Cargando…

CRISPR/dCas9‐mediated activation of multiple endogenous target genes directly converts human foreskin fibroblasts into Leydig‐like cells

Recently, Leydig cell (LC) transplantation has been revealed as a promising strategy for treating male hypogonadism; however, the key problem restricting the application of LC transplantation is a severe lack of seed cells. It seems that targeted activation of endogenous genes may provide a potentia...

Descripción completa

Detalles Bibliográficos
Autores principales: Huang, Hua, Zou, Xiangyu, Zhong, Liang, Hou, Yanping, Zhou, Jin, Zhang, Zhiyuan, Xing, Xiaoyu, Sun, Jie
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6714237/
https://www.ncbi.nlm.nih.gov/pubmed/31264792
http://dx.doi.org/10.1111/jcmm.14470
_version_ 1783447021669056512
author Huang, Hua
Zou, Xiangyu
Zhong, Liang
Hou, Yanping
Zhou, Jin
Zhang, Zhiyuan
Xing, Xiaoyu
Sun, Jie
author_facet Huang, Hua
Zou, Xiangyu
Zhong, Liang
Hou, Yanping
Zhou, Jin
Zhang, Zhiyuan
Xing, Xiaoyu
Sun, Jie
author_sort Huang, Hua
collection PubMed
description Recently, Leydig cell (LC) transplantation has been revealed as a promising strategy for treating male hypogonadism; however, the key problem restricting the application of LC transplantation is a severe lack of seed cells. It seems that targeted activation of endogenous genes may provide a potential alternative. Therefore, the aim of this study was to determine whether targeted activation of Nr5a1, Gata4 and Dmrt1 (NGD) via the CRISPR/dCas9 synergistic activation mediator system could convert human foreskin fibroblasts (HFFs) into functional Leydig‐like cells. We first constructed the stable Hsd3b‐dCas9‐MPH‐HFF cell line using the Hsd3b‐EGFP, dCas9‐VP64 and MS2‐P65‐HSF1 lentiviral vectors and then infected it with single guide RNAs. Next, we evaluated the reprogrammed cells for their reprogramming efficiency, testosterone production characteristics and expression levels of Leydig steroidogenic markers by quantitative real‐time polymerase chain reaction or Western blotting. Our results showed that the reprogramming efficiency was close to 10% and that the reprogrammed Leydig‐like cells secreted testosterone rapidly and, more importantly, responded effectively to stimulation with human chorionic gonadotropin and expressed Leydig steroidogenic markers. Our findings demonstrate that simultaneous targeted activation of the endogenous NGD genes directly reprograms HFFs into functional Leydig‐like cells, providing an innovative technology that may have promising potential for the treatment of male androgen deficiency diseases.
format Online
Article
Text
id pubmed-6714237
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-67142372019-09-05 CRISPR/dCas9‐mediated activation of multiple endogenous target genes directly converts human foreskin fibroblasts into Leydig‐like cells Huang, Hua Zou, Xiangyu Zhong, Liang Hou, Yanping Zhou, Jin Zhang, Zhiyuan Xing, Xiaoyu Sun, Jie J Cell Mol Med Original Articles Recently, Leydig cell (LC) transplantation has been revealed as a promising strategy for treating male hypogonadism; however, the key problem restricting the application of LC transplantation is a severe lack of seed cells. It seems that targeted activation of endogenous genes may provide a potential alternative. Therefore, the aim of this study was to determine whether targeted activation of Nr5a1, Gata4 and Dmrt1 (NGD) via the CRISPR/dCas9 synergistic activation mediator system could convert human foreskin fibroblasts (HFFs) into functional Leydig‐like cells. We first constructed the stable Hsd3b‐dCas9‐MPH‐HFF cell line using the Hsd3b‐EGFP, dCas9‐VP64 and MS2‐P65‐HSF1 lentiviral vectors and then infected it with single guide RNAs. Next, we evaluated the reprogrammed cells for their reprogramming efficiency, testosterone production characteristics and expression levels of Leydig steroidogenic markers by quantitative real‐time polymerase chain reaction or Western blotting. Our results showed that the reprogramming efficiency was close to 10% and that the reprogrammed Leydig‐like cells secreted testosterone rapidly and, more importantly, responded effectively to stimulation with human chorionic gonadotropin and expressed Leydig steroidogenic markers. Our findings demonstrate that simultaneous targeted activation of the endogenous NGD genes directly reprograms HFFs into functional Leydig‐like cells, providing an innovative technology that may have promising potential for the treatment of male androgen deficiency diseases. John Wiley and Sons Inc. 2019-07-02 2019-09 /pmc/articles/PMC6714237/ /pubmed/31264792 http://dx.doi.org/10.1111/jcmm.14470 Text en © 2019 The Authors. Journal of Cellular and Molecular Medicine published by John Wiley & Sons Ltd and Foundation for Cellular and Molecular Medicine. This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Original Articles
Huang, Hua
Zou, Xiangyu
Zhong, Liang
Hou, Yanping
Zhou, Jin
Zhang, Zhiyuan
Xing, Xiaoyu
Sun, Jie
CRISPR/dCas9‐mediated activation of multiple endogenous target genes directly converts human foreskin fibroblasts into Leydig‐like cells
title CRISPR/dCas9‐mediated activation of multiple endogenous target genes directly converts human foreskin fibroblasts into Leydig‐like cells
title_full CRISPR/dCas9‐mediated activation of multiple endogenous target genes directly converts human foreskin fibroblasts into Leydig‐like cells
title_fullStr CRISPR/dCas9‐mediated activation of multiple endogenous target genes directly converts human foreskin fibroblasts into Leydig‐like cells
title_full_unstemmed CRISPR/dCas9‐mediated activation of multiple endogenous target genes directly converts human foreskin fibroblasts into Leydig‐like cells
title_short CRISPR/dCas9‐mediated activation of multiple endogenous target genes directly converts human foreskin fibroblasts into Leydig‐like cells
title_sort crispr/dcas9‐mediated activation of multiple endogenous target genes directly converts human foreskin fibroblasts into leydig‐like cells
topic Original Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6714237/
https://www.ncbi.nlm.nih.gov/pubmed/31264792
http://dx.doi.org/10.1111/jcmm.14470
work_keys_str_mv AT huanghua crisprdcas9mediatedactivationofmultipleendogenoustargetgenesdirectlyconvertshumanforeskinfibroblastsintoleydiglikecells
AT zouxiangyu crisprdcas9mediatedactivationofmultipleendogenoustargetgenesdirectlyconvertshumanforeskinfibroblastsintoleydiglikecells
AT zhongliang crisprdcas9mediatedactivationofmultipleendogenoustargetgenesdirectlyconvertshumanforeskinfibroblastsintoleydiglikecells
AT houyanping crisprdcas9mediatedactivationofmultipleendogenoustargetgenesdirectlyconvertshumanforeskinfibroblastsintoleydiglikecells
AT zhoujin crisprdcas9mediatedactivationofmultipleendogenoustargetgenesdirectlyconvertshumanforeskinfibroblastsintoleydiglikecells
AT zhangzhiyuan crisprdcas9mediatedactivationofmultipleendogenoustargetgenesdirectlyconvertshumanforeskinfibroblastsintoleydiglikecells
AT xingxiaoyu crisprdcas9mediatedactivationofmultipleendogenoustargetgenesdirectlyconvertshumanforeskinfibroblastsintoleydiglikecells
AT sunjie crisprdcas9mediatedactivationofmultipleendogenoustargetgenesdirectlyconvertshumanforeskinfibroblastsintoleydiglikecells