Genomic Determinants of Protein Abundance Variation in Colorectal Cancer Cells

Theodoros I Roumeliotis, Steven P Williams, Emanuel Gonçalves, Clara Alsinet, Martin Del Castillo Velasco-Herrera, Nanne Aben, Fatemeh Zamanzad Ghavidel, Magali Michaut, Michael Schubert, Stacey Price, James C Wright, Lu Yu, Mi Yang, Rodrigo Dienstmann, Justin Guinney, Pedro Beltrao, Alvis Brazma, Mercedes Pardo, Oliver Stegle, David J Adams, Lodewyk Wessels, Julio Saez-Rodriguez, Ultan McDermott, Jyoti S Choudhary, Theodoros I Roumeliotis, Steven P Williams, Emanuel Gonçalves, Clara Alsinet, Martin Del Castillo Velasco-Herrera, Nanne Aben, Fatemeh Zamanzad Ghavidel, Magali Michaut, Michael Schubert, Stacey Price, James C Wright, Lu Yu, Mi Yang, Rodrigo Dienstmann, Justin Guinney, Pedro Beltrao, Alvis Brazma, Mercedes Pardo, Oliver Stegle, David J Adams, Lodewyk Wessels, Julio Saez-Rodriguez, Ultan McDermott, Jyoti S Choudhary

Abstract

Assessing the impact of genomic alterations on protein networks is fundamental in identifying the mechanisms that shape cancer heterogeneity. We have used isobaric labeling to characterize the proteomic landscapes of 50 colorectal cancer cell lines and to decipher the functional consequences of somatic genomic variants. The robust quantification of over 9,000 proteins and 11,000 phosphopeptides on average enabled the de novo construction of a functional protein correlation network, which ultimately exposed the collateral effects of mutations on protein complexes. CRISPR-cas9 deletion of key chromatin modifiers confirmed that the consequences of genomic alterations can propagate through protein interactions in a transcript-independent manner. Lastly, we leveraged the quantified proteome to perform unsupervised classification of the cell lines and to build predictive models of drug response in colorectal cancer. Overall, we provide a deep integrative view of the functional network and the molecular structure underlying the heterogeneity of colorectal cancer cells.

Keywords: CRISPR/cas9; TMT; cell lines; colorectal cancer; drug response; mutations; networks; phosphorylation; protein complexes; proteomics.

Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Global Distributions of Gene-to-Gene Correlations and Protein Co-variation Networks in Colorectal Cancer Cell Lines (A) Distributions of Pearson’s correlation coefficients between protein-protein pairs (left panel) and mRNA-mRNA pairs (right panel) for all pairs (gray) and for pairs with known interactions in the CORUM database (blue). (B) Receiver operating characteristic (ROC) curves illustrating the performance of proteomics- and transcriptomics-based correlations to predict CORUM and high-confident STRING interactions. (C) Protein abundance correlation networks derived from WGCNA analysis for enriched CORUM complexes. The nodes are color-coded according to mRNA-to-protein Pearson correlation. (D) The global structure of the WGCNA network using modules with more than 50 nodes. Protein modules are color coded according to the WGCNA module default name, and representative enriched terms are used for the annotation of the network. See also Figure S3.
Figure 2
Figure 2
The Effect of Colorectal Cancer Driver Mutations on Protein Abundances (A) Association of driver mutations in colorectal cancer genes with the respective protein abundance levels (ANOVA test; permutation-based FDR 

Figure 3

The Global Effects of Genomic…

Figure 3

The Global Effects of Genomic Alterations on Protein and mRNA Abundances (A) Volcano…

Figure 3
The Global Effects of Genomic Alterations on Protein and mRNA Abundances (A) Volcano plot summarizing the effect of missense mutations on the respective protein abundances (ANOVA test). Hits at permutation-based FDR 

Figure 4

The Consequences of Mutations on…

Figure 4

The Consequences of Mutations on Protein Complexes (A) Correlations networks filtered for known…

