miR-182 integrates apoptosis, growth, and differentiation programs in glioblastoma

  1. Alexander H. Stegh1,2,4,5
  1. 1Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, Chicago, Illinois 60611, USA;
  2. 2The Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois 60611, USA;
  3. 3AuraSense Therapeutics, Skokie, Illinois 60077, USA;
  4. 4Department of Chemistry, Northwestern University, Evanston, Illinois 60208, USA;
  5. 5International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, USA;
  6. 6Division Hematology/Oncology, Feinberg School of Medicine, Chicago, Illinois 60611, USA;
  7. 7The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois 60611, USA;
  8. 8Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, Illinois 60611, USA;
  9. 9The Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA;
  10. 10Harvard Medical School, Boston, Massachusetts 02115, USA;
  11. 11Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA;
  12. 12Department of Genomic Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA;
  13. 13Institute for Applied Cancer Science, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA;
  14. 14Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, 60611, USA
  1. Corresponding author: a-stegh{at}northwestern.edu

Abstract

Glioblastoma multiforme (GBM) is a lethal, therapy-resistant brain cancer consisting of numerous tumor cell subpopulations, including stem-like glioma-initiating cells (GICs), which contribute to tumor recurrence following initial response to therapy. Here, we identified miR-182 as a regulator of apoptosis, growth, and differentiation programs whose expression level is correlated with GBM patient survival. Repression of Bcl2-like12 (Bcl2L12), c-Met, and hypoxia-inducible factor 2α (HIF2A) is of central importance to miR-182 anti-tumor activity, as it results in enhanced therapy susceptibility, decreased GIC sphere size, expansion, and stemness in vitro. To evaluate the tumor-suppressive function of miR-182 in vivo, we synthesized miR-182-based spherical nucleic acids (182-SNAs); i.e., gold nanoparticles covalently functionalized with mature miR-182 duplexes. Intravenously administered 182-SNAs penetrated the blood–brain/blood–tumor barriers (BBB/BTB) in orthotopic GBM xenografts and selectively disseminated throughout extravascular glioma parenchyma, causing reduced tumor burden and increased animal survival. Our results indicate that harnessing the anti-tumor activities of miR-182 via safe and robust delivery of 182-SNAs represents a novel strategy for therapeutic intervention in GBM.

Keywords

Footnotes

  • Received February 25, 2014.
  • Accepted February 26, 2015.

This article is distributed exclusively by Cold Spring Harbor Laboratory Press for the first six months after the full-issue publication date (see http://genesdev.cshlp.org/site/misc/terms.xhtml). After six months, it is available under a Creative Commons License (Attribution-NonCommercial 4.0 International), as described at http://creativecommons.org/licenses/by-nc/4.0/.

| Table of Contents

Life Science Alliance