Functional effects of GRM1 suppression in human melanoma cells

Mol Cancer Res. 2012 Nov;10(11):1440-50. doi: 10.1158/1541-7786.MCR-12-0158. Epub 2012 Jul 13.

Abstract

Ectopic expression of a neuronal receptor, metabotropic glutamate receptor 1 (Grm1), in melanocytes has been implicated in melanoma development in mouse models. The human relevance of this receptor's involvement in melanoma pathogenesis was shown by detecting GRM1 expression in subsets of human melanomas, an observation lacking in benign nevi or normal melanocytes. Grm1-transformed mouse melanocytes and a conditional Grm1 transgenic mouse model confirmed a requirement for sustained expression of Grm1 for the maintenance of transformed phenotypes in vitro and tumorigenicity in vivo. Here, we investigate if continued GRM1 expression is also required in human melanoma cell lines by using two inducible, silencing RNA systems: the ecdysone/Ponasterone A and tetracycline on/off approaches to regulate GRM1 expression in the presence of each inducer. Various in vitro assays were conducted to assess the consequences of a reduction in GRM1 expression on cell proliferation, apoptosis, downstream targeted signaling pathways, and in vivo tumorigenesis. We showed that suppression of GRM1 expression in several human melanoma cell lines resulted in a reduction in the number of viable cells and a decrease in stimulated mitogen-activated protein kinase (MAPK) and PI3K/AKT and suppressed tumor progression in vivo. These results reinforce earlier observations where a reduction in cell growth in vitro and tumorigenesis in vivo were correlated with decreased GRM1 activities by pharmacologic inhibitors of the receptor, supporting the notion that GRM1 plays a role in the maintenance of transformed phenotypes in human melanoma cells in vitro and in vivo and could be a potential therapeutic target for the treatment of melanoma.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Apoptosis / genetics
  • Cell Growth Processes / genetics
  • Cell Line, Tumor
  • Doxycycline / pharmacology
  • Ecdysterone / analogs & derivatives
  • Ecdysterone / pharmacology
  • Gene Expression Regulation, Neoplastic
  • Genetic Therapy
  • HEK293 Cells
  • Humans
  • Immunohistochemistry
  • MAP Kinase Signaling System
  • Melanoma / genetics*
  • Melanoma / metabolism
  • Melanoma / pathology
  • Melanoma / therapy
  • Mice
  • Mice, Nude
  • Mice, Transgenic
  • Mitogen-Activated Protein Kinases / metabolism
  • Oncogene Protein v-akt / antagonists & inhibitors
  • Oncogene Protein v-akt / metabolism
  • Phosphatidylinositol 3-Kinases / metabolism
  • Phosphorylation
  • RNA, Small Interfering / administration & dosage
  • RNA, Small Interfering / genetics
  • Random Allocation
  • Receptors, Metabotropic Glutamate / antagonists & inhibitors
  • Receptors, Metabotropic Glutamate / deficiency*
  • Receptors, Metabotropic Glutamate / genetics*
  • Receptors, Metabotropic Glutamate / metabolism
  • Skin Neoplasms / genetics*
  • Skin Neoplasms / metabolism
  • Skin Neoplasms / pathology
  • Skin Neoplasms / therapy
  • Xenograft Model Antitumor Assays

Substances

  • RNA, Small Interfering
  • Receptors, Metabotropic Glutamate
  • metabotropic glutamate receptor type 1
  • Ecdysterone
  • ponasterone A
  • Phosphatidylinositol 3-Kinases
  • Oncogene Protein v-akt
  • Mitogen-Activated Protein Kinases
  • Doxycycline