GET THE APP

Expression and Function of the Eph Receptor Family in Leukemia an
Journal of Leukemia

Journal of Leukemia
Open Access

ISSN: 2329-6917

+44 1300 500008

Review Article - (2013) Volume 1, Issue 1

Expression and Function of the Eph Receptor Family in Leukemia and Hematopoietic Malignancies: Prospects for Targeted Therapies

Sara Charmsaz1* and Andrew W Boyd1,2
1Leukaemia Foundation Research Unit, Queensland Institute of Medical Research, 300 Herston Road, Herston, Queensland 4006, Australia
2Department of Medicine, University of Queensland, Herston, Queensland 4006, Australia
*Corresponding Author: Sara Charmsaz, Leukaemia Foundation Research Unit, Queensland Institute Of Medical Research, P.O. Royal Brisbane Hospital, Queensland, 4029, Brisbane, Australia, Tel: +61-7-33620387, Fax: +61-7-38453509 Email:

Abstract

There has been considerable interest in recent years in the development of therapies, which target Eph receptors or their ephrin ligands. The Eph receptor tyrosine kinases and their membrane bound ephrin ligands are cell surface molecules involved in many biological functions and cell behaviors during embryogenesis and in adult life. However, they are also expressed in an aberrant fashion on many tumors and are re-expressed on normal cells in nonmalignant pathological states. Some of the eph/ephrins including EphA7 and EphA1 protein are thought to function as tumor suppressors in particular cancers. In tumors where Eph/ephrin proteins are expressed at high levels, being expressed on the cell surface these proteins are readily accessible to antibody-mediated therapies several of which are in advanced pre-clinical or early clinical evaluation, including antibodies specific for EphA2, EphA3, and EphB4 which are expressed on many different tumors. We will review the general features of the Eph/ephrin system and discuss the role of this system in normal hematopoiesis before focusing on the role of Eph proteins in leukemia and other hematological malignancies and possible avenues for therapy.

Keywords: Eph receptor; Ephrin; Receptor Tyrosine Kinase; Hematopoiesis; Hematopoietic Stem Cells; Leukemia

Classification, Structure and Binding of Eph/ephrin

The Eph receptors and their membrane bound ephrin ligands represent the largest family of receptor tyrosine kinases (RTK’s). The first member, EphA1, was originally termed Eph, as it was first identified in an Erythropoietin-Producing Hepatocellular (Eph Nomenclature committee, 1997) carcinoma cell line [1]. Fourteen members of the Eph family of RTK have been identified in mammals, which are divided into two groups, the EphA and EphB family. This is based on their sequence homology, ligand specificity and structural features. In mammals there are nine members of the EphA subgroup (EphA1-8 and EphA10) and five EphB receptors (EphB1-4 and EphB6) [2,3].

Eph receptors are type I transmembrane proteins composed of extracellular region including a ligand binding domain with two distinct ligand-binding interfaces (N terminal β jelly roll domain) determining the ephrin binding, and a cysteine-rich region containing an epidermal growth factor (EGF)-like motif that is involved in receptor dimerization followed by two fibronectin type-III domains [4,5]. The intracellular region comprised of a conserved juxtamembrane domain, a tyrosine kinase domain and a sterile alpha motif (SAM) domain, the latter having a potential role in receptor clustering [6,7]. Most of the Eph receptors, possess a C-terminal PDZ (Postsynaptic density protein, Disc large, Zona occludens tight junction protein) binding motif that is involved in signaling at a sub-cellular level and in the assembly of large molecular complexes [3,5,8] (Figure 1a).

leukemia-ephrin-ligand

Figure 1: Schematic representation of Eph receptors and ephrin ligand structure. (A) The structure of the Eph receptor. (B) The structure of the ephrin-A and ephrin-B ligand.

The ephrin ligands of Eph receptors are also membrane bound proteins and like the Ephs they are divided into two groups based on their structural features and preferential binding to either EphA or EphB receptors. The two classes of ephrin ligands are the A-class (ephrinA1-5) and B-class (ephrinB1-3) [3]. The two classes of the ephrin ligands have homology at their N-terminus region while their C-terminal amino acid sequence differs, the ephrin-A ligands that are bound to the plasma membrane by a glycosylphosphatidylinositol (GPI)-linker and the ephrin-B ligands, which possess a transmembrane– spanning region and a highly conserved cytoplasmic tail with a number of highly conserved tyrosine residues and a PDZ-binding motif. The conserved tyrosine residues have been shown to serve as a docking site for proteins, which mediate downstream ephrin signaling [2,9] (Figure 1b).

Generally, the EphA receptors will preferentially bind to members of the ephrin-A ligands (ephrinA1-5) while the EphB receptors will preferentially bind to the ephrin-B ligands (ephrinB1-3) [3,10]. There is a high level of binding promiscuity between the Eph receptors and the ephrin ligands, however for an individual Eph receptor there is a distinct order of affinity of ephrin interactions with affinity constants ranging from 5-500 nM [11]. For instance the EphA1 receptor binds with a high affinity to ephrin-A1 and a lower affinity to other members of the ephrin-A family, including ephrin-A3 and ephrin-A4, and shows essentially no binding to ephrin-A5 [12]. Similarly EphA3 and EphB4 have a higher affinity for ephrin-A5 and ephrin-B2 respectively than for other members of the ephrin family [11,13]. High affinity interaction is also possible between the classes, for example EphA4 binds to both ephrin-B and -A ligands and some of its most important functions depend on interaction with ephrin-B3. Another example is EphB2, which binds to ephrin-A5 as well as ephrin-B ligands [14,15] (Figure 2).

leukemia-Eph-receptor

Figure 2: Schematic of Eph receptor and ephrin binding preference.

Eph/ephrin Activation and Signaling

Eph/ephrin interactions have the capacity to initiate bidirectional signaling. In other words both Ephs and ephrins can act as a ligand as well as a receptor and they both have the ability to initiate signaling. Signaling initiated by the Eph receptor is referred to as forward signaling whereas signaling initiated by the ephrin ligand is termed reverse signaling.

Eph receptor activation occurs following the physical association of the receptor and an ephrin ligand on adjacent cell surfaces mediated by the high affinity binding site (dimerization), this is followed by interaction with a second Eph/ephrin complex, mediated by separate low affinity (tetramerization) binding site to create a hetero-tetrameric complex [16]. The tetramers are then assembled into higher order clusters, which appear to be required for effective forward signaling [17,18]. The clustering of Eph receptors results in ligand-dependent auto-phosphorylation of several tyrosine residues within the cytoplasmic kinase domain and juxtamembrane region of the receptor, which serve as docking sites for downstream signaling proteins including the small GTPases of the Rho and Ras family, focal adhesion kinase (FAK), the Janus kinase/signal transducers and activators of transcription (Jak/Stat) and phosphatidylinositol 3-kinase (P13K) pathways [19].

Eph receptor activation influence cell shape and motility through the regulation of the Rho GTPases, including RhoA, Cdc42, Rac and Ras, via interactions with specific Rho GTPase activating (Rho GAP) and exchange (Rho GEF) factors. The interaction with individual Rho GTPases, mediated via direct binding to the cytoplasmic domain of Eph proteins of Rho GAP and Rho GEF or their recruitment via adaptor proteins, thereby mediating different effects on actin dynamics and cell process formation thus regulating cell shape and movement. For example, activation of EphA4 receptor results in recruitment of ephexin (Rho GEF), leading to activation of RhoA, and α-chimaerin (Rho GAP), leading to inactivation of Rac, jointly resulting in actin depolymerization and retraction of cell processes [15]. Activation through EphB plays a role in actin filament extension, morphogenesis and maturation of dendritic spines. Ras family of GTPases (H- and R- Ras) are also activated through Eph receptors however, unlike the Rho GTPases, the majority of Eph receptors negatively regulate the Ras/Mitogen-activated protein kinase (Ras/MAPK) pathway, with activation normally resulting in regulation of proliferation and migration [15].

