Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Sci Rep ; 13(1): 20840, 2023 11 27.
Article in English | MEDLINE | ID: mdl-38012155

ABSTRACT

One important aim of precision oncology is a personalized treatment of patients. This can be achieved by various biomarkers, especially imaging parameters and gene expression signatures are commonly used. So far, combination approaches are sparse. The aim of the study was to independently validate the prognostic value of the novel positron emission tomography (PET) parameter tumor asphericity (ASP) in non small cell lung cancer (NSCLC) patients and to investigate associations between published gene expression profiles and ASP. This was a retrospective evaluation of PET imaging and gene expression data from three public databases and two institutional datasets. The whole cohort comprised 253 NSCLC patients, all treated with curative intent surgery. Clinical parameters, standard PET parameters and ASP were evaluated in all patients. Additional gene expression data were available for 120 patients. Univariate Cox regression and Kaplan-Meier analysis was performed for the primary endpoint progression-free survival (PFS) and additional endpoints. Furthermore, multivariate cox regression testing was performed including clinically significant parameters, ASP, and the extracellular matrix-related prognostic gene signature (EPPI). In the whole cohort, a significant association with PFS was observed for ASP (p < 0.001) and EPPI (p = 0.012). Upon multivariate testing, EPPI remained significantly associated with PFS (p = 0.018) in the subgroup of patients with additional gene expression data, while ASP was significantly associated with PFS in the whole cohort (p = 0.012). In stage II patients, ASP was significantly associated with PFS (p = 0.009), and a previously published cutoff value for ASP (19.5%) was successfully validated (p = 0.008). In patients with additional gene expression data, EPPI showed a significant association with PFS, too (p = 0.033). The exploratory combination of ASP and EPPI showed that the combinatory approach has potential to further improve patient stratification compared to the use of only one parameter. We report the first successful validation of EPPI and ASP in stage II NSCLC patients. The combination of both parameters seems to be a very promising approach for improvement of risk stratification in a group of patients with urgent need for a more personalized treatment approach.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Prognosis , Lung Neoplasms/pathology , Retrospective Studies , Fluorodeoxyglucose F18/metabolism , Tomography, X-Ray Computed , Precision Medicine , Positron Emission Tomography Computed Tomography
2.
Mol Syst Biol ; 19(11): e11510, 2023 Nov 09.
Article in English | MEDLINE | ID: mdl-37735975

ABSTRACT

For a short period during early development of mammalian embryos, both X chromosomes in females are active, before dosage compensation is ensured through X-chromosome inactivation. In female mouse embryonic stem cells (mESCs), which carry two active X chromosomes, increased X-dosage affects cell signaling and impairs differentiation. The underlying mechanisms, however, remain poorly understood. To dissect X-dosage effects on the signaling network in mESCs, we combine systematic perturbation experiments with mathematical modeling. We quantify the response to a variety of inhibitors and growth factors for cells with one (XO) or two X chromosomes (XX). We then build models of the signaling networks in XX and XO cells through a semi-quantitative modeling approach based on modular response analysis. We identify a novel negative feedback in the PI3K/AKT pathway through GSK3. Moreover, the presence of a single active X makes mESCs more sensitive to the differentiation-promoting Activin A signal and leads to a stronger RAF1-mediated negative feedback in the FGF-triggered MAPK pathway. The differential response to these differentiation-promoting pathways can explain the impaired differentiation propensity of female mESCs.


Subject(s)
Embryonic Stem Cells , Mouse Embryonic Stem Cells , Female , Animals , Male , Mice , Mouse Embryonic Stem Cells/metabolism , Embryonic Stem Cells/metabolism , Sex Characteristics , Glycogen Synthase Kinase 3 , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Cell Differentiation/genetics , Mammals
3.
Cell Death Dis ; 12(12): 1162, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34911941

