Cargando…

PEGylation Potentiates the Effectiveness of an Antagonistic Peptide That Targets the EphB4 Receptor with Nanomolar Affinity

The EphB4 receptor tyrosine kinase together with its preferred ligand, ephrin-B2, regulates a variety of physiological and pathological processes, including tumor progression, pathological forms of angiogenesis, cardiomyocyte differentiation and bone remodeling. We previously reported the identifica...

Descripción completa

Detalles Bibliográficos
Autores principales: Noberini, Roberta, Mitra, Sayantan, Salvucci, Ombretta, Valencia, Fatima, Duggineni, Srinivas, Prigozhina, Natalie, Wei, Ke, Tosato, Giovanna, Huang, Ziwei, Pasquale, Elena B.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2011
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3237458/
https://www.ncbi.nlm.nih.gov/pubmed/22194865
http://dx.doi.org/10.1371/journal.pone.0028611
_version_ 1782218898188271616
author Noberini, Roberta
Mitra, Sayantan
Salvucci, Ombretta
Valencia, Fatima
Duggineni, Srinivas
Prigozhina, Natalie
Wei, Ke
Tosato, Giovanna
Huang, Ziwei
Pasquale, Elena B.
author_facet Noberini, Roberta
Mitra, Sayantan
Salvucci, Ombretta
Valencia, Fatima
Duggineni, Srinivas
Prigozhina, Natalie
Wei, Ke
Tosato, Giovanna
Huang, Ziwei
Pasquale, Elena B.
author_sort Noberini, Roberta
collection PubMed
description The EphB4 receptor tyrosine kinase together with its preferred ligand, ephrin-B2, regulates a variety of physiological and pathological processes, including tumor progression, pathological forms of angiogenesis, cardiomyocyte differentiation and bone remodeling. We previously reported the identification of TNYL-RAW, a 15 amino acid-long peptide that binds to the ephrin-binding pocked of EphB4 with low nanomolar affinity and inhibits ephrin-B2 binding. Although ephrin-B2 interacts promiscuously with all the EphB receptors, the TNYL-RAW peptide is remarkably selective and only binds to EphB4. Therefore, this peptide is a useful tool for studying the biological functions of EphB4 and for imaging EphB4-expressing tumors. Furthermore, TNYL-RAW could be useful for treating pathologies involving EphB4-ephrin-B2 interaction. However, the peptide has a very short half-life in cell culture and in the mouse blood circulation due to proteolytic degradation and clearance by the kidneys and reticuloendothelial system. To overcome these limitations, we have modified TNYL-RAW by fusion with the Fc portion of human IgG1, complexation with streptavidin or covalent coupling to a 40 KDa branched polyethylene glycol (PEG) polymer. These modified forms of TNYL-RAW all have greatly increased stability in cell culture, while retaining high binding affinity for EphB4. Furthermore, PEGylation most effectively increases peptide half-life in vivo. Consistent with increased stability, submicromolar concentrations of PEGylated TNYL-RAW effectively impair EphB4 activation by ephrin-B2 in cultured B16 melanoma cells as well as capillary-like tube formation and capillary sprouting in co-cultures of endothelial and epicardial mesothelial cells. Therefore, PEGylated TNYL-RAW may be useful for inhibiting pathological forms of angiogenesis through a novel mechanism involving disruption of EphB4-ephrin-B2 interactions between endothelial cells and supporting perivascular mesenchymal cells. Furthermore, the PEGylated peptide is suitable for other cell culture and in vivo applications requiring prolonged EphB4 receptor targeting.
format Online
Article
Text
id pubmed-3237458
institution National Center for Biotechnology Information
language English
publishDate 2011
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-32374582011-12-22 PEGylation Potentiates the Effectiveness of an Antagonistic Peptide That Targets the EphB4 Receptor with Nanomolar Affinity Noberini, Roberta Mitra, Sayantan Salvucci, Ombretta Valencia, Fatima Duggineni, Srinivas Prigozhina, Natalie Wei, Ke Tosato, Giovanna Huang, Ziwei Pasquale, Elena B. PLoS One Research Article The EphB4 receptor tyrosine kinase together with its preferred ligand, ephrin-B2, regulates a variety of physiological and pathological processes, including tumor progression, pathological forms of angiogenesis, cardiomyocyte differentiation and bone remodeling. We previously reported the identification of TNYL-RAW, a 15 amino acid-long peptide that binds to the ephrin-binding pocked of EphB4 with low nanomolar affinity and inhibits ephrin-B2 binding. Although ephrin-B2 interacts promiscuously with all the EphB receptors, the TNYL-RAW peptide is remarkably selective and only binds to EphB4. Therefore, this peptide is a useful tool for studying the biological