Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
Add more filters

Publication year range
1.
Cell ; 167(5): 1398-1414.e24, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27863251

ABSTRACT

Characterizing the multifaceted contribution of genetic and epigenetic factors to disease phenotypes is a major challenge in human genetics and medicine. We carried out high-resolution genetic, epigenetic, and transcriptomic profiling in three major human immune cell types (CD14+ monocytes, CD16+ neutrophils, and naive CD4+ T cells) from up to 197 individuals. We assess, quantitatively, the relative contribution of cis-genetic and epigenetic factors to transcription and evaluate their impact as potential sources of confounding in epigenome-wide association studies. Further, we characterize highly coordinated genetic effects on gene expression, methylation, and histone variation through quantitative trait locus (QTL) mapping and allele-specific (AS) analyses. Finally, we demonstrate colocalization of molecular trait QTLs at 345 unique immune disease loci. This expansive, high-resolution atlas of multi-omics changes yields insights into cell-type-specific correlation between diverse genomic inputs, more generalizable correlations between these inputs, and defines molecular events that may underpin complex disease risk.


Subject(s)
Epigenomics , Immune System Diseases/genetics , Monocytes/metabolism , Neutrophils/metabolism , T-Lymphocytes/metabolism , Transcription, Genetic , Adult , Aged , Alternative Splicing , Female , Genetic Predisposition to Disease , Hematopoietic Stem Cells/metabolism , Histone Code , Humans , Male , Middle Aged , Quantitative Trait Loci , Young Adult
2.
Immunity ; 53(4): 824-839.e10, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33053331

ABSTRACT

CD8+ T cells within the tumor microenvironment (TME) are exposed to various signals that ultimately determine functional outcomes. Here, we examined the role of the co-activating receptor CD226 (DNAM-1) in CD8+ T cell function. The absence of CD226 expression identified a subset of dysfunctional CD8+ T cells present in peripheral blood of healthy individuals. These cells exhibited reduced LFA-1 activation, altered TCR signaling, and a distinct transcriptomic program upon stimulation. CD226neg CD8+ T cells accumulated in human and mouse tumors of diverse origin through an antigen-specific mechanism involving the transcriptional regulator Eomesodermin (Eomes). Despite similar expression of co-inhibitory receptors, CD8+ tumor-infiltrating lymphocyte failed to respond to anti-PD-1 in the absence of CD226. Immune checkpoint blockade efficacy was hampered in Cd226-/- mice. Anti-CD137 (4-1BB) agonists also stimulated Eomes-dependent CD226 loss that limited the anti-tumor efficacy of this treatment. Thus, CD226 loss restrains CD8+ T cell function and limits the efficacy of cancer immunotherapy.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/immunology , CD8-Positive T-Lymphocytes/immunology , Neoplasms/immunology , T-Box Domain Proteins/immunology , Animals , Humans , Immune Checkpoint Inhibitors/immunology , Immunotherapy/methods , Mice , Mice, Inbred C57BL , Neoplasms/therapy , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , Transcriptome/immunology , Tumor Microenvironment/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
3.
PLoS Comput Biol ; 19(8): e1011254, 2023 08.
Article in English | MEDLINE | ID: mdl-37561790

ABSTRACT

Inference of gene regulatory networks has been an active area of research for around 20 years, leading to the development of sophisticated inference algorithms based on a variety of assumptions and approaches. With the ever increasing demand for more accurate and powerful models, the inference problem remains of broad scientific interest. The abstract representation of biological systems through gene regulatory networks represents a powerful method to study such systems, encoding different amounts and types of information. In this review, we summarize the different types of inference algorithms specifically based on time-series transcriptomics, giving an overview of the main applications of gene regulatory networks in computational biology. This review is intended to give an updated reference of regulatory networks inference tools to biologists and researchers new to the topic and guide them in selecting the appropriate inference method that best fits their questions, aims, and experimental data.


