. 105, 6788–6794.
specific T cell/tumor interaction. J. Immunol. 151, 7224–7232. Holzmann, B., Gosslar, U., and Bittner, M. (1998). Alpha 4 integrins
Bissell, M.J., and Radisky, D. (2001). Putting tumours in context. and tumor metastasis. Curr. Top. Microbiol. Immunol. 231, 125–
Nat. Rev. Cancer 1, 46–54. 141.
Brooks, P.C., Stromblad, S., Sanders, L.C., von Schalscha, T.L., Ai- Hortsch, M. (1996). The L1 family of neural cell adhesion mole-
mes, R.T., Stetler-Stevenson, W.G., Quigley, J.P., and Cheresh, cules: old proteins performing new tricks. Neuron 17, 587–593.
D.A. (1996). Localization of matrix metalloproteinase MMP-2 to Hsu, M.Y., Shih, D.T., Meier, F.E., Van Belle, P., Hsu, J.Y., Elder,
the surface of invasive cells by interaction with integrin alpha v D.E., Buck, C.A., and Herlyn, M. (1998). Adenoviral gene transfer
beta 3. Cell 85, 683–693. of beta3 integrin subunit induces conversion from radial to verti-
Brummer, J., Ebrahimnejad, A., Flayeh, R., Schumacher, U., Lon- cal growth phase in primary human melanoma. Am. J. Pathol.
ing, T., Bamberger, A.M., and Wagener, C. (2001). cis Interaction 153, 1435–1442.
of the cell adhesion molecule CEACAM1 with integrin beta(3). Hsu, M.Y., Meier, F.E., Nesbit, M., Hsu, J.Y., Van Belle, P., Elder,
Am. J. Pathol. 159, 537–546. D.E., and Herlyn, M. (2000). E-cadherin expression in melanoma
Danen, E.H., Ten Berge, P.J., Van Muijen, G.N., Van’t Hof-Groo- cells restores keratinocyte-mediated growth control and down-
tenboer, A.E., Brocker, E.B., and Ruiter, D.J. (1994). Emergence regulates expression of invasion-related adhesion receptors. Am.
of alpha 5 beta1 fibronectin- and alpha v beta 3 vitronectin-recep- J. Pathol. 156, 1515–1525.

Pigment Cell Res. 18; 150–159 157


Haass et al.

