Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Nat Cancer ; 4(2): 276-289, 2023 02.
Article in English | MEDLINE | ID: mdl-36702933

ABSTRACT

Analysis of mutational signatures can reveal underlying molecular mechanisms of the processes that have imprinted the somatic mutations found in cancer genomes. Here, we analyze single base substitutions and small insertions and deletions in pediatric cancers encompassing 785 whole-genome sequenced tumors from 27 molecularly defined cancer subtypes. We identified only a small number of mutational signatures active in pediatric cancers, compared with previously analyzed adult cancers. Further, we report a significant difference in the proportion of pediatric tumors showing homologous recombination repair defect signatures compared with previous analyses. In pediatric leukemias, we identified an indel signature, not previously reported, characterized by long insertions in nonrepeat regions, affecting mainly intronic and intergenic regions, but also exons of known cancer genes. We provide a systematic overview of COSMIC v.3 mutational signatures active across pediatric cancers, which is highly relevant for understanding tumor biology and enabling future research in defining biomarkers of treatment response.


Subject(s)
Neoplasms , Adult , Humans , Child , Mutation , Neoplasms/genetics , Oncogenes , INDEL Mutation , DNA Repair
2.
Br J Haematol ; 192(5): 832-842, 2021 03.
Article in English | MEDLINE | ID: mdl-33529373

ABSTRACT

In acute myeloid leukaemia (AML) t(8;16)(p11;p13)/MYST3-CREBBP is a very rare abnormality. Previous small series suggested poor outcome. We report on 59 patients with t(8;16) within an international, collaborative study. Median age was 52 (range: 16-75) years. AML was de novo in 58%, therapy-related (t-AML) in 37% and secondary after myelodysplastic syndrome (s-AML) in 5%. Cytogenetics revealed a complex karyotype in 43%. Besides MYST3-CREBBP, whole-genome sequencing on a subset of 10 patients revealed recurrent mutations in ASXL1, BRD3, FLT3, MLH1, POLG, TP53, SAMD4B (n = 3, each), EYS, KRTAP9-1 SPTBN5 (n = 4, each), RUNX1 and TET2 (n = 2, each). Complete remission after intensive chemotherapy was achieved in 84%. Median follow-up was 5·48 years; five-year survival rate was 17%. Patients with s-/t-AML (P = 0·01) and those with complex karyotype (P = 0·04) had an inferior prognosis. Allogeneic haematopoietic cell transplantation (allo-HCT) was performed in 21 (36%) patients, including 15 in first complete remission (CR1). Allo-HCT in CR1 significantly improved survival (P = 0·04); multivariable analysis revealed that allo-HCT in CR1 was effective in de novo AML but not in patients with s-AML/t-AML and less in patients exhibiting a complex karyotype. In summary, outcomes of patients with t(8;16) are dismal with chemotherapy, and may be substantially improved with allo-HCT performed in CR1.


Subject(s)
Chromosomes, Human, Pair 16/ultrastructure , Chromosomes, Human, Pair 8/ultrastructure , Leukemia, Myeloid, Acute/genetics , Translocation, Genetic , Abnormal Karyotype , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Combined Modality Therapy , Consolidation Chemotherapy , Disease Progression , Disease-Free Survival , Female , Follow-Up Studies , Hematopoietic Stem Cell Transplantation , Humans , International Cooperation , Leukemia, Myeloid, Acute/epidemiology , Leukemia, Myeloid, Acute/therapy , Male , Middle Aged , Mutation , Myelodysplastic Syndromes/epidemiology , Neoplasms, Second Primary/chemically induced , Neoplasms, Second Primary/epidemiology , Oncogene Proteins, Fusion/genetics , Remission Induction , Survival Analysis , Whole Genome Sequencing
3.
R Soc Open Sci ; 7(9): 200441, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33047019

