Cebp1 and Cebpβ transcriptional axis controls eosinophilopoiesis in zebrafish

Abstract Eosinophils are a group of granulocytes well known for their capacity to protect the host from parasites and regulate immune function. Diverse biological roles for eosinophils have been increasingly identified, but the developmental pattern and regulation of the eosinophil lineage remain la...

Full description

Bibliographic Details
Main Authors: Gaofei Li, Yicong Sun, Immanuel Kwok, Liting Yang, Wanying Wen, Peixian Huang, Mei Wu, Jing Li, Zhibin Huang, Zhaoyuan Liu, Shuai He, Wan Peng, Jin-Xin Bei, Florent Ginhoux, Lai Guan Ng, Yiyue Zhang
Format: Article
Language:English
Published: Nature Portfolio 2024-01-01
Series:Nature Communications
Online Access:https://doi.org/10.1038/s41467-024-45029-0
_version_ 1797276390820675584
author Gaofei Li
Yicong Sun
Immanuel Kwok
Liting Yang
Wanying Wen
Peixian Huang
Mei Wu
Jing Li
Zhibin Huang
Zhaoyuan Liu
Shuai He
Wan Peng
Jin-Xin Bei
Florent Ginhoux
Lai Guan Ng
Yiyue Zhang
author_facet Gaofei Li
Yicong Sun
Immanuel Kwok
Liting Yang
Wanying Wen
Peixian Huang
Mei Wu
Jing Li
Zhibin Huang
Zhaoyuan Liu
Shuai He
Wan Peng
Jin-Xin Bei
Florent Ginhoux
Lai Guan Ng
Yiyue Zhang
author_sort Gaofei Li
collection DOAJ
description Abstract Eosinophils are a group of granulocytes well known for their capacity to protect the host from parasites and regulate immune function. Diverse biological roles for eosinophils have been increasingly identified, but the developmental pattern and regulation of the eosinophil lineage remain largely unknown. Herein, we utilize the zebrafish model to analyze eosinophilic cell differentiation, distribution, and regulation. By identifying eslec as an eosinophil lineage-specific marker, we establish a Tg(eslec:eGFP) reporter line, which specifically labeled cells of the eosinophil lineage from early life through adulthood. Spatial-temporal analysis of eslec + cells demonstrates their organ distribution from larval stage to adulthood. By single-cell RNA-Seq analysis, we decipher the eosinophil lineage cells from lineage-committed progenitors to mature eosinophils. Through further genetic analysis, we demonstrate the role of Cebp1 in balancing neutrophil and eosinophil lineages, and a Cebp1-Cebpβ transcriptional axis that regulates the commitment and differentiation of the eosinophil lineage. Cross-species functional comparisons reveals that zebrafish Cebp1 is the functional orthologue of human C/EBPεP27 in suppressing eosinophilopoiesis. Our study characterizes eosinophil development in multiple dimensions including spatial-temporal patterns, expression profiles, and genetic regulators, providing for a better understanding of eosinophilopoiesis.
first_indexed 2024-03-07T15:27:37Z
format Article
id doaj.art-3fea63b6b24a49bba081a7c7051025dd
institution Directory Open Access Journal
issn 2041-1723
language English
last_indexed 2024-03-07T15:27:37Z
publishDate 2024-01-01
publisher Nature Portfolio
record_format Article
series Nature Communications
spelling doaj.art-3fea63b6b24a49bba081a7c7051025dd2024-03-05T16:36:00ZengNature PortfolioNature Communications2041-17232024-01-0115111510.1038/s41467-024-45029-0Cebp1 and Cebpβ transcriptional axis controls eosinophilopoiesis in zebrafishGaofei Li0Yicong Sun1Immanuel Kwok2Liting Yang3Wanying Wen4Peixian Huang5Mei Wu6Jing Li7Zhibin Huang8Zhaoyuan Liu9Shuai He10Wan Peng11Jin-Xin Bei12Florent Ginhoux13Lai Guan Ng14Yiyue Zhang15Department of Hematology, the Second Affiliated Hospital, School of Medicine, South China University of TechnologyInnovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of TechnologySingapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research)Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical UniversityInnovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of TechnologyInnovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of TechnologyInnovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of TechnologyInnovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of TechnologyInnovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of TechnologyShanghai Institute of Immunology, Shanghai JiaoTong