ABSTRACT

Resistance against radio(chemo)therapy-induced cell death is a major determinant of oncological treatment failure and remains a perpetual clinical challenge. The underlying mechanisms are manifold and demand for comprehensive, cancer entity- and subtype-specific examination. In the present study, resistance against radiotherapy was systematically assessed in a panel of human head-and-neck squamous cell carcinoma (HNSCC) cell lines and xenotransplants derived thereof with the overarching aim to extract master regulators and potential candidates for mechanism-based pharmacological targeting. Clonogenic survival data were integrated with molecular and functional data on DNA damage repair and different cell fate decisions. A positive correlation between radioresistance and early induction of HNSCC cell senescence accompanied by NF-κB-dependent production of distinct senescence-associated cytokines, particularly ligands of the CXCR2 chemokine receptor, was identified. Time-lapse microscopy and medium transfer experiments disclosed the non-cell autonomous, paracrine nature of these mechanisms, and pharmacological interference with senescence-associated cytokine production by the NF-κB inhibitor metformin significantly improved radiotherapeutic performance in vitro and in vivo. With regard to clinical relevance, retrospective analyses of TCGA HNSCC data and an in-house HNSCC cohort revealed that elevated expression of CXCR2 and/or its ligands are associated with impaired treatment outcome. Collectively, our study identifies radiation-induced tumor cell senescence and the NF-κB-dependent production of distinct senescence-associated cytokines as critical drivers of radioresistance in HNSCC whose therapeutic targeting in the context of multi-modality treatment approaches should be further examined and may be of particular interest for the subgroup of patients with elevated expression of the CXCR2/ligand axis.


Subject(s)
Cellular Senescence , Head and Neck Neoplasms , Radiation Tolerance , Receptors, Interleukin-8B , Squamous Cell Carcinoma of Head and Neck , Cell Line, Tumor , Cytokines , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/radiotherapy , Humans , Ligands , NF-kappa B , Receptors, Interleukin-8B/metabolism , Retrospective Studies , Squamous Cell Carcinoma of Head and Neck/radiotherapy
4.
PLoS Comput Biol ; 17(11): e1009515, 2021 11.
Article in English | MEDLINE | ID: mdl-34735429

ABSTRACT

Very high risk neuroblastoma is characterised by increased MAPK signalling, and targeting MAPK signalling is a promising therapeutic strategy. We used a deeply characterised panel of neuroblastoma cell lines and found that the sensitivity to MEK inhibitors varied drastically between these cell lines. By generating quantitative perturbation data and mathematical modelling, we determined potential resistance mechanisms. We found that negative feedbacks within MAPK signalling and via the IGF receptor mediate re-activation of MAPK signalling upon treatment in resistant cell lines. By using cell-line specific models, we predict that combinations of MEK inhibitors with RAF or IGFR inhibitors can overcome resistance, and tested these predictions experimentally. In addition, phospho-proteomic profiling confirmed the cell-specific feedback effects and synergy of MEK and IGFR targeted treatment. Our study shows that a quantitative understanding of signalling and feedback mechanisms facilitated by models can help to develop and optimise therapeutic strategies. Our findings should be considered for the planning of future clinical trials introducing MEKi in the treatment of neuroblastoma.


Subject(s)
Feedback , Models, Biological , Neuroblastoma/metabolism , Signal Transduction , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Humans , MAP Kinase Signaling System , Neuroblastoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 2/metabolism
5.
Adv Biol Regul ; 79: 100778, 2021 01.
Article in English | MEDLINE | ID: mdl-33431353

ABSTRACT

In colorectal cancer (CRC), the prevalence of NRAS mutations (5-9%) is inferior to that of KRAS mutations (40-50%). NRAS mutations feature lately during tumour progression and drive resistance to anti-EGFR therapy in KRAS wild-type tumours. To elucidate specific functions of NRAS mutations in CRC, we expressed doxycycline-inducible G12D and Q61K mutations in the CRC cell line Caco-2. A focused phospho-proteome analysis based on the Bio-Plex platform, which interrogated the activity of MAPK, PI3K, mTOR, STAT, p38, JNK and ATF2, did not reveal significant differences between Caco-2 cells expressing NRASG12D, NRASQ61K and KRASG12V. However, phenotypic read-outs were different. The NRAS Q61K mutation promoted anchorage-independent proliferation and tumorigenicity, similar to features driven by canonical KRAS mutations. In contrast, expression of NRASG12D resulted in reduced proliferation and apoptosis. At the transcriptome level, we saw upregulation of cytokines and chemokines. IL1A, IL11, CXCL8 (IL-8) and CCL20 exhibited enhanced secretion into the culture medium. In addition, RNA sequencing results indicated activation of the IL1-, JAK/STAT-, NFκB- and TNFα signalling pathways. These results form the basis for an NRASG12D-driven inflammatory phenotype in CRC.


