Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Cell ; 184(10): 2618-2632.e17, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33836156

ABSTRACT

The ongoing pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is currently affecting millions of lives worldwide. Large retrospective studies indicate that an elevated level of inflammatory cytokines and pro-inflammatory factors are associated with both increased disease severity and mortality. Here, using multidimensional epigenetic, transcriptional, in vitro, and in vivo analyses, we report that topoisomerase 1 (TOP1) inhibition suppresses lethal inflammation induced by SARS-CoV-2. Therapeutic treatment with two doses of topotecan (TPT), an FDA-approved TOP1 inhibitor, suppresses infection-induced inflammation in hamsters. TPT treatment as late as 4 days post-infection reduces morbidity and rescues mortality in a transgenic mouse model. These results support the potential of TOP1 inhibition as an effective host-directed therapy against severe SARS-CoV-2 infection. TPT and its derivatives are inexpensive clinical-grade inhibitors available in most countries. Clinical trials are needed to evaluate the efficacy of repurposing TOP1 inhibitors for severe coronavirus disease 2019 (COVID-19) in humans.


Subject(s)
COVID-19 Drug Treatment , DNA Topoisomerases, Type I/metabolism , SARS-CoV-2/metabolism , Topoisomerase I Inhibitors/pharmacology , Topotecan/pharmacology , Animals , COVID-19/enzymology , COVID-19/pathology , Chlorocebus aethiops , Humans , Inflammation/drug therapy , Inflammation/enzymology , Inflammation/pathology , Inflammation/virology , Mesocricetus , Mice , Mice, Transgenic , THP-1 Cells , Vero Cells
2.
Cell ; 167(5): 1369-1384.e19, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27863249

ABSTRACT

Long-range interactions between regulatory elements and gene promoters play key roles in transcriptional regulation. The vast majority of interactions are uncharted, constituting a major missing link in understanding genome control. Here, we use promoter capture Hi-C to identify interacting regions of 31,253 promoters in 17 human primary hematopoietic cell types. We show that promoter interactions are highly cell type specific and enriched for links between active promoters and epigenetically marked enhancers. Promoter interactomes reflect lineage relationships of the hematopoietic tree, consistent with dynamic remodeling of nuclear architecture during differentiation. Interacting regions are enriched in genetic variants linked with altered expression of genes they contact, highlighting their functional role. We exploit this rich resource to connect non-coding disease variants to putative target promoters, prioritizing thousands of disease-candidate genes and implicating disease pathways. Our results demonstrate the power of primary cell promoter interactomes to reveal insights into genomic regulatory mechanisms underlying common diseases.


Subject(s)
Blood Cells/cytology , Disease/genetics , Promoter Regions, Genetic , Cell Lineage , Cell Separation , Chromatin , Enhancer Elements, Genetic , Epigenomics , Genetic Predisposition to Disease , Genome-Wide Association Study , Hematopoiesis , Humans , Polymorphism, Single Nucleotide , Quantitative Trait Loci
3.
Cell ; 148(3): 473-86, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22304916

ABSTRACT

Cell fate decisions are driven through the integration of inductive signals and tissue-specific transcription factors (TFs), although the details on how this information converges in cis remain unclear. Here, we demonstrate that the five genetic components essential for cardiac specification in Drosophila, including the effectors of Wg and Dpp signaling, act as a collective unit to cooperatively regulate heart enhancer activity, both in vivo and in vitro. Their combinatorial binding does not require any specific motif orientation or spacing, suggesting an alternative mode of enhancer function whereby cooperative activity occurs with extensive motif flexibility. A fraction of enhancers co-occupied by cardiogenic TFs had unexpected activity in the neighboring visceral mesoderm but could be rendered active in heart through single-site mutations. Given that cardiac and visceral cells are both derived from the dorsal mesoderm, this "dormant" TF binding signature may represent a molecular footprint of these cells' developmental lineage.


