Guidance molecules in lung cancer

2 downloads 0 Views 1MB Size Report
Oct 21, 2009 - lung mesenchyme while Dll1 expression is restricted to neuroen- docrine cells.76,77 Other genes of the Delta/Notch family such as. Dll4, Hey1 ...
Cell Adhesion & Migration 4:1, 130-145; January/February/March 2009; © 2009 Landes Bioscience

Guidance molecules in lung cancer Patrick Nasarre,1 Vincent Potiron,1 Harry Drabkin1 and Joëlle Roche1,2,* Medical University of South Carolina; Division of Hematology/Oncology; Charleston, SC USA; 2IPBC; Université de Poitiers; CNRS 6187; Poitiers, France

1

Key words: lung cancer, semaphorin, neuropilin, slit, robo, ephrin, netrin, DCC, UNC5, notch Abbreviations: Abl, abelson; ADD, addiction dependence domain; DAPK, death associated protein kinase; DCC, deleted in colon cancer; ECD, extracellular domain; EGF, epidermal growth factor; ERK, extracellular signal-regulated kinase; FAK, focal adhesion kinase; GAP, GTPase activating protein; GEF, guanine nucleotide exchange factor; GPI, glycosylphosphatidylinositol; Ig, immunoglobulin; LOH, loss of heterozygosity; NICD, notch intracellular domain; NRP, neuropilin; NSCLC, non-small cell lung cancer; PDZ, PSD95 discs large zonula occludens; PI3K, phosphoinositide 3-kinase; PSI, plexin semaphorin integrin; RTK, receptor tyrosine kinase; SAM, sterile alpha motif; SCLC, small cell lung cancer; TSG, tumor suppressor gene; UNC5, uncoordinated family member 5; VEGF, vascular endothelial growth factor

Guidance molecules were first described in the nervous system to control axon outgrowth direction. They are also widely expressed outside the nervous system where they control cell migration, tissue development and establishment of the vascular network. In addition, they are involved in cancer development, tumor angiogenesis and metastasis. This review is primarily focused on their functions in lung cancer and their involvement in lung development is also presented. Five guidance molecule families and their corresponding receptors are described, including the semaphorins/neuropilins/plexins, ephrins and Eph receptors, netrin/DCC/UNC5, Slit/Robo and Notch/Delta. In addition, the possibility to target these molecules as a therapeutic approach in cancer is discussed.

Introduction During normal development, timely and spatially-coordinated signals control cell migration, proliferation, fate and specification, thereby governing tissue arrangement and remodeling. Some of these signals are provided by membrane and extracellular cues that carry information to receptive cells in the environment. Such cues are guidance molecules, which administrate appropriate positioning of cell entities. This process is superbly exemplified by axon pathfinding in the nervous system where protrusions from a neuronal body navigate extreme distances to connect with specific neurologic targets.1 Similar guidance processes orchestrate the development of the vasculature network and the patterning of many if not all human tissues.2,3 Thus, it is not surprising if guidance molecules contribute to tumor growth, activation of the microenvironment and metastasis.4,5 We review here the main families of guidance molecules (Table 1) and focus on their participation in lung development and tumorigenesis.

Lung cancer is one of the most frequent malignant diseases. It ranks first in cancer-related deaths in Europe and the United States and is expected to rapidly increase in Asia. Tobacco exposure accounts for 90% of cases, although other factors such as asbestos, benzene and urban pollution are also associated with lung cancer incidence. In addition, lung cancer is an aggressive disease with a survival of less than 15% at 5 years. Non-small cell lung cancer (NSCLC) is more common than small-cell lung cancer (SCLC), accounting for about 80% of all lung cancers. It is mainly epithelial in origin and divided into three predominant types. Adenocarcinoma, the most frequent, has glandular differentiation or mucin production and is usually found in the outer part of the lungs. Squamous cell carcinoma shows keratinization or normal epithelium bridges and is usually found near bronchi. Large cell carcinoma, often with neuroendocrine features, can be found anywhere in the lungs. Frequent molecular alterations of NSCLC include mutations/deletions of FHIT, p53, p16, LKB1 and K-Ras genes, as well as overexpression/ mutation of EGFR or HER2 and increased telomerase activity. Allelic losses are frequently found in 3p, 6q, 8p, 9p, 13p, 17p and 19q. NSCLCs are currently treated by surgical resection, combination chemotherapy and radiation, with the more recent inclusion of EGFR inhibitors and VEGF antibodies (Bevacizumab). SCLC is a fast-growing and highly metastatic form of lung cancer with neuroendocrine origin. SCLCs have been traditionally staged as limited or extensive depending on their ability to be incorporated into a single radiation field. Frequent deletions/mutations are found in the FHIT, p53 and RB genes, while c-MYC and BCL2 genes are overexpressed. Telomerase activity is elevated and allelic 3p loss are found in 100% cases in addition to less frequent loss of 4p, 4q, 8p, 10q, 13q, 17p and 22q. Patients with SCLC do not undergo surgery, since subsequent relapse is almost inevitable, and therefore combination chemotherapy has been the predominant treatment.

*Correspondence to: Joëlle Roche; Email: [email protected] Submitted: 10/21/09; Accepted: 12/07/09 Previously published online: www.landesbioscience.com/journals/ celladhesion/article/10882

130

Cell Adhesion & Migration

Volume 4 Issue 1

special focus: lung cancer

review

Table 1. Canonical ligands and their corresponding major receptors in mammals Canonical ligand

Main receptor

Class 3 semaphorins: SEMA3(A to G)

Neuropilins (NRP1, NRP2), Plexin-A, Integrins

Semaphorins from class 4 to 7: SEMA4 to 7

Plexin-B/C/D

Ephrin-A(1 to 5)

EphA(1 to 8), EphA10

Ephrin-B(1 to 3)

EphB(1 to 4), EphB6

Netrin-1/2/4

DCC, neogenin, UNC5A-D or UNC5H1-H4

Netrin-G1

Unidentified

Netrin-G2

Unidentified

Slit1, Slit2, Slit3

Robo1 to 4

Delta-like (Dll-1/3/4)

Notch1 to 4

Serrate-like (JAG1, JAG2)

Notch1 to 4

This simplified list refers to receptors that ligands most commonly bind to; some exceptions exist. For instance, SEMA3E can bind plexin-D1 rather than NRP, or ephrin-B1 can bind EphA1.

Guidance Molecules: Structure, Pathway and Functions Semaphorins and their receptors. Semaphorins constitute a family of about 30 members. They are divided into eight classes based on structural features: vertebrate semaphorins belong to classes 3–7 and include 21 members. All semaphorins share an extracellular cysteine-rich “sema” domain, which is additionally found in their plexin receptors and the Met/Ron family receptors. Vertebrate semaphorins also possess a plexin-semaphorin-integrin (PSI) domain, and diverge by the presence of an immunoglobulin (Ig)-type domain or thrombospondin repeats. Their main difference however, resides in the C-terminus. Class-3 semaphorins (SEMA3A-G) have a basic region and are secreted proteins, classes 4–6 (SEMA4A-G; SEMA5A-B; SEMA6A-D) are transmembrane proteins with a cytoplasmic region and the single class-7 member (SEMA7A) is glycosylphosphatidylinositol anchored to the plasma membrane. Semaphorins bind and activate plexin receptors. Plexins are transmembrane proteins with the extracellular “sema” domain, PSI and Ig-type domains, followed by an intracellular RasGAP motif, and sometimes by a PSD95 discs large zonula occludens (PDZ) binding motif. Most often, SEMA4-7 bind directly to plexins, while class-3 semaphorins bind to neuropilins (NRP1 or NRP2) which transmit signals to plexins (Table 1). SEMA3E is an exception as this semaphorin binds directly plexin-D1 rather than NRP. In some instances, plexins are associated with receptor tyrosine kinases (RTK) like Met and ErbB2, or other membrane proteins.6 Notably, neuropilins bind both class-3 semaphorins and vascular endothelial growth factor (VEGF).7 In light of this finding, binding competition was proposed to explain some of the anti-angiogenic properties of class-3 semaphorins.8 However, it is still not clear if class-3 semaphorins simply compete with VEGF for NRP1 and NRP2 binding sites or whether they act

www.landesbioscience.com

independently of VEGF. NRP1 and 2 interact with VEGFR-1 and VEGFR-2, and NRP2 interacts also with VEGFR-3, to promote human endothelial cell survival and migration. NRP2 also binds to VEGF-C, a crucial player in lymphangiogenesis with a resulting enhancement of VEGF-C/VEGFR-3 biological effects. This suggests that semaphorin/NRP2 interactions play a role in lymphangiogenesis.9,10 Neuropilins also bind other ligands including placenta growth factor 2 (PlGF-2), fibroblast growth factor (FGF), galectin-1, hepatocyte growth factor (HGF) and transforming growth factor (TGFβ). In addition, NRPs are part of larger complexes that interact with Met and cell adhesion molecules like L1-CAM (L1 cell adhesion molecule).11 Semaphorins are expressed in the nervous, cardiovascular and immune systems, in many organs including lung and kidney, both in the embryo and the adult. They contribute to cell communication processes influencing cell positioning and behavior, and thus tissue morphogenesis. Secreted semaphorins function in a paracrine manner while membrane-bound semaphorins act through cell-cell interactions. However, some semaphorins from class 4 and 6 can be cleaved by proteases and released in the extracellular compartment. Semaphorins employ alternative signaling pathways with a recurrence for small G proteins (Fig. 1).12 To simplify, we can distinguish four main signaling axes. Secreted class-3 semaphorins bind to NRPs, which act via class-A plexins in two directions: either to regulate CRMP proteins and block tubulin assembly (Fig. 1A) or to inhibit R-Ras through plexin RasGAP activity resulting in integrin inactivation and decreased cell adhesion (Fig. 1B). In the same manner, membranous semaphorins can also inhibit R-Ras activity. Moreover, membranous semaphorins often regulate actin dynamics and cell spreading through upstream Rho regulators (GEF or GAP) depending on which semaphorin/plexin or co-receptor is present in the complex (Fig. 1B and C).13 Rho/ Rac pathways also participate in SEMA3 signaling.14 Finally, it is worth noting that some transmembranous semaphorins can signal to the cytoplasm in a reverse manner through their cytoplasmic domain. Examples of effectors in the later case are Abl and EVL, among others (Fig. 1C).15,16 In summary, semaphorins control cell protrusion, spreading and adhesion which, when combined, trigger strong migratory responses, most often in a repulsive manner but also attractive depending on the context.12 In addition, semaphorins can also regulate MAPK or PI3K signaling, proliferation and survival. Ephrins and their receptors. Eph receptors (for erythropoietin-producing human hepatocellular carcinoma) are the largest subgroup of receptor tyrosine kinases (RTK). Both Eph receptors and their ligands, Eph receptor interacting proteins (ephrins), are divided in two classes, A and B, based on sequence conservation and mutual interaction.17 In humans, there are fourteen Eph receptors with nine in class-A and five in class-B (Table 1). Eight members of ephrins belong to either the class-A or class-B. Structurally, Eph receptors are classical RTKs that possess an extracellular domain (ECD), a transmembrane section and cytoplasmic domain with tyrosine kinase activity and a PDZ-binding motif. Ephrin ligands all contain an extracellular receptor-binding domain but differ in their association

Cell Adhesion & Migration

131

Figure 1. Semaphorin signaling. Secreted and membranous semaphorins signal through redundant and alternative pathways. (A) SEMA3-mediated stimulation of plexin-A leads to GSK3-dependent phosphorylation of CRMP and inhibits microtubule assembly by CRMP. Class-3 semaphorins also inhibit Rho-mediated actin polymerization, ERK activation and R-Ras-mediated integrin activation. (B) Upon SEMA4-7 binding, the RasGAP domain of plexin-B/C/D inactivates R-Ras-mediated integrin signaling. This pathway is similarly involved in SEMA3-plexinA signaling. Also, plexin-B/C/D can activate or inhibit RhoA. RhoA regulation sometimes requires the association of the ErbB2 and Met RTK with plexins, which lead to activation of RhoGEF or RhoGAP, respectively. (C) SEMA4-6 reverse signal through their cytoplasmic domain to Abl and EVL to regulate actin dynamics. Gray/black box: R-RasGAP domain. White box: tyrosine kinase domain. Double arrows: proteolytic cleavage. (X) indicates inhibition of the corresponding pathway by SEMA3.