Figure 4
The Consequences of Mutations on Protein Complexes (A) Correlations networks filtered for known STRING interactions of proteins downregulated by LoF mutations at p value 10(p value). CORUM interactions are highlighted as green thick edges, and representative protein complexes are labeled. (B) Protein abundance correlation network of the ARID1A, ARID2, and PBRM1 modules. Green edges denote known CORUM interactions, and the edge thickness is increasing proportionally to the WGCNA interaction weight. (C) Heatmap summarizing the protein and mRNA abundance log2fold-change values in the knockout clones compared to the wild-type (WT) clones for the proteins in the ARID1A, ARID2, and PBRM1 modules. (D) Volcano plots highlighting the differentially regulated mRNAs in the KO samples. (E) Scatterplot illustrating the correlation between protein and mRNA abundance changes in the ARID1A KO. (F) KEGG pathway and CORUM enrichment analysis for the proteomic analysis results of ARID1A, ARID2, and PBRM1 CRISPR-cas9 knockouts in human iPSCs.

Figure 5

Proteome-wide Quantitative Trait Loci Analysis…

Figure 5

Proteome-wide Quantitative Trait Loci Analysis of Cancer Driver Genomic Alterations (A) Identification of…

Figure 5
Proteome-wide Quantitative Trait Loci Analysis of Cancer Driver Genomic Alterations (A) Identification of cis and trans proteome-wide quantitative trait loci (pQTL) in colorectal cancer cell lines considering colorectal cancer driver variants. The p value and genomic coordinates for the most confident non-redundant protein-variant association tests are depicted in the Manhattan plot. (B) Replication rates between independently tested QTL for each phenotype pair using common sets of genes and variants (n = 6,456 genes). (C) Representation of pQTL as 2D plot of variants (x axis) and associated genes (y axis). Associations with q 

Figure 6

Proteomics Subtypes of Colorectal Cancer…

Figure 6

Proteomics Subtypes of Colorectal Cancer Cell Lines and Pathway Analysis (A) Cell lines…

Figure 6
Proteomics Subtypes of Colorectal Cancer Cell Lines and Pathway Analysis (A) Cell lines are represented as columns, horizontally ordered by five color-coded proteomics consensus clusters and aligned with microsatellite instability (MSI), HNF4A protein abundance, cancer driver genomic alterations, and differentially regulated proteins. (B) KEGG pathway and kinase enrichment analysis per cell line. See also Figure S6.

Figure 7

Pharmacoproteomic Models (A) The number…

Figure 7

Pharmacoproteomic Models (A) The number of drugs for which predictive models (i.e., models…

Figure 7
Pharmacoproteomic Models (A) The number of drugs for which predictive models (i.e., models where the Pearson correlation between predicted and observed IC50s exceeds r > 0.4) could be fitted is stratified per data type. (B) Heatmap indicating for each drug and each data type whether a predictive model could be fitted. Most drugs were specifically predicted by one data type. (C) Heatmap of scaled log2 IC50 values for selected drugs displaying significant association (ANOVA FDR < 0.05) between protein abundance of ABCB1, ABCB11, and drug response. (D) Dose-response profiles for colorectal cancer cell lines treated with docetaxel (black line), 2.5 μM tariquidar alone (gray dotted line), or the combination of docetaxel and 2.5 μM tariquidar (orange line). Error bars represent mean ± SEM. See also Figure S7.
All figures (8)
Similar articles
Cited by
References
    1. Aken B.L., Achuthan P., Akanni W., Amode M.R., Bernsdorff F., Bhai J., Billis K., Carvalho-Silva D., Cummins C., Clapham P. Ensembl 2017. Nucleic Acids Res. 2017;45(D1):D635–D642. - PMC - PubMed
    1. Allen J.D., Xie Y., Chen M., Girard L., Xiao G. Comparing statistical methods for constructing large scale gene networks. PLoS ONE. 2012;7:e29348. - PMC - PubMed
    1. Beltrao P., Bork P., Krogan N.J., van Noort V. Evolution and functional cross-talk of protein post-translational modifications. Mol. Syst. Biol. 2013;9:714. - PMC - PubMed
    1. Bertorelle R., Esposito G., Belluco C., Bonaldi L., Del Mistro A., Nitti D., Lise M., Chieco-Bianchi L. p53 gene alterations and protein accumulation in colorectal cancer. Clin. Mol. Pathol. 1996;49:M85–M90. - PMC - PubMed
    1. Boland C.R., Goel A. Microsatellite instability in colorectal cancer. Gastroenterology. 2010;138:2073–2087.e3. - PMC - PubMed
Show all 54 references
MeSH terms
Related information
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 3
Figure 3
The Global Effects of Genomic Alterations on Protein and mRNA Abundances (A) Volcano plot summarizing the effect of missense mutations on the respective protein abundances (ANOVA test). Hits at permutation-based FDR 

