The mRNA m6A reader YTHDF2 suppresses proinflammatory pathways and sustains hematopoietic stem cell function

Author:

Mapperley Christopher12ORCID,van de Lagemaat Louie N.12ORCID,Lawson Hannah2ORCID,Tavosanis Andrea2ORCID,Paris Jasmin12ORCID,Campos Joana2ORCID,Wotherspoon David2ORCID,Durko Jozef2ORCID,Sarapuu Annika2ORCID,Choe Junho34ORCID,Ivanova Ivayla15ORCID,Krause Daniela S.6ORCID,von Kriegsheim Alex7ORCID,Much Christian15ORCID,Morgan Marcos15ORCID,Gregory Richard I.34ORCID,Mead Adam J.8ORCID,O’Carroll Dónal159ORCID,Kranc Kamil R.12ORCID

Affiliation:

1. Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK

2. Laboratory of Haematopoietic Stem Cell and Leukaemia Biology, Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK

3. Stem Cell Program, Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA

4. Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA

5. Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK

6. Georg-Speyer-Haus and Goethe University, Frankfurt, Germany

7. Edinburgh Cancer Research UK Centre, Institute of Genetics and Molecular Medicine, Edinburgh, UK

8. Medical Research Council Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford, UK

9. Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK

Abstract

The mRNA N6-methyladenosine (m6A) modification has emerged as an essential regulator of normal and malignant hematopoiesis. Inactivation of the m6A mRNA reader YTHDF2, which recognizes m6A-modified transcripts to promote m6A-mRNA degradation, results in hematopoietic stem cell (HSC) expansion and compromises acute myeloid leukemia. Here we investigate the long-term impact of YTHDF2 deletion on HSC maintenance and multilineage hematopoiesis. We demonstrate that Ythdf2-deficient HSCs from young mice fail upon serial transplantation, display increased abundance of multiple m6A-modified inflammation-related transcripts, and chronically activate proinflammatory pathways. Consistent with the detrimental consequences of chronic activation of inflammatory pathways in HSCs, hematopoiesis-specific Ythdf2 deficiency results in a progressive myeloid bias, loss of lymphoid potential, HSC expansion, and failure of aged Ythdf2-deficient HSCs to reconstitute multilineage hematopoiesis. Experimentally induced inflammation increases YTHDF2 expression, and YTHDF2 is required to protect HSCs from this insult. Thus, our study positions YTHDF2 as a repressor of inflammatory pathways in HSCs and highlights the significance of m6A in long-term HSC maintenance.

Funder

Cancer Research UK

Medical Research Council

Blood Cancer UK

Barts Charity

Kay Kendall Leukaemia Fund

Wellcome Trust

Wellcome Centre for Cell Biology

European Cooperation in Science and Technology

Publisher

Rockefeller University Press

Subject

Immunology,Immunology and Allergy

Cited by 91 articles. 订阅此论文施引文献 订阅此论文施引文献,注册后可以免费订阅5篇论文的施引文献,订阅后可以查看论文全部施引文献

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3