with the membrane. Ephrins from class-A are GPI-anchored, while ephrins from class-B are transmembrane and possess a short cytoplasmic domain with a PDZ-binding motif. Ephrins from class-A most often bind EphA receptors and vice versa, despite some exceptions (Table 1).17,18 Since both Eph and ephrins are membrane proteins, they must function in a proximal manner through the binding of ephrin ligands from one cell to Eph receptors on another (Fig. 2). Therefore, receptors

132

and ligands are often expressed at tissue boundaries by alternative cell types in juxtaposition. Eph/ephrins are well represented in the nervous system, the vasculature and virtually all organs where cell-cell interactions contribute to proper tissue morphogenesis. An interesting characteristic of the Eph/ephrin pair is the bi-directional signaling. Forward signaling from Eph receptors usually engages tyrosine kinase activity and is initiated by

Cell Adhesion & Migration

Volume 4 Issue 1

Figure 2. Ephrin signaling. Ephrins signal through reverse signaling and through Eph receptors. (A) reverse signaling occurs through focal adhesion kinase FAK, Abl, the Src family kinase Fyn, Axin and RasGAP. These converge to regulate cell migration through cytoskeleton rearrangement and adhesion. (B) Forward signaling from Eph receptors also engages Abl and RasGAP as well as Rho/Rac to regulate integrin-mediated migration. In addition, Ras downstream signaling controls MAPK and proliferation. Of note, Abl and Fyn signaling are targets of Dasatinib, which specifically inhibits proliferation of lung cancer cells with high expression of Eph receptors. White box: tyrosine kinase domain.

ligand-induced receptor clustering, which results in phosphorylation of their cytoplasmic tails.19 Of note, EphA10 and EphB6 are believed to be kinase defective.17 In addition, Eph receptors recruit a variety of adaptors, guanine nucleotide exchange factors (GEFs) and GTPase activating proteins (GAP), which regulate activity of small GTPases such as RhoA and Ras family members (Fig. 2B). Although there is strong promiscuity between signaling

www.landesbioscience.com

cascades, alternative adaptors and pathways can be used depending on the specific EphA or EphB. Reverse signaling occurs from class-A ephrins to Fyn and p120-RasGAP, among others, and it is speculated that an additional membrane-associated protein is necessary because of the absence of cytoplasmic tail on class-A ephrins. In contrast, ephrin-B cytoplasmic domains can be phosphorylated and signaling occurs through Axin, Abl, FAK and others (Fig. 2A). A recurring feature of Eph/ephrin signaling involves cytoskeleton dynamics, cell morphology and migration, which usually but not exclusively leads to repulsion. Also, Eph/ephrin signals act in concert with many RTKs, adhesion molecules, channels and membrane-associated kinases to regulate cell morphology and adhesion.20 Netrins and their receptors. Netrins belong to a family of evolutionarily conserved 60–80 kDa diffusible proteins that present structural similarity with the laminin family of extracellular matrix proteins. Three netrins are expressed in mammals in addition to two related GPI-linked membrane proteins, netrin-G1 and G2 (Table 1). Netrins regulate axon guidance in many organisms and are implicated in axonal outgrowth and orientation of cell migration in the developing nervous system.1 Netrins are bifunctional as they can mediate attraction or repulsion depending on the receptor they bind.21 They are also widely expressed outside the nervous system where they have key roles in branching, morphogenesis and angiogenesis. Netrin receptors include two main families of proteins: DCC (Deleted in Colon Cancer) including the paralog neogenin, and UNC5 (Uncoordinated family member 5 orthologues). In addition, the membrane-associated adenosine A2b receptor may be a DCC co-receptor.22,23 Also, several laminin-binding integrins (α6β4 and α3β1) can act as direct receptors for netrin-1 and as co-receptors with DCC.24,25 In C. elegans, this interaction regulates the stabilization or trafficking of UNC-40 (C. elegans homolog of DCC) during cell invasion and modulates integrin signaling.26 DCC and UNC5 are single-pass transmembrane receptors that possess extracellular immunoglobulin domains. The DCC extracellular domain contains six fibronectin type III-like repeats. Its cytoplasmic domain includes an addiction dependence domain (ADD). UNC5 receptors contain two thrombospondin-like repeats in the extracellular region, and a zonula occludens-1 and a death domain in the intracellular region. DCC and UNC5 can be cleaved by a caspase in their intracellular region.22 In 1998, Mehlen et al. provided evidence for the first time that DCC regulates apoptosis as a dependence receptor.27 These receptors induce programmed cell death in the absence of ligands, whereas in the presence of their trophic ligands, programmed cell death is inhibited and cell survival, invasion and metastasis are enhanced (Fig. 3). Apoptosis induced by DCC/UNC5 would not involve the classical mitochondrial intrinsic and death-receptor extrinsic apoptotic pathways (Fig. 3A). Yet, the pro-apoptotic signaling is still largely unknown.22,23 Upon netrin-1 binding, DCC/UNC5 form homodimers or multimers (Fig. 3B). This multimerization was recently shown to be sufficient to inhibit apoptosis.28 DCC can associate with

Cell Adhesion & Migration

133

Figure 3. Netrin signaling. The dependence receptor model is described. (A) In absence of netrin binding, DCC and UNC5 are cleaved in their intracellular domain by an active caspase. DCC cleavage leads to the exposure of the addiction dependence domain (ADD) upstream of the caspase cleavage site. This may facilitate the binding of caspase 9 in a complex that involves the adaptor protein APPL (also named DIP13α for DCC-interacting protein 13) to further allow caspase-3 activation. Contrary to ADD exposure in DCC, cleavage of UNC5H releases the death domain from the C-terminal region. This peptide might interact with the death associated protein kinase (DAPK), inducing apoptosis through the activation of caspase-3 and 9. It might also interact with the neurotrophin receptor interacting melanoma-associated antigen (MAGE) homolog NRAGE, inducing apoptosis through either inhibition of the caspase inhibitor XIAP or activation of c-JNK (c-JUN N-terminal kinase). (B) As a consequence of netrin-1 binding, DCC/UNC5 caspase cleavage is blocked. Activation of DCC might release the adaptor APPL, and DCC recruits multiple partners (not represented). Caspase-9 is not activated and mitochondrial-mediated apoptosis is blocked. In addition, DCC might trap caspase-3.22,23,164 UNC5B is a p53 target gene, although binding of netrin-1 inhibits p53-induced apoptosis. Association of netrin-1 to integrins modifies cell adhesion and involves small G proteins for actin reorganization. In addition to apoptosis inhibition, consequences of netrin-1 binding to DCC/UNC5B are regulation of cell motility, invasion and morphogenesis of the vascular system through cytoskeletal rearrangements. Gray box: ADD. Double arrows: proteolytic cleavage. Adapted from reference 22.

different partners, including focal adhesion kinase (FAK), the non-catalytic region of tyrosine kinase (Nck1) and phosphatidylinositol transfer protein α (PITPα). UNC5 is a p53 target gene but netrin-1 binding inhibits p53-induced apoptosis.29 As p53 is mutated in about 50% of cancers, UNC5B induction after cellular stress might be impaired. In a tumor context, some functions attributed to netrin-1 could be due, in part, to p53 mutations.30 Multiple pathways are also activated by netrins, including integrins, PI3K-Akt, ERK-1/2 and small GTPase Rac-1/Cdc42. One consequence is the regulation of cell motility, invasion and morphogenesis of the vascular system through cytoskeletal rearrangements. Interestingly, the DCC transmembrane domain is a substrate for g-secretase and the generated fragment (DCC-ICD) is translocated to the nucleus to activate transcription.31 These data indicate that DCC could perform dual roles, both as a cell surface receptor and as a transcriptional coactivator. Slit/Robo. First described in Drosophila, the secreted Slit ligand and its receptor, Robo, prevent developing neurons from inappropriately crossing the midline. Subsequently, a complete Slit/Robo family has been identified in several mammalian tissues (Table 1). Slits are large, secreted multidomain glycoproteins containing a leucine-rich repeat (LRR) that binds the immunoglobulin domain of Robos 1, 2 and 3.32 The human Robos are type I transmembrane receptors consisting of five immunoglobulin (Ig) domains, three fibronectin (Fn) type III repeats in the extracellular domain and four cytoplasmic conserved motifs, although some variations can be found for Robo4.32 Alternatively spliced forms of Robo1 and Robo4 have been identified. The main function of the Slit/Robo family is to regulate cell migration of neurons, leukocytes, endothelial cells and cancer cells (Fig. 4).

134

Several studies have described involvement of Slits and their receptors in angiogenesis.5 Evidence for a pro-angiogenic function comes from cell treatments with soluble Slit, the use of either a soluble extracellular domain of Robo1 (RoboN) or Robo4Fc to inhibit Slit, and by treatment with a monoclonal Robo1 antibody that blocks Robo-Slit interactions.33-35 Among Slit/Robo family members, Slit-2 may have an attractive role on Robo-1 expressing vessels.34 In addition, Robo4 can activate Cdc42 and Rac in endothelial cells leading to the formation of filopodia and lamellipodia.36 However, this mechanism does

Cell Adhesion & Migration

Volume 4 Issue 1

Figure 4. Slit signaling. Slit ligand binding to Robo receptors triggers several pathways that control actin reorganization and cell migration. The Rho family of GTPases appears to play a central role in Slit/Robo function. Part of the signaling involves the GAP proteins Vilse and srGAP (SLIT-ROBO Rho GTPase Activating Protein) that inhibit Rac and Cdc42 depending on the cellular context. The repulsive output of Robo1 signaling also involves the binding of Ena, an actin bundling protein which can be antagonized by Abl binding and phosphorylation. The protein Mena is involved in Robo4-mediated repulsion. Gray/black box: cytoplasmic conserved motifs (CC0-3).

not necessarily involve Slit2, and whether Cdc42 and Rac activation is due to direct binding to Robo4 or inhibition of a GAP is unclear. Contradictory results suggest an anti-angiogenic effect of Slit2. Indeed, when endothelial cells are pretreated with VEGF165, activation of Robo4 by Slit2 inhibits VEGF165-induced endothelial cell migration, tube formation, in vitro permeability and VEGF165-stimulated vascular leak in vivo by blocking Src family kinase activation.37 In this case, ERK and FAK signaling were inhibited, but the mechanistic details are still unknown. Slit/Robo have anti-migratory and pro-apoptotic properties. Indeed, Slit2/Robo1 was shown to counteract HGF-mediated migration by inhibiting Cdc-42 and stimulating Rac-1 in both transformed epithelial cells and cancer cells.38 As a consequence, Slit2 inhibited actin-based protrusive forces and increased the adhesive strength of cadherin-mediated intercellular contacts. This latter effect suggests that in some cancers Slit/Robo could interfere with the epithelial to mesenchymal transition (EMT). In addition, Slit2 inhibits SDF-1-induced chemotaxis of leukocytes

www.landesbioscience.com

in vitro through direct interaction between Robo1 with the SDF-1 receptor, CXCR4.39 Furthermore, Slit triggers RoboDCC interaction with the subsequent loss of responsiveness of DCC to its ligand, netrin-1.40 This loss may activate apoptotic pathways through caspases 3 and 9.41 Delta/Notch. The Notch pathway is an evolutionary conserved signaling system that is mandatory for proper embryonic development and functions to regulate tissue homeostasis and maintenance of stem cells in adults. Interestingly, a number of studies also suggest that Notch and Delta are involved in cell migration and act as guidance cues during nerve development. Indeed, the Notch pathway regulates intersegmental nerve guidance and turning during Drosophila embryonic development42 and is involved in the regulation of microtubule stability and cell migration in vertebrates.43-46 Based on structural homology to Drosophila ligands, the mammalian canonical DSL (Delta, Serrate, Lag2) ligands are Delta-like and Serrate-like Jagged1 (JAG1) and Jagged2 (JAG2) (Table 1).47 The canonical ligands are responsible for the majority of Notch signaling, but an increasing number of non-canonical ligands have also been described. We will focus only on the canonical ligands in lung development and cancer. DSL ligands are type 1 cell-surface proteins containing multiple tandem EGF repeats in their extracellular domains. The DSL domain together with the N-terminal domain and first two EGF repeats are required for DSL ligands to bind Notch. The intracellular regions of DSL ligands lack sequence homology. In mammals, four Notch family members have been identified (Notch1-4). All are single-pass transmembrane proteins that share structural homology and include a large number of extracellular EGF-like repeats followed by a cysteine-rich or LIN12 (LN) domain, a juxtamembrane region with specific proteinase cleavage sites a transmembrane domain including a cleavage site for g-secretase, and a cytoplasmic region that contains several functional domains. Notch signaling is intrinsically asymmetric and its activation requires interaction between a ligand-expressing cell and another cell expressing a Notch receptor (Fig. 5). Notch signal sending cells and receiving cells adopt distinct fates and regulate each other homeostatically. As a consequence, Notch signaling regulates organogenesis and tissue differentiation through three types of developmental events: lateral inhibition, lineage specification and boundary formation.48 In addition, Notch signaling is important during pre- and post-natal life to maintain stem cell viability and alteration of Notch function has been associated with hereditary diseases and several cancers. Guidance Molecules in Lung Development Similar to nerves and vessels, the lung develops by successive branching, generating an architecture of growing complexity until formation of terminal bronchioles (Fig. 6). A push-pull model has been proposed, whereby guidance cues act in concert with other critical growth and morphogenetic factors to sculpt the architecture of the respiratory tree.2

