Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Elena B. Pasquale is active.

Publication


Featured researches published by Elena B. Pasquale.


Oncogene | 2000

The ephrin-A1 ligand and its receptor, EphA2, are expressed during tumor neovascularization

Kazushige Ogawa; Renata Pasqualini; Richard Lindberg; Renate Kain; Andrew L Freeman; Elena B. Pasquale

Eph receptor tyrosine kinases and their ephrin ligands have been implicated in embryonic vascular development and in in vivo models of angiogenesis. Eph proteins may also regulate tumor neovascularization, but this role has not been previously investigated. To screen for Eph proteins expressed in tumor blood vessels, we used tumor xenografts grown in nude mice from MDA-MB-435 human breast cancer cells or KS1767 human Kaposis sarcoma cells. By immunohistochemistry, the ephrin-A1 ligand and one of its receptors, EphA2, were detected throughout tumor vasculature. Double-labeling with anti-CD34 antibodies demonstrated that both ephrin-A1 and EphA2 were expressed in xenograft endothelial cells and also tumor cells. Furthermore, EphA2 was tyrosine-phosphorylated in the xenograft tumors, indicating that it was activated, presumably by interacting with ephrin-A1. Ephrin-A1 and EphA2 were also detected in both the vasculature and tumor cells of surgically removed human cancers. In an in vitro angiogenesis model, a dominant negative form of EphA2 inhibited capillary tube-like formation by human umbilical vein endothelial cells (HUVECs), demonstrating a requirement for EphA receptor signaling. These data suggest that ephrin-A1 and EphA2 play a role in human cancers, at least in part by influencing tumor neovascularization. Eph proteins may represent promising new targets for antiangiogenic cancer treatments.


The Journal of Neuroscience | 2004

Ephrin-A5 Exerts Positive or Inhibitory Effects on Distinct Subsets of EphA4-Positive Motor Neurons

Johann K. Eberhart; Jason Barr; Sinead O'Connell; Alleda E. Flagg; Mary E. Swartz; Karina S. Cramer; Kathryn W. Tosney; Elena B. Pasquale; Catherine E. Krull

Eph receptor tyrosine kinases and ephrins are required for axon patterning and plasticity in the developing nervous system. Typically, Eph–ephrin interactions promote inhibitory events; for example, prohibiting the entry of neural cells into certain embryonic territories. Here, we show that distinct subsets of motor neurons that express EphA4 respond differently to ephrin-A5. EphA4-positive LMC(l) axons avoid entering ephrin-A5-positive hindlimb mesoderm. In contrast, EphA4-positive MMC(m) axons extend through ephrin-A5-positive rostral half-sclerotome. Blocking EphA4 activation in MMC(m) neurons or expanding the domain of ephrin-A5 expression in the somite results in the aberrant growth of MMC(m) axons into the caudal half-sclerotome. Moreover, premature expression of EphA4 in MMC(m) neurons leads to a portion of their axons growing into novel ephrin-A5-positive territories. Together, these results indicate that EphA4-ephrin-A5 signaling acts in a positive manner to constrain MMC(m) axons to the rostral half-sclerotome. Furthermore, we show that Eph activation localizes to distinct subcellular compartments of LMC(l) and MMC(m) neurons, consistent with distinct EphA4 signaling cascades in these neuronal subpopulations.


Cancer Cell | 2012

The EphA2 Receptor Drives Self-Renewal and Tumorigenicity in Stem-Like Tumor-Propagating Cells from Human Glioblastomas

Elena Binda; Alberto Visioli; Fabrizio Giani; Giuseppe Lamorte; Massimiliano Copetti; Ken Pitter; Jason T. Huse; Laura Cajola; Nadia Zanetti; Francesco DiMeco; Lidia De Filippis; Annunziato Mangiola; Giulio Maira; Carmelo Anile; Pasquale De Bonis; Brent A. Reynolds; Elena B. Pasquale; Angelo L. Vescovi

In human glioblastomas (hGBMs), tumor-propagating cells with stem-like characteristics (TPCs) represent a key therapeutic target. We found that the EphA2 receptor tyrosine kinase is overexpressed in hGBM TPCs. Cytofluorimetric sorting into EphA2(High) and EphA2(Low) populations demonstrated that EphA2 expression correlates with the size and tumor-propagating ability of the TPC pool in hGBMs. Both ephrinA1-Fc, which caused EphA2 downregulation in TPCs, and siRNA-mediated knockdown of EPHA2 expression suppressed TPCs self-renewal ex vivo and intracranial tumorigenicity, pointing to EphA2 downregulation as a causal event in the loss of TPCs tumorigenicity. Infusion of ephrinA1-Fc into intracranial xenografts elicited strong tumor-suppressing effects, suggestive of therapeutic applications.


