ArticlePDF AvailableLiterature Review

Semaphorins in cancer

Authors:

Abstract and Figures

The semaphorins are the products of a large family of genes currently containing more than 30 members. These genes are divided into eight classes of which classes 1, 2 and 8 contain invertebrate and viral semaphorins, while classes 3-7 contain the vertebrate semaphorins. The semaphorins have been implicated in diverse developmental processes such as axon guidance during nervous system development and regulation of cell migration. Plexin receptors function as binding and signal transducing receptors for all semaphorins except for the class-3 semaphorins which bind to neuropilins which subsequently activate signaling through associated plexins. The class-3 semaphorins semaphorin-3B (s3b) and semaphorin-3F (s3f) function additionally as potent inhibitors of tumor development in small cell lung carcinoma. Recent evidence indicates that these semaphorins modulate the adhesive and migratory properties of responsive malignant cells. S3f as well as semaphorin-3A (s3a) were also found to function as inhibitors of angiogenesis, and it was shown that the anti-angiogenic properties of s3f contribute significantly to its anti-tumorigenic properties. In contrast with these inhibitory semaphorins, there is some evidence indicating that semaphorins such as semaphorin-3C (s3c), semaphorin-3E (s3e), semaphorin-4D (s4d), semaphorin-5C (s5c) semaphorin-6A (s6a) and semaphorin-6b (s6b) may contribute to tumorigenesis or to tumor progression. In this review we discuss the semaphorins, their receptors and their signal transduction mechanisms, and evidence linking semaphorins to the control of tumorigenesis and tumor progression.
Content may be subject to copyright.
[Frontiers in Bioscience 10, 751-760, January 1, 2005]
751
SEMAPHORINS IN CANCER
Gera Neufeld, Niva Shraga-Heled, Tali Lange, Noga Guttmann-Raviv, Yael Herzog, And Ofra Kessler
Cancer and Vascular Biology Research Center, Rappaport Research Institute in the Medical Sciences, The Bruce Rappaport
Faculty of Medicine, Technion, Israel Institute of Technology, 1 Efron St., P.O. Box 9679, Haifa, 31096, Israel
TABLE OF CONTENTS
1. Abstract
2. Introduction
3. The semaphorin gene family
4. Semaphorin receptors
4.1. Plexins
4.2. Neuropilins
4.3. Additional semaphorin receptors
5. Semaphorin induced signal transduction
6. Semaphorins as modulators of tumor progression
6.1. Class-3 semaphorins in tumorigenesis and tumor progression
6.2. Class 4 semaphorins and their role in tumor progression
6.3. Class 5 and 6 semaphorins and their role in tumor progression
7. Conclusions
8. Acknowledgements
9. References
1. ABSTRACT
The semaphorins are the products of a large
family of genes currently containing more than 30
members. These genes are divided into eight classes of
which classes 1, 2 and 8 contain invertebrate and viral
semaphorins, while classes 3–7 contain the vertebrate
semaphorins. The semaphorins have been implicated in
diverse developmental processes such as axon guidance
during nervous system development and regulation of cell
migration. Plexin receptors function as binding and signal
transducing receptors for all semaphorins except for the
class-3 semaphorins which bind to neuropilins which
subsequently activate signaling through associated plexins.
The class-3 semaphorins semaphorin-3B (s3b) and
semaphorin-3F (s3f) function additionally as potent
inhibitors of tumor development in small cell lung
carcinoma. Recent evidence indicates that these
semaphorins modulate the adhesive and migratory
properties of responsive malignant cells. S3f as well as
semaphorin-3A (s3a) were also found to function as
inhibitors of angiogenesis, and it was shown that the anti-
angiogenic properties of s3f contribute significantly to its
anti-tumorigenic properties. In contrast with these
inhibitory semaphorins, there is some evidence indicating
that semaphorins such as semaphorin-3C (s3c),
semaphorin-3E (s3e), semaphorin-4D (s4d), semaphorin-
5C (s5c) semaphorin-6A (s6a) and semaphorin-6b (s6b)
may contribute to tumorigenesis or to tumor progression. In
this review we discuss the semaphorins, their receptors and
their signal transduction mechanisms, and evidence linking
semaphorins to the control of tumorigenesis and tumor
progression.
2. INTRODUCTION
The semaphorins and their receptors, the
neuropilins and the plexins, were originally characterized
as constituents of the complex regulatory system
responsible for the guidance of growing axons to their
targets during the development of the central nervous
system. However, a growing body of evidence indicates
that the semaphorins and their receptors are implicated in
the regulation of additional developmental processes such
as the migration of neural crest cells and heart development
to name but a few examples (1,2). In addition, there is a
growing body of evidence suggesting that the semaphorins
and their receptors may play a regulatory role in
tumorigenesis and in the process of tumor formation. This
evidence is reviewed in the following sections.
3. THE SEMAPHORIN GENE FAMILY
The semaphorin family consists of more than 30 genes
divided into 8 classes, of which the first two classes are
derived from invertebrates, classes 3-7 are the products of
vertebrate semaphorins, and the 8
th
contains viral
semaphorins (3) (Figure 1). In the literature the
semaphorins are often referred to by an array of confusing
designations. This situation was clarified several years ago
by the adoption of a unified nomenclature for the
semaphorins (3). The semaphorins are characterized by the
presence of a conserved sema domain which is ~500
amino-acids long and is usually located at their N-terminal.
The sema domain is essential for semaphorin signaling and
determines receptor binding specificity (4). The sema
domains of two different semaphorins were recently
characterized at an atomic resolution revealing beta
propeller topology (5-6). In addition, semaphorins contain
additional structural motifs such as immunoglobulin like
domains (classes 2-4 and 7), thrombospondin repeats (class
5) and basic domains (class 3). The semaphorins are
produced either as secreted proteins as in the case of the
class-3 semaphorins or as membrane anchored or trans-
Semaphorins in Cancer
752
Figure 1. The main classes of the semaphorins and their structure. The heavy black line represents the cell membrane.
membrane proteins that can be further processed into
soluble proteins. The active forms of the class-3 and class 4
semaphorins s3a and s4a are homo-dimeric (7,8), and it is
therefore reasonable to assume that the active forms of the
other class-3 and class-4 semaphorins also form
homodimers. Semaphorins have been mainly characterized
as axon guidance factors that function primarily in the
developing nervous system (9). In recent years it was
realized that semaphorins play a role in many
developmental processes outside of the nervous system, in
particular as regulators of cell migration (10), immune
responses (11) and organogenesis (2). It is therefore not
surprising that some of the semaphorins have been recently
found to play important roles in tumor development and
progression.
4. SEMAPHORIN RECEPTORS
4.1 Plexins
The best characterized semaphorin receptors are
encoded by the genes belonging to the plexin family. The
plexins are segregated into four sub-families containing 9
vertebrate plexins. The plexins are transmembrane proteins
containing a cytoplasmic SP domain that includes putative
tyrosine phosphorylation sites but no known enzymatic
activity. Their extracellular domain is distinguished by the
presence of a sema domain, by the presence of a Met
related sequence (MRS) domain and by glycine-proline (G-
P) rich motifs which the plexins share with the tyrosine-
kinase receptors belonging to the Met family of receptors
(Figure 2)(12). Some vertebrate semaphorins belonging to
the 4-7 classes have been shown to bind directly to plexins
and to activate plexin mediated signal transduction as a
result. For example, semaphorin-4D (s4d) binds to plexin-
B1 (13) and semaphorin-6D (s6d) binds to plexin-A1 (14).
In addition, semaphorin-7A as well as several viral
semaphorins were found to bind to plexin-C1 which is
itself a virally encoded plexin like receptor (Table 1) (15).
Plexin-B1 can form a complex with the
hepatocyte growth factor receptor MET. Following s4d
binding to plexin-B1, the MET tyrosine kinase is activated
resulting in the phosphorylation of both receptors (13).
Likewise, s6d potentiates the effects of VEGF as a result of
complex formation between plexin-A1 and VEGF receptor-
2 (Table 1) (14). Plexins were found to associate with
additional types of cell surface receptors. Plexin-A1
associates with the Off-Track (OTK) receptor (16) and the
plexin-A1 ligand s6d induces OTK mediated signaling(14).
4.2. Neuropilins
Class-3 semaphorins differ from other types of
semaphorins by their inability to bind directly to plexins.
The six members of this semaphorin class bind instead to
neuropilin-1 (np1) or to the neuropilin-2 (np2) homo-
dimeric receptors or to heterodimers of these two
neuropilins (17-20). Structurally, these two receptors are
related although the overall homology is only 44% (19).
Semaphorins in Cancer
753
Table 1. The receptors of some selected semaphorins.
Semaphorin
High affinity binding
receptor
Low affinity binding
receptor
Functional receptor References
S3a Np1 Np1/np1/plexin-A(1-2) 24
S3c Np1,Np2 (Np1/Np2)(?)/Plexin-A(?), plexin-D1 20,27, 24
S3f Np2 Np1 Np2/plexin-A(1-2) 24
S4a TIM-2 38
S4b Plexin-B1 Plexin-B1/MET CD-72 13,37
S5A Plexin-B3 Plexin-B3/MET 105
S6d Plexin-A1 Plexin-A1/VEGFR-2 14
Figure 2. The general structure of the neuropilins and their plexin co-receptors.
Their general structure is shown in Figure 2. Although it
was demonstrated that np1 is required for s3a induced
collapse of axonal growth cones, deletion of the
cytoplasmic domain of np1 did not inhibit s3a activity,
suggesting the existence of an independent signal
transducing moiety (21). It was subsequently found that the
four type-A plexins can form complexes with neuropilins in
which the plexins serve as the signal transducing partners
(15,22-24). Plexins belonging to the other three plexin
subfamilies may also be able to form complexes with
neuropilins, as demonstrated in the case of plexin-B1 and
np1 (15,25), and recent data indicates that plexin-D1 can
also transduce signals of class-3 semaphorins (Table-1)
(26,27).