Eph receptors are also important in mediating a number of other molecules involved in cell migration and adhesion due to their ability to regulate signal transduction molecules including integrin signaling pathway elements paxillin, FAK, P130 (Cas) and integrins themselves. The effect of Eph signaling on integrins is complex as they can mediate either promoting or suppressing effects. FAK is important in mediating integrin signaling and Eph receptors can down-regulate this pathway [20].

The Jak/Stat pathway, involved in cell growth and viability, is regulated by EphA receptor activation [21]. EphB receptors mediate cell migration and proliferation through P13K pathway and the protein kinase B/phosphatidylinositol 3-kinase (Akt/ PI3K) pathway is involved in the regulation of cell proliferation and viability. Eph/ephrin also regulates other signaling pathways including Abl/Arg and p53-family of tumor suppressor proteins [19]. A recent study in glioblastoma multiforme (GBM) shows that loss of EphA3 results in elevated MAPK signaling thereby inducing differentiation and reducing proliferation and self-renewal. This study showed that regulation of extracellular signal-regulated kinases/mitogen-activated protein kinases (ERK/ MAPK) signaling by EphA3 is kinase independent of the upstream activators of MAPK signaling [22]. A similar finding has been made for GBM which express EphA2 at high levels [23].

In the ephrin-expressing cells reverse signaling can be induced after Eph/ephrin interaction [19,24]. Ephrin-B reverse signaling partly depends on the tyrosine phosphorylation of conserved residues in the cytoplasmic region, where phosphorylation is mediated by associated tyrosine kinases, most notably members of the Src family (Src, Fyn, Lyn, Yes) of tyrosine kinases. When the ephrin-B ligand is phosphorylated it can bind to cytosolic adaptor molecules via (Src Homology-2) SH-2 and SH-3 domains or PDZ domains [8]. The protein tyrosine phosphatase basophil-like (PTP-BL) has been identified as a negative regulator of ephrin-B signaling and it binds to ephrin-B through its C-terminal PDZ binding motif. De-phosphorylation of the ephrin-B cytoplasmic domain can inactivate the Src family of kinase and therefore cause termination of reverse signaling [25]. In the activated ephrin-B ligand, the PDZ motif plays an important role in assembly of other signaling molecules. Ephrin-B binds to cytoplasmic protein PDZ-RGS3, which contains a PDZ domain and a regulator of G-protein signaling (RGS) domain. Activation of the ephrin-B ligand by EphB receptor via the PDZ-RGS interferes with signaling of the stromal derived factor (SDF)- 1 via its G-protein coupled receptor and the chemokine receptor-4 (CXCR4). Ephrin-B reverse signaling has thus been implicated with regulation of migration in cerebellar development [26].

The ephrin-A reverse signaling mechanism is not well understood, however it is likely that the signaling response is initiated by activity of the Src family of RTK in which transmembrane adaptor molecules, associating with the lipid anchor of the ephrin-A proteins, transmit the signal across the membrane. For example ephrin-A5 induces signaling within the ephrin-A expressing cell when bound to its cognate Eph receptor [27-29].

Expression and Function of the Eph and Ephrin Genes

Eph signaling controls cell adhesion, migration, invasion and morphology by influencing integrin and intercellular adhesion molecule activity and by modification of actin cytoskeleton organization as described above. Through these mechanisms Eph function effects not only the processes of embryogenesis but also specialized cellular function in adult tissues, including bone remodeling, immune function and synaptic plasticity as well as cell proliferation and survival specific tissue stem cells as described further below [27,30].

Both Eph and ephrin proteins are important in regulating cell-cell interactions and their interaction can initiate either cell adhesion or repulsion. Cell repulsion occurs when bidirectional signaling triggers cytoskeletal contraction, loss of focal adhesions, cell rounding and cell detachment, whereas cell attachment occurs when signals are in favor of cell adhesion and migration [31]. This interaction influence many different cell behaviors during embryogenesis and in adult life [27]. Eph/ephrin interactions mediate formation of tissue boundaries (e.g. hindbrain rhombomeres) [32], control axon guidance during development and also tissue morphogenesis and patterning [33,34]. Eph/ephrin interactions are also involved in development of vascular system [35], stem cell biology [36], hematopoiesis, erythropoiesis, immune function and in tumor invasion and metastasis [27,30,37-39].

Many members of the Eph/ephrin family are expressed at high levels in some cancer cells and also elements of the tumor microenvironment, where they influence tumor growth and spread. Specific examples include lung, breast and prostate cancer, as well as melanoma, sarcomas and leukemias [40,41]. There is evidence that the Eph receptors can have either tumor suppressing or tumor promoting activity, depending on the tissue and their expression pattern [27]. Thus, the role and function of Eph/ephrins in cancer is not yet fully elucidated as some tumors show an elevated level of Eph expression while others show a decrease in Eph expression and as yet no single model of their function encompasses all cancers. For example EphA2 is up regulated in many cancers including breast and prostate and its expression is linked to an increase in malignancy [42,43] but it is down regulated in colon cancer [44]. Similarly, EphA1 expression is up-regulated in ovarian cancer [40] but down-regulated in advanced skin and colorectal cancers [45,46]. Studies have also shown the role of EphA7 as a tumor suppressor in follicular lymphoma [47], tumor suppressor function has also been reported for EphB receptors, including EphB2 and EphB3 in colorectal cancer [30,48]. EphB4 is another important example as this gene can act as either a tumor suppressor or an oncogene in different facets of breast cancer progression [49,50]. Table 1 represents the expression of Eph receptors in normal and malignant tissues [51-82].

Eph Tissue expression Expression in cancer References
EphA1 Widely expressed in mouse epithelial tissues evidence of expression in hematopoietic progenitors Over expressed in many different cancers including hepatocellular, prostate, lung, gastric and colon cancer
Down regulated in non-melanoma skin cancer, colorectal cancer
1,12,24,45,46,51-55
EphA2 Expressed highly in adult human epithelial cells and endothelium Over expressed in prostate, breast, melanoma, lung and ovarian cancers
Up-regulated in glioblastoma
Down-regulated in colon cancer
23,24,42,43,54-61
EphA3 Expressed in various stages of embryonic development and in adult central nervous system Expressed in neural cancers, leukemia, lymphomas and sarcomas
Up-regulated in lung, brain, liver and kidney
Over expressed in melanoma
24,54,55,62-66
EphA4 Expressed in development, final stages of embryogenesis and central nervous system Expressed in prostate, pancreatic cancer
Up-regulated in lung cancer
Down-regulated kidney
Over expressed in gastric cancer
24,54,55,62,67-70
EphA5 Expressed in nervous system Expressed in neuroblastomas and neural cancer
Down-regulated in breast cancer
24,54,55,70-72
EphA6 Expressed more prominently in adult tissues than in embryonic tissues Down-regulated in colon cancer and renal carcinoma
Up-regulated in lung and liver cancer
24,54,55
EphA7 Expressed in developing neural tubes, thymus, lymphoid tissues and fetal bone marrow Expressed in colorectal cancer, lung and follicular lymphoma
Up-regulated in ALL1 leukemia
24,44,47,54,55,73-75
EphA8 Expressed in spinal cord and neuronal cells Expressed in colon cancer
Down-regulated in glioblastoma
24,54,55
EphA10 Expressed in testis Over expressed in breast cancer 76,77
EphB1 Expressed in Brain and colon Expressed in lung cancer
Down-regulated in colon carcinoma and in kidney cancer
24,46,54
EphB2 Expressed in epithelial cells, thymus, lymphoid, osteoblastic and osteoclastic cells Over expressed in gastrointestinal
Expressed in colon, ovarian and lung cancer
Up-regulated in colorectal, kidney and hepatocellular cancer
24,54,78
EphB3 Expressed in various tissues Expressed in prostate, lung and melanoma 24,54
EphB4 Expressed in placenta and in range of primary tissues including brain, endothelium, hematopoietic cells Expressed in colon, endometrial, breast, neuroblastoma, glioblastoma and leukemia and lymphoma cancer 24,54,79-81
EphB6 Expressed in various tissues including brain, pancreas, thymus and T-cells Expressed in T cell tumors and leukemia
Up-regulated in colon cancer
Down-regulated in breast, lung and kidney cancer
24,54,82