University School of MedicineSun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineSun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineSun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineSingapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research)Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research)Department of Hematology, the Second Affiliated Hospital, School of Medicine, South China University of TechnologyAbstract Eosinophils are a group of granulocytes well known for their capacity to protect the host from parasites and regulate immune function. Diverse biological roles for eosinophils have been increasingly identified, but the developmental pattern and regulation of the eosinophil lineage remain largely unknown. Herein, we utilize the zebrafish model to analyze eosinophilic cell differentiation, distribution, and regulation. By identifying eslec as an eosinophil lineage-specific marker, we establish a Tg(eslec:eGFP) reporter line, which specifically labeled cells of the eosinophil lineage from early life through adulthood. Spatial-temporal analysis of eslec + cells demonstrates their organ distribution from larval stage to adulthood. By single-cell RNA-Seq analysis, we decipher the eosinophil lineage cells from lineage-committed progenitors to mature eosinophils. Through further genetic analysis, we demonstrate the role of Cebp1 in balancing neutrophil and eosinophil lineages, and a Cebp1-Cebpβ transcriptional axis that regulates the commitment and differentiation of the eosinophil lineage. Cross-species functional comparisons reveals that zebrafish Cebp1 is the functional orthologue of human C/EBPεP27 in suppressing eosinophilopoiesis. Our study characterizes eosinophil development in multiple dimensions including spatial-temporal patterns, expression profiles, and genetic regulators, providing for a better understanding of eosinophilopoiesis.https://doi.org/10.1038/s41467-024-45029-0
spellingShingle Gaofei Li
Yicong Sun
Immanuel Kwok
Liting Yang
Wanying Wen
Peixian Huang
Mei Wu
Jing Li
Zhibin Huang
Zhaoyuan Liu
Shuai He
Wan Peng
Jin-Xin Bei
Florent Ginhoux
Lai Guan Ng
Yiyue Zhang
Cebp1 and Cebpβ transcriptional axis controls eosinophilopoiesis in zebrafish
Nature Communications
title Cebp1 and Cebpβ transcriptional axis controls eosinophilopoiesis in zebrafish
title_full Cebp1 and Cebpβ transcriptional axis controls eosinophilopoiesis in zebrafish
title_fullStr Cebp1 and Cebpβ transcriptional axis controls eosinophilopoiesis in zebrafish
title_full_unstemmed Cebp1 and Cebpβ transcriptional axis controls eosinophilopoiesis in zebrafish
title_short Cebp1 and Cebpβ transcriptional axis controls eosinophilopoiesis in zebrafish
title_sort cebp1 and cebpβ transcriptional axis controls eosinophilopoiesis in zebrafish
url https://doi.org/10.1038/s41467-024-45029-0
work_keys_str_mv AT gaofeili cebp1andcebpbtranscriptionalaxiscontrolseosinophilopoiesisinzebrafish
AT yicongsun cebp1andcebpbtranscriptionalaxiscontrolseosinophilopoiesisinzebrafish
AT immanuelkwok cebp1andcebpbtranscriptionalaxiscontrolseosinophilopoiesisinzebrafish
AT litingyang cebp1andcebpbtranscriptionalaxiscontrolseosinophilopoiesisinzebrafish
AT wanyingwen cebp1andcebpbtranscriptionalaxiscontrolseosinophilopoiesisinzebrafish
AT peixianhuang cebp1andcebpbtranscriptionalaxiscontrolseosinophilopoiesisinzebrafish
AT meiwu cebp1andcebpbtranscriptionalaxiscontrolseosinophilopoiesisinzebrafish
AT jingli cebp1andcebpbtranscriptionalaxiscontrolseosinophilopoiesisinzebrafish
AT zhibinhuang cebp1andcebpbtranscriptionalaxiscontrolseosinophilopoiesisinzebrafish
AT zhaoyuanliu cebp1andcebpbtranscriptionalaxiscontrolseosinophilopoiesisinzebrafish
AT shuaihe cebp1andcebpbtranscriptionalaxiscontrolseosinophilopoiesisinzebrafish
AT wanpeng cebp1andcebpbtranscriptionalaxiscontrolseosinophilopoiesisinzebrafish
AT jinxinbei cebp1andcebpbtranscriptionalaxiscontrolseosinophilopoiesisinzebrafish
AT florentginhoux cebp1andcebpbtranscriptionalaxiscontrolseosinophilopoiesisinzebrafish
AT laiguanng cebp1andcebpbtranscriptionalaxiscontrolseosinophilopoiesisinzebrafish
AT yiyuezhang cebp1andcebpbtranscriptionalaxiscontrolseosinophilopoiesisinzebrafish