You are on page 1of 12

Drug Resistance Updates 29 (2016) 1–12

Contents lists available at ScienceDirect

Drug Resistance Updates


journal homepage: www.elsevier.com/locate/drup

The semaphorins and their receptors as modulators of tumor


progression
Gera Neufeld ∗ , Yelena Mumblat, Tanya Smolkin, Shira Toledano, Inbal Nir-Zvi,
Keren Ziv, Ofra Kessler
The Cancer Research and Vascular Biology Center, The Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa 31096, Israel

a r t i c l e i n f o a b s t r a c t

Article history: The semaphorins were initially characterized as repulsive axon guidance factors. However, they are cur-
Received 22 May 2016 rently also recognized as important regulators of diverse biological processes which include regulation of
Received in revised form 31 July 2016 immune responses, angiogenesis, organogenesis, and a variety of additional physiological and develop-
Accepted 23 August 2016
mental functions. The semaphorin family consists of more than 20 genes divided into seven subfamilies,
all of which contain the sema domain signature. They usually transduce signals by activation of receptors
Keywords:
belonging to the plexin family, either directly, or indirectly following the binding of some semaphorins to
Semaphorins
receptors of the neuropilin family which subsequently associate with plexins. Additional receptors which
Angiogenesis
Cancer
form complexes with these primary semaphorin receptors are also frequently involved in semaphorin sig-
Lymphangiogenesis nalling, and can strongly influence the nature of the biological responses of cells to semaphorins. Recent
evidence suggests that semaphorins play important roles in the etiology of multiple forms of cancer.
Some semaphorins such as some semaphorins belonging to the class-3 semaphorin subfamily, have been
found to function as bona fide tumor suppressors and to inhibit tumor progression by various mech-
anisms. Because these class-3 semaphorins are secreted proteins, these semaphorins may potentially
be used as anti-tumorigenic drugs. Other semaphorins, such as semaphorin-4D, function as inducers
of tumor progression and represent targets for the development of novel anti-tumorigenic drugs. The
mechanisms by which semaphorins affect tumor progression are diverse, ranging from direct effects on
tumor cells to modulation of accessory processes such as modulation of immune responses and inhibition
or promotion of tumor angiogenesis and tumor lymphangiogenesis. This review focuses on the diverse
mechanisms by which semaphorins affect tumor progression.
© 2016 Elsevier Ltd. All rights reserved.

1. The semaphorins and their receptors their N-termini, and by a plexin-semaphorin-integrin (PSI) domain
located downstream to the sema domain. The sema domain has
1.1. The semaphorins a ␤ propeller topology (Love et al., 2003; Antipenko et al., 2003;
Liu et al., 2010), is essential for semaphorin activity, and deter-
Members of the semaphorin family are divided into 8 subclasses mines to some extent, the receptor binding specificity (Feiner
of which subclasses 1 and 2 contain invertebrate semaphorins, et al., 1997). Different semaphorin subclasses can be distinguished
whereas subclasses 3-7 contain the 22 vertebrate semaphorins by class-specific structural motifs. Thus, vertebrate semaphorins
and subclass 8 contains viral semaphorins. In early publications, belonging to classes 4 and 7 contain immunoglobulin-like domains,
semaphorins were assigned confusing names. This situation was class-5 semaphorins contain thrombospondin repeats and class-
rectified by the adoption of a unified semaphorin nomenclature 3 semaphorins contain a basic domain. Class-3 semaphorins are
in which sema is followed by the subclass number and by alpha- the only vertebrate semaphorins produced as secreted proteins
betic designation within the subclass (Goodman et al., 1999). while other vertebrate semaphorins are membrane anchored.
The semaphorins as well as their plexin receptors are character- Some membrane-anchored semaphorins can be further processed
ized by a ∼500 amino acids-long sema domain located close to into soluble forms by proteolytic cleavage (Fig. 1). Some membrane
anchored semaphorins may also be able to function as signal trans-
ducing proteins (Toyofuku et al., 2012; Segarra et al., 2012). The
∗ Corresponding author. active forms of several class-3 and class-6 semaphorins are homod-
E-mail address: gera@tx.technion.ac.il (G. Neufeld). imers (Klostermann et al., 1998; Liu et al., 2010; Janssen et al.,

http://dx.doi.org/10.1016/j.drup.2016.08.001
1368-7646/© 2016 Elsevier Ltd. All rights reserved.
2 G. Neufeld et al. / Drug Resistance Updates 29 (2016) 1–12

Fig. 1. The vertebrate semaphorins: (A) The structural elements of vertebrate semaphorin subclasses are shown. All semaphorins feature the signature N-terminal sema
domain. A conserved stretch of amino-acid residues near the C-terminal of the sema domain bears homology to the N-terminal of ␤-integrins and is designated as the PSI
domain. Class-3 semaphorins are the only secreted semaphorins and are distinguished by a conserved basic domain at their C-termini. Class 4-7 semaphorins are membrane-
anchored. Class 5 semaphorins are distinguished by thrombospondin repeats. All the vertebrate semaphorins except for the class-5 and 6 semaphorins also contain an
immunoglobulin-like domain. The extracellular domains of class-4 semaphorins can be cleaved with metalloproteases generating active soluble extracellular domains.

2010; Nogi et al., 2010), suggesting that the active forms of all the (Franco and Tamagnone, 2008) and to the inactivation of small
semaphorins are dimeric. GTPases that control the polymerization of the actin cytoskeleton
such as Rho as a result of the activation of regulators of Rho activ-
ity including the p190 Rho-GTPase and Rho guanine nucleotide
1.2. Plexin family receptors exchange factors (Puschel, 2007; Worzfeld et al., 2014). However,
semaphorin induced signal transduction is far from being com-
The receptors of the plexin family are segregated into four pletely understood and its thorough description is beyond the
groups consisting of four Type-A plexins, three Type-B plexins, scope of the present review.
and single C and D plexins (Negishi et al., 2005a; Gutmann-Raviv
et al., 2006). Most semaphorins bind to one or to several plexins
(Hota and Buck, 2012). For example, plexin-B1 is a receptor for 1.3. Neuropilins
sema4D (Tamagnone et al., 1999), plexin-A2 and plexin-A4 func-
tion as sema6A and sema6B receptors (Suto et al., 2007; Suto et al., Six of the seven class-3 semaphorins are unable to bind to plex-
2005) and plexin-D1 is a receptor for sema3E and sema4A (Gu et al., ins directly but instead bind to one or to both receptors of the
2005; Toyofuku et al., 2007) (Fig. 2). The sema domain found in the neuropilin family (Gu et al., 2005; Neufeld and Kessler, 2008).
extracellular domain of the plexins serves as an auto-inhibitory Because of their short intracellular domains, neuropilins are not
domain in the basal, non-activated state of the receptor and the able to transduce semaphorin signals independently, and asso-
inhibition is removed following a conformational change induced ciate with type-A plexins or with plexin-D1 after binding a class-3
by the binding of a semaphorin (Takahashi and Strittmatter, 2001). semaphorin to transduce semaphorin signals (Tamagnone et al.,
The intracellular domains of plexins are characterized by the pres- 1999; Takahashi et al., 1999; Gitler et al., 2004). Recent structural
ence of a conserved GTPase activating (GAP) domain (Oinuma et al., studies indicate that functional class-3 semaphorin receptors con-
2004; He et al., 2009; Sakurai et al., 2010). Most of the develop- sist of a tetramer containing a neuropilin homodimer and a plexin
mental effects of plexin-D1 and plexin-B1 are lost when the GAP homodimer that are linked together by the binding of a class-3
domain is mutated suggesting that it plays an essential role in semaphorin homodimer (Janssen et al., 2012). However, there is
plexin mediated semaphorin signal transduction (Worzfeld et al., evidence indicating that functional class-3 semaphorin receptors
2014). Type-A plexins associate spontaneously to form homod- can also contain plexin heterodimers since inhibition of the expres-
imers (Janssen et al., 2010; Nogi et al., 2010) or heterodimers (Kigel sion of either neuropilin-1 or plexin-A1 or plexin-A4 in endothelial
et al., 2011). Recent data indicates that activation of plexin signal- cells was found to be sufficient to completely abrogate sema3A sig-
ing by semaphorins that bind directly to plexins such as sema6A is nal transduction suggesting that the receptor complex in these cells
associated with a conformational change that shifts the conforma- contains plexin-A4 as well as plexin-A1 in addition to neuropilin-
tion of the dimer from the inactive to the active conformation (Liu 1 (Kigel et al., 2011). Similarly, both plexin-A2 and plexin-A4 are
et al., 2010; Nogi et al., 2010). required for sema3B induced signal transduction suggesting that
Activation of plexin signaling by semaphorins activates the GAP functional sema3B receptors also contain more than one type of
domain leading to the inactivation of R-ras, resulting in the subse- plexin (Sabag et al., 2014).
quent inactivation of beta1-integrin, and finally reduced adhesion The neuropilins can perhaps be best described as “scaffold
(Negishi et al., 2005b; Toyofuku et al., 2005; Sakurai et al., 2010; receptors” since they seem to bind to and modulate the activities of
Sakurai et al., 2011). Activation of type-A plexins was also found diverse types of receptors and ligands but do not appear to trans-
to induce the activation of enzymes of the Mical family. These duce signals independently. In addition to class-3 semaphorins,
enzymes perform reduction-oxidation (redox) enzymatic reactions the neuropilins also function as receptors for several types of
and oxidize actin subunits leading to the disassembly of actin growth factors including several members of the vascular endothe-
fibers and to the localized collapse of the actin cytoskeleton of lial growth factor (VEGF) family (Gitay-Goren et al., 1996; Soker
axonal growth cones, thereby contributing to growth cone guid- et al., 1998; Makinen et al., 1999; Karpanen et al., 2006; Migdal
ance (Terman et al., 2002; Hung et al., 2010; Hung et al., 2011; Hung et al., 1998), hepatocyte growth factor (HGF) (Sulpice et al.,
et al., 2013). Activation of plexin signaling by semaphorins also 2007) and TGF-␤ (Glinka and Prud’homme, 2008) to name but
results in the activation of various intracellular tyrosine-kinases a few. The VEGF-A binding domain of neuropilin-1 seems to be
G. Neufeld et al. / Drug Resistance Updates 29 (2016) 1–12 3

Fig. 2. Semaphorin receptors: The different semaphorins are described using a three letter code in which the S stands for semaphorin, the number designates the subfamily
and the following letter the specific sub-family member. Thus, S3a stands for sema3A. The binding receptors for the different semaphorins are shown. Most of the class-3
semaphorins bind to receptors composed of neuropilins and plexins but the primary binding site is located on the neuropilins.

distinct from its semaphorin binding domain (Appleton et al., 2007), the epidermal growth factor (EGF) receptor (Rizzolio et al.,
2007) and it was indeed observed that sema3A and sema3F can 2012) and the hepatocyte growth factor receptor (Matsushita
inhibit VEGF induced activation of ERK1/2 without inhibition et al., 2007).
of VEGF induced auto-phosphorylation of the VEGFR2 receptor Mice lacking functional neuropilin-1 display major defects
which associates with neuropilin-1 (Guttmann-Raviv et al., 2007), in the organization of their nervous system and in their blood
lending support to these observations. However, there is also vessels resulting in embryonic lethality. Mice lacking functional
evidence suggesting that some class-3 semaphorins may com- neuropilin-2 developed almost normally but have defects in
pete with members of the VEGF family for binding to neuropilins lymph vessels (Yuan et al., 2002). The intracellular domain of
(Mumblat et al., 2015) and that post-translational modifications of neuropilin-1 contains a PDZ binding domain which binds synectin
semaphorins such as cleavage by furin like pro-protein convertases (also known as GIPC or NIP) which is important for the formation
(Adams et al., 1997) may modulate their neuropilin binding ability of complexes with VEGFR-2 (Cai and Reed, 1999; Prahst et al.,
and their ability to compete with VEGFs for binding to neuropilins 2008). The neuropilins can also form complexes with adhesion
(Guo et al., 2013). receptors such as L1-CAM which associates with neuropilin-1
In addition to binding several types of diverse ligands, or with Nr-CAM which associates with neuropilin-2, and these
neuropilins are also able to form complexes with diverse interactions were found to modulate signal transduction induced
membrane-anchored receptors in addition to plexins. In this by class-3 semaphorins such as sema3A and sema3F (Castellani
respect, the best studied interaction is with the VEGFR-2 tyrosine- et al., 2002; Falk et al., 2005). Interestingly, the interactions of
kinase receptor in which neuropilin-1 functions as an amplifier the neuropilins are not limited to interactions with proteins.
that enhances VEGF induced pro-angiogenic signaling mediated by Thus, neuropilin-1 was recently found to bind to rare 3-O-sulfate
this receptor (Soker et al., 1998). Similarly, neuropilin-2 interacts moieties of heparan sulfate containing proteoglycans thereby
with the VEGF-C receptor VEGFR-3 and is required for efficient inhibiting sema3A induced signal transduction in axonal growth
transduction of VEGF-C induced pro-lymphangiogenic signals (Xu cones of sema3A responsive neurons (Thacker et al., 2016).
et al., 2010; Karpanen et al., 2006; Favier et al., 2006). Similarly,
neuropilins form complexes and modulate signal transduction 1.4. Additional semaphorin receptors
mediated by several additional receptors including TGF-␤ recep-
tors (Glinka et al., 2011), platelet derived growth factor (PDGF) Some semaphorins bind to additional types of receptors besides
receptors (Cao et al., 2010), neurotrophin receptors (Ben-Zvi et al., plexins and neuropilins. Thus, sema4A also signals using the Tim-2
4 G. Neufeld et al. / Drug Resistance Updates 29 (2016) 1–12

