1,204
Views
17
CrossRef citations to date
0
Altmetric
Review

Regulation and misregulation of Eph/ephrin expression

&
Pages 131-137 | Published online: 30 May 2012

Abstract

The erythropoietin-producing hepatocellular (Eph) receptors form the largest family of receptor tyrosine kinases. Upon interaction of the Eph receptors with their ligands the ephrins, signaling cascades are initiated downstream of both receptor and ligand, a feature known as bidirectional signaling. The Eph receptors and ephrin ligands mediate important roles in embryonic development, particularly in establishing tissue organization by mediating cell adhesion or cell repulsion. In several adult tissues, at least one Eph/ephrin pair is found to play critical roles in tissue physiology and homeostasis. In recent years numerous members of this family have gained considerable attention since changes in their expression levels are a typical feature in cancer cells. Despite the fact that Eph/ephrin developmental expression profiles are well documented, little is known on transcriptional and post-transcriptional mechanisms that permits their highly specific, graded, complementary or overlapping expression patterns. Therefore understanding the transcriptional and post-transcriptional mechanisms regulating Eph/ephrin expression has far-reaching significance in biology. This review provides an overview of the mechanisms regulating Eph/ephrin expression. We highlight important emerging mechanisms of Eph/ephrin regulation or misregulation such as epigenetics and miRNAs.

Introduction

The Eph receptors and their ligands, the ephrins, belong to the largest family of receptor tyrosine kinases.Citation1-Citation3 Ephrin ligands are divided into two subclasses, A and B, depending on their mode of attachment to the plasma membrane. The ephrinA subclass ligands (A1–A5) are tethered to the membrane via a glycosylphosphatidylinositol (GPI) anchor, whereas the ephrinB subclass ligands (B1–B3) are transmembrane proteins.Citation3 Correspondingly, there are two Eph receptor subclasses, A and B, that exhibit preferential affinities for the two ligand subclasses;Citation4 yet promiscuity between ligand and receptor subclasses has also been identified.Citation5 Ephrins and Eph receptors require direct cell-cell contact for their activation. A unique feature of this family is that interaction of Eph receptors with their corresponding ephrin ligands triggers signaling in both the receptor-hosting cell (termed forward signaling) and the ligand-hosting cell (termed reverse signaling).Citation6 Most studies to date have focused on the biological consequences of Eph/ephrin interactions, primarily in a developmental context, where they have been found indispensable for neural crest cell migration, axon guidance, the formation of tissue boundaries and vascular development.Citation1,Citation3 Over the last decade studies have highlighted the involvement of Eph receptors and ephrins in organ function and in disease in the adult.Citation7-Citation12 For instance, Eph/ephrins have been implicated in modulating cell migration, growth and invasiveness in cancer.Citation13

Substantial progress has been made in dissecting the molecular mechanisms downstream of Eph/ephrin bidirectional signaling, several of which have shown regulation of the actin cytoskeleton by small GTPase proteins of the Ras superfamily.Citation14 In addition to the considerable insight gained into the functioning of this family of proteins, these studies have correspondingly exposed the highly complex and diverse gene expression patterns of Eph (eph) receptors and ephrin (efn) ligands. In many tissues, specific Eph receptors and ephrin ligands have complementary domains of expression, whereas in other tissues family members may overlap in their expression.Citation7,Citation15,Citation16 In addition to their complementary or overlapping expression domains, the receptor/ligand pairs can also be expressed in gradientsCitation17 indicating sophisticated control mechanisms. One challenge therefore, has been to understand the transcriptional and post-transcriptional control of Eph receptors and ephrin ligands. Not surprisingly, a large extent of our knowledge on the transcriptional and post-transcriptional control of Eph receptors and ephrins has been gained form studying receptor/ligand expression in development or in tumor growth and metastasis. This review will highlight data that support clear associations between transcriptional and post-transcriptional effectors of Eph receptor and ephrin ligand regulation in developmental processes, adult tissues and in carcinogenesis.

Transcriptional Regulation

Historically, studies have focused on the transcriptional regulation of several of the Eph receptors, mainly in the context of developmental segmentation and patterning. Homeobox (HOX)-containing transcription factors emerged as one of the key regulators of Eph receptor expression.Citation18-Citation33 These transcription factors contain a well conserved DNA binding motif, the homeodomain, which binds to the promoter of target genes to regulate their expression. Their spatially restricted expression patternsCitation34 may have a central role in establishing the graded and segmental gene expression patterns of Eph receptors during embryogenesis. There exists evidence for direct regulation of Eph receptors by HOX proteins. For instance the homeobox proteins HOXA1 and HOXB1 have been shown to activate rhombomere-specific ephA2 expression in the developing mouse brain,Citation31 and HOXA2 was found to regulate ephA4 in rhombomeres 3 and 5.Citation32 While these studies have been performed essentially in the mouse hindbrain, recent studies on endothelial cells and angiogenesis have shown the necessity for HOXA9 in the regulation of ephB4.Citation27 In studying gene regulatory patterns in developing mouse limbs, it was shown that ephA7 is a direct downstream target of HOXD13 and HOXA13.Citation35,Citation36

The efnA2 promoter is targeted by otx2, a homeobox transcription factor.Citation37 In fact, this study demonstrated that efnA2 regulation by otx2 was important for the patterning and morphogenesis of the brain. EphA4 and ephrinA4 genes, ephA4 and efnA4, have been shown to be inversely regulated. Specifically, ephA4 is positively regulated by Twist1 while efnA4 is negatively regulated by Twist1 and the homeobox homolog, MSX2, in coronal suture development.Citation38,Citation39 Additional evidence for the inverse control of ephA4 and efnA expression has been shown in the establishment of topographic motor projections in the limb by Lim homeobox proteins.Citation21 While the exact genetic mechanisms underlying this regulation remain to be identified the authors show that Lim1 promotes ephA4 gene expression in motor neurons, while Lmx1b, the LIM homeobox transcription factor 1-β, inhibits ephrinA expression in the mesenchyme.

