Your browser doesn't support javascript.
loading
Gelatinase Biosensor Reports Cellular Remodeling During Epileptogenesis.
Bouquier, Nathalie; Girard, Benoit; Aparicio Arias, Juri; Fagni, Laurent; Bertaso, Federica; Perroy, Julie.
Afiliação
  • Bouquier N; IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France.
  • Girard B; IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France.
  • Aparicio Arias J; IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France.
  • Fagni L; IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France.
  • Bertaso F; IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France.
  • Perroy J; IGF, Université de Montpellier, CNRS, INSERM, Montpellier, France.
Article em En | MEDLINE | ID: mdl-32372941
ABSTRACT
Epileptogenesis is the gradual process responsible for converting a healthy brain into an epileptic brain. This process can be triggered by a wide range of factors, including brain injury or tumors, infections, and status epilepticus. Epileptogenesis results in aberrant synaptic plasticity, neuroinflammation and seizure-induced cell death. As Matrix Metalloproteinases (MMPs) play a crucial role in cellular plasticity by remodeling the extracellular matrix (ECM), gelatinases (MMP-2 and MMP-9) were recently highlighted as key players in epileptogenesis. In this work, we engineered a biosensor to report in situ gelatinase activity in a model of epileptogenesis. This biosensor encompasses a gelatinase-sensitive activatable cell penetrating peptide (ACPP) coupled to a TAMRA fluorophore, allowing fluorescence uptake in cells displaying endogenous gelatinase activities. In a preclinical mouse model of temporal lobe epilepsy (TLE), the intrahippocampal kainate injection, ACPPs revealed a localized distribution of gelatinase activities, refining temporal cellular changes during epileptogenesis. The activity was found particularly but not only in the ipsilateral hippocampus, starting from the CA1 area and spreading to dentate gyrus from the early stages throughout chronic epilepsy, notably in neurons and microglial cells. Thus, our work shows that ACPPs are suitable molecular imaging probes for detecting the spatiotemporal pattern of gelatinase activity during epileptogenesis, suggesting their possible use as vectors to target cellular reactive changes with treatment for epileptogenesis.
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Prognostic_studies Idioma: En Revista: Front Synaptic Neurosci Ano de publicação: 2020 Tipo de documento: Article País de afiliação: França

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Tipo de estudo: Prognostic_studies Idioma: En Revista: Front Synaptic Neurosci Ano de publicação: 2020 Tipo de documento: Article País de afiliação: França