Invasive Glioblastoma Cells Acquire Stemness and Increased Akt Activation

Glioblastoma multiforme (GBM) is the most frequent and most aggressive brain tumor in adults. The dismal prognosis is due to postsurgery recurrences arising from escaped invasive tumor cells. The signaling pathways activated in invasive cells are under investigation, and models are currently designe...

Full description

Bibliographic Details
Main Authors: Jennifer R. Molina, Yuho Hayashi, Clifton Stephens, Maria-Magdalena Georgescu
Format: Article
Language:English
Published: Elsevier 2010-06-01
Series:Neoplasia: An International Journal for Oncology Research
Online Access:http://www.sciencedirect.com/science/article/pii/S147655861080003X
_version_ 1811245411772399616
author Jennifer R. Molina
Yuho Hayashi
Clifton Stephens
Maria-Magdalena Georgescu
author_facet Jennifer R. Molina
Yuho Hayashi
Clifton Stephens
Maria-Magdalena Georgescu
author_sort Jennifer R. Molina
collection DOAJ
description Glioblastoma multiforme (GBM) is the most frequent and most aggressive brain tumor in adults. The dismal prognosis is due to postsurgery recurrences arising from escaped invasive tumor cells. The signaling pathways activated in invasive cells are under investigation, and models are currently designed in search for therapeutic targets. We developed here an in vivo model of human invasive GBM in mouse brain from a GBM cell line with moderate tumorigenicity that allowed simultaneous primary tumor growth and dispersal of tumor cells in the brain parenchyma. This strategy allowed for the first time the isolation and characterization of matched sets of tumor mass (Core) and invasive (Inv) cells. Both cell populations, but more markedly Inv cells, acquired stem cell markers, neurosphere renewal ability, and resistance to rapamycin-induced apoptosis relative to parental cells. The comparative phenotypic analysis between Inv and Core cells showed significantly increased tumorigenicity in vivo and increased invasion with decreased proliferation in vitro for Inv cells. Examination of a large array of signaling pathways revealed extracellular signal-regulated kinase (Erk) down-modulation and Akt activation in Inv cells and an opposite profile in Core cells. Akt activation correlated with the increased tumorigenicity, stemness, and invasiveness, whereas Erk activation correlated with the proliferation of the cells. These results underscore complementary roles of the Erk and Akt pathways for GBM proliferation and dispersal and raise important implications for a concurrent inhibitory therapy.
first_indexed 2024-04-12T14:39:42Z
format Article
id doaj.art-2f5f88b5c51343fab3ad6ac05b51d4dc
institution Directory Open Access Journal
issn 1476-5586
1522-8002
language English
last_indexed 2024-04-12T14:39:42Z
publishDate 2010-06-01
publisher Elsevier
record_format Article
series Neoplasia: An International Journal for Oncology Research
spelling doaj.art-2f5f88b5c51343fab3ad6ac05b51d4dc2022-12-22T03:28:55ZengElsevierNeoplasia: An International Journal for Oncology Research1476-55861522-80022010-06-0112645346310.1593/neo.10126Invasive Glioblastoma Cells Acquire Stemness and Increased Akt ActivationJennifer R. Molina0Yuho Hayashi1Clifton Stephens2Maria-Magdalena Georgescu3Department of Neuro-oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USADepartment of Neuro-oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USADepartment of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USADepartment of Neuro-oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USAGlioblastoma multiforme (GBM) is the most frequent and most aggressive brain tumor in adults. The dismal prognosis is due to postsurgery recurrences arising from escaped invasive tumor cells. The signaling pathways activated in invasive cells are under investigation, and models are currently designed in search for therapeutic targets. We developed here an in vivo model of human invasive GBM in mouse brain from a GBM cell line with moderate tumorigenicity that allowed simultaneous primary tumor growth and dispersal of tumor cells in the brain parenchyma. This strategy allowed for the first time the isolation and characterization of matched sets of tumor mass (Core) and invasive (Inv) cells. Both cell populations, but more markedly Inv cells, acquired stem cell markers, neurosphere renewal ability, and resistance to rapamycin-induced apoptosis relative to parental cells. The comparative phenotypic analysis between Inv and Core cells showed significantly increased tumorigenicity in vivo and increased invasion with decreased proliferation in vitro for Inv cells. Examination of a large array of signaling pathways revealed extracellular signal-regulated kinase (Erk) down-modulation and Akt activation in Inv cells and an opposite profile in Core cells. Akt activation correlated with the increased tumorigenicity, stemness, and invasiveness, whereas Erk activation correlated with the proliferation of the cells. These results underscore complementary roles of the Erk and Akt pathways for GBM proliferation and dispersal and raise important implications for a concurrent inhibitory therapy.http://www.sciencedirect.com/science/article/pii/S147655861080003X
spellingShingle Jennifer R. Molina
Yuho Hayashi
Clifton Stephens
Maria-Magdalena Georgescu
Invasive Glioblastoma Cells Acquire Stemness and Increased Akt Activation
Neoplasia: An International Journal for Oncology Research
title Invasive Glioblastoma Cells Acquire Stemness and Increased Akt Activation
title_full Invasive Glioblastoma Cells Acquire Stemness and Increased Akt Activation
title_fullStr Invasive Glioblastoma Cells Acquire Stemness and Increased Akt Activation
title_full_unstemmed Invasive Glioblastoma Cells Acquire Stemness and Increased Akt Activation
title_short Invasive Glioblastoma Cells Acquire Stemness and Increased Akt Activation
title_sort invasive glioblastoma cells acquire stemness and increased akt activation
url http://www.sciencedirect.com/science/article/pii/S147655861080003X
work_keys_str_mv AT jenniferrmolina invasiveglioblastomacellsacquirestemnessandincreasedaktactivation
AT yuhohayashi invasiveglioblastomacellsacquirestemnessandincreasedaktactivation
AT cliftonstephens invasiveglioblastomacellsacquirestemnessandincreasedaktactivation
AT mariamagdalenageorgescu invasiveglioblastomacellsacquirestemnessandincreasedaktactivation