In addition to HOX-transcription factors, a handful of other transcription factors have been shown to regulate the expression of Eph/ephrins. The chick brain factor 1 (CBF1) is shown to directly regulate efnA5 expression during development of the retinotectal map in the chick.Citation40 The efnA4 promoter can be bound by the transcriptional activator nuclear factor-Y (NF-Y) in activated lymphocytes.Citation41 Likewise, NF-Y, in addition to Meis1 and MAZ have been shown to physically interact with the efnB2 promoter following extensive analysis of this promoter in prenatal angiogenesis.Citation42 Identified transcriptional regulators of ephB2 include TCF4 and β-catenin in the intestinal epithelium.Citation21,Citation43 The transcription factor valentino (val), the zebrafish ortholog of mouse mafB/Kreisler, a bzip transcription factor, was shown to establish the mutually exclusive expression domains of ephB4 and efnB2 in the zebrafish caudal hindbrain.Citation36 The zinc-finger transcription factor Zic2 was shown to regulate the expression of ephB1 in retinal ganglion cells; a process essential for the development of the ipsilateral projection at the mammalian optic chiasm midline.Citation44 Exciting new evidence shows that Eph/ephrin gene expression may be modulated by mechanical forces in different tissue types, including bone and dental pulp.Citation45,Citation46 The molecular mechanisms underlying mechanical force regulation of Eph/ephrin expression in these contexts, however, are largely unknown. Recent data obtained from endothelial progenitor cells exposed to shear stress showed inverse transcriptional regulation of efnB2 and ephB4.Citation47,Citation48 The increase in efnB2 expression was found to be due to Sp1 activation and binding specifically to the efnB2 promoter. These findings put forward the notion that shear stress induces ephrinB2 gene expression resulting in the differentiation of endothelial progenitor cells into arterial endothelial cells.

Our understanding of the transcriptional regulation of Eph receptors and ephrin ligands in developmental processes is in its early stages. Importantly, over the last decade, our knowledge surrounding transcriptional regulation of Eph/ephrin expression in pathological processes has expanded. These discoveries stem largely from studies on cancer, where misregulation of Eph receptors and ephrin ligands is a typical consequence.

Misregulation

Transcriptional regulation of ephB2 has been studied extensively since alterations in its expression levels occur in several cancers.Citation49-Citation52 Loss of ephB2 expression is observed in many tumors, and is particularly studied in colonic adenomas and carcinomas.Citation53,Citation54 The downregulation of ephB2 in early and advanced colorectal cancers was found to be due to c-Rel binding to a negative regulatory element in the ephB2 promoter.Citation52 On the other hand, upregulation of the EphB2 gene is associated with pancreatic ductal adenocarcinoma where it has been shown to be transcriptionally regulated by the Basic transcription factor 3 (BTF3).Citation55

Another Eph receptor commonly linked to cancer and believed important in tumor metastasis and angiogenesis is ephA2.Citation56,Citation57 The EphA2 promoter was reported to be a direct transcriptional target of the Ras-Raf-MAPK pathway in breast cancer cell lines.Citation58,Citation59 In addition these studies have shown a negative feedback loop between efnA1 expression and ephA2 levels, indicating that this regulation may contribute to receptor-ligand reciprocal expression patterns. Similarly, evidence exists for MAPK dependent regulation of ephA2 in UV radiated induced apoptosis.Citation60 Altogether these studies show several mechanisms underlying the misexpression of Eph receptors and ephrins. In addition to the transcriptional mechanisms controlling Eph/ephrin expression, epigenetic mechanisms have also been studied.

Epigenetic regulation

A major step in epigenetic regulation of gene expression is gene inactivation by hypermethylation of CpG islands located in the promoter regions (for extensive reviews see refs. Citation61 and Citation62). Specific enzymes and methylated DNA binding proteins play a major role in epigenetic regulation of gene expression. Alterations in CpG island methylation can affect gene expression in normal and cancer cells.Citation63-Citation66 The possible regulation of Eph receptors and ephrin ligands by epigenetics has been under considerable study. For example, the ephA5 receptor is a major player in regulating patterning of the topographic connections of retinal ganglion cells during visual system development.Citation67 New findings have shown that CpG islands in the ephA5 promoter display site-specific differences in methylation that could preferentially activate or repress promoter activity and may contribute to graded ephA5 gene expression in the tectum.Citation68 More studies, however, have focused on aberrant methylation patterns of Eph receptor and ephrin genes in cancer. Methylation of CpG promoter sequences of ephA1, ephA2, ephA7, ephB2, ephB4 and ephB6 has been reported in several human tumors, including colorectal, prostate and breast cancer.Citation69-Citation73 The EphA3 gene promoter region isolated from human embryonic kidney cells is also found to be rich in CpG islands that may regulate ephA3 transcription in hematopoietic tumors.Citation74 A recent study using acute lymphoblastic leukemia bone marrow samples and cell lines provided a thorough analysis of CpG island methylation of almost all of the Eph receptors and ephrin ligands.Citation75 Specifically the authors demonstrated hypermethylation of the promoter regions of the ephA2, ephA4, ephA5, ephA6, ephA7, ephA10, ephB1, ephB2, ephB3 and ephB4 receptors, in addition to the efnA1, efnA3, efnA5, efnB1 and efnB2 ligands. More detailed analyses were performed on the EphB4 promoter region showing that hypermethylation renders ephB4 a tumor suppressor gene in acute lymphoblastic leukemia.

Altogether, studies on aberrant promoter methylation of Eph receptors and ephrins are providing promising information on the epigenetic regulation of this family of genes. One example is that hypomethylation of the efnB1 promoter may be an early prognostic event for rheumatoid arthritis.Citation76

Post-Transcriptional Regulation

Over the last few years we and others have shown that ephrins and Eph receptors can be regulated post-transcriptionally. One interesting feature of this large family of proteins is that the 3′-untranslated regions (3′-UTRs) of several of the Eph receptors or the ephrin ligands contain regulatory binding sites that are highly conserved in vertebrates.Citation77,Citation78 An overview of these studies showing Eph receptor or ephrin ligand regulation at the level of mRNA stability or by microRNAs (miRNAs) in physiological and pathological processes is provided below.

mRNA stability

Putative binding sites for RNA-stabilizing and RNA-destabilizing factors have been identified in the 3′ untranslated regions (3′-UTR) sequences of several Eph/ephrin transcripts.Citation77,Citation78 Bioinformatic approaches were used to identify clusters of motifs consisting of cytoplasmic polyadenylation elements (CPEs), AU-rich elements (AREs) and HuR binding sites.Citation78 Despite the presence of these clusters in numerous members of the Eph/ephrin family, only some were validated. Using HeLa cervical cancer cells and U373MG glioma cells, this study showed that the HuR binding sites in the 3′-UTR region of ephA2, ephA4 and efnA2 served to destabilize the transcripts, despite the conventional role of HuR as an mRNA-stabilizing protein. Moreover, the authors report that knockdown of HuR greatly regulates Eph/ephrin expression at both the mRNA and protein levels. Altogether, this study suggests that overexpression of HuR, as found in many progressive tumors, could cause variation in Eph receptor and ephrin ligand expression and therefore result in increased tissue invasiveness.