The neuropilins form complexes with additional
cell surface molecules. Np1 was reported to form
complexes with L1, an adhesion molecule which seems to
be required under certain circumstances for s3a signal
transduction (28,29). Both np1 and np2 were also found to
function as receptors for specific heparin binding splice
forms of the angiogenic factor vascular endothelial growth
factor (VEGF) (30,31). Both neuropilins were observed to
form complexes with the VEGF tyrosine-kinase receptor
VEGFR-1 (32,33). The interaction with VEGFR-1 is
apparently required for np1 mediated repulsion of
migrating neuronal progenitor cells by s3a (34). Likewise,
it was reported that np1 forms complexes with VEGFR-2
(35,36). The formation of such complexes probably
accounts for the potentiation of VEGF induced cell
migration mediated via activation of VEGFR-2 (30).
4.3. Additional semaphorin receptors
S4d and semaphorin-4A (s4a) were recently
found to play important roles in immune responses (11).
CD-72, a membrane bound calcium dependent protein
belonging to the lectin super-family was found to function
as a s4d receptor in the immune system (37). Likewise,
Semaphorins in Cancer
754
TIM-2, a member of the T-cell immunoglobulin and mucin
domain proteins was found to function as a semaphorin-4A
(s4a) receptor in T cells (38).
5. SEMAPHORIN INDUCED SIGNAL
TRANSDUCTION
Activation of plexins by semaphorins, either
directly or indirectly via neuropilins, leads to diverse
biological responses. One of the best studied responses is
the s3a induced repulsion of axonal growth cones. The
repulsion is apparently triggered by local changes in cell
adhesion and actin cytoskeleton organization. Activation of
the plexins, results in phosphorylation of tyrosine residues
in the cytoplasmic domain of the plexins and subsequent
activation of signal transduction. Plexins can be
phosphorylated as a result of complex formation with
tyrosine-kinase receptors as recently demonstrated in the
case of plexin-B1 which is phosphorylated by the
hepatocyte growth factor receptor MET in response to s4d
(13). Complex formation with tyrosine-kinase receptors
also allows s4d to phosphorylate MET and to induce
invasiveness by activation of this receptor. Similarly,
complex formation between the s6d receptor plexin-A1 and
the VEGF receptor VEGFR-2 enables s6d induced
autophophorylation of VEGFR-2 and induction of VEGFR-
2 mediated signaling (14). Phosphorylation of plexins can
also be the result of semaphorin induced recruitment of
cytosolic tyrosine kinases. The binding of s3a to np1
induces the association of the tyrosine-kinase Fes/Fps with
plexin-A1 leading to plexin-A1 phosphorylation. In growth
cones of s3a responsive nerve cells Fes/Fps forms a
complex with the brain specific collapsin response
mediator protein-2 (CRMP-2) and with CRMP associated
molecule (CRAM). These two proteins are required for s3a
signaling to the actin cytoskeleton in responsive nerve cells
and are also phosphorylated by Fes/Fps in response to s3a
although their exact role is still unclear (39). Another
cytosolic tyrosine-kinase that was found to associate with
the intra-cellular part of plexin-A1 as well as plexin-A2 is
fyn. Fyn phosphorylates plexin-A2 in response to s3a and
attracts the cdk5 kinase, which is also phosphorylated by
Fyn. Activation of cdk5 by fyn was also found to be
essential for s3a mediated growth cone repulsion (40).
Another class of intracellular signal transducing
proteins that interact with plexins was found in studies
which utilized the genetic tools available in the drosophila
fruit fly system. It was found that plexin-A, a drosophila
plexin receptor, binds the MICAL-1 protein in response to
the drosophila semaphorin s1a. MICALs interact with
intermediate filaments and actin on the one hand, and are
putative flavoprotein monooxygenases on the other hand.
The oxidative activity of the MICALs may be important for
semaphorin signaling since monooxygenase inhibitors
inhibit MICAL mediated trunsduction of s3a signals
(41,42).
To affect the organization of the actin
cytoskeleton, semaphorin receptors modulate the activity of
the small GTPases RhoA, Rnd1, Rac1 and CDC42.
Activation of Rac and CDC42 usually triggers the
formation of lamellipodia and filopodia, respectively. In
contrast, activation of the Rho family members Rho1 and
Rnd1 leads to the formation of stress fibers. In neuronal
cells the responses are a bit different. RhoA activation
induces neurite retraction and Rac1 activation induces
neurite extension (43). Activation of Plexin-B1 by s4d
inhibits Rac1 signaling while simultaneously activating
Rho thus resulting in the collapse of growing growth cones
(44,45). These responses may well depend on the relative
cytoplasmic concentrations of the GTPases, as recently
demonstrated in the case of the C. elegans Rac1
homologue. In that case it was shown that changes in Rac1
concentration in neuronal cells can turn repulsion into
attraction and vice-versa (46). In contrast to plexin-B1,
plexin-A1 was not observed to bind Rac1 directly even
though Rac1 is known to play a crucial regulatory role in
the induction of s3a induced growth cone collapse (47). Out
of the Rho family GTPases tested only RhoD and Rnd1
were found to bind to plexin-A1. Interestingly, Rnd1
binding to plexin-A1 leads to a collapse of the actin
cytoskeleton even in the absence of s3a. RhoD on the other
hand antagonizes the effects of Rnd1 even though both
GTPases belong to the Rho family of GTPases (48).
Various guanine nucleotide exchange factors (GEFs) and
GTPase activating proteins (GAPs), which function as
regulators of GTPases, also modulate the activity of plexin
binding GTPases (49,50). The GTPases in turn regulate the
activity of downstream effectors such as the LIM kinase
which in turn regulates the phosphorylation state of cofilin,
an actin binding/cleaving protein that is required for s3a
induced growth cone collapse (51).
6. SEMAPHORINS AS MODULATORS OF TUMOR
PROGRESSION
The biological properties of the semaphorins
have been studied extensively with regard to their effects
on axon guidance and nervous system development.
However, it is now clear that their effects are not limited to
the nervous system. Many cell types in addition to nerve
cells express neuropilins and plexins indicating that
semaphorins may be able to affect their behavior. Cell
types expressing neuropilins include neural crest derived
cells (1), immune cells such as dendritic cells and T-cells
(52), neuro-endocrine cells (53,54), Mesothelial cells (55),
endothelial cells (30,56), and bone marrow stromal cells
(57) to name but a few. Plexins are also widely expressed
and are found in endothelial cells (58,59), in bone marrow
cells (60), in epithelial cells of the lung (61) as well as in
many additional types of non-neuronal cells. These
observations suggest that semaphorins may also affect the
behavior of cancer cells derived from cell types that
express semaphorin receptors. Indeed, several semaphorins
have now been found to affect tumor progression and
tumorigenesis.
6.1. Class-3 semaphorins in tumorigenesis and tumor
progression
The discovery of splice form specific vascular
endothelial growth factor receptors on endothelial cells (62)
and their subsequent identification as products of the np1
and np2 genes (30,63), has lead to the identification of both
Semaphorins in Cancer
755
neuropilin receptors as essential regulators of
vasculogenesis (64). These findings suggested that
semaphorins may also function as regulators of
angiogenesis. Since tumor expansion and tumor spread
depend upon tumor angiogenesis (65), it follows that some
class-3 semaphorins may also be able to modulate tumor
progression by modulating tumor angiogenesis. VEGF165
and s3a compete for an overlapping binding site located in
the extra-cellular domain of np1. S3a was indeed able to
inhibit VEGF165 induced migration of endothelial cells as
well as VEGF165 induced in-vitro angiogenesis,
presumably by inhibiting the effects of VEGF (66).
Subsequent experiments have shown that s3a can also
inhibit developmental angiogenesis in chick embryos, a
finding which suggests that s3a should also be able to
inhibit tumor angiogenesis (67,68). However, an effect of
s3a on tumor angiogenesis has not yet been demonstrated.
The effects of s3a may not be limited to the modulation of
tumor angiogenesis. Neuropilins are expressed by tumor cells
derived from prostate cancer (69,70), colon cancer (71),
Melanoma (72), pancreatic carcinoma (73) and breast cancer
(74,75) to name a few examples. MDA-MB-231 breast cancer
cells express large amounts of np1 and plexin-A1 and their
migration and spreading are inhibited by s3a (74,75).
VEGF165 competed with s3a for binding to np1 on these cells
and abrogated the inhibitory effects of s3a, indicating that
semaphorins such as s3a have the potential to function as anti-
metastatic agents, and that by this mechanism VEGF165 may
exert effects that are not dependent upon the presence of the
tyrosine-kinase coupled VEGF receptors (75). S3a was also
found to function as an inducer of neuronal apoptosis (76), a
process that seems to be mediated by MAP kinases (77). This
property may also represent another potential mechanism by
which s3a could affect tumor development and progression.
The genes encoding s3b and s3f have been
originally identified as genes that are lost in small cell lung
carcinoma, and have been therefore characterized as
potential tumor suppressor genes (78-80). It was shown that
the loss of s3b function resulting in tumor development can
also be due to epigenetic reasons such as de-novo promoter
methylation (81). Expression of s3f in small cell lung
carcinoma cells inhibited colony formation in soft agar and
expression of s3f in MCF-7 breast cancer cells inhibited
their adhesion and spreading (82). Over-expression of
either s3b or s3f in small cell lung carcinoma cells inhibits
their in-vivo tumor forming ability (83-85). The
localization of s3f in expressing tumor cells may be
important, as it was shown that in lung tumors cytoplasmic
localization of s3f is correlated with high VEGF expression
levels and increased tumorigenicity, suggesting that failure
to secrete s3f may promote tumor progression (86). In the
case of S3b, it was shown that s3b expression is induced by
the cell cycle gate-keeper gene p53. Thus, external signals
that induce p53 expression and inhibit the entry of cells into
the cell cycle such as UV irradiation also up-regulate s3b,
indicating that s3b may play a role in regulating the
entrance of cells into the cell cycle as part of the p53
associated machinery (87).