Hynes, R.O. (1996). Targeted mutations in cell adhesion genes: Melchiori, A., Mortarini, R., Carlone, S., Marchisio, P.C., Anichini,
what have we learned from them? Dev. Biol. 180, 402–412. A., Noonan, D.M., and Albini, A. (1995). The alpha 3 beta 1 inte-
Hynes, R.O. (2002). Integrins: bidirectional, allosteric signaling grin is involved in melanoma cell migration and invasion. Exp.
machines. Cell 110, 673–687. Cell Res. 219, 233–242.
Jimbow, K., Quevedo, W.C. Jr, Fitzpatrick, T.B., and Szabo, G. Miele, M.E., Bennett, C.F., Miller, B.E., and Welch, D.R. (1994).
(1976). Some aspects if melanin biology: 1950–1975. J. Invest. Enhanced metastatic ability of TNF-alpha-treated malignant mel-
Dermatol. 67, 72–89. anoma cells is reduced by intercellular adhesion molecule-1
Johnson, J.P. (1999). Cell adhesion molecules in the development (ICAM-1, CD54) antisense oligonucleotides. Exp. Cell Res. 214,
and progression of malignant melanoma. Cancer Metastasis Rev. 231–241.
18, 345–357. Montgomery, A.M., Reisfeld, R.A., and Cheresh, D.A. (1994). Inte-
Johnson, J.P., Stade, B.G., Holzmann, B., Schwable, W., and Rieth- grin alpha v beta 3 rescues melanoma cells from apoptosis in
muller, G. (1989). De novo expression of intercellular-adhesion three-dimensional dermal collagen. Proc. Natl Acad. Sci. U. S. A.
molecule 1 in melanoma correlates with increased risk of meta- 91, 8856–8860.
stasis. Proc. Natl Acad. Sci. U. S. A. 86, 641–644. Montgomery, A.M., Becker, J.C., Siu, C.H., Lemmon, V.P., Cher-
Johnson, J.P., Rummel, M.M., Rothbacher, U., and Sers, C. (1996). esh, D.A., Pancook, J.D., Zhao, X., and Reisfeld, R.A. (1996).
MUC18: A cell adhesion molecule with a potential role in tumor Human neural cell adhesion molecule L1 and rat homologue
growth and tumor cell dissemination. Curr. Top. Microbiol. Immu- NILE are ligands for integrin alpha v beta 3. J. Cell Biol. 132,
nol. 213 (Pt 1), 95–105. 475–485.
Johnson, J.P., Bar-Eli, M., Jansen, B., and Markhof, E. (1997). Mel- Moretti, S., Martini, L., Berti, E., Pinzi, C., and Giannotti, B. (1993).
anoma progression-associated glycoprotein MUC18/MCAM Adhesion molecule profile and malignancy of melanocytic
mediates homotypic cell adhesion through interaction with a lesions. Melanoma Res. 3, 235–239.
heterophilic ligand. Int. J. Cancer 73, 769–774. Natali, P., Nicotra, M.R., Cavaliere, R., Bigotti, A., Romano, G.,
van Kempen, L.C., van den Oord, J.J., van Muijen, G.N., Weidle, Temponi, M., and Ferrone, S. (1990). Differential expression of
U.H., Bloemers, H.P., and Swart, G.W. (2000). Activated leuko- intercellular adhesion molecule 1 in primary and metastatic mel-
cyte cell adhesion molecule/CD166, a marker of tumor progres- anoma lesions. Cancer Res. 50, 1271–1278.
sion in primary malignant melanoma of the skin. Am. J. Pathol. Natali, P.G., Nicotra, M.R., Bartolazzi, A., Cavaliere, R., and Bigotti,
156, 769–774. A. (1993). Integrin expression in cutaneous malignant melanoma:
van Kempen, L.C., Meier, F., Egeblad, M., Kersten-Niessen, M.J., association of the alpha 3/beta 1 heterodimer with tumor pro-
Garbe, C., Weidle, U.H., Van Muijen, G.N., Herlyn, M., Bloemers, gression. Int. J. Cancer 54, 68–72.
H.P., and Swart, G.W. (2004). Truncation of activated leukocyte Natali, P.G., Hamby, C.V., Felding-Habermann, B., Liang, B., Nico-
cell adhesion molecule: a gateway to melanoma metastasis. tra, M.R., Di Filippo, F., Giannarelli, D., Temponi, M., and Ferro-
J. Invest. Dermatol. 122, 1293–1301. ne, S. (1997). Clinical significance of alpha(v)beta3 integrin and
Kleinerman, D.I., Troncoso, P., Lin, S.H., Pisters, L.L., Sherwood, intercellular adhesion molecule-1 expression in cutaneous malig-