Fig. 3. The dual role of sema3E in tumor progression: Like all class-3 semaphorins, sema3E contains a conserved cleavage site for furin-like pro-protein convertases (FPPC)
which when cleaved generates the active N-terminal fragment p61-Sema3E (Upper Panel). In contrast to uncleaved full length sema3E, P61-Sema3E induces the association
of the sema3E plexin-D1 receptor with ErbB2 on tumor cells, thus activating “in-trans” ErbB2 mediated signal transduction that promotes tumor metastasis. Full length
sema3E does not promote this association and only inhibits angiogenesis after it binds to its plexin-D1 receptor. FPPC expression is strongly up-regulated in malignant tumor
cells causing most of the sema3E produced by the tumor cells to be cleaved to p61-Sema3E. As a result p61-Sema3E efficiently competes with the remaining full length
sema3E for binding to plexin-D1, resulting in the induction of ErbB2 mediated tumor metastasis.

receptor, a member of the family of T-cell immunoglobulin domain experiments in endothelial cells (Gitay-Goren et al., 1996). They
and mucin domain (Tim) proteins that is expressed on activated were subsequently identified as the products of the neuropilin-
T cells (Kumanogoh et al., 2002a). Sema4D was also found to bind 1 (Soker et al., 1998) and neuropilin-2 (Gluzman-Poltorak et al.,
to the lymphocyte receptor CD-72 (Kumanogoh et al., 2000) in 2000) genes. Since neuropilins have already been identified as
addition to plexin-B1 and plexin-B2. Sema5A and sema3A also class-3 semaphorin receptors (Kolodkin et al., 1997; He and
bind to chondroitin sulfate proteoglycans, an interaction that can Tessier-Lavigne, 1997; Chen et al., 1997), it followed that class-3
convert sema5A from an attractive to an guidance inhibitory cue semaphorins could perhaps modulate VEGF-A induced angiogene-
(Kantor et al., 2004; Dick et al., 2013). Sema4B also binds to a sis. Since angiogenesis is usually required for the development of
CLCP1, a receptor that displays a structural similarity to neuropilins solid tumors (Folkman, 1990), it followed that if the semaphorins
(Nagai et al., 2007). Finally, sema7A binds to ␣-1/␤-1 integrin to do modulate angiogenesis, then they will likely affect tumor
modulate inflammatory responses mediated by these integrins progression. This logic led initially to the identification of the
(Suzuki et al., 2007) (Fig. 2). class-3 semaphorin sema3F as an inhibitor of angiogenesis and
angiogenesis-dependent tumor progression (Kessler et al., 2004;
2. Modulation of angiogenesis by semaphorins Bielenberg et al., 2004). The identification of sema3F as an anti-
angiogenic semaphorin lead to the characterization of additional
2.1. Anti-angiogenic semaphorins class-3 semaphorins, acting as inhibitors of angiogenesis. Indeed,
the class-3 semaphorins sema3A (Acevedo et al., 2008), sema3B
Vascular endothelial growth factor (VEGF-A) had been exten- (Varshavsky et al., 2008), sema3C (Yang et al., 2015; Mumblat
sively characterized as a major angiogenesis promoting factor et al., 2015), sema3D (Sabag et al., 2012) and sema3E (Casazza
(Gospodarowicz et al., 1989; Leung et al., 1989; Keck et al., 1989). et al., 2010) were found to function as inhibitors of angiogene-
VEGF-A is produced in several forms as a result of alternative sis. It is not completely clear how the class-3 semaphorins inhibit
splicing. VEGF-A signals are transduced by two tyrosine kinase angiogenesis. They function on the one hand as repulsive guidance
receptors (VEGFR-1 and VEGFR-2) which bind all the VEGF-A factors and it was found, for example, that expression of sema3E in
splice forms. This indicated that there may exist receptors that somites of developing embryos repels blood vessels and keeps the
are able to distinguish between different VEGF-A splice forms. somites avascular (Gu et al., 2005). On the other hand, prolonged
Such receptors were indeed first identified in binding/cross-linking stimulation of endothelial cells to sema3A and sema3F induces
G. Neufeld et al. / Drug Resistance Updates 29 (2016) 1–12 5

apoptosis and it is possible that part of the anti-angiogenic effect such as sema3A (Casazza et al., 2011), sema3B (Varshavsky et al.,
of semaphorins is due to induction of apoptosis (Guttmann-Raviv 2008) sema3D (Sabag et al., 2012) and sema3F (Kessler et al., 2004;
et al., 2007; Reidy et al., 2009; Maione et al., 2009; Shirvan et al., Bielenberg et al., 2004) inhibit tumor angiogenesis and tumor
1999). development. Down-regulation of sema3A expression in tumor
The development of the retinal vasculature serves as a major cells promotes tumor angiogenesis and tumor progression in
developmental angiogenesis model because the whole network of many types of solid tumors (Vacca et al., 2006; Maione et al., 2009;
blood vessels can be easily observed in retinal whole-mounts. New- Barresi and Tuccari, 2010; Song et al., 2012; Jiang et al., 2015;
born babies as well as newborn mouse pups exposed to high partial Tang et al., 2014; Jiang et al., 2015; Zhou et al., 2014) suggesting
oxygen pressure develop blindness when shifted back to normoxia that sema3A does indeed function as an endogenous negative
(retinopathy of prematurity (ROP)), because of wild growth of new regulator of the angiogenic switch (Hanahan and Folkman, 1996).
blood vessels that is driven by the acute hypoxia felt by astrocytes Likewise, over-expression of sema3A in tumor cells or addition
following the sudden drop in oxygen partial pressure which induces of exogenous sema3A can inhibit tumor angiogenesis and tumor
the astrocytes to express high levels of VEGF (Alon et al., 1995; progression (Kigel et al., 2008; Acevedo et al., 2008; Casazza et al.,
Stone et al., 1995). Sema3A functions as an inhibitor of the wild 2011; Chakraborty et al., 2012; Sabag et al., 2012). Sema3F and
angiogenesis induced by a shift to normoxia as well as an inhibitor sema3B expression in tumor cells also inhibits tumor angiogenesis
of laser coagulation induced choroidal angiogenesis (Yu et al., 2013; and tumor development (Kessler et al., 2004; Ikegami et al.,
Bai et al., 2014). Tip cells are endothelial cells located at the tips 2004; Varshavsky et al., 2008). In the case of sema3F it was also
of the growing angiogenic sprouts. These cells send out filopodia found that its expression is under the control of p53. Loss of p53
and lamellipodia to guide the growing sprout while the stalk cells activity which is frequently encountered in tumor cells results in
which are the endothelial cells that form the main body of the grow- down-regulation of sema3F expression thus alleviating sema3F
ing sprout do not extend such filopodia (Siekmann et al., 2008; induced inhibition of angiogenesis and promoting tumor growth,
Eilken and Adams, 2010). Sema3A expressed by endothelial cells suggesting that sema3F also functions as an endogenous inhibitor
was found to inhibit filopodia formation by vascular tip cells in of angiogenesis (Futamura et al., 2007).
angiogenic sprouts. Consequently, knock-out of the sema3A gene All class-3 semaphorins contain conserved cleavage sites for
in endothelial cells causes a significant increase in the number and proteases belonging to the furin-like pro-protein convertase family
the length of tip cell filopodia at the leading edge of angiogenic (Adams et al., 1997). The expression of some furin-like pro-
sprouts of developing retinal vessels (Ochsenbein et al., 2016). Like- protein convertases is up-regulated by hypoxia (McMahon et al.,
wise, sema3E was also found to inhibit angiogenesis in the ROP 2005) and their expression is up-regulated in malignant cells
model as well as laser- induced choroidal angiogenesis (Suda et al., (Bassi et al., 2005). Cleavage by furin-like pro-protein conver-
2014; Fukushima et al., 2011). Activation of VEGFR-2 in tip cells by tases can be a modality for the inactivation of class-3 semaphorins
VEGF also induces the expression of the notch ligand Dll4 in the thereby relieving anti-angiogenic inhibition as in the case of
tip cells. Dll4 then activates signaling by notch receptors in stalk sema3B (Varshavsky et al., 2008). However, it should be noted
cells located adjacent to the tip cells in angiogenic sprouts. This in that in addition to their direct effects on endothelial cells, anti-
turn down-regulates the expression of VEGFR-2 in the stalk cells, angiogenic semaphorins such as sema3B can also activate biological
resulting in the maintenance of their stalk cell identity (Hellstrom responses in additional types of cells and these may indirectly
et al., 2007; Lobov et al., 2007). Interestingly, VEGF also induces modulate the anti-angiogenic activity of these semaphorins. For
the expression of the plexin-D1 receptor in tip cells of sprouting example, sema3B can also induce the expression of interleukin-
angiogenic retinal blood vessels. Sema3E produced by retinal gan- 8, which in turn, induces the recruitment of tumor-associated
glion cells acts specifically on the plexin-D1 expressing tip cells to macrophages and promotes the metastatic dissemination of tumor
inhibit the VEGF induced expression of Dll4 causing a cell fate shift cells (Rolny et al., 2008). Interleukin-8 is also a well character-
that favours tip cells identity. Thus, sema3E expression is part of ized pro-angiogenic factor (Huang et al., 2002) and it is also likely
a feedback mechanism by which neuronal cells of the retina regu- that induction of interleukin-8 expression induced by sema3B
late the formation of the developing vascular network (Kim et al., may counteract the direct anti-angiogenic effects of sema3B. Sim-
2011). Sema4A, a membrane anchored semaphorin that also uti- ilarly, sema3A was reported to recruit, to developing tumors, a
lizes plexin-D1 as its receptor also functions as an anti-angiogenic neuropilin-1 expressing subpopulation of bone marrow-derived
factor (Toyofuku et al., 2007) although it is not known if it is also part monocytes that promotes the recruitment of pericytes and the sta-
of a similar feedback mechanism. Finally, sema3F was also found bilization and normalization of tumor blood vessels thus protecting
to function as a regulator of angiogenesis during the development them against angiogenesis inhibitors (Zacchigna et al., 2008; Carrer
of the retinal vasculature in which sema3F produced by RPE cells et al., 2012).
in the outer retina forms an anti-angiogenic barrier that inhibits In contrast with class-3 semaphorins such as sema3A or
the growth of choroidal and retinal blood vessels into these layers sema3F which were characterized primarily as anti-tumorigenic
(Buehler et al., 2013). Thus, several class-3 semaphorins function semaphorins, the class-3 semaphorin sema3E was paradoxically
as natural anti-angiogenic agents during embryonic development. found to promote tumor progression despite its anti-angiogenic
Taken together, these observations suggest that anti-angiogenic activity (Christensen et al., 1998). Like other class-3 semaphorins,
class-3 semaphorins may function as endogenous inhibitors of sema3E is cleaved by furin-like pro-protein convertases. One of the
tumor angiogenesis whose function is lost during tumor progres- cleavage products is a shorter peptide (p61-Sema3E) which also
sion, enabling the angiogenic switch thus allowing developing displays anti-angiogenic properties. However, unlike full- length
tumors to initiate angiogenesis-driven growth (Hanahan and sema3E, p61-Sema3E also promotes the association of the sema3E
Folkman, 1996). This hypothesis predicts that class-3 semaphorins receptor plexin-D1 with ErbB2 receptors on tumor cells, resulting
may function as tumor suppressors. Indeed, both sema3F and in the “in-trans” activation of ErbB2 and promotion of tumor metas-
sema3B have been characterized as bona fide tumor suppressor tasis (Casazza et al., 2010). In contrast to p61-Sema3E, full-length
genes (Tomizawa et al., 2001; Sekido et al., 1996; Xiang et al., 1996; sema3E rendered resistant to cleavage by furin-like pro-protein
Xiang et al., 2002). These observations further suggest that class-3 convertases by the introduction of point mutation into the con-
semaphorins could perhaps be utilized as drugs for the treatment of served furin cleavage sites, retained the anti-angiogenic activity
solid tumor development and of diseases characterized by rampant of full length sema3E, but was unable to activate ErbB2 and did
deregulated angiogenesis. Indeed, several class-3 semaphorins not promote tumor metastasis. Furthermore, this point mutant
6 G. Neufeld et al. / Drug Resistance Updates 29 (2016) 1–12