S3f functions as an agonist of the np2 receptor
(19,88) suggesting that s3f may also function as an
inhibitor of angiogenesis. In contrast with s3a, which
inhibits VEGF
165
binding to np1, s3f binding to np2 was
not inhibited by VEGF
165
(63). Nevertheless, s3f inhibited
both VEGF and bFGF induced endothelial cell proliferation
and angiogenesis, as well as the development of tumors
from tumor cells whose proliferation in cell culture was not
inhibited by s3f (58). These experiments suggest that s3f
interferes with the activities of these growth factors using a
mechanism that does not require competition with pro-
angiogenic factors for binding to shared receptors. Rather, the
experiments suggest that s3f inhibits VEGF and bFGF function
downstream of the receptor level, possibly by generating an
inhibitory intracellular signal that counteracts the signals
conveyed by these growth factors in endothelial cells.
S3a, s3b and s3f, have either been shown to
inhibit tumor development, or are expected to inhibit tumor
development based upon experiments suggesting that they
possess anti-angiogenic or anti-tumorigenic properties. The
data regarding other class-3 semaphorins is scant, but
indicates that some of these other class-3 semaphorins may
promote tumor progression rather than inhibit tumor
progression. Thus, S3c expression was found to be up-
regulated in cis-diamminedichloroplatinum (II) (CDDP)-
resistant ovarian cancer TYKnuR cells, in metastatic
human lung adenocarcinoma cells, and in malignant
melanoma cells indicating that up-regulation of s3c
expression may be linked to tumor progression (89-91).
Likewise, high level expression of s3e had been observed
in several metastatic cell lines originating from mouse
mammary adenocarcinoma tumors, indicating that high s3e
expression levels are linked to tumor progression (92).
6.2. Class 4 semaphorins and their role in tumor
progression
The membrane anchored class 4 semaphorin s4d
has recently become a focus of intensive research as a
result of its recently discovered role in immune recognition
(93), and as a result of its newly discovered role as a
regulator of tumor cell invasiveness. HGF/SF (hepatocyte
growth factor/scatter factor) induces scattering, invasion,
proliferation and branching morphogenesis, and plays a
role as a regulator of invasiveness and tumor spread in
many types of tumors (94,95). The HGF/SF tyrosine-kinase
receptor MET as well as MET like receptors such as the
RON receptor for macrophage stimulating protein contain a
conserved sema domain, and were recently found to
associate and form complexes with several types of
receptors belonging to the plexin-B subfamily. When a plexin-
B1/MET complex is challenged with s4d, the tyrosine-kinase
activity of MET is activated, just as if it were activated by
HGF/SF, leading to the phosphorylation of both MET and
plexin-B1. This in turn results in the stimulation of invasive
growth (13,96). These observations implicate s4d as a potential
inducer of tumor invasiveness and tumor progression, although
this has yet to be demonstrated experimentally. HGF/SF also
functions as an inducer of angiogenesis (97,98), indicating that
s4d may also be able to induce angiogenesis via a similar
mechanism.
Recently, the tyrosine-kinase receptor ErbB-2
was also found to form complexes with plexin-B1 and to be
Semaphorins in Cancer
756
phosphorylated in response to s4d (99). Mutations in ErbB-
2 are known to play a role in the induction of tumorigenesis
in breast cancer as well as in other types of cancer (100).
These observations suggest that activating mutations in
plexin-B1 may perhaps be able to induce activation of
ErbB2 and MET in the absence of any ligand and thereby
contribute to tumorigenesis. These possibilities will have to
be tested in the future.
6.3. Class 5 and 6 semaphorins and their role in tumor
progression
Class-5 semaphorins are anchored to cell
membranes and are characterized by seven type 1
thrombospondin repeats functionally important for
tumorigenicity and metastasis (Figure 1) (101). Deletion of
the drosophila lethal giant larvae gene leads to the
generation of highly invasive and widely metastatic tumors
on transplantation into adult flies. Random A p-element
insertion screen was used to identify genes that modulate
tumor progression and tumorigenicity. One of the genes
identified in this screen was the drosophila homologue of
s5c. S5c inactivation was found to inhibit tumor formation
in lethal giant larvae mutants, suggesting that it is required
for tumorigenesis. Further experiments indicate that s5c
probably associates, via its thrombospondin repeats, with
the TGF-β like ligand DPP somehow modulating DPP
induced signal transduction (102).
Vertebrates possess three s5c homologues (s5a,
s5b and s5d) (103,104), indicating that these homologues
may play a role in the development and progression of
human tumors too, although this hypothesis still requires
experimental proof. Recently, this assumption was
strengthened by experiments that have shown that s5a
activates plexin-B3, a receptor that also forms complexes
with the MET tyrosine-kinase receptor. As a result MET
undergoes autophosphorylation and phosphorylates plexin-
B3 (105). This is therefore one more example of a
semaphorin that interacts directly with a B type plexin and
activates as a result the MET tyrosine-kinase receptor
which had been previously shown to play a role in tumor
progression.
S6b belongs to the class-6 membrane anchored
semaphorins. S6b may also be linked to tumor progression
as it was found to be expressed in two different human
glioblastoma cell lines, and its levels were down-regulated
by trans-retinoic acid, an anti-tumorigenic, differentiation
promoting agent (106). In addition, recent evidence has
implicated s6d as a possible regulator of angiogenesis. It is
not known whether tumor cells express s6d, but the fact
that its receptor, plexin-A1 was found to form complexes
with the VEGF receptor VEGFR-2 and to affect
angiogenesis suggests that s6d may have the potential to
affect tumor angiogenesis (14).
7. CONCLUSIONS
During the complex process in which normal
cells turn into malignant cancer cells the tumorigenic cells
acquire an invasive character, become less adhesive and
induce angiogenesis as well as lymphangiogenesis. Normal
cells are also capable of invasive migration and under
appropriate conditions will induce angiogenesis and
lymphangiogenesis. However, in normal tissues there are
multiple regulatory mechanisms that limit and channel
these activities. It is the breakdown of the regulatory
mechanisms that is a hallmark of the tumor tissue formed
by malignant cells. The semaphorins are now emerging as
key regulators of these key processes of tumor progression.
The scanty data that we already have indicates that the
various semaphorins are critical regulators of these
processes, and that the evasion or subversion of semaphorin
mediated regulation of these processes can play a key role
in tumor progression. We have so far only touched the tip
of the iceberg as far as our understanding of the role of the
semaphorins in tumor progression is concerned. The next
few years will likely result in a flood of information, out of
which it is hoped that new and efficient cancer therapies
will emerge.
8. ACKNOWLEDGEMENTS
This work was supported by grants from the
Israel Science Foundation (ISF), By a grant from the
Ministry of Science, Joint program with Germany
(Deutsches Krebsforschungszentrum), By a grant from the
Germany-Israel science Foundation (GIF) and by the
Rappaport Family Institute for Research in the Medical
Sciences of the Faculty of Medicine at the Technion, Israel
Institute of Technology (to G. Neufeld). The costs of
publication of this article were defrayed in part by the
payment of page
9. REFERENCES
1. Eickholt, B. J., S.L.Mackenzie, A.Graham, F.S.Walsh,
and P.Doherty: Evidence for collapsin-1 functioning in the
control of neural crest migration in both trunk and
hindbrain regions. Development 126(10), 2181-2189
(1999)
2. Feiner, L., A.L.Webber, C.B.Brown, M.M.Lu, L.Jia,
P.Feinstein, P.Mombaerts, J.A.Epstein, and J.A.Raper:
Targeted disruption of semaphorin 3C leads to persistent
truncus arteriosus and aortic arch interruption.
Development 128(16), 3061-3070 (2001)
3. Goodman, C. S., A.L.Kolodkin, Y.Luo, A.W.Pueschel,
and J.A.Raper: Unified nomenclature for the semaphorins
collapsins. Cell 97(5), 551-552 (1999)
4. Feiner, L., A.M.Koppel, H.Kobayashi, and J.A.Raper:
Secreted chick semaphorins bind recombinant neuropilin
with similar affinities but bind different subsets of neurons
in situ. Neuron 19(3), 539-545 (1997)
5. Love, C. A., K.Harlos, N.Mavaddat, S.J.Davis,
D.I.Stuart, E.Y.Jones, and R.M.Esnouf: The ligand-binding
face of the semaphorins revealed by the high-resolution
crystal structure of SEMA4D. Nat. Struct. Biol. 10(10),
843-848 (2003)
6. Antipenko, A., J.P.Himanen, K.van Leyen, V.Nardi-Dei,
J.Lesniak, W.A.Barton, K.R.Rajashankar, M.Lu,
C.Hoemme, A.W.Puschel, and D.B.Nikolov: Structure of
the Semaphorin-3A Receptor Binding Module. Neuron
39(4), 589-598 (2003)
Semaphorins in Cancer
757
7. Koppel, A. M. and J.A.Raper: Collapsin-1 covalently
dimerizes, and dimerization is necessary for collapsing
activity. J. Biol. Chem. 273(25), 15708-15713 (1998)
8. Klostermann, A., M.Lohrum, R.H.Adams, and
A.W.Puschel: The chemorepulsive activity of the axonal
guidance signal semaphorin D requires dimerization. J.