ABSTRACT

To elucidate and to inhibit post-surgical fibrotic processes after trabeculectomy in glaucoma therapy, we measured gene expression in a fibrotic cell culture model, based on transforming growth factor TGF-ß induction in primary human tenon fibroblasts (hTFs), and used Connectivity Map (CMap) data for drug repositioning. We found that specific molecular mechanisms behind fibrosis are the upregulation of actins, the downregulation of CD34, and the upregulation of inflammatory cytokines such as IL6, IL11 and BMP6. The macrolide antibiotic Josamycin (JM) reverses these molecular mechanisms according to data from the CMap, and we thus tested JM as an inhibitor of fibrosis. JM was first tested for its toxic effects on hTFs, where it showed no influence on cell viability, but inhibited hTF proliferation in a concentration-dependent manner. We then demonstrated that JM suppresses the synthesis of extracellular matrix (ECM) components. In hTFs stimulated with TGF-ß1, JM specifically inhibited α-smooth muslce actin expression, suggesting that it inhibits the transformation of fibroblasts into fibrotic myofibroblasts. In addition, a decrease of components of the ECM such as fibronectin, which is involved in in vivo scarring, was observed. We conclude that JM may be a promising candidate for the treatment of fibrosis after glaucoma filtration surgery or drainage device implantation in vivo.

4.
Oncogene ; 39(15): 3128-3144, 2020 04.
Article in English | MEDLINE | ID: mdl-32055024

ABSTRACT

NOTCH receptor signaling plays a pivotal role in liver homeostasis and hepatocarcinogenesis. However, the role of NOTCH pathway mutations and the NOTCH target gene HES5 in liver tumorigenesis are poorly understood. Here we performed whole-exome sequencing of 54 human HCC specimens and compared the prevalence of NOTCH pathway component mutations with the TCGA-LIHC cohort (N = 364). In addition, we functionally characterized the NOTCH target HES5 and the patient-derived HES5-R31G mutation in vitro and in an orthotopic mouse model applying different oncogenic backgrounds, to dissect the role of HES5 in different tumor subgroups in vivo. We identified nonsynonymous mutations in 14 immediate NOTCH pathway genes affecting 24.1% and 16.8% of HCC patients in the two independent cohorts, respectively. Among these, the HES5-R31G mutation was predicted in silico to have high biological relevance. Functional analyses in cell culture showed that HES5 reduced cell migration and clonogenicity. Further analyses revealed that the patient-derived HES5-R31G mutant protein was non-functional due to loss of DNA binding and greatly reduced nuclear localization. Furthermore, HES5 exhibited a negative feedback loop by directly inhibiting the NOTCH target HES1 and downregulated the pro-proliferative MYC targets ODC1 and LDHA. Interestingly, HES5 inhibited MYC-dependent hepatocarcinogenesis, whereas it promoted AKT-dependent liver tumor formation and stem cell features in a murine model. Thus, NOTCH pathway component mutations are commonly observed in HCC. Furthermore, the NOTCH target gene HES5 has both pro- and anti-tumorigenic functions in liver cancer proposing a driver gene dependency and it promotes tumorigenesis with its interaction partner AKT.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Receptors, Notch/metabolism , Repressor Proteins/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Cohort Studies , DNA Mutational Analysis , Female , Humans , Liver/pathology , Liver Neoplasms/pathology , Male , Mice , Mutation , Proto-Oncogene Proteins c-akt/metabolism , Repressor Proteins/metabolism , Signal Transduction/genetics , Exome Sequencing , Xenograft Model Antitumor Assays
5.
Oncogene ; 39(4): 814-832, 2020 01.
Article in English | MEDLINE | ID: mdl-31558800

