Dysfunctional bone marrow endothelial progenitor cells are involved in patients with myelodysplastic syndromes

Abstract Background Myelodysplastic syndromes (MDS) are a group of heterogeneous myeloid clonal disorders characterized by ineffective haematopoiesis and immune deregulation. Emerging evidence has shown the effect of bone marrow (BM) endothelial progenitor cells (EPCs) in regulating haematopoiesis a...

Full description

Bibliographic Details
Main Authors: Tong Xing, Zhong-Shi Lyu, Cai-Wen Duan, Hong-Yan Zhao, Shu-Qian Tang, Qi Wen, Yuan-Yuan Zhang, Meng Lv, Yu Wang, Lan-Ping Xu, Xiao-Hui Zhang, Xiao-Jun Huang, Yuan Kong
Format: Article
Language:English
Published: BMC 2022-03-01
Series:Journal of Translational Medicine
Subjects:
Online Access:https://doi.org/10.1186/s12967-022-03354-2
_version_ 1811280211501645824
author Tong Xing
Zhong-Shi Lyu
Cai-Wen Duan
Hong-Yan Zhao
Shu-Qian Tang
Qi Wen
Yuan-Yuan Zhang
Meng Lv
Yu Wang
Lan-Ping Xu
Xiao-Hui Zhang
Xiao-Jun Huang
Yuan Kong
author_facet Tong Xing
Zhong-Shi Lyu
Cai-Wen Duan
Hong-Yan Zhao
Shu-Qian Tang
Qi Wen
Yuan-Yuan Zhang
Meng Lv
Yu Wang
Lan-Ping Xu
Xiao-Hui Zhang
Xiao-Jun Huang
Yuan Kong
author_sort Tong Xing
collection DOAJ
description Abstract Background Myelodysplastic syndromes (MDS) are a group of heterogeneous myeloid clonal disorders characterized by ineffective haematopoiesis and immune deregulation. Emerging evidence has shown the effect of bone marrow (BM) endothelial progenitor cells (EPCs) in regulating haematopoiesis and immune balance. However, the number and functions of BM EPCs in patients with different stages of MDS remain largely unknown. Methods Patients with MDS (N = 30), de novo acute myeloid leukaemia (AML) (N = 15), and healthy donors (HDs) (N = 15) were enrolled. MDS patients were divided into lower-risk MDS (N = 15) and higher-risk MDS (N = 15) groups according to the dichotomization of the Revised International Prognostic Scoring System. Flow cytometry was performed to analyse the number of BM EPCs. Tube formation and migration assays were performed to evaluate the functions of BM EPCs. In order to assess the gene expression profiles of BM EPCs, RNA sequencing (RNA-seq) were performed. BM EPC supporting abilities of haematopoietic stem cells (HSCs), leukaemia cells and T cells were assessed by in vitro coculture experiments. Results Increased but dysfunctional BM EPCs were found in MDS patients compared with HDs, especially in patients with higher-risk MDS. RNA-seq indicated the progressive change and differences of haematopoiesis- and immune-related pathways and genes in MDS BM EPCs. In vitro coculture experiments verified that BM EPCs from HDs, lower-risk MDS, and higher-risk MDS to AML exhibited a progressively decreased ability to support HSCs, manifested as elevated apoptosis rates and intracellular reactive oxygen species (ROS) levels and decreased colony-forming unit plating efficiencies of HSCs. Moreover, BM EPCs from higher-risk MDS patients demonstrated an increased ability to support leukaemia cells, characterized by increased proliferation, leukaemia colony-forming unit plating efficiencies, decreased apoptosis rates and apoptosis-related genes. Furthermore, BM EPCs induced T cell differentiation towards more immune-tolerant cells in higher-risk MDS patients in vitro. In addition, the levels of intracellular ROS and the apoptosis ratios were increased in BM EPCs from MDS patients, especially in higher-risk MDS patients, which may be therapeutic candidates for MDS patients. Conclusion Our results suggest that dysfunctional BM EPCs are involved in MDS patients, which indicates that improving haematopoiesis supporting ability and immuneregulation ability of BM EPCs may represent a promising therapeutic approach for MDS patients.
first_indexed 2024-04-13T01:09:30Z
format Article
id doaj.art-0616e80e7f6d47c58bb61dd8c0c26ef3
institution Directory Open Access Journal
issn 1479-5876
language English
last_indexed 2024-04-13T01:09:30Z
publishDate 2022-03-01
publisher BMC
record_format Article
series Journal of Translational Medicine
spelling doaj.art-0616e80e7f6d47c58bb61dd8c0c26ef32022-12-22T03:09:11ZengBMCJournal of Translational Medicine1479-58762022-03-0120111710.1186/s12967-022-03354-2Dysfunctional bone marrow endothelial progenitor cells are involved in patients with myelodysplastic syndromesTong Xing0Zhong-Shi Lyu1Cai-Wen Duan2Hong-Yan Zhao3Shu-Qian Tang4Qi Wen5Yuan-Yuan Zhang6Meng Lv7Yu Wang8Lan-Ping Xu9Xiao-Hui Zhang10Xiao-Jun Huang11Yuan Kong12Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking UniversityPeking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking UniversityKey Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute, Shanghai Children’s Medical Center, Shanghai Collaborative Innovation Center for Translational Medicine and Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of MedicinePeking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking UniversityPeking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking UniversityPeking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking UniversityPeking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking UniversityPeking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking UniversityPeking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking UniversityPeking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking UniversityPeking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking UniversityPeking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking