Abstract

Impaired glucocorticoid (GC) signaling is a significant factor in aging, stress, and neurodegenerative diseases such as Alzheimer’s disease. Therefore, the study of GC-mediated stress responses to chronic moderately stressful situations, which occur in daily life, is of huge interest for the design of pharmacological strategies toward the prevention of neurodegeneration. To address this issue, SAMP8 mice were exposed to the chronic mild stress (CMS) paradigm for 4 weeks and treated with RL-118, an 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) inhibitor. The inhibition of this enzyme is linked with a reduction in GC levels and cognitive improvement, while CMS exposure has been associated with reduced cognitive performance. The aim of this project was to assess whether RL-118 treatment could reverse the deleterious effects of CMS on cognition and behavioral abilities and to evaluate the molecular mechanisms that compromise healthy aging in SAMP8 mice. First, we confirmed the target engagement between RL-118 and 11β-HSD1. Additionally, we showed that DNA methylation, hydroxymethylation, and histone phosphorylation were decreased by CMS induction, and increased by RL-118 treatment. In addition, CMS exposure caused the accumulation of reactive oxygen species (ROS)-induced damage and increased pro-oxidant enzymes—as well as pro-inflammatory mediators—through the NF-κB pathway and astrogliosis markers, such as GFAP. Of note, these modifications were reversed by 11β-HSD1 inhibition. Remarkably, although CMS altered mTORC1 signaling, autophagy was increased in the SAMP8 RL-118-treated mice. We also showed an increase in amyloidogenic processes and a decrease in synaptic plasticity and neuronal remodeling markers in mice under CMS, which were consequently modified by RL-118 treatment. In conclusion, 11β-HSD1 inhibition through RL-118 ameliorated the detrimental effects induced by CMS, including epigenetic and cognitive disturbances, indicating that GC-excess attenuation shows potential as a therapeutic strategy for age-related cognitive decline and AD.

Highlights

  • The RL-118 target engagement was determined in the TAPS

  • RL-118 Demonstrates Target Engagement with 11β-HSD1 Enzyme in the TAPS Assay quantified was relative to the number of cells and was found to be higher in cells e

  • The in Cr ally, peakfollowing area was significantly higher in 11β-HSD1-positive cells than peak area was significantly higher in 11β-HSD1-positive cells than in Crimson-positive positive cells, indicating that the RL-118 drug is selective for 11β-HSD1

Read more

Summary

Introduction

Stressful situations activate a neuroendocrine response, which leads to the release of Pharmaceuticals 2021, 14, 1040. The deleterious effects of stress and GCs in the brain can be transient, since stress-induced hippocampal atrophy and hippocampal-dependent behavior may be reversed after a stress-free period. A growing body of evidence suggests that high GC exposure in early life can adversely program the release of GCs and increase susceptibility to the development of metabolic, neuropsychiatric, and neurodegenerative diseases [8], as well as induce changes in brain structure—including the generation and loss of neurons and dendritic atrophy—and brain function, affecting electrophysiological activity and cellular signaling [9,10]

Objectives
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