Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Cells ; 11(19)2022 09 21.
Article in English | MEDLINE | ID: mdl-36230912

ABSTRACT

The coronavirus disease 2019 (COVID-19) is accompanied by a cytokine storm with the release of many proinflammatory factors and development of respiratory syndrome. Several SARS-CoV-2 lineages have been identified, and the Delta variant (B.1.617), linked with high mortality risk, has become dominant in many countries. Understanding the immune responses associated with COVID-19 lineages may therefore aid the development of therapeutic and diagnostic strategies. Multiple single-cell gene expression studies revealed innate and adaptive immunological factors and pathways correlated with COVID-19 severity. Additional investigations covering host-pathogen response characteristics for infection caused by different lineages are required. Here, we performed single-cell transcriptome profiling of blood mononuclear cells from the individuals with different severity of the COVID-19 and virus lineages to uncover variant specific molecular factors associated with immunity. We identified significant changes in lymphoid and myeloid cells. Our study highlights that an abundant population of monocytes with specific gene expression signatures accompanies Delta lineage of SARS-CoV-2 and contributes to COVID-19 pathogenesis inferring immune components for targeted therapy.


Subject(s)
COVID-19 , COVID-19/genetics , Gene Expression , Humans , Immunologic Factors , SARS-CoV-2
2.
Cell ; 182(4): 872-885.e19, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32783915

ABSTRACT

Cell function and activity are regulated through integration of signaling, epigenetic, transcriptional, and metabolic pathways. Here, we introduce INs-seq, an integrated technology for massively parallel recording of single-cell RNA sequencing (scRNA-seq) and intracellular protein activity. We demonstrate the broad utility of INs-seq for discovering new immune subsets by profiling different intracellular signatures of immune signaling, transcription factor combinations, and metabolic activity. Comprehensive mapping of Arginase 1-expressing cells within tumor models, a metabolic immune signature of suppressive activity, discovers novel Arg1+ Trem2+ regulatory myeloid (Mreg) cells and identifies markers, metabolic activity, and pathways associated with these cells. Genetic ablation of Trem2 in mice inhibits accumulation of intra-tumoral Mreg cells, leading to a marked decrease in dysfunctional CD8+ T cells and reduced tumor growth. This study establishes INs-seq as a broadly applicable technology for elucidating integrated transcriptional and intra-cellular maps and identifies the molecular signature of myeloid suppressive cells in tumors.


Subject(s)
Membrane Glycoproteins/metabolism , Neoplasms/pathology , RNA, Small Cytoplasmic/chemistry , Receptors, Immunologic/metabolism , Animals , Arginase/genetics , Arginase/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Female , Gene Expression Regulation , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/metabolism , RNA, Small Cytoplasmic/metabolism , Receptors, Immunologic/genetics , Sequence Analysis, RNA , Single-Cell Analysis , Transcription Factors/metabolism , Tumor Microenvironment , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases
3.
Sci Rep ; 8(1): 16169, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30385846

ABSTRACT

Cardiovascular disease associated with metabolic syndrome has a high prevalence, but the mechanistic basis of metabolic cardiomyopathy remains poorly understood. We characterised the cardiac transcriptome in a murine metabolic syndrome (MetS) model (LDLR-/-; ob/ob, DKO) relative to the healthy, control heart (C57BL/6, WT) and the transcriptional changes induced by ACE-inhibition in those hearts. RNA-Seq, differential gene expression and transcription factor analysis identified 288 genes differentially expressed between DKO and WT hearts implicating 72 pathways. Hallmarks of metabolic cardiomyopathy were increased activity in integrin-linked kinase signalling, Rho signalling, dendritic cell maturation, production of nitric oxide and reactive oxygen species in macrophages, atherosclerosis, LXR-RXR signalling, cardiac hypertrophy, and acute phase response pathways. ACE-inhibition had a limited effect on gene expression in WT (55 genes, 23 pathways), and a prominent effect in DKO hearts (1143 genes, 104 pathways). In DKO hearts, ACE-I appears to counteract some of the MetS-specific pathways, while also activating cardioprotective mechanisms. We conclude that MetS and control murine hearts have unique transcriptional profiles and exhibit a partially specific transcriptional response to ACE-inhibition.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Atherosclerosis/genetics , Cardiovascular Diseases/genetics , Metabolic Syndrome/drug therapy , Receptors, LDL/genetics , Aged , Animals , Atherosclerosis/drug therapy , Atherosclerosis/etiology , Atherosclerosis/physiopathology , Cardiotonic Agents/administration & dosage , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/etiology , Cardiovascular Diseases/physiopathology , Disease Models, Animal , Heart/drug effects , Heart/physiopathology , Humans , Metabolic Networks and Pathways/genetics , Metabolic Syndrome/complications , Metabolic Syndrome/genetics , Metabolic Syndrome/physiopathology , Mice , Mice, Knockout , Obesity/drug therapy , Obesity/genetics , Obesity/physiopathology , Peptidyl-Dipeptidase A/genetics , Transcriptome/drug effects , Transcriptome/genetics
4.
Genome Med ; 9(1): 80, 2017 08 30.
Article in English | MEDLINE | ID: mdl-28854983

