Abstract

Preferential neuronal vulnerability is characteristic of several neurodegenerative diseases including the motor neuron disease amyotrophic lateral sclerosis (ALS). It is well established that glia play a critical role in ALS, but it is unknown whether regional differences in the ability of glia to support motor neurons contribute to the specific pattern of neuronal degeneration. In this study, using primary mixed glial cultures from different mouse CNS regions (spinal cord and cortex), we examined whether regional differences exist in key glial pathways that contribute to, or protect against, motor neuron degeneration. Specifically, we examined the NF-κB-mediated inflammatory pathway and the cytoprotective heat shock response (HSR). Glial cultures were treated with pro-inflammatory stimuli, tumour necrosis factor-ɑ/lipopolysaccharide or heat stressed to stimulate the inflammatory and HSR respectively. We found that spinal cord glia expressed more iNOS and produced more NO compared to cortical glia in response to inflammatory stimuli. Intriguingly, we found that expression of ALS-causing SOD1G93A did not elevate the levels of NO in spinal cord glia. However, activation of the stress-responsive HSR was attenuated in SOD1G93A cultures, with a reduced Hsp70 induction in response to stressful stimuli. Exposure of spinal cord glia to heat shock in combination with inflammatory stimuli reduced the activation of the inflammatory response. The results of this study suggest that impaired heat shock response in SOD1G93A glia may contribute to the exacerbated inflammatory reactions observed in ALS mice.Graphical abstractMixed primary glial cultures were established from cortical and spinal cord regions of wild-type mice and mice expressing ALS–causing mutant human SOD1 and the inflammatory and heat shock responses were investigated in these cultures. In the absence of stress, all cultures appeared to have similar cellular composition, levels of inflammatory mediators and similar expression level of heat shock proteins. When stimulated, spinal cord glia were more reactive and activated the inflammatory pathway more readily than cortical glia; this response was similar in wild-type and SOD1G93A glial cultures. Although the heat shock response was similar in spinal cord and cortical glial, in SOD1G93A expressing glia from both the spinal cord and cortex, the induction of heat shock response was diminished. This impaired heat shock response in SOD1G93A glia may therefore contribute to the exacerbated inflammatory reactions observed in ALS mice.

Highlights

  • The stress-induced responses of glia from different regions of the CNS were examined in vitro in mixed glial cultures established from the spinal cord and cortex firstly of neonatal wild-type (WT) mice and, subsequently, in glia derived from SOD1G93A mice that model amyotrophic lateral sclerosis (ALS)

  • FACS analysis of cells double labelled with GFAP and CD11b demonstrated that there were no significant differences between the proportion of astroglia (~ 80%) and microglia (~ 10–20%) in cortical and spinal cord cultures (Fig. 1d, e)

  • The results of this study show that spinal cord–derived glia are intrinsically more prone to activation of the nuclear factor kappa B (NF-κB)-mediated inflammatory pathway and inducible nitric oxide synthase (iNOS) induction than glia derived from the cortex, which may contribute to the selective vulnerability of spinal cord motor neurons in SOD1G93A ALS mice

Read more

Summary

Introduction

We compared the stress responses of primary cortical and spinal cord mixed glial cultures obtained from neonatal mice in response to exposure to heat shock and inflammatory stimuli This model enables the study of the complex interactions between different glial populations, providing a closer in vitro representation of the cortex or spinal cord than pure astroglial or microglial cultures. We explored whether regional differences between cortical and spinal cord glia are altered in cultures obtained from mice expressing mutant SOD1G93A, a model of mutant SOD1–induced ALS in which inflammatory and neuronal damage occurs to a greater extent in the spinal cord than in the cortex. The results of our study provide evidence for the regional specificity of glial stress responses and suggest that impairments in the HSR of SOD1G93A glia might contribute to the excessive neuroinflammation that is known to play a role in motor neuron degeneration in ALS

Results
Discussion
Conclusions
Experimental procedures
Compliance with ethical standards
Full Text
Published version (Free)

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