Your browser doesn't support javascript.
loading
Chromatin accessibility and H3K9me3 landscapes reveal long-term epigenetic effects of fetal-neonatal iron deficiency in rat hippocampus.
Liu, Shirelle X; Ramakrishnan, Aarthi; Shen, Li; Gewirtz, Jonathan C; Georgieff, Michael K; Tran, Phu V.
Afiliação
  • Liu SX; Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA.
  • Ramakrishnan A; Department of Psychology, University of Minnesota, Minneapolis, MN, 55455, USA.
  • Shen L; Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
  • Gewirtz JC; Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
  • Georgieff MK; Department of Psychology, University of Minnesota, Minneapolis, MN, 55455, USA.
  • Tran PV; Department of Pediatrics, University of Minnesota, Minneapolis, MN, 55455, USA.
BMC Genomics ; 25(1): 301, 2024 Mar 21.
Article em En | MEDLINE | ID: mdl-38515015
ABSTRACT

BACKGROUND:

Iron deficiency (ID) during the fetal-neonatal period results in long-term neurodevelopmental impairments associated with pervasive hippocampal gene dysregulation. Prenatal choline supplementation partially normalizes these effects, suggesting an interaction between iron and choline in hippocampal transcriptome regulation. To understand the regulatory mechanisms, we investigated epigenetic marks of genes with altered chromatin accessibility (ATAC-seq) or poised to be repressed (H3K9me3 ChIP-seq) in iron-repleted adult rats having experienced fetal-neonatal ID exposure with or without prenatal choline supplementation.

RESULTS:

Fetal-neonatal ID was induced by limiting maternal iron intake from gestational day (G) 2 through postnatal day (P) 7. Half of the pregnant dams were given supplemental choline (5.0 g/kg) from G11-18. This resulted in 4 groups at P65 (Iron-sufficient [IS], Formerly Iron-deficient [FID], IS with choline [ISch], and FID with choline [FIDch]). Hippocampi were collected from P65 iron-repleted male offspring and analyzed for chromatin accessibility and H3K9me3 enrichment. 22% and 24% of differentially transcribed genes in FID- and FIDch-groups, respectively, exhibited significant differences in chromatin accessibility, whereas 1.7% and 13% exhibited significant differences in H3K9me3 enrichment. These changes mapped onto gene networks regulating synaptic plasticity, neuroinflammation, and reward circuits. Motif analysis of differentially modified genomic sites revealed significantly stronger choline effects than early-life ID and identified multiple epigenetically modified transcription factor binding sites.

CONCLUSIONS:

This study reveals genome-wide, stable epigenetic changes and epigenetically modifiable gene networks associated with specific chromatin marks in the hippocampus, and lays a foundation to further elucidate iron-dependent epigenetic mechanisms that underlie the long-term effects of fetal-neonatal ID, choline, and their interactions.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Deficiências de Ferro / Ferro Limite: Animals / Pregnancy Idioma: En Revista: BMC Genomics Assunto da revista: GENETICA Ano de publicação: 2024 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Deficiências de Ferro / Ferro Limite: Animals / Pregnancy Idioma: En Revista: BMC Genomics Assunto da revista: GENETICA Ano de publicação: 2024 Tipo de documento: Article País de afiliação: Estados Unidos