Subject(s)
Colorectal Neoplasms/genetics , GTP Phosphohydrolases/genetics , Membrane Proteins/genetics , Apoptosis , Caco-2 Cells , Cell Proliferation , Chemokines/genetics , Chemokines/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/physiopathology , Cytokines/genetics , Cytokines/metabolism , GTP Phosphohydrolases/metabolism , Humans , Membrane Proteins/metabolism , Mutation , Oncogenes , Signal Transduction
6.
Mol Brain ; 13(1): 148, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33172478

ABSTRACT

Neuronal activity-regulated gene transcription underlies plasticity-dependent changes in the molecular composition and structure of neurons. A large number of genes regulated by different neuronal plasticity inducing pathways have been identified, but altered gene expression levels represent only part of the complexity of the activity-regulated transcriptional program. Alternative splicing, the differential inclusion and exclusion of exonic sequence in mRNA, is an additional mechanism that is thought to define the activity-dependent transcriptome. Here, we present a genome wide microarray-based survey to identify exons with increased expression levels at 1, 4 or 8 h following neuronal activity in the murine hippocampus provoked by generalized seizures. We used two different bioinformatics approaches to identify alternative activity-induced exon usage and to predict alternative splicing, ANOSVA (ANalysis Of Splicing VAriation) which we here adjusted to accommodate data from different time points and FIRMA (Finding Isoforms using Robust Multichip Analysis). RNA sequencing, in situ hybridization and reverse transcription PCR validate selected activity-dependent splicing events of previously described and so far undescribed activity-regulated transcripts, including Homer1a, Homer1d, Ania3, Errfi1, Inhba, Dclk1, Rcan1, Cda, Tpm1 and Krt75. Taken together, our survey significantly adds to the comprehensive understanding of the complex activity-dependent neuronal transcriptomic signature. In addition, we provide data sets that will serve as rich resources for future comparative expression analyses.


Subject(s)
Alternative Splicing/genetics , Exons/genetics , Neurons/metabolism , Animals , Male , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Reproducibility of Results
7.
Cell Rep ; 32(12): 108184, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32966782

ABSTRACT

Oncoproteins such as the BRAFV600E kinase endow cancer cells with malignant properties, but they also create unique vulnerabilities. Targeting of BRAFV600E-driven cytoplasmic signaling networks has proved ineffective, as patients regularly relapse with reactivation of the targeted pathways. We identify the nuclear protein SFPQ to be synthetically lethal with BRAFV600E in a loss-of-function shRNA screen. SFPQ depletion decreases proliferation and specifically induces S-phase arrest and apoptosis in BRAFV600E-driven colorectal and melanoma cells. Mechanistically, SFPQ loss in BRAF-mutant cancer cells triggers the Chk1-dependent replication checkpoint, results in decreased numbers and reduced activities of replication factories, and increases collision between replication and transcription. We find that BRAFV600E-mutant cancer cells and organoids are sensitive to combinations of Chk1 inhibitors and chemically induced replication stress, pointing toward future therapeutic approaches exploiting nuclear vulnerabilities induced by BRAFV600E.