ABSTRACT

Fusion proteins involving the BRAF serine/threonine kinase occur in many cancers. The oncogenic potential of BRAF fusions has been attributed to the loss of critical N-terminal domains that mediate BRAF autoinhibition. We used whole-exome and RNA sequencing in a patient with glioblastoma multiforme to identify a rearrangement between TTYH3, encoding a membrane-resident, calcium-activated chloride channel, and BRAF intron 1, resulting in a TTYH3-BRAF fusion protein that retained all features essential for BRAF autoinhibition. Accordingly, the BRAF moiety of the fusion protein alone, which represents full-length BRAF without the amino acids encoded by exon 1 (BRAFΔE1), did not induce MEK/ERK phosphorylation or transformation. Likewise, neither the TTYH3 moiety of the fusion protein nor full-length TTYH3 provoked ERK pathway activity or transformation. In contrast, TTYH3-BRAF displayed increased MEK phosphorylation potential and transforming activity, which were caused by TTYH3-mediated tethering of near-full-length BRAF to the (endo)membrane system. Consistent with this mechanism, a synthetic approach, in which BRAFΔE1 was tethered to the membrane by fusing it to the cytoplasmic tail of CD8 also induced transformation. Furthermore, we demonstrate that TTYH3-BRAF signals largely independent of a functional RAS binding domain, but requires an intact BRAF dimer interface and activation loop phosphorylation sites. Cells expressing TTYH3-BRAF exhibited increased MEK/ERK signaling, which was blocked by clinically achievable concentrations of sorafenib, trametinib, and the paradox breaker PLX8394. These data provide the first example of a fully autoinhibited BRAF protein whose oncogenic potential is dictated by a distinct fusion partner and not by a structural change in BRAF itself.


Subject(s)
Glioblastoma/genetics , Glioblastoma/pathology , Heterocyclic Compounds, 2-Ring/pharmacology , Oncogene Proteins, Fusion , Proto-Oncogene Proteins B-raf/genetics , Pyridones/pharmacology , Pyrimidinones/pharmacology , Sulfonamides/pharmacology , Aged , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Chloride Channels/genetics , Chloride Channels/metabolism , Female , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , MAP Kinase Signaling System/drug effects , Phosphorylation , Protein Domains , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Signal Transduction
6.
Biosci Rep ; 39(5)2019 05 31.
Article in English | MEDLINE | ID: mdl-30967499

ABSTRACT

We aimed to identify differences in cytokine/chemokine levels in the aqueous humor (AH) of primary open-angle glaucoma (POAG) patients who suffered from scarring, compared with POAG patients with no scarring after trabeculectomy surgery. Identification of differently expressed cytokines and chemokines may help to understand scarring and fibrotic processes following trabeculectomy, and to make predictions for the outcome of fistulating surgery in the future. Furthermore, the identification of cell signaling pathways involved in fibrosis offers the opportunity for a more specific antifibrotic therapy with reduced side effects, and an improvement in long-term surgical outcome.Eight samples of AH were collected during trabeculectomy surgery and commercially available cytokine/chemokine arrays were used. Specific, differently expressed proteins (cytokines/chemokines) in AH samples from patients with positive and negative surgery outcomes were detected. These proteins were classified based on their known profibrotic, inflammatory, adhesive, and apoptotic properties. Transforming growth factor ß (TGF-ß) and vascular endothelial growth factor (VEGF) were among the most important profibrotic cytokines that we detected. Differences in the fold change of protein expression were highly significant between patients after successful and failed trabeculectomy surgery, and these were processed and visualized using ExprEssence software.This pilot study revealed differences in concentrations of cytokines/chemokines in AH between the two examined groups of patients. Our findings suggest that a positive outcome from trabeculectomy is strongly related to an inhibition of the fibrosis process.


Subject(s)
Chemokines/genetics , Cytokines/genetics , Glaucoma, Open-Angle/surgery , Trabeculectomy/adverse effects , Adolescent , Adult , Aged , Aqueous Humor/metabolism , Child , Female , Glaucoma, Open-Angle/genetics , Glaucoma, Open-Angle/metabolism , Glaucoma, Open-Angle/pathology , Humans , Male , Middle Aged , Pilot Projects , Transforming Growth Factor beta/genetics , Vascular Endothelial Growth Factor A/genetics
7.
Nat Commun ; 10(1): 1635, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30967556