Subject(s)
Gene Regulatory Networks , Transcriptome , Gene Regulatory Networks/genetics , Transcriptome/genetics , Gene Expression Profiling , Algorithms , Computational Biology/methods
4.
Nucleic Acids Res ; 50(21): 12149-12165, 2022 11 28.
Article in English | MEDLINE | ID: mdl-36453993

ABSTRACT

In mammalian cells, chromosomal replication starts at thousands of origins at which replisomes are assembled. Replicative stress triggers additional initiation events from 'dormant' origins whose genomic distribution and regulation are not well understood. In this study, we have analyzed origin activity in mouse embryonic stem cells in the absence or presence of mild replicative stress induced by aphidicolin, a DNA polymerase inhibitor, or by deregulation of origin licensing factor CDC6. In both cases, we observe that the majority of stress-responsive origins are also active in a small fraction of the cell population in a normal S phase, and stress increases their frequency of activation. In a search for the molecular determinants of origin efficiency, we compared the genetic and epigenetic features of origins displaying different levels of activation, and integrated their genomic positions in three-dimensional chromatin interaction networks derived from high-depth Hi-C and promoter-capture Hi-C data. We report that origin efficiency is directly proportional to the proximity to transcriptional start sites and to the number of contacts established between origin-containing chromatin fragments, supporting the organization of origins in higher-level DNA replication factories.


Subject(s)
Chromatin , Replication Origin , Animals , Mice , Replication Origin/genetics , Chromatin/genetics , Mouse Embryonic Stem Cells/metabolism , DNA Replication/genetics , Cell Cycle Proteins/metabolism , Mammals/genetics
5.
Mol Ther ; 30(4): 1553-1563, 2022 04 06.
Article in English | MEDLINE | ID: mdl-35038581

ABSTRACT

Toll-like receptors (TLRs) are key players in the innate immune system. Recent studies have suggested that they may affect the growth of pancreatic cancer, a disease with no cure. Among them, TLR7 shows promise for therapy but may also promotes tumor growth. Thus, we aimed to clarify the therapeutic potential of TLR7 ligands in experimental pancreatic cancer models, to open the door for clinical applications. In vitro, we found that TLR7 ligands strongly inhibit the proliferation of both human and murine pancreatic cancer cells, compared with TLR2 agonists. Hence, TLR7 treatment alters cancer cells' cell cycle and induces cell death by apoptosis. In vivo, TLR7 agonist therapy significantly delays the growth of murine pancreatic tumors engrafted in immunodeficient mice. Remarkably, TLR7 ligands administration instead increases tumor growth and accelerates animal death when tumors are engrafted in immunocompetent models. Further investigations revealed that TLR7 agonists modulate the intratumoral content and phenotype of macrophages and that depleting such tumor-associated macrophages strongly hampers TLR7 agonist-induced tumor growth. Collectively, our findings shine a light on the duality of action of TLR7 agonists in experimental cancer models and call into question their use for pancreatic cancer therapy.


Subject(s)
Pancreatic Neoplasms , Toll-Like Receptor 7 , Animals , Humans , Ligands , Macrophages/metabolism , Membrane Glycoproteins , Mice , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/metabolism , Tumor Microenvironment , Pancreatic Neoplasms
6.
Nucleic Acids Res ; 49(19): 11005-11021, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34648034

ABSTRACT

Cohesin exists in two variants containing STAG1 or STAG2. STAG2 is one of the most mutated genes in cancer and a major bladder tumor suppressor. Little is known about how its inactivation contributes to tumorigenesis. Here, we analyze the genomic distribution of STAG1 and STAG2 and perform STAG2 loss-of-function experiments using RT112 bladder cancer cells; we then analyze the genomic effects by integrating gene expression and chromatin interaction data. Functional compartmentalization exists between the cohesin complexes: cohesin-STAG2 displays a distinctive genomic distribution and mediates short and mid-ranged interactions that engage genes at higher frequency than those established by cohesin-STAG1. STAG2 knockdown results in down-regulation of the luminal urothelial signature and up-regulation of the basal transcriptional program, mirroring differences between STAG2-high and STAG2-low human bladder tumors. This is accompanied by rewiring of DNA contacts within topological domains, while compartments and domain boundaries remain refractive. Contacts lost upon depletion of STAG2 are assortative, preferentially occur within silent chromatin domains, and are associated with de-repression of lineage-specifying genes. Our findings indicate that STAG2 participates in the DNA looping that keeps the basal transcriptional program silent and thus sustains the luminal program. This mechanism may contribute to the tumor suppressor function of STAG2 in the urothelium.