functions of EphB4 and for imaging EphB4-expressing tumors. Furthermore, TNYL-RAW could be useful for treating pathologies involving EphB4-ephrin-B2 interaction. However, the peptide has a very short half-life in cell culture and in the mouse blood circulation due to proteolytic degradation and clearance by the kidneys and reticuloendothelial system. To overcome these limitations, we have modified TNYL-RAW by fusion with the Fc portion of human IgG1, complexation with streptavidin or covalent coupling to a 40 KDa branched polyethylene glycol (PEG) polymer. These modified forms of TNYL-RAW all have greatly increased stability in cell culture, while retaining high binding affinity for EphB4. Furthermore, PEGylation most effectively increases peptide half-life in vivo. Consistent with increased stability, submicromolar concentrations of PEGylated TNYL-RAW effectively impair EphB4 activation by ephrin-B2 in cultured B16 melanoma cells as well as capillary-like tube formation and capillary sprouting in co-cultures of endothelial and epicardial mesothelial cells. Therefore, PEGylated TNYL-RAW may be useful for inhibiting pathological forms of angiogenesis through a novel mechanism involving disruption of EphB4-ephrin-B2 interactions between endothelial cells and supporting perivascular mesenchymal cells. Furthermore, the PEGylated peptide is suitable for other cell culture and in vivo applications requiring prolonged EphB4 receptor targeting. Public Library of Science 2011-12-14 /pmc/articles/PMC3237458/ /pubmed/22194865 http://dx.doi.org/10.1371/journal.pone.0028611 Text en This is an open-access article, free of all copyright, and may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose. The work is made available under the Creative Commons CC0 public domain dedication. https://creativecommons.org/publicdomain/zero/1.0/ This is an open-access article distributed under the terms of the Creative Commons Public Domain declaration, which stipulates that, once placed in the public domain, this work may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose.
spellingShingle Research Article
Noberini, Roberta
Mitra, Sayantan
Salvucci, Ombretta
Valencia, Fatima
Duggineni, Srinivas
Prigozhina, Natalie
Wei, Ke
Tosato, Giovanna
Huang, Ziwei
Pasquale, Elena B.
PEGylation Potentiates the Effectiveness of an Antagonistic Peptide That Targets the EphB4 Receptor with Nanomolar Affinity
title PEGylation Potentiates the Effectiveness of an Antagonistic Peptide That Targets the EphB4 Receptor with Nanomolar Affinity
title_full PEGylation Potentiates the Effectiveness of an Antagonistic Peptide That Targets the EphB4 Receptor with Nanomolar Affinity
title_fullStr PEGylation Potentiates the Effectiveness of an Antagonistic Peptide That Targets the EphB4 Receptor with Nanomolar Affinity
title_full_unstemmed PEGylation Potentiates the Effectiveness of an Antagonistic Peptide That Targets the EphB4 Receptor with Nanomolar Affinity
title_short PEGylation Potentiates the Effectiveness of an Antagonistic Peptide That Targets the EphB4 Receptor with Nanomolar Affinity
title_sort pegylation potentiates the effectiveness of an antagonistic peptide that targets the ephb4 receptor with nanomolar affinity
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3237458/
https://www.ncbi.nlm.nih.gov/pubmed/22194865
http://dx.doi.org/10.1371/journal.pone.0028611
work_keys_str_mv AT noberiniroberta pegylationpotentiatestheeffectivenessofanantagonisticpeptidethattargetstheephb4receptorwithnanomolaraffinity
AT mitrasayantan pegylationpotentiatestheeffectivenessofanantagonisticpeptidethattargetstheephb4receptorwithnanomolaraffinity
AT salvucciombretta pegylationpotentiatestheeffectivenessofanantagonisticpeptidethattargetstheephb4receptorwithnanomolaraffinity
AT valenciafatima pegylationpotentiatestheeffectivenessofanantagonisticpeptidethattargetstheephb4receptorwithnanomolaraffinity
AT dugginenisrinivas pegylationpotentiatestheeffectivenessofanantagonisticpeptidethattargetstheephb4receptorwithnanomolaraffinity
AT prigozhinanatalie pegylationpotentiatestheeffectivenessofanantagonisticpeptidethattargetstheephb4receptorwithnanomolaraffinity
AT weike pegylationpotentiatestheeffectivenessofanantagonisticpeptidethattargetstheephb4receptorwithnanomolaraffinity
AT tosatogiovanna pegylationpotentiatestheeffectivenessofanantagonisticpeptidethattargetstheephb4receptorwithnanomolaraffinity
AT huangziwei pegylationpotentiatestheeffectivenessofanantagonisticpeptidethattargetstheephb4receptorwithnanomolaraffinity
AT pasqualeelenab pegylationpotentiatestheeffectivenessofanantagonisticpeptidethattargetstheephb4receptorwithnanomolaraffinity