In Vitro Organoid-Based Assays Reveal SMAD4 Tumor-Suppressive Mechanisms for Serrated Colorectal Cancer Invasion

Author:

Tong Kevin12345ORCID,Bandari Manisha1ORCID,Carrick Jillian N.14,Zenkevich Anastasia4,Kothari Om A.1,Shamshad Eman1,Stefanik Katarina16,Haro Katherine S.1,Perekatt Ansu O.17,Verzi Michael P.1238

Affiliation:

1. Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA

2. Human Genetics Institute of New Jersey, Piscataway, NJ 08854, USA

3. Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA

4. Hackensack Meridian Health Center for Discovery and Innovation, Nutley, NJ 07110, USA

5. Department of Medical Sciences, Hackensack Meridian Health School of Medicine, Nutley, NJ 07110, USA

6. Department of Biology, The College of New Jersey, Ewing Township, NJ 08618, USA

7. Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ 07030, USA

8. Rutgers Center for Lipid Research, New Brunswick, NJ 08901, USA

Abstract

Colon cancer is the third most prominent cancer and second leading cause of cancer-related deaths in the United States. Up to 20% of colon cancers follow the serrated tumor pathway driven by mutations in the MAPK pathway. Loss of SMAD4 function occurs in the majority of late-stage colon cancers and is associated with aggressive cancer progression. Therefore, it is important to develop technology to accurately model and better understand the genetic mechanisms behind cancer invasion. Organoids derived from tumors found in the Smad4KO BRAFV600E/+ mouse model present multiple phenotypes characteristic of invasion both in ex vivo and in vivo systems. Smad4KO BRAFV600E/+ tumor organoids can migrate through 3D culture and infiltrate through transwell membranes. This invasive behavior can be suppressed when SMAD4 is re-expressed in the tumor organoids. RNA-Seq analysis reveals that SMAD4 expression in organoids rapidly regulates transcripts associated with extracellular matrix and secreted proteins, suggesting that the mechanisms employed by SMAD4 to inhibit invasion are associated with regulation of extracellular matrix and secretory pathways. These findings indicate new models to study SMAD4 regulation of tumor invasion and an additional layer of complexity in the tumor-suppressive function of the SMAD4/Tgfβ pathway.

Funder

NCI

NIDDK

Rutgers Summer Undergraduate Research Fellowship Awards

Human Genetics Institute of New Jersey MacMillan Cancer Genetics Summer Undergraduate Research Fellowships

Publisher

MDPI AG

Subject

Cancer Research,Oncology

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