Abstract

Primary blast neurotrauma represents a unique injury paradigm characterized by high-rate overpressure effects on brain tissue. One major hallmark of blast neurotrauma is glial reactivity, notably prolonged astrocyte activation. This cellular response has been mainly defined in primary blast neurotrauma by increased intermediate filament expression. Because the intermediate filament networks physically interface with transmembrane proteins for junctional support, it was hypothesized that cell junction regulation is altered in the reactive phenotype as well. This would have implications for downstream transcriptional regulation via signal transduction pathways like nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Therefore, a custom high-rate overpressure simulator was built for in vitro testing using mechanical conditions based on intracranial pressure measurements in a rat model of blast neurotrauma. Primary rat astrocytes were exposed to isolated high-rate mechanical stimulation to study cell junction dynamics in relation to their mechano-activation. First, a time course for “classical” features of reactivity was devised by evaluation of glial fibrillary acidic protein (GFAP) and proliferating cell nuclear antigen (PCNA) expression. This was followed by gene and protein expression for both gap junction (connexins) and anchoring junction proteins (integrins and cadherins). Signal transduction analysis was carried out by nuclear localization of two molecules, NF-κB p65 and mitogen-activated protein kinase (MAPK) p38. Results indicated significant increases in connexin-43 expression and PCNA first at 24 h post-overpressure (p < 0.05), followed by structural reactivity (via increased GFAP, p < 0.05) corresponding to increased anchoring junction dynamics at 48 h post-overpressure (p < 0.05). Moreover, increased phosphorylation of focal adhesion kinase (FAK) was observed in addition to increased nuclear localization of both p65 and p38 (p < 0.05) during the period of structural reactivity. To evaluate the transcriptional activity of p65 in the nucleus, electrophoretic mobility shift assay was conducted for a binding site on the promoter region for intracellular adhesion molecule-1 (ICAM-1), an antagonist of tight junctions. A significant increase in the interaction of nuclear proteins with the NF-κB site on the ICAM-1 corresponded to increased gene and protein expression of ICAM-1 (p < 0.05). Altogether, these results indicate multiple targets and corresponding signaling pathways which involve cell junction dynamics in the mechano-activation of astrocytes following high-rate overpressure.

Highlights

  • Traumatic brain injury (TBI) has proven challenging to treat clinically because of the disparity amongst injury modes and severities

  • It should be noted that despite increased proliferating cell nuclear antigen (PCNA), there were no detectable changes in MTT metabolism until 48 h at which point there was a significant increase in enzymatic activity (p = 0.0005; Figure 4) Otherwise, glial fibrillary acidic protein (GFAP) expression had a delayed increase at 48 h post high-rate overpressure exposure

  • Astrocytes have become an important focus in neurotrauma research because of their ability to influence many of these aspects of secondary injury sequelae [29, 64, 65]

Read more

Summary

Introduction

Traumatic brain injury (TBI) has proven challenging to treat clinically because of the disparity amongst injury modes and severities. There is a growing appreciation for the need to better characterize and understand how brain cells, individually and collectively, respond to mechanical damage. It is especially critical to understand the potential differences in cellular and molecular hallmarks of TBI within the context of the mechanical injury itself. The toxic environment associated with acute secondary injury acts as the initiator for prolonged neural cell dysfunction and cognitive deficits. This cascade of early cellular events offers the greatest promise for therapeutic intervention but is highly complex, interdependent, and heterogeneous across TBI modes [1,2,3,4,5]

Objectives
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