Cargando…

Generation of macrophages with altered viral sensitivity from genome-edited rhesus macaque iPSCs to model human disease

Because of their close biological similarity to humans, non-human primate (NHP) models are very useful for the development of induced pluripotent stem cell (iPSC)-based cell and regenerative organ transplantation therapies. However, knowledge on the establishment, differentiation, and genetic modifi...

Descripción completa

Detalles Bibliográficos
Autores principales: Iwamoto, Yoshihiro, Seki, Yohei, Taya, Kahoru, Tanaka, Masahiro, Iriguchi, Shoichi, Miyake, Yasuyuki, Nakayama, Emi E., Miura, Tomoyuki, Shioda, Tatsuo, Akari, Hirofumi, Takaori-Kondo, Akifumi, Kaneko, Shin
Formato: Online Artículo Texto
Lenguaje:English
Publicado: American Society of Gene & Cell Therapy 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8039773/
https://www.ncbi.nlm.nih.gov/pubmed/33869654
http://dx.doi.org/10.1016/j.omtm.2021.03.008
_version_ 1783677665185628160
author Iwamoto, Yoshihiro
Seki, Yohei
Taya, Kahoru
Tanaka, Masahiro
Iriguchi, Shoichi
Miyake, Yasuyuki
Nakayama, Emi E.
Miura, Tomoyuki
Shioda, Tatsuo
Akari, Hirofumi
Takaori-Kondo, Akifumi
Kaneko, Shin
author_facet Iwamoto, Yoshihiro
Seki, Yohei
Taya, Kahoru
Tanaka, Masahiro
Iriguchi, Shoichi
Miyake, Yasuyuki
Nakayama, Emi E.
Miura, Tomoyuki
Shioda, Tatsuo
Akari, Hirofumi
Takaori-Kondo, Akifumi
Kaneko, Shin
author_sort Iwamoto, Yoshihiro
collection PubMed
description Because of their close biological similarity to humans, non-human primate (NHP) models are very useful for the development of induced pluripotent stem cell (iPSC)-based cell and regenerative organ transplantation therapies. However, knowledge on the establishment, differentiation, and genetic modification of NHP-iPSCs, especially rhesus macaque iPSCs, is limited. We succeeded in establishing iPSCs from the peripheral blood of rhesus macaques (Rh-iPSCs) by combining the Yamanaka reprograming factors and two inhibitors (GSK-3 inhibitor [CHIR 99021] and MEK1/2 inhibitor [PD0325901]) and differentiated the cells into functional macrophages through hematopoietic progenitor cells. To confirm feasibility of the Rh-iPSC-derived macrophages as a platform for bioassays to model diseases, we knocked out TRIM5 gene in Rh-iPSCs by CRISPR-Cas9, which is a species-specific HIV resistance factor. TRIM5 knockout (KO) iPSCs had the same differentiation potential to macrophages as did Rh-iPSCs, but the differentiated macrophages showed a gain of sensitivity to HIV infection in vitro. Our reprogramming, gene editing, and differentiation protocols used to obtain Rh-iPSC-derived macrophages can be applied to other gene mutations, expanding the number of NHP gene therapy models.
format Online
Article
Text
id pubmed-8039773
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher American Society of Gene & Cell Therapy
record_format MEDLINE/PubMed
spelling pubmed-80397732021-04-16 Generation of macrophages with altered viral sensitivity from genome-edited rhesus macaque iPSCs to model human disease Iwamoto, Yoshihiro Seki, Yohei Taya, Kahoru Tanaka, Masahiro Iriguchi, Shoichi Miyake, Yasuyuki Nakayama, Emi E. Miura, Tomoyuki Shioda, Tatsuo Akari, Hirofumi Takaori-Kondo, Akifumi Kaneko, Shin Mol Ther Methods Clin Dev Original Article Because of their close biological similarity to humans, non-human primate (NHP) models are very useful for the development of induced pluripotent stem cell (iPSC)-based cell and regenerative organ transplantation therapies. However, knowledge on the establishment, differentiation, and genetic modification of NHP-iPSCs, especially rhesus macaque iPSCs, is limited. We succeeded in establishing iPSCs from the peripheral blood of rhesus macaques (Rh-iPSCs) by combining the Yamanaka reprograming factors