Cell and Tissue Research | 1999

Signal transfer by Eph receptors

Matthew S. Kalo; Elena B. Pasquale

Abstract The Eph receptors are a unique family of receptor tyrosine kinases that enforce cellular position in tissues through mainly repulsive signals generated upon cell-cell contact. Together, Eph receptors and their membrane-anchored ligands, the ephrins, are key molecules for establishing tissue organization through signaling pathways that control axonal projection, cell migration, and the maintenance of cellular boundaries. Through their SH2 (Src Homology 2) and PDZ (postsynaptic density protein, disks large, zona occludens) domains, several signaling molecules have been demonstrated to interact with the activated cytoplasmic domain of Eph receptors by using the yeast two-hybrid system and in vitro biochemical assays. Most proteins found to interact with Eph receptors are well-known regulators of cytoskeletal organization and cell adhesion, and also cell proliferation. Promoting growth, however, does not appear to be a primary role of Eph receptors. Explaining which signaling interactions identified for the Eph receptors have physiological significance, how Eph receptor signaling cascades are propagated, and characterizing the intrinsic signaling properties of the ephrins are all exciting questions currently being investigated.


Cancer Discovery | 2015

Ligand independent EphA2 signaling drives the adoption of a targeted therapy-mediated metastatic melanoma phenotype

Kim H. T. Paraiso; Meghna Das Thakur; Bin Fang; John M. Koomen; Inna V. Fedorenko; Jobin K. John; Hensin Tsao; Keith T. Flaherty; Vernon K. Sondak; Jane L. Messina; Elena B. Pasquale; Alejandro Villagra; Uma N. M. Rao; John M. Kirkwood; Friedegund Meier; Sarah Sloot; Geoffrey T. Gibney; Darrin Stuart; Hussein Tawbi; Keiran S.M. Smalley

UNLABELLED Many patients with BRAF inhibitor resistance can develop disease at new sites, suggesting that drug-induced selection pressure drives metastasis. Here, we used mass spectrometry-based phosphoproteomic screening to uncover ligand-independent EPHA2 signaling as an adaptation to BRAF inhibitor therapy that led to the adoption of a metastatic phenotype. The EPHA2-mediated invasion was AKT-dependent and readily reversible upon removal of the drug as well as through PI3K and AKT inhibition. In xenograft models, BRAF inhibition led to the development of EPHA2-positive metastases. A retrospective analysis of patients with melanoma on BRAF inhibitor therapy showed that 68% of those failing therapy develop metastases at new disease sites, compared with 35% of patients on dacarbazine. Further IHC staining of melanoma specimens taken from patients on BRAF inhibitor therapy as well as metastatic samples taken from patients failing therapy showed increased EPHA2 staining. We suggest that inhibition of ligand-independent EPHA2 signaling may limit metastases associated with BRAF inhibitor therapy. SIGNIFICANCE This study provides evidence that BRAF inhibition promotes the adoption of a reversible, therapy-driven metastatic phenotype in melanoma. The cotargeting of ligand-independent EPHA2 signaling and BRAF may be one strategy to prevent the development of therapy-mediated disease at new sites.


Journal of Biological Chemistry | 2015

EphA2 Receptor Unliganded Dimers Suppress EphA2 Pro-tumorigenic Signaling

Deo R. Singh; Fozia Ahmed; Christopher King; Nisha Gupta; Matt Salotto; Elena B. Pasquale; Kalina Hristova

Background: The EphA2 receptor tyrosine kinase can promote cell migration and cancer malignancy in the absence of ligand binding. Results: We uncover a correlation between unliganded dimerization and tumorigenic signaling. Conclusion: EphA2 pro-tumorigenic signaling is likely mediated by the EphA2 monomer. Significance: A therapeutic strategy that aims at the stabilization of EphA2 dimers may be beneficial for the treatment of cancers linked to EphA2 overexpression. The EphA2 receptor tyrosine kinase promotes cell migration and cancer malignancy through a ligand- and kinase-independent distinctive mechanism that has been linked to high Ser-897 phosphorylation and low tyrosine phosphorylation. Here, we demonstrate that EphA2 forms dimers in the plasma membrane of HEK293T cells in the absence of ephrin ligand binding, suggesting that the current seeding mechanism model of EphA2 activation is incomplete. We also characterize a dimerization-deficient EphA2 mutant that shows enhanced ability to promote cell migration, concomitant with increased Ser-897 phosphorylation and decreased tyrosine phosphorylation compared with EphA2 wild type. Our data reveal a correlation between unliganded dimerization and tumorigenic signaling and suggest that EphA2 pro-tumorigenic activity is mediated by the EphA2 monomer. Thus, a therapeutic strategy that aims at the stabilization of EphA2 dimers may be beneficial for the treatment of cancers linked to EphA2 overexpression.


