Modulation of Rho-Rock signaling pathway protects oligodendrocytes against cytokine toxicity via PPAR-α-dependent mechanism

Glia. 2013 Sep;61(9):1500-1517. doi: 10.1002/glia.22537. Epub 2013 Jul 10.

Abstract

We earlier documented that lovastatin (LOV)-mediated inhibition of small Rho GTPases activity protects vulnerable oligodendrocytes (OLs) in mixed glial cell cultures stimulated with Th1 cytokines and in a murine model of multiple sclerosis (MS). However, the precise mechanism of OL protection remains unclear. We here employed genetic and biochemical approaches to elucidate the underlying mechanism that protects LOV treated OLs from Th1 (tumor necrosis factor-α) and Th17 (interleukin-17) cytokines toxicity in in vitro. Cytokines enhanced the reactive oxygen species (ROS) generation and mitochondrial membrane depolarization with corresponding lowering of glutathione (reduced) level in OLs and that were reverted by LOV. In addition, the expression of ROS detoxifying enzymes (catalase and superoxide-dismutase 2) and the transactivation of peroxisome proliferators-activated receptor (PPAR)-α/-β/-γ including PPAR-γ coactivator-1α were enhanced by LOV in similarly treated OLs. Interestingly, LOV-mediated inhibition of small Rho GTPases, i.e., RhoA and cdc42, and Rho-associated kinase (ROCK) activity enhanced the levels of PPAR ligands in OLs via extracellular signal regulated kinase (1/2)/p38 mitogen-activated protein kinase/cytoplasmic phospholipase 2/cyclooxygenase-2 signaling cascade activation. Small hairpin RNA transfection-based studies established that LOV mainly enhances PPAR-α and less so of PPAR-β and PPAR-γ transactivation that enhances ROS detoxifying defense in OLs. In support of this, the observed LOV-mediated protection was lacking in PPAR-α-deficient OLs exposed to cytokines. Collectively, these data provide unprecedented evidence that LOV-mediated inhibition of the Rho-ROCK signaling pathway boosts ROS detoxifying defense in OLs via PPAR-α-dependent mechanism that has implication in neurodegenerative disorders including MS.

Keywords: EAE/MS; PPAR-α; RhoA-ROCK; differentiation; lovastatin; oligodendrocyte progenitors; survival.

Publication types

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

MeSH terms

  • Analysis of Variance
  • Animals
  • Animals, Newborn
  • Cell Survival
  • Cells, Cultured
  • Cerebral Cortex / cytology
  • Cytokines / toxicity*
  • Enzyme Inhibitors / pharmacology
  • Female
  • Galactosides / metabolism
  • Glutathione / metabolism
  • Hydroxymethylglutaryl-CoA Reductase Inhibitors / pharmacology
  • L-Lactate Dehydrogenase / metabolism
  • Lovastatin / pharmacology
  • Membrane Potential, Mitochondrial / drug effects
  • Membrane Potential, Mitochondrial / physiology
  • Mice
  • Mice, Knockout
  • Neurons / physiology
  • Oligodendroglia / drug effects*
  • PPAR alpha / deficiency
  • PPAR alpha / metabolism*
  • Pregnancy
  • Rats
  • Rats, Sprague-Dawley
  • Reactive Oxygen Species / metabolism
  • Signal Transduction / drug effects
  • Signal Transduction / physiology*
  • Transfection
  • rho GTP-Binding Proteins / metabolism*
  • rho-Associated Kinases / metabolism*

Substances

  • Cytokines
  • Enzyme Inhibitors
  • Galactosides
  • Hydroxymethylglutaryl-CoA Reductase Inhibitors
  • PPAR alpha
  • Reactive Oxygen Species
  • beta-galactoside
  • Lovastatin
  • L-Lactate Dehydrogenase
  • rho-Associated Kinases
  • rho GTP-Binding Proteins
  • Glutathione