inhibited the binding of p61-Sema3E to plexin-D1, and thereby Akt signaling pathway (Basile et al., 2005; Basile et al., 2007a).
prevented the activation of ErbB2 and the induction of tumor It was recently reported that this Met independent activity is
metastasis by p61-Sema3E while simultaneously inhibiting tumor mediated, in addition, by Rho/Rho Kinase (ROK) dependent genera-
angiogenesis and tumor development (Casazza et al., 2012) (Fig. 3). tion of PI(4,5)P(2) upon treatment of endothelial cells with Sema4D
Similarly, sema3C was also characterized as a promoter of tumor (Binmadi et al., 2011; Binmadi et al., 2012). In addition, activation of
progression (Miyato et al., 2012; Man et al., 2014) even though it plexin-B1 by sema4D in endothelial cells can result in the activation
too functions as an anti-angiogenic factor that inhibits, for exam- of NF-kappaB and subsequently in the expression of the angiogenic
ple, oxygen-induced retinal angiogenesis (Yang et al., 2015). In this factor IL-8 (Yang et al., 2011; Strieter et al., 1992). Because sema4D
case too, a point mutated sema3C resistant to cleavage by furin activates angiogenesis using the plexin-B1 receptor and not the
like pro-protein convertases displayed anti-angiogenic properties VEGFR-2 receptor used by VEGF, it follows that sema4D can act
and inhibited tumor growth and metastasis. Preliminary evidence additively with VEGF and that inhibition of sema4D signaling can
suggests that the p65-Sema3C peptide produced from sema3C as represent an alternative anti-angiogenic treatment strategy (Zhou
a result cleavage by furin-like pro-protein convertases functions et al., 2012). In addition, it was observed that sema4D also regulates
as a promoter of tumor cell survival (Mumblat et al., 2015). It is the permeability of blood vessels by enhancing the expression of
thus possible that in the case of sema3C too, cleavage by furin- PDGF-B and angiopoietin like protein-4 in endothelial cells, result-
like pro-protein convertases is responsible for the activation of ing in the dissociation of pericytes (Zhou et al., 2013).
pro-tumorigenic functions. Studies of sema7A focused primarily on its immune modulating
activity. However, recent evidence indicates that sema7A also
2.2. Pro-angiogenic semaphorins functions as a pro-tumorigenic and pro-angiogenic factor. Thus, its
expression is up-regulated in transformed mammary tumor cells.
In contrast to the class-3 semaphorins, which function primar- Macrophages exposed to sema7A expressed elevated levels of
ily as inhibitors of angiogenesis, Sema4D (also known as CD100) CXCL2/MIP-2 and tumor cells in which the expression of sema7A
functions as an inducer of angiogenesis and as a promoter of tumor was inhibited formed less angiogenic tumors (Garcia-Areas et al.,
progression that plays a central role in the development and tumor 2014). Sema7A expression is associated with ductal carcinoma in
progression of head and neck squamous cell carcinomas (Basile situ (DCIS) progression. Enhanced sema7A expression occurs in a
et al., 2004; Basile et al., 2006). Sema4D is a membrane-bound large percentage of breast cancers and is associated with decreased
class-4 semaphorin that binds to plexin-B1, plexin-B2 and to the overall and distant metastasis-free survival and similar results
CD-72 receptors (Tamagnone et al., 1999; Kumanogoh et al., 2000) were reported with regard to oral cancer (Black et al., 2016; Saito
(Fig. 2). The extracellular domain of sema4D can be cleaved and et al., 2015).
released from producing cells by membrane type-1 matrix met-
alloproteinase (MT1-MMP) and by the metalloprotease ADAM17
(TACE). These two metalloproteases are up-regulated in many types 3. Modulation of lymph vessels-mediated tumor
of malignant cells (Strongin, 2010; Zhu et al., 2007; Arribas and progression by semaphorins
Esselens, 2009). Sema4D is stored in platelets and its extracellu-
lar domain can be released from platelets (Zhu et al., 2007). The Lymph vessels drain excessive fluids from tissues. Lymphangio-
cleaved soluble extracellular domain of sema4D retains the biolog- genesis is the process by which new lymph vessels grow out of an
ical activity of the full length sema4D (Basile et al., 2007b) and was existing bed of lymph vessels. Tumor development is frequently
used extensively to study the role of sema4D in immune reactions, accompanied by lymphangiogenesis that is induced by tumor
tumor progression and the control of angiogenesis. derived lymphangiogenic factors such as VEGF-C and VEGF-D, and
The tyrosine-kinase receptor Met is the receptor for hepatocyte in certain forms of cancer such as in head and neck carcinomas or
growth factor/scatter factor (HGF/SF), a potent inducer of tumor in breast cancer tumor cells invade these new lymph vessels and
cell invasiveness and angiogenesis (Bussolino et al., 1992; Trusolino utilize them to migrate to sentinel lymph nodes in which they form
et al., 2010). The soluble extracellular domain of sema4D was found secondary tumors and from these sites further migrate to distant
to bind to plexin-B1 and to induce the association of the sema4D organs (Li and Li, 2014; Podgrabinska and Skobe, 2014; Karaman
receptor plexin-B1 with Met. This association then promotes “in- and Detmar, 2014; Alitalo and Detmar, 2012).
trans” auto-phosphorylation of the Met receptor and induction The discovery of anti-angiogenic semaphorins suggests that
of tumor cells invasiveness (Giordano et al., 2002). It was subse- some semaphorins may also function as regulators of lymph ves-
quently observed that sema4D can also transactivate the related sels function and lymphangiogenesis. Indeed, sema3A and its
macrophage stimulating protein (MSP) receptor Ron (Yao et al., neuropilin-1 and plexin-A1 are required for correct development
2013), and that all three type-B plexins are able to form complexes of lymph vessel valves. In utero treatment of mice with an antibody
with the Met and Ron receptors (Conrotto et al., 2004; Conrotto that blocks Sema3A binding to neuropilin-1, but not treatment
et al., 2005). Sema5A, a semaphorin that binds to the plexin-B3 with an antibody that blocks VEGF-A binding to neuropilin-1, com-
receptor, can also transactivate Met similarly to sema4D (Artigiani promised the function of lymph vessels, and resulted in abnormal
et al., 2004). Since HGF/SF is also a potent inducer of angiogenesis, lymph vessel and lymphatic valve morphology (Jurisic et al., 2012;
these observations suggested that sema4D may also function as a Bouvree et al., 2012).
pro-angiogenic factor and thereby further promote tumor progres- Sema3F repels cultured lymphatic endothelial cells suggest-
sion as was indeed demonstrated (Conrotto et al., 2005). ing that it may function as an anti-lymphangiogenic agent in vivo
However, sema4D was also found to induce angiogenesis inde- (Ikegami et al., 2004). Indeed, it was recently demonstrated that
pendently of Met by utilizing plexin-B1 induced Rho dependent sema3F inhibits tumor lymphangiogenesis and tumor metastasis of
mechanisms (Basile et al., 2004). This mechanism involves the acti- head and neck squamous cell carcinomas, a tumor type that metas-
vation of the PI3K/Akt pathway following the binding of sema4D tasizes primarily via lymph vessels (Doci et al., 2015; Zhang et al.,
to plexin-B1. Activated plexin-B1 activates in turn an intracellu- 2014). Likewise, it was recently found that sema3C also functions
lar tyrosine kinase cascade that involves the sequential activation as a repellant of lymphatic endothelial cells. A mutated form of
of PYK2 and Src which results in the tyrosine phosphorylation sema3C resistant to cleavage by furin-like pro-protein convertases
of Plexin-B1, recruitment of a multimeric signaling complex that strongly reduced the density of lymph vessels in tumors derived
includes PYK2, Src, and PI3K to Plexin-B1 and activation of the from breast cancer cells and strongly inhibited the spontaneous
G. Neufeld et al. / Drug Resistance Updates 29 (2016) 1–12 7

formation of lymph node metastases (Mumblat et al., 2015). In con- surface plexin-B1 receptors (Giordano et al., 2002; Yao et al., 2013).
trast, enhanced expression of sema4D is associated with enhanced Likewise, sema5A, a semaphorin that binds to the plexin-B3 recep-
expression of VEGF-C and VEGF-D and with increased lymph node tor, can also transactivate Met similarly to sema4D (Artigiani et al.,
metastasis of cervical cancer and colon cancer (Liu et al., 2014; Mu 2004). These observations lead to the development of inhibitory
et al., 2014). Likewise, expression of sema7A was also reported to humanized antibodies that are developed as anti-tumorigenic bio-
be associated with increased metastasis and increased lymphan- logicals that target sema4D (Patnaik et al., 2015). However, there
giogenesis of breast cancer tumors (Black et al., 2016). are also opposite observations. In melanocytes and in malignant
melanoma cells sema4D inhibits HGF-induced activation of Met
and the inhibition of plexin-B1 expression in these cells leads to
4. Direct effects of semaphorins on tumor cells and tumor the activation, rather than to the inactivation of Met (McClelland
progression et al., 2011; Soong et al., 2012; Soong and Scott, 2012). In breast car-
cinoma cells, plexin-B1 and plexins-B2 also form complexes with
In addition to their effects on angiogenesis and lymphangio- the ErbB2 tyrosine-kinase receptor, and sema4D as well as sema4C
genesis, semaphorins can also affect the behavior of tumor cells are both able to induce ErbB2 phosphorylation “in-trans” following
directly. Thus, sema3A inhibited the proliferation of gastric can- their binding to plexins-B1 or plexins-B2 receptors (Swiercz et al.,
cer cells and their migration in “in vitro” assays, indicating that in 2004). In these cells, the binding of sema4D to plexin-B1 which
addition to its anti-angiogenic effects it can also affect the behavior is associated with ErbB2, induces cell migration and metastasis;
of tumor cells directly (Tang et al., 2014). Sema3A also inhibited whereas the binding of sema4D to plexin-B1 associated with Met
the anchorage-independent proliferation of triple negative breast causes inhibition of cell migration, indicating that the exchange of
cancer cells as did additional class-3 semaphorins such as sema3D the two receptor tyrosine kinases is sufficient to convert the cellu-
and sema3F but not sema3E (Kigel et al., 2008). The sema3B gene lar response of Sema4D from pro- to anti-migratory and vice versa
was identified along with sema3F as a tumor suppressor whose (Swiercz et al., 2008). Similar observations were also reported in
function is lost in small cell lung carcinoma cells by a variety of prostate cancer cells (Damola et al., 2013). It was recently observed
mechanisms that include promoter methylation and loss of het- that the activation of plexin-B1 by Sema4D in breast carcinoma
erozygosity (Tomizawa et al., 2001; Kuroki et al., 2003; Nair et al., cells results in tyrosine phosphorylation of plexin-B1 by Met, thus
2007; Campioni et al., 2008; Chen et al., 2014; Loginov et al., creating a docking site for the SH2 domain of growth factor recep-
2015; Gao et al., 2015). Single nucleotide polymorphisms in the tor bound-2 (Grb2). Grb2 is thereby recruited into the plexin-B1
sema3B gene were also found to be associated with poor prog- receptor complex and through its SH3 domain, interacts with p190
nosis of prostate cancer (Beuten et al., 2009). Lung cancer and RhoGAP and mediates RhoA deactivation and leads to the subse-
breast cancer cells express neuropilins, and it found that sema3B quent inhibition of breast carcinoma cells motility (Sun et al., 2012).
induces apoptosis of breast and lung cancer cells grown in cell Sema6D is a membrane-anchored semaphorin that binds to the
culture. This pro-apoptotic effect was inhibited by VEGF165 which plexin-A1 receptor. It induces the association of plexin-A1 with
binds to neuropilins but not by VEGF121 which does not, suggest- the VEGFR-2 VEGF tyrosine-kinase receptors and activates signal
ing that VEGF165 functions as a survival factor for these cells and transduction by this receptor (Toyofuku et al., 2004). Sema6D
that sema3B can inhibit this effect of VEGF165 (Castro-Rivera et al., functions as a survival and metastasis inducing gene in malignant
2004). Similarly, sema3F causes non small cell lung carcinoma cells pleural mesothelioma in a plexin-A1 and VEGFR-2-dependent
to round up and lose extracellular contacts (Kusy et al., 2005) and manner (Catalano et al., 2009). In addition, sema6D is important
inhibited the anchorage-independent growth of breast cancer cells for tumor progression in a subtype of triple negative breast cancer
(Kigel et al., 2008). In breast cancer cells, the expression of sema3F is (Chen et al., 2015). Another family member, sema6B, was recently
regulated by retinoid orphan nuclear receptor ␣ (ROR␣). Reduced described as an inducer of metastasis in gastric cancer (Ge et al.,
ROR␣ expression correlated with diminished sema3F expression 2013) but was also reported as a possible inhibitor of breast
and poor prognosis while restoration of ROR␣ expression repro- cancer progression (D’Apice et al., 2013). Sema5A is a semaphorin
grammed breast cancer cells to form non-invasive structures in 3D that binds to the plexin-A1, plexin-A3 and plexin-B3 receptors
culture (Xiong et al., 2012). Likewise Sema3F inhibited the attach- (Matsuoka et al., 2011; Artigiani et al., 2004). It was recently found
ment and spreading of MCF-7 breast cancer cells and inhibited the to promote tumor metastasis in gastric cancer (Pan et al., 2012)
expression of E-cadherin thus contributing to epithelial to mes- and in pancreatic cancer (Sadanandam et al., 2012) but inhibited
enchymal transition (EMT) (Perl et al., 1998), a process of cardinal the motility of glioma cells (Li and Lee, 2010). Sema5B may play
importance for the conversion of tumor cells into metastatic cells a role in renal cell carcinoma since down-regulation of sema5B
(Nasarre et al., 2003; Nasarre et al., 2005). Knockdown of SEMA3F expression in renal cell carcinoma cells significantly compromises
also promoted the self-renewal and tumorigenicity of colorectal their viability (Hirota et al., 2006).
cancer cells and increased the expression of stemness-associated
genes while over-expression of SEMA3F reduced the expression
of stemness genes (Rao et al., 2014). In addition, Sema3F inhib- 5. Modulation of tumor progression by semaphorins that
ited integrin-␤1-mediated attachment of A375 melanoma cells by a alter immune responses or by semaphorins expressed in
neuropilin-2-mediated mechanism and suppressed the metastatic immune cells
spread of cells from tumors derived from these cells (Bielenberg
et al., 2004). SEMA3F inhibits phosphatidyl inositol-3 kinase (PI3K) Tumor associated macrophages (TAMs) secrete a variety of pro-
and Akt activity in a variety of target cells including endothelial angiogenic factors including VEGF and PlGF, which render them key
cells and several tumor cell types. These responses were associated promoters of tumor angiogenesis. Normally TAMs are character-
with the disruption of mTOR/rictor assembly and mTOR-dependent ized as M1 TAMs that express CD11c which function as inhibitors
activation of the RhoA GTPase (Nakayama et al., 2015). of tumor progression. However, in the tumor microenvironment
In contrast with the above mentioned class-3 semaphorins, they frequently change their gene expression profile and behave
sema4D acts as a pro-tumorigenic factor. Sema4D can also pro- as M2 macrophages that secrete pro-angiogenic factors such as
mote tumor progression in “trans” by activation of the Met and Ron VEGF, PlGF and sema4D to promote tumor angiogenesis and tumor
tyrosine-kinase receptors and induction of epithelial to mesenchy- progression (Sierra et al., 2008; Mantovani and Sica, 2010). M2
mal transition (EMT) following the binding of sema4D to tumor cell macrophages also suppress anti-tumor immunity by preventing
8 G. Neufeld et al. / Drug Resistance Updates 29 (2016) 1–12

