Glucocorticoid Receptors Drive Breast Cancer Cell Migration and Metabolic Reprogramming via PDK4

Author:

Dwyer Amy R12ORCID,Perez Kerkvliet Carlos1,Truong Thu H1ORCID,Hagen Kyla M1,Krutilina Raisa I3,Parke Deanna N3,Oakley Robert H4,Liddle Christopher5,Cidlowski John A4,Seagroves Tiffany N3,Lange Carol A16ORCID

Affiliation:

1. Masonic Cancer Center, University of Minnesota , Minneapolis, MN, 55455 , USA

2. Dame Roma Mitchell Cancer Research Laboratories, University of Adelaide , Adelaide, SA 5005 , Australia

3. Department of Pathology and Laboratory Medicine and Center for Cancer Research, College of Medicine, University of Tennessee Health Science Center , Memphis, TN, 38163 , USA

4. Signal Transduction Laboratory, Department of Health and Human Services, National Institute of Environmental Health Sciences, National Institutes of Health , Research Triangle Park, NC, 27709 , USA

5. Storr Liver Centre, The Westmead Institute for Medical Research and University of Sydney School of Medicine , Darlington, NSW, 2006 , Australia

6. Departments of Medicine (Division of Hematology, Oncology, and Transplantation) and Pharmacology, University of Minnesota , Minneapolis, MN, 55455 , USA

Abstract

Abstract Corticosteroids act on the glucocorticoid receptor (GR; NR3C1) to resolve inflammation and are routinely prescribed to breast cancer patients undergoing chemotherapy treatment to alleviate side effects. Triple-negative breast cancers (TNBCs) account for 15% to 20% of diagnoses and lack expression of estrogen and progesterone receptors as well as amplified HER2, but they often express high GR levels. GR is a mediator of TNBC progression to advanced metastatic disease; however, the mechanisms underpinning this transition to more aggressive behavior remain elusive. We previously showed that tissue/cellular stress (hypoxia, chemotherapies) as well as factors in the tumor microenvironment (transforming growth factor β [TGF-β], hepatocyte growth factor [HGF]) activate p38 mitogen-activated protein kinase (MAPK), which phosphorylates GR on Ser134. In the absence of ligand, pSer134-GR further upregulates genes important for responses to cellular stress, including key components of the p38 MAPK pathway. Herein, we show that pSer134-GR is required for TNBC metastatic colonization to the lungs of female mice. To understand the mechanisms of pSer134-GR action in the presence of GR agonists, we examined glucocorticoid-driven transcriptomes in CRISPR knock-in models of TNBC cells expressing wild-type or phospho-mutant (S134A) GR. We identified dexamethasone- and pSer134-GR-dependent regulation of specific gene sets controlling TNBC migration (NEDD9, CSF1, RUNX3) and metabolic adaptation (PDK4, PGK1, PFKFB4). TNBC cells harboring S134A-GR displayed metabolic reprogramming that was phenocopied by pyruvate dehydrogenase kinase 4 (PDK4) knockdown. PDK4 knockdown or chemical inhibition also blocked cancer cell migration. Our results reveal a convergence of GR agonists (ie, host stress) with cellular stress signaling whereby pSer134-GR critically regulates TNBC metabolism, an exploitable target for the treatment of this deadly disease.

Funder

National Institutes of Health's

National Center for Advancing Translational Sciences

Intramural Research Program

NIEHS

Breast Cancer Research

Metavivor Early Investigator Award

National Breast Cancer Foundation

Fellowship Award

Publisher

The Endocrine Society

Subject

Endocrinology

Cited by 4 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