Cargando…

Multiparameter Evaluation of the Heterogeneity of Circulating Tumor Cells Using Integrated RNA In Situ Hybridization and Immunocytochemical Analysis

Circulating tumor cells (CTCs) are routinely identified as cytokeratin (CK)-positive, epithelial cell adhesion molecule (EpCAM)-positive, and CD45-negative and are enriched based on EpCAM. However, there are a number of methodological challenges regarding both isolation and characterization of these...

Descripción completa

Detalles Bibliográficos
Autores principales: Wu, Yongqi, Park, Kyoung-Joo Jenny, Deighan, Clayton, Amaya, Peter, Miller, Brandon, Pan, Quintin, Zborowski, Maciej, Lustberg, Maryam, Chalmers, Jeffery
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5099140/
https://www.ncbi.nlm.nih.gov/pubmed/27878106
http://dx.doi.org/10.3389/fonc.2016.00234
_version_ 1782465883957886976
author Wu, Yongqi
Park, Kyoung-Joo Jenny
Deighan, Clayton
Amaya, Peter
Miller, Brandon
Pan, Quintin
Zborowski, Maciej
Lustberg, Maryam
Chalmers, Jeffery
author_facet Wu, Yongqi
Park, Kyoung-Joo Jenny
Deighan, Clayton
Amaya, Peter
Miller, Brandon
Pan, Quintin
Zborowski, Maciej
Lustberg, Maryam
Chalmers, Jeffery
author_sort Wu, Yongqi
collection PubMed
description Circulating tumor cells (CTCs) are routinely identified as cytokeratin (CK)-positive, epithelial cell adhesion molecule (EpCAM)-positive, and CD45-negative and are enriched based on EpCAM. However, there are a number of methodological challenges regarding both isolation and characterization of these rare CTCs including downregulation or absence of EpCAM in a variety of solid tumors leading to the omission of subpopulations of CTCs, difficulties in analyzing RNA and protein targets in CTCs due to the rarity of these cells, and low levels of targets and technological limitations of visualizing the targets of interest on each individual cell. Building on our previous CTC research on CD45-based negative magnetic separation and four-color fluorescent immunocytochemical (ICC) staining, RNA in situ hybridization (ISH) was applied to fluorescently target mRNA sequences corresponding to tumor-related genes at the single CTC level. Multiple categories of markers are targeted including CK, human epidermal growth factor receptor family markers, Hedgehog pathway markers, human papillomavirus markers, and protein arginine methyltransferase 5. In addition, an integrated method of RNA ISH and fluorescent ICC staining was developed to visualize CTCs on both mRNA and protein levels. The robustness of the integrated co-ICC and RNA ISH staining was demonstrated by a series of tests on representative tumor markers of different categories. The integrated staining can incorporate the advantages of both RNA ISH and fluorescent ICC staining and provide more intense signals and more specific bindings. With this integrated staining methodology, distinct staining patterns were applied in this report to facilitate the searching and characterization of rare subgroups of CTCs. These results support the existence of diverse groups of CTCs at both protein and mRNA transcript levels and provide an analytical tool for the research on CTCs of rare subgroups.
format Online
Article
Text
id pubmed-5099140
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-50991402016-11-22 Multiparameter Evaluation of the Heterogeneity of Circulating Tumor Cells Using Integrated RNA In Situ Hybridization and Immunocytochemical Analysis Wu, Yongqi Park, Kyoung-Joo Jenny Deighan, Clayton Amaya, Peter Miller, Brandon Pan, Quintin Zborowski, Maciej Lustberg, Maryam Chalmers, Jeffery Front Oncol Oncology Circulating tumor cells (CTCs) are routinely identified as cytokeratin (CK)-positive, epithelial cell adhesion molecule (EpCAM)-positive, and CD45-negative and are enriched based on EpCAM. However, there are a number of methodological challenges regarding both isolation and characterization of these rare CTCs including downregulation or absence of EpCAM in a variety of solid tumors leading to the omission of subpopulations of CTCs, difficulties in analyzing RNA and protein targets in CTCs due to the rarity of these cells, and low levels of targets and technological limitations of visualizing the targets of interest on each individual cell. Building on our previous CTC research on CD45-based negative magnetic separation and