and two inhibitors (GSK-3 inhibitor [CHIR 99021] and MEK1/2 inhibitor [PD0325901]) and differentiated the cells into functional macrophages through hematopoietic progenitor cells. To confirm feasibility of the Rh-iPSC-derived macrophages as a platform for bioassays to model diseases, we knocked out TRIM5 gene in Rh-iPSCs by CRISPR-Cas9, which is a species-specific HIV resistance factor. TRIM5 knockout (KO) iPSCs had the same differentiation potential to macrophages as did Rh-iPSCs, but the differentiated macrophages showed a gain of sensitivity to HIV infection in vitro. Our reprogramming, gene editing, and differentiation protocols used to obtain Rh-iPSC-derived macrophages can be applied to other gene mutations, expanding the number of NHP gene therapy models. American Society of Gene & Cell Therapy 2021-03-17 /pmc/articles/PMC8039773/ /pubmed/33869654 http://dx.doi.org/10.1016/j.omtm.2021.03.008 Text en © 2021 The Authors https://creativecommons.org/licenses/by/4.0/This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
spellingShingle Original Article
Iwamoto, Yoshihiro
Seki, Yohei
Taya, Kahoru
Tanaka, Masahiro
Iriguchi, Shoichi
Miyake, Yasuyuki
Nakayama, Emi E.
Miura, Tomoyuki
Shioda, Tatsuo
Akari, Hirofumi
Takaori-Kondo, Akifumi
Kaneko, Shin
Generation of macrophages with altered viral sensitivity from genome-edited rhesus macaque iPSCs to model human disease
title Generation of macrophages with altered viral sensitivity from genome-edited rhesus macaque iPSCs to model human disease
title_full Generation of macrophages with altered viral sensitivity from genome-edited rhesus macaque iPSCs to model human disease
title_fullStr Generation of macrophages with altered viral sensitivity from genome-edited rhesus macaque iPSCs to model human disease
title_full_unstemmed Generation of macrophages with altered viral sensitivity from genome-edited rhesus macaque iPSCs to model human disease
title_short Generation of macrophages with altered viral sensitivity from genome-edited rhesus macaque iPSCs to model human disease
title_sort generation of macrophages with altered viral sensitivity from genome-edited rhesus macaque ipscs to model human disease
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8039773/
https://www.ncbi.nlm.nih.gov/pubmed/33869654
http://dx.doi.org/10.1016/j.omtm.2021.03.008
work_keys_str_mv AT iwamotoyoshihiro generationofmacrophageswithalteredviralsensitivityfromgenomeeditedrhesusmacaqueipscstomodelhumandisease
AT sekiyohei generationofmacrophageswithalteredviralsensitivityfromgenomeeditedrhesusmacaqueipscstomodelhumandisease
AT tayakahoru generationofmacrophageswithalteredviralsensitivityfromgenomeeditedrhesusmacaqueipscstomodelhumandisease
AT tanakamasahiro generationofmacrophageswithalteredviralsensitivityfromgenomeeditedrhesusmacaqueipscstomodelhumandisease
AT iriguchishoichi generationofmacrophageswithalteredviralsensitivityfromgenomeeditedrhesusmacaqueipscstomodelhumandisease
AT miyakeyasuyuki generationofmacrophageswithalteredviralsensitivityfromgenomeeditedrhesusmacaqueipscstomodelhumandisease
AT nakayamaemie generationofmacrophageswithalteredviralsensitivityfromgenomeeditedrhesusmacaqueipscstomodelhumandisease
AT miuratomoyuki generationofmacrophageswithalteredviralsensitivityfromgenomeeditedrhesusmacaqueipscstomodelhumandisease
AT shiodatatsuo generationofmacrophageswithalteredviralsensitivityfromgenomeeditedrhesusmacaqueipscstomodelhumandisease
AT akarihirofumi generationofmacrophageswithalteredviralsensitivityfromgenomeeditedrhesusmacaqueipscstomodelhumandisease
AT takaorikondoakifumi generationofmacrophageswithalteredviralsensitivityfromgenomeeditedrhesusmacaqueipscstomodelhumandisease
AT kanekoshin generationofmacrophageswithalteredviralsensitivityfromgenomeeditedrhesusmacaqueipscstomodelhumandisease