Abstract

There is currently no therapeutic drug treatment for traumatic brain injury (TBI) despite decades of experimental clinical trials. This may be because the mechanistic pathways for improving TBI outcomes have yet to be identified and exploited. As such, there remains a need to seek out new molecular targets and their drug candidates to find new treatments for TBI. This review presents supporting evidence for cathepsin B, a cysteine protease, as a potentially important drug target for TBI. Cathepsin B expression is greatly up-regulated in TBI animal models, as well as in trauma patients. Importantly, knockout of the cathepsin B gene in TBI mice results in substantial improvements of TBI-caused deficits in behavior, pathology, and biomarkers, as well as improvements in related injury models. During the process of TBI-induced injury, cathepsin B likely escapes the lysosome, its normal subcellular location, into the cytoplasm or extracellular matrix (ECM) where the unleashed proteolytic power causes destruction via necrotic, apoptotic, autophagic, and activated glia-induced cell death, together with ECM breakdown and inflammation. Significantly, chemical inhibitors of cathepsin B are effective for improving deficits in TBI and related injuries including ischemia, cerebral bleeding, cerebral aneurysm, edema, pain, infection, rheumatoid arthritis, epilepsy, Huntington’s disease, multiple sclerosis, and Alzheimer’s disease. The inhibitor E64d is unique among cathepsin B inhibitors in being the only compound to have demonstrated oral efficacy in a TBI model and prior safe use in man and as such it is an excellent tool compound for preclinical testing and clinical compound development. These data support the conclusion that drug development of cathepsin B inhibitors for TBI treatment should be accelerated.

Highlights

  • Traumatic brain injury (TBI) occurs when an external force, such as that due to a vehicular accident, a football collision, or a bullet, causes brain dysfunction and pathology

  • The E64d-treated cathepsin B knockout animals performed significantly (20%) better in the neuromotor assay than carrier-treated cathepsin B knockout animals on day 1 post-TBI [48]. These results suggest that E64d produced an additional benefit in behavior from inhibiting other proteases in addition to cathepsin B

  • Deleting or inhibiting cathepsin B improves outcomes in injury models related to TBI including epilepsy, aneurysm, ischemia, pain, surgical trauma, spinal cord trauma, infectious disease, and neurodegeneration

Read more

Summary

Introduction

Traumatic brain injury (TBI) occurs when an external force, such as that due to a vehicular accident, a football collision, or a bullet, causes brain dysfunction and pathology. While it has been suggested that autophagy might provide neuroprotection after TBI [239], administration of the autophagy inhibitor 3-methyladenine (3-MA) to TBI animal models reduced the TBI-increased LC3II and Beclin 1 levels and restored the reduced P62 levels that resulted from trauma, while improving memory and neuromotor defects and reducing brain lesion volume, neuronal cell death, cathepsin B activity, and caspase 3 activity [241]. Cathepsin B inhibitors inhibit lysosomal cysteine proteases, which reduces autophagy function [246] and thereby may improve TBI outcomes.

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