Raft-dependent endocytosis of autocrine motility factor/phosphoglucose isomerase: a potential drug delivery route for tumor cells.

Autocrine motility factor/phosphoglucose isomerase (AMF/PGI) is the extracellular ligand for the gp78/AMFR receptor overexpressed in a variety of human cancers. We showed previously that raft-dependent internalization of AMF/PGI is elevated in metastatic MDA-435 cells, but not metastatic, caveolin-1...

Full description

Bibliographic Details
Main Authors: Liliana D Kojic, Sam M Wiseman, Fariba Ghaidi, Bharat Joshi, Hinyu Nedev, H Uri Saragovi, Ivan R Nabi
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2008-01-01
Series:PLoS ONE
Online Access:http://europepmc.org/articles/PMC2575378?pdf=render
id doaj-2d0956353d504cb8bc55f4f52ecc493f
record_format Article
spelling doaj-2d0956353d504cb8bc55f4f52ecc493f2020-11-25T00:23:25ZengPublic Library of Science (PLoS)PLoS ONE1932-62032008-01-01310e359710.1371/journal.pone.0003597Raft-dependent endocytosis of autocrine motility factor/phosphoglucose isomerase: a potential drug delivery route for tumor cells.Liliana D KojicSam M WisemanFariba GhaidiBharat JoshiHinyu NedevH Uri SaragoviIvan R NabiAutocrine motility factor/phosphoglucose isomerase (AMF/PGI) is the extracellular ligand for the gp78/AMFR receptor overexpressed in a variety of human cancers. We showed previously that raft-dependent internalization of AMF/PGI is elevated in metastatic MDA-435 cells, but not metastatic, caveolin-1-expressing MDA-231 cells, relative to non-metastatic MCF7 and dysplastic MCF10A cells suggesting that it might represent a tumor cell-specific endocytic pathway.Similarly, using flow cytometry, we demonstrate that raft-dependent endocytosis of AMF/PGI is increased in metastatic HT29 cancer cells expressing low levels of caveolin-1 relative to metastatic, caveolin-1-expressing, HCT116 colon cells and non-metastatic Caco-2 cells. Therefore, we exploited the raft-dependent internalization of AMF/PGI as a potential tumor-cell specific targeting mechanism. We synthesized an AMF/PGI-paclitaxel conjugate and found it to be as efficient as free paclitaxel in inducing cytotoxicity and apoptosis in tumor cells that readily internalize AMF/PGI compared to tumor cells that poorly internalize AMF/PGI. Murine K1735-M1 and B16-F1 melanoma cells internalize FITC-conjugated AMF/PGI and are acutely sensitive to AMF/PGI-paclitaxel mediated cytotoxicity in vitro. Moreover, following in vivo intratumoral injection, FITC-conjugated AMF/PGI is internalized in K1735-M1 tumors. Intratumoral injection of AMF/PGI-paclitaxel induced significantly higher tumor regression compared to free paclitaxel, even in B16-F1 cells, known to be resistant to taxol treatment. Treatment with AMF/PGI-paclitaxel significantly prolonged the median survival time of tumor bearing mice. Free AMF/PGI exhibited a pro-survival role, reducing the cytotoxic effect of both AMF/PGI-paclitaxel and free paclitaxel suggesting that AMF/PGI-paclitaxel targets a pathway associated with resistance to chemotherapeutic agents. AMF/PGI-FITC uptake by normal murine spleen and thymus cells was negligible both in vitro and following intravenous injection in vivo where AMF/PGI-FITC was selectively internalized by subcutaneous B16-F1 tumor cells.The raft-dependent endocytosis of AMF/PGI may therefore represent a tumor cell specific endocytic pathway of potential value for drug delivery to tumor cells.http://europepmc.org/articles/PMC2575378?pdf=render
collection DOAJ
language English
format Article
sources DOAJ
author Liliana D Kojic
Sam M Wiseman
Fariba Ghaidi
Bharat Joshi
Hinyu Nedev
H Uri Saragovi
Ivan R Nabi
spellingShingle Liliana D Kojic
Sam M Wiseman
Fariba Ghaidi
Bharat Joshi
Hinyu Nedev
H Uri Saragovi
Ivan R Nabi
Raft-dependent endocytosis of autocrine motility factor/phosphoglucose isomerase: a potential drug delivery route for tumor cells.
PLoS ONE
author_facet Liliana D Kojic
Sam M Wiseman
Fariba Ghaidi
Bharat Joshi
Hinyu Nedev
H Uri Saragovi
Ivan R Nabi
author_sort Liliana D Kojic
title Raft-dependent endocytosis of autocrine motility factor/phosphoglucose isomerase: a potential drug delivery route for tumor cells.