activation of dendritic cells (DCs), cytotoxic T lymphocytes (CTLs), proliferation. Sema7A also plays a role in the adaptive immune sys-
and natural killer (NK) cells (Mantovani and Sica, 2010). tem in vitro by promoting transmigration of poly morphonuclear
Treatment of macrophages and monocytes with thioglycolate granulocytes across endothelial cells into hypoxic areas via acti-
or LPS induces the activation of toll like receptors (TLRs) and vation of its Plexin-C1 receptor. Thus up-regulation of sema7A
increases the expression levels of SEMA4A and its receptors Plexin- appears to enhance the immune response at hypoxic sites such as
B2 and plexin-D1 specifically in Ly6Chigh inflammatory monocytes those found in tumors by enhancing the accumulation of polymor-
but not in Ly6Clow resident monocytes. In turn, Sema4A increases phonuclear granulocytes (Morote-Garcia et al., 2012).
the migratory capacity of macrophages and induces VEGF expres-
sion as a result of the activation of plexin-D1, thereby enhancing 6. Conclusions and future perspectives
angiogenesis. Since inflammation and tumor progression are inti-
mately linked (Ben-Neriah and Karin, 2011; Mantovani et al., 2008), Initially it was thought that the semaphorins would function
these observations suggest that sema4A produced by recruited primarily as inhibitors of tumor progression and tumor angiogen-
monocytes and macrophages can also indirectly promote tumor esis. This turned out not to be the case and nowadays several
progression. Unlike other class-4 semaphorins, sema4A also uti- semaphorins have been found to promote tumor progression
lizes neuropilin-1 as a binding/signaling receptor. Neuropilin-1 and to enhance angiogenesis. Furthermore, several semaphorins
is required by Treg cells to limit anti-tumor immune responses. were reported to both induce and inhibit tumor progression.
Sema4A binds to neuropilin-1 expressed on T-regulatory cells These different activities seem context-dependent and evidence
(Tregs) and as a result supports their survival (Delgoffe et al., 2013). exists suggesting that interactions between semaphorin receptors
An important aspect of tumor progression is the ability of and apparently unrelated receptors such as various tyrosine-
cancer cells to escape detection and clearance by the immune sys- kinase receptors as well as post-translational modifications of the
tem. Interestingly, some tumor cells express sema3A despite its semaphorins and their receptors can profoundly affect their biolog-
anti-angiogenic properties. T cells express the neuropilin-1 and ical activities as exemplified in the case of sema3E (Casazza et al.,
plexin-A4 sema3A receptors, and Sema3A containing conditioned 2010; Chauvet et al., 2007; Luchino et al., 2013; Rizzolio et al., 2012).
medium from such tumor cells was observed to inhibit the prolif- These interactions and modifications can in turn profoundly affect
eration of these cells and their activation by anti-Cd3 antibodies. In the course of diseases such as cancer, and a better understanding of
contrast, suppression of Sema-3A expression in tumor cells using these interactions and post translational modifications is required
a specific siRNA augmented T-cell activation (Catalano et al., 2006; if one considers the development of anti-tumorigenic and anti-
Yamamoto et al., 2008). Thus, sema3A seems to interfere with the angiogenic therapeutic agents that target or utilize semaphorin
recognition of tumor cells by the immune system. signal transduction. Thus, research aimed at a better understand-
Sema4D was also identified as a critical regulator of T cell func- ing of the processing of semaphorins and their receptors and better
tion. In the tumor microenvironment Sema4D is mainly expressed characterization of the cross-talk between semaphorins and their
by TAMs. CD4+ T-cells derived from mice lacking a functional receptors and other signal transduction pathways is likely to be a
sema4D gene display impaired proliferation and impaired cytokine focus of attention for some time to come. In addition to cancer, it
production in response to antigen stimulation (Kumanogoh et al., seems that semaphorins play major regulatory roles in the devel-
2002b), while transgenic mice overexpressing a soluble extra- opment and maintenance of the vascular and neuronal networks of
cellular domain of sema4D display enhanced T-cell responses organs such as the retina and the kidney as well as in the immune
(Watanabe et al., 2001). Likewise, down-regulation of sema4D system. It is likely that the study of the role of the semaphorins in
expression in CD8+ T-cells is correlated with impaired T-cells func- the development of vascular diseases such as complications of dia-
tion. Dendritic cells (DC) express the sema4D receptor CD-72 and betes including diabetic retinopathy or diabetic nephropathy will
the binding of sema4D to DC induces their maturation which in also become a focus of intensive research in the near future.
turn results in enhanced activation of T-cells (Ishida et al., 2003;
Li et al., 2006). Natural killer cells express sema4D as well as Conflict of interest
CD-72, and stimulation by sema4D enhanced target cell killing
(Lammie et al., 1991). These activities of sema4D are apparently The authors report no conflict of interest.
in conflict with the pro-angiogenic and pro-tumorigenic functions
of sema4D and how these activities are balanced remains to be Acknowledgements
elucidated. Interestingly, sema4D not only affects the activities of
immune cells but also their recruitment to the tumor microenvi- This work was supported by grants from the Israel Science Foun-
ronment. Strong expression of SEMA4D at the invasive margins of dation (ISF) and by a grant from the Rappaport Family Institute for
actively growing tumors influences the infiltration and distribu- Research in the Medical Sciences.
tion of leukocytes in the tumor microenvironment. Neutralization
of sema4D function by blocking antibodies disrupts this gradient References
of expression and enhances recruitment of activated monocytes
and lymphocytes into the tumor. This shifts the balance of cells Acevedo, L.M., Barillas, S., Weis, S.M., Gothert, J.R., Cheresh, D.A., 2008. Semaphorin
3A suppresses VEGF-mediated angiogenesis yet acts as a vascular permeability
and cytokines in a pro-inflammatory and anti-tumorigenic direc- factor. Blood 111, 2674–2680.
tion and was found to be associated with durable tumor rejection in Adams, R.H., Lohrum, M., Klostermann, A., Betz, H., Puschel, A.W., 1997. The
murine Colon26 and ERBB2+ mammary carcinoma models (Evans chemorepulsive activity of secreted semaphorins is regulated by furin-
dependent proteolytic processing. EMBO J. 16, 6077–6086.
et al., 2015). The migration of immune cells can also be controlled
Alitalo, A., Detmar, M., 2012. Interaction of tumor cells and lymphatic vessels in
by other semaphorins such as sema3F which was recently found to cancer progression. Oncogene 31, 4499–4508.
repulse thymocytes and T-cells (Mendes-Da-Cruz et al., 2014). Alon, T., Hemo, I., Itin, A., Peer, J., Stone, J., Keshet, E., 1995. Vascular endothelial
growth factor acts as a survival factor for newly formed retinal vessels and has
Another semaphorin that has distinct roles in immune
implications for retinopathy of prematurity. Nat. Med. 1, 1024–1028.
responses is sema7A. Activated T-cells express sema7A, and Antipenko, A., Himanen, J.P., van Leyen, K., Nardi-Dei, V., Lesniak, J., Barton, W.A.,
sema7A in turn induces the expression of pro-inflammatory fac- et al., 2003. Structure of the semaphorin-3A receptor binding module. Neuron
tors in macrophages (Suzuki et al., 2007; Holmes et al., 2002). 39, 589–598.
Appleton, B.A., Wu, P., Maloney, J., Yin, J., Liang, W.C., Stawicki, S., et al., 2007.
Sema7A produced by T-cells inhibits the activation of T-cells Structural studies of neuropilin/antibody complexes provide insights into
since T-cells lacking sema7A display enhanced antigen induced semaphorin and VEGF binding. EMBO J. 26, 4902–4912.
G. Neufeld et al. / Drug Resistance Updates 29 (2016) 1–12 9