ABSTRACT

Chordomas are rare bone tumors with few therapeutic options. Here we show, using whole-exome and genome sequencing within a precision oncology program, that advanced chordomas (n = 11) may be characterized by genomic patterns indicative of defective homologous recombination (HR) DNA repair and alterations affecting HR-related genes, including, for example, deletions and pathogenic germline variants of BRCA2, NBN, and CHEK2. A mutational signature associated with HR deficiency was significantly enriched in 72.7% of samples and co-occurred with genomic instability. The poly(ADP-ribose) polymerase (PARP) inhibitor olaparib, which is preferentially toxic to HR-incompetent cells, led to prolonged clinical benefit in a patient with refractory chordoma, and whole-genome analysis at progression revealed a PARP1 p.T910A mutation predicted to disrupt the autoinhibitory PARP1 helical domain. These findings uncover a therapeutic opportunity in chordoma that warrants further exploration, and provide insight into the mechanisms underlying PARP inhibitor resistance.


Subject(s)
Chordoma/drug therapy , Phthalazines/therapeutic use , Piperazines/therapeutic use , Poly (ADP-Ribose) Polymerase-1/genetics , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Recombinational DNA Repair/genetics , Adult , Aged , Chordoma/genetics , Chordoma/pathology , Chromosome Mapping , DNA Breaks, Double-Stranded , DNA Mutational Analysis , Drug Resistance, Neoplasm/genetics , Female , Genomic Instability , Humans , Male , Middle Aged , Phthalazines/pharmacology , Piperazines/pharmacology , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Precision Medicine/methods , Protein Domains/genetics , Treatment Outcome , Exome Sequencing
8.
Sci Rep ; 8(1): 7477, 2018 05 10.
Article in English | MEDLINE | ID: mdl-29748622

ABSTRACT

A venous tumor thrombus (VTT) is a potentially lethal complication of renal cell carcinoma (RCC) but virtually nothing is known about the underlying natural history. Based on our observation that venous thrombi contain significant numbers of viable tumor cells, we applied multiregion whole exome sequencing to a total of 37 primary tumor and VTT samples including normal tissue specimens from five consecutive patients. Our findings demonstrate mutational heterogeneity between primary tumor and VTT with 106 of 483 genes (22%) harboring functional SNVs and/or indels altered in either primary tumor or thrombus. Reconstruction of the clonal phylogeny showed clustering of tumor samples and VTT samples, respectively, in the majority of tumors. However, no new subclones were detected suggesting that pre-existing subclones of the primary tumor drive VTT formation. Importantly, we found several lines of evidence for "BRCAness" in a subset of tumors. These included mutations in genes that confer "BRCAness", a mutational signature and an increase of small indels. Re-analysis of SNV calls from the TCGA KIRC-US cohort confirmed a high frequency of the "BRCAness" mutational signature AC3 in clear cell RCC. Our findings warrant further pre-clinical experiments and may lead to novel personalized therapies for RCC patients.


Subject(s)
Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Renal Veins/pathology , Transcriptome , Venous Thrombosis/genetics , Aged , Aged, 80 and over , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/complications , Carcinoma, Renal Cell/pathology , Cohort Studies , DNA Mutational Analysis , Female , Gene Expression Regulation, Neoplastic , Genomics , Humans , Kidney Neoplasms/blood supply , Kidney Neoplasms/complications , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis , Venous Thrombosis/complications , Venous Thrombosis/pathology , Exome Sequencing
10.
Sci Rep ; 7(1): 8576, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28819298

