Cargando…

Regulatory T-cells regulate neonatal heart regeneration by potentiating cardiomyocyte proliferation in a paracrine manner

The neonatal mouse heart is capable of transiently regenerating after injury from postnatal day (P) 0-7 and macrophages are found important in this process. However, whether macrophages alone are sufficient to orchestrate this regeneration; what regulates cardiomyocyte proliferation; why cardiomyocy...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Jiatao, Yang, Kevin Y., Tam, Rachel Chun Yee, Chan, Vicken W., Lan, Hui Yao, Hori, Shohei, Zhou, Bin, Lui, Kathy O.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6599663/
https://www.ncbi.nlm.nih.gov/pubmed/31285764
http://dx.doi.org/10.7150/thno.32734
_version_ 1783430978630320128
author Li, Jiatao
Yang, Kevin Y.
Tam, Rachel Chun Yee
Chan, Vicken W.
Lan, Hui Yao
Hori, Shohei
Zhou, Bin
Lui, Kathy O.
author_facet Li, Jiatao
Yang, Kevin Y.
Tam, Rachel Chun Yee
Chan, Vicken W.
Lan, Hui Yao
Hori, Shohei
Zhou, Bin
Lui, Kathy O.
author_sort Li, Jiatao
collection PubMed
description The neonatal mouse heart is capable of transiently regenerating after injury from postnatal day (P) 0-7 and macrophages are found important in this process. However, whether macrophages alone are sufficient to orchestrate this regeneration; what regulates cardiomyocyte proliferation; why cardiomyocytes do not proliferate after P7; and whether adaptive immune cells such as regulatory T-cells (Treg) influence neonatal heart regeneration have less studied. Methods: We employed both loss- and gain-of-function transgenic mouse models to study the role of Treg in neonatal heart regeneration. In loss-of-function studies, we treated mice with the lytic anti-CD25 antibody that specifically depletes Treg; or we treated FOXP3(DTR) with diphtheria toxin that specifically ablates Treg. In gain-of-function studies, we adoptively transferred hCD2(+) Treg from NOD.Foxp3(hCD2) to NOD/SCID that contain Treg as the only T-cell population. Furthermore, we performed single-cell RNA-sequencing of Treg to uncover paracrine factors essential for cardiomyocyte proliferation. Results: Unlike their wild type counterparts, NOD/SCID mice that are deficient in T-cells but harbor macrophages fail to regenerate their injured myocardium at as early as P3. During the first week of injury, Treg are recruited to the injured cardiac muscle but their depletion contributes to more severe cardiac fibrosis. On the other hand, adoptive transfer of Treg results in mitigated fibrosis and enhanced proliferation and function of the injured cardiac muscle. Mechanistically, single-cell transcriptomic profiling reveals that Treg could be a source of regenerative factors. Treg directly promote proliferation of both mouse and human cardiomyocytes in a paracrine manner; and their secreted factors such as CCL24, GAS6 or AREG potentiate neonatal cardiomyocyte proliferation. By comparing the regenerating P3 and non-regenerating P8 heart, there is a significant increase in the absolute number of intracardiac Treg but the whole transcriptomes of these Treg do not differ regardless of whether the neonatal heart regenerates. Furthermore, even adult Treg, given sufficient quantity, possess the same regenerative capability. Conclusion: Our results demonstrate a regenerative role of Treg in neonatal heart regeneration. Treg can directly facilitate cardiomyocyte proliferation in a paracrine manner.
