4.4 Article

Pharmacological or genetic depletion of senescent astrocytes prevents whole brain irradiation-induced impairment of neurovascular coupling responses protecting cognitive function in mice

Journal

GEROSCIENCE
Volume 42, Issue 2, Pages 409-428

Publisher

SPRINGER
DOI: 10.1007/s11357-020-00154-8

Keywords

Senescence; WBI; WBRT; Whole brain radiation therapy; Aging; Vascular cognitive impairment; Functional hyperemia; Radiation; Dementia

Funding

  1. American Heart Association
  2. Oklahoma Center for the Advancement of Science and Technology
  3. National Institute on Aging [R01-AG047879, R01-AG038747, R01-AG055395]
  4. National Institute of Neurological Disorders and Stroke [R01-NS056218, R01-NS100782]
  5. Oklahoma Shared Clinical and Translational Resources (OSCTR) program - National Institute of General Medical Sciences [GM104938]
  6. Presbyterian Health Foundation
  7. NIA [T32AG052363]
  8. Oklahoma Nathan Shock Center [P30AG050911]
  9. Cellular and Molecular GeroScience CoBRE [1P20GM125528, 5337]

Ask authors/readers for more resources

Whole brain irradiation (WBI, also known as whole brain radiation therapy or WBRT) is a mainstream therapy for patients with identifiable brain metastases and as a prophylaxis for microscopic malignancies. WBI accelerates brain aging, causing progressive cognitive dysfunction in similar to 50% of surviving patients, thus compromising quality of life. The mechanisms responsible for this WBI side effect remain obscure, and there are no effective treatments or prevention strategies. Here, we test the hypothesis that WBI induces astrocyte senescence, which contributes to impaired astrocytic neurovascular coupling (NVC) responses and the genesis of cognitive decline. To achieve this goal, we used transgenic p16-3MR mice, which allows the detection and selective elimination of senescent cells. We subjected these mice to a clinically relevant protocol of fractionated WBI (5 Gy twice weekly for 4 weeks). WBI-treated and control mice were tested for spatial memory performance (radial arm water maze), astrocyte-dependent NVC responses (whisker-stimulation-induced increases in cerebral blood flow, assessed by laser speckle contrast imaging), NVC-related gene expression, astrocytic release of eicosanoid gliotransmitters and the presence of senescent astrocytes (by flow cytometry, immunohistochemistry and gene expression profiling) at 6 months post-irradiation. WBI induced senescence in astrocytes, which associated with NVC dysfunction and impaired performance on cognitive tasks. To establish a causal relationship between WBI-induced senescence and NVC dysfunction, senescent cells were depleted from WBI-treated animals (at 3 months post-WBI) by genetic (ganciclovir treatment) or pharmacological (treatment with the BCL-2/BCL-xL inhibitor ABT263/Navitoclax, a known senolytic drug) means. In WBI-treated mice, both treatments effectively eliminated senescent astrocytes, rescued NVC responses, and improved cognitive performance. Our findings suggest that the use of senolytic drugs can be a promising strategy for preventing the cognitive impairment associated with WBI.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.4
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available