Along the same lines, post-transcriptional regulation of ephB2 may also be important in cancer metastasis. The EphB2 gene has been implicated as a tumor suppressor gene altered in both prostate cancer and colorectal cancer.Citation79 Huusko and colleaguesCitation80 reported that the DU 145 prostate cancer cell line carries a truncating mutation of ephB2 and a deletion of the remaining allele. This truncated version was subject to Nonsense-Mediated Decay (NMD). Furthermore, the authors identified other missense and nonsense mutations from clinical prostate cancer samples. Overall, the authors proposed that EphB2 may have an essential role in cell migration and maintenance of normal tissue architecture, and that mutational inactivation of ephB2 may be important in the progression and metastasis of prostate cancer. Nonsense and frameshift mutations in the efnb1 gene were reported to undergo NMD.Citation81 In fact these mutations are considered to be contributors to the pathogenic mechanisms reported in the human X-linked malformation syndrome, craniofrontonasal syndrome (CFNS). Therefore, accumulating evidence implicates mRNA stability as a possible regulatory or deregulatory process in Eph/ephrin expression in cancer and other pathologies. In addition to these processes miRNAs have also emerged as post-transcriptional regulators of Eph/ephrin expression in both developmental and pathophysiological conditions.

miRNAs

Post-transcriptional regulation by miRNAs is important for many aspects of development, homeostasis and disease. MicroRNAs constitute a family of short noncoding RNA molecules of 20 to 25 nucleotides in length that regulate gene expression at the post-transcriptional level.Citation82,Citation83 In animals, miRNAs typically target sequences in the transcript 3′-UTRs that are only partially complementary to the miRNA, thereby causing a repression in translation of the mRNA.Citation84 Eph receptors and ephrin ligands have emerged as potential targets for miRNA regulation. A number of studies have correlated changes in the expression levels of Eph receptors or ephrin ligands to modulations in specific miRNA levels. For example, the upregulation of miR-223 following hepatic ischemia injury in mice resulted in a downregulation of the ephrin-A1 transcript, efnA1.Citation85 Similarly, miRNA expression profiling coupled with proteomic analysis following different dietetic regimens has inferred efnA1 and ephA2 as potential targets for miR-122, miR-451 and miR-27.Citation86

To date there is little evidence for the direct regulation of Eph receptors and ephrin ligands by miRNAs, yet, several members of the family have predicted miRNA binding sites (). This indeed may be a reflection of the complexity of the gradient, complementary and overlapping expression domains of Eph receptors and ephrin ligands. A pioneering study in 2008 showed that ephrin-A3 gene, efnA3, was a direct target of miR-210.Citation87 The authors concluded that efnA3 modulation by miR-210 had significant functional consequences for endothelial cell response to hypoxia, affecting cell survival, migration and differentiation. A recent report has further shown that miR-210 modulation of efnA3 underlies one of the molecular mechanisms in preeclampsia.Citation88 We have identified miR-124 as a post-transcriptional repressor of efnB1 expression in neural stem cells.Citation77 In addition we presented evidence for the regulation of miR-124 levels by ephrin-B1 reverse signaling, thus revealing the existence of a mutually repressive interaction between ephrin-B1 and miR-124. More recently it has been shown that ephA2 is a direct target of miR-26b.Citation89 EphA2 is expressed in a number of cancers and has potential roles in the regulation of cancer cell growth, survival, migration, invasion and angiogenesis.Citation90,Citation91 The authors demonstrated that miR-26b may act as a tumor suppressor in glioma by directly regulating ephA2 expression. Therefore, miRNAs, which are involved in the control of a wide range of biological functions and processes, have now been implicated in post-transcriptionally regulating members of the Eph/ephrin family.

Table 1. Representation of predicted and validated miRNAs targeting Eph receptors and ephrin ligands

The challenge to identify additional post-transcriptional regulating mechanisms for Eph/ephrins continues. Undoubtedly, this will be fraught by the complexity in Eph/ephrin expression patterns, and the means to identify cell-specific, context-dependent regulation. Therefore, future analyses will be valuable for understanding the changes in Eph/ephrin expression levels in normal and disease states.

Concluding Remarks

We are only beginning to understand the regulatory mechanisms governing the expression of Eph receptors and ephrins in development, in adult tissues, and their misregulation in disease. While HOX transcription factors have emerged as one of the key transcriptional regulators of Eph receptor genes in development, data shows that tissue-specific expression of each member of the Eph/ephrin family is also dependent on distinct molecular effectors. This indeed is surprising, given the high conservation between Eph/ephrin subclass members and the noted redundancy in their function. As we are only beginning to unravel the transcriptional control of Ephs and ephrins, it is possible that future studies may uncover common regulatory mechanisms.

There is an increasing body of evidence for inverse regulation between Eph receptors and ephrin ligands. Several of the examples demonstrating an inverse regulation between Eph receptors and ephrin ligands in developmental processes and in cancer are discussed in this review.Citation38,Citation39,Citation47,Citation48 Fewer findings exist for co-regulation of the receptor/ligand pair. One such example revealed that Eph/ephrins act synergistically in the mouse skin to induce angiogenesis in response to local hypoxia.Citation92 Curiously, one emerging notion is that activation of bidirectional signaling may be one of the cell-specific mechanisms employed to establish positive (or negative) feedback loops regulating Eph/ephrin expression. Our findings for the auto-regulation of efnB1 following ephrin-B1 activation in trans by EphB2 in neural progenitorsCitation77 strongly suggest that one of the several outcomes of the Eph receptor/ephrin bidirectional activation (or inhibition) may lead to establishing their own highly precise and combinatorial expression patterns. If indeed additional future studies corroborate that Eph/ephrin activity-dependent regulation is involved in establishing Eph/ephrin complementary expression domains, these discoveries will help identify the molecular basis of topographic positioning within the developing embryo and adult. Moreover, such findings may help explain the prominent misregulation of the receptors and ligands in cancer.

