Small Extracellular Vesicles from Failing Heart Accelerate Tumor Growth

Author:

Caller Tal,Rotem Itai,Shaihov–Teper Olga,Lendengolts Daria,Schary Yeshai,Shai Ruty,Glick-Saar Efrat,Dominissini Dan,Motie Menachem,Katzir Idan,Popovtzer Rachela,Nahmoud Merav,Boomgarden Alex,D’Souza-Schorey Crislyn,Naftali-Shani Nili,Leor Jonathan

Abstract

ABSTRACTBackgroundMyocardial infarction (MI) and heart failure (HF) are associated with an increased incidence of cancer. The mechanism is complex and unclear. Here, we aimed to test our hypothesis that cardiac small extracellular vesicles (sEVs), particularly cardiac mesenchymal stromal cells-derived sEVs (cMSC-sEVs), contribute to the link between post-MI HF and cancer.MethodsWe purified and characterized sEVs from the whole heart and cultured cMSCs. Then, we analyzed cMSC-EV cargo and pro-neoplastic effects on several types of cancer cell lines, macrophages, and endothelial cells. Next, we modeled post-MI HF along with heterotopic and orthotopic lung and breast cancer tumors in mice. We used cMSC-sEV transfer to assess sEV biodistribution and its effect on tumor growth. Finally, we tested the effects of sEV depletion and spironolactone treatment on cMSC-EV release and tumor growth.ResultsPost-MI hearts, particularly cMSCs, produced more sEVs with pro-neoplastic cargo than non-failing hearts did. Proteomic analysis revealed unique protein profiles and higher quantities of tumor-promoting cytokines, proteins, and microRNAs in cMSC-sEVs from failing hearts. The pro-neoplastic effects of cMSC-sEVs varied with different types of cancer cells, substantially affecting lung cancer cells relative to other more aggressive cancer cell lines. We also found that post-MI cMSC-sEVs activated resting macrophages into pro-angiogenic and pro-tumorigenic states in vitro. At 28-day follow-up analysis, mice with post-MI HF developed larger lung tumors than did sham-MI mice. Adoptive transfer of cMSC-sEVs from failing hearts accelerated lung tumor growth, and biodistribution analysis revealed an accumulating cMSC- sEVs in tumor cells along with accelerated tumor cell proliferation. Significantly, sEV depletion reduced the tumor-promoting effects of HF, and adoptive transfer of cMSC-sEVs from failing hearts partially restored it. Finally, post-MI spironolactone treatment reduced the number of cMSC-sEVs and suppressed tumor growth during post-MI HF.ConclusionsFor the first time, we show that cardiac sEVs, specifically cMSC-sEVs from post-MI failing hearts, carry multiple pro-tumorigenic factors. Uptake of cMSC-sEVs by cancer cells accelerates tumor growth. Post-MI spironolactone treatment reduces the associated tumor growth. Thus, we provide new insight into the link between post-MI HF and cancer and propose a translational option to mitigate this deadly association.

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