Arribas, J., Esselens, C., 2009. ADAM17 as a therapeutic target in multiple diseases. Castro-Rivera, E., Ran, S., Thorpe, P., Minna, J.D., 2004. Semaphorin 3B (SEMA3B)
Curr. Pharm. Des. 15, 2319–2335. induces apoptosis in lung and breast cancer, whereas VEGF165 antagonizes this
Artigiani, S., Conrotto, P., Fazzari, P., Gilestro, G.F., Barberis, D., Giordano, S., et al., effect. Proc. Natl. Acad. Sci. U. S. A. 101, 11432–11437.
2004. Plexin-B3 is a functional receptor for semaphorin 5A. EMBO Rep. 5, Catalano, A., Caprari, P., Moretti, S., Faronato, M., Tamagnone, L., Procopio, A., 2006.
710–714. Semaphorin-3A is expressed by tumor cells and alters T-cell signal transduction
Bai, Y., Liang, S., Yu, W., Zhao, M., Huang, L., Zhao, M., et al., 2014. Semaphorin and function. Blood 107, 3321–3329.
3A blocks the formation of pathologic choroidal neovascularization induced by Catalano, A., Lazzarini, R., Di, N.S., Orciari, S., Procopio, A., 2009. The Plexin-A1
transforming growth factor beta. Mol. Vis. 20, 1258–1270, eCollection@2014, receptor activates vascular endothelial growth factor-receptor 2 and nuclear
1258–1270. factor-{kappa}B to mediate survival and anchorage-independent growth of
Barresi, V., Tuccari, G., 2010. Increased ratio of vascular endothelial growth fac- malignant mesothelioma cells. Cancer Res. 69, 1485–1493.
tor to semaphorin3A is a negative prognostic factor in human meningiomas. Chakraborty, G., Kumar, S., Mishra, R., Patil, T.V., Kundu, G.C., 2012. Semaphorin 3A
Neuropathology 30, 537–546. suppresses tumor growth and metastasis in mice melanoma model. PLoS ONE
Basile, J.R., Afkhami, T., Gutkind, J.S., 2005. Semaphorin 4D/plexin-B1 induces 7, e33633.
endothelial cell migration through the activation of PYK2, Src, and the phos- Chauvet, S., Cohen, S., Yoshida, Y., Fekrane, L., Livet, J., Gayet, O., et al., 2007. Gat-
phatidylinositol 3-kinase-Akt pathway. Mol. Cell Biol. 25, 6889–6898. ing of Sema3E/PlexinD1 signaling by neuropilin-1 switches axonal repulsion to
Basile, J.R., Barac, A., Zhu, T., Guan, K.L., Gutkind, J.S., 2004. Class IV semaphorins attraction during brain development. Neuron 56, 807–822.
promote angiogenesis by stimulating rho-initiated pathways through plexin-B. Chen, D., Li, Y., Wang, L., Jiao, K., 2015. SEMA6D expression and patient survival in
Cancer Res. 64, 5212–5224. breast invasive carcinoma. Int. J. Breast Cancer, article 539721.
Basile, J.R., Castilho, R.M., Williams, V.P., Gutkind, J.S., 2006. Semaphorin 4D provides Chen, H., Chedotal, A., He, Z., Goodman, C.S., Tessier-Lavigne, M., 1997. Neuropilin-
a link between axon guidance processes and tumor-induced angiogenesis. Proc. 2, a novel member of the neuropilin family, is a high affinity receptor for the
Natl. Acad. Sci. U. S. A. 103, 9017–9022. semaphorins Sema E and Sema IV but not Sema III. Neuron 19, 547–559.
Basile, J.R., Gavard, J., Gutkind, J.S., 2007a. Plexin-B1 utilizes RhoA and Rho kinase to Chen, R., Zhuge, X., Huang, Z., Lu, D., Ye, X., Chen, C., et al., 2014. Analysis of SEMA3B
promote the integrin-dependent activation of Akt and ERK and endothelial cell methylation and expression patterns in gastric cancer tissue and cell lines. Oncol.
motility. J. Biol. Chem. 282, 34888–34895. Rep. 31, 1211–1218.
Basile, J.R., Holmbeck, K., Bugge, T.H., Gutkind, J.S., 2007b. MT1-MMP controls tumor- Christensen, C.R., Klingelhofer, J., Tarabykina, S., Hulgaard, E.F., Kramerov, D., Lukani-
induced angiogenesis through the release of semaphorin 4D. J. Biol. Chem. 282, din, E., 1998. Transcription of a novel mouse semaphorin gene, M-semaH,
6899–6905. correlates with the metastatic ability of mouse tumor cell lines. Cancer Res.
Bassi, D.E., Fu, J., Lopez de, C.R., Klein-Szanto, A.J., 2005. Proprotein convertases: 58, 1238–1244.
“master switches” in the regulation of tumor growth and progression. Mol. Conrotto, P., Corso, S., Gamberini, S., Comoglio, P.M., Giordano, S., 2004. Interplay
Carcinog. 44, 151–161. between scatter factor receptors and B plexins controls invasive growth. Onco-
Ben-Neriah, Y., Karin, M., 2011. Inflammation meets cancer, with NF-kappaB as the gene 23, 5131–5137.
matchmaker. Nat. Immunol. 12, 715–723. Conrotto, P., Valdembri, D., Corso, S., Serini, G., Tamagnone, L., Comoglio, P.M., et al.,
Ben-Zvi, A., Ben-Gigi, L., Klein, H., Behar, O., 2007. Modulation of semaphorin3A 2005. Sema4D induces angiogenesis through Met recruitment by Plexin B1.
activity by p75 neurotrophin receptor influences peripheral axon patterning. J. Blood 105, 4321–4329.
Neurosci. 27, 13000–13011. D’Apice, L., Costa, V., Valente, C., Trovato, M., Pagani, A., Manera, S., et al., 2013.
Beuten, J., Garcia, D., Brand, T.C., He, X., Balic, I., Canby-Hagino, E., et al., 2009. Analysis of SEMA6B gene expression in breast cancer: identification of a new
Semaphorin 3B and 3F single nucleotide polymorphisms are associated with isoform. Biochim. Biophys. Acta 1830, 4543–4553.
prostate cancer risk and poor prognosis. J. Urol. 182, 1614–1620. Damola, A., Legendre, A., Ball, S., Masters, J.R., Williamson, M., 2013. Function of
Bielenberg, D.R., Hida, Y., Shimizu, A., Kaipainen, A., Kreuter, M., Kim, C.C., et al., mutant and wild-type plexinb1 in prostate cancer cells. Prostate 73, 1326–1335.
2004. Semaphorin 3F, a chemorepulsant for endothelial cells, induces a poorly Delgoffe, G.M., Woo, S.R., Turnis, M.E., Gravano, D.M., Guy, C., Overacre, A.E., et al.,
vascularized, encapsulated, nonmetastatic tumor phenotype. J. Clin. Invest. 114, 2013. Stability and function of regulatory T cells is maintained by a neuropilin-
1260–1271. 1-semaphorin-4a axis. Nature 501, 252–256.
Binmadi, N.O., Proia, P., Zhou, H., Yang, Y.H., Basile, J.R., 2011. Rho-mediated acti- Dick, G., Tan, C.L., Alves, J.N., Ehlert, E.M., Miller, G.M., Hsieh-Wilson, L.C., et al., 2013.
vation of PI(4)P5K and lipid second messengers is necessary for promotion of Semaphorin 3A binds to the perineuronal nets via chondroitin sulfate type E
angiogenesis by Semaphorin 4D. Angiogenesis 14, 309–319. motifs in rodent brains. J. Biol. Chem. 288, 27384–27395.
Binmadi, N.O., Yang, Y.H., Zhou, H., Proia, P., Lin, Y.L., Batista De Paula, A.M., et al., Doci, C.L., Mikelis, C.M., Lionakis, M.S., Molinolo, A.A., Gutkind, J.S., 2015. Genetic
2012. Plexin-B1 and Semaphorin 4D cooperate to promote perineural invasion identification of SEMA3F as an anti-lymphangiogenic metastasis suppressor
in a RhoA/ROK-dependent manner. Am. J. Pathol. 180, 1232–1242. gene in head and neck squamous carcinoma. Cancer Res. 75, 2937–2948.
Black, S.A., Nelson, A.C., Gurule, N.J., Futscher, B.W., Lyons, T.R., 2016. Semaphorin Eilken, H.M., Adams, R.H., 2010. Dynamics of endothelial cell behavior in sprouting
7a exerts pleiotropic effects to promote breast tumor progression. Oncogene (in angiogenesis. Curr. Opin. Cell Biol. 22, 617–625.
press). Evans, E.E., Jonason Jr., A.S., Bussler, H., Torno, S., Veeraraghavan, J., Reilly, C., et al.,
Bouvree, K., Brunet, I., Del, T.R., Gordon, E., Prahst, C., Cristofaro, B., et al., 2012. 2015. Antibody blockade of semaphorin 4D promotes immune infiltration into
Semaphorin3A, neuropilin-1, and PlexinA1 are required for lymphatic valve tumor and enhances response to other immunomodulatory therapies. Cancer
formation. Circ. Res. 111, 437–445. Immunol. Res. 3, 689–701.
Buehler, A., Sitaras, N., Favret, S., Bucher, F., Berger, S., Pielen, A., et al., 2013. Falk, J., Bechara, A., Fiore, R., Nawabi, H., Zhou, H., Hoyo-Becerra, C., et al., 2005. Dual
Semaphorin 3F forms an anti-angiogenic barrier in outer retina. FEBS Lett. 587, functional activity of semaphorin 3B is required for positioning the anterior
1650–1655. commissure. Neuron 48, 63–75.
Bussolino, F., Di Renzo, M.F., Ziche, M., Bocchietto, E., Olivero, M., Naldini, L., et al., Favier, B., Alam, A., Barron, P., Bonnin, J., Laboudie, P., Fons, P., et al., 2006. Neuropilin-
1992. Hepatocyte growth factor is a potent angiogenic factor which stimulates 2 interacts with VEGFR-2 and VEGFR-3 and promotes human endothelial cell
endothelial cell motility and growth. J. Cell Biol. 119, 629–641. survival and migration. Blood 108, 1243–1250.
Cai, H.B., Reed, R.R., 1999. Cloning and characterization of neuropilin-1-interacting Feiner, L., Koppel, A.M., Kobayashi, H., Raper, J.A., 1997. Secreted chick semaphorins
protein: a PSD-95/Dlg/ZO-1 domain-containing protein that interacts with the bind recombinant neuropilin with similar affinities but bind different subsets of
cytoplasmic domain of neuropilin-1. J. Neurosci. 19, 6519–6527. neurons in situ. Neuron 19, 539–545.
Campioni, M., Ambrogi, V., Pompeo, E., Citro, G., Castelli, M., Spugnini, E.P., et al., Folkman, J., 1990. What is the evidence that tumors are angiogenesis dependent. J.
2008. Identification of genes down-regulated during lung cancer progression: a Nat. Cancer Inst. 82, 4–7.
cDNA array study. J. Exp. Clin. Cancer Res. 27, 38. Franco, M., Tamagnone, L., 2008. Tyrosine phosphorylation in semaphorin signalling:
Cao, S., Yaqoob, U., Das, A., Shergill, U., Jagavelu, K., Huebert, R.C., et al., 2010. shifting into overdrive. EMBO Rep. 9, 865–871.
Neuropilin-1 promotes cirrhosis of the rodent and human liver by enhancing Fukushima, Y., Okada, M., Kataoka, H., Hirashima, M., Yoshida, Y., Mann, F., et al.,
PDGF/TGF-beta signaling in hepatic stellate cells. J. Clin. Invest. 120, 2379–2394. 2011. Sema3E-PlexinD1 signaling selectively suppresses disoriented angiogen-
Carrer, A., Moimas, S., Zacchigna, S., Pattarini, L., Zentilin, L., Ruozi, G., et al., esis in ischemic retinopathy in mice. J. Clin. Invest. 121, 1974–1985.
2012. Neuropilin-1 identifies a subset of bone marrow Gr1-monocytes that can Futamura, M., Kamino, H., Miyamoto, Y., Kitamura, N., Nakamura, Y., Ohnishi, S.,
induce tumor vessel normalization and inhibit tumor growth. Cancer Res. 15, et al., 2007. Possible role of semaphorin 3F, a candidate tumor suppressor gene
6371–6381. at 3p21.3, in p53-regulated tumor angiogenesis suppression. Cancer Res. 67,
Casazza, A., Finisguerra, V., Capparuccia, L., Camperi, A., Swiercz, J.M., Rizzolio, S., 1451–1460.
et al., 2010. Sema3E-Plexin D1 signaling drives human cancer cell invasiveness Gao, X., Tang, C., Shi, W., Feng, S., Qin, W., Jiang, T., et al., 2015. Semaphorin-3F
and metastatic spreading in mice. J. Clin. Invest. 120, 2684–2698. functions as a tumor suppressor in colorectal cancer due to regulation by DNA
Casazza, A., Fu, X., Johansson, I., Capparuccia, L., Andersson, F., Giustacchini, A., et al., methylation. Int. J. Clin. Exp. Pathol. 8, 12766–12774.
2011. Systemic and targeted delivery of semaphorin 3A inhibits tumor angio- Garcia-Areas, R., Libreros, S., Amat, S., Keating, P., Carrio, R., Robinson, P., et al., 2014.
genesis and progression in mouse tumor Models. Arterioscler. Thromb. Vasc. Semaphorin7A promotes tumor growth and exerts a pro-angiogenic effect in
Biol., 741–749. macrophages of mammary tumor-bearing mice. Front Physiol. 5, 17.
Casazza, A., Kigel, B., Maione, F., Capparuccia, L., Kessler, O., Giraudo, E., et al., Ge, C., Li, Q., Wang, L., Xu, X., 2013. The role of axon guidance factor semaphorin
2012. Tumour growth inhibition and anti-metastatic activity of a mutated furin- 6B in the invasion and metastasis of gastric cancer. J. Int. Med. Res. 41,
resistant Semaphorin 3E isoform. EMBO Mol. Med. 4, 234–250. 284–292.
Castellani, V., De Angelis, E., Kenwrick, S., Rougon, G., 2002. Cis and trans interactions Giordano, S., Corso, S., Conrotto, P., Artigiani, S., Gilestro, G., Barberis, D., et al., 2002.
of L1 with neuropilin-1 control axonal responses to semaphorin 3A. EMBO J. 21, The Semaphorin 4D receptor controls invasive growth by coupling with Met.
6348–6357. Nat. Cell Biol. 4, 720–724.
10 G. Neufeld et al. / Drug Resistance Updates 29 (2016) 1–12