Subject(s)
Colorectal Neoplasms/genetics , Mutation/genetics , PTB-Associated Splicing Factor/metabolism , Proto-Oncogene Proteins B-raf/genetics , Synthetic Lethal Mutations/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Checkpoint Kinase 1/metabolism , Colorectal Neoplasms/pathology , DNA Damage , DNA Repair/drug effects , DNA Repair/genetics , DNA Replication/drug effects , DNA Replication/genetics , Female , Humans , Hydroxyurea/pharmacology , Mice, Nude , Rad51 Recombinase/metabolism , Reproducibility of Results , S Phase/drug effects , S Phase/genetics , Stress, Physiological/drug effects , Tumor Suppressor p53-Binding Protein 1/metabolism
8.
Cell Death Dis ; 11(7): 499, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32612138

ABSTRACT

To unravel vulnerabilities of KRAS-mutant CRC cells, a shRNA-based screen specifically inhibiting MAPK pathway components and targets was performed in CaCo2 cells harboring conditional oncogenic KRASG12V. The custom-designed shRNA library comprised 121 selected genes, which were previously identified to be strongly regulated in response to MEK inhibition. The screen showed that CaCo2 cells expressing KRASG12V were sensitive to the suppression of the DNA replication licensing factor minichromosome maintenance complex component 7 (MCM7), whereas KRASwt CaCo2 cells were largely resistant to MCM7 suppression. Similar results were obtained in an isogenic DLD-1 cell culture model. Knockdown of MCM7 in a KRAS-mutant background led to replication stress as indicated by increased nuclear RPA focalization. Further investigation showed a significant increase in mitotic cells after simultaneous MCM7 knockdown and KRASG12V expression. The increased percentage of mitotic cells coincided with strongly increased DNA damage in mitosis. Taken together, the accumulation of DNA damage in mitotic cells is due to replication stress that remained unresolved, which results in mitotic catastrophe and cell death. In summary, the data show a vulnerability of KRAS-mutant cells towards suppression of MCM7 and suggest that inhibiting DNA replication licensing might be a viable strategy to target KRAS-mutant cancers.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Mitosis , Mutation/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Replication Origin , Caco-2 Cells , Cell Death , Cell Proliferation , Cellular Senescence , DNA Damage , DNA Replication , Gene Knockdown Techniques , Humans , Minichromosome Maintenance Complex Component 7/metabolism
9.
Nucleic Acids Res ; 48(W1): W307-W312, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32313938

ABSTRACT

Extracting signalling pathway activities from transcriptome data is important to infer mechanistic origins of transcriptomic dysregulation, for example in disease. A popular method to do so is by enrichment analysis of signature genes in e.g. differentially regulated genes. Previously, we derived signatures for signalling pathways by integrating public perturbation transcriptome data and generated a signature database called SPEED (Signalling Pathway Enrichment using Experimental Datasets), for which we here present a substantial upgrade as SPEED2. This web server hosts consensus signatures for 16 signalling pathways that are derived from a large number of transcriptomic signalling perturbation experiments. When providing a gene list of e.g. differentially expressed genes, the web server allows to infer signalling pathways that likely caused these genes to be deregulated. In addition to signature lists, we derive 'continuous' gene signatures, in a transparent and automated fashion without any fine-tuning, and describe a new algorithm to score these signatures.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , Signal Transduction , Software , Algorithms , Databases, Genetic
10.
Nat Commun ; 10(1): 2919, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31266962

ABSTRACT

Oncogenic mutations in KRAS or BRAF are frequent in colorectal cancer and activate the ERK kinase. Here, we find graded ERK phosphorylation correlating with cell differentiation in patient-derived colorectal cancer organoids with and without KRAS mutations. Using reporters, single cell transcriptomics and mass cytometry, we observe cell type-specific phosphorylation of ERK in response to transgenic KRASG12V in mouse intestinal organoids, while transgenic BRAFV600E activates ERK in all cells. Quantitative network modelling from perturbation data reveals that activation of ERK is shaped by cell type-specific MEK to ERK feed forward and negative feedback signalling. We identify dual-specificity phosphatases as candidate modulators of ERK in the intestine. Furthermore, we find that oncogenic KRAS, together with ß-Catenin, favours expansion of crypt cells with high ERK activity. Our experiments highlight key differences between oncogenic BRAF and KRAS in colorectal cancer and find unexpected heterogeneity in a signalling pathway with fundamental relevance for cancer therapy.