ABSTRACT

The identification of functional non-coding mutations is a key challenge in the field of genomics. Here we introduce µ-cisTarget to filter, annotate and prioritize cis-regulatory mutations based on their putative effect on the underlying "personal" gene regulatory network. We validated µ-cisTarget by re-analyzing the TAL1 and LMO1 enhancer mutations in T-ALL, and the TERT promoter mutation in melanoma. Next, we re-sequenced the full genomes of ten cancer cell lines and used matched transcriptome data and motif discovery to identify master regulators with de novo binding sites that result in the up-regulation of nearby oncogenic drivers. µ-cisTarget is available from http://mucistarget.aertslab.org .


Subject(s)
DNA Mutational Analysis/methods , Gene Regulatory Networks , Genes, Neoplasm , Mutation , Neoplasms/genetics , Regulatory Sequences, Nucleic Acid , Algorithms , Binding Sites , Cell Line, Tumor , Female , Gene Expression Profiling , Genomics/methods , Humans , Male , Neoplasms/metabolism , Precision Medicine/methods , Transcription Factors/metabolism
5.
Genome Res ; 26(7): 882-95, 2016 07.
Article in English | MEDLINE | ID: mdl-27197205

ABSTRACT

Transcription factors regulate their target genes by binding to regulatory regions in the genome. Although the binding preferences of TP53 are known, it remains unclear what distinguishes functional enhancers from nonfunctional binding. In addition, the genome is scattered with recognition sequences that remain unoccupied. Using two complementary techniques of multiplex enhancer-reporter assays, we discovered that functional enhancers could be discriminated from nonfunctional binding events by the occurrence of a single TP53 canonical motif. By combining machine learning with a meta-analysis of TP53 ChIP-seq data sets, we identified a core set of more than 1000 responsive enhancers in the human genome. This TP53 cistrome is invariably used between cell types and experimental conditions, whereas differences among experiments can be attributed to indirect nonfunctional binding events. Our data suggest that TP53 enhancers represent a class of unsophisticated cell-autonomous enhancers containing a single TP53 binding site, distinct from complex developmental enhancers that integrate signals from multiple transcription factors.


Subject(s)
Enhancer Elements, Genetic , Transcriptional Activation , Tumor Suppressor Protein p53/physiology , Binding Sites , Biological Assay , Genes, Reporter , Humans , MCF-7 Cells , Protein Binding
6.
PLoS Comput Biol ; 11(11): e1004590, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26562774