Gitay-Goren, H., Cohen, T., Tessler, S., Soker, S., Gengrinovitch, S., Rockwell, P., et al., Kantor, D.B., Chivatakarn, O., Peer, K.L., Oster, S.F., Inatani, M., Hansen, M.J., et al.,
1996. Selective binding of VEGF121 to one of the three VEGF receptors of vascular 2004. Semaphorin 5A is a bifunctional axon guidance cue regulated by heparan
endothelial cells. J. Biol. Chem. 271, 5519–5523. and chondroitin sulfate proteoglycans. Neuron 44, 961–975.
Gitler, A.D., Lu, M.M., Epstein, J.A., 2004. PlexinD1 and Semaphorin signaling Karaman, S., Detmar, M., 2014. Mechanisms of lymphatic metastasis. J. Clin. Invest.
are required in endothelial cells for cardiovascular development. Dev. Cell 7, 124, 922–928.
107–116. Karpanen, T., Heckman, C.A., Keskitalo, S., Jeltsch, M., Ollila, H., Neufeld, G., et al.,
Glinka, Y., Prud’homme, G.J., 2008. Neuropilin-1 is a receptor for transforming 2006. Functional interaction of VEGF-C and VEGF-D with neuropilin receptors.
growth factor {beta}-1, activates its latent form, and promotes regulatory T cell FASEB J. 20, 1462–1472.
activity. J. Leukoc. Biol. 84, 302–310. Keck, P.J., Hauser, S.D., Krivi, G., Sanzo, K., Warren, T., Feder, J., et al., 1989. Vascular
Glinka, Y., Stoilova, S., Mohammed, N., Prud’homme, G.J., 2011. Neuropilin-1 exerts permeability factor, an endothelial cell mitogen related to PDGF. Science 246,
coreceptor function for TGF-beta-1 on the membrane of cancer cells and 1309–1312.
enhances responses to both latent and active TGF-beta. Carcinogenesis 32, Kessler, O., Shraga-Heled, N., Lange, T., Gutmann-Raviv, N., Sabo, E., Baruch, L., et al.,
613–621. 2004. Semaphorin-3F is an inhibitor of tumor angiogenesis. Cancer Res. 64,
Gluzman-Poltorak, Z., Cohen, T., Herzog, Y., Neufeld, G., 2000. Neuropilin-2 and 1008–1015.
Neuropilin-1 are receptors for 165-amino acid long form of vascular endothe- Kigel, B., Rabinowicz, N., Varshavsky, A., Kessler, O., Neufeld, G., 2011. Plexin-A4
lial growth factor (VEGF) and of placenta growth factor-2, but only neuropilin-2 promotes tumor progression and tumor angiogenesis by enhancement of VEGF
functions as a receptor for the 145 amino acid form of VEGF. J. Biol. Chem. 275, and bFGF signaling. Blood 118, 4285–4296.
18040–18045. Kigel, B., Varshavsky, A., Kessler, O., Neufeld, G., 2008. Successful inhibition of tumor
Goodman, C.S., Kolodkin, A.L., Luo, Y., Pueschel, A.W., Raper, J.A., 1999. Unified development by specific class-3 semaphorins is associated with expression of
nomenclature for the semaphorins collapsins. Cell 97, 551–552. appropriate semaphorin receptors by tumor cells. PLoS ONE 3, e3287.
Gospodarowicz, D., Abraham, J.A., Schilling, J., 1989. Isolation and characterization Kim, J., Oh, W.J., Gaiano, N., Yoshida, Y., Gu, C., 2011. Semaphorin 3E-Plexin-D1 sig-
of a vascular endothelial cell mitogen produced by pituitary-derived folliculo naling regulates VEGF function in developmental angiogenesis via a feedback
stellate cells. Proc. Natl. Acad. Sci. U. S. A. 86, 7311–7315. mechanism. Genes Dev. 25, 1399–1411.
Gu, C., Yoshida, Y., Livet, J., Reimert, D.V., Mann, F., Merte, J., et al., 2005. Semaphorin Klostermann, A., Lohrum, M., Adams, R.H., Puschel, A.W., 1998. The chemorepulsive
3E and plexin-D1 control vascular pattern independently of neuropilins. Science activity of the axonal guidance signal semaphorin D requires dimerization. J.
307, 265–268. Biol. Chem. 273, 7326–7331.
Guo, H.F., Li, X., Parker, M.W., Waltenberger, J., Becker, P.M., Vander Kooi, C.W., Kolodkin, A.L., Levengood, D.V., Rowe, E.G., Tai, Y.T., Giger, R.J., Ginty, D.D., 1997.
2013. Mechanistic basis for the potent anti-angiogenic activity of Semaphorin Neuropilin is a semaphorin III receptor. Cell 90, 753–762.
3F. Biochemistry 52, 7551–7558. Kumanogoh, A., Marukawa, S., Suzuki, K., Takegahara, N., Watanabe, C., Ch’ng, E.,
Gutmann-Raviv, N., Kessler, O., Shraga-Heled, N., Lange, T., Herzog, Y., Neufeld, G., et al., 2002a. Class IV semaphorin Sema4A enhances T-cell activation and inter-
2006. The Neuropilins and their roll in tumorigenesis and tumor progression. acts with Tim-2. Nature 419, 629–633.
Cancer Lett. 231, 1–11. Kumanogoh, A., Suzuki, K., Ch’ng, E., Watanabe, C., Marukawa, S., Takegahara, N.,
Guttmann-Raviv, N., Shraga-Heled, N., Varshavsky, A., Guimaraes-Sternberg, C., et al., 2002b. Requirement for the lymphocyte semaphorin, CD100, in the induc-
Kessler, O., Neufeld, G., 2007. Semaphorin-3A and Semaphorin-3F work together tion of antigen-specific T cells and the maturation of dendritic cells. J. Immunol.
to repel endothelial cells and to inhibit their survival by induction of apoptosis. 169, 1175–1181.
J. Biol. Chem. 282, 26294–26305. Kumanogoh, A., Watanabe, C., Lee, I., Wang, X., Shi, W., Araki, H., et al., 2000. Iden-
Hanahan, D., Folkman, J., 1996. Patterns and emerging mechanisms of the angiogenic tification of CD72 as a lymphocyte receptor for the class IV semaphorin CD100:
switch during tumorigenesis. Cell 86, 353–364. a novel mechanism for regulating B cell signaling. Immunity 13, 621–631.
He, H., Yang, T., Terman, J.R., Zhang, X., 2009. Crystal structure of the plexin A3 Kuroki, T., Trapasso, F., Yendamuri, S., Matsuyama, A., Alder, H., Williams, N.N., et al.,
intracellular region reveals an autoinhibited conformation through active site 2003. Allelic loss on chromosome 3p21.3 and promoter hypermethylation of
sequestration. Proc. Natl. Acad. Sci. U. S. A. 106, 15610–15615. Semaphorin 3B in non-small cell lung cancer. Cancer Res. 63, 3352–3355.
He, Z., Tessier-Lavigne, M., 1997. Neuropilin is a receptor for the axonal chemore- Kusy, S., Nasarre, P., Chan, D., Potiron, V., Meyronet, D., Gemmill, R.M., et al., 2005.
pellent Semaphorin III. Cell 90, 739–751. Selective suppression of in vivo tumorigenicity by semaphorin SEMA3F in lung
Hellstrom, M., Phng, L.K., Hofmann, J.J., Wallgard, E., Coultas, L., Lindblom, P., et al., cancer cells. Neoplasia 7, 457–465.
2007. Dll4 signalling through Notch1 regulates formation of tip cells during Lammie, G.A., Fantl, V., Smith, R., Schuuring, E., Brookes, S., Michalides, R., et al.,
angiogenesis. Nature 445, 776–780. 1991. D11S287, a putative oncogene on chromosome 11q13, is amplified and
Hirota, E., Yan, L., Tsunoda, T., Ashida, S., Fujime, M., Shuin, T., et al., 2006. Genome- expressed in squamous cell and mammary carcinomas and linked to BCL-1.
wide gene expression profiles of clear cell renal cell carcinoma: identification Oncogene 6, 439–444.
of molecular targets for treatment of renal cell carcinoma. Int. J. Oncol. 29, Leung, D.W., Cachianes, G., Kuang, W.J., Goeddel, D.V., Ferrara, N., 1989. Vascu-
799–827. lar endothelial growth factor is a secreted angiogenic mitogen. Science 246,
Holmes, S., Downs, A., Fosberry, A., Hayes, P., Michalovich, D., Murdoch, P., 1306–1309.
et al., 2002. Sema7A is a potent monocyte stimulator. Scand. J. Immunol. 56, Li, M., O’Sullivan, K.M., Jones, L.K., Semple, T., Kumanogoh, A., Kikutani, H., et al., 2006.
270–275. CD100 enhances dendritic cell and CD4+ cell activation leading to pathogenetic
Hota, P.K., Buck, M., 2012. Plexin structures are coming: opportunities for multilevel humoral responses and immune complex glomerulonephritis. J. Immunol. 177,
investigations of semaphorin guidance receptors, their cell signaling mecha- 3406–3412.
nisms, and functions. Cell Mol. Life Sci. 69, 3765–3805. Li, S., Li, Q., 2014. Cancer stem cells, lymphangiogenesis, and lymphatic metastasis.
Huang, S., Mills, L., Mian, B., Tellez, C., Mccarty, M., Yang, X.D., et al., 2002. Fully Cancer Lett. 357, 438–447.
humanized neutralizing antibodies to interleukin-8 (ABX-IL8) inhibit angiogen- Li, X., Lee, A.Y., 2010. Semaphorin 5A and plexin-B3 inhibit human glioma cell motil-
esis, tumor growth, and metastasis of human melanoma. Am. J. Pathol. 161, ity through RhoGDI{alpha}-mediated inactivation of Rac1 GTPase. J. Biol. Chem.
125–134. 285, 32436–32445.
Hung, R.J., Pak, C.W., Terman, J.R., 2011. Direct redox regulation of f-actin assembly Liu, H., Juo, Z.S., Shim, A.H., Focia, P.J., Chen, X., Garcia, K.C., et al., 2010. Structural
and disassembly by Mical. Science 334, 1710–1713. basis of Semaphorin-Plexin recognition and viral mimicry from Sema7A and
Hung, R.J., Spaeth, C.S., Yesilyurt, H.G., Terman, J.R., 2013. SelR reverses Mical- A39R complexes with PlexinC1. Cell 142, 749–761.
mediated oxidation of actin to regulate F-actin dynamics. Nat. Cell Biol. 15, Liu, H., Yang, Y., Xiao, J., Yang, S., Liu, Y., Kang, W., et al., 2014. Semaphorin 4D
1445–1454. expression is associated with a poor clinical outcome in cervical cancer patients.
Hung, R.J., Yazdani, U., Yoon, J., Wu, H., Yang, T., Gupta, N., et al., 2010. Mical links Microvasc. Res. 93, 1–8.
semaphorins to F-actin disassembly. Nature 463, 823–827. Lobov, I.B., Renard, R.A., Papadopoulos, N., Gale, N.W., Thurston, G., Yancopoulos,
Ikegami, R., Zheng, H., Ong, S.H., Culotti, J., 2004. Integration of Semaphorin-2A/MAB- G.D., et al., 2007. Delta-like ligand 4 (Dll4) is induced by VEGF as a negative
20, ephrin-4, and UNC-129 TGF-beta signaling pathways regulates sorting of regulator of angiogenic sprouting. Proc. Natl. Acad. Sci. U. S. A. 104, 3219–
distinct sensory rays in C. elegans. Dev. Cell 6, 383–395. 3224.
Ishida, I., Kumanogoh, A., Suzuki, K., Akahani, S., Noda, K., Kikutani, H., 2003. Involve- Loginov, V.I., Dmitriev, A.A., Senchenko, V.N., Pronina, I.V., Khodyrev, D.S., Kudryavt-
ment of CD100, a lymphocyte semaphorin, in the activation of the human seva, A.V., et al., 2015. Tumor suppressor function of the SEMA3B gene in human
immune system via CD72: implications for the regulation of immune and inflam- lung and renal cancers. PLOS ONE 10, e0123369.
matory responses. Int. Immunol. 15, 1027–1034. Love, C.A., Harlos, K., Mavaddat, N., Davis, S.J., Stuart, D.I., Jones, E.Y., et al., 2003. The
Janssen, B.J., Malinauskas, T., Weir, G.A., Cader, M.Z., Siebold, C., Jones, E.Y., 2012. ligand-binding face of the semaphorins revealed by the high-resolution crystal
Neuropilins lock secreted semaphorins onto plexins in a ternary signaling com- structure of SEMA4D. Nat. Struct. Biol. 10, 843–848.
plex. Nat. Struct. Mol. Biol. 19, 1293–1299. Luchino, J., Hocine, M., Amoureux, M.C., Gibert, B., Bernet, A., Royet, A., et al., 2013.
Janssen, B.J., Robinson, R.A., Perez-Branguli, F., Bell, C.H., Mitchell, K.J., Siebold, Semaphorin 3E suppresses tumor cell death triggered by the Plexin D1 depen-
C., et al., 2010. Structural basis of semaphorin-plexin signalling. Nature 467, dence receptor in metastatic breast cancers. Cancer Cell 24, 673–685.
1118–1122. Maione, F., Molla, F., Meda, C., Latini, R., Zentilin, L., Giacca, M., et al., 2009.
Jiang, H., Qi, L., Wang, F., Sun, Z., Huang, Z., Xi, Q., 2015. Decreased semaphorin 3A Semaphorin 3A is an endogenous angiogenesis inhibitor that blocks tumor
expression is associated with a poor prognosis in patients with epithelial ovarian growth and normalizes tumor vasculature in transgenic mouse models. J. Clin.
carcinoma. Int. J. Mol. Med. 35, 1374–1380. Invest. 119, 3356–3372.
Jurisic, G., Maby-El, H.H., Karaman, S., Ochsenbein, A.M., Alitalo, A., Siddiqui, S.S., Makinen, T., Olofsson, B., Karpanen, T., Hellman, U., Soker, S., Klagsbrun, M., et al.,
et al., 2012. An unexpected role of Semaphorin3A/Neuropilin-1 signaling in 1999. Differential binding of vascular endothelial growth factor B splice and
lymphatic vessel maturation and valve formation. Circ. Res. 111, 426–436. proteolytic isoforms to neuropilin-1. J. Biol. Chem. 274, 21217–21222.
G. Neufeld et al. / Drug Resistance Updates 29 (2016) 1–12 11