Subject(s)
Colonic Neoplasms/enzymology , Intestinal Mucosa/enzymology , Mitogen-Activated Protein Kinase Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase Kinases/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Species Specificity
11.
Life Sci Alliance ; 2(4)2019 08.
Article in English | MEDLINE | ID: mdl-31253656

ABSTRACT

Tumors of different molecular subtypes can show strongly deviating responses to drug treatment, making stratification of patients based on molecular markers an important part of cancer therapy. Pharmacogenomic studies have led to the discovery of selected genomic markers (e.g., BRAFV600E), whereas transcriptomic and proteomic markers so far have been largely absent in clinical use, thus constituting a potentially valuable resource for further substratification of patients. To systematically assess the explanatory power of different -omics data types, we assembled a panel of 49 melanoma cell lines, including genomic, transcriptomic, proteomic, and pharmacological data, showing that drug sensitivity models trained on transcriptomic or proteomic data outperform genomic-based models for most drugs. These results were confirmed in eight additional tumor types using published datasets. Furthermore, we show that drug sensitivity models can be transferred between tumor types, although after correcting for training sample size, transferred models perform worse than within-tumor-type predictions. Our results suggest that transcriptomic/proteomic signals may be alternative biomarker candidates for the stratification of patients without known genomic markers.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Proteome/drug effects , Transcriptome/drug effects , Biomarkers, Tumor/genetics , Cell Line, Tumor , Computer Simulation , Endometrial Neoplasms/metabolism , Female , Humans , Melanoma/genetics , Melanoma/metabolism , Models, Biological , PTEN Phosphohydrolase/metabolism , Proteome/genetics , Proteomics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Transcriptome/genetics , Exome Sequencing
12.
Mol Biol Cell ; 30(9): 1108-1117, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30785867

ABSTRACT

HRAS, NRAS, and KRAS isoforms are almost identical proteins that are ubiquitously expressed and activate a common set of effectors. In vivo studies have revealed that they are not biologically redundant; however, the isoform specificity of Ras signaling remains poorly understood. Using a novel panel of isogenic SW48 cell lines endogenously expressing wild-type or G12V-mutated activated Ras isoforms, we have performed a detailed characterization of endogenous isoform-specific mutant Ras signaling. We find that despite displaying significant Ras activation, the downstream outputs of oncogenic Ras mutants are minimal in the absence of growth factor inputs. The lack of mutant KRAS-induced effector activation observed in SW48 cells appears to be representative of a broad panel of colon cancer cell lines harboring mutant KRAS. For MAP kinase pathway activation in KRAS-mutant cells, the requirement for coincident growth factor stimulation occurs at an early point in the Raf activation cycle. Finally, we find that Ras isoform-specific signaling was highly context dependent and did not conform to the dogma derived from ectopic expression studies.


Subject(s)
ras Proteins/genetics , ras Proteins/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Genes, ras , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Mutation , Protein Isoforms , Signal Transduction/physiology
13.
Bioinformatics ; 34(17): i997-i1004, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30423075

ABSTRACT

Motivation: Signal-transduction networks are often aberrated in cancer cells, and new anti-cancer drugs that specifically target oncogenes involved in signaling show great clinical promise. However, the effectiveness of such targeted treatments is often hampered by innate or acquired resistance due to feedbacks, crosstalks or network adaptations in response to drug treatment. A quantitative understanding of these signaling networks and how they differ between cells with different oncogenic mutations or between sensitive and resistant cells can help in addressing this problem. Results: Here, we present Comparative Network Reconstruction (CNR), a computational method to reconstruct signaling networks based on possibly incomplete perturbation data, and to identify which edges differ quantitatively between two or more signaling networks. Prior knowledge about network topology is not required but can straightforwardly be incorporated. We extensively tested our approach using simulated data and applied it to perturbation data from a BRAF mutant, PTPN11 KO cell line that developed resistance to BRAF inhibition. Comparing the reconstructed networks of sensitive and resistant cells suggests that the resistance mechanism involves re-establishing wild-type MAPK signaling, possibly through an alternative RAF-isoform. Availability and implementation: CNR is available as a python module at https://github.com/NKI-CCB/cnr. Additionally, code to reproduce all figures is available at https://github.com/NKI-CCB/CNR-analyses. Supplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
Software , Neural Networks, Computer , Signal Transduction
14.
Essays Biochem ; 62(4): 535-547, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30315094

