Cerebral vascular and blood brain‐barrier abnormalities in a mouse model of epilepsy and tuberous sclerosis complex

Abstract

Objective

Tuberous sclerosis complex (TSC) is a genetic disorder, characterized by tumor formation in the brain and other organs, and severe neurological symptoms, such as epilepsy. Abnormal vascular endothelial growth factor (VEGF) expression may promote angiogenesis in kidney and lung tumors in TSC and has been identified in brain specimens from TSC patients, but the role of VEGF and vascular abnormalities in neurological manifestations of TSC is poorly defined. In this study, we investigated abnormalities in brain VEGF expression, cerebral blood vessel anatomy, and blood-brain barrier (BBB) structure and function in a mouse model of TSC.

Methods

Tsc1
GFAPCKO mice were used to investigate VEGF expression and vascular abnormalities in the brain by western blotting and immunohistochemical analysis of vascular and BBB markers. In vivo two-photon imaging was used to assess BBB permeability to normally impenetrable fluorescently-labeled compounds. The effect of mechanistic target of rapamycin (mTOR) pathway inhibitors, VEGF receptor antagonists (apatinib), or BBB stabilizers (RepSox) were assessed in some of these assays, as well as on seizures by video-EEG.

Results

VEGF expression was elevated in cortex of Tsc1
GFAPCKO mice, which was reversed by the mTOR inhibitor, rapamycin. Tsc1
GFAPCKO mice exhibited increased cerebral angiogenesis and vascular complexity in cortex and hippocampus, which were reversed by the VEGF receptor antagonist, apatinib. BBB permeability was abnormally increased and BBB-related tight junction proteins, occludin and claudin-5, were decreased in Tsc1
GFAPCKO mice, also in an apatinib- and RepSox-dependent manner. The BBB stabilizer (RepSox), but not the VEGF receptor antagonist (apatinib), decreased seizures and improved survival in Tsc1
GFAPCKO mice.

Significance

Increased brain VEGF expression is dependent on mTOR pathway activation and promotes cerebral vascular abnormalities and increased BBB permeability in a mouse model of TSC. BBB modulation may affect epileptogenesis and represent a rationale treatment for epilepsy in TSC.

0