Cargando…

Restorative functions of Autologous Stem Leydig Cell transplantation in a Testosterone-deficient non-human primate model

Rationale: Stem Leydig cells (SLCs) transplantation can restore testosterone production in rodent models and is thus a potential solution for treating testosterone deficiency (TD). However, it remains unknown whether these favorable effects will be reproduced in more clinically relevant large-animal...

Descripción completa

Detalles Bibliográficos
Autores principales: Xia, Kai, Chen, Hong, Wang, Jiancheng, Feng, Xin, Gao, Yong, Wang, Yi, Deng, Rongda, Wu, Chunxing, Luo, Peng, Zhang, Min, Wang, Chao, Zhang, Yong, Zhang, Yadong, Liu, Guihua, Tu, Xiang'an, Sun, Xiangzhou, Li, Weiqiang, Ke, Qiong, Deng, Chunhua, Xiang, Andy Peng
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7392013/
https://www.ncbi.nlm.nih.gov/pubmed/32754273
http://dx.doi.org/10.7150/thno.46854
_version_ 1783564765048602624
author Xia, Kai
Chen, Hong
Wang, Jiancheng
Feng, Xin
Gao, Yong
Wang, Yi
Deng, Rongda
Wu, Chunxing
Luo, Peng
Zhang, Min
Wang, Chao
Zhang, Yong
Zhang, Yadong
Liu, Guihua
Tu, Xiang'an
Sun, Xiangzhou
Li, Weiqiang
Ke, Qiong
Deng, Chunhua
Xiang, Andy Peng
author_facet Xia, Kai
Chen, Hong
Wang, Jiancheng
Feng, Xin
Gao, Yong
Wang, Yi
Deng, Rongda
Wu, Chunxing
Luo, Peng
Zhang, Min
Wang, Chao
Zhang, Yong
Zhang, Yadong
Liu, Guihua
Tu, Xiang'an
Sun, Xiangzhou
Li, Weiqiang
Ke, Qiong
Deng, Chunhua
Xiang, Andy Peng
author_sort Xia, Kai
collection PubMed
description Rationale: Stem Leydig cells (SLCs) transplantation can restore testosterone production in rodent models and is thus a potential solution for treating testosterone deficiency (TD). However, it remains unknown whether these favorable effects will be reproduced in more clinically relevant large-animal models. Therefore, we assessed the feasibility, safety and efficacy of autologous SLCs transplantation in a testosterone-deficient non-human primate (NHP) model. Methods: Cynomolgus monkey SLCs (CM-SLCs) were isolated from testis biopsies of elderly (> 19 years) cynomolgus monkeys by flow cytometry. Autologous CM-SLCs were injected into the testicular interstitium of 7 monkeys. Another 4 monkeys were injected the same way with cynomolgus monkey dermal fibroblasts (CM-DFs) as controls. The animals were then examined for sex hormones, semen, body composition, grip strength, and exercise activity. Results: We first isolated CD271(+) CM-SLCs which were confirmed to expand continuously and show potential to differentiate into testosterone-producing Leydig cells (LCs) in vitro. Compared with CM-DFs transplantation, engraftment of autologous CM-SLCs into elderly monkeys could significantly increase the serum testosterone level in a physiological pattern for 8 weeks, without any need for immunosuppression. Importantly, CM-SLCs transplantation recovered spermatogenesis and ameliorated TD-related symptoms, such as those related to body fat mass, lean mass, bone mineral density, strength and exercise capacity. Conclusion: For the first time, our short-term observations demonstrated that autologous SLCs can increase testosterone levels and ameliorate relevant TD symptoms in primate models. A larger cohort with long-term follow-up will be required to assess the translational potential of autologous SLCs for TD therapy.
format Online
Article
Text
id pubmed-7392013
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-73920132020-08-03 Restorative functions of Autologous Stem Leydig Cell transplantation in a Testosterone-deficient non-human primate model Xia, Kai Chen, Hong Wang, Jiancheng Feng, Xin Gao, Yong Wang, Yi Deng, Rongda Wu, Chunxing Luo, Peng Zhang, Min Wang, Chao Zhang, Yong Zhang, Yadong Liu, Guihua Tu, Xiang'an Sun, Xiangzhou Li, Weiqiang Ke, Qiong Deng, Chunhua Xiang, Andy Peng Theranostics Research Paper Rationale: Stem Leydig cells (SLCs) transplantation can restore testosterone production in rodent models and is thus a potential solution for treating testosterone deficiency (TD). However, it remains unknown whether these favorable effects will be reproduced in more clinically relevant large-animal models. Therefore, we assessed the feasibility, safety and efficacy of autologous SLCs transplantation in a testosterone-deficient non-human primate (NHP) model. Methods: Cynomolgus monkey SLCs (CM-SLCs) were isolated