Further studies on Eph/ephrin RNA processing mechanisms, including the control of intracellular localization of Eph/ephrin mRNAs and association with translating ribosomes, and the post-transcriptional processing of the different members are necessary to fully understand the mechanisms controlling their expression. Indeed, it is likely an amalgamation of all these factors, genetic and epigenetic, that leads to the highly precise and combinatorial expression patterns of ephs and ephrins. Unraveling the mechanisms that control Eph receptor and ephrin ligand expression during development will undoubtedly yield important insight into tumorigenesis, as it is probable that tumors exploit similar regulatory mechanisms. It is important to take note that mechanisms beyond transcriptional and post-transcriptional regulatory processes have been presented in the literature. For instance, the attenuation of EphA3 function by ephrinA5 in cis can result in a loss of sensitivity of retinal axons to ephrinAs.Citation94 A similar cis-mediated attenuation of Eph receptors function has been reported during spinal motor neuron selection of a limb trajectory.Citation95 Interestingly, cis attenuation of Eph signaling by co-expressed ephrins has not been widely reported in cancer settings, perhaps owing to the fact that expression of receptors and ligands is usually inversely regulated. Data also exists for bidirectional activity dependent regulation in Eph/ephrin localization and expression. One example of this complexity is found in the work published by Bush and SorianoCitation93 showing that the mosaic expression of efnB1 in ephrin-B1 heterozygote animals leads to upregulation of the EphB3 receptor in adjacent, non-ephrin-B1 expressing cells through relief of EphB3 endocytosis and degradation. This finding supports the notion that the Eph/ephrin signaling cascade is involved in setting up complementary domains via post-transductional mechanisms. These data highlight how Eph/ephrin interactions can regulate their functional expression patterns and more importantly offer an additional level in the complexity of understanding the establishment and maintenance in Eph/ephrin expression domains.

The changes in expression profiles of Eph receptors and ephrin ligands in several cancers have made this family of proteins prime targets for cancer prognosis and therapies.Citation96-Citation99 However, what is important to note is that Eph/ephrins are expressed in numerous organs through adulthood; therefore, any therapy aimed at reducing their expression systemically to treat pathologies will need to account for their requirement in normal tissues. Therefore, details on tissue-specific molecular mechanisms of Eph/ephrin gene regulation will be important to develop more sophisticated and precise therapeutics. Future work should address these important questions to resolve how Eph/ephrin gene expression is controlled in highly diverse biological settings and in cancer.

Acknowledgments

We have chosen to restrict our review to studies that reported direct regulation of eph and/or efn gene expression and have therefore purposefully omitted global transcriptomic studies. We acknowledge all of our colleagues whose important work was not directly cited here because of space limitations. This work was supported by a CDA Award from Human Frontier Science Program Organization to A.D. and by a Fondation pour la Recherche Medicale fellowship to D.N.A.

