Skip to main content

Lund University Publications

LUND UNIVERSITY LIBRARIES

Transcriptional reactivation of OTX2, RX1 and SIX3 during reprogramming contributes to the generation of RPE cells from human iPSCs

Li, Peng ; Sun, Xiaofeng ; Ma, Zhizhong ; Liu, Yinan ; Jin, Ying ; Ge, Ruimin LU ; Hao, Limin ; Ma, Yanling ; Han, Shuo and Sun, Haojie , et al. (2016) In International Journal of Biological Sciences 12(5). p.505-517
Abstract

Directed differentiation of human induced pluripotent stem cells (iPSCs) into retinal pigmented epithelium (RPE) holds great promise in cell replacement therapy for patients suffering from degenerative eye diseases, including age-related macular degeneration (AMD). In this study, we generated iPSCs from human dermal fibroblasts (HDFs) by electroporation with episomal plasmid vectors encoding OCT4, SOX2, KLF4, L-MYC together with p53 suppression. Intriguingly, cell reprogramming resulted in a metastable transcriptional activation and selective demethylation of neural and retinal specification-associated genes, such as OTX2, RX1 and SIX3. In contrast, RPE progenitor genes were transcriptionally silent in HDFs and descendant iPSCs.... (More)

Directed differentiation of human induced pluripotent stem cells (iPSCs) into retinal pigmented epithelium (RPE) holds great promise in cell replacement therapy for patients suffering from degenerative eye diseases, including age-related macular degeneration (AMD). In this study, we generated iPSCs from human dermal fibroblasts (HDFs) by electroporation with episomal plasmid vectors encoding OCT4, SOX2, KLF4, L-MYC together with p53 suppression. Intriguingly, cell reprogramming resulted in a metastable transcriptional activation and selective demethylation of neural and retinal specification-associated genes, such as OTX2, RX1 and SIX3. In contrast, RPE progenitor genes were transcriptionally silent in HDFs and descendant iPSCs. Overexpression of OCT4 and SOX2 directly stimulated the expression of OTX2, RX1 and SIX3 in HDFs and iPSCs. Luciferase and chromatin immunoprecipitation (ChIP) assays further identified an OCT4- and two SOX2-binding sites located in the proximal promoter of OTX2. Histone acetylation and methylation on the local promoter also participated in the reactivation of OTX2. The transcriptional conversion of RX1 and SIX3 genes partially attributed to DNA demethylation. Subsequently, iPSCs were induced into the RPE cells displaying the characteristics of polygonal shapes and pigments, and expressing typical RPE cell markers. Taken together, our results establish readily efficient and safe protocols to produce iPSCs and iPSC-derived RPE cells, and underline that the reactivation of anterior neural transcription factor OTX2, eye field transcription factor RX1 and SIX3 in iPSCs is a feature of pluripotency acquisition and predetermines the potential of RPE differentiation.

(Less)
Please use this url to cite or link to this publication:
author
; ; ; ; ; ; ; ; and , et al. (More)
; ; ; ; ; ; ; ; ; ; ; ; and (Less)
organization
publishing date
type
Contribution to journal
publication status
published
subject
keywords
Epigenetic modification, Induced pluripotent stem cells, Reprogramming, Retinal pigment epithelium
in
International Journal of Biological Sciences
volume
12
issue
5
pages
13 pages
publisher
Ivyspring International Publisher
external identifiers
  • scopus:84960444340
  • pmid:27019633
  • wos:000371709100003
ISSN
1449-2288
DOI
10.7150/ijbs.14212
language
English
LU publication?
yes
id
4417d02f-dfb7-45cc-b9bd-e80deebf4aba
date added to LUP
2016-07-14 13:34:56
date last changed
2024-01-19 06:51:51
@article{4417d02f-dfb7-45cc-b9bd-e80deebf4aba,
  abstract     = {{<p>Directed differentiation of human induced pluripotent stem cells (iPSCs) into retinal pigmented epithelium (RPE) holds great promise in cell replacement therapy for patients suffering from degenerative eye diseases, including age-related macular degeneration (AMD). In this study, we generated iPSCs from human dermal fibroblasts (HDFs) by electroporation with episomal plasmid vectors encoding OCT4, SOX2, KLF4, L-MYC together with p53 suppression. Intriguingly, cell reprogramming resulted in a metastable transcriptional activation and selective demethylation of neural and retinal specification-associated genes, such as OTX2, RX1 and SIX3. In contrast, RPE progenitor genes were transcriptionally silent in HDFs and descendant iPSCs. Overexpression of OCT4 and SOX2 directly stimulated the expression of OTX2, RX1 and SIX3 in HDFs and iPSCs. Luciferase and chromatin immunoprecipitation (ChIP) assays further identified an OCT4- and two SOX2-binding sites located in the proximal promoter of OTX2. Histone acetylation and methylation on the local promoter also participated in the reactivation of OTX2. The transcriptional conversion of RX1 and SIX3 genes partially attributed to DNA demethylation. Subsequently, iPSCs were induced into the RPE cells displaying the characteristics of polygonal shapes and pigments, and expressing typical RPE cell markers. Taken together, our results establish readily efficient and safe protocols to produce iPSCs and iPSC-derived RPE cells, and underline that the reactivation of anterior neural transcription factor OTX2, eye field transcription factor RX1 and SIX3 in iPSCs is a feature of pluripotency acquisition and predetermines the potential of RPE differentiation.</p>}},
  author       = {{Li, Peng and Sun, Xiaofeng and Ma, Zhizhong and Liu, Yinan and Jin, Ying and Ge, Ruimin and Hao, Limin and Ma, Yanling and Han, Shuo and Sun, Haojie and Zhang, Mingzhi and Li, Ruizhi and Li, Tao and Shen, Li}},
  issn         = {{1449-2288}},
  keywords     = {{Epigenetic modification; Induced pluripotent stem cells; Reprogramming; Retinal pigment epithelium}},
  language     = {{eng}},
  month        = {{02}},
  number       = {{5}},
  pages        = {{505--517}},
  publisher    = {{Ivyspring International Publisher}},
  series       = {{International Journal of Biological Sciences}},
  title        = {{Transcriptional reactivation of OTX2, RX1 and SIX3 during reprogramming contributes to the generation of RPE cells from human iPSCs}},
  url          = {{http://dx.doi.org/10.7150/ijbs.14212}},
  doi          = {{10.7150/ijbs.14212}},
  volume       = {{12}},
  year         = {{2016}},
}