Man, J., Shoemake, J., Zhou, W., Fang, X., Wu, Q., Rizzo, A., et al., 2014. Sema3C Reidy, K.J., Villegas, G., Teichman, J., Veron, D., Shen, W., Jimenez, J., et al., 2009.
promotes the survival and tumorigenicity of glioma stem cells through Rac1 Semaphorin3a regulates endothelial cell number and podocyte differentiation
activation. Cell Rep. 9, 1812–1826. during glomerular development. Development 136, 3979–3989.
Mantovani, A., Allavena, P., Sica, A., Balkwill, F., 2008. Cancer-related inflammation. Rizzolio, S., Rabinoviz, N., Rainero, E., Lanzetti, L., Serini, G., Norman, J.C., et al., 2012.
Nature 454, 436–444. Neuropilin-1 dependent regulation of EGF-receptor signaling. Cancer Res. 72,
Mantovani, A., Sica, A., 2010. Macrophages, innate immunity and cancer: balance, 5801–5811.
tolerance, and diversity. Curr. Opin. Immunol. 22, 231–237. Rolny, C., Capparuccia, L., Casazza, A., Mazzone, M., Vallario, A., Cignetti, A., et al.,
Matsuoka, R.L., Chivatakarn, O., Badea, T.C., Samuels, I.S., Cahill, H., Katayama, K., 2008. The tumor suppressor semaphorin 3B triggers a prometastatic program
et al., 2011. Class 5 transmembrane semaphorins control selective Mammalian mediated by interleukin 8 and the tumor microenvironment. J. Exp. Med. 205,
retinal lamination and function. Neuron 71, 460–473. 1155–1171.
Matsushita, A., Gotze, T., Korc, M., 2007. Hepatocyte growth factor-mediated cell Sabag, A.D., Bode, J., Fink, D., Kigel, B., Kugler, W., Neufeld, G., 2012. Semaphorin-3D
invasion in pancreatic cancer cells is dependent on neuropilin-1. Cancer Res. 67, and Semaphorin-3E inhibit the development of tumors from glioblastoma cells
10309–10316. implanted in the cortex of the brain. PLoS ONE 7, e42912.
McClelland, L., Chen, Y., Soong, J., Kuo, I., Scott, G., 2011. Plexin B1 inhibits integrin- Sabag, A.D., Smolkin, T., Mumblat, Y., Ueffing, M., Kessler, O., Gloeckner, C.J., et al.,
dependent pp125(FAK) and Rho activity in melanoma. Pigment Cell Melanoma 2014. The role of the plexin-A2 receptor in Sema3A and Sema3B signal trans-
Res. 24, 165–174. duction. J. Cell Sci. 127, 5240–5252.
McMahon, S., Grondin, F., McDonald, P.P., Richard, D.E., Dubois, C.M., 2005. Hypoxia- Sadanandam, A., Sidhu, S.S., Wullschleger, S., Singh, S., Varney, M.L., Yang, C.S.,
enhanced expression of the proprotein convertase furin is mediated by hypoxia- et al., 2012. Secreted semaphorin 5A suppressed pancreatic tumour burden
inducible factor-1: impact on the bioactivation of proproteins. J. Biol. Chem. 280, but increased metastasis and endothelial cell proliferation. Br. J. Cancer 107,
6561–6569. 501–507.
Mendes-Da-Cruz, D.A., Brignier, A.C., Asnafi, V., Baleydier, F., Messias, C.V., Lepel- Saito, T., Kasamatsu, A., Ogawara, K., Miyamoto, I., Saito, K., Iyoda, M., et al., 2015.
letier, Y., et al., 2014. Semaphorin 3F and neuropilin-2 control the migration of Semaphorin7A promotion of tumoral growth and metastasis in human oral
human T-cell precursors. PLOS ONE 9, e103405. cancer by regulation of G1 cell cycle and matrix metalloproteases: possible
Migdal, M., Huppertz, B., Tessler, S., Comforti, A., Shibuya, M., Reich, R., et al., contribution to tumoral angiogenesis. PLOS ONE 10, e0137923.
1998. Neuropilin-1 is a placenta growth factor-2 receptor. J. Biol. Chem. 273, Sakurai, A., Gavard, J., nnas-Linhares, Y., Basile, J.R., Amornphimoltham, P., Palmby,
22272–22278. T.R., et al., 2010. Semaphorin 3E initiates anti-angiogenic signaling through
Miyato, H., Tsuno, N.H., Kitayama, J., 2012. Semaphorin 3C is involved in the pro- Plexin-D1 by regulating Arf6 and R-Ras. Mol. Cell Biol. 30, 3086–3098.
gression of gastric cancer. Cancer Sci. 103, 1961–1966. Sakurai, A., Jian, X., Lee, C.J., Manavski, Y., Chavakis, E., Donaldson, J., et al., 2011.
Morote-Garcia, J.C., Napiwotzky, D., Kohler, D., Rosenberger, P., 2012. Endothelial Phosphatidylinositol-4-phosphate 5-kinase and GEP100/Brag2 protein mediate
Semaphorin 7A promotes neutrophil migration during hypoxia. Proc. Natl. Acad. antiangiogenic signaling by Semaphorin 3E-Plexin-D1 through Arf6 protein. J.
Sci. U. S. A. 109, 14146–14151. Biol. Chem. 286, 34335–34345.
Mu, L., Wang, J., Chen, Y., Li, L., Guo, X., Zheng, S., et al., 2014. Hypoxia-inducible Segarra, M., Ohnuki, H., Maric, D., Salvucci, O., Hou, X., Kumar, A., et al., 2012.
factor-1alpha and semaphorin4D genes involved with tumor-associated Semaphorin 6A regulates angiogenesis by modulating VEGF signaling. Blood
macrophage-induced metastatic behavior and clinical significance in colon can- 120, 4104–4115.
cer. Chin Med. J. (Engl.) 127, 3568–3575. Sekido, Y., Bader, S., Latif, F., Chen, J.Y., Duh, F.M., Wei, M.H., et al., 1996. Human
Mumblat, Y., Kessler, O., Ilan, N., Neufeld, G., 2015. Full length semaphorin-3C func- semaphorins A(V) and IV reside in the 3p21.3 small cell lung cancer deletion
tions as an inhibitor of tumor lymphangiogenesis and tumor metastasis. Cancer region and demonstrate distinct expression patterns. Proc. Natl. Acad. Sci. U. S.
Res. 75, 2177–2186. A. 93, 4120–4125.
Nagai, H., Sugito, N., Matsubara, H., Tatematsu, Y., Hida, T., Sekido, Y., et al., 2007. Shirvan, A., Ziv, I., Fleminger, G., Shina, R., He, Z.G., Brudo, I., et al., 1999. Semaphorins
CLCP1 interacts with semaphorin 4B and regulates motility of lung cancer cells. as mediators of neuronal apoptosis. J. Neurochem. 73, 961–971.
Oncogene 26, 4025–4031. Siekmann, A.F., Covassin, L., Lawson, N.D., 2008. Modulation of VEGF signalling out-
Nair, P.N., McArdle, L., Cornell, J., Cohn, S.L., Stallings, R.L., 2007. High-resolution put by the Notch pathway. Bioessays 30, 303–313.
analysis of 3p deletion in neuroblastoma and differential methylation of the Sierra, J.R., Corso, S., Caione, L., Cepero, V., Conrotto, P., Cignetti, A., et al., 2008. Tumor
SEMA3B tumor suppressor gene. Cancer Genet. Cytogenet. 174, 100–110. angiogenesis and progression are enhanced by Sema4D produced by tumor-
Nakayama, H., Bruneau, S., Kochupurakkal, N., Coma, S., Briscoe, D.M., Klagsbrun, M., associated macrophages. J. Exp. Med. 205, 1673–1685.
2015. Regulation of mTOR signaling by Semaphorin 3F-Neuropilin 2 interactions Soker, S., Takashima, S., Miao, H.Q., Neufeld, G., Klagsbrun, M., 1998. Neuropilin-1
in vitro and in vivo. Sci. Rep. 5, 11789. is expressed by endothelial and tumor cells as an isoform specific receptor for
Nasarre, P., Constantin, B., Rouhaud, L., Harnois, T., Raymond, G., Drabkin, H.A., et al., vascular endothelial growth factor. Cell 92, 735–745.
2003. Semaphorin SEMA3F and VEGF have opposing effects on cell attachment Song, X., Zhang, W., Zhang, Y., Zhang, H., Fu, Z., Ye, J., et al., 2012. Expres-
and spreading. Neoplasia 5, 83–92. sion of semaphorin 3A and neuropilin 1 with clinicopathological features
Nasarre, P., Kusy, S., Constantin, B., Castellani, V., Drabkin, H.A., Bagnard, D., et al., and survival in human tongue cancer. Med. Oral Patol. Oral Cir. Bucal. 17,
2005. Semaphorin SEMA3F has a repulsing activity on breast cancer cells and e962–e968.
inhibits E-cadherin-mediated cell adhesion. Neoplasia 7, 180–189. Soong, J., Chen, Y., Shustef, E.M., Scott, G.A., 2012. Sema4D, the ligand for Plexin B1,
Negishi, M., Oinuma, I., Katoh, H., 2005a. Plexins: axon guidance and signal trans- suppresses c-Met activation and migration and promotes melanocyte survival
duction. Cell Mol. Life Sci. 62 (12), 1363–1371, 1363-1371. and growth. J. Invest. Dermatol. 132, 1230–1238.
Negishi, M., Oinuma, I., Katoh, H., 2005b. R-ras as a key player for signaling pathway Soong, J., Scott, G., 2012. Plexin B1 inhibits MET through direct association and
of plexins. Mol. Neurobiol. 32, 217–222. regulates Shp2 expression in melanocytes. J. Cell Sci. 126, 688–695.
Neufeld, G., Kessler, O., 2008. The semaphorins: versatile regulators of tumour pro- Stone, J., Itin, A., Alon, T., Peer, J., Gnessin, H., Chanling, T., et al., 1995. Development of
gression and tumour angiogenesis. Nat. Rev. Cancer 8, 632–645. retinal vasculature is mediated by hypoxia- induced vascular endothelial growth
Nogi, T., Yasui, N., Mihara, E., Matsunaga, Y., Noda, M., Yamashita, N., et al., 2010. factor (VEGF) expression by neuroglia. J. Neurosci. 15, 4738–4747.
Structural basis for semaphorin signalling through the plexin receptor. Nature Strieter, R.M., Kunkel, S.L., Elner, V.M., Martonyi, C.L., Koch, A.E., Polverini, P.J., et al.,
467, 1123–1127. 1992. Interleukin-8 – a corneal factor that induces neovascularization. Am. J.
Ochsenbein, A.M., Karaman, S., Proulx, S.T., Berchtold, M., Jurisic, G., Stoeckli, E.T., Pathol. 141, 1279–1284.
et al., 2016. Endothelial cell-derived semaphorin 3A inhibits filopodia formation Strongin, A.Y., 2010. Proteolytic and non-proteolytic roles of membrane type-1
by blood vascular tip cells. Development 143, 589–594. matrix metalloproteinase in malignancy. Biochim. Biophys. Acta 1803, 133–141.
Oinuma, I., Ishikawa, Y., Katoh, H., Negishi, M., 2004. The Semaphorin 4D receptor Suda, K., Guo, C., Oishi, A., Ikeda, S., Uemura, A., Yoshimura, N., 2014. Therapeutic
Plexin-B1 is a GTPase activating protein for R-Ras. Science 305, 862–865. potential of semaphorin 3E for the treatment of choroidal neovascularization.
Pan, G., Zhang, X., Ren, J., Lu, J., Li, W., Fu, H., et al., 2012. Semaphorin 5A, an axon guid- Invest Ophthalmol. Vis. Sci. 55, 4700–4706.
ance molecule, enhances the invasion and metastasis of human gastric cancer Sulpice, E., Plouet, J., Berge, M., Allanic, D., Tobelem, G., Merkulova-Rainon, T., 2007.
through activation of MMP9. Pathol. Oncol. Res. 19, 11–18. Neuropilin-1 and neuropilin-2 act as coreceptors, potentiating proangiogenic
Patnaik, A., Weiss, G.J., Leonard, J.E., Rasco, D., Sachdev, J.C., Fisher, T.L., et al., activity. Blood 111, 2036–2045.
2015. Safety, pharmacokinetics and pharmacodynamics of a humanized anti- Sun, T., Krishnan, R., Swiercz, J.M., 2012. Grb2 mediates semaphorin-4D-dependent
Semaphorin 4D antibody, in a first-in-human study of patients with advanced RhoA inactivation. J. Cell Sci. 125, 3557–3567.
solid tumors. Clin. Cancer Res. 22, 827–836. Suto, F., Ito, K., Uemura, M., Shimizu, M., Shinkawa, Y., Sanbo, M., et al., 2005. Plexin-
Perl, A.K., Wilgenbus, P., Dahl, U., Semb, H., Christofori, G., 1998. A causal role for a4 mediates axon-repulsive activities of both secreted and transmembrane
E-cadherin in the transition from adenoma to carcinoma. Nature 392, 190–193. semaphorins and plays roles in nerve fiber guidance. J. Neurosci. 25, 3628–3637.
Podgrabinska, S., Skobe, M., 2014. Role of lymphatic vasculature in regional and Suto, F., Tsuboi, M., Kamiya, H., Mizuno, H., Kiyama, Y., Komai, S., et al., 2007.
distant metastases. Microvasc. Res. 95C, 46–52. Interactions between Plexin-A2, Plexin-A4, and Semaphorin 6A control lamina-
Prahst, C., Heroult, M., Lanahan, A.A., Uziel, N., Kessler, O., Shraga-Heled, N., et al., restricted projection of hippocampal mossy fibers. Neuron 53, 535–547.
2008. Neuropilin-1/VEGFR-2 complexing requires the PDZ-binding domain of Suzuki, K., Okuno, T., Yamamoto, M., Pasterkamp, R.J., Takegahara, N., Takamatsu, H.,
neuropilin-1. J. Biol. Chem. 283, 25110–25114. et al., 2007. Semaphorin 7A initiates T-cell-mediated inflammatory responses
Puschel, A.W., 2007. GTPases in semaphorin signaling. Adv. Exp. Med. Biol. 600, through alpha1beta1 integrin. Nature 446, 680–684.
12–23. Swiercz, J.M., Kuner, R., Offermanns, S., 2004. Plexin-B1/RhoGEF-mediated RhoA
Rao, J., Zhou, Z., Yang, J., Shi, Y., Xu, S., Wang, B., et al., 2014. Semaphorin-3F sup- activation involves the receptor tyrosine kinase ErbB-2. J. Cell Biol. 165, 869–880.
presses the stemness of colorectal cancer cells by inactivating Rac1. Cancer Lett. Swiercz, J.M., Worzfeld, T., Offermanns, S., 2008. ERBB-2 and met reciprocally regu-
358, 76–84. late cellular signaling via plexin-B1. J. Biol. Chem. 283, 1893–1901.
12 G. Neufeld et al. / Drug Resistance Updates 29 (2016) 1–12