Table 1: Eph expression in normal and malignant tissues.

The Eph/ephrin in HSC and Leukemia

Eph/ephrin expression in HSC and progenitors

The expression of Eph/ephrin has been detected on purified population of hematopoietic stem cells (HSCs) in both human and mouse. Gene expression analysis of HSC showed expression of ephrin-B2, indicating that it may be involved in signaling between HSC and their microenvironment. Other array based studies on primary human HSC (CD34+ hematopoietic cells) shows expression of the EphA1 protein and its ligands ephrin-A3 and ephrin-A4 suggesting that their interaction may play a role in hematopoietic stem and progenitor cell positioning and function [83,84]. Further analysis of CD133+ and CD34+ hematopoietic stem cells in peripheral blood showed expression of EphA2 in all CD34+ cells and the majority of CD133+ cells however EphB2 was expressed in all CD133+ cells and fifty percent of CD34+ cells, these data suggest that a number of elements of the Eph/ephrin system may have a role in HSC function through regulatory effects on cell adhesion, migration and differentiation but also that there may be a degree of functional redundancy between several Eph proteins [85].

Real-time quantitative PCR of mouse Lin-ckit+sca1+ (KLS) showed detectable expression of all EphA receptors except EphA6 and EphA8, along with ephrin-A ligands, with ephrin-A4 and ephrin-A5 being the most highly expressed ligands on purified HSCs in the mouse bone marrow [86]. Flow cytometric analysis of EphA2, A3, A4 and A5 along with ephrinA1-5 showed that EphA2 and EphA3 were the highest expressing EphA receptors. Expression of EphA2, A3, A4 and A5 was also detected in the mouse stromal cell lines however human stromal cell line showed only EphA2 expression at moderate levels [86]. Whilst the function has not been fully investigated, a role in HSC trafficking was demonstrated by treatment of mice with an Eph/ephrin inhibitor, EphA3-Fc, which resulted in mobilization of bone marrow progenitor cells into peripheral blood [86].

Some of the members of the Eph/ephrin family are also involved in development and regulation of mature hematopoietic cells. For example, EphA4 and EphB1 receptors along with ephrin-B1 ligand are expressed on human platelets [87], these studies also shows that EphA4 is involved in regulation of platelet aggregation and adhesion to fibrinogen, a process dependent on integrin αIIbβ3 engagement [39].

EphB4 was originally identified on human bone marrow CD34+ cells and its expression has been reported on erythroid progenitor cells in early stages of red blood cell development. Significantly, the EphB4 ligand, ephrin-B2, is expressed on bone marrow stromal cells [88] where it has been reported to be involved with regulating erythropoiesis via interaction with EphB4 [89]. Studies by Suenobu et al. [89] showed co-culturing hematopoietic progenitor cells expressing EphB4 with stromal cells expressing ephrin-B2 results in hematopoietic progenitor cells detachment from stromal layer and differentiation into a mature erythroid cells accompanied by EphB4 down regulation, however coculturing these cell with ephrin-B2-negative stromal cells resulted in less maturation of erythroid cells and no change to EphB4 expression [89]. Ephrin-B2 ectopic expression in stromal cells increased adhesion of hematopoietic cells to stromal calls and decreased transmigration of hematopoietic cells beneath a stromal cell monolayer. These findings strongly support a role for the EphB4/ephrin-B2 interaction in migration and colonization of stem/progenitors cells in the bone marrow microenvironment [90].

Some of the Eph/ephrin molecules are also involved in lymphoid development. The expression of Eph/ephrin has been studied extensively in T-lymphocytes and expression of some of the members of this family including EphA1, EphA2, EphA3, EphA4, EphA7, EphB2, EphB6, ephrin-A1 ephrin-A3, ephrin-A5 and ephrin-B1 has been reported in the thymus, both on thymic stroma and lymphoid cells suggesting a role in T-cell development [91-93]. Interestingly, there are no reports of defective T lineage development in knockout mice, perhaps a result of there being multiple Eph receptors with overlapping functions in the T cell compartment.

As well as T-lymphocytes the expression of Eph/ephrin gene has been also reported in B-lymphocytes [38,94,95]. EphA7 and EphA4 transcripts were found in human fetal bone marrow pro-B and pre-B cells. EphA4 expression is found in both adult and fetal pro-B and pre-B lineage cells with high levels of expression in peripheral blood. The full length EphA7 transcript, however, was not found in mature fetal B-lineage and adult B-lineage cells. This suggests that EphA7 may be involved in expansion and /or differentiation of pre/pro B-cell but is lost on mature B cells [95]. Further studies show that there are different types of EphA7 mRNA, one of which encodes the full length EphA7 and another splice variant encodes a truncated, soluble protein that lacks the cytoplasmic domain. Studies by Dawson et al. [74] showed that normal lymphocytes express and secrete the truncated form of EphA7 [74], which have been shown to have tumor suppressive effects in lymphoma [47].

Eph/Ephrin in vascular development

In the context of hematopoiesis, the vascular system is crucial in development of blood cells within the bone marrow and also in the function and migration of mature hematopoietic cells. The vascular system arises from two distinct processes known as vasculogenesis and angiogenesis. Vasculogenesis is an early event in embryonic development involving mesodermal cells differentiation to form a vascular plexus in embryonic tissues and angiogenesis is a process in which new blood vessels are formed from existing blood vessels. Angiogenesis has a role both in early development and in the adult hematopoietic system, it also promotes sprouting of new blood vessels in embryonic and postnatal vasculature and it has been shown to be important in development and metastasis of solid tumors [96]. Some members of Eph/ephrin family have established roles in vasculogenesis and angiogenesis. The expression analysis of Eph/ephrin using realtime polymerase chain reaction (RT-PCR) has shown expression of EphB2, EphB3, EphB4, ephrin-B1 and ephrin-B2 in the yolk sac [97]. Ephrin-B1 expression has been detected on both arteries and veins while the high affinity ligand for EphB4, ephrin-B2, is only detected on arteries and EphB4 expression is only detected on veins. Knock out ephrin-B2 mice and some of the EphB2 and EphB3 double mutants mice have defects in embryonic vasculature and therefore these mice are embryonically lethal. EphB2 and ephrin-B2 expression in mesenchyme adjacent to vessels and the vascular defects in EphB2/EphB3 double mutants indicate a requirement for Eph/ephrin signaling between endothelial cells and surrounding mesenchymal cells [97]. EphA2 has also been reported to have a role in angiogenesis and the expression of EphA2 and its ligand ephrin-A1 has been reported in both human and mouse tumor vasculature [61].