format Online
Article
Text
id pubmed-6599663
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-65996632019-07-08 Regulatory T-cells regulate neonatal heart regeneration by potentiating cardiomyocyte proliferation in a paracrine manner Li, Jiatao Yang, Kevin Y. Tam, Rachel Chun Yee Chan, Vicken W. Lan, Hui Yao Hori, Shohei Zhou, Bin Lui, Kathy O. Theranostics Research Paper The neonatal mouse heart is capable of transiently regenerating after injury from postnatal day (P) 0-7 and macrophages are found important in this process. However, whether macrophages alone are sufficient to orchestrate this regeneration; what regulates cardiomyocyte proliferation; why cardiomyocytes do not proliferate after P7; and whether adaptive immune cells such as regulatory T-cells (Treg) influence neonatal heart regeneration have less studied. Methods: We employed both loss- and gain-of-function transgenic mouse models to study the role of Treg in neonatal heart regeneration. In loss-of-function studies, we treated mice with the lytic anti-CD25 antibody that specifically depletes Treg; or we treated FOXP3(DTR) with diphtheria toxin that specifically ablates Treg. In gain-of-function studies, we adoptively transferred hCD2(+) Treg from NOD.Foxp3(hCD2) to NOD/SCID that contain Treg as the only T-cell population. Furthermore, we performed single-cell RNA-sequencing of Treg to uncover paracrine factors essential for cardiomyocyte proliferation. Results: Unlike their wild type counterparts, NOD/SCID mice that are deficient in T-cells but harbor macrophages fail to regenerate their injured myocardium at as early as P3. During the first week of injury, Treg are recruited to the injured cardiac muscle but their depletion contributes to more severe cardiac fibrosis. On the other hand, adoptive transfer of Treg results in mitigated fibrosis and enhanced proliferation and function of the injured cardiac muscle. Mechanistically, single-cell transcriptomic profiling reveals that Treg could be a source of regenerative factors. Treg directly promote proliferation of both mouse and human cardiomyocytes in a paracrine manner; and their secreted factors such as CCL24, GAS6 or AREG potentiate neonatal cardiomyocyte proliferation. By comparing the regenerating P3 and non-regenerating P8 heart, there is a significant increase in the absolute number of intracardiac Treg but the whole transcriptomes of these Treg do not differ regardless of whether the neonatal heart regenerates. Furthermore, even adult Treg, given sufficient quantity, possess the same regenerative capability. Conclusion: Our results demonstrate a regenerative role of Treg in neonatal heart regeneration. Treg can directly facilitate cardiomyocyte proliferation in a paracrine manner. Ivyspring International Publisher 2019-06-09 /pmc/articles/PMC6599663/ /pubmed/31285764 http://dx.doi.org/10.7150/thno.32734 Text en © Ivyspring International Publisher This is an open access article distributed under the terms of the Creative Commons Attribution (CC BY-NC) license (https://creativecommons.org/licenses/by-nc/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Li, Jiatao
Yang, Kevin Y.
Tam, Rachel Chun Yee
Chan, Vicken W.
Lan, Hui Yao
Hori, Shohei
Zhou, Bin
Lui, Kathy O.
Regulatory T-cells regulate neonatal heart regeneration by potentiating cardiomyocyte proliferation in a paracrine manner
title Regulatory T-cells regulate neonatal heart regeneration by potentiating cardiomyocyte proliferation in a paracrine manner
title_full Regulatory T-cells regulate neonatal heart regeneration by potentiating cardiomyocyte proliferation in a paracrine manner
title_fullStr Regulatory T-cells regulate neonatal heart regeneration by potentiating cardiomyocyte proliferation in a paracrine manner
title_full_unstemmed Regulatory T-cells regulate neonatal heart regeneration by potentiating cardiomyocyte proliferation in a paracrine manner
title_short Regulatory T-cells regulate neonatal heart regeneration by potentiating cardiomyocyte proliferation in a paracrine manner
title_sort regulatory t-cells regulate neonatal heart regeneration by potentiating cardiomyocyte proliferation in a paracrine manner
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6599663/
https://www.ncbi.nlm.nih.gov/pubmed/31285764
http://dx.doi.org/10.7150/thno.32734
work_keys_str_mv AT lijiatao regulatorytcellsregulateneonatalheartregenerationbypotentiatingcardiomyocyteproliferationinaparacrinemanner
AT yangkeviny regulatorytcellsregulateneonatalheartregenerationbypotentiatingcardiomyocyteproliferationinaparacrinemanner
AT tamrachelchunyee regulatorytcellsregulateneonatalheartregenerationbypotentiatingcardiomyocyteproliferationinaparacrinemanner
AT chanvickenw regulatorytcellsregulateneonatalheartregenerationbypotentiatingcardiomyocyteproliferationinaparacrinemanner
AT lanhuiyao regulatorytcellsregulateneonatalheartregenerationbypotentiatingcardiomyocyteproliferationinaparacrinemanner
AT horishohei regulatorytcellsregulateneonatalheartregenerationbypotentiatingcardiomyocyteproliferationinaparacrinemanner
AT zhoubin regulatorytcellsregulateneonatalheartregenerationbypotentiatingcardiomyocyteproliferationinaparacrinemanner
AT luikathyo regulatorytcellsregulateneonatalheartregenerationbypotentiatingcardiomyocyteproliferationinaparacrinemanner