Cell Adhesion & Migration

135

Semaphorins and their receptors. Semaphorins are expressed and function in the development or maintenance of multiple organs and physiological systems such as the central and peripheral nervous systems, cardiovascular network, lung, kidney, muscle, bone morphogenesis and immune surveillance. Few studies have directly addressed the function of semaphorins in lung morphogenesis. However, in mice, Sema3A is expressed in the mesenchyme and restrains lung budding while Sema3C and Sema3F in the epithelial compartment promote lung branching and support the development of a mature arborescence.49,50 Accordingly, we observed expression of SEMA3F at the membrane of type I and II epithelial cells in normal human lungs.51 In murine lungs, Sema3A, 3C and 3F are expressed at specific locations and times during development, and this seems to correlate with concomitant regulation of their receptors NRP1, NRP2 and plexin-A1. Semaphorins also participate in lung vasculogenesis and angiogenesis. Ephrins and their receptors. Almost all the Eph receptors and ephrin ligands are normally expressed during lung development, but their specific role has not been well characterized. Ephrin-B2, important during lung development, is expressed in branching epithelial buds and distal epithelia in embryonic stages and later localizes to microvascular beds from birth to adultFigure 5. Notch signaling. Notch is presented to its ligand as a heterodimer resulthood.52 Mice with mutant ephrin-B2 lacking the PDZing from processing by a furin-like protease during transit to the plasma membinding domain, show changes in extracellular matrix brane.165 Ligand binding triggers intra-membrane regulated cleavage of Notch composition which lead to reduced secondary septation within the juxtamembrane and transmembrane domains, ultimately leading to and the formation of enlarged alveolae. Ephrin-B2 recepthe release of the Notch intracellular domain (NICD). NICD then translocates to the tors EphA4, B2 and B4 are simultaneously expressed in nucleus where it directly interacts with the CSL (CBF1, Su(H), LAG1) transcription factor. The transcriptional repressor CoR is released. Recruitment of co-activators, the vasculature. In contrast, EphB3 was found in interincluding Mastermind, turns on expression of Notch target genes. These targets stitial cells and loss of function induced disorganized include a set of transcriptional repressors (Hes1, Hes5, Hey1, Hey2 and HeyL) of the matrix. For several other ephrins, not much more than basic helix-loop-helix (bHLH) family that repress expression of other bHLH genes expression data are available. Ephrin-A1, B1, B2 and involved in cell differentiation like the ubiquituous E2A factors and Mash1 (mamB3 are expressed in fetal and/or adult lungs.53-59 Eph malian achaete-scute homolog-1), MyoD and others. The ultimate consequence is the regulation of cell fate and differentiation status. Gray box: NICD. receptors EphA1, A2, A3 and A4, EphB2 and B4 transcripts are observed at moderate to high levels in lung tissues.53,60 However, EphA8 and EphB1 were not found in this study. to associate with microvessels.65 EphB4 negatively regulates vesPerhaps most interesting, Eph/ephrins are well-established sel branching and permeability and promotes circumferential regulators of vasculature development and angiogenesis. Since growth.66 Conversely, EphA2 increases lung blood vessel perthe endothelial-epithelial interface is critical to oxygen assimi- meability.67 More information about the role of these molecules lation in distal airways, Eph-ephrin contribution to lung vas- and other guidance molecules in vasculature has been published culature also may regulate lung tissue morphogenesis. Indeed, elsewhere.2,68 Eph/ephrins are expressed in the blood vessel compartment of Netrins and their receptors. Netrin-1 and its receptors control the lung and among them, ephrin-A1, B1, B2 and EphB4 are morphology of endothelial cells and vascular smooth muscle cells. among the most enriched in alveolar capillary endothelial cells They are implicated in the reorganization of the cytoskeleton, as in mice.61 However, few specific functions in lung vasculariza- well as epithelial cell adhesion and migration in lungs, mammary tion have been reported. In angiogenesis, ephrin-B2 expression glands and pancreas.2 Netrins are expressed during early stages of is restricted to arteries or neovascularization sites whereas EphB4 murine embryonic development and their localization becomes expression is restricted to veins.62,63 In developing vessels of the restricted to the epithelium of large airways at embryonic day 18 lung epithelium, there is co-expression of ephrin-B2 and EphB4, (E18).69 They are located at the basement membrane and/or bind which suggests a temporary lack of specification in early lung locally to epithelial cells at the neck region of elongating endoderm embryogenesis.64 Later, smooth muscle cells utilize ephrin-B2 buds to restrict ectopic budding.70 DCC expression peaks early in

136

Cell Adhesion & Migration

Volume 4 Issue 1

Figure 6. Lung development. Human lung development is organized in five distinct periods.166 The first embryonic period (weeks 3–7) leads to the formation of two lung buds at the ventral-lateral aspect of the foregut, followed by the formation of the major bronchi and division of tracheal-esophageal tube. During the second pseudoglandular period (weeks 5–17), bronchial branches, acinar tubules and buds proliferate, and vasculogenesis and innervation occur. Later (weeks 16–26), in the canalicular period, pulmonary vascular bed and acinus organize and innervation increases. Then, the saccular period (weeks 24–38) is characterized by dilation of peripheral airspaces, differentiation of the respiratory epithelium, an increased vascularity of the saccules and surfactant synthesis. Finally, growth and septation of alveoli, and maturation of the pulmonary vascular system take place from week 38 to maturity (alveolar period).

development with a subsequent switch from localization in the mesenchyme to the epithelium in late development. UNC5H2 is expressed late in development, first localized to both basal and apical surfaces of airway epithelium while later the expression is apical only.69 Netrins modulate the morphogenetic response of lung endoderm to exogenous fibroblast growth factors (FGFs). This effect involves inhibition of localized changes in cell shape and decrease of local ERK1/2 activity. According to this model, the basal lamina plus netrins acts as a kind of “corset,” restricting morphogenesis of the merging bud. Slit/Robo. The first evidence for involvement of the Slit/ Robo family in lung development comes from a Robo1 knockdown mouse model.71 Heterozygous mice born at the expected frequency have no obvious phenotype. However, newborn Robo1-/- mice are usually inactive with labored breathing and 63% die within the first 24 h after birth. Appearance of the lung structures is consistent with a developmental delay. In addition, in an in vitro model using co-culture of endothelial cells and pulmonary epithelial cells on reconstituted basement membrane, Slit3 expression is upregulated during the alignment process of airway epithelium with endothelium.72 Altogether, these results suggest that Slit3 and Robo1 may be key players involved in the maturation of lung tissue during embryonic development. The expression levels and localization of Slit2, Slit3, Robo1 and Robo3 proteins have been studied throughout lung development in embryonic mice and early postnatal life.72-74 However, none of these studies really addressed the exact role of Slit/Robo during lung development.

www.landesbioscience.com

Delta/Notch. Quantitative expression studies from the developing mouse lung demonstrate a progressive increase of Notch and Notch ligands from E11.5 to adulthood. All Notch molecules are expressed in the lung, Notch 4 being endothelial specific.75-79 Interestingly, by inhibiting g-secretase, Notch signaling was shown to be unnecessary for lung bud initiation, but rather required to maintain a balance of proximal-distal cell fates in early lung development stages.80 Regarding Notch ligands, JAG1 and JAG2 are expressed in lung mesenchyme while Dll1 expression is restricted to neuroendocrine cells.76,77 Other genes of the Delta/Notch family such as Dll4, Hey1, Jagged2, Notch1, Numb and Siah1b are also expressed in lung capillaries, further supporting a role for Notch signaling in angiogenesis.61 Early lethality of mice with homozygous deletions for Notch1, Notch2, Dll1 and Jagged1 has limited the assessment of these genetic alterations in lung development, forcing researchers to look towards alterations of Notch targets. Among those targets, Hes1 is expressed in Notch1 and Notch3 positive nonendocrine airway epithelial cells, and Hes1-reactive cells are destined to become Clara cells.76 Furthermore, Hes1 appears to be necessary for mediating the inhibition of neuroendocrine phenotype. Compared to Hes1, Mash1 expression is restricted to neuroepithelial bodies and pulmonary neuroendocrine cells (PNEC), and Mash1 appears to be essential for the expression of Dll1 and the differentiation of PNECs in the developing lung.76,81 Other Notch/Delta targets such as HeyL, Hey1 and Hey2 have been identified in the developing lung but their

Cell Adhesion & Migration

137

function remains unknown.82,83 Notch signaling can also affect the immune response in adult and alteration of this pathway has been implicated in some lung diseases such as asthma, “Ondine’s curse” congenital hypoventilation syndrome, chronic obstructive pulmonary disease and inflammation due to microbial infection.84-87 Guidance Molecules in Lung Cancer Semaphorins and their receptors. Detailed reviews regarding the involvement of semaphorins and neuropilins in different types of cancer, including lung, have been recently published.6,88-91 Because of space, we refer the readers to these reviews. The first evidence suggesting semaphorin involvement in cancer was the cloning of SEMA3B and SEMA3F from the chromosome 3p21.3 region, which undergoes frequent loss of heterozygosity (LOH) and less frequent homozygous deletion in lung cancer (Table 2).92-94 Since then, most semaphorin studies in lung cancer have focused on these two SEMA3s, which are often downregulated by allelic loss of one allele and hypermethylation of the second. Also, SEMA3B and SEMA3F are regulated by the p53 tumor suppressor, which is frequently mutated or lost in cancer. Furthermore, SEMA3F is a target of ZEB-1, a transcriptional repressor involved in the epithelial to mesenchymal transition (EMT).95 In human lung tumors, SEMA3F is lost or delocalized in the cytoplasm, whereas in normal tissues it is found at the membrane of epithelial cells.51 In addition, loss of SEMA3F correlates with increased membrane VEGF staining and higher tumor grade. In contrast to the loss of SEMA3F, NRP1 and NRP2 levels increase from dysplasia to microinvasive carcinoma and correlate with VEGF expression.96 Both SEMA3B and SEMA3F have demonstrated tumor-suppressor activity in vitro and in experimental xenograft models. For instance, SEMA3B transfection inhibited lung cancer cell growth and was associated with apoptosis and PI3K signaling inhibition.97 This SEMA3B growth inhibitory effect is exerted through induction of insulin-like growth factor-binding protein-6 (IGFBP-6) which is considered to be a TSG.98 One new finding is that while SEMA3B inhibits primary tumor growth, it may be associated with increased metastasis, since it leads to increased IL-8 expression and recruitment of tumor-associated macrophages.99 Effective lung tumor suppression by SEMA3F has been reported by several studies. One hypothesis is that SEMA3F inhibits tumor-associated angiogenesis. Also, SEMA3F reexpression has multiple inhibitory effects in lung cancer cells including lower integrin activation, reduced MAPK signaling and loss of both HIF-1α expression and VEGF secretion. While much attention has been placed on SEMA3B and 3F, other semaphorins or partners may contribute to lung tumorigenesis. For example, SEMA3C is upregulated in lung cancer cells with higher metastatic capability. SEMA4B promotes migration and invasion, and its receptor, CLCP1 (a NRP-like protein), is upregulated in metastatic lung cancer cells. SEMA4D and its receptor, plexin-B1, are overexpressed in lung cancer.

