Abstract

Enhanced levels of Matrix Metalloproteinase-9 (MMP-9) have been implicated in the pathogenesis of epilepsy in humans and rodents. Lack of Mmp-9 impoverishes, whereas excess of Mmp-9 facilitates epileptogenesis. Epigenetic mechanisms driving the epileptogenesis-related upregulation of MMP-9 expression are virtually unknown. The aim of this study was to reveal these mechanisms. We analyzed hippocampi extracted from adult and pediatric patients with temporal lobe epilepsy as well as from partially and fully pentylenetetrazole kindled rats. We used a unique approach to the analysis of the kindling model results (inclusion in the analysis of rats being during kindling, and not only a group of fully kindled animals), which allowed us to separate the molecular effects exerted by the epileptogenesis from those related to epilepsy and epileptic activity. Consequently, it allowed for a disclosure of molecular mechanisms underlying causes, and not consequences, of epilepsy. Our data show that the epileptogenesis-evoked upregulation of Mmp-9 expression is regulated by removal from Mmp-9 gene proximal promoter of the two, interweaved potent silencing mechanisms–DNA methylation and Polycomb Repressive Complex 2 (PRC2)-related repression. Demethylation depends on a gradual dissociation of the DNA methyltransferases, Dnmt3a and Dnmt3b, and on progressive association of the DNA demethylation promoting protein Gadd45β to Mmp-9 proximal gene promoter in vivo. The PRC2-related mechanism relies on dissociation of the repressive transcription factor YY1 and the dissipation of the PRC2-evoked trimethylation on Lys27 of the histone H3 from the proximal Mmp-9 promoter chromatin in vivo. Moreover, we show that the DNA hydroxymethylation, a new epigenetic DNA modification, which is localized predominantly in the gene promoters and is particularly abundant in the brain, is not involved in a regulation of MMP-9 expression during the epileptogenesis in the rat hippocampus as well as in the hippocampi of pediatric and adult epileptic patients. Additionally, we have also found that despite of its transient nature, the histone modification H3S10ph is strongly and gradually accumulated during epileptogenesis in the cell nuclei and in the proximal Mmp-9 gene promoter in the hippocampus, which suggests that H3S10ph can be involved in DNA demethylation in mammals, and not only in Neurospora. The study identifies MMP-9 as the first protein coding gene which expression is regulated by DNA methylation in human epilepsy. We present a detailed epigenetic model of the epileptogenesis-evoked upregulation of MMP-9 expression in the hippocampus. To our knowledge, it is the most complex and most detailed mechanism of epigenetic regulation of gene expression ever revealed for a particular gene in epileptogenesis. Our results also suggest for the first time that dysregulation of DNA methylation found in epilepsy is a cause rather than a consequence of this condition.

Highlights

  • One of the most prominent pathologic features of epilepsy is aberrant synaptic plasticity [1]

  • To determine whether Matrix Metalloproteinase-9 (MMP-9) is abnormally expressed in patients with the mesial temporal sclerosis, we analyzed MMP-9 mRNA levels using reverse transcription quantitative PCR (RTqPCR) in the samples of hippocampi surgically dissected from adult and pediatric epileptic patients as well as from autopsy controls, who died from disorders unrelated to the nervous system (Table 1)

  • To evaluate the possibility that this phenomenon is involved in the control of MMP-9 mRNA upregulation in human epilepsy, we investigated by methylated DNA immunoprecipitation (MeDIP) whether the proximal MMP-9 promoter was demethylated in epileptic patients

Read more

Summary

Introduction

One of the most prominent pathologic features of epilepsy is aberrant synaptic plasticity [1]. The synaptic plasticity–related protease Matrix Metalloproteinase-9 (MMP-9) [2,3] is an important stimulant for the development of epilepsy in humans and rodents [4,5]. It is upregulated in epilepsy; lack of Mmp-9 impoverishes, whereas excess of Mmp-9 facilitates epileptogenesis [4]. Changes in DNA methylation are strongly involved in physiological and aberrant synaptic plasticity as well as in epilepsy development [6,7]

Objectives
Methods
Results
Conclusion
Full Text
Paper version not known

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