UniversityPeking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Collaborative Innovation Center of Hematology, Peking UniversityAbstract Background Myelodysplastic syndromes (MDS) are a group of heterogeneous myeloid clonal disorders characterized by ineffective haematopoiesis and immune deregulation. Emerging evidence has shown the effect of bone marrow (BM) endothelial progenitor cells (EPCs) in regulating haematopoiesis and immune balance. However, the number and functions of BM EPCs in patients with different stages of MDS remain largely unknown. Methods Patients with MDS (N = 30), de novo acute myeloid leukaemia (AML) (N = 15), and healthy donors (HDs) (N = 15) were enrolled. MDS patients were divided into lower-risk MDS (N = 15) and higher-risk MDS (N = 15) groups according to the dichotomization of the Revised International Prognostic Scoring System. Flow cytometry was performed to analyse the number of BM EPCs. Tube formation and migration assays were performed to evaluate the functions of BM EPCs. In order to assess the gene expression profiles of BM EPCs, RNA sequencing (RNA-seq) were performed. BM EPC supporting abilities of haematopoietic stem cells (HSCs), leukaemia cells and T cells were assessed by in vitro coculture experiments. Results Increased but dysfunctional BM EPCs were found in MDS patients compared with HDs, especially in patients with higher-risk MDS. RNA-seq indicated the progressive change and differences of haematopoiesis- and immune-related pathways and genes in MDS BM EPCs. In vitro coculture experiments verified that BM EPCs from HDs, lower-risk MDS, and higher-risk MDS to AML exhibited a progressively decreased ability to support HSCs, manifested as elevated apoptosis rates and intracellular reactive oxygen species (ROS) levels and decreased colony-forming unit plating efficiencies of HSCs. Moreover, BM EPCs from higher-risk MDS patients demonstrated an increased ability to support leukaemia cells, characterized by increased proliferation, leukaemia colony-forming unit plating efficiencies, decreased apoptosis rates and apoptosis-related genes. Furthermore, BM EPCs induced T cell differentiation towards more immune-tolerant cells in higher-risk MDS patients in vitro. In addition, the levels of intracellular ROS and the apoptosis ratios were increased in BM EPCs from MDS patients, especially in higher-risk MDS patients, which may be therapeutic candidates for MDS patients. Conclusion Our results suggest that dysfunctional BM EPCs are involved in MDS patients, which indicates that improving haematopoiesis supporting ability and immuneregulation ability of BM EPCs may represent a promising therapeutic approach for MDS patients.https://doi.org/10.1186/s12967-022-03354-2Myelodysplastic syndromesEndothelial progenitor cellsHaematopoiesisImmune regulation
spellingShingle Tong Xing
Zhong-Shi Lyu
Cai-Wen Duan
Hong-Yan Zhao
Shu-Qian Tang
Qi Wen
Yuan-Yuan Zhang
Meng Lv
Yu Wang
Lan-Ping Xu
Xiao-Hui Zhang
Xiao-Jun Huang
Yuan Kong
Dysfunctional bone marrow endothelial progenitor cells are involved in patients with myelodysplastic syndromes
Journal of Translational Medicine
Myelodysplastic syndromes
Endothelial progenitor cells
Haematopoiesis
Immune regulation
title Dysfunctional bone marrow endothelial progenitor cells are involved in patients with myelodysplastic syndromes
title_full Dysfunctional bone marrow endothelial progenitor cells are involved in patients with myelodysplastic syndromes
title_fullStr Dysfunctional bone marrow endothelial progenitor cells are involved in patients with myelodysplastic syndromes
title_full_unstemmed Dysfunctional bone marrow endothelial progenitor cells are involved in patients with myelodysplastic syndromes
title_short Dysfunctional bone marrow endothelial progenitor cells are involved in patients with myelodysplastic syndromes
title_sort dysfunctional bone marrow endothelial progenitor cells are involved in patients with myelodysplastic syndromes
topic Myelodysplastic syndromes
Endothelial progenitor cells
Haematopoiesis
Immune regulation
url https://doi.org/10.1186/s12967-022-03354-2
work_keys_str_mv AT tongxing dysfunctionalbonemarrowendothelialprogenitorcellsareinvolvedinpatientswithmyelodysplasticsyndromes
AT zhongshilyu dysfunctionalbonemarrowendothelialprogenitorcellsareinvolvedinpatientswithmyelodysplasticsyndromes
AT caiwenduan dysfunctionalbonemarrowendothelialprogenitorcellsareinvolvedinpatientswithmyelodysplasticsyndromes
AT hongyanzhao dysfunctionalbonemarrowendothelialprogenitorcellsareinvolvedinpatientswithmyelodysplasticsyndromes
AT shuqiantang dysfunctionalbonemarrowendothelialprogenitorcellsareinvolvedinpatientswithmyelodysplasticsyndromes
AT qiwen dysfunctionalbonemarrowendothelialprogenitorcellsareinvolvedinpatientswithmyelodysplasticsyndromes
AT yuanyuanzhang dysfunctionalbonemarrowendothelialprogenitorcellsareinvolvedinpatientswithmyelodysplasticsyndromes
AT menglv dysfunctionalbonemarrowendothelialprogenitorcellsareinvolvedinpatientswithmyelodysplasticsyndromes
AT yuwang dysfunctionalbonemarrowendothelialprogenitorcellsareinvolvedinpatientswithmyelodysplasticsyndromes
AT lanpingxu dysfunctionalbonemarrowendothelialprogenitorcellsareinvolvedinpatientswithmyelodysplasticsyndromes
AT xiaohuizhang dysfunctionalbonemarrowendothelialprogenitorcellsareinvolvedinpatientswithmyelodysplasticsyndromes
AT xiaojunhuang dysfunctionalbonemarrowendothelialprogenitorcellsareinvolvedinpatientswithmyelodysplasticsyndromes
AT yuankong dysfunctionalbonemarrowendothelialprogenitorcellsareinvolvedinpatientswithmyelodysplasticsyndromes