138

In contrast, SEMA6A, which has anti-angiogenic properties, is downregulated in some cancer cell lines. One cause of this downregulation is LOH at 5q23.1 where SEMA6A is localized. Expression of the VEGF/SEMA3 receptor, NRP1, correlates with poor survival in lung cancer and coexpression of NRP1 and 2 is associated with tumor vascularization. In the semaphorin pathway, it was reported that the expression of LCRMP-1, the long form of collapsin response mediator protein-1, is associated with clinical outcome and lymph node metastasis in NSCLC patients.100 Ephrins and their receptors. The EPH receptors and ephrin ligands have been implicated in multiple cancers and their expression is deregulated by oncogenic signals, microenvironment changes and inflammation. Special attention has been focused on EPH/ephrins in colorectal, pancreatic, ovarian and breast cancers, melanoma and glioblastoma and is reviewed elsewhere.17 In lung cancer, EPHB2 is overexpressed in a subset of adenocarcinoma while no membranous expression was observed in normal lungs.101 In SCLC cell lines and tumor specimens, expression of EPHB1, B2, B3, B4 and B6 was detected with EPHB2 being the most prominent, although it is not clear how these levels relate to normal tissues. Also, ephrin-B1, B2 and B3 were expressed, suggesting that autocrine loops may be activated. In advanced NSCLC tumor stages, EPHA2 expression is increased and correlates with survival. In lung cancer, ephrin-A3 expression was found to be upregulated 26-fold compared to benign tissue. One mechanistic suggestion is that EPHB6 activates MAPK signaling in lung adenocarcinoma.102 MAPK regulation by EPH/ephrins is common in other cancers, and we can only speculate that other EPH receptors exploit this signaling pathway in lung cancer. Signaling from the PDZ-binding domain of ephrin-B may also be involved, since it is known to play a role in normal lung cells. One mechanism by which NSCLC tumor cells overexpress EPH/ephrins is chromosomal gain. Increased gene copy numbers of EPHA3, A5 and A8 have been reported in NSCLC and EPHA3 silencing by shRNA reduced the viability of cells displaying gene amplifications (Table 2).103 Strikingly, ten EPH receptor genes are mutated at moderate-high frequency in lung adenocarcinoma, notably EPHA3 and EPHA5, plus EPHA7, B1 and B6.104 Mutations arise in the extracellular domain, presumably affecting ligand binding, and in the kinase domain sometimes at a speculated molecular breakpoint. Overall, 16% of adenocarcinomas have mutations or amplifications in the EPH pathway, supporting the notion that EPHs might be proto-oncogenes. Finally, a few studies have focused on mesothelioma, a particular type of lung cancer associated with asbestos exposure. EPHA2 seems to be more expressed in malignant mesothelioma than in normal pleural mesothelial cells and gene silencing reduces cell growth.105 Moreover, stimulation by ephrin-A1 decreases EPHA2 levels and mesothelioma growth.105 About 26% of mesotheliomas have fair to moderate expression of EPHB2, but the significance is unknown.101 Another way by which EPH/ephrins may contribute to lung tumorigenesis is their capacity to control tumor angiogenesis. For example, EPHB4 and ephrin-B2 participate in postnatal

Cell Adhesion & Migration

Volume 4 Issue 1

Table 2. Chromosomal localization of human guidance ligands and their major receptors Chromosome

Localization

Gene

1

1p36 1p36.1-p35 1p36.12 1p34.3 1p13-p11 1q21.2 1q21-q22 1q22 1q32.2

EPHA2 EPHB2 EPHA8 EPHA10 NOTCH2 SEMA6C Ephrin-A1, -A3, -A4 SEMA4A PLXNA2

2

2p13.1 2q11.2 2q33.3 2q36.1

SEMA4F SEMA4C NRP2 EPHA4

3

3p21.31 3p21.3 3p21.1 3p12.3 3p12 3p11.2 3q11.2 3q21.1 3q21.3 3q22.1 3q21-23 3q21-qter

PLXNB1 SEMA3F, SEMA3B SEMA3G ROBO2 ROBO1 EPHA3 EPHA6 SEMA5B PLXNA1 PLXND1 EPHB1 EPHB3

4

4p15.2 4q13.1 4q21-q23

SLIT2 EPHA5 UNC5C

5

5p15.2 5q21 5q23.1 5q35 5q35.2

SEMA5A Ephrin-A5 SEMA6A SLIT3 UNC5A

6

6p21.3 6q16.1 6q27

NOTCH4 EPHA7 DLL1

7

7p12.1 7q21.11 7q21-q31 7q22 7q32.3 7q34 7q33-q35

SEMA3A SEMA3D, SEMA3E SEMA3C EPHB4 PLXNA4 EPHA1 EPHB6

8

8p12

UNC5D

9

9q22.2 9q34.3

SEMA4D NOTCH1

10

10p12 10q22.1 10q23.3-q24 10q24.31

NRP1 UNC5B SLIT1 SEMA4G

11

11q24.2

ROBO3, ROBO4

12

12q22-q23 12q23.3

Netrin-4 PLXNC1

13

13q33

Ephrin-B2

14

14q32

JAG2

www.landesbioscience.com

Table 2. Chromosomal localization of human guidance ligands and their major receptors

15

15q14 15q21.1 15q22.3-q23 15q25

DLL4 SEMA6D SEMA7A SEMA4B

16

16p13.3

Netrin-3

17

17p13-p12 17p13.1-p11.2

Netrin-1 Ephrin-B3

18

18q21.3

DCC

19

19q13 19p13.2-p13.1 19p13.3

DLL3 NOTCH3 SEMA6B

20

20p12.1-p11.23

JAG1

22

22q13.33

PLXNB2

X

Xq12 Xq28

Ephrin-B1 PLXNA3, PLXNB3

and neoplastic angiogenesis, suggesting that normally extinct embryonic EPH/ephrin pathways are reactivated by tumor cells to regulate neoangiogenesis.66 Ephrin-A1 and EPHA2 have been detected in the vasculature of surgically resected human tumors and are essential for maximal effects of VEGF.106 Netrins and their receptors. DCC was identified in 18q21.1, a region of common LOH in colon cancer as well as in many cancers (Table 2).107 It was first suggested that DCC was a tumor suppressor gene. However, this hypothesis was questioned for several reasons, i.e., DCC mutations are rare, DCC mutant mice were not predisposed to cancers and loss of DCC had not been shown to give a selective advantage to tumor cells. Furthermore, SMAD2 and SMAD4, two other tumor suppressor candidates, are also localized in this region affected by LOH and SMAD mutations were frequently identified in pancreatic cancer. However, a number of experiments including forced DCC expression and DCC inhibition have documented roles of DCC as a tumor suppressor gene.22 Over the past decade, evidence has accumulated indicating that netrin-1 and its receptors have an important role in tumor biology. Loss of the apoptotic activity of dependence receptors is advantageous for tumor cell survival and could be achieved by three different mechanisms: loss of the receptors, overexpression of the ligand or inhibition of downstream signaling. The first of these, loss or reduced expression of DCC/UNC5H, was reported in colorectal cancers in addition to various other cancers either through genetic or epigenetic processes.23,107,108 In a panel of 25 lung carcinomas with corresponding adjacent normal tissues, DCC expression was decreased in tumors but netrin-1 expression was increased.109 However, at least two of the UNC5H receptors were always present in tumors with high netrin-1 expression. Secondly, overexpression of netrin-1 was reported in metastatic breast cancer and colon cancer where it confers a selective advantage for tumor cell survival and invasion.110,111 In addition, netrin-1 overexpression was associated with a worse outcome in poorly differentiated pancreatic adenocarcinoma.112 In lung cancer, 92 NSCLC tumors were examined by immunohistochemistry

Cell Adhesion & Migration

139

for netrin-1 expression.109 Netrin-1 was absent or low in normal bronchial and alveolar epithelial cells, whereas both adenocarcinomas and squamous carcinomas expressed high levels of netrin-1, which like in the embryonic pattern was localized in the cytoplasm and in both apical and basal membranes. No statistically significant differences were observed at different stages, but netrin-1 expression was more frequent and more intense in adenocarcinomas than squamous cell carcinomas. In situ hybridization indicated that netrin-1 is not expressed in the stroma cells but is expressed in tumor cells. By either inhibiting netrin-1 expression or its interaction with receptors in two lung cancer cell lines, apoptosis was demonstrated to be dependent on UNC5H1 and UNC5H2 receptors but not DCC.109 In addition, blocking caspase-9 resulted in inhibition of apoptotic activity by the UNC5H receptors. The netrin-1 gain of expression in tumor cells of epithelial origin may have two additive effects in an autocrine manner. Inhibiting dependence receptor-induced cell death confers a selective advantage to tumor cells. In addition, it would favor blood vessel maintenance and development, as netrin-1 was shown to control survival of endothelial cells and promote angiogenesis despite apparently contradictory observations resolved by the dependence receptor hypothesis.113-116 The third mechanism to inhibit apoptotic activity is downregulation of DAPK, the downstream effector of UNC5H2-induced cell death.117 In the lung, aggressive metastatic mouse carcinoma clones did not express DAPK in contrast to their low-metastatic counterparts. DAPK restoration in highly-metastatic Lewis lung carcinoma cells suppressed their ability to form lung metastases and delayed local tumor growth.118 The authors proposed a model where loss of DAPK expression provides a unique mechanism that links suppression of apoptosis to metastasis. DAPK can be also inhibited by DNA methylation in lung cancer.119 Slit/Robo. Slit/Robo are suspected to have pro-tumoral and pro-angiogenic functions in different cancer models.34,120 However, a Robo1-/- mouse model suggested that Slit/Robo signaling functions as tumor suppressor in lung cancer. In this model, two-thirds of the mice that survived to adulthood showed early signs of morbidity. At autopsy, bronchial epithelial hyperplasia and focal dysplasia were observed in most mice.71 In human lung cancer, Slit/Robo expression is often reduced either because of LOH or inactivation by DNA methylation. Of note, ROBO1, SLIT2 and SLIT3 are located in 3p12.3,121,122 4p15.2123,124 and 5q35-q34125 respectively and these locations are affected by homozygous deletions or frequent LOH in lung cancer (Table 2). However, no substantial frequency of mutations was found for either ROBO1 or SLIT2.71,121 Methylation of the ROBO1 promoter and SLIT3 CpG islands are also rare events in lung cancer, but SLIT2 promoter methylation occurs in almost one-half of lung and breast primary tumors.126 Nevertheless, no experimental data clearly prove that Slit/Robo are TSGs in lung cancer. The fact that ROBO4 levels, as measured in the serum of patients with advanced NSCLC, correlated with poor survival127 is unconvincing, since it is not known if the circulating ROBO4 represented a degraded form, and whether its increased levels correlate with impaired angiogenesis in tumors.

