Cargando…

Chimerism through the activation of invariant natural killer T cells prolongs graft survival after transplantation of induced pluripotent stem cell–derived allogeneic cardiomyocytes

The loss of functional cells through immunological rejection after transplantation reduces the efficacy of regenerative therapies for cardiac failure that use allogeneic induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). Recently, mixed-chimera mice with donor-specific immunotolerance...

Descripción completa

Detalles Bibliográficos
Autores principales: Yoshida, Shohei, Miyagawa, Shigeru, Matsuzaki, Takashi, Ishii, Yasuyuki, Fukuda-Kawaguchi, Emi, Kawamura, Takuji, Kawamura, Ai, Nakamura, Yuki, Toda, Koichi, Sawa, Yoshiki
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8890721/
https://www.ncbi.nlm.nih.gov/pubmed/35235568
http://dx.doi.org/10.1371/journal.pone.0264317
_version_ 1784661704815149056
author Yoshida, Shohei
Miyagawa, Shigeru
Matsuzaki, Takashi
Ishii, Yasuyuki
Fukuda-Kawaguchi, Emi
Kawamura, Takuji
Kawamura, Ai
Nakamura, Yuki
Toda, Koichi
Sawa, Yoshiki
author_facet Yoshida, Shohei
Miyagawa, Shigeru
Matsuzaki, Takashi
Ishii, Yasuyuki
Fukuda-Kawaguchi, Emi
Kawamura, Takuji
Kawamura, Ai
Nakamura, Yuki
Toda, Koichi
Sawa, Yoshiki
author_sort Yoshida, Shohei
collection PubMed
description The loss of functional cells through immunological rejection after transplantation reduces the efficacy of regenerative therapies for cardiac failure that use allogeneic induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). Recently, mixed-chimera mice with donor-specific immunotolerance have been established using the RGI-2001 (liposomal formulation of α-galactosyl ceramide) ligand, which activates invariant natural killer T (iNKT) cells. The present study aimed to investigate whether mixed chimerism, established using RGI-2001, prolongs graft survival in allogeneic iPSC-CM transplantation. Mixed-chimera mice were established via combinatorial treatment with RGI-2001 and anti-CD154 antibodies in an irradiated murine bone marrow transplant model. Luciferase-expressing allogeneic iPSC-CMs were transplanted into mixed-chimera and untreated mice, followed by in vivo imaging. RGI-2001 enhanced iNKT cell activation in mice, and mixed chimerism was successfully established. In vivo imaging revealed that while the allografts were completely obliterated within 2 weeks when transplanted to untreated mice, their survivals were not affected in the mixed-chimera mice. Furthermore, numerous CD3+ cells infiltrated allografts in untreated mice, but fewer CD3+ cells were present in mixed-chimera mice. We conclude that mixed-chimera mice established using RGI-2001 showed prolonged graft survival after allogeneic iPSC-CM transplantation. This donor-specific immunotolerance might increase the efficacy of regenerative therapies for heart failure with allogeneic iPSC-CMs.
format Online
Article
Text
id pubmed-8890721
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-88907212022-03-03 Chimerism through the activation of invariant natural killer T cells prolongs graft survival after transplantation of induced pluripotent stem cell–derived allogeneic cardiomyocytes Yoshida, Shohei Miyagawa, Shigeru Matsuzaki, Takashi Ishii, Yasuyuki Fukuda-Kawaguchi, Emi Kawamura, Takuji Kawamura, Ai Nakamura, Yuki Toda, Koichi Sawa, Yoshiki PLoS One Research Article The loss of functional cells through immunological rejection after transplantation reduces the efficacy of regenerative therapies for cardiac failure that use allogeneic induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). Recently, mixed-chimera mice with donor-specific immunotolerance have been established using the RGI-2001 (liposomal formulation of α-galactosyl ceramide) ligand, which activates invariant natural killer T (iNKT) cells. The present study aimed to investigate whether mixed chimerism, established using RGI-2001, prolongs graft survival in allogeneic iPSC-CM transplantation. Mixed-chimera mice were established via combinatorial treatment with RGI-2001 and anti-CD154 antibodies in an irradiated murine bone marrow transplant model. Luciferase-expressing allogeneic iPSC-CMs were transplanted into mixed-chimera and untreated mice, followed by in vivo imaging. RGI-2001 enhanced iNKT cell activation in mice, and mixed chimerism was