from testis biopsies of elderly (> 19 years) cynomolgus monkeys by flow cytometry. Autologous CM-SLCs were injected into the testicular interstitium of 7 monkeys. Another 4 monkeys were injected the same way with cynomolgus monkey dermal fibroblasts (CM-DFs) as controls. The animals were then examined for sex hormones, semen, body composition, grip strength, and exercise activity. Results: We first isolated CD271(+) CM-SLCs which were confirmed to expand continuously and show potential to differentiate into testosterone-producing Leydig cells (LCs) in vitro. Compared with CM-DFs transplantation, engraftment of autologous CM-SLCs into elderly monkeys could significantly increase the serum testosterone level in a physiological pattern for 8 weeks, without any need for immunosuppression. Importantly, CM-SLCs transplantation recovered spermatogenesis and ameliorated TD-related symptoms, such as those related to body fat mass, lean mass, bone mineral density, strength and exercise capacity. Conclusion: For the first time, our short-term observations demonstrated that autologous SLCs can increase testosterone levels and ameliorate relevant TD symptoms in primate models. A larger cohort with long-term follow-up will be required to assess the translational potential of autologous SLCs for TD therapy. Ivyspring International Publisher 2020-07-09 /pmc/articles/PMC7392013/ /pubmed/32754273 http://dx.doi.org/10.7150/thno.46854 Text en © The author(s) This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Xia, Kai
Chen, Hong
Wang, Jiancheng
Feng, Xin
Gao, Yong
Wang, Yi
Deng, Rongda
Wu, Chunxing
Luo, Peng
Zhang, Min
Wang, Chao
Zhang, Yong
Zhang, Yadong
Liu, Guihua
Tu, Xiang'an
Sun, Xiangzhou
Li, Weiqiang
Ke, Qiong
Deng, Chunhua
Xiang, Andy Peng
Restorative functions of Autologous Stem Leydig Cell transplantation in a Testosterone-deficient non-human primate model
title Restorative functions of Autologous Stem Leydig Cell transplantation in a Testosterone-deficient non-human primate model
title_full Restorative functions of Autologous Stem Leydig Cell transplantation in a Testosterone-deficient non-human primate model
title_fullStr Restorative functions of Autologous Stem Leydig Cell transplantation in a Testosterone-deficient non-human primate model
title_full_unstemmed Restorative functions of Autologous Stem Leydig Cell transplantation in a Testosterone-deficient non-human primate model
title_short Restorative functions of Autologous Stem Leydig Cell transplantation in a Testosterone-deficient non-human primate model
title_sort restorative functions of autologous stem leydig cell transplantation in a testosterone-deficient non-human primate model
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7392013/
https://www.ncbi.nlm.nih.gov/pubmed/32754273
http://dx.doi.org/10.7150/thno.46854
work_keys_str_mv AT xiakai restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel
AT chenhong restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel
AT wangjiancheng restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel
AT fengxin restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel
AT gaoyong restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel
AT wangyi restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel
AT dengrongda restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel
AT wuchunxing restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel
AT luopeng restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel
AT zhangmin restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel
AT wangchao restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel
AT zhangyong restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel
AT zhangyadong restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel
AT liuguihua restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel
AT tuxiangan restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel
AT sunxiangzhou restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel
AT liweiqiang restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel
AT keqiong restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel
AT dengchunhua restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel
AT xiangandypeng restorativefunctionsofautologousstemleydigcelltransplantationinatestosteronedeficientnonhumanprimatemodel