Cargando…

Large-scale integration of small molecule-induced genome-wide transcriptional responses, Kinome-wide binding affinities and cell-growth inhibition profiles reveal global trends characterizing systems-level drug action

The Library of Integrated Network-based Cellular Signatures (LINCS) project is a large-scale coordinated effort to build a comprehensive systems biology reference resource. The goals of the program include the generation of a very large multidimensional data matrix and informatics and computational...

Descripción completa

Detalles Bibliográficos
Autores principales: Vidović, Dušica, Koleti, Amar, Schürer, Stephan C.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2014
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4179751/
https://www.ncbi.nlm.nih.gov/pubmed/25324859
http://dx.doi.org/10.3389/fgene.2014.00342
_version_ 1782337143285219328
author Vidović, Dušica
Koleti, Amar
Schürer, Stephan C.
author_facet Vidović, Dušica
Koleti, Amar
Schürer, Stephan C.
author_sort Vidović, Dušica
collection PubMed
description The Library of Integrated Network-based Cellular Signatures (LINCS) project is a large-scale coordinated effort to build a comprehensive systems biology reference resource. The goals of the program include the generation of a very large multidimensional data matrix and informatics and computational tools to integrate, analyze, and make the data readily accessible. LINCS data include genome-wide transcriptional signatures, biochemical protein binding profiles, cellular phenotypic response profiles and various other datasets for a wide range of cell model systems and molecular and genetic perturbations. Here we present a partial survey of this data facilitated by data standards and in particular a robust compound standardization workflow; we integrated several types of LINCS signatures and analyzed the results with a focus on mechanism of action (MoA) and chemical compounds. We illustrate how kinase targets can be related to disease models and relevant drugs. We identified some fundamental trends that appear to link Kinome binding profiles and transcriptional signatures to chemical information and biochemical binding profiles to transcriptional responses independent of chemical similarity. To fill gaps in the datasets we developed and applied predictive models. The results can be interpreted at the systems level as demonstrated based on a large number of signaling pathways. We can identify clear global relationships, suggesting robustness of cellular responses to chemical perturbation. Overall, the results suggest that chemical similarity is a useful measure at the systems level, which would support phenotypic drug optimization efforts. With this study we demonstrate the potential of such integrated analysis approaches and suggest prioritizing further experiments to fill the gaps in the current data.
format Online
Article
Text
id pubmed-4179751
institution National Center for Biotechnology Information
language English
publishDate 2014
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-41797512014-10-16 Large-scale integration of small molecule-induced genome-wide transcriptional responses, Kinome-wide binding affinities and cell-growth inhibition profiles reveal global trends characterizing systems-level drug action Vidović, Dušica Koleti, Amar Schürer, Stephan C. Front Genet Physiology The Library of Integrated Network-based Cellular Signatures (LINCS) project is a large-scale coordinated effort to build a comprehensive systems biology reference resource. The goals of the program include the generation of a very large multidimensional data matrix and informatics and computational tools to integrate, analyze, and make the data readily accessible. LINCS data include genome-wide transcriptional signatures, biochemical protein binding profiles, cellular phenotypic response profiles and various other datasets for a wide range of cell model systems and molecular and genetic perturbations. Here we present a partial survey of this data facilitated by data standards and in particular a robust compound standardization workflow; we integrated several types of LINCS signatures and analyzed the results with a focus on mechanism of action (MoA) and chemical compounds. We illustrate how kinase targets can be related to disease models and relevant drugs. We identified some fundamental trends that appear to link Kinome binding profiles and transcriptional signatures to chemical information and biochemical binding profiles to transcriptional responses independent of chemical similarity. To fill gaps in the datasets we developed and applied predictive models. The results can be interpreted at the systems level as demonstrated based on a large number of signaling pathways. We can identify clear global relationships, suggesting robustness of cellular responses to chemical perturbation. Overall, the results suggest that chemical similarity is a useful measure at the systems level, which would support phenotypic drug optimization efforts. With this study we demonstrate the potential of such integrated analysis approaches and suggest prioritizing further experiments to fill the gaps in the current data. Frontiers Media S.A. 2014-09-30 /pmc/articles/PMC4179751/ /pubmed/25324859 http://dx.doi.org/10.3389/fgene.2014.00342 Text en Copyright © 2014 Vidović, Koleti and Schürer. 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 Physiology
Vidović, Dušica
Koleti, Amar
Schürer, Stephan C.
Large-scale integration of small molecule-induced genome-wide transcriptional responses, Kinome-wide binding affinities and cell-growth inhibition profiles reveal global trends characterizing systems-level drug action
title Large-scale integration of small molecule-induced genome-wide transcriptional responses, Kinome-wide binding affinities and cell-growth inhibition profiles reveal global trends characterizing systems-level drug action
title_full Large-scale integration of small molecule-induced genome-wide transcriptional responses, Kinome-wide binding affinities and cell-growth inhibition profiles reveal global trends characterizing systems-level drug action
title_fullStr Large-scale integration of small molecule-induced genome-wide transcriptional responses, Kinome-wide binding affinities and cell-growth inhibition profiles reveal global trends characterizing systems-level drug action
title_full_unstemmed Large-scale integration of small molecule-induced genome-wide transcriptional responses, Kinome-wide binding affinities and cell-growth inhibition profiles reveal global trends characterizing systems-level drug action
title_short Large-scale integration of small molecule-induced genome-wide transcriptional responses, Kinome-wide binding affinities and cell-growth inhibition profiles reveal global trends characterizing systems-level drug action
title_sort large-scale integration of small molecule-induced genome-wide transcriptional responses, kinome-wide binding affinities and cell-growth inhibition profiles reveal global trends characterizing systems-level drug action
topic Physiology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4179751/
https://www.ncbi.nlm.nih.gov/pubmed/25324859
http://dx.doi.org/10.3389/fgene.2014.00342
work_keys_str_mv AT vidovicdusica largescaleintegrationofsmallmoleculeinducedgenomewidetranscriptionalresponseskinomewidebindingaffinitiesandcellgrowthinhibitionprofilesrevealglobaltrendscharacterizingsystemsleveldrugaction
AT koletiamar largescaleintegrationofsmallmoleculeinducedgenomewidetranscriptionalresponseskinomewidebindingaffinitiesandcellgrowthinhibitionprofilesrevealglobaltrendscharacterizingsystemsleveldrugaction
AT schurerstephanc largescaleintegrationofsmallmoleculeinducedgenomewidetranscriptionalresponseskinomewidebindingaffinitiesandcellgrowthinhibitionprofilesrevealglobaltrendscharacterizingsystemsleveldrugaction