Figure 4

The Consequences of Mutations on…

Figure 4

The Consequences of Mutations on Protein Complexes (A) Correlations networks filtered for known…

Figure 4
The Consequences of Mutations on Protein Complexes (A) Correlations networks filtered for known STRING interactions of proteins downregulated by LoF mutations at p value 10(p value). CORUM interactions are highlighted as green thick edges, and representative protein complexes are labeled. (B) Protein abundance correlation network of the ARID1A, ARID2, and PBRM1 modules. Green edges denote known CORUM interactions, and the edge thickness is increasing proportionally to the WGCNA interaction weight. (C) Heatmap summarizing the protein and mRNA abundance log2fold-change values in the knockout clones compared to the wild-type (WT) clones for the proteins in the ARID1A, ARID2, and PBRM1 modules. (D) Volcano plots highlighting the differentially regulated mRNAs in the KO samples. (E) Scatterplot illustrating the correlation between protein and mRNA abundance changes in the ARID1A KO. (F) KEGG pathway and CORUM enrichment analysis for the proteomic analysis results of ARID1A, ARID2, and PBRM1 CRISPR-cas9 knockouts in human iPSCs.

Figure 5

Proteome-wide Quantitative Trait Loci Analysis…

Figure 5

Proteome-wide Quantitative Trait Loci Analysis of Cancer Driver Genomic Alterations (A) Identification of…

Figure 5
Proteome-wide Quantitative Trait Loci Analysis of Cancer Driver Genomic Alterations (A) Identification of cis and trans proteome-wide quantitative trait loci (pQTL) in colorectal cancer cell lines considering colorectal cancer driver variants. The p value and genomic coordinates for the most confident non-redundant protein-variant association tests are depicted in the Manhattan plot. (B) Replication rates between independently tested QTL for each phenotype pair using common sets of genes and variants (n = 6,456 genes). (C) Representation of pQTL as 2D plot of variants (x axis) and associated genes (y axis). Associations with q 

Figure 6

Proteomics Subtypes of Colorectal Cancer…

Figure 6

Proteomics Subtypes of Colorectal Cancer Cell Lines and Pathway Analysis (A) Cell lines…

Figure 6
Proteomics Subtypes of Colorectal Cancer Cell Lines and Pathway Analysis (A) Cell lines are represented as columns, horizontally ordered by five color-coded proteomics consensus clusters and aligned with microsatellite instability (MSI), HNF4A protein abundance, cancer driver genomic alterations, and differentially regulated proteins. (B) KEGG pathway and kinase enrichment analysis per cell line. See also Figure S6.