four-color fluorescent immunocytochemical (ICC) staining, RNA in situ hybridization (ISH) was applied to fluorescently target mRNA sequences corresponding to tumor-related genes at the single CTC level. Multiple categories of markers are targeted including CK, human epidermal growth factor receptor family markers, Hedgehog pathway markers, human papillomavirus markers, and protein arginine methyltransferase 5. In addition, an integrated method of RNA ISH and fluorescent ICC staining was developed to visualize CTCs on both mRNA and protein levels. The robustness of the integrated co-ICC and RNA ISH staining was demonstrated by a series of tests on representative tumor markers of different categories. The integrated staining can incorporate the advantages of both RNA ISH and fluorescent ICC staining and provide more intense signals and more specific bindings. With this integrated staining methodology, distinct staining patterns were applied in this report to facilitate the searching and characterization of rare subgroups of CTCs. These results support the existence of diverse groups of CTCs at both protein and mRNA transcript levels and provide an analytical tool for the research on CTCs of rare subgroups. Frontiers Media S.A. 2016-11-08 /pmc/articles/PMC5099140/ /pubmed/27878106 http://dx.doi.org/10.3389/fonc.2016.00234 Text en Copyright © 2016 Wu, Park, Deighan, Amaya, Miller, Pan, Zborowski, Lustberg and Chalmers. http://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) or licensor are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Oncology
Wu, Yongqi
Park, Kyoung-Joo Jenny
Deighan, Clayton
Amaya, Peter
Miller, Brandon
Pan, Quintin
Zborowski, Maciej
Lustberg, Maryam
Chalmers, Jeffery
Multiparameter Evaluation of the Heterogeneity of Circulating Tumor Cells Using Integrated RNA In Situ Hybridization and Immunocytochemical Analysis
title Multiparameter Evaluation of the Heterogeneity of Circulating Tumor Cells Using Integrated RNA In Situ Hybridization and Immunocytochemical Analysis
title_full Multiparameter Evaluation of the Heterogeneity of Circulating Tumor Cells Using Integrated RNA In Situ Hybridization and Immunocytochemical Analysis
title_fullStr Multiparameter Evaluation of the Heterogeneity of Circulating Tumor Cells Using Integrated RNA In Situ Hybridization and Immunocytochemical Analysis
title_full_unstemmed Multiparameter Evaluation of the Heterogeneity of Circulating Tumor Cells Using Integrated RNA In Situ Hybridization and Immunocytochemical Analysis
title_short Multiparameter Evaluation of the Heterogeneity of Circulating Tumor Cells Using Integrated RNA In Situ Hybridization and Immunocytochemical Analysis
title_sort multiparameter evaluation of the heterogeneity of circulating tumor cells using integrated rna in situ hybridization and immunocytochemical analysis
topic Oncology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5099140/
https://www.ncbi.nlm.nih.gov/pubmed/27878106
http://dx.doi.org/10.3389/fonc.2016.00234
work_keys_str_mv AT wuyongqi multiparameterevaluationoftheheterogeneityofcirculatingtumorcellsusingintegratedrnainsituhybridizationandimmunocytochemicalanalysis
AT parkkyoungjoojenny multiparameterevaluationoftheheterogeneityofcirculatingtumorcellsusingintegratedrnainsituhybridizationandimmunocytochemicalanalysis
AT deighanclayton multiparameterevaluationoftheheterogeneityofcirculatingtumorcellsusingintegratedrnainsituhybridizationandimmunocytochemicalanalysis
AT amayapeter multiparameterevaluationoftheheterogeneityofcirculatingtumorcellsusingintegratedrnainsituhybridizationandimmunocytochemicalanalysis
AT millerbrandon multiparameterevaluationoftheheterogeneityofcirculatingtumorcellsusingintegratedrnainsituhybridizationandimmunocytochemicalanalysis
AT panquintin multiparameterevaluationoftheheterogeneityofcirculatingtumorcellsusingintegratedrnainsituhybridizationandimmunocytochemicalanalysis
AT zborowskimaciej multiparameterevaluationoftheheterogeneityofcirculatingtumorcellsusingintegratedrnainsituhybridizationandimmunocytochemicalanalysis
AT lustbergmaryam multiparameterevaluationoftheheterogeneityofcirculatingtumorcellsusingintegratedrnainsituhybridizationandimmunocytochemicalanalysis
AT chalmersjeffery multiparameterevaluationoftheheterogeneityofcirculatingtumorcellsusingintegratedrnainsituhybridizationandimmunocytochemicalanalysis