140

Delta/Notch. Notch signaling in lung cancer exhibits properties suggesting both tumor promotion and inhibition, depending on the cell type. Notch1 and Notch2 are frequently expressed in NSCLC; however, the role of Notch1 in lung cancer remains obscure. On one hand, Notch1 inhibits growth of A549 adenocarcinoma cells in vitro and in vivo.128 On the other, hypoxia upregulates Notch1 and sensitizes cells to small molecule g-secretase inhibitors.129 Notch2 is frequently overexpressed in lung cancers as the result of genetic alterations affecting chromosome 1p (Table 2).130 Notch3 mRNA expression was detected in 7 of 25 NSCLC but not in SCLC lines.131,132 In addition, Notch3 is the target of chromosome 19 translocations in NSCLCs. Interestingly, inhibition of Notch3 increases growth factor dependence sensitivity to EGF receptor inhibitors and is associated with a reduced phosphorylation of MAPK.132 The g-secretase inhibitor, MRK-003, also inhibits Notch3 activation and induces apoptosis of lung tumor cells in vitro and in vivo.133 Together, these results suggest that at least Notch2 and Notch3 may have pro-tumor capabilities in NSCLC. Notch1 is rarely expressed in SCLC, whereas a subset of SCLC exhibits Notch2 expression. In contrast to NSCLC, SCLC appears to be growth inhibited by overexpression of Notch1 and Notch2.134,135 Indeed, activated Notch1 and Notch2 cause a marked G1 arrest in SCLC cells, accompanied by upregulation of p21waf1/cip1. A prominent function of Notch signaling is to inhibit the transcriptional activities of the widely expressed E2A proteins. This inhibition may occur as a consequence of forming inhibitory complexes of E2A protein with Hes/HERP/HEY proteins, as well as promotion of E2A protein ubiquitinylation and degradation by Notch.136 Indeed, when cultured SCLC cells are induced to overexpress the E2A protein, E12, the ability of Notch1 to induce hASH1 degradation is blunted.135 Together, these observations indicate that Notch signaling rapidly induces degradation along with inactivation of E-proteins and tissue-specific bHLH proteins such as hASH1. Interestingly, Notch1 was much more potent than Hes1 in causing hASH1 silencing and proteasomal degradation, as well as SCLC G1 cell cycle arrest.134,135 These findings are consistent with a model suggesting that Hes-Hey heterodimers may form a more potent Notch effector complex than Hes1 alone. Thus, loss of hASH1 may be critical in mediating the growth inhibitory effect of Notch1 in SCLC. A recent study supporting the idea that Notch1 could have anti-tumor properties on neuroendocrine tumors shows that the induction of Notch1 by valproic acid inhibits carcinoid cell proliferation in vitro and carcinoid tumor growth in vivo.137 The role of Notch ligands in lung cancer has been poorly studied, yet a recent report shows that among Notch ligands, JAG1, JAG2, Dll1 and Dll3 are highly expressed in lung cancer cell lines.138 Moreover, JAG1 expression is dependent upon EGFR activation whereas JAG2 is not. JAG1 depletion (but not JAG2) induces apoptosis, whereas JAG2 depletion induces increased expression of inflammatory related genes. These results suggest that in the same cell type, JAG1 and JAG2 may have distinct functions and that JAG2 can regulate cytokines involved in antitumor immunity.

Cell Adhesion & Migration

Volume 4 Issue 1

Are Guidance Molecules and their Receptors Targets for Lung Cancer Therapy? These five families of guidance molecules are involved in tumor development and regulate tumor cell growth, migration, metastasis or angiogenesis. Thus, their manipulation represents new approaches in cancer therapy. However, given the complexity of the various systems in different contexts, it will be necessary to precisely understand the nature of the alteration in order to best take advantage of therapeutic opportunities. Semaphorins and their receptors. Since semaphorins and their receptors have emerged as regulators of lung tumor development and progression, there is currently strong emphasis on elaborating therapeutic molecules that affect this pathway. To date, significant progress has been achieved although most interest has been centered on neuropilins as downstream VEGF receptors.88,139 Researchers have come up with different strategies to impair the VEGF-neuropilin pathway, such as polysaccharide sulfateinduced neuropilin internalization, neuropilin dimerization inhibiting peptides and VEGF-binding blocking peptides.140-142 One of the most promising studies demonstrated that neuropilin antibodies that specifically prevent VEGF binding to NRP1 or NRP2 efficiently reduce tumor growth, angiogenesis and tumor metastasis in animal models.143,144 Metastasis could also be inhibited by blocking lymphangiogenesis with an anti-NRP2 treatment that blocks VEGF-C binding to NRP2. This treatment did not affect the primary tumor growth but reduced metastasis to the lung without affecting normal lymphatics.143,144 Such a treatment might avoid side effects like lymphedema. Moreover, these antibodies were shown to have additive effects with anti-VEGF antibodies. Thus, there is substantial enthusiasm for targeting neuropilin in cancer.88 Semaphorin-based therapies could also prove valuable in clinical practice. For instance, injection of the extracellular domain of SEMA6D reduced tumor growth and angiogenesis in mice. In some situations where semaphorin inhibition is desired, antibodies that block semaphorin-neuropilin binding have been designed and their efficacy as therapeutic agents will likely be evaluated in the future. One option that has not been reported to our knowledge is to target plexins, possibly by screening chemical libraries for plexin agonists or small molecule inhibitors. This might be of interest considering the recent findings that plexins are mutated in some cancers.145,146 Given that there are two neuropilins, but nine plexins, it appears reasonable that therapeutic targeting of the latter will be more specific. Alternatively, development of specific inhibitors of semaphorins proved to be a feasible approach, as evidenced by independent reports in the nervous system.147,148 Interestingly, a dual VEGF-SEMA3A signaling inhibitor, ZD4190, decreases lung tumor xenograft growth in mice and has additive effects with a Src kinase inhibitor.149 Therefore, there is hope to target the semaphorin pathway for cancer patient treatment, even if this field is still on the emerging slope. Ephrins and their receptors. As described, a subset of lung cancers exhibit Eph/ephrin mutations or amplifications. Therefore,

www.landesbioscience.com

these molecules might represent therapeutic targets, at least in patients whose tumors contain relevant molecular alterations. For instance, lung cancer cell lines with high copy number of EPHA3, A5 or A8 are more sensitive to the broad-spectrum Bcr/ Abl-Src kinase inhibitor, Dasatinib.103 Interestingly, EPH/ephrins are known to signal through Abl and the Src family kinase FYN (Fig. 2), suggesting that Dasatinib directly inhibits ephrin signaling. Alternatively, overexpression of the EGFR family member, ErbB2, in a mouse cancer model correlates with sensitivity to EphA2 inhibition since EphA2 amplifies ErbB2 signals.150 These findings might be relevant to lung cancer where EGFR pathways are commonly affected. Targeting EphA2 appears promising since it would simultaneously block several aspects of tumor progression involving tumor growth, vasculature and microenvironment.151 An effort has been placed on engineering potential therapeutic agents that target the ephrin pathway and several strategies have been employed. For example, a small molecule inhibitor competitively blocks ephrin-A5 binding to EphA2 and EphA4 receptors.152 Conversely, ephrin-mimetic peptides allow specific drug delivery to EphA2 expressing tumor cells. Drug-conjugated human monoclonal antibodies that selectively recognize EphA2 have also been used as “trojan horse”.153 Other efforts, such as an ephrin-A1 cytotoxin and manipulation of EphA2 function have been reported.154 Finally, EphB2 receptor can be inhibited using azurin, a metalloprotein with structural homology with the ephrin-B2 receptor-binding region.155 Also, a soluble monomeric EphB4 inhibits the tri-dimensional and in vivo growth of cancer cells.156 In summary, there is considerable enthusiasm that agents targeting the Eph/ephrin system will find their way to the clinic for the treatment of lung cancer. Netrins and their receptors. Targeting netrin-1 might be attractive as a treatment for lung cancer either by inhibiting netrin-1 expression or by preventing its interaction with receptors. For example, DCC-5Fbn, a 100 amino-acid decoy DCC fragment, affects the ability of netrin-1 to trigger receptor multimerization thereby inhibiting its antiapoptotic effect.110 Inhibition of netrin-1 binding to its receptors might benefit patients whose primary tumors show high levels of netrin-1.109 Such an approach might not only eradicate tumor epithelial cells but also angiogenic vessels. However, induction of apoptotic death in netrin-1 producing cells by interfering with netrin-1 activity is restricted to human cells and animal models that fail to recapitulate the process of NSCLC in humans. Better understanding of the roles of netrins in cancer will be necessary to induce apoptosis specifically in tumor cells. Slit/Robo. Some molecules interacting and interfering with Slit/Robo have proved to have potential anti-tumor capabilities in animal models by inhibiting tumor angiogenesis. Despite the fact that lung cancer therapy has not been studied yet, results obtained in melanoma and in chemically induced oral carcinoma, are promising for lung cancer. In these models, both the extracellular domain of Robo1 and a monoclonal antibody directed against the first immunoglobulin domain of Robo1 (R5) inhibited tumor angiogenesis and tumor growth in vivo.34,35 Interestingly, interfering with Slit2/Robo1 signaling did not affect

Cell Adhesion & Migration

141

the expression profile of VEGF. This suggests that inhibiting Slit-Robo signaling could potentially address the clinical problem of drug resistance in patients treated with VEGF antagonists, such as bevacizumab, in addition to metastasis promotion and perturbation of normal angiogenesis.157,158 Soluble Robo4 could have potential for cancer therapy as Robo4 is specifically expressed on endothelial cells.159,160 Delta/Notch. Although not reported in lung cancer patients, a few clinical trials involving the g-secretase inhibitor MK-0752 are ongoing for patients with breast cancers, leukemia and lymphoma. Valproic acid, which induces Notch1 expression in carcinoid cells and subsequent G1 arrest, is also under investigation in several clinical trials, including one for patients with SCLC, and it is used in combination with other molecules.137 The use of compounds affecting Notch signaling in angiogenesis might have potential for future therapies.47 Dll4 is expressed specifically in tumor vasculature and weaker in adjacent normal vessels, suggesting that therapies directed against Dll4 signaling, as opposed to therapies affecting VEGF signaling, might specifically affect tumor growth without altering other organ functions. Blockade of Dll4-Notch has been tested in various tumor cell lines grown in mice, with reduction of tumor growth from 50 to 90%. Remarkably, the reduced tumor growth is associated with an increase of non-productive vessels together with a concomitant decrease of ephrin-B2. Furthermore, inhibiting Dll4-Notch interaction using Dll4-Fc or anti-Dll4 antibody affects growth of tumors that were resistant to anti-VEGF treatment. Conclusion

Acknowledgements

It has become apparent in the last several years that guidance molecules are involved in lung cancer development/progression through interacting with cell survival, migration and tumor angiogenic pathways. The observation that alterations such as LOH or amplification frequently affect the chromosomal locations of several guidance genes in lung cancer also suggests their References 1.

2.

3.

4.

5.

6.

7.

142

Tessier-Lavigne M, Goodman CS. The molecular biology of axon guidance. Science 1996; 274:112333. Hinck L. The versatile roles of “axon guidance” cues in tissue morphogenesis. Dev Cell 2004; 7:78393. Suchting S, Bicknell R, Eichmann A. Neuronal clues to vascular guidance. Exp Cell Res 2006; 312:66875. Chedotal A, Kerjan G, Moreau-Fauvarque C. The brain within the tumor: new roles for axon guidance molecules in cancers. Cell Death Differ 2005; 12:104456. Klagsbrun M, Eichmann A. A role for axon guidance receptors and ligands in blood vessel development and tumor angiogenesis. Cytokine Growth Factor Rev 2005; 16:535-48. Potiron VA, Roche J, Drabkin HA. Semaphorins and their receptors in lung cancer. Cancer Lett 2009; 273:114. Soker S, Takashima S, Miao HQ, Neufeld G, Klagsbrun M. Neuropilin-1 is expressed by endothelial and tumor cells as an isoform-specific receptor for vascular endothelial growth factor. Cell 1998; 92:735-45.

importance in this disease (Table 2). Moreover, mutations and inactivation by DNA methylation have also been reported for some genes. In further support of the importance of these molecules in lung cancer, some therapeutic success has been achieved against specific targets. Most of these approaches have been carried out on human cancer cell lines and in animal models. For instance, targeting the neuropilins as VEGF co-receptors appears to be an effective strategy both for angiogenesis as well as lymphangiogenesis. In addition, there is enthusiasm for modulating the semaphorin pathway: while class-3 semaphorins have received the greatest attention because of their interaction with the neuropilins, other components can be targeted. It is also our opinion that the other guidance molecules, including the EPH/ephrin, netrin/DCC/UNC5, Slit/Robo and Delta/JAG/Notch pathways provide equally interesting targets. On a cautionary note, there is considerable complexity in these pathways. For example, one ligand can bind different receptors and one receptor can bind different ligands with different biologic consequences. Thus, predicting the biologic consequences of any given intervention may be difficult and considerable experimentation will be required. Moreover, there appears to be cross-talk between pathways. Finally, with increasing knowledge come unexpected consequences, such as reports that VEGF blockade is associated with increased invasion and metastases, at least in experimental models.157,161-163 Nevertheless, this represents an exciting area of investigation and promise for the future.

8.

