Cargando…

Impact of Variable RNA-Sequencing Depth on Gene Expression Signatures and Target Compound Robustness: Case Study Examining Brain Tumor (Glioma) Disease Progression

PURPOSE: Gene expression profiling can uncover biologic mechanisms underlying disease and is important in drug development. RNA sequencing (RNA-seq) is routinely used to assess gene expression, but costs remain high. Sample multiplexing reduces RNA-seq costs; however, multiplexed samples have lower...

Descripción completa

Detalles Bibliográficos
Autores principales: Stupnikov, Alexey, O’Reilly, Paul G., McInerney, Caitriona E., Roddy, Aideen C., Dunne, Philip D., Gilmore, Alan, Ellis, Hayley P., Flannery, Tom, Healy, Estelle, McIntosh, Stuart A., Savage, Kienan, Kurian, Kathreena M., Emmert-Streib, Frank, Prise, Kevin M., Salto-Tellez, Manuel, McArt, Darragh G.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: American Society of Clinical Oncology 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6186166/
https://www.ncbi.nlm.nih.gov/pubmed/30324181
http://dx.doi.org/10.1200/PO.18.00014
_version_ 1783362824702001152
author Stupnikov, Alexey
O’Reilly, Paul G.
McInerney, Caitriona E.
Roddy, Aideen C.
Dunne, Philip D.
Gilmore, Alan
Ellis, Hayley P.
Flannery, Tom
Healy, Estelle
McIntosh, Stuart A.
Savage, Kienan
Kurian, Kathreena M.
Emmert-Streib, Frank
Prise, Kevin M.
Salto-Tellez, Manuel
McArt, Darragh G.
author_facet Stupnikov, Alexey
O’Reilly, Paul G.
McInerney, Caitriona E.
Roddy, Aideen C.
Dunne, Philip D.
Gilmore, Alan
Ellis, Hayley P.
Flannery, Tom
Healy, Estelle
McIntosh, Stuart A.
Savage, Kienan
Kurian, Kathreena M.
Emmert-Streib, Frank
Prise, Kevin M.
Salto-Tellez, Manuel
McArt, Darragh G.
author_sort Stupnikov, Alexey
collection PubMed
description PURPOSE: Gene expression profiling can uncover biologic mechanisms underlying disease and is important in drug development. RNA sequencing (RNA-seq) is routinely used to assess gene expression, but costs remain high. Sample multiplexing reduces RNA-seq costs; however, multiplexed samples have lower cDNA sequencing depth, which can hinder accurate differential gene expression detection. The impact of sequencing depth alteration on RNA-seq–based downstream analyses such as gene expression connectivity mapping is not known, where this method is used to identify potential therapeutic compounds for repurposing. METHODS: In this study, published RNA-seq profiles from patients with brain tumor (glioma) were assembled into two disease progression gene signature contrasts for astrocytoma. Available treatments for glioma have limited effectiveness, rendering this a disease of poor clinical outcome. Gene signatures were subsampled to simulate sequencing alterations and analyzed in connectivity mapping to investigate target compound robustness. RESULTS: Data loss to gene signatures led to the loss, gain, and consistent identification of significant connections. The most accurate gene signature contrast with consistent patient gene expression profiles was more resilient to data loss and identified robust target compounds. Target compounds lost included candidate compounds of potential clinical utility in glioma (eg, suramin, dasatinib). Lost connections may have been linked to low-abundance genes in the gene signature that closely characterized the disease phenotype. Consistently identified connections may have been related to highly expressed abundant genes that were ever-present in gene signatures, despite data reductions. Potential noise surrounding findings included false-positive connections that were gained as a result of gene signature modification with data loss. CONCLUSION: Findings highlight the necessity for gene signature accuracy for connectivity mapping, which should improve the clinical utility of future target compound discoveries.
format Online
Article
Text
id pubmed-6186166
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher American Society of Clinical Oncology
record_format MEDLINE/PubMed
spelling pubmed-61861662018-10-13 Impact of Variable RNA-Sequencing Depth on Gene Expression Signatures and Target Compound Robustness: Case Study Examining Brain Tumor (Glioma) Disease Progression Stupnikov, Alexey O’Reilly, Paul G. McInerney, Caitriona E. Roddy, Aideen C. Dunne, Philip D. Gilmore, Alan Ellis, Hayley P. Flannery, Tom Healy, Estelle McIntosh, Stuart A. Savage, Kienan Kurian, Kathreena M. Emmert-Streib, Frank Prise, Kevin M. Salto-Tellez, Manuel McArt, Darragh G. JCO Precis Oncol Original Report PURPOSE: Gene expression profiling can uncover biologic mechanisms underlying disease and is important in drug development. RNA sequencing (RNA-seq) is routinely used to assess gene expression, but costs remain high. Sample multiplexing reduces RNA-seq costs; however, multiplexed samples have lower cDNA sequencing depth, which can hinder accurate differential gene expression detection. The impact of sequencing depth alteration on RNA-seq–based downstream analyses such as gene expression connectivity mapping is not known, where this method is used to identify potential therapeutic compounds for repurposing. METHODS: In this study, published RNA-seq profiles from patients with brain tumor (glioma) were assembled into two disease progression gene signature contrasts for astrocytoma. Available treatments for glioma have limited effectiveness, rendering this a disease of poor clinical outcome. Gene signatures were subsampled to simulate sequencing alterations and analyzed in connectivity mapping to investigate target compound robustness. RESULTS: Data