L-Type Ca2+ Channel Inhibition Rescues the LPS-Induced Neuroinflammatory Response and Impairments in Spatial Memory and Dendritic Spine Formation

Int J Mol Sci. 2022 Nov 6;23(21):13606. doi: 10.3390/ijms232113606.

Abstract

Ca2+ signaling is implicated in the transition between microglial surveillance and activation. Several L-type Ca2+ channel blockers (CCBs) have been shown to ameliorate neuroinflammation by modulating microglial activity. In this study, we examined the effects of the L-type CCB felodipine on LPS-mediated proinflammatory responses. We found that felodipine treatment significantly diminished LPS-evoked proinflammatory cytokine levels in BV2 microglial cells in an L-type Ca2+ channel-dependent manner. In addition, felodipine leads to the inhibition of TLR4/AKT/STAT3 signaling in BV2 microglial cells. We further examined the effects of felodipine on LPS-stimulated neuroinflammation in vivo and found that daily administration (3 or 7 days, i.p.) significantly reduced LPS-mediated gliosis and COX-2 and IL-1β levels in C57BL/6 (wild-type) mice. Moreover, felodipine administration significantly reduced chronic neuroinflammation-induced spatial memory impairment, dendritic spine number, and microgliosis in C57BL/6 mice. Taken together, our results suggest that the L-type CCB felodipine could be repurposed for the treatment of neuroinflammation/cognitive function-associated diseases.

Keywords: Ca2+ channel blocker; LPS; felodipine; gliosis; neuroinflammation; spatial memory.

MeSH terms

  • Animals
  • Dendritic Spines
  • Felodipine / adverse effects
  • Inflammation / chemically induced
  • Inflammation / drug therapy
  • Lipopolysaccharides* / toxicity
  • Mice
  • Mice, Inbred C57BL
  • Microglia
  • Spatial Memory*

Substances

  • Lipopolysaccharides
  • Felodipine