ABSTRACT

Cancer genomes contain vast amounts of somatic mutations, many of which are passenger mutations not involved in oncogenesis. Whereas driver mutations in protein-coding genes can be distinguished from passenger mutations based on their recurrence, non-coding mutations are usually not recurrent at the same position. Therefore, it is still unclear how to identify cis-regulatory driver mutations, particularly when chromatin data from the same patient is not available, thus relying only on sequence and expression information. Here we use machine-learning methods to predict functional regulatory regions using sequence information alone, and compare the predicted activity of the mutated region with the reference sequence. This way we define the Predicted Regulatory Impact of a Mutation in an Enhancer (PRIME). We find that the recently identified driver mutation in the TAL1 enhancer has a high PRIME score, representing a "gain-of-target" for MYB, whereas the highly recurrent TERT promoter mutation has a surprisingly low PRIME score. We trained Random Forest models for 45 cancer-related transcription factors, and used these to score variations in the HeLa genome and somatic mutations across more than five hundred cancer genomes. Each model predicts only a small fraction of non-coding mutations with a potential impact on the function of the encompassing regulatory region. Nevertheless, as these few candidate driver mutations are often linked to gains in chromatin activity and gene expression, they may contribute to the oncogenic program by altering the expression levels of specific oncogenes and tumor suppressor genes.


Subject(s)
Models, Statistical , Mutation/genetics , Neoplasms/genetics , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors/genetics , Algorithms , Binding Sites/genetics , Computational Biology/methods , Genome , HeLa Cells , Humans , Machine Learning
7.
Nat Commun ; 6: 6683, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25865119

ABSTRACT

Transcriptional reprogramming of proliferative melanoma cells into a phenotypically distinct invasive cell subpopulation is a critical event at the origin of metastatic spreading. Here we generate transcriptome, open chromatin and histone modification maps of melanoma cultures; and integrate this data with existing transcriptome and DNA methylation profiles from tumour biopsies to gain insight into the mechanisms underlying this key reprogramming event. This shows thousands of genomic regulatory regions underlying the proliferative and invasive states, identifying SOX10/MITF and AP-1/TEAD as regulators, respectively. Knockdown of TEADs shows a previously unrecognized role in the invasive gene network and establishes a causative link between these transcription factors, cell invasion and sensitivity to MAPK inhibitors. Using regulatory landscapes and in silico analysis, we show that transcriptional reprogramming underlies the distinct cellular states present in melanoma. Furthermore, it reveals an essential role for the TEADs, linking it to clinically relevant mechanisms such as invasion and resistance.


Subject(s)
Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Melanoma/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Transcriptome , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cellular Reprogramming/genetics , Chromatin/chemistry , Chromatin/metabolism , DNA Methylation , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Histones/genetics , Histones/metabolism , Humans , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Neoplasm Invasiveness , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Signal Transduction , TEA Domain Transcription Factors , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Transcription, Genetic
8.
PLoS Comput Biol ; 10(7): e1003731, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25058159

ABSTRACT

Identifying master regulators of biological processes and mapping their downstream gene networks are key challenges in systems biology. We developed a computational method, called iRegulon, to reverse-engineer the transcriptional regulatory network underlying a co-expressed gene set using cis-regulatory sequence analysis. iRegulon implements a genome-wide ranking-and-recovery approach to detect enriched transcription factor motifs and their optimal sets of direct targets. We increase the accuracy of network inference by using very large motif collections of up to ten thousand position weight matrices collected from various species, and linking these to candidate human TFs via a motif2TF procedure. We validate iRegulon on gene sets derived from ENCODE ChIP-seq data with increasing levels of noise, and we compare iRegulon with existing motif discovery methods. Next, we use iRegulon on more challenging types of gene lists, including microRNA target sets, protein-protein interaction networks, and genetic perturbation data. In particular, we over-activate p53 in breast cancer cells, followed by RNA-seq and ChIP-seq, and could identify an extensive up-regulated network controlled directly by p53. Similarly we map a repressive network with no indication of direct p53 regulation but rather an indirect effect via E2F and NFY. Finally, we generalize our computational framework to include regulatory tracks such as ChIP-seq data and show how motif and track discovery can be combined to map functional regulatory interactions among co-expressed genes. iRegulon is available as a Cytoscape plugin from http://iregulon.aertslab.org.


Subject(s)
Computational Biology/methods , Gene Expression Regulation/genetics , Gene Regulatory Networks/genetics , Transcription Factors/genetics , Breast Neoplasms , Cell Line, Tumor , Chromatin Immunoprecipitation , Databases, Genetic , Genes, p53 , Humans , Models, Genetic , Sequence Analysis, RNA
SELECTION OF CITATIONS
SEARCH DETAIL
...