9.

10.

11.

12.

13.

14.

We would like to apologize for not citing all relevant articles due to space limitations. Our work was supported by University of Poitiers, CNRS, La Ligue Contre le Cancer and L’Association pour la Recherche sur le Cancer for P.N., V.P., J.R. and National Institutes of Health CA58187 (Colorado Lung Cancer Specialized Program of Research Excellence for P.N., V.P., J.R., H.D.).

Miao HQ, Soker S, Feiner L, Alonso JL, Raper JA, Klagsbrun M. Neuropilin-1 mediates collapsin-1/semaphorin III inhibition of endothelial cell motility: functional competition of collapsin-1 and vascular endothelial growth factor-165. J Cell Biol 1999; 146:23342. Favier B, Alam A, Barron P, Bonnin J, Laboudie P, Fons P, et al. Neuropilin-2 interacts with VEGFR2 and VEGFR-3 and promotes human endothelial cell survival and migration. Blood 2006; 108:124350. Karpanen T, Heckman CA, Keskitalo S, Jeltsch M, Ollila H, Neufeld G, et al. Functional interaction of VEGF-C and VEGF-D with neuropilin receptors. Faseb J 2006; 20:1462-72. Uniewicz KA, Fernig DG. Neuropilins: a versatile partner of extracellular molecules that regulate development and disease. Front Biosci 2008; 13:4339-60. Kruger RP, Aurandt J, Guan KL. Semaphorins command cells to move. Nat Rev Mol Cell Biol 2005; 6:789-800. Swiercz JM, Worzfeld T, Offermanns S. ErbB-2 and met reciprocally regulate cellular signaling via plexinB1. J Biol Chem 2008; 283:1893-901. Puschel AW. GTPases in semaphorin signaling. Adv Exp Med Biol 2007; 600:12-23.

Cell Adhesion & Migration

15. Klostermann A, Lutz B, Gertler F, Behl C. The orthologous human and murine semaphorin 6A-1 proteins (SEMA6A-1/Sema6A-1) bind to the enabled/vasodilator-stimulated phosphoprotein-like protein (EVL) via a novel carboxyl-terminal zyxin-like domain. J Biol Chem 2000; 275:39647-53. 16. Toyofuku T, Zhang H, Kumanogoh A, Takegahara N, Yabuki M, Harada K, et al. Guidance of myocardial patterning in cardiac development by Sema6D reverse signalling. Nat Cell Biol 2004; 6:1204-11. 17. Surawska H, Ma PC, Salgia R. The role of ephrins and Eph receptors in cancer. Cytokine Growth Factor Rev 2004; 15:419-33. 18. Zhang J, Hughes S. Role of the ephrin and Eph receptor tyrosine kinase families in angiogenesis and development of the cardiovascular system. J Pathol 2006; 208:453-61. 19. Pasquale EB. Eph-ephrin bidirectional signaling in physiology and disease. Cell 2008; 133:38-52. 20. Arvanitis D, Davy A. Eph/ephrin signaling: networks. Genes Dev 2008; 22:416-29. 21. Hong K, Hinck L, Nishiyama M, Poo MM, TessierLavigne M, Stein E. A ligand-gated association between cytoplasmic domains of UNC5 and DCC family receptors converts netrin-induced growth cone attraction to repulsion. Cell 1999; 97:927-41.

Volume 4 Issue 1

22. Arakawa H. Netrin-1 and its receptors in tumorigenesis. Nat Rev Cancer 2004; 4:978-87. 23. Bernet A, Fitamant J. Netrin-1 and its receptors in tumour growth promotion. Expert Opin Ther Targets 2008; 12:995-1007. 24. Nikolopoulos SN, Giancotti FG. Netrin-integrin signaling in epithelial morphogenesis, axon guidance and vascular patterning. Cell Cycle 2005; 4:131-5. 25. Yebra M, Montgomery AM, Diaferia GR, Kaido T, Silletti S, Perez B, et al. Recognition of the neural chemoattractant Netrin-1 by integrins alpha6beta4 and alpha3beta1 regulates epithelial cell adhesion and migration. Dev Cell 2003; 5:695-707. 26. Hagedorn EJ, Yashiro H, Ziel JW, Ihara S, Wang Z, Sherwood DR. Integrin acts upstream of netrin signaling to regulate formation of the anchor cell’s invasive membrane in C. elegans. Dev Cell 2009; 17:187-98. 27. Mehlen P, Rabizadeh S, Snipas SJ, Assa-Munt N, Salvesen GS, Bredesen DE. The DCC gene product induces apoptosis by a mechanism requiring receptor proteolysis. Nature 1998; 395:801-4. 28. Mille F, Llambi F, Guix C, Delloye-Bourgeois C, Guenebeaud C, Castro-Obregon S, et al. Interfering with multimerization of netrin-1 receptors triggers tumor cell death. Cell Death Differ 2009; 16:134451. 29. Tanikawa C, Matsuda K, Fukuda S, Nakamura Y, Arakawa H. p53RDL1 regulates p53-dependent apoptosis. Nat Cell Biol 2003; 5:216-23. 30. Schon MP, Schon M. To die or not to die, that’s the question—and the answer may depend on netrin-1. J Natl Cancer Inst 2009; 101:217-9. 31. Taniguchi Y, Kim SH, Sisodia SS. Presenilin-dependent “gamma-secretase” processing of deleted in colorectal cancer (DCC). J Biol Chem 2003; 278:304258. 32. Legg JA, Herbert JM, Clissold P, Bicknell R. Slits and Roundabouts in cancer, tumour angiogenesis and endothelial cell migration. Angiogenesis 2008; 11:1321. 33. Suchting S, Heal P, Tahtis K, Stewart LM, Bicknell R. Soluble Robo4 receptor inhibits in vivo angiogenesis and endothelial cell migration. Faseb J 2005; 19:121-3. 34. Wang B, Xiao Y, Ding BB, Zhang N, Yuan X, Gui L, et al. Induction of tumor angiogenesis by Slit-Robo signaling and inhibition of cancer growth by blocking Robo activity. Cancer Cell 2003; 4:19-29. 35. Wang LJ, Zhao Y, Han B, Ma YG, Zhang J, Yang DM, et al. Targeting Slit-Roundabout signaling inhibits tumor angiogenesis in chemical-induced squamous cell carcinogenesis. Cancer Sci 2008; 99:510-7. 36. Kaur S, Castellone MD, Bedell VM, Konar M, Gutkind JS, Ramchandran R. Robo4 signaling in endothelial cells implies attraction guidance mechanisms. J Biol Chem 2006; 281:11347-56. 37. Jones CA, London NR, Chen H, Park KW, Sauvaget D, Stockton RA, et al. Robo4 stabilizes the vascular network by inhibiting pathologic angiogenesis and endothelial hyperpermeability. Nat Med 2008; 14:44853. 38. Stella MC, Trusolino L, Comoglio PM. The Slit/Robo system suppresses hepatocyte growth factor-dependent invasion and morphogenesis. Mol Biol Cell 2009; 20:642-57. 39. Wu JY, Feng L, Park HT, Havlioglu N, Wen L, Tang H, et al. The neuronal repellent Slit inhibits leukocyte chemotaxis induced by chemotactic factors. Nature 2001; 410:948-52. 40. Stein E, Tessier-Lavigne M. Hierarchical organization of guidance receptors: silencing of netrin attraction by slit through a Robo/DCC receptor complex. Science 2001; 291:1928-38. 41. Forcet C, Ye X, Granger L, Corset V, Shin H, Bredesen DE, et al. The dependence receptor DCC (deleted in colorectal cancer) defines an alternative mechanism for caspase activation. Proc Natl Acad Sci USA 2001; 98:3416-21.

www.landesbioscience.com

42. Giniger E, Jan LY, Jan YN. Specifying the path of the intersegmental nerve of the Drosophila embryo: a role for Delta and Notch. Development 1993; 117:43140. 43. De Bellard ME, Ching W, Gossler A, Bronner-Fraser M. Disruption of segmental neural crest migration and ephrin expression in delta-1 null mice. Dev Biol 2002; 249:121-30. 44. Ferrari-Toninelli G, Bonini SA, Bettinsoli P, Uberti D, Memo M. Microtubule stabilizing effect of notch activation in primary cortical neurons. Neuroscience 2008; 154:946-52. 45. Fuss B, Josten F, Feix M, Hoch M. Cell movements controlled by the Notch signalling cascade during foregut development in Drosophila. Development 2004; 131:1587-95. 46. Hashimoto-Torii K, Torii M, Sarkisian MR, Bartley CM, Shen J, Radtke F, et al. Interaction between Reelin and Notch signaling regulates neuronal migration in the cerebral cortex. Neuron 2008; 60:273-84. 47. Thurston G, Noguera-Troise I, Yancopoulos GD. The Delta paradox: DLL4 blockade leads to more tumour vessels but less tumour growth. Nat Rev Cancer 2007; 7:327-31. 48. Bray S. Notch signalling in Drosophila: three ways to use a pathway. Semin Cell Dev Biol 1998; 9: 591-7. 49. Ito T, Kagoshima M, Sasaki Y, Li C, Udaka N, Kitsukawa T, et al. Repulsive axon guidance molecule Sema3A inhibits branching morphogenesis of fetal mouse lung. Mech Dev 2000; 97:35-45. 50. Kagoshima M, Ito T. Diverse gene expression and function of semaphorins in developing lung: positive and negative regulatory roles of semaphorins in lung branching morphogenesis. Genes Cells 2001; 6:55971. 51. Brambilla E, Constantin B, Drabkin H, Roche J. Semaphorin SEMA3F localization in malignant human lung and cell lines: A suggested role in cell adhesion and cell migration. Am J Pathol 2000; 156:939-50. 52. Wilkinson GA, Schittny JC, Reinhardt DP, Klein R. Role for ephrinB2 in postnatal lung alveolar development and elastic matrix integrity. Dev Dyn 2008; 237:2220-34. 53. Wohlfahrt JG, Karagiannidis C, Kunzmann S, Epstein MM, Kempf W, Blaser K, et al. Ephrin-A1 suppresses Th2 cell activation and provides a regulatory link to lung epithelial cells. J Immunol 2004; 172:843-50. 54. Aasheim HC, Pedeutour F, Grosgeorge J, Logtenberg T. Cloning, chromosal mapping and tissue expression of the gene encoding the human Eph-family kinase ligand ephrin-A2. Biochem Biophys Res Commun 1998; 252:378-82. 55. Fox GM, Holst PL, Chute HT, Lindberg RA, Janssen AM, Basu R, et al. cDNA cloning and tissue distribution of five human EPH-like receptor protein-tyrosine kinases. Oncogene 1995; 10:897-905. 56. Beckmann MP, Cerretti DP, Baum P, Vanden Bos T, James L, Farrah T, et al. Molecular characterization of a family of ligands for eph-related tyrosine kinase receptors. EMBO J 1994; 13:3757-62. 57. Bohme B, Holtrich U, Wolf G, Luzius H, Grzeschik KH, Strebhardt K, et al. PCR mediated detection of a new human receptor-tyrosine-kinase, HEK 2. Oncogene 1993; 8:2857-62. 58. Cerretti DP, Vanden Bos T, Nelson N, Kozlosky CJ, Reddy P, Maraskovsky E, et al. Isolation of LERK-5: a ligand of the eph-related receptor tyrosine kinases. Mol Immunol 1995; 32:1197-205. 59. Fletcher FA, Carpenter MK, Shilling H, Baum P, Ziegler SF, Gimpel S, et al. LERK-2, a binding protein for the receptor-tyrosine kinase ELK, is evolutionarily conserved and expressed in a developmentally regulated pattern. Oncogene 1994; 9:3241-7. 60. Hafner C, Schmitz G, Meyer S, Bataille F, Hau P, Langmann T, et al. Differential gene expression of Eph receptors and ephrins in benign human tissues and cancers. Clin Chem 2004; 50:490-9.

