Paternal nicotine exposure defines different behavior in subsequent generation via hyper-methylation of mmu-miR-15b

Abstract The neurobehavioral effects of paternal smoking and nicotine use have not been widely reported. In the present study, nicotine exposure induced depression in the paternal generation, but reduced depression and promoted hyperactivity in F1 offspring. While this intergenerational effect was n...

Descripción completa

Guardado en:
Detalles Bibliográficos
Autores principales: Jingbo Dai, Zhaoxia Wang, Wangjie Xu, Meixing Zhang, Zijue Zhu, Xianglong Zhao, Dong Zhang, Dongsheng Nie, Lianyun Wang, Zhongdong Qiao
Formato: article
Lenguaje:EN
Publicado: Nature Portfolio 2017
Materias:
R
Q
Acceso en línea:https://doaj.org/article/283c5422c4ab4114b86d90348d3fd0f8
Etiquetas: Agregar Etiqueta
Sin Etiquetas, Sea el primero en etiquetar este registro!
id oai:doaj.org-article:283c5422c4ab4114b86d90348d3fd0f8
record_format dspace
spelling oai:doaj.org-article:283c5422c4ab4114b86d90348d3fd0f82021-12-02T12:31:50ZPaternal nicotine exposure defines different behavior in subsequent generation via hyper-methylation of mmu-miR-15b10.1038/s41598-017-07920-32045-2322https://doaj.org/article/283c5422c4ab4114b86d90348d3fd0f82017-08-01T00:00:00Zhttps://doi.org/10.1038/s41598-017-07920-3https://doaj.org/toc/2045-2322Abstract The neurobehavioral effects of paternal smoking and nicotine use have not been widely reported. In the present study, nicotine exposure induced depression in the paternal generation, but reduced depression and promoted hyperactivity in F1 offspring. While this intergenerational effect was not passed down to the F2 generation. Further studies revealed that nicotine induced the down-regulation of mmu-miR-15b expression due to hyper-methylation in the CpG island shore region of mmu-miR-15b in both the spermatozoa of F0 mice and the brains of F1 mice. As the target gene of mmu-miR-15b, Wnt4 expression was elevated in the thalamus of F1 mice due to the inheritance of DNA methylation patterns from the paternal generation. Furthermore, the increased expression of Wnt4 elevated the phosphorylation level of its downstream protein GSK-3 through the canonical WNT4 pathway which involved in the behavioral alterations observed in F1 mice. Moreover, in vivo stereotaxic brain injections were used to induce the overexpression of mmu-miR-15b and WNT4 and confirm the neurobehavioral effects in vitro. The behavioral phenotype of the F1 mice resulting from paternal nicotine exposure could be attenuated by viral manipulation of mmu-miR-15b in the thalamus.Jingbo DaiZhaoxia WangWangjie XuMeixing ZhangZijue ZhuXianglong ZhaoDong ZhangDongsheng NieLianyun WangZhongdong QiaoNature PortfolioarticleMedicineRScienceQENScientific Reports, Vol 7, Iss 1, Pp 1-15 (2017)
institution DOAJ
collection DOAJ
language EN
topic Medicine
R
Science
Q
spellingShingle Medicine
R
Science
Q
Jingbo Dai
Zhaoxia Wang
Wangjie Xu
Meixing Zhang
Zijue Zhu
Xianglong Zhao
Dong Zhang
Dongsheng Nie
Lianyun Wang
Zhongdong Qiao
Paternal nicotine exposure defines different behavior in subsequent generation via hyper-methylation of mmu-miR-15b
description Abstract The neurobehavioral effects of paternal smoking and nicotine use have not been widely reported. In the present study, nicotine exposure induced depression in the paternal generation, but reduced depression and promoted hyperactivity in F1 offspring. While this intergenerational effect was not passed down to the F2 generation. Further studies revealed that nicotine induced the down-regulation of mmu-miR-15b expression due to hyper-methylation in the CpG island shore region of mmu-miR-15b in both the spermatozoa of F0 mice and the brains of F1 mice. As the target gene of mmu-miR-15b, Wnt4 expression was elevated in the thalamus of F1 mice due to the inheritance of DNA methylation patterns from the paternal generation. Furthermore, the increased expression of Wnt4 elevated the phosphorylation level of its downstream protein GSK-3 through the canonical WNT4 pathway which involved in the behavioral alterations observed in F1 mice. Moreover, in vivo stereotaxic brain injections were used to induce the overexpression of mmu-miR-15b and WNT4 and confirm the neurobehavioral effects in vitro. The behavioral phenotype of the F1 mice resulting from paternal nicotine exposure could be attenuated by viral manipulation of mmu-miR-15b in the thalamus.
format article
author Jingbo Dai
Zhaoxia Wang
Wangjie Xu
Meixing Zhang
Zijue Zhu
Xianglong Zhao
Dong Zhang
Dongsheng Nie
Lianyun Wang
Zhongdong Qiao
author_facet Jingbo Dai
Zhaoxia Wang
Wangjie Xu
Meixing Zhang
Zijue Zhu
Xianglong Zhao
Dong Zhang
Dongsheng Nie
Lianyun Wang
Zhongdong Qiao
author_sort Jingbo Dai
title Paternal nicotine exposure defines different behavior in subsequent generation via hyper-methylation of mmu-miR-15b
title_short Paternal nicotine exposure defines different behavior in subsequent generation via hyper-methylation of mmu-miR-15b
title_full Paternal nicotine exposure defines different behavior in subsequent generation via hyper-methylation of mmu-miR-15b
title_fullStr Paternal nicotine exposure defines different behavior in subsequent generation via hyper-methylation of mmu-miR-15b
title_full_unstemmed Paternal nicotine exposure defines different behavior in subsequent generation via hyper-methylation of mmu-miR-15b
title_sort paternal nicotine exposure defines different behavior in subsequent generation via hyper-methylation of mmu-mir-15b
publisher Nature Portfolio
publishDate 2017
url https://doaj.org/article/283c5422c4ab4114b86d90348d3fd0f8
work_keys_str_mv AT jingbodai paternalnicotineexposuredefinesdifferentbehaviorinsubsequentgenerationviahypermethylationofmmumir15b
AT zhaoxiawang paternalnicotineexposuredefinesdifferentbehaviorinsubsequentgenerationviahypermethylationofmmumir15b
AT wangjiexu paternalnicotineexposuredefinesdifferentbehaviorinsubsequentgenerationviahypermethylationofmmumir15b
AT meixingzhang paternalnicotineexposuredefinesdifferentbehaviorinsubsequentgenerationviahypermethylationofmmumir15b
AT zijuezhu paternalnicotineexposuredefinesdifferentbehaviorinsubsequentgenerationviahypermethylationofmmumir15b
AT xianglongzhao paternalnicotineexposuredefinesdifferentbehaviorinsubsequentgenerationviahypermethylationofmmumir15b
AT dongzhang paternalnicotineexposuredefinesdifferentbehaviorinsubsequentgenerationviahypermethylationofmmumir15b
AT dongshengnie paternalnicotineexposuredefinesdifferentbehaviorinsubsequentgenerationviahypermethylationofmmumir15b
AT lianyunwang paternalnicotineexposuredefinesdifferentbehaviorinsubsequentgenerationviahypermethylationofmmumir15b
AT zhongdongqiao paternalnicotineexposuredefinesdifferentbehaviorinsubsequentgenerationviahypermethylationofmmumir15b
_version_ 1718394265457917952