Biol. Chem. 273(13), 7326-7331 (1998)
9. De Wit, J. and J.Verhaagen: Role of semaphorins in the
adult nervous system. Prog. Neurobiol. 71(2-3), 249-267
(2003)
10. Tamagnone, L. and P.M.Comoglio: To move or not to
move? EMBO Rep. 5(4), 356-361 (2004)
11. Kumanogoh, A. and H.Kikutani: Roles of the
semaphorin family in immune regulation. Adv. Immunol.
81:173-98.173-198 (2003)
12. Trusolino, L. and P.M.Comoglio: Scatter-factor and
semaphorin receptors: cell signalling for invasive growth.
Nat. Rev. Cancer 2(4), 289-300 (2002)
13. Giordano, S., S.Corso, P.Conrotto, S.Artigiani,
G.Gilestro, D.Barberis, L.Tamagnone, and P.M.Comoglio:
The Semaphorin 4D receptor controls invasive growth by
coupling with Met. Nat. Cell Biol. 4(9), 720-724 (2002)
14. Toyofuku, T., H.Zhang, A.Kumanogoh, N.Takegahara,
F.Suto, J.Kamei, K.Aoki, M.Yabuki, M.Hori, H.Fujisawa,
and H.Kikutani: Dual roles of Sema6D in cardiac
morphogenesis through region-specific association of its
receptor, Plexin-A1, with off-tack and vascular endothelial
growth factor receptor type 2. Genes Dev. 18(4), 435-447
(2004)
15. Tamagnone, L., S.Artigiani, H.Chen, Z.He, G.I.Ming,
H.Song, A.Chedotal, M.L.Winberg, C.S.Goodman, M.Poo,
M.Tessier-Lavigne, and P.M.Comoglio: Plexins are a large
family of receptors for transmembrane, secreted, and GPI-
anchored semaphorins in vertebrates. Cell 99(1), 71-80
(1999)
16. Winberg, M. L., L.Tamagnone, J.Bai, P.M.Comoglio,
D.Montell, and C.S.Goodman: The transmembrane protein
off-track associates with plexins and functions downstream
of semaphorin signaling during axon guidance. Neuron
32(1), 53-62 (2001)
17. He, Z. and M.Tessier-Lavigne: Neuropilin is a receptor
for the axonal chemorepellent Semaphorin III. Cell 90(4),
739-751 (1997)
18. Kolodkin, A. L., D.V.Levengood, E.G.Rowe, Y.T.Tai,
R.J.Giger, and D.D.Ginty: Neuropilin is a semaphorin III
receptor. Cell 90(4), 753-762 (1997)
19. Giger, R. J., E.R.Urquhart, S.K.Gillespie, D.V.Levengood,
D.D.Ginty, and A.L.Kolodkin: Neuropilin-2 is a receptor for
semaphorin IV: insight into the structural basis of receptor
function and specificity. Neuron 21(5), 1079-1092 (1998)
20. Takahashi, T., F.Nakamura, Z.Jin, R.G.Kalb, and
S.M.Strittmatter: Semaphorins A and E act as antagonists of
neuropilin-1 and agonists of neuropilin-2 receptors. Nat.
Neurosci. 1487-493 (1998)
21. Nakamura, F., M.Tanaka, T.Takahashi, R.G.Kalb, and
S.M.Strittmatter: Neuropilin-1 extracellular domains mediate
semaphorin D/III- induced growth cone collapse. Neuron
21(5), 1093-1100 (1998)
22. Takahashi, T., A.Fournier, F.Nakamura, L.H.Wang,
Y.Murakami, R.G.Kalb, H.Fujisawa, and S.M.Strittmatter:
Plexin-neuropilin-1 complexes form functional
semaphorin-3A receptors. Cell 99(1), 59-69 (1999)
23. Suto, F., Y.Murakami, F.Nakamura, Y.Goshima, and
H.Fujisawa: Identification and characterization of a novel
mouse plexin, plexin-A4. Mech. Dev. 120(3), 385-396
(2003)
24. Takahashi, T. and S.M.Strittmatter: PlexinA1
Autoinhibition by the Plexin Sema Domain. Neuron 29(2),
429-439 (2001)
25. Rohm, B., A.Ottemeyer, M.Lohrum, and
A.W.Pueschel: Plexin/neuropilin complexes mediate
repulsion by the axonal guidance signal semaphorin 3A.
Mech. Dev. 93(1-2), 95-104 (2000)
26. Torres-Vazquez, J., A.D.Gitler, S.D.Fraser, J.D.Berk,
N.P.Van, M.C.Fishman, S.Childs, J.A.Epstein, and
B.M.Weinstein: Semaphorin-plexin signaling guides
patterning of the developing vasculature. Dev. Cell 7(1),
117-123 (2004)
27. Gitler, A. D., M.M.Lu, and J.A.Epstein: PlexinD1 and
Semaphorin Signaling Are Required in Endothelial Cells
for Cardiovascular Development. Dev. Cell 7(1), 107-116
(2004)
28. Castellani, V., A.Chedotal, M.Schachner, C.Faivre-
Sarrailh, and G.Rougon: Analysis of the L1-deficient
mouse phenotype reveals cross-talk between Sema3A and
L1 signaling pathways in axonal guidance. Neuron 27(2),
237-249 (2000)
29. Castellani, V., E.De Angelis, S.Kenwrick, and
G.Rougon: Cis and trans interactions of L1 with neuropilin-
1 control axonal responses to semaphorin 3A. EMBO J.
21(23), 6348-6357 (2002)
30. Soker, S., S.Takashima, H.Q.Miao, G.Neufeld, and
M.Klagsbrun: Neuropilin-1 is expressed by endothelial and
tumor cells as an isoform specific receptor for vascular
endothelial growth factor. Cell 92735-745 (1998)
31. Gluzman-Poltorak, Z., T.Cohen, Y.Herzog, and
G.Neufeld: Neuropilin-2 and Neuropilin-1 are receptors for
165-amino acid long form of vascular endothelial growth
factor (VEGF) and of placenta growth factor-2, but only
neuropilin-2 functions as a receptor for the 145 amino acid
form of VEGF. J Biol Chem 27518040-18045 (2000)
32. Gluzman-Poltorak, Z., T.Cohen, M.Shibuya, and
G.Neufeld: Vascular endothelial growth factor receptor-1
and neuropilin-2 form complexes. J. Biol. Chem. 276(22),
18688-18694 (2001)
33. Fuh, G., K.C.Garcia, and A.M.De Vos: The Interaction of
Neuropilin-1 with Vascular Endothelial Growth Factor and Its
Receptor Flt-1. J. Biol. Chem. 275(35), 26690-26695 (2000)
34. Bagnard, D., C.Vaillant, S.T.Khuth, N.Dufay, M.Lohrum,
A.W.Puschel, M.F.Belin, J.Bolz, and N.Thomasset:
Semaphorin 3A-Vascular Endothelial Growth Factor-165
Balance Mediates Migration and Apoptosis of Neural
Progenitor Cells by the Recruitment of Shared Receptor. J.
Neurosci. 21(10), 3332-3341 (2001)
35. Whitaker, G. B., B.J.Limberg, and J.S.Rosenbaum:
Vascular endothelial growth factor receptor-2 and neuropilin-1
form a receptor complex that is responsible for the differential
signaling potency of VEGF(165) and VEGF(121). J. Biol.
Chem. 276(27), 25520-25531 (2001)
36. Soker, S., H.Q.Miao, M.Nomi, S.Takashima, and
M.Klagsbrun: VEGF(165) mediates formation of complexes
containing VEGFR-2 and neuropilin-1 that enhance
VEGF(165)-receptor binding. J. Cell Biochem. 85(2), 357-
368 (2002)
Semaphorins in Cancer
758
37. Kumanogoh, A., C.Watanabe, I.Lee, X.Wang, W.Shi,
H.Araki, H.Hirata, K.Iwahori, J.Uchida, T.Yasui,
M.Matsumoto, K.Yoshida, H.Yakura, C.Pan, J.R.Parnes,
and H.Kikutani: Identification of CD72 as a lymphocyte
receptor for the class IV semaphorin CD100: a novel
mechanism for regulating B cell signaling. Immunity.
13(5), 621-631 (2000)
38. Kumanogoh, A., S.Marukawa, K.Suzuki,
N.Takegahara, C.Watanabe, E.Ch'ng, I.Ishida, H.Fujimura,
S.Sakoda, K.Yoshida, and H.Kikutani: Class IV
semaphorin Sema4A enhances T-cell activation and
interacts with Tim-2. Nature 419(6907), 629-633 (2002)
39. Mitsui, N., R.Inatome, S.Takahashi, Y.Goshima,
H.Yamamura, and S.Yanagi: Involvement of Fes/Fps
tyrosine kinase in semaphorin3A signaling. EMBO J.
21(13), 3274-3285 (2002)
40. Sasaki, Y., C.Cheng, Y.Uchida, O.Nakajima,
T.Ohshima, T.Yagi, M.Taniguchi, T.Nakayama, R.Kishida,
Y.Kudo, S.Ohno, F.Nakamura, and Y.Goshima: Fyn and
Cdk5 Mediate Semaphorin-3A Signaling, Which Is
Involved in Regulation of Dendrite Orientation in Cerebral
Cortex. Neuron 35(5), 907 (2002)
41. Terman, J. R., T.Mao, R.J.Pasterkamp, H.H.Yu, and
A.L.Kolodkin: MICALs, a family of conserved
flavoprotein oxidoreductases, function in plexin-mediated
axonal repulsion. Cell 109(7), 887-900 (2002)
42. Suzuki, T., T.Nakamoto, S.Ogawa, S.Seo, T.Matsumura,
K.Tachibana, C.Morimoto, and H.Hirai: MICAL, a novel
CasL interacting molecule, associates with vimentin. J. Biol.