Figure 7

Pharmacoproteomic Models (A) The number…

Figure 7

Pharmacoproteomic Models (A) The number of drugs for which predictive models (i.e., models…

Figure 7
Pharmacoproteomic Models (A) The number of drugs for which predictive models (i.e., models where the Pearson correlation between predicted and observed IC50s exceeds r > 0.4) could be fitted is stratified per data type. (B) Heatmap indicating for each drug and each data type whether a predictive model could be fitted. Most drugs were specifically predicted by one data type. (C) Heatmap of scaled log2 IC50 values for selected drugs displaying significant association (ANOVA FDR < 0.05) between protein abundance of ABCB1, ABCB11, and drug response. (D) Dose-response profiles for colorectal cancer cell lines treated with docetaxel (black line), 2.5 μM tariquidar alone (gray dotted line), or the combination of docetaxel and 2.5 μM tariquidar (orange line). Error bars represent mean ± SEM. See also Figure S7.
All figures (8)
Similar articles
Cited by
References
    1. Aken B.L., Achuthan P., Akanni W., Amode M.R., Bernsdorff F., Bhai J., Billis K., Carvalho-Silva D., Cummins C., Clapham P. Ensembl 2017. Nucleic Acids Res. 2017;45(D1):D635–D642. - PMC - PubMed
    1. Allen J.D., Xie Y., Chen M., Girard L., Xiao G. Comparing statistical methods for constructing large scale gene networks. PLoS ONE. 2012;7:e29348. - PMC - PubMed
    1. Beltrao P., Bork P., Krogan N.J., van Noort V. Evolution and functional cross-talk of protein post-translational modifications. Mol. Syst. Biol. 2013;9:714. - PMC - PubMed
    1. Bertorelle R., Esposito G., Belluco C., Bonaldi L., Del Mistro A., Nitti D., Lise M., Chieco-Bianchi L. p53 gene alterations and protein accumulation in colorectal cancer. Clin. Mol. Pathol. 1996;49:M85–M90. - PMC - PubMed
    1. Boland C.R., Goel A. Microsatellite instability in colorectal cancer. Gastroenterology. 2010;138:2073–2087.e3. - PMC - PubMed
Show all 54 references
MeSH terms
Related information
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 4
Figure 4
The Consequences of Mutations on Protein Complexes (A) Correlations networks filtered for known STRING interactions of proteins downregulated by LoF mutations at p value 10(p value). CORUM interactions are highlighted as green thick edges, and representative protein complexes are labeled. (B) Protein abundance correlation network of the ARID1A, ARID2, and PBRM1 modules. Green edges denote known CORUM interactions, and the edge thickness is increasing proportionally to the WGCNA interaction weight. (C) Heatmap summarizing the protein and mRNA abundance log2fold-change values in the knockout clones compared to the wild-type (WT) clones for the proteins in the ARID1A, ARID2, and PBRM1 modules. (D) Volcano plots highlighting the differentially regulated mRNAs in the KO samples. (E) Scatterplot illustrating the correlation between protein and mRNA abundance changes in the ARID1A KO. (F) KEGG pathway and CORUM enrichment analysis for the proteomic analysis results of ARID1A, ARID2, and PBRM1 CRISPR-cas9 knockouts in human iPSCs.
Figure 5
Figure 5
Proteome-wide Quantitative Trait Loci Analysis of Cancer Driver Genomic Alterations (A) Identification of cis and trans proteome-wide quantitative trait loci (pQTL) in colorectal cancer cell lines considering colorectal cancer driver variants. The p value and genomic coordinates for the most confident non-redundant protein-variant association tests are depicted in the Manhattan plot. (B) Replication rates between independently tested QTL for each phenotype pair using common sets of genes and variants (n = 6,456 genes). (C) Representation of pQTL as 2D plot of variants (x axis) and associated genes (y axis). Associations with q 

Figure 6

Proteomics Subtypes of Colorectal Cancer…

Figure 6

Proteomics Subtypes of Colorectal Cancer Cell Lines and Pathway Analysis (A) Cell lines…

Figure 6
Proteomics Subtypes of Colorectal Cancer Cell Lines and Pathway Analysis (A) Cell lines are represented as columns, horizontally ordered by five color-coded proteomics consensus clusters and aligned with microsatellite instability (MSI), HNF4A protein abundance, cancer driver genomic alterations, and differentially regulated proteins. (B) KEGG pathway and kinase enrichment analysis per cell line. See also Figure S6.

