Abstract

The endocannabinoid CB₂ receptor system has been implicated in the neuropathology of Alzheimer's disease (AD). In order to investigate the impact of the CB₂ receptor system on AD pathology, a colony of mice with a deleted CB₂ receptor gene, CNR2, was established on a transgenic human mutant APP background for pathological comparison with CB₂ receptor-sufficient transgenic mice. J20 APP (PDGFB-APPSwInd) mice were bred over two generations with CNR2⁻/⁻ (Cnr2(tm1Dgen)/J) mice to produce a colony of J20 CNR2⁺/⁺ and J20 CNR2⁻/⁻ mice. Seventeen J20 CNR2⁺/⁺ mice (12 females, 5 males) and 16 J20 CNR2⁻/⁻ mice (11 females, 5 males) were killed at 12 months, and their brains were interrogated for AD-related pathology with both biochemistry and immunocytochemistry (ICC). In addition to amyloid-dependent endpoints such as soluble Aβ production and plaque deposition quantified with 6E10 staining, the effect of CB2 receptor deletion on total soluble mouse tau production was assayed by using a recently developed high-sensitivity assay. Results revealed that soluble Aβ42 and plaque deposition were significantly increased in J20 CNR2⁻/⁻ mice relative to CNR2⁺/⁺ mice. Microgliosis, quantified with ionized calcium-binding adapter molecule 1 (Iba-1) staining, did not differ between groups, whereas plaque associated microglia was more abundant in J20 CNR2⁻/⁻ mice. Total tau was significantly suppressed in J20 CNR2⁻/⁻ mice relative to J20 CNR2⁺/⁺ mice. The results confirm the constitutive role of the CB₂ receptor system both in reducing amyloid plaque pathology in AD and also support tehpotential of cannabinoid therapies targeting CB₂ to reduce Aβ; however, the results suggest that interventions may have a divergent effect on tau pathology.

Highlights

  • The primary neuropathological hallmarks of Alzheimer’s disease (AD) comprise amyloid-β (Aβ) deposits and neurofibrillary tangles composed of abnormally phosphorylated tau protein in the brain parenchyma [1]

  • Because there have been reports of amyloid production being influenced by gender in transgenic models [34], the groups were matched for gender

  • When AD brains are compared with controls, both levels of soluble and insoluble Aβ increase, but there is a shift from soluble forms to insoluble forms such that, in AD, insoluble Aβ dominates the total fraction of amyloid in the brain, whereas in healthy controls, soluble forms predominate [36]

Read more

Summary

Introduction

The primary neuropathological hallmarks of Alzheimer’s disease (AD) comprise amyloid-β (Aβ) deposits and neurofibrillary tangles composed of abnormally phosphorylated tau protein in the brain parenchyma [1]. Deposition of Aβ in brain is known to induce microgliosis [2] and attendant cytokine production [3], events believed to be important drivers of the pathogenic cascade [4]. The endocannabinoid system comprises at least two biolipid signaling molecules produced on demand in a wide spectrum of cell types and tissues, in-. Cluding neurons and immune cells of the central nervous system, that bind to at least two cannabinoid receptors, CB1 and CB2 [5]. The CB2 receptor is expressed primarily in immune-competent cells and functions as a feedback inhibitor of inflammatory cytokine production [6]. This study was followed by an investigation of mRNA expression in the blood of subjects with AD in which expression of CB2 RECEPTOR DEFICIENCY IN AN AMYLOID MOUSE MODEL

Objectives
Methods
Results
Discussion
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