Impact of ambient temperature on inflammation-induced encephalopathy in endotoxemic mice-role of phosphoinositide 3-kinase gamma

J Neuroinflammation. 2020 Oct 7;17(1):292. doi: 10.1186/s12974-020-01954-7.

Abstract

Background: Sepsis-associated encephalopathy (SAE) is an early and frequent event of infection-induced systemic inflammatory response syndrome. Phosphoinositide 3-kinase γ (PI3Kγ) is linked to neuroinflammation and inflammation-related microglial activity. In homeotherms, variations in ambient temperature (Ta) outside the thermoneutral zone lead to thermoregulatory responses, mainly driven by a gradually increasing sympathetic activity, and may affect disease severity. We hypothesized that thermoregulatory response to hypothermia (reduced Ta) aggravates SAE in PI3Kγ-dependent manner.

Methods: Experiments were performed in wild-type, PI3Kγ knockout, and PI3Kγ kinase-dead mice, which were kept at neutral (30 ± 0.5 °C) or moderately lowered (26 ± 0.5 °C) Ta. Mice were exposed to lipopolysaccharide (LPS, 10 μg/g, from Escherichia coli serotype 055:B5, single intraperitoneal injection)-evoked systemic inflammatory response (SIR) and monitored 24 h for thermoregulatory response and blood-brain barrier integrity. Primary microglial cells and brain tissue derived from treated mice were analyzed for inflammatory responses and related cell functions. Comparisons between groups were made with one-way or two-way analysis of variance, as appropriate. Post hoc comparisons were made with the Holm-Sidak test or t tests with Bonferroni's correction for adjustments of multiple comparisons. Data not following normal distribution was tested with Kruskal-Wallis test followed by Dunn's multiple comparisons test.

Results: We show that a moderate reduction of ambient temperature triggers enhanced hypothermia of mice undergoing LPS-induced systemic inflammation by aggravated SAE. PI3Kγ deficiency enhances blood-brain barrier injury and upregulation of matrix metalloproteinases (MMPs) as well as an impaired microglial phagocytic activity.

Conclusions: Thermoregulatory adaptation in response to ambient temperatures below the thermoneutral range exacerbates LPS-induced blood-brain barrier injury and neuroinflammation. PI3Kγ serves a protective role in suppressing release of MMPs, maintaining microglial motility and reinforcing phagocytosis leading to improved brain tissue integrity. Thus, preclinical research targeting severe brain inflammation responses is seriously biased when basic physiological prerequisites of mammal species such as preferred ambient temperature are ignored.

Keywords: Ambient temperature; Blood–brain barrier; Matrix metalloproteinases; Microglia; Migration; Neuroinflammation; Phagocytosis; Phosphoinositide 3-kinase γ.

MeSH terms

  • Animals
  • Animals, Newborn
  • Blood-Brain Barrier / drug effects
  • Blood-Brain Barrier / enzymology
  • Blood-Brain Barrier / physiopathology
  • Body Temperature / drug effects
  • Body Temperature / physiology
  • Body Temperature Regulation / drug effects
  • Body Temperature Regulation / physiology*
  • Cells, Cultured
  • Class Ib Phosphatidylinositol 3-Kinase / deficiency*
  • Female
  • Lipopolysaccharides / toxicity*
  • Male
  • Mice
  • Mice, Inbred C57BL
  • Mice, Knockout
  • Sepsis-Associated Encephalopathy / chemically induced
  • Sepsis-Associated Encephalopathy / enzymology*
  • Sepsis-Associated Encephalopathy / physiopathology*

Substances

  • Lipopolysaccharides
  • Class Ib Phosphatidylinositol 3-Kinase
  • Pik3cg protein, mouse