Figure 7

Pharmacoproteomic Models (A) The number…

Figure 7

Pharmacoproteomic Models (A) The number of drugs for which predictive models (i.e., models…

Figure 7
Pharmacoproteomic Models (A) The number of drugs for which predictive models (i.e., models where the Pearson correlation between predicted and observed IC50s exceeds r > 0.4) could be fitted is stratified per data type. (B) Heatmap indicating for each drug and each data type whether a predictive model could be fitted. Most drugs were specifically predicted by one data type. (C) Heatmap of scaled log2 IC50 values for selected drugs displaying significant association (ANOVA FDR < 0.05) between protein abundance of ABCB1, ABCB11, and drug response. (D) Dose-response profiles for colorectal cancer cell lines treated with docetaxel (black line), 2.5 μM tariquidar alone (gray dotted line), or the combination of docetaxel and 2.5 μM tariquidar (orange line). Error bars represent mean ± SEM. See also Figure S7.
All figures (8)
Figure 6
Figure 6
Proteomics Subtypes of Colorectal Cancer Cell Lines and Pathway Analysis (A) Cell lines are represented as columns, horizontally ordered by five color-coded proteomics consensus clusters and aligned with microsatellite instability (MSI), HNF4A protein abundance, cancer driver genomic alterations, and differentially regulated proteins. (B) KEGG pathway and kinase enrichment analysis per cell line. See also Figure S6.
Figure 7
Figure 7
Pharmacoproteomic Models (A) The number of drugs for which predictive models (i.e., models where the Pearson correlation between predicted and observed IC50s exceeds r > 0.4) could be fitted is stratified per data type. (B) Heatmap indicating for each drug and each data type whether a predictive model could be fitted. Most drugs were specifically predicted by one data type. (C) Heatmap of scaled log2 IC50 values for selected drugs displaying significant association (ANOVA FDR < 0.05) between protein abundance of ABCB1, ABCB11, and drug response. (D) Dose-response profiles for colorectal cancer cell lines treated with docetaxel (black line), 2.5 μM tariquidar alone (gray dotted line), or the combination of docetaxel and 2.5 μM tariquidar (orange line). Error bars represent mean ± SEM. See also Figure S7.