Subject(s)
Cell Cycle Proteins/genetics , Chromatin/chemistry , Loss of Function Mutation , Nuclear Proteins/genetics , Transcription, Genetic , Urinary Bladder Neoplasms/genetics , Base Sequence , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Ontology , HEK293 Cells , Histones/genetics , Histones/metabolism , Humans , Molecular Sequence Annotation , Nuclear Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology
7.
Bioinformatics ; 37(21): 3989-3991, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34213523

ABSTRACT

SUMMARY: Networks provide a powerful framework to analyze spatial omics experiments. However, we lack tools that integrate several methods to easily reconstruct networks for further analyses with dedicated libraries. In addition, choosing the appropriate method and parameters can be challenging. We propose tysserand, a Python library to reconstruct spatial networks from spatially resolved omics experiments. It is intended as a common tool to which the bioinformatics community can add new methods to reconstruct networks, choose appropriate parameters, clean resulting networks and pipe data to other libraries. AVAILABILITY AND IMPLEMENTATION: tysserand software and tutorials with a Jupyter notebook to reproduce the results are available at https://github.com/VeraPancaldiLab/tysserand. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Biological Products , Libraries , Software , Gene Library
8.
Nucleic Acids Res ; 48(8): 4066-4080, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32182345

ABSTRACT

We introduce an R package and a web-based visualization tool for the representation, analysis and integration of epigenomic data in the context of 3D chromatin interaction networks. GARDEN-NET allows for the projection of user-submitted genomic features on pre-loaded chromatin interaction networks, exploiting the functionalities of the ChAseR package to explore the features in combination with chromatin network topology properties. We demonstrate the approach using published epigenomic and chromatin structure datasets in haematopoietic cells, including a collection of gene expression, DNA methylation and histone modifications data in primary healthy myeloid cells from hundreds of individuals. These datasets allow us to test the robustness of chromatin assortativity, which highlights which epigenomic features, alone or in combination, are more strongly associated with 3D genome architecture. We find evidence for genomic regions with specific histone modifications, DNA methylation, and gene expression levels to be forming preferential contacts in 3D nuclear space, to a different extent depending on the cell type and lineage. Finally, we examine replication timing data and find it to be the genomic feature most strongly associated with overall 3D chromatin organization at multiple scales, consistent with previous results from the literature.


Subject(s)
Chromatin/metabolism , Epigenesis, Genetic , Hematopoietic Stem Cells/metabolism , Software , B-Lymphocytes/metabolism , DNA Methylation , DNA Replication Timing , Gene Expression , Histone Code , Humans , Neutrophils/metabolism , Promoter Regions, Genetic
9.
Int J Mol Sci ; 22(9)2021 May 07.
Article in English | MEDLINE | ID: mdl-34066960

ABSTRACT

DNA replication timing (RT), reflecting the temporal order of origin activation, is known as a robust and conserved cell-type specific process. Upon low replication stress, the slowing of replication forks induces well-documented RT delays associated to genetic instability, but it can also generate RT advances that are still uncharacterized. In order to characterize these advanced initiation events, we monitored the whole genome RT from six independent human cell lines treated with low doses of aphidicolin. We report that RT advances are cell-type-specific and involve large heterochromatin domains. Importantly, we found that some major late to early RT advances can be inherited by the unstressed next-cellular generation, which is a unique process that correlates with enhanced chromatin accessibility, as well as modified replication origin landscape and gene expression in daughter cells. Collectively, this work highlights how low replication stress may impact cellular identity by RT advances events at a subset of chromosomal domains.


Subject(s)
DNA Replication Timing , Stress, Physiological , Aphidicolin/pharmacology , Cell Line, Tumor , Chromatin/metabolism , DNA Damage , DNA Replication Timing/genetics , Epigenesis, Genetic/drug effects , Genetic Loci , Histone Code , Humans , Models, Biological , Stress, Physiological/genetics
10.
Lancet Oncol ; 21(7): 914-922, 2020 07.
Article in English | MEDLINE | ID: mdl-32539942