Chem. 277(17), 14933-14941 (2002)
43. Kozma, R., S.Sarner, S.Ahmed, and L.Lim: Rho family
GTPases and neuronal growth cone remodelling: relationship
between increased complexity induced by Cdc42Hs, Rac1, and
acetylcholine and collapse induced by RhoA and
lysophosphatidic acid . Mol. Cell. Biol. 171201-1211 (997)
44. Driessens, M. H., H.Hu, C.D.Nobes, A.Self, I.Jordens,
C.S.Goodman, and A.Hall: Plexin-B semaphorin receptors
interact directly with active Rac and regulate the actin
cytoskeleton by activating Rho. Curr. Biol. 11(5), 339-344
(2001)
45. Vikis, H. G., W.Li, and K.L.Guan: The plexin-B1/Rac
interaction inhibits PAK activation and enhances Sema4D
ligand binding. Genes Dev. 16(7), 836-845 (2002)
46. Dalpe, G., L.W.Zhang, H.Zheng, and J.G.Culotti:
Conversion of cell movement responses to Semaphorin-1 and
Plexin-1 from attraction to repulsion by lowered levels of
specific RAC GTPases in C. elegans. Development 131(9),
2073-2088 (2004)
47. Jin, Z. and S.M.Strittmatter: Rac1 mediates collapsin-1-
induced growth cone collapse. J. Neurosci. 17(16), 6256-6263
(1997)
48. Zanata, S. M., I.Hovatta, B.Rohm, and A.W.Puschel:
Antagonistic effects of Rnd1 and RhoD GTPases regulate
receptor activity in Semaphorin 3A-induced cytoskeletal
collapse. J. Neurosci. 22(2), 471-477 (2002)
49. Aurandt, J., H.G.Vikis, J.S.Gutkind, N.Ahn, and
K.L.Guan: The semaphorin receptor plexin-B1 signals through
a direct interaction with the Rho-specific nucleotide exchange
factor, LARG. Proc. Natl. Acad. Sci. U. S. A 99(19), 12085-
12090 (2002)
50. Perrot, V., J.Vazquez-Prado, and J.S.Gutkind: Plexin B
regulates Rho through the guanine nucleotide exchange
factors Leukemia-associated RhoGEF (LARG) and PDZ-
RhoGEF. J. Biol. Chem. 278(28), 26111-26119 (2002)
51. Aizawa, H., S.Wakatsuki, A.Ishii, K.Moriyama,
Y.Sasaki, K.Ohashi, Y.Sekine-Aizawa, A.Sehara-Fujisawa,
K.Mizuno, Y.Goshima, and I.Yahara: Phosphorylation of
cofilin by LIM-kinase is necessary for semaphorin 3A-
induced growth cone collapse. Nat. Neurosci. 4(4), 367-373
(2001)
52. Tordjman, R., Y.Lepelletier, V.Lemarchandel,
M.Cambot, P.Gaulard, O.Hermine, and P.H.Romeo: A
neuronal receptor, neuropilin-1, is essential for the
initiation of the primary immune response. Nat. Immunol.
3(5), 477-482 (2002)
53. Cohen, T., Z.Gluzman-Poltorak, A.Brodzky, V.Meytal,
E.Sabo, I.Misselevich, M.Hassoun, J.H.Boss, M.Resnick,
D.Shneyvas, S.Eldar, and G.Neufeld: Neuroendocrine Cells
along the Digestive Tract Express Neuropilin-2. Biochem.
Biophys. Res. Commun. 284(2), 395-403 (2001)
54. Cohen, T., Y.Herzog, A.Brodzky, J.K.Greenson,
S.Eldar, Z.Gluzman-Poltorak, G.Neufeld, and
M.B.Resnick: Neuropilin-2 is a novel marker expressed in
pancreatic islet cells and endocrine pancreatic tumours. J.
Pathol. 198(1), 77-82 (2002)
55. Catalano, A., P.Caprari, S.Rodilossi, P.Betta,
M.Castellucci, A.Casazza, L.Tamagnone, and A.Procopio:
Cross-talk between vascular endothelial growth factor and
semaphorin-3A pathway in the regulation of normal and
malignant mesothelial cell proliferation. FASEB J. 18(2),
358-360 (2004)
56. Herzog, Y., C.Kalcheim, N.Kahane, R.Reshef, and
G.Neufeld: Differential expression of neuropilin-1 and
neuropilin-2 in arteries and veins. Mech. Dev. 109115-119
(2001)
57. Tordjman, R., N.Ortega, L.Coulombel, J.Plouet,
P.H.Romeo, and V.Lemarchandel: Neuropilin-1 is
expressed on bone marrow stromal cells: A novel
interaction with hematopoietic cells? Blood 94(7), 2301-
2309 (1999)
58. Kessler, O., N.Shraga-Heled, T.Lange, N.Gutmann-
Raviv, E.Sabo, L.Baruch, M.Machluf, and G.Neufeld:
Semaphorin-3F Is an Inhibitor of Tumor Angiogenesis.
Cancer Res. 64(3), 1008-1015 (2004)
59. Van Der, Z. B., A.J.Hellemons, W.P.Leenders,
J.P.Burbach, H.G.Brunner, G.W.Padberg, and H.Van
Bokhoven: PLEXIN-D1, a novel plexin family member, is
expressed in vascular endothelium and the central nervous
system during mouse embryogenesis. Dev. Dyn. 225(3), 336-
343 (2002)
60. Granziero, L., P.Circosta, C.Scielzo, E.Frisaldi, S.Stella,
M.Geuna, S.Giordano, P.Ghia, and F.Caligaris-Cappio:
CD100/Plexin-B1 interactions sustain proliferation and
survival of normal and leukemic CD5+ B lymphocytes. Blood
101(5), 1962-1969 (2003)
61. Kagoshima, M. and T.Ito: Diverse gene expression and
function of semaphorins in developing lung: positive and
negative regulatory roles of semaphorins in lung branching
morphogenesis. Genes Cells 6(6), 559-571 (2001)
62. Gitay-Goren, H., T.Cohen, S.Tessler, S.Soker,
S.Gengrinovitch, P.Rockwell, M.Klagsbrun, B.-Z.Levi, and
G.Neufeld: Selective binding of VEGF121 to one of the three
VEGF receptors of vascular endothelial cells. J. Biol.
Chem. 2715519-5523 (1996)
Semaphorins in Cancer
759
63. Gluzman-Poltorak, Z., T.Cohen, Y.Herzog, and
G.Neufeld: Neuropilin-2 is a receptor for the vascular
endothelial growth factor (VEGF) forms VEGF-145 and
VEGF-165 [corrected]. J. Biol. Chem. 275(24), 18040-
18045 (2000)
64. Takashima, S., M.Kitakaze, M.Asakura, H.Asanuma,
S.Sanada, F.Tashiro, H.Niwa, J.J.Miyazaki, S.Hirota,
Y.Kitamura, T.Kitsukawa, H.Fujisawa, M.Klagsbrun, and
M.Hori: Targeting of both mouse neuropilin-1 and
neuropilin-2 genes severely impairs developmental yolk sac
and embryonic angiogenesis. Proc. Natl. Acad. Sci. U. S. A
99(6), 3657-3662 (2002)
65. Folkman, J.: What is the evidence that tumors are
angiogenesis dependent. J. Nat. Cancer Inst. 824-7 (1990)
66. Miao, H. Q., S.Soker, L.Feiner, J.L.Alonso, J.A.Raper,
and M.Klagsbrun: Neuropilin-1 Mediates Collapsin-
1/Semaphorin III Inhibition of Endothelial Cell Motility.
Functional competition of collapsin-1 and vascular
endothelial growth factor-165. J. Cell Biol. 146(1), 233-242
(1999)
67. Serini, G., D.Valdembri, S.Zanivan, G.Morterra,
C.Burkhardt, F.Caccavari, L.Zammataro, L.Primo,
L.Tamagnone, M.Logan, M.Tessier-Lavigne, M.Taniguchi,
A.W.Puschel, and F.Bussolino: Class 3 semaphorins
control vascular morphogenesis by inhibiting integrin
function. Nature 424(6947), 391-397 (2003)
68. Bates, D., G.I.Taylor, J.Minichiello, P.Farlie,
A.Cichowitz, N.Watson, M.Klagsbrun, R.Mamluk, and
D.F.Newgreen: Neurovascular congruence results from a
shared patterning mechanism that utilizes Semaphorin3A
and Neuropilin-1. Dev. Biol. 255(1), 77-98 (2003)
69. Miao, H. Q., P.Lee, H.Lin, S.Soker, and M.Klagsbrun:
Neuropilin-1 expression by tumor cells promotes tumor
angiogenesis and progression. FASEB J. 14(15), 2532-
2539 (2000)
70. Latil, A., I.Bieche, S.Pesche, A.Valeri, G.Fournier,
O.Cussenot, and R.Lidereau: VEGF overexpression in
clinically localized prostate tumors and neuropilin-1
overexpression in metastatic forms. Int. J. Cancer 89(2),
167-171 (2000)
71. Parikh, A. A., F.Fan, W.B.Liu, S.A.Ahmad,
O.Stoeltzing, N.Reinmuth, D.Bielenberg, C.D.Bucana,
M.Klagsbrun, and L.M.Ellis: Neuropilin-1 in human colon
cancer: expression, regulation, and role in induction of
angiogenesis. Am. J. Pathol. 164(6), 2139-2151 (2004)
72. Lacal, P. M., C.M.Failla, E.Pagani, T.Odorisio,
C.Schietroma, S.Falcinelli, G.Zambruno, and S.D'Atri:
Human melanoma cells secrete and respond to placenta
growth factor and vascular endothelial growth factor. J.