loss to gene signatures led to the loss, gain, and consistent identification of significant connections. The most accurate gene signature contrast with consistent patient gene expression profiles was more resilient to data loss and identified robust target compounds. Target compounds lost included candidate compounds of potential clinical utility in glioma (eg, suramin, dasatinib). Lost connections may have been linked to low-abundance genes in the gene signature that closely characterized the disease phenotype. Consistently identified connections may have been related to highly expressed abundant genes that were ever-present in gene signatures, despite data reductions. Potential noise surrounding findings included false-positive connections that were gained as a result of gene signature modification with data loss. CONCLUSION: Findings highlight the necessity for gene signature accuracy for connectivity mapping, which should improve the clinical utility of future target compound discoveries. American Society of Clinical Oncology 2018-09-13 /pmc/articles/PMC6186166/ /pubmed/30324181 http://dx.doi.org/10.1200/PO.18.00014 Text en © 2018 by American Society of Clinical Oncology https://creativecommons.org/licenses/by/4.0/ Licensed under the Creative Commons Attribution 4.0 License: https://creativecommons.org/licenses/by/4.0/.
spellingShingle Original Report
Stupnikov, Alexey
O’Reilly, Paul G.
McInerney, Caitriona E.
Roddy, Aideen C.
Dunne, Philip D.
Gilmore, Alan
Ellis, Hayley P.
Flannery, Tom
Healy, Estelle
McIntosh, Stuart A.
Savage, Kienan
Kurian, Kathreena M.
Emmert-Streib, Frank
Prise, Kevin M.
Salto-Tellez, Manuel
McArt, Darragh G.
Impact of Variable RNA-Sequencing Depth on Gene Expression Signatures and Target Compound Robustness: Case Study Examining Brain Tumor (Glioma) Disease Progression
title Impact of Variable RNA-Sequencing Depth on Gene Expression Signatures and Target Compound Robustness: Case Study Examining Brain Tumor (Glioma) Disease Progression
title_full Impact of Variable RNA-Sequencing Depth on Gene Expression Signatures and Target Compound Robustness: Case Study Examining Brain Tumor (Glioma) Disease Progression
title_fullStr Impact of Variable RNA-Sequencing Depth on Gene Expression Signatures and Target Compound Robustness: Case Study Examining Brain Tumor (Glioma) Disease Progression
title_full_unstemmed Impact of Variable RNA-Sequencing Depth on Gene Expression Signatures and Target Compound Robustness: Case Study Examining Brain Tumor (Glioma) Disease Progression
title_short Impact of Variable RNA-Sequencing Depth on Gene Expression Signatures and Target Compound Robustness: Case Study Examining Brain Tumor (Glioma) Disease Progression
title_sort impact of variable rna-sequencing depth on gene expression signatures and target compound robustness: case study examining brain tumor (glioma) disease progression
topic Original Report
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6186166/
https://www.ncbi.nlm.nih.gov/pubmed/30324181
http://dx.doi.org/10.1200/PO.18.00014
work_keys_str_mv AT stupnikovalexey impactofvariablernasequencingdepthongeneexpressionsignaturesandtargetcompoundrobustnesscasestudyexaminingbraintumorgliomadiseaseprogression
AT oreillypaulg impactofvariablernasequencingdepthongeneexpressionsignaturesandtargetcompoundrobustnesscasestudyexaminingbraintumorgliomadiseaseprogression
AT mcinerneycaitrionae impactofvariablernasequencingdepthongeneexpressionsignaturesandtargetcompoundrobustnesscasestudyexaminingbraintumorgliomadiseaseprogression
AT roddyaideenc impactofvariablernasequencingdepthongeneexpressionsignaturesandtargetcompoundrobustnesscasestudyexaminingbraintumorgliomadiseaseprogression
AT dunnephilipd impactofvariablernasequencingdepthongeneexpressionsignaturesandtargetcompoundrobustnesscasestudyexaminingbraintumorgliomadiseaseprogression
AT gilmorealan impactofvariablernasequencingdepthongeneexpressionsignaturesandtargetcompoundrobustnesscasestudyexaminingbraintumorgliomadiseaseprogression
AT ellishayleyp impactofvariablernasequencingdepthongeneexpressionsignaturesandtargetcompoundrobustnesscasestudyexaminingbraintumorgliomadiseaseprogression
AT flannerytom impactofvariablernasequencingdepthongeneexpressionsignaturesandtargetcompoundrobustnesscasestudyexaminingbraintumorgliomadiseaseprogression
AT healyestelle impactofvariablernasequencingdepthongeneexpressionsignaturesandtargetcompoundrobustnesscasestudyexaminingbraintumorgliomadiseaseprogression
AT mcintoshstuarta impactofvariablernasequencingdepthongeneexpressionsignaturesandtargetcompoundrobustnesscasestudyexaminingbraintumorgliomadiseaseprogression
AT savagekienan impactofvariablernasequencingdepthongeneexpressionsignaturesandtargetcompoundrobustnesscasestudyexaminingbraintumorgliomadiseaseprogression
AT kuriankathreenam impactofvariablernasequencingdepthongeneexpressionsignaturesandtargetcompoundrobustnesscasestudyexaminingbraintumorgliomadiseaseprogression
AT emmertstreibfrank impactofvariablernasequencingdepthongeneexpressionsignaturesandtargetcompoundrobustnesscasestudyexaminingbraintumorgliomadiseaseprogression
AT prisekevinm impactofvariablernasequencingdepthongeneexpressionsignaturesandtargetcompoundrobustnesscasestudyexaminingbraintumorgliomadiseaseprogression
AT saltotellezmanuel impactofvariablernasequencingdepthongeneexpressionsignaturesandtargetcompoundrobustnesscasestudyexaminingbraintumorgliomadiseaseprogression
AT mcartdarraghg impactofvariablernasequencingdepthongeneexpressionsignaturesandtargetcompoundrobustnesscasestudyexaminingbraintumorgliomadiseaseprogression