ABSTRACT

BACKGROUND: Early reports on patients with cancer and COVID-19 have suggested a high mortality rate compared with the general population. Patients with thoracic malignancies are thought to be particularly susceptible to COVID-19 given their older age, smoking habits, and pre-existing cardiopulmonary comorbidities, in addition to cancer treatments. We aimed to study the effect of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection on patients with thoracic malignancies. METHODS: The Thoracic Cancers International COVID-19 Collaboration (TERAVOLT) registry is a multicentre observational study composed of a cross-sectional component and a longitudinal cohort component. Eligibility criteria were the presence of any thoracic cancer (non-small-cell lung cancer [NSCLC], small-cell lung cancer, mesothelioma, thymic epithelial tumours, and other pulmonary neuroendocrine neoplasms) and a COVID-19 diagnosis, either laboratory confirmed with RT-PCR, suspected with symptoms and contacts, or radiologically suspected cases with lung imaging features consistent with COVID-19 pneumonia and symptoms. Patients of any age, sex, histology, or stage were considered eligible, including those in active treatment and clinical follow-up. Clinical data were extracted from medical records of consecutive patients from Jan 1, 2020, and will be collected until the end of pandemic declared by WHO. Data on demographics, oncological history and comorbidities, COVID-19 diagnosis, and course of illness and clinical outcomes were collected. Associations between demographic or clinical characteristics and outcomes were measured with odds ratios (ORs) with 95% CIs using univariable and multivariable logistic regression, with sex, age, smoking status, hypertension, and chronic obstructive pulmonary disease included in multivariable analysis. This is a preliminary analysis of the first 200 patients. The registry continues to accept new sites and patient data. FINDINGS: Between March 26 and April 12, 2020, 200 patients with COVID-19 and thoracic cancers from eight countries were identified and included in the TERAVOLT registry; median age was 68·0 years (61·8-75·0) and the majority had an Eastern Cooperative Oncology Group performance status of 0-1 (142 [72%] of 196 patients), were current or former smokers (159 [81%] of 196), had non-small-cell lung cancer (151 [76%] of 200), and were on therapy at the time of COVID-19 diagnosis (147 [74%] of 199), with 112 (57%) of 197 on first-line treatment. 152 (76%) patients were hospitalised and 66 (33%) died. 13 (10%) of 134 patients who met criteria for ICU admission were admitted to ICU; the remaining 121 were hospitalised, but were not admitted to ICU. Univariable analyses revealed that being older than 65 years (OR 1·88, 95% 1·00-3·62), being a current or former smoker (4·24, 1·70-12·95), receiving treatment with chemotherapy alone (2·54, 1·09-6·11), and the presence of any comorbidities (2·65, 1·09-7·46) were associated with increased risk of death. However, in multivariable analysis, only smoking history (OR 3·18, 95% CI 1·11-9·06) was associated with increased risk of death. INTERPRETATION: With an ongoing global pandemic of COVID-19, our data suggest high mortality and low admission to intensive care in patients with thoracic cancer. Whether mortality could be reduced with treatment in intensive care remains to be determined. With improved cancer therapeutic options, access to intensive care should be discussed in a multidisciplinary setting based on cancer specific mortality and patients' preference. FUNDING: None.


Subject(s)
Coronavirus Infections/epidemiology , Pneumonia, Viral/epidemiology , Registries/statistics & numerical data , Thoracic Neoplasms/epidemiology , Aged , Betacoronavirus , COVID-19 , Cause of Death , Coronavirus Infections/mortality , Coronavirus Infections/pathology , Cross-Sectional Studies , Female , Hospitalization/statistics & numerical data , Humans , Longitudinal Studies , Male , Middle Aged , Pandemics , Pneumonia, Viral/mortality , Pneumonia, Viral/pathology , Risk Factors , SARS-CoV-2 , Thoracic Neoplasms/mortality , Thoracic Neoplasms/pathology , Thoracic Neoplasms/therapy
11.
Bioessays ; 40(2)2018 02.
Article in English | MEDLINE | ID: mdl-29251357

ABSTRACT

Epigenetic and transcriptional variability contribute to the vast diversity of cellular and organismal phenotypes and are key in human health and disease. In this review, we describe different types, sources, and determinants of epigenetic and transcriptional variability, enabling cells and organisms to adapt and evolve to a changing environment. We highlight the latest research and hypotheses on how chromatin structure and the epigenome influence gene expression variability. Further, we provide an overview of challenges in the analysis of biological variability. An improved understanding of the molecular mechanisms underlying epigenetic and transcriptional variability, at both the intra- and inter-individual level, provides great opportunity for disease prevention, better therapeutic approaches, and personalized medicine.