Eph/Ephrin on bone remodeling and formation

The bone marrow is the principal site of hematopoiesis in adult animals and requires both vascular and other stromal cell types to create the hematopoietic niche. Important amongst these are the osteoblasts and osteoclasts, which mediate bone formation and remodeling. Bone is constantly remodeled through resorption of mineralized bone by osteoclast and formation of new bone by osteoblast. Coupling of bone resorption and formation is critical during normal bone remodeling and it is necessary for bone growth, any deregulation in this process will result in pathological bone disease [19,78].

Eph receptors and ephrin ligands are important in bone remodeling and homeostasis during this process Eph/ephrin bidirectional signaling regulates differentiation and function of the bone cells. Real time PCR (RT-PCR) analysis of the Eph/ephrin showed mRNA expression of ephrin ligands, ephrin-B1, ephrin-B2 and ephrin-A1, A2, A4 and A5, as well as Eph receptors including EphB2-4, EphB6, EphA2-4 and EphA7 receptors on osteoblastic and osteoclastic cells [98-100]. Expression of EphB4 is observed on the osteoblasts and forward signaling through EphB4 results in bone formation and reverse signaling through ephrin-B2 inhibit bone resorption therefore ephrin-B2/EphB4 act as coupling stimulator [98]. Expression of ephrin-A2 was observed during early osteoclastogenesis and unlike the ephrin-B2 it acts as coupling inhibitor as reverse signaling through ephrin-A2 result osteoclastogenesis and EphA2 forward signaling into osteoblast inhibit osteoblastic bone formation and mineralization [99]. Ephrin-A2/ EphA2 bidirectional signaling facilitates bone remodeling at initiation phase, forward signaling through EphA2 receptor on osteoblast inhibit osteoblastic differentiation and bone formation and reverse signaling into osteoclast through ephrin-A2 promote osteoclast differentiation [78,99]. Ephrin-B1 full knockout mice are prenatally lethal and they have skeletal defects. Studies on disruption of ephrin-B1 on collagen I producing cells result in reduced bone formation and skull defect and studies on ephrin-B1 conditional knockout mice shows defects in osteoblastic mediated bone formation with no increase in osteoclastic bone resorption and this condition results in reduction in bone size and density [101,102].

The importance of Eph/ephrin interactions has also been shown in various stem cell niches, including neural, dental and intestinal stem cell compartments [103,104]. More recent studies show their involvement in bone homeostasis and mesenchymal stem cell (MSC) regulation. Arthur et al. [104] showed increase in osteogenic differentiation upon ephrin-B1 and/or ephrin-B2 expression by MSC. They also showed that ephrin-B1 activation promoted chondrogenic differentiation; therefore EphB/ephrin-B interactions may be involved in recruitment, migration and differentiation of MSC during bone repair [104]. Studies by Ting et al. [86] shows that ephrin-A signaling interact with stem/progenitor cells in the bone marrow niche as it’s signaling mediates the release of progenitor cells from hematopoietic niche [86].

Interestingly, Eph/ephrin interactions are also involved in bone malignancies and tumors, osteocarcinoma is a malignant bone tumor in adolescence and microarray analysis studies show increased expression of EphA2, EphA4, ephrin-B1 and ephrin-A1 in osteosarcoma cells [105,106].

Eph/Ephrin expression in leukemia and other hematopoietic tumors

Both chronic and acute myeloid leukemia are malignant diseases of the hematopoietic system which in most cases are believed to arise through the abnormal proliferation of either uncommitted or partially committed HSC [107]. The origin of other types of leukemia such as promyelocytic leukemia, pre-B acute lymphoblastic leukemia (ALL) and T-ALL and chronic lymphoblastic leukemia (CLL) are more likely due to malignant transformation of more mature progenitor cells. Expression of elements of the Eph/ephrin system has been detected on many types of human leukemia. One of the best studied is EphA3, which was originally identified in the LK63 pre-B ALL cell line and further investigations revealed its expression in T-cell leukemia cell lines such as Jurkat, JM and HSB-2 [65,108]. It has been shown that EphA3 can induce both adhesive and cell repulsive responses in different cell types [109]. In analyzing ephrin induced cell adhesion in LK63 cells, a critical role was identified for protein phosphatase activity, which prevented EphA3 phosphorylation and hence maintained the Eph/ephrin adhesive bond and prevented initiation of the signaling mechanisms leading to cell repulsion [31]. In leukemia, EphA3 is expressed at significantly higher levels compared to normal blood cells, elevated expression of EphA3 being detected in a proportion of clinical samples from cases of lymphoid and myeloid leukemias 111. Elevated EphA3 expression has also been detected on other cancers such as lung cancer, melanoma and brain tumors [22,64,65,109], whereas expression was found to be absent or low in corresponding normal tissues [54,65,110]. Recent array based studies also showed EphA3 as one of the genes with copy number alteration (CNA) in the genome of acute myeloid leukemia (AML) patients [111]. Further studies by Guan et al. [112] showed copy number variation (CNV) of EphA3 to be associated with various types hematological malignancies and therefore CNV of EphA3 could be used as a diagnostic indicator for different types of leukemia [112]. Many cancers, including leukemia, require multiple cooperative oncogene mutations for malignant cell transformation. Specific sets of synergistically dysregulated by cooperative oncogenes are known as cooperative response gene (CRGs), which regulate leukemia stem cell (LSC) growth and survival. Studies by Ashton et al. [113], where stem cells were retrovirally transduced with two fusion genes found in human myeloid leukemias. NUP98-HOXA9 and BCR-ABL, have identified EphA3 as a common CRG. They showed that shRNA knock down of EphA3 in leukemic stem cells reduced leukemic cell engraftment, concluding that this gene may be responsible for leukemia stem cell growth and survival in bone marrow microenvironment [113]. With the involvement of EphA3 in many different types of leukemia a high affinity monoclonal antibody to EphA3 (IIIA4) [65] has been fully humaneered by Kalabios and the resulting antibody, KB004, is now in phase I clinical trial in leukemia and other hematological cancers [114].

As mentioned previously EphB4 was originally identified in human bone marrow CD34+ cells and its expression been reported in erythroid progenitor cells, however it’s ligand ephrin-B2 is expressed in bone marrow stromal cells [88]. Co-expression of EphB4 and ephrin-B2 is found in the yolk sac, which is the first site of hematopoiesis and vascular development during embryogenesis. EphB4/ephrin-B2 expression has been shown in the majority of leukemia and lymphoma cell lines although expression in clinical samples appears less prominent [81]. Antibodies to EphB4 have undergone extensive pre-clinical evaluation and shown good anti-tumor effects in solid tumors, which over-express EphB4 and by inhibition of angiogenesis, although no efficacy has been shown in hematopoietic tumors to date [115]. Nevertheless, these antibodies may have the potential to be developed to target EphB4 in leukemia and related blood cancers.