References

  • Kullander K, Klein R. Mechanisms and functions of Eph and ephrin signalling. Nat Rev Mol Cell Biol 2002; 3:475 - 86; http://dx.doi.org/10.1038/nrm856; PMID: 12094214
  • Murai KK, Pasquale EB. ‘Eph’ective signaling: forward, reverse and crosstalk. J Cell Sci 2003; 116:2823 - 32; http://dx.doi.org/10.1242/jcs.00625; PMID: 12808016
  • Pasquale EB. Eph receptor signalling casts a wide net on cell behaviour. Nat Rev Mol Cell Biol 2005; 6:462 - 75; http://dx.doi.org/10.1038/nrm1662; PMID: 15928710
  • Pasquale EB. The Eph family of receptors. Curr Opin Cell Biol 1997; 9:608 - 15; http://dx.doi.org/10.1016/S0955-0674(97)80113-5; PMID: 9330863
  • Haramis AP, Perrakis A. Selectivity and promiscuity in Eph receptors. Structure 2006; 14:169 - 71; http://dx.doi.org/10.1016/j.str.2006.01.005; PMID: 16472735
  • Davy A, Soriano P. Ephrin signaling in vivo: look both ways. Dev Dyn 2005; 232:1 - 10; http://dx.doi.org/10.1002/dvdy.20200; PMID: 15580616
  • Augustin HG, Reiss Y. EphB receptors and ephrinB ligands: regulators of vascular assembly and homeostasis. Cell Tissue Res 2003; 314:25 - 31; http://dx.doi.org/10.1007/s00441-003-0770-9; PMID: 12905065
  • Klein R. Excitatory Eph receptors and adhesive ephrin ligands. Curr Opin Cell Biol 2001; 13:196 - 203; http://dx.doi.org/10.1016/S0955-0674(00)00197-6; PMID: 11248553
  • Pasquale EB. Eph-ephrin bidirectional signaling in physiology and disease. Cell 2008; 133:38 - 52; http://dx.doi.org/10.1016/j.cell.2008.03.011; PMID: 18394988
  • Goldshmit Y, McLenachan S, Turnley A. Roles of Eph receptors and ephrins in the normal and damaged adult CNS. Brain Res Rev 2006; 52:327 - 45; http://dx.doi.org/10.1016/j.brainresrev.2006.04.006; PMID: 16774788
  • Merlos-Súrez A, Batlle E. Eph-ephrin signalling in adult tissues and cancer. Curr Opin Cell Biol 2008; 20:194 - 200; http://dx.doi.org/10.1016/j.ceb.2008.01.011; PMID: 18353626
  • Kuijper S, Turner CJ, Adams RH. Regulation of angiogenesis by Eph-ephrin interactions. Trends Cardiovasc Med 2007; 17:145 - 51; http://dx.doi.org/10.1016/j.tcm.2007.03.003; PMID: 17574121
  • Pasquale EB. Eph receptors and ephrins in cancer: bidirectional signalling and beyond. Nat Rev Cancer 2010; 10:165 - 80; http://dx.doi.org/10.1038/nrc2806; PMID: 20179713
  • Noren NK, Pasquale EB. Eph receptor-ephrin bidirectional signals that target Ras and Rho proteins. Cell Signal 2004; 16:655 - 66; http://dx.doi.org/10.1016/j.cellsig.2003.10.006; PMID: 15093606
  • Xu Q, Mellitzer G, Wilkinson DG. Roles of Eph receptors and ephrins in segmental patterning. Philos Trans R Soc Lond B Biol Sci 2000; 355:993 - 1002; http://dx.doi.org/10.1098/rstb.2000.0635; PMID: 11128993
  • Wilkinson DG. Eph receptors and ephrins: regulators of guidance and assembly. Int Rev Cytol 2000; 196:177 - 244; http://dx.doi.org/10.1016/S0074-7696(00)96005-4; PMID: 10730216
  • Feldheim DA, O’Leary DD. Visual map development: bidirectional signaling, bifunctional guidance molecules, and competition. Cold Spring Harb Perspect Biol 2010; 2:a001768; http://dx.doi.org/10.1101/cshperspect.a001768; PMID: 20880989
  • Luria V, Krawchuk D, Jessell TM, Laufer E, Kania A. Specification of motor axon trajectory by ephrin-B:EphB signaling: symmetrical control of axonal patterning in the developing limb. Neuron 2008; 60:1039 - 53; http://dx.doi.org/10.1016/j.neuron.2008.11.011; PMID: 19109910
  • Studer M, Gavalas A, Marshall H, Ariza-McNaughton L, Rijli FM, Chambon P, et al. Genetic interactions between Hoxa1 and Hoxb1 reveal new roles in regulation of early hindbrain patterning. Development 1998; 125:1025 - 36; PMID: 9463349
  • Luria V, Laufer E. Lateral motor column axons execute a ternary trajectory choice between limb and body tissues. Neural Dev 2007; 2:13; http://dx.doi.org/10.1186/1749-8104-2-13; PMID: 17605791
  • Kania A, Jessell TM. Topographic motor projections in the limb imposed by LIM homeodomain protein regulation of ephrin-A:EphA interactions. Neuron 2003; 38:581 - 96; http://dx.doi.org/10.1016/S0896-6273(03)00292-7; PMID: 12765610
  • Schulte D, Furukawa T, Peters MA, Kozak CA, Cepko CL. Misexpression of the Emx-related homeobox genes cVax and mVax2 ventralizes the retina and perturbs the retinotectal map. Neuron 1999; 24:541 - 53; http://dx.doi.org/10.1016/S0896-6273(00)81111-3; PMID: 10595508
  • Schneider-Maunoury S, Seitanidou T, Topilko P, Vesque C, Frain M, Gilardi-Hebenstreit P, et al. [Role of the Krox-20 gene in the development of rhombencephalon]. C R Seances Soc Biol Fil 1997; 191:91 - 4; PMID: 9181130
  • Richmond CA, Breault DT. Regulation of gene expression in the intestinal epithelium. Prog Mol Biol Transl Sci 2010; 96:207 - 29; http://dx.doi.org/10.1016/B978-0-12-381280-3.00009-9; PMID: 21075346
  • Suzuki A, Urushitani H, Sato T, Kobayashi T, Watanabe H, Ohta Y, et al. Gene expression change in the Müllerian duct of the mouse fetus exposed to diethylstilbestrol in utero. Exp Biol Med (Maywood) 2007; 232:503 - 14; PMID: 17392486
  • Hernandez RE, Rikhof HA, Bachmann R, Moens CB. vhnf1 integrates global RA patterning and local FGF signals to direct posterior hindbrain development in zebrafish. Development 2004; 131:4511 - 20; http://dx.doi.org/10.1242/dev.01297; PMID: 15342476
  • Bruhl T, Urbich C, Aicher D, Acker-Palmer A, Zeiher AM, Dimmeler S. Homeobox A9 transcriptionally regulates the EphB4 receptor to modulate endothelial cell migration and tube formation. Circ Res 2004; 94:743 - 51; http://dx.doi.org/10.1161/01.RES.0000120861.27064.09; PMID: 14764452