Subject(s)
Adaptation, Physiological/genetics , Biological Variation, Population/genetics , Epigenesis, Genetic , Genetic Variation , Transcription, Genetic , Biological Variation, Individual , Chromatin/genetics , Humans , Precision Medicine
12.
Nucleic Acids Res ; 45(16): 9244-9259, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28934481

ABSTRACT

Hematopoiesis is one of the best characterized biological systems but the connection between chromatin changes and lineage differentiation is not yet well understood. We have developed a bioinformatic workflow to generate a chromatin space that allows to classify 42 human healthy blood epigenomes from the BLUEPRINT, NIH ROADMAP and ENCODE consortia by their cell type. This approach let us to distinguish different cells types based on their epigenomic profiles, thus recapitulating important aspects of human hematopoiesis. The analysis of the orthogonal dimension of the chromatin space identify 32,662 chromatin determinant regions (CDRs), genomic regions with different epigenetic characteristics between the cell types. Functional analysis revealed that these regions are linked with cell identities. The inclusion of leukemia epigenomes in the healthy hematological chromatin sample space gives us insights on the healthy cell types that are more epigenetically similar to the disease samples. Further analysis of tumoral epigenetic alterations in hematopoietic CDRs points to sets of genes that are tightly regulated in leukemic transformations and commonly mutated in other tumors. Our method provides an analytical approach to study the relationship between epigenomic changes and cell lineage differentiation. Method availability: https://github.com/david-juan/ChromDet.


Subject(s)
Chromatin/metabolism , Epigenesis, Genetic , Epigenomics/methods , Hematopoiesis/genetics , Binding Sites , Hematopoietic Stem Cells/metabolism , Histone Code , Humans , Leukemia/genetics , Transcription Factors/metabolism
13.
Int J Mol Sci ; 20(13)2019 Jun 26.
Article in English | MEDLINE | ID: mdl-31247897

ABSTRACT

Matrix factorization (MF) is an established paradigm for large-scale biological data analysis with tremendous potential in computational biology. Here, we challenge MF in depicting the molecular bases of epidemiologically described disease-disease (DD) relationships. As a use case, we focus on the inverse comorbidity association between Alzheimer's disease (AD) and lung cancer (LC), described as a lower than expected probability of developing LC in AD patients. To this day, the molecular mechanisms underlying DD relationships remain poorly explained and their better characterization might offer unprecedented clinical opportunities. To this goal, we extend our previously designed MF-based framework for the molecular characterization of DD relationships. Considering AD-LC inverse comorbidity as a case study, we highlight multiple molecular mechanisms, among which we confirm the involvement of processes related to the immune system and mitochondrial metabolism. We then distinguish mechanisms specific to LC from those shared with other cancers through a pan-cancer analysis. Additionally, new candidate molecular players, such as estrogen receptor (ER), cadherin 1 (CDH1) and histone deacetylase (HDAC), are pinpointed as factors that might underlie the inverse relationship, opening the way to new investigations. Finally, some lung cancer subtype-specific factors are also detected, also suggesting the existence of heterogeneity across patients in the context of inverse comorbidity.


Subject(s)
Alzheimer Disease/epidemiology , Computational Biology , Lung Neoplasms/epidemiology , Models, Biological , Algorithms , Alzheimer Disease/complications , Alzheimer Disease/etiology , Comorbidity , Computational Biology/methods , Humans , Lung Neoplasms/complications , Lung Neoplasms/etiology
14.
BMC Evol Biol ; 15: 24, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25888139

