Differential chromatin accessibility and transcriptional dynamics define breast cancer subtypes and their lineages

Author:

Iglesia Michael D.,Jayasinghe Reyka G.,Chen Siqi,Terekhanova Nadezhda V.,Herndon John M.,Storrs Erik,Karpova Alla,Zhou Daniel Cui,Al Deen Nataly Naser,Shinkle Andrew T.,Lu Rita Jui-Hsien,Caravan Wagma,Houston Andrew,Zhao Yanyan,Sato Kazuhito,Lal Preet,Street Cherease,Rodrigues Fernanda Martins,Southard-Smith Austin N.,Targino da Costa André Luiz N.,Zhu Houxiang,Mo Chia-Kuei,Crowson Lisa,Fulton Robert S.,Wyczalkowski Matthew A.,Fronick Catrina C.,Fulton Lucinda A.,Sun Hua,Davies Sherri R.,Appelbaum Elizabeth L.,Chasnoff Sara E.,Carmody Madelyn,Brooks Candace,Liu Ruiyang,Wendl Michael C.,Oh Clara,Bender Diane,Cruchaga CarlosORCID,Harari Oscar,Bredemeyer Andrea,Lavine Kory,Bose Ron,Margenthaler Julie,Held Jason M.,Achilefu Samuel,Ademuyiwa Foluso,Aft Rebecca,Ma Cynthia,Colditz Graham A.,Ju Tao,Oh Stephen T.ORCID,Fitzpatrick James,Hwang E. Shelley,Shoghi Kooresh I.,Chheda Milan G.,Veis Deborah J.,Chen Feng,Fields Ryan C.,Gillanders William E.,Ding Li

Abstract

ABSTRACTBreast cancer is a heterogeneous disease, and treatment is guided by biomarker profiles representing distinct molecular subtypes. Breast cancer arises from the breast ductal epithelium, and experimental data suggests breast cancer subtypes have different cells of origin within that lineage. The precise cells of origin for each subtype and the transcriptional networks that characterize these tumor-normal lineages are not established. In this work, we applied bulk, single-cell (sc), and single-nucleus (sn) multi-omic techniques as well as spatial transcriptomics and multiplex imaging on 61 samples from 37 breast cancer patients to show characteristic links in gene expression and chromatin accessibility between breast cancer subtypes and their putative cells of origin. We applied the PAM50 subtyping algorithm in tandem with bulk RNA-seq and snRNA-seq to reliably subtype even low-purity tumor samples and confirm promoter accessibility using snATAC. Trajectory analysis of chromatin accessibility and differentially accessible motifs clearly connected progenitor populations with breast cancer subtypes supporting the cell of origin for basal-like and luminal A and B tumors. Regulatory network analysis of transcription factors underscored the importance of BHLHE40 in luminal breast cancer and luminal mature cells, and KLF5 in basal-like tumors and luminal progenitor cells. Furthermore, we identify key genes defining the basal-like (PRKCA,SOX6,RGS6,KCNQ3) and luminal A/B (FAM155A,LRP1B) lineages, with expression in both precursor and cancer cells and further upregulation in tumors. Exhausted CTLA4-expressing CD8+ T cells were enriched in basal-like breast cancer, suggesting altered means of immune dysfunction among breast cancer subtypes. We used spatial transcriptomics and multiplex imaging to provide spatial detail for key markers of benign and malignant cell types and immune cell colocation. These findings demonstrate analysis of paired transcription and chromatin accessibility at the single cell level is a powerful tool for investigating breast cancer lineage development and highlight transcriptional networks that define basal and luminal breast cancer lineages.

Publisher

Cold Spring Harbor Laboratory

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