Biochemical Journal | 2015

Unliganded EphA3 dimerization promoted by the SAM domain.

Deo R. Singh; Qingqing Cao; Christopher King; Matt Salotto; Fozia Ahmed; Xiang Yang Zhou; Elena B. Pasquale; Kalina Hristova

Erythropoietin-producing hepatocellular carcinoma A3 (EphA3) can form dimers in the absence of ligand binding, which are stabilized by the sterile α-motif (SAM) domain. This challenges the current understanding of EphA3 activation events and establishes a new role for the EphA3 SAM domain in receptor-receptor interactions.


Biochimica et Biophysica Acta | 2017

The SAM domain inhibits EphA2 interactions in the plasma membrane.

Deo R. Singh; Fozia Ahmed; Michael D. Paul; Manasee Gedam; Elena B. Pasquale; Kalina Hristova

All members of the Eph receptor family of tyrosine kinases contain a SAM domain near the C terminus, which has been proposed to play a role in receptor homotypic interactions and/or interactions with binding partners. The SAM domain of EphA2 is known to be important for receptor function, but its contribution to EphA2 lateral interactions in the plasma membrane has not been determined. Here we use a FRET-based approach to directly measure the effect of the SAM domain on the stability of EphA2 dimers on the cell surface in the absence of ligand binding. We also investigate the functional consequences of EphA2 SAM domain deletion. Surprisingly, we find that the EphA2 SAM domain inhibits receptor dimerization and decreases EphA2 tyrosine phosphorylation. This role is dramatically different from the role of the SAM domain of the related EphA3 receptor, which we previously found to stabilize EphA3 dimers and increase EphA3 tyrosine phosphorylation in cells in the absence of ligand. Thus, the EphA2 SAM domain likely contributes to a unique mode of EphA2 interaction that leads to distinct signaling outputs.


Biochimica et Biophysica Acta | 2016

A small peptide promotes EphA2 kinase-dependent signaling by stabilizing EphA2 dimers

Deo R. Singh; Elena B. Pasquale; Kalina Hristova

BACKGROUND The EphA2 receptor tyrosine kinase is known to promote cancer cell malignancy in the absence of activation by ephrin ligands. This behavior depends on high EphA2 phosphorylation on Ser897 and low tyrosine phosphorylation, resulting in increased cell migration and invasiveness. We have previously shown that EphA2 forms dimers in the absence of ephrin ligand binding, and that dimerization of unliganded EphA2 can decrease EphA2 Ser897 phosphorylation. We have also identified a small peptide called YSA, which binds EphA2 and competes with the naturally occurring ephrin ligands. METHODS Here, we investigate the effect of YSA on EphA2 dimer stability and EphA2 function using quantitative FRET techniques, Western blotting, and cell motility assays. RESULTS We find that the YSA peptide stabilizes the EphA2 dimer, increases EphA2 Tyr phosphorylation, and decreases both Ser897 phosphorylation and cell migration. CONCLUSIONS The experiments demonstrate that the small peptide ligand YSA reduces EphA2 Ser897 pro-tumorigenic signaling by stabilizing the EphA2 dimer. GENERAL SIGNIFICANCE This work is a proof-of-principle demonstration that EphA2 homointeractions in the plasma membrane can be pharmacologically modulated to decrease the pro-tumorigenic signaling of the receptor.


Journal of Cell Biology | 2016

Exosomes expand the sphere of influence of Eph receptors and ephrins

Elena B. Pasquale

Membrane-anchored Eph receptors and ephrins represent a ubiquitous intercellular communication system that typically engages at sites of cell–cell contact to initiate bidirectional signaling. Gong et al. (2016. J. Cell Biol. http://dx.doi.org/10.1083/jcb.201601085) show that cells can deploy the EphB2 receptor on exosomes to activate ephrinB signaling and collapse the growth cones of distant neurons.

Collaboration


Dive into the Elena B. Pasquale's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Deo R. Singh

University of Wisconsin–Milwaukee

View shared research outputs
Top Co-Authors

Avatar

Fozia Ahmed

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Matt Salotto

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar

Nisha Gupta

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Manasee Gedam

Johns Hopkins University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qingqing Cao

Johns Hopkins University

View shared research outputs
Researchain Logo
Decentralizing Knowledge