Studies by Nakanishi et al. [75] shows EphA7 up regulation in the ALL1 associated leukemia (ALL1/AF4 and ALL1/AF9). They also showed that EphA7 up-regulation was associated with phosphorylation of ERK and treatment with a phosphorylated ERK blocking drug resulted in apoptotic cell death in ALL1/AF4 leukemic blast cells [75]. Thus, anti-EphA7 antibodies or other inhibitors may well have a role in leukemia associated with this translocation. In contra-distinction to this positive role in leukemia, EphA7 is lost in lymphomas, where the gene is hypermethylated and repressed in germinal center B-cell non-Hodgkins lymphomas and this has a potential to influence tumor progression and spread [74]. In this study a soluble form of EphA7 was shown to inhibit lymphoma in a mouse model, a chimeric protein consisting of soluble EphA7 and CD20 antibody had still greater therapeutic effect. Further studies show EphA7 as targeted tumor suppressor gene in T-cell lymphoblastic leukemia and lymphoma (T-LBL) and follicular B cell lymphoma [47,116].

EphB6 expression has been observed in normal human tissue and over-expression of EphB6 has also been reported in both myeloid [117] and lymphoid leukemias [82,118]. The expression level of EphB6 decreases with maturation of the cells in T-cell derived leukemiacells, therefore suggesting that EphB6 expression regulates T-cell development but has less significant role in mature T cells [82]. To date there are no reports of experimental therapies targeting EphB6. Table 2 [110,119,120] represents the summary of Eph receptors used as therapy target in various malignancies.

Eph Targeted therapy References
EphA1 Tumor suppressor in colorectal cancer 44
EphA2 EphA2 targeting reagents in ovarian cancer therapy 119
EphA3 Therapeutic target in leukemia and glioblastoma 22,110
EphA7 Tumor suppressor in T-LBL and follicular lymphoma 47,120
EphB4 EphB4 antibody to inhibit solid tumor growth 115

Table 2: Eph receptors as a therapy target for cancer.

Summary

In summary, the aberrant expression of Eph receptors in hematopoietic tumors reflects the spectrum of functions of these receptors in all cancers. In some cancers these genes act as tumor suppressor, examples being EphA1 in colorectal cancer and EphA7 in follicular lymphomas. On the other hand these proteins can also have oncogenic effects, examples being the expression of EphA2 and EphA3 in glioma and the over-expression of EphA3 in leukemia. In terms of therapy, the over-expression in certain tumors, taken together with the surface expression of these proteins, makes a strong case for targeted therapies. This is particularly the case when expression on normal tissues is minimal; this is exemplified by EphA3 and EphB4 where no toxicity was evident in pre-clinical testing of potential therapeutic antibodies. These studies reveal the therapeutic potential of targeting components of the Eph/ephrin system in leukemia and other cancers. These results should prompt further research into the specific roles of these proteins in different cancers as a prelude to designing and optimizing the therapeutic targeting of these proteins.