Cell Adhesion & Migration

61. Favre CJ, Mancuso M, Maas K, McLean JW, Baluk P, McDonald DM. Expression of genes involved in vascular development and angiogenesis in endothelial cells of adult lung. Am J Physiol Heart Circ Physiol 2003; 285:1917-38. 62. Gale NW, Baluk P, Pan L, Kwan M, Holash J, DeChiara TM, et al. Ephrin-B2 selectively marks arterial vessels and neovascularization sites in the adult, with expression in both endothelial and smooth-muscle cells. Dev Biol 2001; 230:151-60. 63. Wang HU, Chen ZF, Anderson DJ. Molecular distinction and angiogenic interaction between embryonic arteries and veins revealed by ephrin-B2 and its receptor Eph-B4. Cell 1998; 93:741-53. 64. Schwarz MA, Caldwell L, Cafasso D, Zheng H. Emerging pulmonary vasculature lacks fate specification. Am J Physiol Lung Cell Mol Physiol 2009; 296:71-81. 65. Foo SS, Turner CJ, Adams S, Compagni A, Aubyn D, Kogata N, et al. Ephrin-B2 controls cell motility and adhesion during blood-vessel-wall assembly. Cell 2006; 124:161-73. 66. Erber R, Eichelsbacher U, Powajbo V, Korn T, Djonov V, Lin J, et al. EphB4 controls blood vascular morphogenesis during postnatal angiogenesis. EMBO J 2006; 25:628-41. 67. Larson J, Schomberg S, Schroeder W, Carpenter TC. Endothelial EphA receptor stimulation increases lung vascular permeability. Am J Physiol Lung Cell Mol Physiol 2008; 295:431-9. 68. Eichmann A, Makinen T, Alitalo K. Neural guidance molecules regulate vascular remodeling and vessel navigation. Genes Dev 2005; 19:1013-21. 69. Dalvin S, Anselmo MA, Prodhan P, Komatsuzaki K, Schnitzer JJ, Kinane TB. Expression of Netrin-1 and its two receptors DCC and UNC5H2 in the developing mouse lung. Gene Expr Patterns 2003; 3:279-83. 70. Liu Y, Stein E, Oliver T, Li Y, Brunken WJ, Koch M, et al. Novel role for Netrins in regulating epithelial behavior during lung branching morphogenesis. Curr Biol 2004; 14:897-905. 71. Xian J, Clark KJ, Fordham R, Pannell R, Rabbitts TH, Rabbitts PH. Inadequate lung development and bronchial hyperplasia in mice with a targeted deletion in the Dutt1/Robo1 gene. Proc Natl Acad Sci USA 2001; 98:15062-6. 72. Greenberg JM, Thompson FY, Brooks SK, Shannon JM, Akeson AL. Slit and robo expression in the developing mouse lung. Dev Dyn 2004; 230:350-60. 73. Anselmo MA, Dalvin S, Prodhan P, Komatsuzaki K, Aidlen JT, Schnitzer JJ, et al. Slit and robo: expression patterns in lung development. Gene Expr Patterns 2003; 3:13-9. 74. Clark K, Hammond E, Rabbitts P. Temporal and spatial expression of two isoforms of the Dutt1/Robo1 gene in mouse development. FEBS Lett 2002; 523:12-6. 75. Guseh JS, Bores SA, Stanger BZ, Zhou Q, Anderson WJ, Melton DA, et al. Notch signaling promotes airway mucous metaplasia and inhibits alveolar development. Development 2009; 136:1751-9. 76. Ito T, Udaka N, Yazawa T, Okudela K, Hayashi H, Sudo T, et al. Basic helix-loop-helix transcription factors regulate the neuroendocrine differentiation of fetal mouse pulmonary epithelium. Development 2000; 127:3913-21. 77. Post LC, Ternet M, Hogan BL. Notch/Delta expression in the developing mouse lung. Mech Dev 2000; 98:958. 78. Shan L, Aster JC, Sklar J, Sunday ME. Notch-1 regulates pulmonary neuroendocrine cell differentiation in cell lines and in transgenic mice. Am J Physiol Lung Cell Mol Physiol 2007; 292:500-9. 79. Tsao PN, Vasconcelos M, Izvolsky KI, Qian J, Lu J, Cardoso WV. Notch signaling controls the balance of ciliated and secretory cell fates in developing airways. Development 2009; 136:2297-307.

143

80. Tsao PN, Chen F, Izvolsky KI, Walker J, Kukuruzinska MA, Lu J, et al. Gamma-secretase activation of notch signaling regulates the balance of proximal and distal fates in progenitor cells of the developing lung. J Biol Chem 2008; 283:29532-44. 81. Borges M, Linnoila RI, van de Velde HJ, Chen H, Nelkin BD, Mabry M, et al. An achaete-scute homologue essential for neuroendocrine differentiation in the lung. Nature 1997; 386:852-5. 82. Leimeister C, Schumacher N, Steidl C, Gessler M. Analysis of HeyL expression in wild-type and Notch pathway mutant mouse embryos. Mech Dev 2000; 98:175-8. 83. Steidl C, Leimeister C, Klamt B, Maier M, Nanda I, Dixon M, et al. Characterization of the human and mouse HEY1, HEY2 and HEYL genes: cloning, mapping, and mutation screening of a new bHLH gene family. Genomics 2000; 66:195-203. 84. de Pontual L, Nepote V, Attie-Bitach T, Al Halabiah H, Trang H, Elghouzzi V, et al. Noradrenergic neuronal development is impaired by mutation of the proneural HASH-1 gene in congenital central hypoventilation syndrome (Ondine’s curse). Hum Mol Genet 2003; 12:3173-80. 85. Huang MT, Dai YS, Chou YB, Juan YH, Wang CC, Chiang BL. Regulatory T cells negatively regulate neovasculature of airway remodeling via DLL4-notch signaling. J Immunol 2009; 183:4745-54. 86. Raymond T, Schaller M, Hogaboam CM, Lukacs NW, Rochford R, Kunkel SL. Toll-like receptors, Notch ligands and cytokines drive the chronicity of lung inflammation. Proc Am Thorac Soc 2007; 4:635-41. 87. Tilley AE, Harvey BG, Heguy A, Hackett NR, Wang R, O’Connor TP, et al. Downregulation of the notch pathway in human airway epithelium in association with smoking and chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2009; 179:457-66. 88. Bagri A, Tessier-Lavigne M, Watts RJ. Neuropilins in tumor biology. Clin Cancer Res 2009; 15: 1860-4. 89. Capparuccia L, Tamagnone L. Semaphorin signaling in cancer cells and in cells of the tumor microenvironment: two sides of a coin. J Cell Sci 2009; 122:172336. 90. Geretti E, Shimizu A, Klagsbrun M. Neuropilin structure governs VEGF and semaphorin binding and regulates angiogenesis. Angiogenesis 2008; 11:31-9. 91. Neufeld G, Kessler O. The semaphorins: versatile regulators of tumour progression and tumour angiogenesis. Nat Rev Cancer 2008; 8:632-45. 92. Roche J, Boldog F, Robinson M, Robinson L, VarellaGarcia M, Swanton M, et al. Distinct 3p21.3 deletions in lung cancer and identification of a new human semaphorin. Oncogene 1996; 12:1289-97. 93. Sekido Y, Bader S, Latif F, Chen JY, Duh FM, Wei MH, et al. Human semaphorins A(V) and IV reside in the 3p21.3 small cell lung cancer deletion region and demonstrate distinct expression patterns. Proc Natl Acad Sci USA 1996; 93:4120-5. 94. Xiang RH, Hensel CH, Garcia DK, Carlson HC, Kok K, Daly MC, et al. Isolation of the human semaphorin III/F gene (SEMA3F) at chromosome 3p21, a region deleted in lung cancer. Genomics 1996; 32:39-48. 95. Clarhaut J, Gemmill RM, Potiron VA, Ait-Si-Ali S, Imbert J, Drabkin HA, et al. ZEB-1, a repressor of the semaphorin 3F tumor suppressor gene in lung cancer cells. Neoplasia 2009; 11:157-66. 96. Lantuejoul S, Constantin B, Drabkin H, Brambilla C, Roche J, Brambilla E. Expression of VEGF, semaphorin SEMA3F, and their common receptors neuropilins NP1 and NP2 in preinvasive bronchial lesions, lung tumours, and cell lines. J Pathol 2003; 200:336-47. 97. Castro-Rivera E, Ran S, Brekken RA, Minna JD. Semaphorin 3B inhibits the phosphatidylinositol 3-kinase/Akt pathway through neuropilin-1 in lung and breast cancer cells. Cancer Res 2008; 68:8295303.

144

98. Koyama N, Zhang J, Huqun, Miyazawa H, Tanaka T, Su X, et al. Identification of IGFBP-6 as an effector of the tumor suppressor activity of SEMA3B. Oncogene 2008; 27:6581-9. 99. Rolny C, Capparuccia L, Casazza A, Mazzone M, Vallario A, Cignetti A, et al. The tumor suppressor semaphorin 3B triggers a prometastatic program mediated by interleukin 8 and the tumor microenvironment. J Exp Med 2008; 205:1155-71. 100. Pan SH, Chao YC, Chen HY, Hung PF, Lin PY, Lin CW, et al. Long form collapsin response mediator protein-1 (LCRMP-1) expression is associated with clinical outcome and lymph node metastasis in non-small cell lung cancer patients. Lung Cancer 2010; 67:93-100. 101. Lugli A, Spichtin H, Maurer R, Mirlacher M, Kiefer J, Huusko P, et al. EphB2 expression across 138 human tumor types in a tissue microarray: high levels of expression in gastrointestinal cancers. Clin Cancer Res 2005; 11:6450-8. 102. Yu J, Bulk E, Ji P, Hascher A, Koschmieder S, Berdel WE, et al. The kinase defective EPHB6 receptor tyrosine kinase activates MAP kinase signaling in lung adenocarcinoma. Int J Oncol 2009; 35: 175-9. 103. Sos ML, Michel K, Zander T, Weiss J, Frommolt P, Peifer M, et al. Predicting drug susceptibility of nonsmall cell lung cancers based on genetic lesions. J Clin Invest 2009; 119:1727-40. 104. Ding L, Getz G, Wheeler DA, Mardis ER, McLellan MD, Cibulskis K, et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature 2008; 455:1069-75. 105. Nasreen N, Mohammed KA, Lai Y, Antony VB. Receptor EphA2 activation with ephrinA1 suppresses growth of malignant mesothelioma (MM). Cancer Lett 2007; 258:215-22. 106. Ogawa K, Pasqualini R, Lindberg RA, Kain R, Freeman AL, Pasquale EB. The ephrin-A1 ligand and its receptor, EphA2, are expressed during tumor neovascularization. Oncogene 2000; 19:6043-52. 107. Fearon ER, Cho KR, Nigro JM, Kern SE, Simons JW, Ruppert JM, et al. Identification of a chromosome 18q gene that is altered in colorectal cancers. Science 1990; 247:49-56. 108. Shin SK, Nagasaka T, Jung BH, Matsubara N, Kim WH, Carethers JM, et al. Epigenetic and genetic alterations in Netrin-1 receptors UNC5C and DCC in human colon cancer. Gastroenterology 2007; 133:1849-57. 109. Delloye-Bourgeois C, Brambilla E, Coissieux MM, Guenebeaud C, Pedeux R, Firlej V, et al. Interference with netrin-1 and tumor cell death in non-small cell lung cancer. J Natl Cancer Inst 2009; 101:237-47. 110. Fitamant J, Guenebeaud C, Coissieux MM, Guix C, Treilleux I, Scoazec JY, et al. Netrin-1 expression confers a selective advantage for tumor cell survival in metastatic breast cancer. Proc Natl Acad Sci USA 2008; 105:4850-5. 111. Rodrigues S, De Wever O, Bruyneel E, Rooney RJ, Gespach C. Opposing roles of netrin-1 and the dependence receptor DCC in cancer cell invasion, tumor growth and metastasis. Oncogene 2007; 26:5615-25. 112. Link BC, Reichelt U, Schreiber M, Kaifi JT, Wachowiak R, Bogoevski D, et al. Prognostic implications of netrin-1 expression and its receptors in patients with adenocarcinoma of the pancreas. Ann Surg Oncol 2007; 14:2591-9. 113. Castets M, Coissieux MM, Delloye-Bourgeois C, Bernard L, Delcros JG, Bernet A, et al. Inhibition of endothelial cell apoptosis by netrin-1 during angiogenesis. Dev Cell 2009; 16:614-20. 114. Park KW, Crouse D, Lee M, Karnik SK, Sorensen LK, Murphy KJ, et al. The axonal attractant Netrin-1 is an angiogenic factor. Proc Natl Acad Sci USA 2004; 101:16210-5.