  • Durbin L, Sordino P, Barrios A, Gering M, Thisse C, Thisse B, et al. Anteroposterior patterning is required within segments for somite boundary formation in developing zebrafish. Development 2000; 127:1703 - 13; PMID: 10725246
  • Niederreither K, Vermot J, Schuhbaur B, Chambon P, Dollé P. Retinoic acid synthesis and hindbrain patterning in the mouse embryo. Development 2000; 127:75 - 85; PMID: 10654602
  • Manzanares M, Trainor PA, Nonchev S, Ariza-McNaughton L, Brodie J, Gould A, et al. The role of kreisler in segmentation during hindbrain development. Dev Biol 1999; 211:220 - 37; http://dx.doi.org/10.1006/dbio.1999.9318; PMID: 10395784
  • Chen J, Ruley HE. An enhancer element in the EphA2 (Eck) gene sufficient for rhombomere-specific expression is activated by HOXA1 and HOXB1 homeobox proteins. J Biol Chem 1998; 273:24670 - 5; http://dx.doi.org/10.1074/jbc.273.38.24670; PMID: 9733765
  • Theil T, Frain M, Gilardi-Hebenstreit P, Flenniken A, Charnay P, Wilkinson DG. Segmental expression of the EphA4 (Sek-1) receptor tyrosine kinase in the hindbrain is under direct transcriptional control of Krox-20. Development 1998; 125:443 - 52; PMID: 9425139
  • Taneja R, Thisse B, Rijli FM, Thisse C, Bouillet P, Dollé P, et al. The expression pattern of the mouse receptor tyrosine kinase gene MDK1 is conserved through evolution and requires Hoxa-2 for rhombomere-specific expression in mouse embryos. Dev Biol 1996; 177:397 - 412; http://dx.doi.org/10.1006/dbio.1996.0173; PMID: 8806819
  • Shashikant CS, Utset MF, Violette SM, Wise TL, Einat P, Einat M, et al. Homeobox genes in mouse development. Crit Rev Eukaryot Gene Expr 1991; 1:207 - 45; PMID: 1686978
  • Salsi V, Zappavigna V. Hoxd13 and Hoxa13 directly control the expression of the EphA7 Ephrin tyrosine kinase receptor in developing limbs. J Biol Chem 2006; 281:1992 - 9; http://dx.doi.org/10.1074/jbc.M510900200; PMID: 16314414
  • Cooke J, Moens C, Roth L, Durbin L, Shiomi K, Brennan C, et al. Eph signalling functions downstream of Val to regulate cell sorting and boundary formation in the caudal hindbrain. Development 2001; 128:571 - 80; PMID: 11171340
  • Rhinn M, Dierich A, Le Meur M, Ang S. Cell autonomous and non-cell autonomous functions of Otx2 in patterning the rostral brain. Development 1999; 126:4295 - 304; PMID: 10477297
  • Ting MC, Wu NL, Roybal PG, Sun J, Liu L, Yen Y, et al. EphA4 as an effector of Twist1 in the guidance of osteogenic precursor cells during calvarial bone growth and in craniosynostosis. Development 2009; 136:855 - 64; http://dx.doi.org/10.1242/dev.028605; PMID: 19201948
  • Merrill AE, Bochukova EG, Brugger SM, Ishii M, Pilz DT, Wall SA, et al. Cell mixing at a neural crest-mesoderm boundary and deficient ephrin-Eph signaling in the pathogenesis of craniosynostosis. Hum Mol Genet 2006; 15:1319 - 28; http://dx.doi.org/10.1093/hmg/ddl052; PMID: 16540516
  • Takahashi H, Shintani T, Sakuta H, Noda M. CBF1 controls the retinotectal topographical map along the anteroposterior axis through multiple mechanisms. Development 2003; 130:5203 - 15; http://dx.doi.org/10.1242/dev.00724; PMID: 12954716
  • Munthe E, Aasheim HC. Characterization of the human ephrin-A4 promoter. Biochem J 2002; 366:447 - 58; http://dx.doi.org/10.1042/BJ20011693; PMID: 12030849
  • Sohl M, Lanner F, Farnebo F. Characterization of the murine Ephrin-B2 promoter. Gene 2009; 437:54 - 9; http://dx.doi.org/10.1016/j.gene.2009.02.017; PMID: 19268698
  • Batlle E, Henderson JT, Beghtel H, van den Born MM, Sancho E, Huls G, et al. Beta-catenin and TCF mediate cell positioning in the intestinal epithelium by controlling the expression of EphB/ephrinB. Cell 2002; 111:251 - 63; http://dx.doi.org/10.1016/S0092-8674(02)01015-2; PMID: 12408869
  • García-Frigola C, Carreres MI, Vegar C, Mason C, Herrera E. Zic2 promotes axonal divergence at the optic chiasm midline by EphB1-dependent and -independent mechanisms. Development 2008; 135:1833 - 41; http://dx.doi.org/10.1242/dev.020693; PMID: 18417618
  • Xing W, Baylink D, Kesavan C, Hu Y, Kapoor S, Chadwick RB, et al. Global gene expression analysis in the bones reveals involvement of several novel genes and pathways in mediating an anabolic response of mechanical loading in mice. J Cell Biochem 2005; 96:1049 - 60; http://dx.doi.org/10.1002/jcb.20606; PMID: 16149068
  • Diercke K, Sen S, Kohl A, Lux CJ, Erber R. Compression-dependent up-regulation of ephrin-A2 in PDL fibroblasts attenuates osteogenesis. J Dent Res 2011; 90:1108 - 15; http://dx.doi.org/10.1177/0022034511413926; PMID: 21724962
  • Obi S, Yamamoto K, Shimizu N, Kumagaya S, Masumura T, Sokabe T, et al. Fluid shear stress induces arterial differentiation of endothelial progenitor cells. J Appl Physiol 2009; 106:203 - 11; http://dx.doi.org/10.1152/japplphysiol.00197.2008; PMID: 18988767
  • Sohl M, Lanner F, Farnebo F. Sp1 mediate hypoxia induced ephrinB2 expression via a hypoxia-inducible factor independent mechanism. Biochem Biophys Res Commun 2010; 391:24 - 7; http://dx.doi.org/10.1016/j.bbrc.2009.10.146; PMID: 19883626
  • 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; http://dx.doi.org/10.1158/1078-0432.CCR-04-2458; PMID: 16166419
  • Wu Q, Suo Z, Risberg B, Karlsson MG, Villman K, Nesland JM. Expression of Ephb2 and Ephb4 in breast carcinoma. Pathol Oncol Res 2004; 10:26 - 33; http://dx.doi.org/10.1007/BF02893405; PMID: 15029258
  • Jubb AM, Zhong F, Bheddah S, Grabsch HI, Frantz GD, Mueller W, et al. EphB2 is a prognostic factor in colorectal cancer. Clin Cancer Res 2005; 11:5181 - 7; http://dx.doi.org/10.1158/1078-0432.CCR-05-0143; PMID: 16033834
