Low-level laser therapy regulates microglial function through Src-mediated signaling pathways: implications for neurodegenerative diseases

J Neuroinflammation. 2012 Sep 18:9:219. doi: 10.1186/1742-2094-9-219.

Abstract

Background: Activated microglial cells are an important pathological component in brains of patients with neurodegenerative diseases. The purpose of this study was to investigate the effect of He-Ne (632.8 nm, 64.6 mW/cm2) low-level laser therapy (LLLT), a non-damaging physical therapy, on activated microglia, and the subsequent signaling events of LLLT-induced neuroprotective effects and phagocytic responses.

Methods: To model microglial activation, we treated the microglial BV2 cells with lipopolysaccharide (LPS). For the LLLT-induced neuroprotective study, neuronal cells with activated microglial cells in a Transwell™ cell-culture system were used. For the phagocytosis study, fluorescence-labeled microspheres were added into the treated microglial cells to confirm the role of LLLT.

Results: Our results showed that LLLT (20 J/cm2) could attenuate toll-like receptor (TLR)-mediated proinflammatory responses in microglia, characterized by down-regulation of proinflammatory cytokine expression and nitric oxide (NO) production. LLLT-triggered TLR signaling inhibition was achieved by activating tyrosine kinases Src and Syk, which led to MyD88 tyrosine phosphorylation, thus impairing MyD88-dependent proinflammatory signaling cascade. In addition, we found that Src activation could enhance Rac1 activity and F-actin accumulation that typify microglial phagocytic activity. We also found that Src/PI3K/Akt inhibitors prevented LLLT-stimulated Akt (Ser473 and Thr308) phosphorylation and blocked Rac1 activity and actin-based microglial phagocytosis, indicating the activation of Src/PI3K/Akt/Rac1 signaling pathway.

Conclusions: The present study underlines the importance of Src in suppressing inflammation and enhancing microglial phagocytic function in activated microglia during LLLT stimulation. We have identified a new and important neuroprotective signaling pathway that consists of regulation of microglial phagocytosis and inflammation under LLLT treatment. Our research may provide a feasible therapeutic approach to control the progression of neurodegenerative diseases.

Publication types

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

MeSH terms

  • Actins / metabolism
  • Analysis of Variance
  • Animals
  • Animals, Newborn
  • Brain / cytology
  • Cells, Cultured
  • Chromones / pharmacology
  • Cyclic N-Oxides / pharmacology
  • Cytokines / metabolism
  • Cytotoxicity Tests, Immunologic
  • Enzyme Inhibitors / pharmacology
  • Free Radical Scavengers / pharmacology
  • Gene Expression Regulation / drug effects
  • Gene Expression Regulation / radiation effects
  • Humans
  • Imidazoles / pharmacology
  • Lipopolysaccharides / pharmacology
  • Low-Level Light Therapy*
  • Mice
  • Mice, Inbred C57BL
  • Microglia / radiation effects*
  • Microscopy, Confocal
  • Morpholines / pharmacology
  • Myeloid Differentiation Factor 88 / metabolism
  • Neuroblastoma / pathology
  • Nitric Oxide / metabolism
  • Phagocytosis / drug effects
  • Phagocytosis / radiation effects
  • Phalloidine / metabolism
  • Proto-Oncogene Proteins pp60(c-src) / metabolism*
  • Signal Transduction / drug effects
  • Signal Transduction / physiology*
  • Signal Transduction / radiation effects
  • Statistics as Topic
  • Time Factors
  • Transfection
  • Tyrosine / metabolism
  • rac1 GTP-Binding Protein / metabolism

Substances

  • Actins
  • Chromones
  • Cyclic N-Oxides
  • Cytokines
  • Enzyme Inhibitors
  • Free Radical Scavengers
  • Imidazoles
  • LY 290042
  • Lipopolysaccharides
  • Morpholines
  • Myd88 protein, mouse
  • Myeloid Differentiation Factor 88
  • Phalloidine
  • 2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide
  • Nitric Oxide
  • Tyrosine
  • Proto-Oncogene Proteins pp60(c-src)
  • rac1 GTP-Binding Protein