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 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.

[1]  L. Chin,et al.  Malignant astrocytic glioma: genetics, biology, and paths to treatment. , 2007, Genes & development.

[2]  Yuri Kotliarov,et al.  Tumor stem cells derived from glioblastomas cultured in bFGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum-cultured cell lines. , 2006, Cancer cell.

[3]  M. Westphal,et al.  Glioma invasion in the central nervous system. , 1996, Neurosurgery.

[4]  H. Hanafusa,et al.  Biological Effects of c-Mer Receptor Tyrosine Kinase in Hematopoietic Cells Depend on the Grb2 Binding Site in the Receptor and Activation of NF-κB , 1999, Molecular and Cellular Biology.

[5]  H. Hanafusa,et al.  PTEN Induces Cell Cycle Arrest by Decreasing the Level and Nuclear Localization of Cyclin D1 , 2003, Molecular and Cellular Biology.

[6]  Ali H. Brivanlou,et al.  Signaling Pathways in Cancer and Embryonic Stem Cells , 2007, Stem Cell Reviews.

[7]  P. Kleihues,et al.  Genetic pathways to primary and secondary glioblastoma. , 2007, The American journal of pathology.

[8]  H. Clevers,et al.  Stem cells, self-renewal, and differentiation in the intestinal epithelium. , 2009, Annual review of physiology.

[9]  I. Jonassen,et al.  Angiogenesis-independent tumor growth mediated by stem-like cancer cells , 2006, Proceedings of the National Academy of Sciences.

[10]  Erwin G. Van Meir,et al.  Tumor initiating cells in malignant gliomas: biology and implications for therapy , 2009, Journal of Molecular Medicine.

[11]  M. Georgescu,et al.  Overexpression of ezrin inactivates NF2 tumor suppressor in glioblastoma. , 2010, Neuro-oncology.

[12]  Seung-Hoon Lee,et al.  Role of hyaluronan in glioma invasion , 2008, Cell adhesion & migration.

[13]  Yoko Takahashi,et al.  PTEN tumor suppressor associates with NHERF proteins to attenuate PDGF receptor signaling , 2006, The EMBO journal.

[14]  H. Hanafusa,et al.  The tumor-suppressor activity of PTEN is regulated by its carboxyl-terminal region. , 1999, Proceedings of the National Academy of Sciences of the United States of America.

[15]  L. S. Harrington,et al.  Restraining PI3K: mTOR signalling goes back to the membrane. , 2005, Trends in biochemical sciences.

[16]  Khalid Shah,et al.  Human glioblastoma-derived cancer stem cells: establishment of invasive glioma models and treatment with oncolytic herpes simplex virus vectors. , 2009, Cancer research.

[17]  E. Radaelli,et al.  Immunohistopathological and neuroimaging characterization of murine orthotopic xenograft models of glioblastoma multiforme recapitulating the most salient features of human disease. , 2009, Histology and histopathology.

[18]  K. Moelling,et al.  Phosphorylation and regulation of Raf by Akt (protein kinase B). , 1999, Science.

[19]  Magnus Bosse,et al.  Regulation of Raf-Akt Cross-talk , 2002, The Journal of Biological Chemistry.

[20]  Michael E. Berens,et al.  Glioma Cell Motility is Associated with Reduced Transcription of Proapoptotic and Proliferation Genes: A cDNA Microarray Analysis , 2001, Journal of Neuro-Oncology.

[21]  Yoko Takahashi,et al.  Ezrin-radixin-moesin (ERM)-binding phosphoprotein 50 organizes ERM proteins at the apical membrane of polarized epithelia. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[22]  M. Georgescu,et al.  Random selection: a model for poliovirus infection of the central nervous system. , 1997, The Journal of general virology.

[23]  A. Newton,et al.  PHLPP: a phosphatase that directly dephosphorylates Akt, promotes apoptosis, and suppresses tumor growth. , 2005, Molecular cell.

[24]  Daniel J Brat,et al.  Microregional extracellular matrix heterogeneity in brain modulates glioma cell invasion. , 2004, The international journal of biochemistry & cell biology.

[25]  R. Bjerkvig,et al.  Stimulation of extracellular matrix components in the normal brain by invading glioma cells , 1998, International journal of cancer.

[26]  R. Bjerkvig,et al.  Migratory pattern of fetal rat brain cells and human glioma cells in the adult rat brain. , 1993, Cancer research.

[27]  R. Beroukhim,et al.  Proteasomal and genetic inactivation of the NF1 tumor suppressor in gliomagenesis. , 2009, Cancer cell.

[28]  W. Arap,et al.  Bottom-Up Assembly of Hydrogels from Bacteriophage and Au Nanoparticles: The Effect of Cis- and Trans-Acting Factors , 2008, PloS one.

[29]  K. Kalland,et al.  Highly infiltrative brain tumours show reduced chemosensitivity associated with a stem cell‐like phenotype , 2009 .

[30]  I. Weissman,et al.  Stem cells, cancer, and cancer stem cells , 2001, Nature.

[31]  A. Newton,et al.  PHLPP and a second isoform, PHLPP2, differentially attenuate the amplitude of Akt signaling by regulating distinct Akt isoforms. , 2007, Molecular cell.

[32]  I. Bayazitov,et al.  A perivascular niche for brain tumor stem cells. , 2007, Cancer cell.

[33]  M. Sulis,et al.  PTEN: from pathology to biology. , 2003, Trends in cell biology.

[34]  Yong Yang,et al.  Isolation and characterization of cancer stem like cells in human glioblastoma cell lines. , 2009, Cancer letters.

[35]  W. K. Alfred Yung,et al.  Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers , 1997, Nature Genetics.

[36]  G. Fuller,et al.  An implantable guide-screw system for brain tumor studies in small animals. , 2000, Journal of neurosurgery.

[37]  James A Bankson,et al.  Three-dimensional tissue culture based on magnetic cell levitation. , 2010, Nature nanotechnology.

[38]  M. Berens,et al.  Autocrine factors that sustain glioma invasion and paracrine biology in the brain microenvironment. , 2007, Journal of the National Cancer Institute.