  • Fu T, Li P, Wang H, He Y, Luo D, Zhang A, et al. c-Rel is a transcriptional repressor of EPHB2 in colorectal cancer. J Pathol 2009; 219:103 - 13; http://dx.doi.org/10.1002/path.2590; PMID: 19621336
  • Guo DL, Zhang J, Yuen ST, Tsui WY, Chan AS, Ho C, et al. Reduced expression of EphB2 that parallels invasion and metastasis in colorectal tumours. Carcinogenesis 2006; 27:454 - 64; http://dx.doi.org/10.1093/carcin/bgi259; PMID: 16272170
  • Senior PV, Zhang BX, Chan ST. Loss of cell-surface receptor EphB2 is important for the growth, migration, and invasiveness of a colon cancer cell line. Int J Colorectal Dis 2010; 25:687 - 94; http://dx.doi.org/10.1007/s00384-010-0916-7; PMID: 20339854
  • Kusumawidjaja G, Kayed H, Giese N, Bauer A, Erkan M, Giese T, et al. Basic transcription factor 3 (BTF3) regulates transcription of tumor-associated genes in pancreatic cancer cells. Cancer Biol Ther 2007; 6:367 - 76; http://dx.doi.org/10.4161/cbt.6.3.3704; PMID: 17312387
  • Kataoka H, Igarashi H, Kanamori M, Ihara M, Wang JD, Wang YJ, et al. Correlation of EPHA2 overexpression with high microvessel count in human primary colorectal cancer. Cancer Sci 2004; 95:136 - 41; http://dx.doi.org/10.1111/j.1349-7006.2004.tb03194.x; PMID: 14965363
  • Brannan JM, Sen B, Saigal B, Prudkin L, Behrens C, Solis L, et al. EphA2 in the early pathogenesis and progression of non-small cell lung cancer. Cancer Prev Res (Phila) 2009; 2:1039 - 49; http://dx.doi.org/10.1158/1940-6207.CAPR-09-0212; PMID: 19934338
  • Menges CW, McCance DJ. Constitutive activation of the Raf-MAPK pathway causes negative feedback inhibition of Ras-PI3K-AKT and cellular arrest through the EphA2 receptor. Oncogene 2008; 27:2934 - 40; http://dx.doi.org/10.1038/sj.onc.1210957; PMID: 18059341
  • Macrae M, Neve RM, Rodriguez-Viciana P, Haqq C, Yeh J, Chen C, et al. A conditional feedback loop regulates Ras activity through EphA2. Cancer Cell 2005; 8:111 - 8; http://dx.doi.org/10.1016/j.ccr.2005.07.005; PMID: 16098464
  • Zhang G, Njauw CN, Park JM, Naruse C, Asano M, Tsao H. EphA2 is an essential mediator of UV radiation-induced apoptosis. Cancer Res 2008; 68:1691 - 6; http://dx.doi.org/10.1158/0008-5472.CAN-07-2372; PMID: 18339848
  • Nakao M. Epigenetics: interaction of DNA methylation and chromatin. Gene 2001; 278:25 - 31; http://dx.doi.org/10.1016/S0378-1119(01)00721-1; PMID: 11707319
  • Novik KL, Nimmrich I, Genc B, Maier S, Piepenbrock C, Olek A, et al. Epigenomics: genome-wide study of methylation phenomena. Curr Issues Mol Biol 2002; 4:111 - 28; PMID: 12432963
  • Duthie SJ. Epigenetic modifications and human pathologies: cancer and CVD. Proc Nutr Soc 2011; 70:47 - 56; http://dx.doi.org/10.1017/S0029665110003952; PMID: 21067630
  • Delgado MD, León J. Gene expression regulation and cancer. Clin Transl Oncol 2006; 8:780 - 7; http://dx.doi.org/10.1007/s12094-006-0132-7; PMID: 17134965
  • Grønbaek K, Hother C, Jones PA. Epigenetic changes in cancer. APMIS 2007; 115:1039 - 59; http://dx.doi.org/10.1111/j.1600-0463.2007.apm_636.xml.x; PMID: 18042143
  • Strathdee G, Sim A, Brown R. Control of gene expression by CpG island methylation in normal cells. Biochem Soc Trans 2004; 32:913 - 5; http://dx.doi.org/10.1042/BST0320913; PMID: 15506922
  • Zhou R. Regulation of topographic projection by the Eph family receptor Bsk (EphA5) and its ligands. Cell Tissue Res 1997; 290:251 - 9; http://dx.doi.org/10.1007/s004410050929; PMID: 9321686
  • Petkova TD, Seigel GM, Otteson DC. A role for DNA methylation in regulation of EphA5 receptor expression in the mouse retina. Vision Res 2011; 51:260 - 8; http://dx.doi.org/10.1016/j.visres.2010.09.022; PMID: 20875442
  • Davalos V, Dopeso H, Castaño J, Wilson AJ, Vilardell F, Romero-Gimenez J, et al. EPHB4 and survival of colorectal cancer patients. Cancer Res 2006; 66:8943 - 8; http://dx.doi.org/10.1158/0008-5472.CAN-05-4640; PMID: 16982731
  • Ogino S, Hazra A, Tranah GJ, Kirkner GJ, Kawasaki T, Nosho K, et al. MGMT germline polymorphism is associated with somatic MGMT promoter methylation and gene silencing in colorectal cancer. Carcinogenesis 2007; 28:1985 - 90; http://dx.doi.org/10.1093/carcin/bgm160; PMID: 17621591
  • Wang J, Kataoka H, Suzuki M, Sato N, Nakamura R, Tao H, et al. Downregulation of EphA7 by hypermethylation in colorectal cancer. Oncogene 2005; 24:5637 - 47; http://dx.doi.org/10.1038/sj.onc.1208720; PMID: 16007213
  • Alazzouzi H, Davalos V, Kokko A, Domingo E, Woerner SM, Wilson AJ, et al. Mechanisms of inactivation of the receptor tyrosine kinase EPHB2 in colorectal tumors. Cancer Res 2005; 65:10170 - 3; http://dx.doi.org/10.1158/0008-5472.CAN-05-2580; PMID: 16288001
  • Herath NI, Doecke J, Spanevello MD, Leggett BA, Boyd AW. Epigenetic silencing of EphA1 expression in colorectal cancer is correlated with poor survival. Br J Cancer 2009; 100:1095 - 102; http://dx.doi.org/10.1038/sj.bjc.6604970; PMID: 19277044
  • Dottori M, Down M, Hüttmann A, Fitzpatrick DR, Boyd AW. Cloning and characterization of EphA3 (Hek) gene promoter: DNA methylation regulates expression in hematopoietic tumor cells. Blood 1999; 94:2477 - 86; PMID: 10498621
  • Kuang SQ, Bai H, Fang ZH, Lopez G, Yang H, Tong W, et al. Aberrant DNA methylation and epigenetic inactivation of Eph receptor tyrosine kinases and ephrin ligands in acute lymphoblastic leukemia. Blood 2010; 115:2412 - 9; http://dx.doi.org/10.1182/blood-2009-05-222208; PMID: 20061560