Subject(s)
Drosophila melanogaster/cytology , Gene Regulatory Networks , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Mesoderm/cytology , Mesoderm/metabolism , Myocardium/cytology , Myocardium/metabolism , Transcription Factors/metabolism
4.
Mol Cell ; 66(3): 420-435.e5, 2017 May 04.
Article in English | MEDLINE | ID: mdl-28475875

ABSTRACT

Interactions between transcriptional promoters and their distal regulatory elements play an important role in transcriptional regulation; however, the extent to which these interactions are subject to rapid modulations in response to signals is unknown. Here, we use promoter capture Hi-C to demonstrate a rapid reorganization of promoter-anchored chromatin loops within 4 hr after inducing differentiation of 3T3-L1 preadipocytes. The establishment of new promoter-enhancer loops is tightly coupled to activation of poised (histone H3 lysine 4 mono- and dimethylated) enhancers, as evidenced by the acquisition of histone H3 lysine 27 acetylation and the binding of MED1, SMC1, and P300 proteins to these regions, as well as to activation of target genes. Intriguingly, formation of loops connecting activated enhancers and promoters is also associated with extensive recruitment of corepressors such as NCoR and HDACs, indicating that this class of coregulators may play a previously unrecognized role during enhancer activation.


Subject(s)
Adipocytes/metabolism , Adipogenesis , Chromatin Assembly and Disassembly , Chromatin/metabolism , Promoter Regions, Genetic , 3T3-L1 Cells , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin/chemistry , Chromatin/genetics , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/metabolism , Enhancer Elements, Genetic , Mediator Complex Subunit 1/genetics , Mediator Complex Subunit 1/metabolism , Mice , Nucleic Acid Conformation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Analysis, RNA , Time Factors , Transcription, Genetic , Transcriptional Activation
5.
Cell ; 132(3): 422-33, 2008 Feb 08.
Article in English | MEDLINE | ID: mdl-18237772

ABSTRACT

Cohesins mediate sister chromatid cohesion, which is essential for chromosome segregation and postreplicative DNA repair. In addition, cohesins appear to regulate gene expression and enhancer-promoter interactions. These noncanonical functions remained unexplained because knowledge of cohesin-binding sites and functional interactors in metazoans was lacking. We show that the distribution of cohesins on mammalian chromosome arms is not driven by transcriptional activity, in contrast to S. cerevisiae. Instead, mammalian cohesins occupy a subset of DNase I hypersensitive sites, many of which contain sequence motifs resembling the consensus for CTCF, a DNA-binding protein with enhancer blocking function and boundary-element activity. We find cohesins at most CTCF sites and show that CTCF is required for cohesin localization to these sites. Recruitment by CTCF suggests a rationale for noncanonical cohesin functions and, because CTCF binding is sensitive to DNA methylation, allows cohesin positioning to integrate DNA sequence and epigenetic state.


Subject(s)
Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Mammalian/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Animals , Base Sequence , CCCTC-Binding Factor , Cell Differentiation , Cell Line , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , Cytokines/genetics , Deoxyribonuclease I/metabolism , Gene Expression , Humans , Mice , Mice, Transgenic , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Cohesins
6.
Cell Mol Life Sci ; 78(19-20): 6453-6485, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34414474

ABSTRACT

Transcriptional enhancers play a key role in the initiation and maintenance of gene expression programmes, particularly in metazoa. How these elements control their target genes in the right place and time is one of the most pertinent questions in functional genomics, with wide implications for most areas of biology. Here, we synthesise classic and recent evidence on the regulatory logic of enhancers, including the principles of enhancer organisation, factors that facilitate and delimit enhancer-promoter communication, and the joint effects of multiple enhancers. We show how modern approaches building on classic insights have begun to unravel the complexity of enhancer-promoter relationships, paving the way towards a quantitative understanding of gene control.


