Progesterone Receptors Promote Quiescence and Ovarian Cancer Cell Phenotypes via DREAM in p53-Mutant Fallopian Tube Models

Author:

Mauro Laura J12ORCID,Seibel Megan I1,Diep Caroline H1,Spartz Angela1,Perez Kerkvliet Carlos1,Singhal Hari3,Swisher Elizabeth M4,Schwartz Lauren E5,Drapkin Ronny6,Saini Siddharth7,Sesay Fatmata7,Litovchick Larisa7,Lange Carol A18ORCID

Affiliation:

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

2. University of Minnesota, Department of Animal Science, St. Paul, MN 55108, USA

3. Northwestern University, Department of Surgery, Feinberg School of Medicine, Chicago, IL 60611, USA

4. University of Washington Seattle, Dept Obstetrics & Gynecology, Division of Gynecologic Oncology, Seattle, WA 98109, USA

5. University of Pennsylvania, Dept of Pathology and Laboratory Medicine, Philadelphia, PA 19104, USA

6. University of Pennsylvania, Penn Ovarian Cancer Research Center, Dept Obstetrics & Gynecology, Philadelphia, PA 19104, USA

7. Virginia Commonwealth University, Massey Cancer Center, Dept. Internal Medicine, Division of Hematology, Oncology & Palliative Care, Richmond, VA 23298, USA

8. University of Minnesota, Dept Medicine, Division of Hematology, Oncology & Transplantation, Minneapolis, MN 55455, USA

Abstract

Abstract Context The ability of ovarian steroids to modify ovarian cancer (OC) risk remains controversial. Progesterone is considered to be protective; recent studies indicate no effect or enhanced OC risk. Knowledge of progesterone receptor (PR) signaling during altered physiology that typifies OC development is limited. Objective This study defines PR-driven oncogenic signaling mechanisms in p53-mutant human fallopian tube epithelia (hFTE), a precursor of the most aggressive OC subtype. Methods PR expression in clinical samples of serous tubal intraepithelial carcinoma (STIC) lesions and high-grade serous OC (HGSC) tumors was analyzed. Novel PR-A and PR-B isoform-expressing hFTE models were characterized for gene expression and cell cycle progression, emboli formation, and invasion. PR regulation of the DREAM quiescence complex and DYRK1 kinases was established. Results STICs and HGSC express abundant activated phospho-PR. Progestin promoted reversible hFTE cell cycle arrest, spheroid formation, and invasion. RNAseq/biochemical studies revealed potent ligand-independent/-dependent PR actions, progestin-induced regulation of the DREAM quiescence complex, and cell cycle target genes through enhanced complex formation and chromatin recruitment. Disruption of DREAM/DYRK1s by pharmacological inhibition, HPV E6/E7 expression, or DYRK1A/B depletion blocked progestin-induced cell arrest and attenuated PR-driven gene expression and associated OC phenotypes. Conclusion Activated PRs support quiescence and pro-survival/pro-dissemination cell behaviors that may contribute to early HGSC progression. Our data support an alternative perspective on the tenet that progesterone always confers protection against OC. STICs can reside undetected for decades prior to invasive disease; our studies reveal clinical opportunities to prevent the ultimate development of HGSC by targeting PRs, DREAM, and/or DYRKs.

Funder

Minnesota Ovarian Cancer Alliance

University of Minnesota

National Institutes of Health

National Cancer Institute

Dr. Miriam and Sheldon G. Adelson Medical Research Foundation

Claneil Foundation

Publisher

The Endocrine Society

Subject

Biochemistry (medical),Clinical Biochemistry,Endocrinology,Biochemistry,Endocrinology, Diabetes and Metabolism

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