Takahashi, T., Fournier, A., Nakamura, F., Wang, L.H., Murakami, Y., Kalb, R.G., et al., Xiang, R., Davalos, A.R., Hensel, C.H., Zhou, X.J., Tse, C., Naylor, S.L., 2002. Semaphorin
1999. Plexin-neuropilin-1 complexes form functional semaphorin-3A receptors. 3F gene from human 3p21.3 suppresses tumor formation in nude mice. Cancer
Cell 99, 59–69. Res. 62, 2637–2643.
Takahashi, T., Strittmatter, S.M., 2001. PlexinA1 autoinhibition by the Plexin Sema Xiang, R.H., Hensel, C.H., Garcia, D.K., Carlson, H.C., Kok, K., Daly, M.C., et al., 1996.
domain. Neuron 29, 429–439. Isolation of the human semaphorin III/F gene (SEMA3F) at chromosome 3p21, a
Tamagnone, L., Artigiani, S., Chen, H., He, Z., Ming, G.I., Song, H., et al., 1999. Plexins region deleted in lung cancer. Genomics 32, 39–48.
are a large family of receptors for transmembrane, secreted, and GPI-anchored Xiong, G., Wang, C., Evers, B.M., Zhou, B.P., Xu, R., 2012. RORalpha suppresses breast
semaphorins in vertebrates. Cell 99, 71–80. tumor invasion by inducing SEMA3F expression. Cancer Res. 72, 1728–1739.
Tang, C., Gao, X., Liu, H., Jiang, T., Zhai, X., 2014. Decreased expression of SEMA3A is Xu, Y., Yuan, L., Mak, J., Pardanaud, L., Caunt, M., Kasman, I., et al., 2010. Neuropilin-
associated with poor prognosis in gastric carcinoma. Int. J. Clin. Exp. Pathol. 7, 2 mediates VEGF-C-induced lymphatic sprouting together with VEGFR3. J. Cell
4782–4794. Biol. 188, 115–130.
Terman, J.R., Mao, T., Pasterkamp, R.J., Yu, H.H., Kolodkin, A.L., 2002. MICALs, a family Yamamoto, M., Suzuki, K., Okuno, T., Ogata, T., Takegahara, N., Takamatsu, H., et al.,
of conserved flavoprotein oxidoreductases, function in plexin-mediated axonal 2008. Plexin-A4 negatively regulates T lymphocyte responses. Int. Immunol. 20,
repulsion. Cell 109, 887–900. 413–420.
Thacker, B.E., Seamen, E., Lawrence, R., Parker, M.W., Xu, Y., Liu, J., et al., 2016. Yang, W.J., Hu, J., Uemura, A., Tetzlaff, F., Augustin, H.G., Fischer, A., 2015.
Expanding the 3-O-sulfate proteome – enhanced binding of neuropilin-1 to Semaphorin-3C signals through Neuropilin-1 and PlexinD1 receptors to inhibit
3-O-sulfated heparan sulfate modulates its activity. ACS Chem. Biol. 11, 971–980. pathological angiogenesis. EMBO Mol. Med., e201404922.
Tomizawa, Y., Sekido, Y., Kondo, M., Gao, B., Yokota, J., Roche, J., et al., 2001. Inhibi- Yang, Y.H., Zhou, H., Binmadi, N.O., Proia, P., Basile, J.R., 2011. Plexin-B1 activates
tion of lung cancer cell growth and induction of apoptosis after reexpression of NF-kappaB and IL-8 to promote a pro-angiogenic response in endothelial cells.
3p21.3 candidate tumor suppressor gene SEMA3B. Proc. Natl. Acad. Sci. U. S. A. PLoS ONE 6, e25826.
98, 13954–13959. Yao, H.P., Zhou, Y.Q., Zhang, R., Wang, M.H., 2013. MSP-RON signalling in cancer:
Toyofuku, T., Nojima, S., Ishikawa, T., Takamatsu, H., Tsujimura, T., Uemura, A., et al., pathogenesis and therapeutic potential. Nat. Rev. Cancer 13, 466–481.
2012. Endosomal sorting by Semaphorin 4A in retinal pigment epithelium sup- Yu, W., Bai, Y., Han, N., Wang, F., Zhao, M., Huang, L., et al., 2013. Inhibition of patho-
ports photoreceptor survival. Genes Dev. 26, 816–829. logical retinal neovascularization by semaphorin 3A. Mol. Vis. 19, 1397–1405,
Toyofuku, T., Yabuki, M., Kamei, J., Kamei, M., Makino, N., Kumanogoh, A., et al., Print@2013, 1397–1405.
2007. Semaphorin-4A, an activator for T-cell-mediated immunity, suppresses Yuan, L., Moyon, D., Pardanaud, L., Breant, C., Karkkainen, M.J., Alitalo, K., et al.,
angiogenesis via Plexin-D1. EMBO J. 26, 1373–1384. 2002. Abnormal lymphatic vessel development in neuropilin 2 mutant mice.
Toyofuku, T., Yoshida, J., Sugimoto, T., Zhang, H., Kumanogoh, A., Hori, M., et al., 2005. Development 129, 4797–4806.
FARP2 triggers signals for Sema3A-mediated axonal repulsion. Nat. Neurosci. 8, Zacchigna, S., Pattarini, L., Zentilin, L., Moimas, S., Carrer, A., Sinigaglia, M., et al.,
1712–1719. 2008. Bone marrow cells recruited through the neuropilin-1 receptor promote
Toyofuku, T., Zhang, H., Kumanogoh, A., Takegahara, N., Suto, F., Kamei, J., et al., arterial formation at the sites of adult neoangiogenesis in mice. J. Clin. Invest.
2004. Dual roles of Sema6D in cardiac morphogenesis through region-specific 118, 2062–2075.
association of its receptor, Plexin-A1, with off-track and vascular endothelial Zhang, B., Gao, Z., Sun, M., Li, H., Fan, H., Chen, D., et al., 2014. Prognostic significance
growth factor receptor type 2. Genes Dev. 18, 435–447. of VEGF-C, semaphorin 3F, and neuropilin-2 expression in oral squamous cell
Trusolino, L., Bertotti, A., Comoglio, P.M., 2010. MET signalling: principles and func- carcinomas and their relationship with lymphangiogenesis. J. Surg. Oncol. 111,
tions in development, organ regeneration and cancer. Nat. Rev. Mol. Cell Biol. 382–388.
11, 834–848. Zhou, H., Binmadi, N.O., Yang, Y.H., Proia, P., Basile, J.R., 2012. Semaphorin 4D coop-
Vacca, A., Scavelli, C., Serini, G., Di, P.G., Cirulli, T., Merchionne, F., et al., 2006. Loss of erates with VEGF to promote angiogenesis and tumor progression. Angiogenesis
inhibitory semaphorin 3A (SEMA3A) autocrine loops in bone marrow endothe- 15, 391–407.
lial cells of patients with multiple myeloma. Blood 108, 1661–1667. Zhou, H., Wu, A., Fu, W., Lv, Z., Zhang, Z., 2014. Significance of semaphorin-3A
Varshavsky, A., Kessler, O., Abramovitch, S., Kigel, B., Zaffryar, S., Akiri, G., et al., 2008. and MMP-14 protein expression in non-small cell lung cancer. Oncol. Lett. 7,
Semaphorin-3B is an angiogenesis inhibitor that is inactivated by furin-like pro- 1395–1400.
protein convertases. Cancer Res. 68, 6922–6931. Zhou, H., Yang, Y.H., Basile, J.R., 2013. The Semaphorin 4D-Plexin-B1-RhoA signaling
Watanabe, C., Kumanogoh, A., Shi, W., Suzuki, K., Yamada, S., Okabe, M., et al., 2001. axis recruits pericytes and regulates vascular permeability through endothelial
Enhanced immune responses in transgenic mice expressing a truncated form of production of PDGF-B and ANGPTL4. Angiogenesis 17, 261–274.
the lymphocyte semaphorin cd100. J. Immunol. 167, 4321–4328. Zhu, L., Bergmeier, W., Wu, J., Jiang, H., Stalker, T.J., Cieslak, M., et al., 2007. Reg-
Worzfeld, T., Swiercz, J.M., Senturk, A., Genz, B., Korostylev, A., Deng, S., et al., 2014. ulated surface expression and shedding support a dual role for semaphorin
Genetic dissection of plexin signaling in vivo. Proc. Natl. Acad. Sci. U. S. A. 111, 4D in platelet responses to vascular injury. Proc. Natl. Acad. Sci. U. S. A. 104,
2194–2199. 1621–1626.

You might also like