Invest Dermatol. 115(6), 1000-1007 (2000)
73. Fukahi, K., M.Fukasawa, G.Neufeld, J.Itakura, and
M.Korc: Aberrant expression of neuropilin-1 and -2 in
human pancreatic cancer cells. Clin. Cancer Res. 10(2),
581-590 (2004)
74. Soker, S., H.Fidder, G.Neufeld, and M.Klagsbrun:
Characterization of novel VEGF binding proteins
associated with tumor cells that bind VEGF165 but not
VEGF121. J. Biol. Chem. 2715761-5767 (1996)
75. Bachelder, R. E., E.A.Lipscomb, X.Lin, M.A.Wendt,
N.H.Chadborn, B.J.Eickholt, and A.M.Mercurio:
Competing Autocrine Pathways Involving Alternative
Neuropilin-1 Ligands Regulate Chemotaxis of Carcinoma
Cells. Cancer Res. 63(17), 5230-5233 (2003)
76. Shirvan, A., I.Ziv, G.Fleminger, R.Shina, Z.G.He,
I.Brudo, E.Melamed, and A.Barzilai: Semaphorins as
mediators of neuronal apoptosis. J. Neurochem. 73(3), 961-
971 (1999)
77. Bagnard, D., N.Sainturet, D.Meyronet, M.Perraut,
M.Miehe, G.Roussel, D.Aunis, M.F.Belin, and
N.Thomasset: Differential MAP kinases activation during
semaphorin3A-induced repulsion or apoptosis of neural
progenitor cells. Mol. Cell Neurosci. 25(4), 722-731 (2004)
78. Roche, J., F.Boldog, M.Robinson, L.Robinson,
M.Varella-Garcia, M.Swanton, B.Waggoner, R.Fishel,
W.Franklin, R.Gemmill, and H.Drabkin: Distinct 3p21.3
deletions in lung cancer and identification of a new human
semaphorin. Oncogene 12(6), 1289-1297 (1996)
79. Sekido, Y., S.Bader, F.Latif, J.Y.Chen, F.M.Duh,
M.H.Wei, J.P.Albanesi, C.C.Lee, M.I.Lerman, and
J.D.Minna: Human semaphorins A(V) and IV reside in the
3p21.3 small cell lung cancer deletion region and
demonstrate distinct expression patterns. Proc. Natl. Acad.
Sci. U. S. A 93(9), 4120-4125 (1996)
80. Roche, J. and H.A.Drabkin: The role of semaphorins in
lung cancer. Clin. Lung Cancer 3(2), 145-150 (2001)
81. Kuroki, T., F.Trapasso, S.Yendamuri, A.Matsuyama,
H.Alder, N.N.Williams, L.R.Kaiser, and C.M.Croce:
Allelic Loss on Chromosome 3p21.3 and Promoter
Hypermethylation of Semaphorin 3B in Non-Small Cell
Lung Cancer. Cancer Res. 63(12), 3352-3355 (2003)
82. Nasarre, P., B.Constantin, L.Rouhaud, T.Harnois,
G.Raymond, H.A.Drabkin, N.Bourmeyster, and J.Roche:
Semaphorin SEMA3F and VEGF Have Opposing Effects
on Cell Attachment and Spreading. Neoplasia. 5(1), 83-92
(2003)
83. Xiang, R., A.R.Davalos, C.H.Hensel, X.J.Zhou, C.Tse,
and S.L.Naylor: Semaphorin 3F Gene from Human 3p21.3
Suppresses Tumor Formation in Nude Mice. Cancer Res.
62(9), 2637-2643 (2002)
84. Tomizawa, Y., Y.Sekido, M.Kondo, B.Gao, J.Yokota,
J.Roche, H.Drabkin, M.I.Lerman, A.F.Gazdar, and
J.D.Minna: Inhibition of lung cancer cell growth and
induction of apoptosis after reexpression of 3p21.3
candidate tumor suppressor gene SEMA3B. Proc. Natl.
Acad. Sci. U. S. A 98(24), 13954-13959 (2001)
85. Tse, C., R.H.Xiang, T.Bracht, and S.L.Naylor: Human
Semaphorin 3B (SEMA3B) Located at Chromosome
3p21.3 Suppresses Tumor Formation in an
Adenocarcinoma Cell Line. Cancer Res. 62(2), 542-546
(2002)
86. Brambilla, E., B.Constantin, H.Drabkin, and J.Roche:
Semaphorin SEMA3F localization in malignant human
lung and cell lines: A suggested role in cell adhesion and
cell migration. Am. J. Pathol. 156(3), 939-950 (2000)
87. Ochi, K., T.Mori, Y.Toyama, Y.Nakamura, and
H.Arakawa: Identification of semaphorin3B as a direct
target of p53. Neoplasia. 4(1), 82-87 (2002)
88. Chen, H., A.Chedotal, Z.He, C.S.Goodman, and
M.Tessier-Lavigne: Neuropilin-2, a novel member of the
neuropilin family, is a high affinity receptor for the
semaphorins Sema E and Sema IV but not Sema III.
Neuron 19(3), 547-559 (1997)
Semaphorins in Cancer
760
89. Yamada, T., R.Endo, M.Gotoh, and S.Hirohashi:
Identification of semaphorin E as a non-MDR drug
resistance gene of human cancers. Proc. Natl. Acad. Sci.
USA 94(26), 14713-14718 (1997)
90. Martin-Satue, M. and J.Blanco: Identification of
semaphorin E gene expression in metastatic human lung
adenocarcinoma cells by mRNA differential display. J.
Surg. Oncol. 72(1), 18-23 (1999)
91. Rieger, J., W.Wick, and M.Weller: Human malignant
glioma cells express semaphorins and their receptors,
neuropilins and plexins. GLIA 42(4), 379-389 (2003)
92. Christensen, C. R., J.Klingelhofer, S.Tarabykina,
E.F.Hulgaard, D.Kramerov, and E.Lukanidin: Transcription
of a novel mouse semaphorin gene, M-semaH, correlates
with the metastatic ability of mouse tumor cell lines.
Cancer Res. 58(6), 1238-1244 (1998)
93. Kikutani, H. and A.Kumanogoh: Semaphorins in
interactions between T cells and antigen-presenting cells.
Nat. Rev. Immunol. 3(2), 159-167 (2003)
94. Zhang, Y. W. and G.F.Vande Woude: HGF/SF-met
signaling in the control of branching morphogenesis and
invasion. J. Cell Biochem. 88(2), 408-417 (2003)
95. van, D., V, T.E.Taher, P.W.Derksen, M.Spaargaren,
N.R.van der, and S.T.Pals: The hepatocyte growth
factor/Met pathway in development, tumorigenesis, and B-
cell differentiation. Adv. Cancer Res. 79:39-90.39-90
(2000)
96. Conrotto, P., S.Corso, S.Gamberini, P.M.Comoglio,
and S.Giordano: Interplay between scatter factor receptors
and B plexins controls invasive growth. Oncogene 23(30),
5131-5137 (2004)
97. Grant, D. S., H.K.Kleinman, I.D.Goldberg,
M.M.Bhargava, B.J.Nickoloff, J.L.Kinsella, P.Polverini,
and E.M.Rosen: Scatter Factor Induces Blood Vessel
Formation Invivo. Proc. Natl. Acad. Sci. USA 901937-
1941 (1993)
98. Rosen, E. M. and I.D.Goldberg: Regulation of
angiogenesis by scatter factor. EXS. 79193-208 (1997)
99. Swiercz, J. M., R.Kuner, and S.Offermanns: Plexin-
B1/RhoGEF-mediated RhoA activation involves the
receptor tyrosine kinase ErbB-2. J. Cell Biol. 165(6), 869-
880 (2004)
100. Peles, E. and Y.Yarden: Neu and its ligands: from an
oncogene to neural factors. Bioessays 15(12), 815-824
(1993)
101. Nakamura, F., R.G.Kalb, and S.M.Strittmatter:
Molecular basis of semaphorin-mediated axon guidance. J.
Neurobiol. 44(2), 219-229 (2000)
102. Woodhouse, E. C., A.Fisher, R.W.Bandle, B.Bryant-
Greenwood, L.Charboneau, E.F.Petricoin, III, and
L.A.Liotta: Drosophila screening model for metastasis:
Semaphorin 5c is required for l(2)gl cancer phenotype.
Proc. Natl. Acad. Sci. U. S. A 100(20), 11463-11468
(2003)
103. Inagaki, S., T.Furuyama, and Y.Iwahashi:
Identification of a member of mouse semaphorin family.
FEBS Lett. 370(3), 269-272 (1995)
104. Furuyama, T., S.Inagaki, A.Kosugi, S.Noda, S.Saitoh,
M.Ogata, Y.Iwahashi, N.Miyazaki, T.Hamaoka, and
M.Tohyama: Identification of a novel transmembrane
semaphorin expressed on lymphocytes. J. Biol. Chem.
271(52), 33376-33381 (1996)
105. Artigiani, S., P.Conrotto, P.Fazzari, G.F.Gilestro,
D.Barberis, S.Giordano, P.M.Comoglio, and L.Tamagnone:
Plexin-B3 is a functional receptor for semaphorin 5A.
EMBO Rep. 5(7), 710-714 (2004)
106. Correa, R. G., R.M.Sasahara, M.H.Bengtson,
M.L.Katayama, A.C.Salim, M.M.Brentani, M.C.Sogayar,
S.J.de Souza, and A.J.Simpson: Human semaphorin 6B
[(HSA)SEMA6B], a novel human class 6 semaphorin gene:
alternative splicing and all-trans-retinoic acid-dependent
downregulation in glioblastoma cell lines. Genomics 73(3),
343-348 (2001)
Abbreviations: The names of all semaphorins are
abbreviated according to the following rules: S always
appears as the first denoting a semaphorin. The number
after the S designates the semaphorin to a specific class,
and the final letter designates its place within the class.