References

  1. Hirai H, Maru Y, Hagiwara K, Nishida J, Takaku F (1987) A novel putative tyrosine kinase receptor encoded by the eph gene. Science 238: 1717-1720.
  2. Himanen JP, Nikolov DB (2003) Eph signaling: a structural view. Trends Neurosci 26: 46-51.
  3. Pasquale EB (2005) Eph receptor signalling casts a wide net on cell behaviour. Nat Rev Mol Cell Biol 6: 462-475.
  4. Pasquale EB (1997) The Eph family of receptors. Curr Opin Cell Biol 9: 608-615.
  5. Himanen JP, Nikolov DB (2003) Eph receptors and ephrins. Int J Biochem Cell Biol 35: 130-134.
  6. Holland SJ, Gale NW, Gish GD, Roth RA, Songyang Z, et al. (1997) Juxtamembrane tyrosine residues couple the Eph family receptor EphB2/Nuk to specific SH2 domain proteins in neuronal cells. EMBO J 16: 3877-3888.
  7. Stapleton D, Balan I, Pawson T, Sicheri F (1999) The crystal structure of an Eph receptor SAM domain reveals a mechanism for modular dimerization. Nat Struct Biol 6: 44-49.
  8. Torres R, Firestein BL, Dong H, Staudinger J, Olson EN, et al. (1998) PDZ proteins bind, cluster, and synaptically colocalize with Eph receptors and their ephrin ligands. Neuron 21: 1453-1463.
  9. Cerretti DP, Vanden Bos T, Nelson N, Kozlosky CJ, Reddy P, et al. (1995) Isolation of LERK-5: a ligand of the eph-related receptor tyrosine kinases. Mol Immunol 32: 1197-1205.
  10. Boyd AW, Lackmann M (2001) Signals from Eph and ephrin proteins: a developmental tool kit. Sci STKE 2001: re20.
  11. Lackmann M, Mann RJ, Kravets L, Smith FM, Bucci TA, et al. (1997) Ligand for EPH-related kinase (LERK) 7 is the preferred high affinity ligand for the HEK receptor. J Biol Chem 272: 16521-16530.
  12. Coulthard MG, Lickliter JD, Subanesan N, Chen K, Webb GC, et al. (2001) Characterization of the Epha1 receptor tyrosine kinase: expression in epithelial tissues. Growth Factors 18: 303-317.
  13. Blits-Huizinga CT, Nelersa CM, Malhotra A, Liebl DJ (2004) Ephrins and their receptors: binding versus biology. IUBMB Life 56: 257-265.
  14. Mosch B, Reissenweber B, Neuber C, Pietzsch J (2010) Eph receptors and ephrin ligands: important players in angiogenesis and tumor angiogenesis. J Oncol 2010: 135285.
  15. Lackmann M, Boyd AW (2008) Eph, a protein family coming of age: more confusion, insight, or complexity? Sci Signal 1: re2.
  16. Himanen JP, Rajashankar KR, Lackmann M, Cowan CA, Henkemeyer M, et al. (2001) Crystal structure of an Eph receptor-ephrin complex. Nature 414: 933-938.
  17. Wimmer-Kleikamp SH, Janes PW, Squire A, Bastiaens PI, Lackmann M (2004) Recruitment of Eph receptors into signaling clusters does not require ephrin contact. J Cell Biol 164: 661-666.
  18. Smith FM, Vearing C, Lackmann M, Treutlein H, Himanen J, et al. (2004) Dissecting the EphA3/Ephrin-A5 interactions using a novel functional mutagenesis screen. J Biol Chem 279: 9522-9531.
  19. Edwards CM, Mundy GR (2008) Eph receptors and ephrin signaling pathways: a role in bone homeostasis. Int J Med Sci 5: 263-272.
  20. Miao H, Burnett E, Kinch M, Simon E, Wang B (2000) Activation of EphA2 kinase suppresses integrin function and causes focal-adhesion-kinase dephosphorylation. Nat Cell Biol 2: 62-69.
  21. Lai KO, Chen Y, Po HM, Lok KC, Gong K, et al. (2004) Identification of the Jak/Stat proteins as novel downstream targets of EphA4 signaling in muscle: implications in the regulation of acetylcholinesterase expression. J Biol Chem 279: 13383-13392.
  22. Day BW, Stringer BW, Al-Ejeh F, Ting MJ, Wilson J, et al. (2013) EphA3 maintains tumorigenicity and is a therapeutic target in glioblastoma multiforme. Cancer Cell 23: 238-248.
  23. Binda E, Visioli A, Giani F, Lamorte G, Copetti M, et al. (2012) The EphA2 receptor drives self-renewal and tumorigenicity in stem-like tumor-propagating cells from human glioblastomas. Cancer Cell 22: 765-780.
  24. Surawska H, Ma PC, Salgia R (2004) The role of ephrins and Eph receptors in cancer. Cytokine Growth Factor Rev 15: 419-433.
  25. Palmer A, Zimmer M, Erdmann KS, Eulenburg V, Porthin A, et al. (2002) EphrinB phosphorylation and reverse signaling: regulation by Src kinases and PTP-BL phosphatase. Mol Cell 9: 725-737.
  26. Lu Q, Sun EE, Klein RS, Flanagan JG (2001) Ephrin-B reverse signaling is mediated by a novel PDZ-RGS protein and selectively inhibits G protein-coupled chemoattraction. Cell 105: 69-79.
  27. Pasquale EB (2008) Eph-ephrin bidirectional signaling in physiology and disease. Cell 133: 38-52.
  28. Davy A, Gale NW, Murray EW, Klinghoffer RA, Soriano P, et al. (1999) Compartmentalized signaling by GPI-anchored ephrin-A5 requires the Fyn tyrosine kinase to regulate cellular adhesion. Genes Dev 13: 3125-3135.
  29. Davy A, Robbins SM (2000) Ephrin-A5 modulates cell adhesion and morphology in an integrin-dependent manner. EMBO J 19: 5396-5405.
  30. Pasquale EB (2010) Eph receptors and ephrins in cancer: bidirectional signalling and beyond. Nat Rev Cancer 10: 165-180.
  31. Wimmer-Kleikamp SH, Nievergall E, Gegenbauer K, Adikari S, Mansour M, et al. (2008) Elevated protein tyrosine phosphatase activity provokes Eph/ephrin-facilitated adhesion of pre-B leukemia cells. Blood 112: 721-732.
  32. Dahmann C, Oates AC, Brand M (2011) Boundary formation and maintenance in tissue development. Nat Rev Genet 12: 43-55.
  33. Scicolone G, Ortalli AL, Carri NG (2009) Key roles of Ephs and ephrins in retinotectal topographic map formation. Brain Res Bull 79: 227-247.
  34. Coulthard MG, Duffy S, Down M, Evans B, Power M, et al. (2002) The role of the Eph-ephrin signalling system in the regulation of developmental patterning. Int J Dev Biol 46: 375-384.
  35. Adams RH, Eichmann A (2010) Axon guidance molecules in vascular patterning. Cold Spring Harb Perspect Biol 2: a001875.
  36. Murai KK, Pasquale EB (2010) Restraining stem cell niche plasticity: a new talent of Eph receptors. Cell Stem Cell 7: 647-648.
  37. Rivera J, Lozano ML, Navarro-Núñez L, Vicente V (2009) Platelet receptors and signaling in the dynamics of thrombus formation. Haematologica 94: 700-711.
  38. Ting MJ, Boyd AW (2008) The Role of Eph Receptor Tyrosine Kinases and Ephrin Ligands in Hematopoietic Cell Development and Function. The Open Hematology Journal 2: 103-110.
  39. Prévost N, Woulfe DS, Jiang H, Stalker TJ, Marchese P, et al. (2005) Eph kinases and ephrins support thrombus growth and stability by regulating integrin outside-in signaling in platelets. Proc Natl Acad Sci U S A 102: 9820-9825.
  40. Herath NI, Spanevello MD, Sabesan S, Newton T, Cummings M, et al. (2006) Over-expression of Eph and ephrin genes in advanced ovarian cancer: ephrin gene expression correlates with shortened survival. BMC Cancer 6: 144.
  41. Noberini R, Pasquale EB (2009) Proliferation and tumor suppression: not mutually exclusive for Eph receptors. Cancer Cell 16: 452-454.
  42. Walker-Daniels J, Coffman K, Azimi M, Rhim JS, Bostwick DG, et al. (1999) Overexpression of the EphA2 tyrosine kinase in prostate cancer. Prostate 41: 275-280.
  43. Zelinski DP, Zantek ND, Stewart JC, Irizarry AR, Kinch MS (2001) EphA2 overexpression causes tumorigenesis of mammary epithelial cells. Cancer Res 61: 2301-2306.
  44. Herath NI, Spanevello MD, Doecke JD, Smith FM, Pouponnot C, et al. (2012) Complex expression patterns of Eph receptor tyrosine kinases and their ephrin ligands in colorectal carcinogenesis. Eur J Cancer 48: 753-762.
