Targeted Rapamycin Delivery via Magnetic Nanoparticles to Address Stenosis in a 3D Bioprinted in Vitro Model of Pulmonary Veins

Author:

Ning Liqun12,Zanella Stefano1,Tomov Martin L.1,Amoli Mehdi Salar1,Jin Linqi1,Hwang Boeun1,Saadeh Maher1,Chen Huang1,Neelakantan Sunder3,Dasi Lakshmi Prasad1,Avazmohammadi Reza34,Mahmoudi Morteza5,Bauser‐Heaton Holly D.1678,Serpooshan Vahid167ORCID

Affiliation:

1. Wallace H. Coulter Department of Biomedical Engineering Emory University School of Medicine and Georgia Institute of Technology Atlanta GA 30322 USA

2. Department of Mechanical Engineering Cleveland State University Cleveland OH 44115 USA

3. Department of Biomedical Engineering Texas A&M University College Station TX 77843 USA

4. J. Mike Walker ’66 Department of Mechanical Engineering Texas A&M University College Station TX 77840 USA

5. Department of Radiology and Precision Health Program Michigan State University East Landing MI 48824 USA

6. Department of Pediatrics Emory University School of Medicine Atlanta GA 30322 USA

7. Children's Healthcare of Atlanta Atlanta GA 30322 USA

8. Sibley Heart Center at Children's Healthcare of Atlanta Atlanta GA 30322 USA

Abstract

AbstractVascular cell overgrowth and lumen size reduction in pulmonary vein stenosis (PVS) can result in elevated PV pressure, pulmonary hypertension, cardiac failure, and death. Administration of chemotherapies such as rapamycin have shown promise by inhibiting the vascular cell proliferation; yet clinical success is limited due to complications such as restenosis and off‐target effects. The lack of in vitro models to recapitulate the complex pathophysiology of PVS has hindered the identification of disease mechanisms and therapies. This study integrated 3D bioprinting, functional nanoparticles, and perfusion bioreactors to develop a novel in vitro model of PVS. Bioprinted bifurcated PV constructs are seeded with endothelial cells (ECs) and perfused, demonstrating the formation of a uniform and viable endothelium. Computational modeling identified the bifurcation point at high risk of EC overgrowth. Application of an external magnetic field enabled targeting of the rapamycin‐loaded superparamagnetic iron oxide nanoparticles at the bifurcation site, leading to a significant reduction in EC proliferation with no adverse side effects. These results establish a 3D bioprinted in vitro model to study PV homeostasis and diseases, offering the potential for increased throughput, tunability, and patient specificity, to test new or more effective therapies for PVS and other vascular diseases.

Funder

National Institutes of Health

National Science Foundation

Publisher

Wiley

Cited by 1 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