Thus, S3F means semaphorin-3F while S4D means
semaphorin-4D.
Key Words: Tumor progression, tumorigenesis,
semaphorin, review, neuropilin, plexins, angiogenesis,
metastasis, invasion, Review
Send correspondence to: Dr Gera Neufeld, Cancer and
Vascular Biology Research Center, Rappaport Research
Institute in the Medical Sciences, The Bruce Rappaport
Faculty of Medicine, Technion, Israel Institute of
Technology, 1 Efron St., P.O. Box 9679, Haifa, 31096,
Israel, Tel: 972-4-8523672, Fax:972-4-8523947, E-mail:
gera@techunix.technion.ac.il
http://www.bioscience.org/current/vol10.htm
... 28, 2019; cancer patients and the largest number of breast cancer-related deaths. The semaphorin family of proteins have multiple, and variable, described roles in cancer with some demonstrating tumorsuppressive functions and others demonstrating tumor-promotional roles 21 . Our lab and others have recently begun to specifically characterize SEMA7A as a mediator of breast/mammary cancers, melanoma, and lung cancer [9][10][11]22 . ...
Preprint
Approximately 70% of all breast cancers are estrogen receptor positive (ER+BC) and endocrine therapy has improved survival for patients with ER+BC. Yet, up to half of these tumors recur within 20 years. Recurrent ER+BCs develop resistance to endocrine therapy; thus, novel targets are needed to treat recurrent ER+BC. We found that semaphorin 7A (SEMA7A) confers significantly decreased patient survival rates in ER+BC. We show that SEMA7A is hormonally regulated in ER+BC, but its expression does not uniformly decrease with anti-estrogen treatments. Additionally, overexpression of SEMA7A in ER+ cell lines drives increased in vitro growth in the presence of estrogen-deprivation, tamoxifen, and fulvestrant. In in vivo studies, we found that SEMA7A confers primary tumor resistance to fulvestrant and, importantly, induced lung metastases. Finally, we identify pro-survival signaling as a therapeutic vulnerability of ER+SEMA7A+ tumors and propose that targeting with inhibitors of survival signaling such as venetoclax may have efficacy for treating SEMA7A+ tumors. SIGNIFICANCE We report that SEMA7A predicts for, and likely contributes to, poor response to standard-of-care therapies and suggest that patients with SEMA7A+ER+ tumors may benefit from alternative therapeutic strategies.
... It was proved that for gastric and OC the overall survival of the underexpressed SEMA3A group of patients was significantly shorter comparing to the group with positive SEMA3A expression. It may suggest that SEMA3A may be an inhibitor of tumors of epithelial origin [15,17,18,21,22,[24][25][26]. It is of general knowledge that metastatic and poorly differentiated tumor cells are usually more resistant to cytotoxic drugs. ...
Article
Full-text available
Ovarian cancer is the fifth leading cause of cancer-related deaths in women. Its high mortality rate results from lack of adequate and sensitive methods allowing for the detection of the early stages of the disease, as well as low efficiency of the treatment, caused by the cytotoxic drug resistance of cancer cells. Unfortunately, tumours are able to develop new pathways and protective mechanisms that allow them to survive toxic conditions of chemotherapy. Therefore, intensive search for new genes and proteins involved in resistance to cytotoxic drugs is still needed, especially from a clinical point of view. The article presents an overview of the available literature on the role of semaphorin 3A (SEMA3A), protocadherin 9 (PCDH9), and S100 calcium binding protein A3 (S100A3) in carcinogenesis and chemoresistance of various tumors including ovarian cancer. As it turns out, the role of described genes/proteins is not limited only to their native biological activity but they function also as an oncogenic or suppressor factors in the tumor development. Moreover, they can also play an important role in development of drug resistance, as it was shown in ovarian cancer cell lines.
Article
Semaphorin family members (Semaphorins; SEMAs) consist of a large family that includes both secreted and membrane‐associated proteins that are originally found to provide axon guidance in selected areas for neural development. The SEMAs have diverse biologic activities such as neuronal cellular migration, axon guidance, vasculogenesis, branching morphogenesis, and cardiac organogenesis. Recent studies have reported that SEMAs also play crucial roles in various carcinomas. However, the association and their role in oral squamous cell carcinoma (OSCC) are still poorly understood. In this study, we evaluated SEMAs mRNA expression in OSCC‐derived cell lines using quantitative reverse transcriptase‐polymerase chain reaction (qRT‐PCR) analysis and revealed the altered gene expression profile of the SEMAs in OSCC cell lines compared with human normal oral keratinocytes. We found that tendency for up‐regulation of SEMA3E, 4C, 5A, 6B, and 7A in OSCC‐derived cell lines. Especially, SEMA3E and SEMA7A mRNA were significantly up‐regulated in all OSCC‐derived cell lines. On the other hands, SEMA3A, 3C, 3F, and 6A mRNA were significantly down‐regulated in all OSCC‐derived cell lines. The expression and role of SEMAs in OSCC depend on the forms and may provide novel insights and therapeutic target for OSCCs.
Article
Full-text available
Hepatocellular carcinoma (HCC) is responsible for the second-leading cancer-related death toll worldwide. Although sorafenib and levantinib as frontline therapy and regorafenib, cabazantinib and ramicurimab have now been approved for second-line therapy, the therapeutic benefit is in the range of only a few months with respect to prolongation of survival. Aggressiveness of HCC is mediated by metastasis. Intrahepatic metastases and distant metastasis to the lungs, lymph nodes, bones, omentum, adrenal gland and brain have been observed. Therefore, the identification of metastasis-related new targets and treatment modalities is of paramount importance. In this review, we focus on metastasis-related microRNAs (miRs) as therapeutic targets for HCC. We describe miRs which mediate or repress HCC metastasis in mouse xenograft models. We discuss 18 metastasis-promoting miRs and 35 metastasis-inhibiting miRs according to the criteria as outlined. Six of the metastasis-promoting miRs (miR-29a, -219-5p, -331-3p, 425-5p, -487a and -1247-3p) are associated with unfavourable clinical prognosis. Another set of six down-regulated miRs (miR-101, -129-3p, -137, -149, -503, and -630) correlate with a worse clinical prognosis. We discuss the corresponding metastasis-related targets as well as their potential as therapeutic modalities for treatment of HCC-related metastasis. A subset of up-regulated miRs -29a, -219-5p and -425-5p and down-regulated miRs -129-3p and -630 were evaluated in orthotopic metastasis-related models which are suitable to mimic HCC-related metastasis. Those miRNAs may represent prioritized targets emerging from our survey.
Article
Vascular endothelial growth factor (VEGF) inhibition forms the basis for anti-angiogenic therapies. With the methods based on the monoclonal antibody-mediated typical VEGF blockade, pathological angiogenesis in the tumor microenvironment is inhibited and the limitation of tumor growth is provided; however, the existing tumor tissue cannot be intervened. In this study, the anti-angiogenic effects of Semaphorin (SEMA) 3F, which has frequently been reported to have tumor suppressive properties, on a chick chorioallantoic membrane model as well as in vitro cell-cell interactions were investigated and comparatively assessed using anti-VEGF antibody. Vascular endothelial cells and chick embryos were stimulated with 10–16 ng/mL VEGF165 prior to SEMA 3F administration in order to generate pathological vascularization conditions. Both in vitro and in ovo results revealed that SEMA 3F suppressed VEGF165-induced abnormal vascularization more effectively than anti-VEGF. Moreover, the required dose of SEMA 3F was significantly lower than that of anti-VEGF (10³ times less under in ovo conditions). In light of these results, SEMA 3F is recommended as an important therapeutic agent for the prevention of pathological angiogenesis. SEMA 3F may offer an effective and efficient anti-angiogenic intervention that can be administered at a lower dose alternative to typical VEGF blocking agents.
Article
Full-text available
The semaphorins represent a large family of signaling molecules with crucial roles in neuronal and cardiac development. While normal semaphorin function pertains largely to development, their involvement in malignancy is becoming increasingly evident. One member, Semaphorin 3C (SEMA3C), has been shown to drive a number of oncogenic programs, correlate inversely with cancer prognosis, and promote the progression of multiple different cancer types. This report surveys the body of knowledge surrounding SEMA3C as a therapeutic target in cancer. In particular, we summarize SEMA3C’s role as an autocrine andromedin in prostate cancer growth and survival and provide an overview of other cancer types that SEMA3C has been implicated in including pancreas, brain, breast, and stomach. We also propose molecular strategies that could potentially be deployed against SEMA3C as anticancer agents such as biologics, small molecules, monoclonal antibodies and antisense oligonucleotides. Finally, we discuss important considerations for the inhibition of SEMA3C as a cancer therapeutic agent.
Article
Full-text available
The inflammatory and immune response elicited by the growth of cancer cells is a major element conditioning the tumor microenvironment, impinging on disease progression and patients’ prognosis. Semaphorin receptors are widely expressed in inflammatory cells, and their ligands are provided by tumor cells, featuring an intense signaling cross-talk at local and systemic levels. Moreover, diverse semaphorins control both cells of the innate and the antigen-specific immunity. Notably, semaphorin signals acting as inhibitors of anti-cancer immune response are often dysregulated in human tumors, and may represent potential therapeutic targets. In this mini-review, we provide a survey of the best known semaphorin regulators of inflammatory and immune cells, and discuss their functional impact in the tumor microenvironment.
