Abstract

Toll-like receptor (TLR)3 activation during the neonatal period produces responses linked to the origins of neuropsychiatric disorders. Although there is sexual dimorphism in neuropsychiatric disorders, it is unknown if brain responses to TLR3 activation are sex-specific. We hypothesized that poly I:C in a post-natal day (P)8 model induces a sexually dimorphic inflammatory responses. C57BL6 mice received intraperitoneal injection of poly I:C (10 mg/kg) or vehicle [normal saline (NS)] at P8. Pups were killed at 6 or 14 h for caspase 3 and 8 activity assays, NFkB ELISA, IRF3, AP1, and GFAP western blotting and cytokines/chemokines gene expression real time qRT-PCR (4–6/group). A second group of pups were killed at 24 h (P9) or 7 days (P15) after poly I:C to assess astrocytic (GFAP) and microglia (Iba1) activation in the hippocampus, thalamus and cortex using immunohistochemistry, and gene and protein expression of cytokines/chemokines using real time RT-PCR and MSD, respectively (4–6/group). Non-parametric analysis was applied. Six hours after poly I:C, caspase-3 and -8 activities in cytosolic fractions were 1.6 and 2.8-fold higher in poly I:C-treated than in NS-treated female mice, respectively, while gene expressions of pro-inflammatory cytokines were upregulated in both sexes. After poly I:C, IRF3 nuclear translocation occurred earlier (6 h) in female mice and later (14 h) in male mice. At 14 h after poly I:C, only male mice also had increased nuclear NFκB levels (88%, p < 0.001) and GFAP expression coinciding with persistent IL-6 and FAS gene upregulation (110 and 77%, respectively; p < 0.001) and IL-10 gene downregulation (-42%, p < 0.05). At 24 h after poly I:C, IL-1β, CXCL-10, TNF-α, and MCP-1 were similarly increased in both sexes but at 7 days after exposure, CXCL-10 and INFγ were increased and IL-10 was decreased only in female mice. Accordingly, microglial activation persisted at 7 days after poly I:C in the hippocampus, thalamus and cortex of female mice. This preliminary study suggests that TLR3 activation may produce in the developing neonatal mouse brain a sexually dimorphic response with early activation of caspase-dependent pathways in female mice, activation of inflammatory cascades in both sexes, which then persists in female mice. Further well-powered studies are essential to confirm these sex-specific findings.

Highlights

  • Toll-like receptors (TLRs) are a family of innate immune system receptors that react to both microbial stimulation and to molecules released upon tissue injury

  • Our pilot results suggest that TLR3 activation affects the developing brain differently in male and female mice

  • IRF-3 nuclear translocation downstream of TLR3 activation leads to INF-β upregulation 6 h after polycytidylic acid (poly I):C in female mice, while delayed nuclear factor κB (NFκB)-IRF-3 nuclear translocation leads to INF-β upregulation 14 h after exposure in male mice

Read more

Summary

INTRODUCTION

Toll-like receptors (TLRs) are a family of innate immune system receptors that react to both microbial stimulation and to molecules released upon tissue injury. Following TLR3 activation, neuronal death may proceed via: (i) apoptosis with release of apoptotic bodies containing DNA fragments, or (ii) necrosis (or necrosis-like) with release of free suspended dead cell materials rich in DAMPs (Blasius and Beutler, 2010). In the injured developing brain, activated astrocytes and microglia engulf DAMPs forming intracellular vesicles, which fuse with TLR3-cointaing acidic endosomes (Rossi and Volterra, 2009; Ransohoff and Brown, 2012; Wakida et al, 2018). Persistent ER stress may modulate the response to many intracellular TLR ligands, including the TLR3 ligand polyinosine-polycytidylic acid (poly I:C) (Randow and Seed, 2001; Tabeta et al, 2006; Yang et al, 2007). We studied the potential role of ER stress in sex-specific responses to TLR3 activation

MATERIALS AND METHODS
RESULTS
DISCUSSION
ETHICS STATEMENT
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.