ABSTRACT

BACKGROUND: Modelling genetic phenomena affecting biological traits is important for the development of agriculture as it allows breeders to predict the potential of breeding for certain traits. One such phenomenon is heterosis or hybrid vigor: crossing individuals from genetically distinct populations often results in improvements in quantitative traits, such as growth rate, biomass production and stress resistance. Heterosis has become a very useful tool in global agriculture, but its genetic basis remains controversial and its effects hard to predict. We have taken a computational approach to studying heterosis, developing a simulation of evolution, independent reassortment of alleles and hybridization of Gene Regulatory Networks (GRNs) in a Boolean framework. These artificial regulatory networks exhibit topological properties that reflect those observed in biology, and fitness is measured as the ability of a network to respond to external inputs in a pre-defined way. RESULTS: Our model reproduced common experimental observations on heterosis using only biologically justified parameters, such as mutation rates. Hybrid vigor was observed and its extent was seen to increase as parental populations diverged, up until a point of sudden collapse of hybrid fitness. Thus, the model also describes a process akin to speciation due to genetic incompatibility of the separated populations. We also reproduce, for the first time in a model, the fact that hybrid vigor cannot easily be fixed by within a breeding line, currently an important limitation of the use of hybrid crops. The simulation allowed us to study the effects of three standard models for the genetic basis of heterosis: dominance, over-dominance, and epistasis. CONCLUSION: This study describes the most detailed simulation of heterosis using gene regulatory networks to date and reproduces several phenomena associated with heterosis for the first time in a model. The level of detail in our model allows us to suggest possible warning signs of the impending collapse of hybrid vigor in breeding. In addition, the simulation provides a framework that can be extended to study other aspects of heterosis and alternative evolutionary scenarios.


Subject(s)
Crops, Agricultural/genetics , Gene Regulatory Networks , Hybrid Vigor , Models, Genetic , Breeding , Computer Simulation , Crops, Agricultural/classification , Epistasis, Genetic , Genetic Fitness , Genetic Speciation , Heterozygote , Hybridization, Genetic
15.
Cancer Res ; 84(7): 1013-1028, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38294491

ABSTRACT

Cytidine deaminase (CDA) functions in the pyrimidine salvage pathway for DNA and RNA syntheses and has been shown to protect cancer cells from deoxycytidine-based chemotherapies. In this study, we observed that CDA was overexpressed in pancreatic adenocarcinoma from patients at baseline and was essential for experimental tumor growth. Mechanistic investigations revealed that CDA localized to replication forks where it increased replication speed, improved replication fork restart efficiency, reduced endogenous replication stress, minimized DNA breaks, and regulated genetic stability during DNA replication. In cellular pancreatic cancer models, high CDA expression correlated with resistance to DNA-damaging agents. Silencing CDA in patient-derived primary cultures in vitro and in orthotopic xenografts in vivo increased replication stress and sensitized pancreatic adenocarcinoma cells to oxaliplatin. This study sheds light on the role of CDA in pancreatic adenocarcinoma, offering insights into how this tumor type modulates replication stress. These findings suggest that CDA expression could potentially predict therapeutic efficacy and that targeting CDA induces intolerable levels of replication stress in cancer cells, particularly when combined with DNA-targeted therapies. SIGNIFICANCE: Cytidine deaminase reduces replication stress and regulates DNA replication to confer resistance to DNA-damaging drugs in pancreatic cancer, unveiling a molecular vulnerability that could enhance treatment response.


Subject(s)
Adenocarcinoma , Cytidine Deaminase , Nucleic Acid Synthesis Inhibitors , Pancreatic Neoplasms , Humans , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Cytidine Deaminase/metabolism , DNA , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , DNA Replication , Nucleic Acid Synthesis Inhibitors/therapeutic use
16.
Bioinformatics ; 28(16): 2137-45, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22718785

ABSTRACT

MOTIVATION: It has been recognized that the topology of molecular networks provides information about the certainty and nature of individual interactions. Thus, network motifs have been used for predicting missing links in biological networks and for removing false positives. However, various different measures can be inferred from the structure of a given network and their predictive power varies depending on the task at hand. RESULTS: Herein, we present a systematic assessment of seven different network features extracted from the topology of functional genetic networks and we quantify their ability to classify interactions into different types of physical protein associations. Using machine learning, we combine features based on network topology with non-network features and compare their importance of the classification of interactions. We demonstrate the utility of network features based on human and budding yeast networks; we show that network features can distinguish different sub-types of physical protein associations and we apply the framework to fission yeast, which has a much sparser known physical interactome than the other two species. Our analysis shows that network features are at least as predictive for the tasks we tested as non-network features. However, feature importance varies between species owing to different topological characteristics of the networks. The application to fission yeast shows that small maps of physical interactomes can be extended based on functional networks, which are often more readily available. AVAILABILITY AND IMPLEMENTATION: The R-code for computing the network features is available from www.cellularnetworks.org