title_short Raft-dependent endocytosis of autocrine motility factor/phosphoglucose isomerase: a potential drug delivery route for tumor cells.
title_full Raft-dependent endocytosis of autocrine motility factor/phosphoglucose isomerase: a potential drug delivery route for tumor cells.
title_fullStr Raft-dependent endocytosis of autocrine motility factor/phosphoglucose isomerase: a potential drug delivery route for tumor cells.
title_full_unstemmed Raft-dependent endocytosis of autocrine motility factor/phosphoglucose isomerase: a potential drug delivery route for tumor cells.
title_sort raft-dependent endocytosis of autocrine motility factor/phosphoglucose isomerase: a potential drug delivery route for tumor cells.
publisher Public Library of Science (PLoS)
series PLoS ONE
issn 1932-6203
publishDate 2008-01-01
description Autocrine motility factor/phosphoglucose isomerase (AMF/PGI) is the extracellular ligand for the gp78/AMFR receptor overexpressed in a variety of human cancers. We showed previously that raft-dependent internalization of AMF/PGI is elevated in metastatic MDA-435 cells, but not metastatic, caveolin-1-expressing MDA-231 cells, relative to non-metastatic MCF7 and dysplastic MCF10A cells suggesting that it might represent a tumor cell-specific endocytic pathway.Similarly, using flow cytometry, we demonstrate that raft-dependent endocytosis of AMF/PGI is increased in metastatic HT29 cancer cells expressing low levels of caveolin-1 relative to metastatic, caveolin-1-expressing, HCT116 colon cells and non-metastatic Caco-2 cells. Therefore, we exploited the raft-dependent internalization of AMF/PGI as a potential tumor-cell specific targeting mechanism. We synthesized an AMF/PGI-paclitaxel conjugate and found it to be as efficient as free paclitaxel in inducing cytotoxicity and apoptosis in tumor cells that readily internalize AMF/PGI compared to tumor cells that poorly internalize AMF/PGI. Murine K1735-M1 and B16-F1 melanoma cells internalize FITC-conjugated AMF/PGI and are acutely sensitive to AMF/PGI-paclitaxel mediated cytotoxicity in vitro. Moreover, following in vivo intratumoral injection, FITC-conjugated AMF/PGI is internalized in K1735-M1 tumors. Intratumoral injection of AMF/PGI-paclitaxel induced significantly higher tumor regression compared to free paclitaxel, even in B16-F1 cells, known to be resistant to taxol treatment. Treatment with AMF/PGI-paclitaxel significantly prolonged the median survival time of tumor bearing mice. Free AMF/PGI exhibited a pro-survival role, reducing the cytotoxic effect of both AMF/PGI-paclitaxel and free paclitaxel suggesting that AMF/PGI-paclitaxel targets a pathway associated with resistance to chemotherapeutic agents. AMF/PGI-FITC uptake by normal murine spleen and thymus cells was negligible both in vitro and following intravenous injection in vivo where AMF/PGI-FITC was selectively internalized by subcutaneous B16-F1 tumor cells.The raft-dependent endocytosis of AMF/PGI may therefore represent a tumor cell specific endocytic pathway of potential value for drug delivery to tumor cells.
url http://europepmc.org/articles/PMC2575378?pdf=render
work_keys_str_mv AT lilianadkojic raftdependentendocytosisofautocrinemotilityfactorphosphoglucoseisomeraseapotentialdrugdeliveryroutefortumorcells
AT sammwiseman raftdependentendocytosisofautocrinemotilityfactorphosphoglucoseisomeraseapotentialdrugdeliveryroutefortumorcells
AT faribaghaidi raftdependentendocytosisofautocrinemotilityfactorphosphoglucoseisomeraseapotentialdrugdeliveryroutefortumorcells
AT bharatjoshi raftdependentendocytosisofautocrinemotilityfactorphosphoglucoseisomeraseapotentialdrugdeliveryroutefortumorcells
AT hinyunedev raftdependentendocytosisofautocrinemotilityfactorphosphoglucoseisomeraseapotentialdrugdeliveryroutefortumorcells
AT hurisaragovi raftdependentendocytosisofautocrinemotilityfactorphosphoglucoseisomeraseapotentialdrugdeliveryroutefortumorcells
AT ivanrnabi raftdependentendocytosisofautocrinemotilityfactorphosphoglucoseisomeraseapotentialdrugdeliveryroutefortumorcells
_version_ 1725357244181118976