Article
Full-text available
During development, neurons form synapses with their fate-determined targets. While we begin to elucidate the mechanisms by which extracellular ligand-receptor interactions enhance synapse specificity by inhibiting synaptogenesis, our knowledge about their intracellular mechanisms remains limited. Here we show that Rap2 GTPase (rap-2) and its effector, TNIK (mig-15), act genetically downstream of Plexin (plx-1) to restrict presynaptic assembly and to form tiled synaptic innervation in C. elegans. Both constitutively GTP- and GDP-forms of rap-2 mutants exhibit synaptic tiling defects as plx-1 mutants, suggesting that cycling of the RAP-2 nucleotide state is critical for synapse inhibition. Consistently, PLX-1 suppresses local RAP-2 activity. Excessive ectopic synapse formation in mig-15 mutants causes a severe synaptic tiling defect. Conversely, overexpression of mig-15 strongly inhibited synapse formation, suggesting that mig-15 is a negative regulator of synapse formation. These results reveal that subcellular regulation of small GTPase activity by Plexin shapes proper synapse patterning in vivo.
Article
Full-text available
Background Semaphorin 5A has been linked to tumor growth, invasion, and metastasis in pancreatic cancer. However, the role of semaphorin 5A in cervical cancer is not known. Our aim is to investigate the prognostic value of semaphorin 5A and its potential role in lymphangiogenesis and invasion in cervical cancer. Methods In this study, pathological features and clinical data of 232 cervical cancer patients were retrospectively reviewed. Semaphorin 5A protein and mRNA expression was detected by immunohistochemistry and quantitative real-time reverse transcription-polymerase chain reaction, respectively. In vitro, we determined the role and mechanistic pathways of semaphorin 5A in tumor progression in cervical carcinoma cell lines. ResultsSemaphorin 5A expression was significantly higher in stage IIb tumors than in stage Ia, Ib, and IIa tumors. High semaphorin 5A expression was significantly associated with pelvic lymph node metastasis, lymphovascular permeation, and poor survival. Semaphorin 5A induced lymphangiogenesis through a plexin-B/Met/vascular endothelial growth factor-C pathway. Semaphorin 5A also increased cervical cancer cell invasion by stimulating the expression and activity of matrix metalloproteinase-2 and matrix metalloproteinase-9 via PI3K/AKT and plexin-B3. Conclusion Our findings indicate that semaphorin 5A may represent a poor prognostic biomarker and anti-metastasis therapeutic target in cervical cancer.
Article
Full-text available
Semaphorins are a large family of secreted and transmembrane signaling proteins that repel axons in the developing nervous system. Recent studies have shown that Semaphorins are involved in the development of lung and pulmonary diseases through regulating cell proliferation, adhesion, motility and apoptosis. This review focuses on the emerging roles of the Semaphorins in lung branching morphogenesis, angiogenesis and pulmonary diseases.
Article
Full-text available
A cDNA microarray analysis indicated that Semaphorin3B. (20Sema3B), a gene whose product is involved in axon guidance and axonal repulsion, is inducible by p53. Introduction of exogenous p53 into a glioblastoma cell line lacking wild-type p53. (20U373MG) dramatically induced expression of Sema3B mRNA. An electrophoretic mobility shift assay and a reporter assay confirmed that a potential p53 binding site present in the promoter region had p53-dependent transcriptional activity. Expression of endogenous Sema3B was induced in response to genotoxic stresses caused by adriamycin treatment or UV irradiation in a p53-dependent manner. Ectopic expression of Sema3B in p53-defective cells reduced the number of colonies in colony formation assays. These results suggest that Sema3B might play some role in regulating cell growth as a mediator of p53 tumor- suppressor activity.
Article
Full-text available
Neuropilin-1 (NP1), in conjunction with plexins, promotes axon repulsion by binding to semaphorin 3A (SEMA3A). Although NP1 is expressed in carcinoma cells, its functions have remained elusive, and neither SEMA3A nor plexin expression has been explored in cancer. Here we provide evidence that breast carcinoma cells support an autocrine pathway involving SEMA3A, plexin-A1, and NP1 that impedes their ability to chemotax. Reducing SEMA3A or NP1 expression by RNA interference or inhibiting plexin-A1 signaling enhanced migration. Conversely, expression of constitutively active plexin-A1 impaired chemotaxis. The paradox of how breast carcinoma cells expressing these endogenous chemotaxis inhibitors are able to migrate is explained by their expression of vascular endothelial growth factor (VEGF), a NP1 ligand that competes with SEMA3A for receptor binding. Finally, we establish that the ratio of endogenous VEGF and SEMA3A concentrations in carcinoma cells determines their chemotactic rate. Our findings lead to the surprising conclusion that opposing autocrine loops involving NP1 regulate the chemotaxis of breast carcinoma cells. Moreover, our data indicate a novel autocrine function for VEGF in chemotaxis.
Article
Full-text available
Neuropilin-1 (NRP1) is a receptor for two unrelated ligands with disparate activities, vascular endothelial growth factor-165 (VEGF165), an angiogenesis factor, and semaphorin/collapsins, mediators of neuronal guidance. To determine whether semaphorin/collapsins could interact with NRP1 in nonneuronal cells, the effects of recombinant collapsin-1 on endothelial cells (EC) were examined. Collapsin-1 inhibited the motility of porcine aortic EC (PAEC) expressing NRP1 alone; coexpressing KDR and NRP1 (PAEC/KDR/NRP1), but not parental PAEC; or PAEC expressing KDR alone. The motility of PAEC expressing NRP1 was inhibited by 65-75% and this inhibition was abrogated by anti-NRP1 antibody. In contrast, VEGF165 stimulated the motility of PAEC/KDR/NRP1. When VEGF165 and collapsin-1 were added simultaneously to PAEC/KDR/NRP1, dorsal root ganglia (DRG), and COS-7/NRP1 cells, they competed with each other in EC motility, DRG collapse, and NRP1-binding assays, respectively, suggesting that the two ligands have overlapping NRP1 binding sites. Collapsin-1 rapidly disrupted the formation of lamellipodia and induced depolymerization of F-actin in an NRP1-dependent manner. In an in vitro angiogenesis assay, collapsin-1 inhibited the capillary sprouting of EC from rat aortic ring segments. These results suggest that collapsin-1 can inhibit EC motility as well as axon motility, that these inhibitory effects on motility are mediated by NRP1, and that VEGF165 and collapsin-1 compete for NRP1-binding sites.
Article
Full-text available
Semaphorins SEMA3B and its homologue SEMA3F are 3p21.3 candidate tumor suppressor genes (TSGs), the expression of which is frequently lost in lung cancers. To test the TSG candidacy of SEMA3B and SEMA3F, we transfected them into lung cancer NCI-H1299 cells, which do not express either gene. Colony formation of H1299 cells was reduced 90% after transfection with wild-type SEMA3B compared with the control vector. By contrast, only 30–40% reduction in colony formation was seen after the transfection of SEMA3F or SEMA3B variants carrying lung cancer-associated single amino acid missense mutations. H1299 cells transfected with wild-type but not mutant SEMA3B underwent apoptosis. We found that lung cancers (n = 34) always express the neuropilin-1 receptor for secreted semaphorins, whereas 82% expressed the neuropilin-2 receptor. Because SEMA3B and SEMA3F are secreted proteins, we tested conditioned medium from COS-7 cells transfected with SEMA3B and SEMA3F and found that medium from wild-type SEMA3B transfectants reduced the growth of several lung cancer lines 30–90%, whereas SEMA3B mutants or SEMA3F had little effect in the same assay. Sequencing of sodium bisulfite-treated DNA showed dense methylation of CpG sites in the SEMA3B 5′ region of lung cancers not expressing SEMA3B but no methylation in SEMA3B-expressing tumors. These results are consistent with SEMA3B functioning as a TSG, the expression of which is inactivated frequently in lung cancers by allele loss and promoter region methylation.
Article
In the May 2002 issue of Nature Immunology, an incorrect version of Figure 3c was supplied. The correct version is shown below. This mistake does not affect the main message of the paper.
Article
Mutations in the L1 gene induce a spectrum of human neurological disorders due to abnormal development of several brain structures and fiber tracts. Among its binding partners, L1 immunoglobulin superfamily adhesion molecule (Ig CAM) associates with neuropilin-1 (NP-1) to form a semaphorin3A (Sema3A) receptor and soluble L1 converts Sema3A-induced axonal repulsion into attraction. Using L1 constructs containing missense pathological mutations, we show here that this reversion is initiated by a specific trans binding of L1 to NP-1, but not to L1 or other Ig CAMs, and leads to activation of the NO/cGMP pathway. We identified the L1–NP-1-binding site in a restricted sequence of L1 Ig domain 1, as a peptide derived from this region could reverse Sema3A repulsive effects. A pathological L1 missense mutation located in this sequence specifically disrupts both L1–NP-1 complex formation and Sema3A reversion, suggesting that the cross-talk between L1 and Sema3A might participate in human brain development.
Article
The semaphorins are a family of secreted, transmembrane, and membrane-associated proteins initially identified as causing the repulsion of nerve growth cone guidance. However, subsequent experiments have demonstrated that they can induce retraction in non-neural cells and affect the development of non-neural organs. Two related secreted semaphorins, SEMA3F and SEMA3B, were isolated from a recurrent 3p21.3 homozygous deletion region in small-cell lung cancer (SCLC) cell lines. Moreover, a functionally related molecule, Roundabout/DUTT1, was identified as the target of a more proximal chromosome 3p deletion also involving SCLCs. Based on current data, it is likely that the loss of these semaphorins or Roundabout may affect cell migration, metastasis, and apoptosis. In addition, receptors for the secreted semaphorins, neuropilin-1 and neuropilin-2, have been shown to function as coreceptors for a subset of vascular endothelial growth factor (VEGF) isoforms and related growth factors. Since semaphorins and VEGF bind antagonistically to neuropilins, their loss is likely to facilitate angiogenesis. In lung cancer specimens, antibody staining against SEMA3F has been shown to correlate with stage and histologic subtypes with more aggressive tumors showing increased VEGF and decreased SEMA3F staining. This review is focused on a basic understanding of these pathways with an emphasis on their role in lung cancer.