  45. Hafner C, Becker B, Landthaler M, Vogt T (2006) Expression profile of Eph receptors and ephrin ligands in human skin and downregulation of EphA1 in nonmelanoma skin cancer. Mod Pathol 19: 1369-1377.
  46. Herath NI, Doecke J, Spanevello MD, Leggett BA, Boyd AW (2009) Epigenetic silencing of EphA1 expression in colorectal cancer is correlated with poor survival. Br J Cancer 100: 1095-1102.
  47. Oricchio E, Nanjangud G, Wolfe AL, Schatz JH, Mavrakis KJ, et al. (2011) The Eph-receptor A7 is a soluble tumor suppressor for follicular lymphoma. Cell 147: 554-564.
  48. Schneikert J, Behrens J (2007) The canonical Wnt signalling pathway and its APC partner in colon cancer development. Gut 56: 417-425.
  49. Noren NK, Foos G, Hauser CA, Pasquale EB (2006) The EphB4 receptor suppresses breast cancer cell tumorigenicity through an Abl-Crk pathway. Nat Cell Biol 8: 815-825.
  50. Noren NK, Pasquale EB (2007) Paradoxes of the EphB4 receptor in cancer. Cancer Res 67: 3994-3997.
  51. Nakamoto M, Bergemann AD (2002) Diverse roles for the Eph family of receptor tyrosine kinases in carcinogenesis. Microsc Res Tech 59: 58-67.
  52. Robinson D, He F, Pretlow T, Kung HJ (1996) A tyrosine kinase profile of prostate carcinoma. Proc Natl Acad Sci U S A 93: 5958-5962.
  53. Kao HW, Chen HC, Wu CW, Lin WC (2003) Tyrosine-kinase expression profiles in human gastric cancer cell lines and their modulations with retinoic acids. Br J Cancer 88: 1058-1064.
  54. Hafner C, Schmitz G, Meyer S, Bataille F, Hau P, et al. (2004) Differential gene expression of Eph receptors and ephrins in benign human tissues and cancers. Clin Chem 50: 490-499.
  55. Böhme B, Holtrich U, Wolf G, Luzius H, Grzeschik KH, et al. (1993) PCR mediated detection of a new human receptor-tyrosine-kinase, HEK 2. Oncogene 8: 2857-2862.
  56. Coulthard MG, Morgan M, Woodruff TM, Arumugam TV, Taylor SM, et al. (2012) Eph/Ephrin signaling in injury and inflammation. Am J Pathol 181: 1493-1503.
  57. Hess AR, Seftor EA, Gardner LM, Carles-Kinch K, Schneider GB, et al. (2001) Molecular regulation of tumor cell vasculogenic mimicry by tyrosine phosphorylation: role of epithelial cell kinase (Eck/EphA2). Cancer Res 61: 3250-3255.
  58. Kinch MS, Moore MB, Harpole DH Jr (2003) Predictive value of the EphA2 receptor tyrosine kinase in lung cancer recurrence and survival. Clin Cancer Res 9: 613-618.
  59. Thaker PH, Deavers M, Celestino J, Thornton A, Fletcher MS, et al. (2004) EphA2 expression is associated with aggressive features in ovarian carcinoma. Clin Cancer Res 10: 5145-5150.
  60. Easty DJ, Herlyn M, Bennett DC (1995) Abnormal protein tyrosine kinase gene expression during melanoma progression and metastasis. Int J Cancer 60: 129-136.
  61. Ogawa K, Pasqualini R, Lindberg RA, Kain R, Freeman AL, et al. (2000) The ephrin-A1 ligand and its receptor, EphA2, are expressed during tumor neovascularization. Oncogene 19: 6043-6052.
  62. Kilpatrick TJ, Brown A, Lai C, Gassmann M, Goulding M, et al. (1996) Expression of the Tyro4/Mek4/Cek4 gene specifically marks a subset of embryonic motor neurons and their muscle targets. Mol Cell Neurosci 7: 62-74.
  63. Dottori M, Down M, Hüttmann A, Fitzpatrick DR, Boyd AW (1999) Cloning and characterization of EphA3 (Hek) gene promoter: DNA methylation regulates expression in hematopoietic tumor cells. Blood 94: 2477-2486.
  64. Chiari R, Hames G, Stroobant V, Texier C, Maillère B, et al. (2000) Identification of a tumor-specific shared antigen derived from an Eph receptor and presented to CD4 T cells on HLA class II molecules. Cancer Res 60: 4855-4863.
  65. Boyd AW, Ward LD, Wicks IP, Simpson RJ, Salvaris E, et al. (1992) Isolation and characterization of a novel receptor-type protein tyrosine kinase (hek) from a human pre-B cell line. J Biol Chem 267: 3262-3267.
  66. Salvaris E, Novotny JR, Welch K, Campbell L, Boyd AW (1992) Characterization of two novel pre-B-cell lines (LK63 and LiLa-1): potential models of pre-B-cell differentiation. Leuk Res 16: 655-663.
  67. Iiizumi M, Hosokawa M, Takehara A, Chung S, Nakamura T, et al. (2006) EphA4 receptor, overexpressed in pancreatic ductal adenocarcinoma, promotes cancer cell growth. Cancer Sci 97: 1211-1216.
  68. Dottori M, Hartley L, Galea M, Paxinos G, Polizzotto M, et al. (1998) EphA4 (Sek1) receptor tyrosine kinase is required for the development of the corticospinal tract. Proc Natl Acad Sci U S A 95: 13248-13253.
  69. Oki M, Yamamoto H, Taniguchi H, Adachi Y, Imai K, et al. (2008) Overexpression of the receptor tyrosine kinase EphA4 in human gastric cancers. World J Gastroenterol 14: 5650-5656.
  70. Olivieri G, Miescher GC (1999) Immunohistochemical localization of EphA5 in the adult human central nervous system. J Histochem Cytochem 47: 855-861.
  71. Fu DY, Wang ZM, Wang BL, Chen L, Yang WT, et al. (2010) Frequent epigenetic inactivation of the receptor tyrosine kinase EphA5 by promoter methylation in human breast cancer. Hum Pathol 41: 48-58.
  72. Szabó PM, Pintér M, Szabó DR, Zsippai A, Patócs A, et al. (2012) Integrative analysis of neuroblastoma and pheochromocytoma genomics data. BMC Med Genomics 5: 48.
  73. Vergara-Silva A, Schaefer KL, Berg LJ (2002) Compartmentalized Eph receptor and ephrin expression in the thymus. Mech Dev 119 Suppl 1: S225-229.
  74. Dawson DW, Hong JS, Shen RR, French SW, Troke JJ, et al. (2007) Global DNA methylation profiling reveals silencing of a secreted form of Epha7 in mouse and human germinal center B-cell lymphomas. Oncogene 26: 4243-4252.
  75. Nakanishi H, Nakamura T, Canaani E, Croce CM (2007) ALL1 fusion proteins induce deregulation of EphA7 and ERK phosphorylation in human acute leukemias. Proc Natl Acad Sci U S A 104: 14442-14447.
  76. Aasheim HC, Patzke S, Hjorthaug HS, Finne EF (2005) Characterization of a novel Eph receptor tyrosine kinase, EphA10, expressed in testis. Biochim Biophys Acta 1723: 1-7.
  77. Fox BP, Kandpal RP (2011) A paradigm shift in EPH receptor interaction: biological relevance of EPHB6 interaction with EPHA2 and EPHB2 in breast carcinoma cell lines. Cancer Genomics Proteomics 8: 185-193.
  78. Matsuo K, Otaki N (2012) Bone cell interactions through Eph/ephrin: bone modeling, remodeling and associated diseases. Cell Adh Migr 6: 148-156.
  79. Andres AC, Reid HH, Zürcher G, Blaschke RJ, Albrecht D, et al. (1994) Expression of two novel eph-related receptor protein tyrosine kinases in mammary gland development and carcinogenesis. Oncogene 9: 1461-1467.
  80. Stephenson SA, Slomka S, Douglas EL, Hewett PJ, Hardingham JE (2001) Receptor protein tyrosine kinase EphB4 is up-regulated in colon cancer. BMC Mol Biol 2: 15.
  81. Steube KG, Meyer C, Habig S, Uphoff CC, Drexler HG (1999) Expression of receptor tyrosine kinase HTK (hepatoma transmembrane kinase) and HTK ligand by human leukemia-lymphoma cell lines. Leuk Lymphoma 33: 371-376.
  82. Shimoyama M, Matsuoka H, Tamekane A, Ito M, Iwata N, et al. (2000) T-cell-specific expression of kinase-defective Eph-family receptor protein, EphB6 in normal as well as transformed hematopoietic cells. Growth Factors 18: 63-78.
  83. Ivanova NB, Dimos JT, Schaniel C, Hackney JA, Moore KA, et al. (2002) A stem cell molecular signature. Science 298: 601-604.
  84. Steidl U, Bork S, Schaub S, Selbach O, Seres J, et al. (2004) Primary human CD34+ hematopoietic stem and progenitor cells express functionally active receptors of neuromediators. Blood 104: 81-88.