References

    1. Aken B.L., Achuthan P., Akanni W., Amode M.R., Bernsdorff F., Bhai J., Billis K., Carvalho-Silva D., Cummins C., Clapham P. Ensembl 2017. Nucleic Acids Res. 2017;45(D1):D635–D642.
    1. Allen J.D., Xie Y., Chen M., Girard L., Xiao G. Comparing statistical methods for constructing large scale gene networks. PLoS ONE. 2012;7:e29348.
    1. Beltrao P., Bork P., Krogan N.J., van Noort V. Evolution and functional cross-talk of protein post-translational modifications. Mol. Syst. Biol. 2013;9:714.
    1. Bertorelle R., Esposito G., Belluco C., Bonaldi L., Del Mistro A., Nitti D., Lise M., Chieco-Bianchi L. p53 gene alterations and protein accumulation in colorectal cancer. Clin. Mol. Pathol. 1996;49:M85–M90.
    1. Boland C.R., Goel A. Microsatellite instability in colorectal cancer. Gastroenterology. 2010;138:2073–2087.e3.
    1. Cairns B.R. Emerging roles for chromatin remodeling in cancer biology. Trends Cell Biol. 2001;11:S15–S21.
    1. Chan C.W.M., Wong N.A., Liu Y., Bicknell D., Turley H., Hollins L., Miller C.J., Wilding J.L., Bodmer W.F. Gastrointestinal differentiation marker Cytokeratin 20 is regulated by homeobox gene CDX1. Proc. Natl. Acad. Sci. USA. 2009;106:1936–1941.
    1. Chen Z., Shi T., Zhang L., Zhu P., Deng M., Huang C., Hu T., Jiang L., Li J. Mammalian drug efflux transporters of the ATP binding cassette (ABC) family in multidrug resistance: A review of the past decade. Cancer Lett. 2016;370:153–164.
    1. Chen W.H., Lu G., Chen X., Zhao X.M., Bork P. OGEE v2: an update of the online gene essentiality database with special focus on differentially essential genes in human cancer cell lines. Nucleic Acids Res. 2017;45(D1):D940–D944.
    1. Costanzo M., VanderSluis B., Koch E.N., Baryshnikova A., Pons C., Tan G., Wang W., Usaj M., Hanchard J., Lee S.D. A global genetic interaction network maps a wiring diagram of cellular function. Science. 2016;353:aaf1420.
    1. Costello J.C., Heiser L.M., Georgii E., Gönen M., Menden M.P., Wang N.J., Bansal M., Ammad-ud-din M., Hintsanen P., Khan S.A., NCI DREAM Community A community effort to assess and improve drug sensitivity prediction algorithms. Nat. Biotechnol. 2014;32:1202–1212.
    1. Davis M.A., Larimore E.A., Fissel B.M., Swanger J., Taatjes D.J., Clurman B.E. The SCF-Fbw7 ubiquitin ligase degrades MED13 and MED13L and regulates CDK8 module association with Mediator. Genes Dev. 2013;27:151–156.
    1. De Sousa E Melo F., Wang X., Jansen M., Fessler E., Trinh A., de Rooij L.P., de Jong J.H., de Boer O.J., van Leersum R., Bijlsma M.F. Poor-prognosis colon cancer is defined by a molecularly distinct subtype and develops from serrated precursor lesions. Nat. Med. 2013;19:614–618.
    1. Dix B., Robbins P., Carrello S., House A., Iacopetta B. Comparison of p53 gene mutation and protein overexpression in colorectal carcinomas. Br. J. Cancer. 1994;70:585–590.
    1. Forester C.M., Maddox J., Louis J.V., Goris J., Virshup D.M. Control of mitotic exit by PP2A regulation of Cdc25C and Cdk1. Proc. Natl. Acad. Sci. USA. 2007;104:19867–19872.
    1. Garnett M.J., Edelman E.J., Heidorn S.J., Greenman C.D., Dastur A., Lau K.W., Greninger P., Thompson I.R., Luo X., Soares J. Systematic identification of genomic markers of drug sensitivity in cancer cells. Nature. 2012;483:570–575.
    1. Garrison W.D., Battle M.A., Yang C., Kaestner K.H., Sladek F.M., Duncan S.A. Hepatocyte nuclear factor 4alpha is essential for embryonic development of the mouse colon. Gastroenterology. 2006;130:1207–1220.
    1. Gholami A.M., Hahne H., Wu Z., Auer F.J., Meng C., Wilhelm M., Kuster B. Global proteome analysis of the NCI-60 cell line panel. Cell Rep. 2013;4:609–620.