ABSTRACT

Accumulating evidence suggests that dysregulation of hypoxia-regulated transcriptional mechanisms is involved in development of chronic kidney diseases (CKD). However, it remains unclear how hypoxia-induced transcription factors (HIFs) and subsequent biological processes contribute to CKD development and progression. In our study, genome-wide expression profiles of more than 200 renal biopsies from patients with different CKD stages revealed significant correlation of HIF-target genes with eGFR in glomeruli and tubulointerstitium. These correlations were positive and negative and in part compartment-specific. Microarrays of proximal tubular cells and podocytes with stable HIF1α and/or HIF2α suppression displayed cell type-specific HIF1/HIF2-dependencies as well as dysregulation of several pathways. WGCNA analysis identified gene sets that were highly coregulated within modules. Characterization of the modules revealed common as well as cell group- and condition-specific pathways, GO-Terms and transcription factors. Gene expression analysis of the hypoxia-interconnected pathways in patients with different CKD stages revealed an increased dysregulation with loss of renal function. In conclusion, our data clearly point to a compartment- and cell type-specific dysregulation of hypoxia-associated gene transcripts and might help to improve the understanding of hypoxia, HIF dysregulation, and transcriptional program response in CKD.


Subject(s)
Gene Regulatory Networks , Kidney/metabolism , Podocytes/metabolism , Transcriptome , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia , Cell Line , Cells, Cultured , Gene Expression Profiling , Gene Ontology , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology
11.
Sci Rep ; 7: 42638, 2017 02 16.
Article in English | MEDLINE | ID: mdl-28205611

ABSTRACT

To identify genes contributing to disease phenotypes remains a challenge for bioinformatics. Static knowledge on biological networks is often combined with the dynamics observed in gene expression levels over disease development, to find markers for diagnostics and therapy, and also putative disease-modulatory drug targets and drugs. The basis of current methods ranges from a focus on expression-levels (Limma) to concentrating on network characteristics (PageRank, HITS/Authority Score), and both (DeMAND, Local Radiality). We present an integrative approach (the FocusHeuristics) that is thoroughly evaluated based on public expression data and molecular disease characteristics provided by DisGeNet. The FocusHeuristics combines three scores, i.e. the log fold change and another two, based on the sum and difference of log fold changes of genes/proteins linked in a network. A gene is kept when one of the scores to which it contributes is above a threshold. Our FocusHeuristics is both, a predictor for gene-disease-association and a bioinformatics method to reduce biological networks to their disease-relevant parts, by highlighting the dynamics observed in expression data. The FocusHeuristics is slightly, but significantly better than other methods by its more successful identification of disease-associated genes measured by AUC, and it delivers mechanistic explanations for its choice of genes.


Subject(s)
Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Genetic Predisposition to Disease , Algorithms , Biomarkers , Cluster Analysis , Humans , Leukemia, Megakaryoblastic, Acute/diagnosis , Leukemia, Megakaryoblastic, Acute/genetics , Leukemia, Megakaryoblastic, Acute/metabolism , Prognosis , ROC Curve
12.
Nat Commun ; 7: ncomms11845, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27291893

ABSTRACT

Intratumoural heterogeneity (ITH) is a major cause of cancer-associated lethality. Extensive genomic ITH has previously been reported in clear cell renal cell carcinoma (ccRCC). Here we address the question whether ITH increases with malignant progression and can hence be exploited as a prognostic marker. Unexpectedly, precision quantitative image analysis reveals that the degree of functional ITH is virtually identical between primary ccRCCs of the lowest stage and advanced, metastatic tumours. Functional ITH was found to show a stage-independent topological pattern with peak proliferative and signalling activities almost exclusively in the tumour periphery. Exome sequencing of matching peripheral and central primary tumour specimens reveals various region-specific mutations. However, these mutations cannot directly explain the zonal pattern suggesting a role of microenvironmental factors in shaping functional ITH. In conclusion, our results indicate that ITH is an early and general characteristic of malignant growth rather than a consequence of malignant progression.


