Abstract

Astrocytes have emerged as critical elements for the maintenance and function of the central nervous system. The expression on their cell membrane of RAGE and TLR4 receptors makes astrocytes susceptible to High-mobility group box 1 (HMGB1), a nuclear protein typically released in the extracellular milieu by living cells experiencing physiological stress conditions or by damaged cells. Here, we show that the interaction of HMGB1 with normal spinal cord astrocytes induces the astrocytic production of neurotrophic factors, particularly brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF). Multiple investigations suggest a role for HMGB1 in amyotrophic lateral sclerosis (ALS). Yet, no mechanistic information on the implication of HMGB1 signaling in this disorder is currently available. We demonstrate that non-transgenic and transgenic SOD1WT spinal motor neurons exhibit only a basal nucleus-to-cytoplasm shuttling of the HMGB1 protein. Conversely, in SOD1G93A ALS mouse spinal cords, HMGB1 significantly translocates from the nucleus to the cytoplasm of motor neurons, thereby suggesting that it may be eventually released in the extracellular environment during the progression of the disease. We postulate that extracellular HMGB1 can paracrinally interact with the neighboring astrocytes in an attempt to counteract the neurodegenerative process. Yet, at variance with normal cells, SOD1G93A-expressing astrocytes show impaired capacity to raise BDNF and GDNF levels upon HMGB1 stimulation. Our data suggest that HMGB1 have a potential to promote neuroprotective actions by healthy astrocytes. However, this neurotrophic response is disrupted in ALS astrocytes. This indicates that diseased astroglial cells may exacerbate motor neuron degeneration in ALS because of the loss of their neurosupportive functions.

Highlights

  • Astrocytes are the most abundant glial cell population in the central nervous system (CNS) and are essential to maintain CNS homeostasis and function

  • To directly test the hypothesis that healthy astrocytes can respond to High-mobility group box 1 (HMGB1) stimulation by increasing their neurotrophic properties, we first treated primary astrocytes from wild-type mouse spinal cord with disulfide-HMGB1 (3 μg/ml, 6 h) and we analyzed the astrocytic expression of brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF) at the messenger RNA and protein level

  • reverse transcription quantitative polymerase chain reaction (RT-qPCR) analysis revealed that receptor for Advanced Glycation End-Products (RAGE) and TLR4 are both implicated in the trophic factor expression, as indicated by the fact that both receptor antagonists significantly suppressed the HMGB1-dependent increment in BDNF and GDNF messenger RNA (mRNA) levels (Figure 1)

Read more

Summary

Introduction

Astrocytes are the most abundant glial cell population in the central nervous system (CNS) and are essential to maintain CNS homeostasis and function. Reduced cysteines render HMGB1 chemoattractant, whereas the formation of an intramolecular disulfide bond enables the partially oxidized HMGB1 isoform, named “disulfide-HMGB1,” a pro-inflammatory cytokinestimulating mediator (e.g., increasing the production of Tumor Necrosis Factor α, TNFα) (Venereau et al, 2012; Yang et al, 2012; reviewed in Vénéreau et al, 2015) Consistent with this latter function, disulfide-HMGB1 has been described to sustain neuroinflammation and related neurodamaging events in various CNS disorders (Fang et al, 2012). In addition to release a plethora of pro-inflammatory mediators, they can secrete a variety of trophic factors These include brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF) (Schaar et al, 1993; Appel et al, 1997; Dougherty et al, 2000; Ikeda et al, 2001; Chen et al, 2006; reviewed in Rossi, 2015; Colombo and Farina, 2016). We found that normal astrocytes, but not astroglial cells from a mouse model of ALS, respond to HMGB1 stimulation by enhancing their production of BDNF and GDNF

Methods
Results
Conclusion
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