E.R., Brooks, T., von Eschenbach, A.C., and Hsieh, J.T. (1995). nant melanoma lesions. Cancer Res. 57, 1554–1560.
Consistent expression of an epithelial cell adhesion molecule Nesbit, M., and Herlyn, M. (1994). Adhesion receptors in human
(C-CAM) during human prostate development and loss of expres- melanoma progression. Invasion Metastasis 14, 131–146.
sion in prostate cancer: implication as a tumor suppressor. Can- Nikkola, J., Vihinen, P., Vlaykova, T., Hahka-Kemppinen, M., Heino,
cer Res. 55, 1215–1220. J., and Pyrhonen, S. (2004). Integrin chains beta1 and alphav as
Kraus, A., Masat, L., and Johnson, J.P. (1997). Analysis of the prognostic factors in human metastatic melanoma. Melanoma
expression of intercellular adhesion molecule-1 and MUC18 on Res. 14, 29–37.
benign and malignant melanocytic lesions using monoclonal anti- Nollau, P., Scheller, H., Kona-Horstmann, M., Rohde, S., Hagenmul-
bodies directed against distinct epitopes and recognizing dena- ler, F., Wagener, C., and Neumaier, M. (1997). Expression of
tured, non-glycosylated antigen. Melanoma Res. 7(Suppl. 2), CD66a (human C-CAM) and other members of the carcinoembry-
S75–S81. onic antigen gene family of adhesion molecules in human colo-
Kuphal, S., Bauer, R., and Bosserhoff, A.K. (in press) Integrin signa- rectal adenomas. Cancer Res. 57, 2354–2357.
ling in malignant melanoma. Cancer Metastasis Rev. Nolte, C., Moos, M., and Schachner, M. (1999). Immunolocalization
Lehmann, J.M., Riethmuller, G., and Johnson, J.P. (1989). MUC18, a of the neural cell adhesion molecule L1 in epithelia of rodents.
marker of tumor progression in human melanoma, shows sequence Cell Tissue Res. 298, 261–273.
similarity to the neural cell adhesion molecules of the immuno- Palecek, S.P., Huttenlocher, A., Horwitz, A.F., and Lauffenburger,
globulin superfamily. Proc. Natl Acad. Sci. U. S. A. 86, 9891–9895. D.A. (1998). Physical and biochemical regulation of integrin
Li, G., and Herlyn, M. (2000). Dynamics of intercellular communica- release during rear detachment of migrating cells. J. Cell Sci.
tion during melanoma development. Mol. Med. Today 6, 163–169. 111 (Pt 7), 929–940.
Li, X., Chen, B., Blystone, S.D., McHugh, K.P., Ross, F.P., and Ra- Palecek, S.P., Horwitz, A.F., and Lauffenburger, D.A. (1999). Kinetic
mos, D.M. (1998). Differential expression of alphav integrins in model for integrin-mediated adhesion release during cell migra-
K1735 melanoma cells. Invasion Metastasis 18, 1–14. tion. Ann. Biomed. Eng. 27, 219–235.
Li, X., Regezi, J., Ross, F.P., Blystone, S., Ilic, D., Leong, S.P., and Park, C.C., Bissell, M.J., and Barcellos-Hoff, M.H. (2000). The influ-
Ramos, D.M. (2001). Integrin alphavbeta3 mediates K1735 mu- ence of the microenvironment on the malignant phenotype. Mol.
rine melanoma cell motility in vivo and in vitro. J. Cell Sci. 114, Med. Today 6, 324–329.
2665–2672. Patel, D.D., Wee, S.F., Whichard, L.P., Bowen, M.A., Pesando,
McGary, E.C., Lev, D.C., and Bar-Eli, M. (2002). Cellular adhesion J.M., Aruffo, A., and Haynes, B.F. (1995). Identification and char-
pathways and metastatic potential of human melanoma. Cancer acterization of a 100-kD ligand for CD6 on human thymic epithe-
Biol. Ther. 1, 459–465. lial cells. J. Exp. Med. 181, 1563–1568.
Meier, F., Nesbit, M., Hsu, M.Y. et al. (2000). Human melanoma Petitclerc, E., Stromblad, S., von Schalscha, T.L., Mitjans, F., Piul-
progression in skin reconstructs: biological significance of bFGF. ats, J., Montgomery, A.M., Cheresh, D.A., and Brooks, P.C.
Am. J. Pathol. 156, 193–200. (1999). Integrin alpha(v)beta3 promotes M21 melanoma growth