Subject(s)
Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Genetic Heterogeneity , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Tumor Microenvironment , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Humans , Mutation/genetics , Neoplasm Staging , Phenotype , Prognosis , Signal Transduction , Exome Sequencing
13.
PLoS One ; 8(12): e81784, 2013.
Article in English | MEDLINE | ID: mdl-24349128

ABSTRACT

In silico approaches are increasingly considered to improve breast cancer treatment. One of these treatments, neoadjuvant TFAC chemotherapy, is used in cases where application of preoperative systemic therapy is indicated. Estimating response to treatment allows or improves clinical decision-making and this, in turn, may be based on a good understanding of the underlying molecular mechanisms. Ever increasing amounts of high throughput data become available for integration into functional networks. In this study, we applied our software tool ExprEssence to identify specific mechanisms relevant for TFAC therapy response, from a gene/protein interaction network. We contrasted the resulting active subnetwork to the subnetworks of two other such methods, OptDis and KeyPathwayMiner. We could show that the ExprEssence subnetwork is more related to the mechanistic functional principles of TFAC therapy than the subnetworks of the other two methods despite the simplicity of ExprEssence. We were able to validate our method by recovering known mechanisms and as an application example of our method, we identified a mechanism that may further explain the synergism between paclitaxel and doxorubicin in TFAC treatment: Paclitaxel may attenuate MELK gene expression, resulting in lower levels of its target MYBL2, already associated with doxorubicin synergism in hepatocellular carcinoma cell lines. We tested our hypothesis in three breast cancer cell lines, confirming it in part. In particular, the predicted effect on MYBL2 could be validated, and a synergistic effect of paclitaxel and doxorubicin could be demonstrated in the breast cancer cell lines SKBR3 and MCF-7.


Subject(s)
Breast Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks , Neoadjuvant Therapy , Software , Biomarkers, Pharmacological/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Doxorubicin/administration & dosage , Drug Synergism , Female , Humans , Paclitaxel/administration & dosage , Protein Interaction Mapping , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
14.
Kidney Int ; 84(1): 104-15, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23552858

ABSTRACT

Interactions between proteins crucially determine cellular structure and function. Differential analysis of the interactome may help elucidate molecular mechanisms during disease development; however, this analysis necessitates mapping of expression data on protein-protein interaction networks. These networks do not exist for the podocyte; therefore, we built PodNet, a literature-based mouse podocyte network in Cytoscape format. Using database protein-protein interactions, we expanded PodNet to XPodNet with enhanced connectivity. In order to test the performance of XPodNet in differential interactome analysis, we examined podocyte developmental differentiation and the effect of cell culture. Transcriptomes of podocytes in 10 different states were mapped on XPodNet and analyzed with the Cytoscape plugin ExprEssence, based on the law of mass action. Interactions between slit diaphragm proteins are most significantly upregulated during podocyte development and most significantly downregulated in culture. On the other hand, our analysis revealed that interactions lost during podocyte differentiation are not regained in culture, suggesting a loss rather than a reversal of differentiation for podocytes in culture. Thus, we have developed PodNet as a valuable tool for differential interactome analysis in podocytes, and we have identified established and unexplored regulated interactions in developing and cultured podocytes.


Subject(s)
Cell Differentiation , Databases, Protein , Podocytes/metabolism , Protein Interaction Mapping , Protein Interaction Maps , Proteins/metabolism , Animals , Cells, Cultured , Gene Expression Regulation , Mice , Principal Component Analysis , Proteins/genetics , Signal Transduction
15.
PLoS One ; 7(4): e34907, 2012.
Article in English | MEDLINE | ID: mdl-22529955

ABSTRACT

Parietal epithelial cells (PECs) are crucially involved in the pathogenesis of rapidly progressive glomerulonephritis (RPGN) as well as in focal and segmental glomerulosclerosis (FSGS). In this study, transgenic mouse lines were used to isolate pure, genetically tagged primary cultures of PECs or podocytes using FACsorting. By this approach, the morphology of primary glomerular epithelial cells in culture could be resolved: Primary podocytes formed either large cells with intracytoplasmatic extensions or smaller spindle shaped cells, depending on specific culture conditions. Primary PECs were small and exhibited a spindle-shaped or polygonal morphology. In the very early phases of primary culture, rapid changes in gene expression (e.g. of WT-1 and Pax-2) were observed. However, after prolonged culture primary PECs and podocytes still segregated clearly in a transcriptome analysis--demonstrating that the origin of primary cell cultures is important. Of the classical markers, synaptopodin and podoplanin expression were differentially regulated the most in primary PEC and podocyte cultures. However, no expression of any endogenous gene allowed to differentiate between the two cell types in culture. Finally, we show that the transcription factor WT1 is also expressed by PECs. In summary, genetic tagging of PECs and podocytes is a novel and necessary tool to derive pure primary cultures with proven origin. These cultures will be a powerful tool for the emerging field of parietal epithelial cell biology.


