SUMOylation Regulates Growth Factor Independence 1 in Transcriptional Control and Hematopoiesis

Author:

Andrade Daniel1,Velinder Matthew1,Singer Jason1,Maese Luke23,Bareyan Diana1,Nguyen Hong4,Chandrasekharan Mahesh B.5,Lucente Helena1,McClellan David1,Jones David1,Sharma Sunil67,Liu Fang8,Engel Michael E.1239

Affiliation:

1. Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, Utah, USA

2. Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, USA

3. Primary Children's Hospital, Salt Lake City, Utah, USA

4. Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA

5. Department of Radiation Oncology, University of Utah School of Medicine, Salt Lake City, Utah, USA

6. Department of Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA

7. Center for Investigational Therapeutics, Huntsman Cancer Institute, Salt Lake City, Utah, USA

8. Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA

9. Nuclear Control Program, Huntsman Cancer Institute, Salt Lake City, Utah, USA

Abstract

ABSTRACT Cell fate specification requires precise coordination of transcription factors and their regulators to achieve fidelity and flexibility in lineage allocation. The transcriptional repressor growth factor independence 1 (GFI1) is comprised of conserved Snail/Slug/Gfi1 (SNAG) and zinc finger motifs separated by a linker region poorly conserved with GFI1B, its closest homolog. Moreover, GFI1 and GFI1B coordinate distinct developmental fates in hematopoiesis, suggesting that their functional differences may derive from structures within their linkers. We show a binding interface between the GFI1 linker and the SP-RING domain of PIAS3, an E3-SUMO (small ubiquitin-related modifier) ligase. The PIAS3 binding region in GFI1 contains a conserved type I SUMOylation consensus element, centered on lysine-239 (K239). In silico prediction algorithms identify K239 as the only high-probability site for SUMO modification. We show that GFI1 is modified by SUMO at K239. SUMOylation-resistant derivatives of GFI1 fail to complement Gfi1 depletion phenotypes in zebrafish primitive erythropoiesis and granulocytic differentiation in cultured human cells. LSD1/CoREST recruitment and MYC repression by GFI1 are profoundly impaired for SUMOylation-resistant GFI1 derivatives, while enforced expression of MYC blocks granulocytic differentiation. These findings suggest that SUMOylation within the GFI1 linker favors LSD1/CoREST recruitment and MYC repression to govern hematopoietic differentiation.

Funder

HHS | NIH | National Cancer Institute

HHS | NIH | National Institute of Diabetes and Digestive and Kidney Diseases

St. Baldrick's Foundation

Publisher

American Society for Microbiology

Subject

Cell Biology,Molecular Biology

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