  • Kitamura T, Kabuyama Y, Kamataki A, Homma MK, Kobayashi H, Aota S, et al. Enhancement of lymphocyte migration and cytokine production by ephrinB1 system in rheumatoid arthritis. Am J Physiol Cell Physiol 2008; 294:C189 - 96; http://dx.doi.org/10.1152/ajpcell.00314.2007; PMID: 17942634
  • Arvanitis DN, Jungas T, Behar A, Davy A. Ephrin-B1 reverse signaling controls a posttranscriptional feedback mechanism via miR-124. Mol Cell Biol 2010; 30:2508 - 17; http://dx.doi.org/10.1128/MCB.01620-09; PMID: 20308325
  • Winter J, Roepcke S, Krause S, Müller EC, Otto A, Vingron M, et al. Comparative 3’UTR analysis allows identification of regulatory clusters that drive Eph/ephrin expression in cancer cell lines. PLoS One 2008; 3:e2780; http://dx.doi.org/10.1371/journal.pone.0002780; PMID: 18648668
  • Kokko A, Laiho P, Lehtonen R, Korja S, Carvajal-Carmona LG, Järvinen H, et al. EPHB2 germline variants in patients with colorectal cancer or hyperplastic polyposis. BMC Cancer 2006; 6:145; http://dx.doi.org/10.1186/1471-2407-6-145; PMID: 16740153
  • Huusko P, Ponciano-Jackson D, Wolf M, Kiefer JA, Azorsa DO, Tuzmen S, et al. Nonsense-mediated decay microarray analysis identifies mutations of EPHB2 in human prostate cancer. Nat Genet 2004; 36:979 - 83; http://dx.doi.org/10.1038/ng1408; PMID: 15300251
  • Wieland I, Makarov R, Reardon W, Tinschert S, Goldenberg A, Thierry P, et al. Dissecting the molecular mechanisms in craniofrontonasal syndrome: differential mRNA expression of mutant EFNB1 and the cellular mosaic. Eur J Hum Genet 2008; 16:184 - 91; http://dx.doi.org/10.1038/sj.ejhg.5201968; PMID: 18043713
  • Ambros V. The functions of animal microRNAs. Nature 2004; 431:350 - 5; http://dx.doi.org/10.1038/nature02871; PMID: 15372042
  • Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004; 116:281 - 97; http://dx.doi.org/10.1016/S0092-8674(04)00045-5; PMID: 14744438
  • Filipowicz W, Bhattacharyya SN, Sonenberg N. Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight?. Nat Rev Genet 2008; 9:102 - 14; http://dx.doi.org/10.1038/nrg2290; PMID: 18197166
  • Yu CH, Xu CF, Li YM. Association of MicroRNA-223 expression with hepatic ischemia/reperfusion injury in mice. Dig Dis Sci 2009; 54:2362 - 6; http://dx.doi.org/10.1007/s10620-008-0629-8; PMID: 19104939
  • Alisi A, Da Sacco L, Bruscalupi G, Piemonte F, Panera N, De Vito R, et al. Mirnome analysis reveals novel molecular determinants in the pathogenesis of diet-induced nonalcoholic fatty liver disease. Lab Invest 2011; 91:283 - 93; http://dx.doi.org/10.1038/labinvest.2010.166; PMID: 20956972
  • Fasanaro P, D’Alessandra Y, Di Stefano V, Melchionna R, Romani S, Pompilio G, et al. MicroRNA-210 modulates endothelial cell response to hypoxia and inhibits the receptor tyrosine kinase ligand Ephrin-A3. J Biol Chem 2008; 283:15878 - 83; http://dx.doi.org/10.1074/jbc.M800731200; PMID: 18417479
  • Zhang Y, Fei M, Xue G, Zhou Q, Jia Y, Li L, et al. Elevated levels of hypoxia-inducible microRNA-210 in pre-eclampsia: new insights into molecular mechanisms for the disease. J Cell Mol Med 2012; 16:249 - 59; http://dx.doi.org/10.1111/j.1582-4934.2011.01291.x; PMID: 21388517
  • Wu N, Zhao X, Liu M, Liu H, Yao W, Zhang Y, et al. Role of microRNA-26b in glioma development and its mediated regulation on EphA2. PLoS One 2011; 6:e16264; http://dx.doi.org/10.1371/journal.pone.0016264; PMID: 21264258
  • Walker-Daniels J, Riese DJ 2nd, Kinch MS. c-Cbl-dependent EphA2 protein degradation is induced by ligand binding. Mol Cancer Res 2002; 1:79 - 87; PMID: 12496371
  • Walker-Daniels J, Hess AR, Hendrix MJ, Kinch MS. Differential regulation of EphA2 in normal and malignant cells. Am J Pathol 2003; 162:1037 - 42; http://dx.doi.org/10.1016/S0002-9440(10)63899-0; PMID: 12651595
  • Vihanto MM, Plock J, Erni D, Frey BM, Frey FJ, Huynh-Do U. Hypoxia up-regulates expression of Eph receptors and ephrins in mouse skin. FASEB J 2005; 19:1689 - 91; PMID: 16081502
  • Bush JO, Soriano P. Ephrin-B1 forward signaling regulates craniofacial morphogenesis by controlling cell proliferation across Eph-ephrin boundaries. Genes Dev 2010; 24:2068 - 80; http://dx.doi.org/10.1101/gad.1963210; PMID: 20844017
  • Carvalho RF, Beutler M, Marler KJ, Knöll B, Becker-Barroso E, Heintzmann R, et al. Silencing of EphA3 through a cis interaction with ephrinA5. Nat Neurosci 2006; 9:322 - 30; http://dx.doi.org/10.1038/nn1655; PMID: 16491080
  • Kao TJ, Kania A. Ephrin-mediated cis-attenuation of Eph receptor signaling is essential for spinal motor axon guidance. Neuron 2011; 71:76 - 91; http://dx.doi.org/10.1016/j.neuron.2011.05.031; PMID: 21745639
  • Hatano M, Eguchi J, Tatsumi T, Kuwashima N, Dusak JE, Kinch MS, et al. EphA2 as a glioma-associated antigen: a novel target for glioma vaccines. Neoplasia 2005; 7:717 - 22; http://dx.doi.org/10.1593/neo.05277; PMID: 16207473
  • Oricchio E, Nanjangud G, Wolfe AL, Schatz JH, Mavrakis KJ, Jiang M, et al. The Eph-receptor A7 is a soluble tumor suppressor for follicular lymphoma. Cell 2011; 147:554 - 64; http://dx.doi.org/10.1016/j.cell.2011.09.035; PMID: 22036564
  • Lee JW, Han HD, Shahzad MM, Kim SW, Mangala LS, Nick AM, et al. EphA2 immunoconjugate as molecularly targeted chemotherapy for ovarian carcinoma. J Natl Cancer Inst 2009; 101:1193 - 205; http://dx.doi.org/10.1093/jnci/djp231; PMID: 19641174
  • Kiewlich D, Zhang J, Gross C, Xia W, Larsen B, Cobb RR, et al. Anti-EphA2 antibodies decrease EphA2 protein levels in murine CT26 colorectal and human MDA-231 breast tumors but do not inhibit tumor growth. Neoplasia 2006; 8:18 - 30; http://dx.doi.org/10.1593/neo.05544; PMID: 16533422

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.