Subject(s)
Podocytes/cytology , Podocytes/metabolism , Animals , Cell Lineage , Cluster Analysis , Female , Gene Expression Profiling , Gene Order , Kidney Glomerulus/cytology , Kidney Glomerulus/growth & development , Kidney Glomerulus/metabolism , Mice , Mice, Transgenic , PAX2 Transcription Factor/genetics , PAX2 Transcription Factor/metabolism , Primary Cell Culture , Transcriptome , WT1 Proteins/genetics , WT1 Proteins/metabolism
16.
BMC Syst Biol ; 4: 164, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-21118483

ABSTRACT

BACKGROUND: Experimentalists are overwhelmed by high-throughput data and there is an urgent need to condense information into simple hypotheses. For example, large amounts of microarray and deep sequencing data are becoming available, describing a variety of experimental conditions such as gene knockout and knockdown, the effect of interventions, and the differences between tissues and cell lines. RESULTS: To address this challenge, we developed a method, implemented as a Cytoscape plugin called ExprEssence. As input we take a network of interaction, stimulation and/or inhibition links between genes/proteins, and differential data, such as gene expression data, tracking an intervention or development in time. We condense the network, highlighting those links across which the largest changes can be observed. Highlighting is based on a simple formula inspired by the law of mass action. We can interactively modify the threshold for highlighting and instantaneously visualize results. We applied ExprEssence to three scenarios describing kidney podocyte biology, pluripotency and ageing: 1) We identify putative processes involved in podocyte (de-)differentiation and validate one prediction experimentally. 2) We predict and validate the expression level of a transcription factor involved in pluripotency. 3) Finally, we generate plausible hypotheses on the role of apoptosis, cell cycle deregulation and DNA repair in ageing data obtained from the hippocampus. CONCLUSION: Reducing the size of gene/protein networks to the few links affected by large changes allows to screen for putative mechanistic relationships among the genes/proteins that are involved in adaptation to different experimental conditions, yielding important hypotheses, insights and suggestions for new experiments. We note that we do not focus on the identification of 'active subnetworks'. Instead we focus on the identification of single links (which may or may not form subnetworks), and these single links are much easier to validate experimentally than submodules. ExprEssence is available at http://sourceforge.net/projects/expressence/.


Subject(s)
Computational Biology/methods , Gene Regulatory Networks , Proteins/metabolism , Software , Aging/genetics , Aging/metabolism , Animals , Cell Line , Humans , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Podocytes/metabolism , Proteins/genetics , Reproducibility of Results
17.
PLoS One ; 5(12): e15165, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-21179244

