Inhibition of Autotaxin and Lysophosphatidic Acid Receptor 5 Attenuates Neuroinflammation in LPS-Activated BV-2 Microglia and a Mouse Endotoxemia Model

Int J Mol Sci. 2021 Aug 7;22(16):8519. doi: 10.3390/ijms22168519.

Abstract

Increasing evidence suggests that systemic inflammation triggers a neuroinflammatory response that involves sustained microglia activation. This response has deleterious consequences on memory and learning capability in experimental animal models and in patients. However, the mechanisms connecting systemic inflammation and microglia activation remain poorly understood. Here, we identify the autotaxin (ATX)/lysophosphatidic acid (LPA)/LPA-receptor axis as a potential pharmacological target to modulate the LPS-mediated neuroinflammatory response in vitro (the murine BV-2 microglia cell line) and in vivo (C57BL/6J mice receiving a single i.p. LPS injection). In LPS-stimulated (20 ng/mL) BV-2 cells, we observed increased phosphorylation of transcription factors (STAT1, p65, and c-Jun) that are known to induce a proinflammatory microglia phenotype. LPS upregulated ATX, TLR4, and COX2 expression, amplified NO production, increased neurotoxicity of microglia conditioned medium, and augmented cyto-/chemokine concentrations in the cellular supernatants. PF8380 (a type I ATX inhibitor, used at 10 and 1 µM) and AS2717638 (an LPA5 antagonist, used at 1 and 0.1 µM) attenuated these proinflammatory responses, at non-toxic concentrations, in BV-2 cells. In vivo, we demonstrate accumulation of PF8380 in the mouse brain and an accompanying decrease in LPA concentrations. In vivo, co-injection of LPS (5 mg/kg body weight) and PF8380 (30 mg/kg body weight), or LPS/AS2717638 (10 mg/kg body weight), significantly attenuated LPS-induced iNOS, TNFα, IL-1β, IL-6, and CXCL2 mRNA expression in the mouse brain. On the protein level, PF8380 and AS2717638 significantly reduced TLR4, Iba1, GFAP and COX2 expression, as compared to LPS-only injected animals. In terms of the communication between systemic inflammation and neuroinflammation, both inhibitors significantly attenuated LPS-mediated systemic TNFα and IL-6 synthesis, while IL-1β was only reduced by PF8380. Inhibition of ATX and LPA5 may thus provide an opportunity to protect the brain from the toxic effects that are provoked by systemic endotoxemia.

Keywords: AS2717638; PF8380; chemokines; cytokines; neurotoxicity.

MeSH terms

  • Animals
  • Benzoxazoles / pharmacology*
  • Brain / metabolism*
  • Brain / pathology
  • Cell Line
  • Disease Models, Animal
  • Endotoxemia* / chemically induced
  • Endotoxemia* / metabolism
  • Endotoxemia* / pathology
  • Inflammation / chemically induced
  • Inflammation / metabolism
  • Inflammation / pathology
  • Isoquinolines / pharmacology*
  • Lipopolysaccharides / toxicity*
  • Mice
  • Microglia / metabolism*
  • Microglia / pathology
  • Phosphoric Diester Hydrolases / metabolism*
  • Piperazines / pharmacology*
  • Piperidines / pharmacology*
  • Receptors, Lysophosphatidic Acid* / antagonists & inhibitors
  • Receptors, Lysophosphatidic Acid* / metabolism

Substances

  • 6-(3-(piperazin-1-yl)propanoyl)benzo(d)oxazol-2(3H)-one
  • AS2717638
  • Benzoxazoles
  • Isoquinolines
  • LPAR5 protein, mouse
  • Lipopolysaccharides
  • Piperazines
  • Piperidines
  • Receptors, Lysophosphatidic Acid
  • Phosphoric Diester Hydrolases
  • alkylglycerophosphoethanolamine phosphodiesterase