Connective tissue growth factor (CTGF/CCN2) is negatively regulated during neuron-glioblastoma interaction

PLoS One. 2013;8(1):e55605. doi: 10.1371/journal.pone.0055605. Epub 2013 Jan 31.

Abstract

Connective-tissue growth factor (CTGF/CCN2) is a matricellular-secreted protein involved in complex processes such as wound healing, angiogenesis, fibrosis and metastasis, in the regulation of cell proliferation, migration and extracellular matrix remodeling. Glioblastoma (GBM) is the major malignant primary brain tumor and its adaptation to the central nervous system microenvironment requires the production and remodeling of the extracellular matrix. Previously, we published an in vitro approach to test if neurons can influence the expression of the GBM extracellular matrix. We demonstrated that neurons remodeled glioma cell laminin. The present study shows that neurons are also able to modulate CTGF expression in GBM. CTGF immnoreactivity and mRNA levels in GBM cells are dramatically decreased when these cells are co-cultured with neonatal neurons. As proof of particular neuron effects, neonatal neurons co-cultured onto GBM cells also inhibit the reporter luciferase activity under control of the CTGF promoter, suggesting inhibition at the transcription level. This inhibition seems to be contact-mediated, since conditioned media from embryonic or neonatal neurons do not affect CTGF expression in GBM cells. Furthermore, the inhibition of CTGF expression in GBM/neuronal co-cultures seems to affect the two main signaling pathways related to CTGF. We observed inhibition of TGFβ luciferase reporter assay; however phopho-SMAD2 levels did not change in these co-cultures. In addition levels of phospho-p44/42 MAPK were decreased in co-cultured GBM cells. Finally, in transwell migration assay, CTGF siRNA transfected GBM cells or GBM cells co-cultured with neurons showed a decrease in the migration rate compared to controls. Previous data regarding laminin and these results demonstrating that CTGF is down-regulated in GBM cells co-cultured with neonatal neurons points out an interesting view in the understanding of the tumor and cerebral microenvironment interactions and could open up new strategies as well as suggest a new target in GBM control.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Cell Communication*
  • Cell Line, Tumor
  • Cell Movement
  • Coculture Techniques
  • Connective Tissue Growth Factor / genetics
  • Connective Tissue Growth Factor / metabolism*
  • Gene Expression
  • Gene Expression Regulation, Neoplastic
  • Glioblastoma / genetics
  • Glioblastoma / metabolism*
  • Mitogen-Activated Protein Kinase 3 / metabolism
  • Neurons / metabolism*
  • Phosphorylation
  • Primary Cell Culture
  • Promoter Regions, Genetic
  • Rats
  • Signal Transduction
  • Smad2 Protein / metabolism
  • Smad3 Protein / metabolism
  • Transcriptional Activation
  • Transforming Growth Factor beta / metabolism

Substances

  • Smad2 Protein
  • Smad3 Protein
  • Transforming Growth Factor beta
  • Connective Tissue Growth Factor
  • Mitogen-Activated Protein Kinase 3

Grants and funding

This work was funded by Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq); Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ); Fundação Ary Frauzino para Pesquisa e Controle do Câncer (FAF); Programa de Núcleos de Excelência (PRONEX); Programa de Ciências Biomédicas (PCM); Instituto Nacional de Neurociência Translacional (INNT). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.