ABSTRACT

BACKGROUND: Analysis of the mechanisms underlying pluripotency and reprogramming would benefit substantially from easy access to an electronic network of genes, proteins and mechanisms. Moreover, interpreting gene expression data needs to move beyond just the identification of the up-/downregulation of key genes and of overrepresented processes and pathways, towards clarifying the essential effects of the experiment in molecular terms. METHODOLOGY/PRINCIPAL FINDINGS: We have assembled a network of 574 molecular interactions, stimulations and inhibitions, based on a collection of research data from 177 publications until June 2010, involving 274 mouse genes/proteins, all in a standard electronic format, enabling analyses by readily available software such as Cytoscape and its plugins. The network includes the core circuit of Oct4 (Pou5f1), Sox2 and Nanog, its periphery (such as Stat3, Klf4, Esrrb, and c-Myc), connections to upstream signaling pathways (such as Activin, WNT, FGF, BMP, Insulin, Notch and LIF), and epigenetic regulators as well as some other relevant genes/proteins, such as proteins involved in nuclear import/export. We describe the general properties of the network, as well as a Gene Ontology analysis of the genes included. We use several expression data sets to condense the network to a set of network links that are affected in the course of an experiment, yielding hypotheses about the underlying mechanisms. CONCLUSIONS/SIGNIFICANCE: We have initiated an electronic data repository that will be useful to understand pluripotency and to facilitate the interpretation of high-throughput data. To keep up with the growth of knowledge on the fundamental processes of pluripotency and reprogramming, we suggest to combine Wiki and social networking software towards a community curation system that is easy to use and flexible, and tailored to provide a benefit for the scientist, and to improve communication and exchange of research results. A PluriNetWork tutorial is available at http://www.ibima.med.uni-rostock.de/IBIMA/PluriNetWork/.


Subject(s)
Pluripotent Stem Cells/cytology , Algorithms , Animals , Cell Differentiation/genetics , Epigenomics , Gene Expression Profiling , Gene Expression Regulation , Gene Expression Regulation, Developmental , Kruppel-Like Factor 4 , Mice , Models, Biological , Models, Genetic , Protein Binding/genetics , Signal Transduction , Software , Transcription, Genetic
18.
Mol Genet Genomics ; 278(3): 317-30, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17588177

ABSTRACT

In the yeast Saccharomyces cerevisiae, structural genes of phospholipid biosynthesis are activated by a heterodimer of basic helix-loop-helix proteins, Ino2 and Ino4, which bind to the inositol/choline-responsive element (ICRE) UAS element. In silico, we identified Candida albicans genes, which encode proteins similar to Ino2 and Ino4 (designated CaIno2 and CaIno4). CaINO4 contains an intron with an unusual branch point sequence. Although neither CaINO2 nor CaINO4 could individually complement S. cerevisiae mutations ino2 and ino4, respectively, coexpression of both CaINO2 and CaINO4 restored inositol auxotrophy of an ino2 ino4 double mutant. CaIno2 and CaIno4 could interact in vivo as well as in vitro and together were able to bind to the ICRE from S. cerevisiae INO1. Similar to Ino2 of S. cerevisiae, CaIno2 contains two transcriptional activation domains. CaIno2 and CaIno4 interact with CaSua7 (basal transcription factor TFIIB) but not with Sua7 from S. cerevisiae. Surprisingly, CaIno2 + CaIno4 were unable to stimulate expression of a CaINO1-lacZ reporter gene while an INO1-lacZ fusion was efficiently activated. This result agrees with the finding that promoter scanning of the CaINO1 upstream region gave no evidence for CaIno2 + CaIno4 binding in vitro. We derived a consensus binding site for CaIno2 + CaIno4 (BWTCASRTG), which could be detected upstream of 25 ribosomal protein genes. Since we failed to obtain homozygous deletion mutations for CaINO2 and CaINO4, we conclude that CaIno2 and CaIno4 acquired new essential target genes among which may be ribosomal protein genes.


Subject(s)
Candida albicans/genetics , Candida albicans/metabolism , Fungal Proteins/genetics , Fungal Proteins/metabolism , Genes, Fungal , Ribosomal Proteins/genetics , Trans-Activators/metabolism , Amino Acid Sequence , Base Sequence , Basic Helix-Loop-Helix Transcription Factors , Binding Sites/genetics , DNA, Fungal/genetics , DNA, Fungal/metabolism , Gene Expression Regulation, Fungal , Genetic Complementation Test , Models, Genetic , Molecular Sequence Data , Promoter Regions, Genetic , Repressor Proteins/genetics , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Homology, Amino Acid , Trans-Activators/genetics , Transcription Factor TFIIB/genetics , Transcription Factor TFIIB/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...