    1. Guinney J., Dienstmann R., Wang X., de Reyniès A., Schlicker A., Soneson C., Marisa L., Roepman P., Nyamundanda G., Angelino P. The consensus molecular subtypes of colorectal cancer. Nat. Med. 2015;21:1350–1356.
    1. Hein M.Y., Hubner N.C., Poser I., Cox J., Nagaraj N., Toyoda Y., Gak I.A., Weisswange I., Mansfeld J., Buchholz F. A human interactome in three quantitative dimensions organized by stoichiometries and abundances. Cell. 2015;163:712–723.
    1. Hodges C., Kirkland J.G., Crabtree G.R. The many roles of BAF (mSWI/SNF) and PBAF complexes in cancer. Cold Spring Harb. Perspect. Med. 2016;6:a026930.
    1. Hornbeck P.V., Zhang B., Murray B., Kornhauser J.M., Latham V., Skrzypek E. PhosphoSitePlus, 2014: mutations, PTMs and recalibrations. Nucleic Acids Res. 2015;43:D512–D520.
    1. Huttlin E.L., Ting L., Bruckner R.J., Gebreab F., Gygi M.P., Szpyt J., Tam S., Zarraga G., Colby G., Baltier K. The BioPlex network: a systematic exploration of the human interactome. Cell. 2015;162:425–440.
    1. Iorio F., Knijnenburg T.A., Vis D.J., Bignell G.R., Menden M.P., Schubert M., Aben N., Gonçalves E., Barthorpe S., Lightfoot H. A landscape of pharmacogenomic interactions in cancer. Cell. 2016;166:740–754.
    1. Jones M.F., Hara T., Francis P., Li X.L., Bilke S., Zhu Y., Pineda M., Subramanian M., Bodmer W.F., Lal A. The CDX1-microRNA-215 axis regulates colorectal cancer stem cell differentiation. Proc. Natl. Acad. Sci. USA. 2015;112:E1550–E1558.
    1. Kaelin W.G., Jr. The concept of synthetic lethality in the context of anticancer therapy. Nat. Rev. Cancer. 2005;5:689–698.
    1. Kent W.J., Sugnet C.W., Furey T.S., Roskin K.M., Pringle T.H., Zahler A.M., Haussler D. The human genome browser at UCSC. Genome Res. 2002;12:996–1006.
    1. Kuleshov M.V., Jones M.R., Rouillard A.D., Fernandez N.F., Duan Q., Wang Z., Koplev S., Jenkins S.L., Jagodnik K.M., Lachmann A. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016;44(W1):W90–W97.
    1. Langfelder P., Horvath S. Eigengene networks for studying the relationships between co-expression modules. BMC Syst. Biol. 2007;1:54.
    1. Langfelder P., Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics. 2008;9:559.
    1. Lei M., Tye B.K. Initiating DNA synthesis: from recruiting to activating the MCM complex. J. Cell Sci. 2001;114:1447–1454.
    1. Li J., Zhao W., Akbani R., Liu W., Ju Z., Ling S., Vellano C.P., Roebuck P., Yu Q., Eterovic A.K. Characterization of human cancer cell lines by reverse-phase protein arrays. Cancer Cell. 2017;31:225–239.
    1. McAlister G.C., Nusinow D.P., Jedrychowski M.P., Wühr M., Huttlin E.L., Erickson B.K., Rad R., Haas W., Gygi S.P. MultiNotch MS3 enables accurate, sensitive, and multiplexed detection of differential expression across cancer cell line proteomes. Anal. Chem. 2014;86:7150–7158.
    1. Medico E., Russo M., Picco G., Cancelliere C., Valtorta E., Corti G., Buscarino M., Isella C., Lamba S., Martinoglio B. The molecular landscape of colorectal cancer cell lines unveils clinically actionable kinase targets. Nat. Commun. 2015;6:7002.
    1. Mertins P., Mani D.R., Ruggles K.V., Gillette M.A., Clauser K.R., Wang P., Wang X., Qiao J.W., Cao S., Petralia F., NCI CPTAC Proteogenomics connects somatic mutations to signalling in breast cancer. Nature. 2016;534:55–62.
    1. Millevoi S., Loulergue C., Dettwiler S., Karaa S.Z., Keller W., Antoniou M., Vagner S. An interaction between U2AF 65 and CF I(m) links the splicing and 3′ end processing machineries. EMBO J. 2006;25:4854–4864.
