Persistent accumulation of unrepaired DNA damage in rat cortical neurons: nuclear organization and ChIP-seq analysis of damaged DNA

Acta Neuropathol Commun. 2018 Jul 26;6(1):68. doi: 10.1186/s40478-018-0573-6.

Abstract

Neurons are highly vulnerable to DNA damage induced by genotoxic agents such as topoisomerase activity, oxidative stress, ionizing radiation (IR) and chemotherapeutic drugs. To avert the detrimental effects of DNA lesions in genome stability, transcription and apoptosis, neurons activate robust DNA repair mechanisms. However, defective DNA repair with accumulation of unrepaired DNA are at the basis of brain ageing and several neurodegenerative diseases. Understanding the mechanisms by which neurons tolerate DNA damage accumulation as well as defining the genomic regions that are more vulnerable to DNA damage or refractory to DNA repair and therefore constitute potential targets in neurodegenerative diseases are essential issues in the field. In this work we investigated the nuclear topography and organization together with the genome-wide distribution of unrepaired DNA in rat cortical neurons 15 days upon IR. About 5% of non-irradiated and 55% of irradiated cells accumulate unrepaired DNA within persistent DNA damage foci (PDDF) of chromatin. These PDDF are featured by persistent activation of DNA damage/repair signaling, lack of transcription and localization in repressive nuclear microenvironments. Interestingly, the chromatin insulator CTCF is concentrated at the PDDF boundaries, likely contributing to isolate unrepaired DNA from intact transcriptionally active chromatin. By confining damaged DNA, PDDF would help preserving genomic integrity and preventing the production of aberrant proteins encoded by damaged genes.ChIP-seq analysis of genome-wide γH2AX distribution revealed a number of genomic regions enriched in γH2AX signal in IR-treated cortical neurons. Some of these regions are in close proximity to genes encoding essential proteins for neuronal functions and human neurodegenerative disorders such as epm2a (Lafora disease), serpini1 (familial encephalopathy with neuroserpin inclusion bodies) and il1rpl1 (mental retardation, X-linked 21). Persistent γH2AX signal close to those regions suggests that nearby genes could be either more vulnerable to DNA damage or more refractory to DNA repair.

Keywords: DNA damage- ionizing radiation- cortical neurons- persistent DNA damage foci- transcription silencing- CTCF- γH2AX genomic distribution; Neurodegenerative diseases.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Antineoplastic Agents / therapeutic use
  • Calcium-Binding Proteins / metabolism
  • Cell Nucleus / metabolism*
  • Cell Nucleus / radiation effects
  • Cerebral Cortex / cytology
  • Chromatin Immunoprecipitation
  • DNA Damage / radiation effects*
  • DNA Repair / drug effects
  • DNA Repair / radiation effects*
  • Folic Acid / analogs & derivatives
  • Folic Acid / therapeutic use
  • Gene Expression Regulation / radiation effects
  • Glial Fibrillary Acidic Protein / metabolism
  • Histones / metabolism
  • Male
  • Microfilament Proteins / metabolism
  • Microscopy, Electron
  • Neurons / radiation effects*
  • Neurons / ultrastructure
  • Quinazolines / therapeutic use
  • RNA, Messenger / genetics
  • RNA, Messenger / metabolism
  • Rats
  • Rats, Sprague-Dawley
  • Time Factors
  • X-Rays / adverse effects*

Substances

  • Aif1 protein, rat
  • Antineoplastic Agents
  • Calcium-Binding Proteins
  • Glial Fibrillary Acidic Protein
  • Histones
  • Microfilament Proteins
  • Quinazolines
  • RNA, Messenger
  • CB 3717
  • Folic Acid