Matrix degradation and cell proliferation are coupled to promote invasion and escape from an engineered human breast microtumor

Author:

Rabie Emann M12,Zhang Sherry X3,Kourouklis Andreas P3,Kilinc A Nihan3,Simi Allison K3,Radisky Derek C4,Tien Joe56,Nelson Celeste M23

Affiliation:

1. Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA

2. Department of Molecular Biology, Princeton University, Princeton, NJ, USA

3. Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ, USA

4. Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA

5. Department of Biomedical Engineering, Boston University, Boston, MA, USA

6. Division of Materials Science and Engineering, Boston University, Boston, MA, USA

Abstract

Abstract Metastasis, the leading cause of mortality in cancer patients, depends upon the ability of cancer cells to invade into the extracellular matrix that surrounds the primary tumor and to escape into the vasculature. To investigate the features of the microenvironment that regulate invasion and escape, we generated solid microtumors of MDA-MB-231 human breast carcinoma cells within gels of type I collagen. The microtumors were formed at defined distances adjacent to an empty cavity, which served as an artificial vessel into which the constituent tumor cells could escape. To define the relative contributions of matrix degradation and cell proliferation on invasion and escape, we used pharmacological approaches to block the activity of matrix metalloproteinases (MMPs) or to arrest the cell cycle. We found that blocking MMP activity prevents both invasion and escape of the breast cancer cells. Surprisingly, blocking proliferation increases the rate of invasion but has no effect on that of escape. We found that arresting the cell cycle increases the expression of MMPs, consistent with the increased rate of invasion. To gain additional insight into the role of cell proliferation in the invasion process, we generated microtumors from cells that express the fluorescent ubiquitination-based cell cycle indicator. We found that the cells that initiate invasions are preferentially quiescent, whereas cell proliferation is associated with the extension of invasions. These data suggest that matrix degradation and cell proliferation are coupled during the invasion and escape of human breast cancer cells and highlight the critical role of matrix proteolysis in governing tumor phenotype.

Funder

National Cancer Institute

National Institutes of Health

National Research Service Award fellowship

National Institute of General Medical Sciences

Publisher

Oxford University Press (OUP)

Subject

Biochemistry,Biophysics

Cited by 7 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

1. TME-Related Biomimetic Strategies Against Cancer;International Journal of Nanomedicine;2024-01

2. Protein-Based Microfluidic Models for Biomedical Applications;Handbook of the Extracellular Matrix;2023

3. Cell Patterning to Mimic Tumor Anatomy;Biomaterial Based Approaches to Study the Tumour Microenvironment;2022-12-07

4. Role of Lymphatic Endothelium in Vascular Escape of Engineered Human Breast Microtumors;Cellular and Molecular Bioengineering;2022-11-07

5. Exosomal MMP-1 transfers metastasis potential in triple-negative breast cancer through PAR1-mediated EMT;Breast Cancer Research and Treatment;2022-03-07

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