    1. Narlikar G.J., Sundaramoorthy R., Owen-Hughes T. Mechanisms and functions of ATP-dependent chromatin-remodeling enzymes. Cell. 2013;154:490–503.
    1. Ochoa D., Jonikas M., Lawrence R.T., El Debs B., Selkrig J., Typas A., Villén J., Santos S.D., Beltrao P. An atlas of human kinase regulation. Mol. Syst. Biol. 2016;12:888.
    1. Ohta S., Tatsumi Y., Fujita M., Tsurimoto T., Obuse C. The ORC1 cycle in human cells: II. Dynamic changes in the human ORC complex during the cell cycle. J. Biol. Chem. 2003;278:41535–41540.
    1. Petryszak R., Keays M., Tang Y.A., Fonseca N.A., Barrera E., Burdett T., Füllgrabe A., Fuentes A.M., Jupp S., Koskinen S. Expression Atlas update--an integrated database of gene and protein expression in humans, animals and plants. Nucleic Acids Res. 2016;44(D1):D746–D752.
    1. Petsalaki E., Helbig A.O., Gopal A., Pasculescu A., Roth F.P., Pawson T. SELPHI: correlation-based identification of kinase-associated networks from global phospho-proteomics data sets. Nucleic Acids Res. 2015;43(W1):W276–W282.
    1. Popat S., Hubner R., Houlston R.S. Systematic review of microsatellite instability and colorectal cancer prognosis. J. Clin. Oncol. 2005;23:609–618.
    1. Prasanth S.G., Prasanth K.V., Stillman B. Orc6 involved in DNA replication, chromosome segregation, and cytokinesis. Science. 2002;297:1026–1031.
    1. Robinson J.T., Thorvaldsdóttir H., Winckler W., Guttman M., Lander E.S., Getz G., Mesirov J.P. Integrative genomics viewer. Nat. Biotechnol. 2011;29:24–26.
    1. Ruepp A., Waegele B., Lechner M., Brauner B., Dunger-Kaltenbach I., Fobo G., Frishman G., Montrone C., Mewes H.W. CORUM: the comprehensive resource of mammalian protein complexes--2009. Nucleic Acids Res. 2010;38:D497–D501.
    1. Sadanandam A., Lyssiotis C.A., Homicsko K., Collisson E.A., Gibb W.J., Wullschleger S., Ostos L.C.G., Lannon W.A., Grotzinger C., Del Rio M. A colorectal cancer classification system that associates cellular phenotype and responses to therapy. Nat. Med. 2013;19:619–625.
    1. Stefely J.A., Kwiecien N.W., Freiberger E.C., Richards A.L., Jochem A., Rush M.J.P., Ulbrich A., Robinson K.P., Hutchins P.D., Veling M.T. Mitochondrial protein functions elucidated by multi-omic mass spectrometry profiling. Nat. Biotechnol. 2016;34:1191–1197.
    1. Szklarczyk D., Franceschini A., Wyder S., Forslund K., Heller D., Huerta-Cepas J., Simonovic M., Roth A., Santos A., Tsafou K.P. STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 2015;43:D447–D452.
    1. Thompson M. Polybromo-1: the chromatin targeting subunit of the PBAF complex. Biochimie. 2009;91:309–319.
    1. Wang J., Ma Z., Carr S.A., Mertins P., Zhang H., Zhang Z., Chan D.W., Ellis M.J., Townsend R.R., Smith R.D. Proteome profiling outperforms transcriptome profiling for coexpression based gene function prediction. Mol. Cell. Proteomics. 2017;16:121–134.
    1. Wickramasinghe V.O., Laskey R.A. Control of mammalian gene expression by selective mRNA export. Nat. Rev. Mol. Cell Biol. 2015;16:431–442.
    1. Wilkerson M.D., Hayes D.N. ConsensusClusterPlus: a class discovery tool with confidence assessments and item tracking. Bioinformatics. 2010;26:1572–1573.
    1. Zhang B., Wang J., Wang X., Zhu J., Liu Q., Shi Z., Chambers M.C., Zimmerman L.J., Shaddox K.F., Kim S., NCI CPTAC Proteogenomic characterization of human colon and rectal cancer. Nature. 2014;513:382–387.
    1. Zhang H., Liu T., Zhang Z., Payne S.H., Zhang B., McDermott J.E., Zhou J.Y., Petyuk V.A., Chen L., Ray D., CPTAC investigators Integrated proteogenomic characterization of human high-grade serous ovarian cancer. Cell. 2016;166:755–765.

Source: PubMed

3
Abonnere