Integrated in silico and 3D in vitro model of macrophage migration in response to physical and chemical factors in the tumor microenvironment

Author:

Lee Sharon Wei Ling123ORCID,Seager R J4,Litvak Felix4,Spill Fabian456,Sieow Je Lin3,Leong Penny Hweixian3,Kumar Dillip3,Tan Alrina Shin Min3,Wong Siew Cheng23,Adriani Giulia3,Zaman Muhammad Hamid47,Kamm and Roger D58ORCID

Affiliation:

1. BioSystems and Micromechanics IRG, Singapore-MIT Alliance for Research and Technology, Singapore, 138602, Singapore

2. Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, 117597, Singapore

3. Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research (A*STAR), Singapore

4. Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA

5. Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA

6. School of Mathematics, University of Birmingham, Birmingham, B15 2TT, UK

7. Howard Hughes Medical Institute, Boston University, Boston, MA, 02215, USA

8. Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA

Abstract

AbstractMacrophages are abundant in the tumor microenvironment (TME), serving as accomplices to cancer cells for their invasion. Studies have explored the biochemical mechanisms that drive pro-tumor macrophage functions; however the role of TME interstitial flow (IF) is often disregarded. Therefore, we developed a three-dimensional microfluidic-based model with tumor cells and macrophages to study how IF affects macrophage migration and its potential contribution to cancer invasion. The presence of either tumor cells or IF individually increased macrophage migration directedness and speed. Interestingly, there was no additive effect on macrophage migration directedness and speed under the simultaneous presence of tumor cells and IF. Further, we present an in silico model that couples chemokine-mediated signaling with mechanosensing networks to explain our in vitro observations. In our model design, we propose IL-8, CCL2, and β-integrin as key pathways that commonly regulate various Rho GTPases. In agreement, in vitro macrophage migration remained elevated when exposed to a saturating concentration of recombinant IL-8 or CCL2 or to the co-addition of a sub-saturating concentration of both cytokines. Moreover, antibody blockade against IL-8 and/or CCL2 inhibited migration that could be restored by IF, indicating cytokine-independent mechanisms of migration induction. Importantly, we demonstrate the utility of an integrated in silico and 3D in vitro approach to aid the design of tumor-associated macrophage-based immunotherapeutic strategies.

Funder

National Cancer Institute

Biomedical Research Council

Publisher

Oxford University Press (OUP)

Subject

Biochemistry,Biophysics

Cited by 40 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