158 Pigment Cell Res. 18; 150–159


Adhesion, migration and communication in melanocytes and melanoma

in human skin by regulating tumor cell survival. Cancer Res. 59, Sienel, W., Dango, S., Woelfle, U., Morresi-Hauf, A., Wagener, C.,
2724–2730. Brummer, J., Mutschler, W., Passlick, B., and Pantel, K. (2003).
Raucher, D., and Sheetz, M.P. (2000). Cell spreading and lamellipo- Promotes progression of non-small cell lung cancer. Clin. Cancer
dial extension rate is regulated by membrane tension. J. Cell Res. 1, 2260–2266.
Biol. 148, 127–136. van de Stolpe, A., and van der Saag, P.T. (1996). Intercellular adhe-
Riethdorf, L., Lisboa, B.W., Henkel, U., Naumann, M., Wagener, sion molecule-1. J. Mol. Med. 74, 13–33.
C., and Loning, T. (1997). Differential expression of CD66a Swart, G.W., Lunter, P.C., van Kilsdonk, J.W.J., and van Kempen,
(BGP), a cell adhesion molecule of the carcinoembryonic anti- L.C.L.T. (in press) Activated Leukocyte Cell Adhesion Molecule
gen family, in benign, premalignant, and malignant lesions of (ALCAM/CD166): Signaling at the Divide of Melanoma Cell Clus-
the human mammary gland. J. Histochem. Cytochem. 45, 957– tering and Cell Migration? Cancer Metastasis Rev.
963. Thies, A., Moll, I., Berger, J., Wagener, C., Brummer, J., Schulze,
Satyamoorthy, K., Muyrers, J., Meier, F., Patel, D., and Herlyn, M. H.J., Brunner, G., and Schumacher, U. (2002a). CEACAM1
(2001). Mel-CAM-specific genetic suppressor elements inhibit expression in cutaneous malignant melanoma predicts the devel-
melanoma growth and invasion through loss of gap junctional opment of metastatic disease. J. Clin. Oncol. 20, 2530–2536.
communication. Oncogene 20, 4676–4684. Thies, A., Schachner, M., Moll, I., Berger, J., Schulze, H.J., Brun-
Schadendorf, D., Gawlik, C., Haney, U., Ostmeier, H., Suter, L., ner, G., and Schumacher, U. (2002b). Overexpression of the cell
and Czarnetzki, B.M. (1993). Tumour progression and metastatic adhesion molecule L1 is associated with metastasis in cutaneous
behaviour in vivo correlates with integrin expression on melano- malignant melanoma. Eur. J. Cancer 38, 1708–1716.
cytic tumours. J. Pathol. 170, 429–434. Van Belle, P.A., Elenitsas, R., Satyamoorthy, K. et al. (1999). Pro-
Schadendorf, D., Heidel, J., Gawlik, C., Suter, L., and Czarnetzki, gression-related expression of beta3 integrin in melanomas and
B.M. (1995). Association with clinical outcome of expression of nevi. Hum. Pathol. 30, 562–567.
VLA-4 in primary cutaneous malignant melanoma as well as P-se- Voura, E.B., Ramjeesingh, R.A., Montgomery, A.M., and Siu, C.H.
lectin and E-selectin on intratumoral vessels. J. Natl Cancer Inst. (2001). Involvement of integrin alpha(v)beta(3) and cell adhesion
87, 366–371. molecule L1 in transendothelial migration of melanoma cells.
Schlagbauer-Wadl, H., Jansen, B., Muller, M., Polterauer, P., Wolff, Mol. Biol. Cell 12, 2699–2710.
K., Eichler, H.G., Pehamberger, H., Konak, E., and Johnson, J.P. Xie, S., Luca, M., Huang, S., Gutman, M., Reich, R., Johnson, J.P.,
(1999). Influence of MUC18/MCAM/CD146 expression on human and Bar-Eli, M. (1997). Expression of MCAM/MUC18 by human
melanoma growth and metastasis in SCID mice. Int. J. Cancer melanoma cells leads to increased tumor growth and metastasis.
81, 951–955. Cancer Res. 57, 2295–2303.
Schumacher, D., and Schaumburg-Lever, G. (1999). Ultrastructural Yoshinaga, I.G., Vink, J., Dekker, S.K., Mihm, M.C. Jr, and Byers,
localization of alpha-3 integrin subunit in malignant melanoma H.R. (1993). Role of alpha 3 beta 1 and alpha 2 beta 1 integrins
and adjacent epidermis. J. Cutan. Pathol. 26, 321–326. in melanoma cell migration. Melanoma Res. 3, 435–441.
Shih, I.M., Elder, D.E., Speicher, D., Johnson, J.P., and Herlyn, M. Zhu, N., Eves, P.C., Katerinaki, E., Szabo, M., Morandini, R., Gha-
(1994). Isolation and functional characterization of the A32 melan- nem, G., Lorigan, P., MacNeil, S., and Haycock, J.W. (2002).
oma-associated antigen. Cancer Res. 54, 2514–2520. Melanoma cell attachment, invasion, and integrin expression is
Shih, I.M., Speicher, D., Hsu, M.Y., Levine, E., and Herlyn, M. upregulated by tumor necrosis factor alpha and suppressed by
(1997a). Melanoma cell–cell interactions are mediated through alpha melanocyte stimulating hormone. J. Invest. Dermatol. 119,
heterophilic Mel-CAM/ligand adhesion. Cancer Res. 57, 3835– 1165–1171.
3840. Ziober, B.L., Chen, Y.Q., Ramos, D.M., Waleh, N., and Kramer,
Shih, L.M., Hsu, M.Y., Palazzo, J.P., and Herlyn, M. (1997b). The R.H. (1999). Expression of the alpha7beta1 laminin receptor sup-
cell–cell adhesion receptor Mel-CAM acts as a tumor suppressor presses melanoma growth and metastatic potential. Cell Growth
in breast carcinoma. Am. J. Pathol. 151, 745–751. Differ. 10, 479–490.

Pigment Cell Res. 18; 150–159 159

Das könnte Ihnen auch gefallen