Intratumoral Heterogeneity Promotes Collective Cancer Invasion through NOTCH1 Variation

Cellular and molecular heterogeneity within tumors has long been associated with the progression of cancer to an aggressive phenotype and a poor prognosis. However, how such intratumoral heterogeneity contributes to the invasiveness of cancer is largely unknown. Here, using a tumor bioengineering ap...

Full description

Bibliographic Details
Main Authors: Peter Torab, Yue Yan, Mona Ahmed, Hironobu Yamashita, Joshua I. Warrick, Jay D. Raman, David J. DeGraff, Pak Kin Wong
Format: Article
Language:English
Published: MDPI AG 2021-11-01
Series:Cells
Subjects:
Online Access:https://www.mdpi.com/2073-4409/10/11/3084
_version_ 1797510810371620864
author Peter Torab
Yue Yan
Mona Ahmed
Hironobu Yamashita
Joshua I. Warrick
Jay D. Raman
David J. DeGraff
Pak Kin Wong
author_facet Peter Torab
Yue Yan
Mona Ahmed
Hironobu Yamashita
Joshua I. Warrick
Jay D. Raman
David J. DeGraff
Pak Kin Wong
author_sort Peter Torab
collection DOAJ
description Cellular and molecular heterogeneity within tumors has long been associated with the progression of cancer to an aggressive phenotype and a poor prognosis. However, how such intratumoral heterogeneity contributes to the invasiveness of cancer is largely unknown. Here, using a tumor bioengineering approach, we investigate the interaction between molecular subtypes within bladder microtumors and the corresponding effects on their invasiveness. Our results reveal heterogeneous microtumors formed by multiple molecular subtypes possess enhanced invasiveness compared to individual cells, even when both cells are not invasive individually. To examine the molecular mechanism of intratumoral heterogeneity mediated invasiveness, live single cell biosensing, RNA interference, and CRISPR-Cas9 gene editing approaches were applied to investigate and control the composition of the microtumors. An agent-based computational model was also developed to evaluate the influence of NOTCH1 variation on DLL4 expression within a microtumor. The data indicate that intratumoral variation in NOTCH1 expression can lead to upregulation of DLL4 expression within the microtumor and enhancement of microtumor invasiveness. Overall, our results reveal a novel mechanism of heterogeneity mediated invasiveness through intratumoral variation of gene expression.
first_indexed 2024-03-10T05:36:32Z
format Article
id doaj.art-9fd3d75d6c6547998b2dfb18ad573bc7
institution Directory Open Access Journal
issn 2073-4409
language English
last_indexed 2024-03-10T05:36:32Z
publishDate 2021-11-01
publisher MDPI AG
record_format Article
series Cells
spelling doaj.art-9fd3d75d6c6547998b2dfb18ad573bc72023-11-22T22:51:04ZengMDPI AGCells2073-44092021-11-011011308410.3390/cells10113084Intratumoral Heterogeneity Promotes Collective Cancer Invasion through NOTCH1 VariationPeter Torab0Yue Yan1Mona Ahmed2Hironobu Yamashita3Joshua I. Warrick4Jay D. Raman5David J. DeGraff6Pak Kin Wong7Department of Mechanical Engineering, The Pennsylvania State University, University Park, PA 16802, USADepartment of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USADepartment of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USADepartment of Pathology and Laboratory Medicine, The Pennsylvania State University, Hershey, PA 17033, USADepartment of Pathology and Laboratory Medicine, The Pennsylvania State University, Hershey, PA 17033, USAPenn State Health Milton S., Hershey Medical Center, Department of Surgery, Hershey, PA 17033, USADepartment of Pathology and Laboratory Medicine, The Pennsylvania State University, Hershey, PA 17033, USADepartment of Mechanical Engineering, The Pennsylvania State University, University Park, PA 16802, USACellular and molecular heterogeneity within tumors has long been associated with the progression of cancer to an aggressive phenotype and a poor prognosis. However, how such intratumoral heterogeneity contributes to the invasiveness of cancer is largely unknown. Here, using a tumor bioengineering approach, we investigate the interaction between molecular subtypes within bladder microtumors and the corresponding effects on their invasiveness. Our results reveal heterogeneous microtumors formed by multiple molecular subtypes possess enhanced invasiveness compared to individual cells, even when both cells are not invasive individually. To examine the molecular mechanism of intratumoral heterogeneity mediated invasiveness, live single cell biosensing, RNA interference, and CRISPR-Cas9 gene editing approaches were applied to investigate and control the composition of the microtumors. An agent-based computational model was also developed to evaluate the influence of NOTCH1 variation on DLL4 expression within a microtumor. The data indicate that intratumoral variation in NOTCH1 expression can lead to upregulation of DLL4 expression within the microtumor and enhancement of microtumor invasiveness. Overall, our results reveal a novel mechanism of heterogeneity mediated invasiveness through intratumoral variation of gene expression.https://www.mdpi.com/2073-4409/10/11/3084single cell analysistumor subtypesbasalluminaltumor-on-chipbiosensing
spellingShingle Peter Torab
Yue Yan
Mona Ahmed
Hironobu Yamashita
Joshua I. Warrick
Jay D. Raman
David J. DeGraff
Pak Kin Wong
Intratumoral Heterogeneity Promotes Collective Cancer Invasion through NOTCH1 Variation
Cells
single cell analysis
tumor subtypes
basal
luminal
tumor-on-chip
biosensing
title Intratumoral Heterogeneity Promotes Collective Cancer Invasion through NOTCH1 Variation
title_full Intratumoral Heterogeneity Promotes Collective Cancer Invasion through NOTCH1 Variation
title_fullStr Intratumoral Heterogeneity Promotes Collective Cancer Invasion through NOTCH1 Variation
title_full_unstemmed Intratumoral Heterogeneity Promotes Collective Cancer Invasion through NOTCH1 Variation
title_short Intratumoral Heterogeneity Promotes Collective Cancer Invasion through NOTCH1 Variation
title_sort intratumoral heterogeneity promotes collective cancer invasion through notch1 variation
topic single cell analysis
tumor subtypes
basal
luminal
tumor-on-chip
biosensing
url https://www.mdpi.com/2073-4409/10/11/3084
work_keys_str_mv AT petertorab intratumoralheterogeneitypromotescollectivecancerinvasionthroughnotch1variation
AT yueyan intratumoralheterogeneitypromotescollectivecancerinvasionthroughnotch1variation
AT monaahmed intratumoralheterogeneitypromotescollectivecancerinvasionthroughnotch1variation
AT hironobuyamashita intratumoralheterogeneitypromotescollectivecancerinvasionthroughnotch1variation
AT joshuaiwarrick intratumoralheterogeneitypromotescollectivecancerinvasionthroughnotch1variation
AT jaydraman intratumoralheterogeneitypromotescollectivecancerinvasionthroughnotch1variation
AT davidjdegraff intratumoralheterogeneitypromotescollectivecancerinvasionthroughnotch1variation
AT pakkinwong intratumoralheterogeneitypromotescollectivecancerinvasionthroughnotch1variation