Subject(s)
Artificial Intelligence , Computational Biology/methods , Protein Interaction Mapping/methods , Proteins/chemistry , Area Under Curve , Humans , Protein Binding , ROC Curve , Saccharomyces cerevisiae , Schizosaccharomyces , Software
17.
Nucleic Acids Res ; 39(14): 5826-36, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21459850

ABSTRACT

Post-transcriptional gene regulation is mediated through complex networks of protein-RNA interactions. The targets of only a few RNA binding proteins (RBPs) are known, even in the well-characterized budding yeast. In silico prediction of protein-RNA interactions is therefore useful to guide experiments and to provide insight into regulatory networks. Computational approaches have identified RBP targets based on sequence binding preferences. We investigate here to what extent RBP-RNA interactions can be predicted based on RBP and mRNA features other than sequence motifs. We analyze global relationships between gene and protein properties in general and between selected RBPs and known mRNA targets in particular. Highly translated RBPs tend to bind to shorter transcripts, and transcripts bound by the same RBP show high expression correlation across different biological conditions. Surprisingly, a given RBP preferentially binds to mRNAs that encode interaction partners for this RBP, suggesting coordinated post-transcriptional auto-regulation of protein complexes. We apply a machine-learning approach to predict specific RBP targets in yeast. Although this approach performs well for RBPs with known targets, predictions for uncharacterized RBPs remain challenging due to limiting experimental data. We also predict targets of fission yeast RBPs, indicating that the suggested framework could be applied to other species once more experimental data are available.


Subject(s)
Artificial Intelligence , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Genes, Fungal , Humans , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics
18.
Curr Opin Genet Dev ; 80: 102051, 2023 06.
Article in English | MEDLINE | ID: mdl-37245241

ABSTRACT

Increasing numbers of datasets and experimental assays that capture the organization of chromatin inside the nucleus warrant an effort to develop tools to visualize and analyze these structures. Alongside polymer physics or constraint-based modeling, network theory approaches to describe 3D epigenome organization have gained in popularity. Representing genomic regions as nodes in a network enables visualization of 1D epigenomics datasets in the context of chromatin structure maps, while network theory metrics can be used to describe 3D epigenome organization and dynamics. In this review, we summarize the most salient applications of network theory to the study of chromatin contact maps, demonstrating its potential in revealing epigenomic patterns and relating them to cellular phenotypes.


Subject(s)
Chromatin , Chromosomes , Chromatin/genetics , Cell Nucleus , Genome , Epigenome , Epigenomics
19.
bioRxiv ; 2023 Mar 20.
Article in English | MEDLINE | ID: mdl-36993595

ABSTRACT

Single-cell spatially resolved proteomic or transcriptomic methods offer the opportunity to discover cell types interactions of biological or clinical importance. To extract relevant information from these data, we present mosna, a Python package to analyze spatially resolved experiments and discover patterns of cellular spatial organization. It includes the detection of preferential interactions between specific cell types and the discovery of cellular niches. We exemplify the proposed analysis pipeline on spatially resolved proteomic data from cancer patient samples annotated with clinical response to immunotherapy, and we show that mosna can identify a number of features describing cellular composition and spatial distribution that can provide biological hypotheses regarding factors that affect response to therapies.

20.
Nat Commun ; 14(1): 6678, 2023 10 21.
Article in English | MEDLINE | ID: mdl-37865700

ABSTRACT

In mammals, insulators contribute to the regulation of loop extrusion to organize chromatin into topologically associating domains. In Drosophila the role of insulators in 3D genome organization is, however, under current debate. Here, we addressed this question by combining bioinformatics analysis and multiplexed chromatin imaging. We describe a class of Drosophila insulators enriched at regions forming preferential chromatin interactions genome-wide. Notably, most of these 3D interactions do not involve TAD borders. Multiplexed imaging shows that these interactions occur infrequently, and only rarely involve multiple genomic regions coalescing together in space in single cells. Finally, we show that non-border preferential 3D interactions enriched in this class of insulators are present before TADs and transcription during Drosophila development. Our results are inconsistent with insulators forming stable hubs in single cells, and instead suggest that they fine-tune existing 3D chromatin interactions, providing an additional regulatory layer for transcriptional regulation.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Drosophila/genetics , Drosophila/metabolism , Chromatin/genetics , Gene Expression Regulation , Genome , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Mammals/genetics
SELECTION OF CITATIONS
SEARCH DETAIL