  85. Lazarova P, Wu Q, Kvalheim G, Suo Z, Haakenstad KW, et al. (2006) Growth factor receptors in hematopoietic stem cells: EPH family expression in CD34+ and CD133+ cell populations from mobilized peripheral blood. Int J Immunopathol Pharmacol 19: 49-56.
  86. Ting MJ, Day BW, Spanevello MD, Boyd AW (2010) Activation of ephrin A proteins influences hematopoietic stem cell adhesion and trafficking patterns. Exp Hematol 38: 1087-1098.
  87. Prevost N, Woulfe D, Tanaka T, Brass LF (2002) Interactions between Eph kinases and ephrins provide a mechanism to support platelet aggregation once cell-to-cell contact has occurred. Proc Natl Acad Sci U S A 99: 9219-9224.
  88. Wang Z, Cohen K, Shao Y, Mole P, Dombkowski D, et al. (2004) Ephrin receptor, EphB4, regulates ES cell differentiation of primitive mammalian hemangioblasts, blood, cardiomyocytes, and blood vessels. Blood 103: 100-109.
  89. Suenobu S, Takakura N, Inada T, Yamada Y, Yuasa H, et al. (2002) A role of EphB4 receptor and its ligand, ephrin-B2, in erythropoiesis. Biochem Biophys Res Commun 293: 1124-1131.
  90. Okubo T, Yanai N, Obinata M (2006) Stromal cells modulate ephrinB2 expression and transmigration of hematopoietic cells. Exp Hematol 34: 330-338.
  91. Muñoz JJ, Alonso-C LM, Sacedón R, Crompton T, Vicente A, et al. (2002) Expression and function of the Eph A receptors and their ligands ephrins A in the rat thymus. J Immunol 169: 177-184.
  92. Yu G, Mao J, Wu Y, Luo H, Wu J (2006) Ephrin-B1 is critical in T-cell development. J Biol Chem 281: 10222-10229.
  93. Shimoyama M, Matsuoka H, Nagata A, Iwata N, Tamekane A, et al. (2002) Developmental expression of EphB6 in the thymus: lessons from EphB6 knockout mice. Biochem Biophys Res Commun 298: 87-94.
  94. Trinidad EM, Zapata AG, Alonso-Colmenar LM (2010) Eph-ephrin bidirectional signaling comes into the context of lymphocyte transendothelial migration. Cell Adh Migr 4: 363-367.
  95. Aasheim HC, Terstappen LW, Logtenberg T (1997) Regulated expression of the Eph-related receptor tyrosine kinase Hek11 in early human B lymphopoiesis. Blood 90: 3613-3622.
  96. Brantley DM, Cheng N, Thompson EJ, Lin Q, Brekken RA, et al. (2002) Soluble Eph A receptors inhibit tumor angiogenesis and progression in vivo. Oncogene 21: 7011-7026.
  97. Adams RH, Wilkinson GA, Weiss C, Diella F, Gale NW, et al. (1999) Roles of ephrinB ligands and EphB receptors in cardiovascular development: demarcation of arterial/venous domains, vascular morphogenesis, and sprouting angiogenesis. Genes Dev 13: 295-306.
  98. Zhao C, Irie N, Takada Y, Shimoda K, Miyamoto T, et al. (2006) Bidirectional ephrinB2-EphB4 signaling controls bone homeostasis. Cell Metab 4: 111-121.
  99. Irie N, Takada Y, Watanabe Y, Matsuzaki Y, Naruse C, et al. (2009) Bidirectional signaling through ephrinA2-EphA2 enhances osteoclastogenesis and suppresses osteoblastogenesis. J Biol Chem 284: 14637-14644.
  100. Allan EH, Häusler KD, Wei T, Gooi JH, Quinn JM, et al. (2008) EphrinB2 regulation by PTH and PTHrP revealed by molecular profiling in differentiating osteoblasts. J Bone Miner Res 23: 1170-1181.
  101. Xing W, Kim J, Wergedal J, Chen ST, Mohan S (2010) Ephrin B1 regulates bone marrow stromal cell differentiation and bone formation by influencing TAZ transactivation via complex formation with NHERF1. Mol Cell Biol 30: 711-721.
  102. Cheng S, Zhao SL, Nelson B, Kesavan C, Qin X, et al. (2012) Targeted disruption of ephrin B1 in cells of myeloid lineage increases osteoclast differentiation and bone resorption in mice. PLoS One 7: e32887.
  103. Lathia JD, Rao MS, Mattson MP, Ffrench-Constant C (2007) The microenvironment of the embryonic neural stem cell: lessons from adult niches? Dev Dyn 236: 3267-3282.
  104. Arthur A, Zannettino A, Panagopoulos R, Koblar SA, Sims NA, et al. (2011) EphB/ephrin-B interactions mediate human MSC attachment, migration and osteochondral differentiation. Bone 48: 533-542.
  105. Kiyokawa E, Takai S, Tanaka M, Iwase T, Suzuki M, et al. (1994) Overexpression of ERK, an EPH family receptor protein tyrosine kinase, in various human tumors. Cancer Res 54: 3645-3650.
  106. Varelias A, Koblar SA, Cowled PA, Carter CD, Clayer M (2002) Human osteosarcoma expresses specific ephrin profiles: implications for tumorigenicity and prognosis. Cancer 95: 862-869.
  107. McKenzie (1996). Textbook of Hematology. (2ndedn), Williams & Wilkins Publishers, USA.
  108. Wicks IP, Wilkinson D, Salvaris E, Boyd AW (1992) Molecular cloning of HEK, the gene encoding a receptor tyrosine kinase expressed by human lymphoid tumor cell lines. Proc Natl Acad Sci U S A 89: 1611-1615.
  109. Lawrenson ID, Wimmer-Kleikamp SH, Lock P, Schoenwaelder SM, Down M, et al. (2002) Ephrin-A5 induces rounding, blebbing and de-adhesion of EphA3-expressing 293T and melanoma cells by CrkII and Rho-mediated signalling. J Cell Sci 115: 1059-1072.
  110. Vearing C, Lee FT, Wimmer-Kleikamp S, Spirkoska V, To C, et al. (2005) Concurrent binding of anti-EphA3 antibody and ephrin-A5 amplifies EphA3 signaling and downstream responses: potential as EphA3-specific tumor-targeting reagents. Cancer Res 65: 6745-6754.
  111. Walter MJ, Payton JE, Ries RE, Shannon WD, Deshmukh H, et al. (2009) Acquired copy number alterations in adult acute myeloid leukemia genomes. Proc Natl Acad Sci U S A 106: 12950-12955.
  112. Guan M, Liu L, Zhao X, Wu Q, Yu B, et al. (2011) Copy number variations of EphA3 are associated with multiple types of hematologic malignancies. Clin Lymphoma Myeloma Leuk 11: 50-53.
  113. Ashton JM, Balys M, Neering SJ, Hassane DC, Cowley G, et al. (2012) Gene sets identified with oncogene cooperativity analysis regulate in vivo growth and survival of leukemia stem cells. Cell Stem Cell 11: 359-372.
  114. Hagey A, et al. (2011) A recombinant antibody to EPHA3 for the treatment of hematologic malignancies: research update and interim Phase 1 study results. ASH Annual Meeting Abstracts.
  115. Krasnoperov V, Kumar SR, Ley E, Li X, Scehnet J, et al. (2010) Novel EphB4 monoclonal antibodies modulate angiogenesis and inhibit tumor growth. Am J Pathol 176: 2029-2038.
  116. López-Nieva P, Vaquero C, Fernández-Navarro P, González-Sánchez L, Villa-Morales M, et al. (2012) EPHA7, a new target gene for 6q deletion in T-cell lymphoblastic lymphomas. Carcinogenesis 33: 452-458.
  117. Müller-Tidow C, Schwäble J, Steffen B, Tidow N, Brandt B, et al. (2004) High-throughput analysis of genome-wide receptor tyrosine kinase expression in human cancers identifies potential novel drug targets. Clin Cancer Res 10: 1241-1249.
  118. Alonso-C LM, Trinidad EM, de Garcillan B, Ballesteros M, Castellanos M, et al. (2009) Expression profile of Eph receptors and ephrin ligands in healthy human B lymphocytes and chronic lymphocytic leukemia B-cells. Leuk Res 33: 395-406.
  119. Landen CN, Kinch MS, Sood AK (2005) EphA2 as a target for ovarian cancer therapy. Expert Opin Ther Targets 9: 1179-1187.
  120. Traylor RN, Fan Z, Hudson B, Rosenfeld JA, Shaffer LG, et al. (2009) Microdeletion of 6q16.1 encompassing EPHA7 in a child with mild neurological abnormalities and dysmorphic features: case report. Mol Cytogenet 2: 17.
Citation: Charmsaz S, Boyd AW (2013) Expression and Function of the Eph Receptor Family in Leukemia and Hematopoietic Malignancies: Prospects for Targeted Therapies. J Leuk (Los Angel) 1:107.

Copyright: © 2013 Charmsaz S, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top