successfully established. In vivo imaging revealed that while the allografts were completely obliterated within 2 weeks when transplanted to untreated mice, their survivals were not affected in the mixed-chimera mice. Furthermore, numerous CD3+ cells infiltrated allografts in untreated mice, but fewer CD3+ cells were present in mixed-chimera mice. We conclude that mixed-chimera mice established using RGI-2001 showed prolonged graft survival after allogeneic iPSC-CM transplantation. This donor-specific immunotolerance might increase the efficacy of regenerative therapies for heart failure with allogeneic iPSC-CMs. Public Library of Science 2022-03-02 /pmc/articles/PMC8890721/ /pubmed/35235568 http://dx.doi.org/10.1371/journal.pone.0264317 Text en © 2022 Yoshida et al https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/) , which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Article
Yoshida, Shohei
Miyagawa, Shigeru
Matsuzaki, Takashi
Ishii, Yasuyuki
Fukuda-Kawaguchi, Emi
Kawamura, Takuji
Kawamura, Ai
Nakamura, Yuki
Toda, Koichi
Sawa, Yoshiki
Chimerism through the activation of invariant natural killer T cells prolongs graft survival after transplantation of induced pluripotent stem cell–derived allogeneic cardiomyocytes
title Chimerism through the activation of invariant natural killer T cells prolongs graft survival after transplantation of induced pluripotent stem cell–derived allogeneic cardiomyocytes
title_full Chimerism through the activation of invariant natural killer T cells prolongs graft survival after transplantation of induced pluripotent stem cell–derived allogeneic cardiomyocytes
title_fullStr Chimerism through the activation of invariant natural killer T cells prolongs graft survival after transplantation of induced pluripotent stem cell–derived allogeneic cardiomyocytes
title_full_unstemmed Chimerism through the activation of invariant natural killer T cells prolongs graft survival after transplantation of induced pluripotent stem cell–derived allogeneic cardiomyocytes
title_short Chimerism through the activation of invariant natural killer T cells prolongs graft survival after transplantation of induced pluripotent stem cell–derived allogeneic cardiomyocytes
title_sort chimerism through the activation of invariant natural killer t cells prolongs graft survival after transplantation of induced pluripotent stem cell–derived allogeneic cardiomyocytes
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8890721/
https://www.ncbi.nlm.nih.gov/pubmed/35235568
http://dx.doi.org/10.1371/journal.pone.0264317
work_keys_str_mv AT yoshidashohei chimerismthroughtheactivationofinvariantnaturalkillertcellsprolongsgraftsurvivalaftertransplantationofinducedpluripotentstemcellderivedallogeneiccardiomyocytes
AT miyagawashigeru chimerismthroughtheactivationofinvariantnaturalkillertcellsprolongsgraftsurvivalaftertransplantationofinducedpluripotentstemcellderivedallogeneiccardiomyocytes
AT matsuzakitakashi chimerismthroughtheactivationofinvariantnaturalkillertcellsprolongsgraftsurvivalaftertransplantationofinducedpluripotentstemcellderivedallogeneiccardiomyocytes
AT ishiiyasuyuki chimerismthroughtheactivationofinvariantnaturalkillertcellsprolongsgraftsurvivalaftertransplantationofinducedpluripotentstemcellderivedallogeneiccardiomyocytes
AT fukudakawaguchiemi chimerismthroughtheactivationofinvariantnaturalkillertcellsprolongsgraftsurvivalaftertransplantationofinducedpluripotentstemcellderivedallogeneiccardiomyocytes
AT kawamuratakuji chimerismthroughtheactivationofinvariantnaturalkillertcellsprolongsgraftsurvivalaftertransplantationofinducedpluripotentstemcellderivedallogeneiccardiomyocytes
AT kawamuraai chimerismthroughtheactivationofinvariantnaturalkillertcellsprolongsgraftsurvivalaftertransplantationofinducedpluripotentstemcellderivedallogeneiccardiomyocytes
AT nakamurayuki chimerismthroughtheactivationofinvariantnaturalkillertcellsprolongsgraftsurvivalaftertransplantationofinducedpluripotentstemcellderivedallogeneiccardiomyocytes
AT todakoichi chimerismthroughtheactivationofinvariantnaturalkillertcellsprolongsgraftsurvivalaftertransplantationofinducedpluripotentstemcellderivedallogeneiccardiomyocytes
AT sawayoshiki chimerismthroughtheactivationofinvariantnaturalkillertcellsprolongsgraftsurvivalaftertransplantationofinducedpluripotentstemcellderivedallogeneiccardiomyocytes