ABSTRACT

Gene regulatory networks control the cellular phenotype by changing the RNA and protein composition. Despite its importance, the gene regulatory network in higher organisms is only partly mapped out. Here, we investigate the potential of reverse engineering methods to unravel the structure of these networks. Particularly, we focus on modular response analysis (MRA), a method that can disentangle networks from perturbation data. We benchmark a version of MRA that was previously successfully applied to reconstruct a signalling-driven genetic network, termed MLMSMRA, to test cases mimicking various aspects of gene regulatory networks. We then investigate the performance in comparison with other MRA realisations and related methods. The benchmark shows that MRA has the potential to predict functional interactions, but also shows that successful application of MRA is restricted to small sparse networks and to data with a low signal-to-noise ratio.


Subject(s)
Gene Regulatory Networks , Genetic Engineering , Systems Biology , Computer Simulation , Signal-To-Noise Ratio
15.
Bioinformatics ; 34(23): 4079-4086, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29931053

ABSTRACT

Motivation: Intracellular signalling is realized by complex signalling networks, which are almost impossible to understand without network models, especially if feedbacks are involved. Modular Response Analysis (MRA) is a convenient modelling method to study signalling networks in various contexts. Results: We developed the software package STASNet (STeady-STate Analysis of Signalling Networks) that provides an augmented and extended version of MRA suited to model signalling networks from incomplete perturbation schemes and multi-perturbation data. Using data from the Dialogue on Reverse Engineering Assessment and Methods challenge, we show that predictions from STASNet models are among the top-performing methods. We applied the method to study the effect of SHP2, a protein that has been implicated in resistance to targeted therapy in colon cancer, using a novel dataset from the colon cancer cell line Widr and a SHP2-depleted derivative. We find that SHP2 is required for mitogen-activated protein kinase signalling, whereas AKT signalling only partially depends on SHP2. Availability and implementation: An R-package is available at https://github.com/molsysbio/STASNet. Supplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
Signal Transduction , Software , Cell Line, Tumor , Colonic Neoplasms , Computational Biology , Humans , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics
16.
Nat Commun ; 9(1): 20, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29295995

ABSTRACT

Aberrant cell signaling can cause cancer and other diseases and is a focal point of drug research. A common approach is to infer signaling activity of pathways from gene expression. However, mapping gene expression to pathway components disregards the effect of post-translational modifications, and downstream signatures represent very specific experimental conditions. Here we present PROGENy, a method that overcomes both limitations by leveraging a large compendium of publicly available perturbation experiments to yield a common core of Pathway RespOnsive GENes. Unlike pathway mapping methods, PROGENy can (i) recover the effect of known driver mutations, (ii) provide or improve strong markers for drug indications, and (iii) distinguish between oncogenic and tumor suppressor pathways for patient survival. Collectively, these results show that PROGENy accurately infers pathway activity from gene expression in a wide range of conditions.


Subject(s)
Gene Expression , Genes, Neoplasm , Genomics/methods , Neoplasms/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm , HEK293 Cells , Humans , Mutation , Neoplasms/mortality
18.
Mol Syst Biol ; 13(5): 928, 2017 05 03.
Article in English | MEDLINE | ID: mdl-28468958