Cell Adhesion & Migration

115. Lu X, Le Noble F, Yuan L, Jiang Q, De Lafarge B, Sugiyama D, et al. The netrin receptor UNC5B mediates guidance events controlling morphogenesis of the vascular system. Nature 2004; 432:179-86. 116. Wilson BD, Ii M, Park KW, Suli A, Sorensen LK, Larrieu-Lahargue F, et al. Netrins promote developmental and therapeutic angiogenesis. Science 2006; 313:640-4. 117. Llambi F, Lourenco FC, Gozuacik D, Guix C, Pays L, Del Rio G, et al. The dependence receptor UNC5H2 mediates apoptosis through DAP-kinase. EMBO J 2005; 24:1192-201. 118. Inbal B, Cohen O, Polak-Charcon S, Kopolovic J, Vadai E, Eisenbach L, et al. DAP kinase links the control of apoptosis to metastasis. Nature 1997; 390:1804. 119. Yanagawa N, Tamura G, Oizumi H, Kanauchi N, Endoh M, Sadahiro M, et al. Promoter hypermethylation of RASSF1A and RUNX3 genes as an independent prognostic prediction marker in surgically resected nonsmall cell lung cancers. Lung Cancer 2007; 58:131-8. 120. Abdollahi A, Schwager C, Kleeff J, Esposito I, Domhan S, Peschke P, et al. Transcriptional network governing the angiogenic switch in human pancreatic cancer. Proc Natl Acad Sci USA 2007; 104:12890-5. 121. Dallol A, Forgacs E, Martinez A, Sekido Y, Walker R, Kishida T, et al. Tumour specific promoter region methylation of the human homologue of the Drosophila Roundabout gene DUTT1 (ROBO1) in human cancers. Oncogene 2002; 21:3020-8. 122. Sundaresan V, Chung G, Heppell-Parton A, Xiong J, Grundy C, Roberts I, et al. Homozygous deletions at 3p12 in breast and lung cancer. Oncogene 1998; 17:1723-9. 123. Georgas K, Burridge L, Smith K, Holmes GP, ChenevixTrench G, Ioannou PA, et al. Assignment of the human slit homologue SLIT2 to human chromosome band 4p15.2. Cytogenet Cell Genet 1999; 86:246-7. 124. Shivapurkar N, Virmani AK, Wistuba II, Milchgrub S, Mackay B, Minna JD, et al. Deletions of chromosome 4 at multiple sites are frequent in malignant mesothelioma and small cell lung carcinoma. Clin Cancer Res 1999; 5:17-23. 125. Girard L, Zochbauer-Muller S, Virmani AK, Gazdar AF, Minna JD. Genome-wide allelotyping of lung cancer identifies new regions of allelic loss, differences between small cell lung cancer and non-small cell lung cancer, and loci clustering. Cancer Res 2000; 60:4894906. 126. Dallol A, Da Silva NF, Viacava P, Minna JD, Bieche I, Maher ER, et al. SLIT2, a human homologue of the Drosophila Slit2 gene, has tumor suppressor activity and is frequently inactivated in lung and breast cancers. Cancer Res 2002; 62:5874-80. 127. Gorn M, Anige M, Burkholder I, Muller B, Scheffler A, Edler L, et al. Serum levels of Magic Roundabout protein in patients with advanced non-small cell lung cancer (NSCLC). Lung Cancer 2005; 49:71-6. 128. Zheng Q, Qin H, Zhang H, Li J, Hou L, Wang H, et al. Notch signaling inhibits growth of the human lung adenocarcinoma cell line A549. Oncol Rep 2007; 17:847-52. 129. Chen Y, De Marco MA, Graziani I, Gazdar AF, Strack PR, Miele L, et al. Oxygen concentration determines the biological effects of NOTCH-1 signaling in adenocarcinoma of the lung. Cancer Res 2007; 67:7954-9. 130. Garnis C, Campbell J, Davies JJ, Macaulay C, Lam S, Lam WL. Involvement of multiple developmental genes on chromosome 1p in lung tumorigenesis. Hum Mol Genet 2005; 14:475-82. 131. Dang TP, Gazdar AF, Virmani AK, Sepetavec T, Hande KR, Minna JD, et al. Chromosome 19 translocation, overexpression of Notch3, and human lung cancer. J Natl Cancer Inst 2000; 92:1355-7.

Volume 4 Issue 1

132. Haruki N, Kawaguchi KS, Eichenberger S, Massion PP, Olson S, Gonzalez A, et al. Dominant-negative Notch3 receptor inhibits mitogen-activated protein kinase pathway and the growth of human lung cancers. Cancer Res 2005; 65:3555-61. 133. Konishi J, Kawaguchi KS, Vo H, Haruki N, Gonzalez A, Carbone DP, et al. Gamma-secretase inhibitor prevents Notch3 activation and reduces proliferation in human lung cancers. Cancer Res 2007; 67:80517. 134. Sriuranpong V, Borges MW, Ravi RK, Arnold DR, Nelkin BD, Baylin SB, et al. Notch signaling induces cell cycle arrest in small cell lung cancer cells. Cancer Res 2001; 61:3200-5. 135. Sriuranpong V, Borges MW, Strock CL, Nakakura EK, Watkins DN, Blaumueller CM, et al. Notch signaling induces rapid degradation of achaete-scute homolog 1. Mol Cell Biol 2002; 22:3129-39. 136. Nie L, Xu M, Vladimirova A, Sun XH. Notch-induced E2A ubiquitination and degradation are controlled by MAP kinase activities. EMBO J 2003; 22:5780-92. 137. Greenblatt DY, Vaccaro AM, Jaskula-Sztul R, Ning L, Haymart M, Kunnimalaiyaan M, et al. Valproic acid activates notch-1 signaling and regulates the neuroendocrine phenotype in carcinoid cancer cells. Oncologist 2007; 12:942-51. 138. Choi K, Ahn YH, Gibbons DL, Tran HT, Creighton CJ, Girard L, et al. Distinct biological roles for the notch ligands Jagged-1 and Jagged-2. J Biol Chem 2009; 284:17766-74. 139. Geretti E, Klagsbrun M. Neuropilins: novel targets for anti-angiogenesis therapies. Cell Adh Migr 2007; 1:5661. 140. Binetruy-Tournaire R, Demangel C, Malavaud B, Vassy R, Rouyre S, Kraemer M, et al. Identification of a peptide blocking vascular endothelial growth factor (VEGF)-mediated angiogenesis. EMBO J 2000; 19:1525-33. 141. Narazaki M, Segarra M, Tosato G. Sulfated polysaccharides identified as inducers of neuropilin-1 internalization and functional inhibition of VEGF165 and semaphorin3A. Blood 2008; 111:4126-36. 142. Roth L, Nasarre C, Dirrig-Grosch S, Aunis D, Cremel G, Hubert P, et al. Transmembrane domain interactions control biological functions of neuropilin-1. Mol Biol Cell 2008; 19:646-54.

www.landesbioscience.com

143. Caunt M, Mak J, Liang WC, Stawicki S, Pan Q, Tong RK, et al. Blocking neuropilin-2 function inhibits tumor cell metastasis. Cancer Cell 2008; 13:331-42. 144. Pan Q, Chanthery Y, Liang WC, Stawicki S, Mak J, Rathore N, et al. Blocking neuropilin-1 function has an additive effect with anti-VEGF to inhibit tumor growth. Cancer Cell 2007; 11:53-67. 145. Balakrishnan A, Penachioni JY, Lamba S, Bleeker FE, Zanon C, Rodolfo M, et al. Molecular profiling of the “plexinome” in melanoma and pancreatic cancer. Hum Mutat 2009; 30:1167-74. 146. Wong OG, Nitkunan T, Oinuma I, Zhou C, Blanc V, Brown RS, et al. Plexin-B1 mutations in prostate cancer. Proc Natl Acad Sci USA 2007; 104:19040-5. 147. Kaneko S, Iwanami A, Nakamura M, Kishino A, Kikuchi K, Shibata S, et al. A selective Sema3A inhibitor enhances regenerative responses and functional recovery of the injured spinal cord. Nat Med 2006; 12:1380-9. 148. Montolio M, Messeguer J, Masip I, Guijarro P, Gavin R, Antonio Del Rio J, et al. A semaphorin 3A inhibitor blocks axonal chemorepulsion and enhances axon regeneration. Chem Biol 2009; 16:691-701. 149. Nguyen QD, Rodrigues S, Rodrigue CM, Rivat C, Grijelmo C, Bruyneel E, et al. Inhibition of vascular endothelial growth factor (VEGF)-165 and semaphorin 3A-mediated cellular invasion and tumor growth by the VEGF signaling inhibitor ZD4190 in human colon cancer cells and xenografts. Mol Cancer Ther 2006; 5:2070-7. 150. Brantley-Sieders DM, Zhuang G, Hicks D, Fang WB, Hwang Y, Cates JM, et al. The receptor tyrosine kinase EphA2 promotes mammary adenocarcinoma tumorigenesis and metastatic progression in mice by amplifying ErbB2 signaling. J Clin Invest 2008; 118:64-78. 151. Ireton RC, Chen J. EphA2 receptor tyrosine kinase as a promising target for cancer therapeutics. Curr Cancer Drug Targets 2005; 5:149-57. 152. Noberini R, Koolpe M, Peddibhotla S, Dahl R, Su Y, Cosford ND, et al. Small molecules can selectively inhibit ephrin binding to the EphA4 and EphA2 receptors. J Biol Chem 2008; 283:29461-72. 153. Jackson D, Gooya J, Mao S, Kinneer K, Xu L, Camara M, et al. A human antibody-drug conjugate targeting EphA2 inhibits tumor growth in vivo. Cancer Res 2008; 68:9367-74.

Cell Adhesion & Migration

154. Wykosky J, Debinski W. The EphA2 receptor and ephrinA1 ligand in solid tumors: function and therapeutic targeting. Mol Cancer Res 2008; 6:1795-806. 155. Chaudhari A, Mahfouz M, Fialho AM, Yamada T, Granja AT, Zhu Y, et al. Cupredoxin-cancer interrelationship: azurin binding with EphB2, interference in EphB2 tyrosine phosphorylation, and inhibition of cancer growth. Biochemistry 2007; 46:1799-810. 156. Martiny-Baron G, Korff T, Schaffner F, Esser N, Eggstein S, Marme D, et al. Inhibition of tumor growth and angiogenesis by soluble EphB4. Neoplasia 2004; 6:248-57. 157. Ebos JM, Lee CR, Cruz-Munoz W, Bjarnason GA, Christensen JG, Kerbel RS. Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell 2009; 15:232-9. 158. Kamba T, McDonald DM. Mechanisms of adverse effects of anti-VEGF therapy for cancer. Br J Cancer 2007; 96:1788-95. 159. Huminiecki L, Bicknell R. In silico cloning of novel endothelial-specific genes. Genome Res 2000; 10:1796806. 160. Neri D, Bicknell R. Tumour vascular targeting. Nat Rev Cancer 2005; 5:436-46. 161. Greenberg JI, Shields DJ, Barillas SG, Acevedo LM, Murphy E, Huang J, et al. A role for VEGF as a negative regulator of pericyte function and vessel maturation. Nature 2008; 456:809-13. 162. Paez-Ribes M, Allen E, Hudock J, Takeda T, Okuyama H, Vinals F, et al. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell 2009; 15:220-31. 163. Stockmann C, Doedens A, Weidemann A, Zhang N, Takeda N, Greenberg JI, et al. Deletion of vascular endothelial growth factor in myeloid cells accelerates tumorigenesis. Nature 2008; 456:814-8. 164. Round J, Stein E. Netrin signaling leading to directed growth cone steering. Curr Opin Neurobiol 2007; 17:15-21. 165. Nichols JT, Miyamoto A, Weinmaster G. Notch signaling—constantly on the move. Traffic 2007; 8:959-69. 166. Maeda Y, Dave V, Whitsett JA. Transcriptional control of lung morphogenesis. Physiol Rev 2007; 87:219-44.

145