Basic Study
Copyright ©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Stem Cells. Aug 26, 2022; 14(8): 599-615
Published online Aug 26, 2022. doi: 10.4252/wjsc.v14.i8.599
Sinomenine promotes differentiation of induced pluripotent stem cells into immature dendritic cells with high induction of immune tolerance
Xiao-Yan Huang, Zhan-Kui Jin, Meng Dou, Bing-Xuan Zheng, Xiang-Rong Zhao, Qing Feng, Yang-Meng Feng, Xiang-Long Duan, Pu-Xun Tian, Cui-Xiang Xu
Xiao-Yan Huang, Xiang-Rong Zhao, Qing Feng, Yang-Meng Feng, Cui-Xiang Xu, Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People’s Hospital, Xi'an 710068, Shaanxi Province, China
Zhan-Kui Jin, Department of Orthopedics, Shaanxi Provincial People’s Hospital, Xi'an 710068, Shaanxi Province, China
Meng Dou, Bing-Xuan Zheng, Pu-Xun Tian, Department of Kidney Transplantation, The First Affiliated Hospital of Xi’an Jiaotong University, Xi'an 710061, Shaanxi Province, China
Xiang-Long Duan, Second Department of General Surgery, Shaanxi Provincial People's Hospital, Xi'an 710068, Shaanxi Province, China
Author contributions: Huang XY, Jin ZK, Tian PX and Xu CX conceived and designed the study, and drafted the article; Dou M, Zhao XR, Feng Q, Feng YM and Duan XL performed the experiments; Huang XY and Xu CX analyzed the data; Huang XY, Jin ZK, Dou M and Xu CX performed data analysis; all authors contributed to the collection of literature data, reviewed the manuscript, and approved the version to be published.
Supported by National Natural Science Foundation of China, No. 81900686; Science and Technology Incubation Fund Project of Shaanxi Provincial People’s Hospital, No. 2020YXM-08; Technology Talent Support Program of Shaanxi Provincial People’s Hospital, No. 2021BJ-07; Key Projects of Shaanxi Provincial Department of Education, No. 21JS038; and Medical Research Development Fund of Beijing Kangmeng Charity Foundation, No. 7B202010.
Institutional animal care and use committee statement: All of the procedures and ethics of animal use were reviewed and approved by the Biomedical Ethics Committee of Shaanxi Provicial People’s Hospital, No. 2021-198.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Data sharing statement: No additional data are available.
ARRIVE guidelines statement: The authors have read the ARRIVE guidelines, and the manuscript was prepared and revised according to the ARRIVE guidelines.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Cui-Xiang Xu, PhD, Chief Doctor, Doctor, Senior Researcher, Shaanxi Provincial Key Laboratory of Infection and Immune Diseases, Shaanxi Provincial People’s Hospital, No. 256 West Road, Beilin District, Xi'an 710068, Shaanxi Province, China. xucuixiang1129@163.com
Received: February 25, 2022
Peer-review started: February 25, 2022
First decision: April 19, 2022
Revised: May 16, 2022
Accepted: July 11, 2022
Article in press: July 11, 2022
Published online: August 26, 2022
Abstract
BACKGROUND

Immature dendritic cells (imDCs) play an important role in the induction of donor-specific transplant immunotolerance. However, these cells have limitations, such as rapid maturation and a short lifespan in vivo. In previous studies, induced pluripotent stem cells (iPSCs) differentiated into imDCs, and sinomenine (SN) was used to inhibit the maturation of imDCs.

AIM

To study the capacity of SN to maintain iPSC-derived imDCs (SN-iPSCs-imDCs) in an immature state and the mechanism by which SN-iPSCs-imDCs induce immunotolerance.

METHODS

In this study, mouse iPSCs were induced to differentiate into imDCs in culture medium without or with SN (iPSCs-imDCs and SN-iPSCs-imDCs). The imDC-related surface markers, endocytotic capacity of fluorescein isothiocyanate-Dextran and apoptosis were analyzed by flow cytometry. The effects of iPSCs-imDCs and SN-iPSCs-imDCs on T-cell stimulatory function, and regulatory T (Treg) cell proliferative function in vitro were analyzed by mixed lymphocyte reaction. Cytokine expression was detected by ELISA. The apoptosis-related proteins of iPSCs-DCs and SN-iPSCs-DCs were analyzed by western blotting. The induced immunotolerance of SN-iPSCs-DCs was evaluated by treating recipient Balb/c skin graft mice. Statistical evaluation of graft survival was performed using Kaplan–Meier curves.

RESULTS

Both iPSCs-imDCs and SN-iPSCs-imDCs were successfully obtained, and their biological characteristics and ability to induce immunotolerance were compared. SN-iPSCs-imDCs exhibited higher CD11c levels and lower CD80 and CD86 levels compared with iPSCs-imDCs. Reduced major histocompatibility complex II expression, worse T-cell stimulatory function, higher Treg cell proliferative function and stronger endocytotic capacity were observed with SN-iPSCs-imDCs (P < 0.05). The levels of interleukin (IL)-2, IL-12, interferon-γ in SN-iPSCs-imDCs were lower than those in iPSCs-imDCs, whereas IL-10 and transforming growth factor-β levels were higher (P < 0.05). The apoptosis rate of these cells was significantly higher (P < 0.05), and the expression levels of cleaved caspase3, Bax and cleaved poly(ADP-ribose) polymerase were higher after treatment with lipopolysaccharides, but Bcl-2 was reduced. In Balb/c mice recipients immunized with iPSCs-imDCs or SN-iPSCs-imDCs 7 d before skin grafting, the SN-iPSCs-imDCs group showed lower ability to inhibit donor-specific CD4+ T-cell proliferation (P < 0.05) and a higher capacity to induce CD4+CD25+FoxP3+ Treg cell proliferation in the spleen (P < 0.05). The survival span of C57bl/6 skin grafts was significantly prolonged in immunized Balb/c recipients with a donor-specific pattern.

CONCLUSION

This study demonstrated that SN-iPSCs-imDCs have potential applications in vitro and in vivo for induction of immunotolerance following organ transplantation.

Keywords: Immature dendritic cells, Induced pluripotent stem cells, Sinomenine, Immune tolerance, Organ transplantation

Core Tip: Immature dendritic cells (imDCs) play an important role in the induction of donor-specific transplant immune tolerance. However, these cells have limitations, such as rapid maturation and a short lifespan. This study focused on exploring sinomenine (SN) to promote differentiation of induced pluripotent stem cells (iPSCs) into imDCs (SN-iPSCs-imDCs), and found SN-iPSCs-imDCs with worse donor-specific T-cell stimulatory function, and higher regulatory T-cell proliferative function in vitro and in vivo to induce high immune tolerance. It provided a new idea for the application of the combination of traditional Chinese medicine and modern new technologies in transplantation immunity.