ABSTRACT

The RAF-MEK-ERK signalling pathway controls fundamental, often opposing cellular processes such as proliferation and apoptosis. Signal duration has been identified to play a decisive role in these cell fate decisions. However, it remains unclear how the different early and late responding gene expression modules can discriminate short and long signals. We obtained both protein phosphorylation and gene expression time course data from HEK293 cells carrying an inducible construct of the proto-oncogene RAF By mathematical modelling, we identified a new gene expression module of immediate-late genes (ILGs) distinct in gene expression dynamics and function. We find that mRNA longevity enables these ILGs to respond late and thus translate ERK signal duration into response amplitude. Despite their late response, their GC-rich promoter structure suggested and metabolic labelling with 4SU confirmed that transcription of ILGs is induced immediately. A comparative analysis shows that the principle of duration decoding is conserved in PC12 cells and MCF7 cells, two paradigm cell systems for ERK signal duration. Altogether, our findings suggest that ILGs function as a gene expression module to decode ERK signal duration.


Subject(s)
Gene Expression Regulation , MAP Kinase Signaling System/genetics , RNA, Messenger/metabolism , Animals , Computer Simulation , GC Rich Sequence , HEK293 Cells , Half-Life , Humans , MCF-7 Cells , Models, Theoretical , Multigene Family , PC12 Cells , Promoter Regions, Genetic , Proto-Oncogene Mas , Rats , Signal Transduction/genetics , raf Kinases/genetics
19.
Cancer Res ; 77(12): 3364-3375, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28381545

ABSTRACT

Genomic features are used as biomarkers of sensitivity to kinase inhibitors used widely to treat human cancer, but effective patient stratification based on these principles remains limited in impact. Insofar as kinase inhibitors interfere with signaling dynamics, and, in turn, signaling dynamics affects inhibitor responses, we investigated associations in this study between cell-specific dynamic signaling pathways and drug sensitivity. Specifically, we measured 14 phosphoproteins under 43 different perturbed conditions (combinations of 5 stimuli and 7 inhibitors) in 14 colorectal cancer cell lines, building cell line-specific dynamic logic models of underlying signaling networks. Model parameters representing pathway dynamics were used as features to predict sensitivity to a panel of 27 drugs. Specific parameters of signaling dynamics correlated strongly with drug sensitivity for 14 of the drugs, 9 of which had no genomic biomarker. Following one of these associations, we validated a drug combination predicted to overcome resistance to MEK inhibitors by coblockade of GSK3, which was not found based on associations with genomic data. These results suggest that to better understand the cancer resistance and move toward personalized medicine, it is essential to consider signaling network dynamics that cannot be inferred from static genotypes. Cancer Res; 77(12); 3364-75. ©2017 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Signal Transduction/drug effects , Cell Line, Tumor , Humans , Models, Statistical , Protein Kinase Inhibitors/pharmacology
20.
Oncotarget ; 7(6): 6693-710, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26760764

ABSTRACT

Despite the approval of numerous molecular targeted drugs, long-term antiproliferative efficacy is rarely achieved and therapy resistance remains a central obstacle of cancer care. Combined inhibition of multiple cancer-driving pathways promises to improve antiproliferative efficacy. HIF-1 is a driver of gastric cancer and considered to be an attractive target for therapy. We noted that gastric cancer cells are able to functionally compensate the stable loss of HIF-1α. Via transcriptomics we identified a group of upregulated genes in HIF-1α-deficient cells and hypothesized that these genes confer survival upon HIF-1α loss. Strikingly, simultaneous knock-down of HIF-1α and Annexin A1 (ANXA1), one of the identified genes, resulted in complete cessation of proliferation. Using stable isotope-resolved metabolomics, oxidative and reductive glutamine metabolism was found to be significantly impaired in HIF-1α/ANXA1-deficient cells, potentially explaining the proliferation defect. In summary, we present a conceptually novel application of stable gene inactivation enabling in-depth deconstruction of resistance mechanisms. In theory, this experimental approach is applicable to any cancer-driving gene or pathway and promises to identify various new targets for combination therapies.


Subject(s)
Annexin A1/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Stomach Neoplasms/metabolism , Animals , Annexin A1/genetics , Cell Line, Tumor , Cell Proliferation/physiology , Female , Heterografts , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunohistochemistry , Mice , Mice, Inbred NOD , Mice, SCID , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...