Subject(s)
Enhancer Elements, Genetic/genetics , Promoter Regions, Genetic/genetics , Transcription, Genetic/genetics , Animals , Communication , Gene Expression Regulation/genetics , Humans
7.
Nucleic Acids Res ; 48(6): 2866-2879, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32112106

ABSTRACT

Identifying DNA cis-regulatory modules (CRMs) that control the expression of specific genes is crucial for deciphering the logic of transcriptional control. Natural genetic variation can point to the possible gene regulatory function of specific sequences through their allelic associations with gene expression. However, comprehensive identification of causal regulatory sequences in brute-force association testing without incorporating prior knowledge is challenging due to limited statistical power and effects of linkage disequilibrium. Sequence variants affecting transcription factor (TF) binding at CRMs have a strong potential to influence gene regulatory function, which provides a motivation for prioritizing such variants in association testing. Here, we generate an atlas of CRMs showing predicted allelic variation in TF binding affinity in human lymphoblastoid cell lines and test their association with the expression of their putative target genes inferred from Promoter Capture Hi-C and immediate linear proximity. We reveal >1300 CRM TF-binding variants associated with target gene expression, the majority of them undetected with standard association testing. A large proportion of CRMs showing associations with the expression of genes they contact in 3D localize to the promoter regions of other genes, supporting the notion of 'epromoters': dual-action CRMs with promoter and distal enhancer activity.


Subject(s)
Gene Expression Regulation , Promoter Regions, Genetic , Transcription Factors/metabolism , Base Sequence , Binding Sites , Chromatin/metabolism , Genes, Reporter , Protein Binding , Quantitative Trait Loci/genetics , Transcription, Genetic
8.
Bioinformatics ; 35(22): 4764-4766, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31197313

ABSTRACT

SUMMARY: Capture Hi-C is a powerful approach for detecting chromosomal interactions involving, at least on one end, DNA regions of interest, such as gene promoters. We present Chicdiff, an R package for robust detection of differential interactions in Capture Hi-C data. Chicdiff enhances a state-of-the-art differential testing approach for count data with bespoke normalization and multiple testing procedures that account for specific statistical properties of Capture Hi-C. We validate Chicdiff on published Promoter Capture Hi-C data in human Monocytes and CD4+ T cells, identifying multitudes of cell type-specific interactions, and confirming the overall positive association between promoter interactions and gene expression. AVAILABILITY AND IMPLEMENTATION: Chicdiff is implemented as an R package that is publicly available at https://github.com/RegulatoryGenomicsGroup/chicdiff. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Chromatin , Chromosomes , Software , Humans , Promoter Regions, Genetic
9.
Blood ; 127(13): e12-23, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-26809507

ABSTRACT

Comprehensive study of transcriptional control processes will be required to enhance our understanding of both normal and malignant hematopoiesis. Modern sequencing technologies have revolutionized our ability to generate genome-scale expression and histone modification profiles, transcription factor (TF)-binding maps, and also comprehensive chromatin-looping information. Many of these technologies, however, require large numbers of cells, and therefore cannot be applied to rare hematopoietic stem/progenitor cell (HSPC) populations. The stem cell factor-dependent multipotent progenitor cell line HPC-7 represents a well-recognized cell line model for HSPCs. Here we report genome-wide maps for 17 TFs, 3 histone modifications, DNase I hypersensitive sites, and high-resolution promoter-enhancer interactomes in HPC-7 cells. Integrated analysis of these complementary data sets revealed TF occupancy patterns of genomic regions involved in promoter-anchored loops. Moreover, preferential associations between pairs of TFs bound at either ends of chromatin loops led to the identification of 4 previously unrecognized protein-protein interactions between key blood stem cell regulators. All HPC-7 data sets are freely available both through standard repositories and a user-friendly Web interface. Together with previously generated genome-wide data sets, this study integrates HPC-7 data into a genomic resource on par with ENCODE tier 1 cell lines and, importantly, is the only current model with comprehensive genome-scale data that is relevant to HSPC biology.


Subject(s)
Gene Expression Regulation , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Regulatory Sequences, Nucleic Acid , Transcription Factors/metabolism , Animals , Binding Sites/genetics , Cells, Cultured , Chromatin Immunoprecipitation , Embryo, Mammalian , Genome , HEK293 Cells , Humans , Mice , Mice, Transgenic , Promoter Regions, Genetic , Protein Binding/genetics , Transcription Factors/genetics
10.
J Cell Sci ; 128(20): 3707-13, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26345368

ABSTRACT

Endocytosis is essential for uptake of many substances into the cell, but how it links to nutritional signalling is poorly understood. Here, we show a new role for endocytosis in regulating the response to low phosphate in Schizosaccharomyces pombe. Loss of function of myosin I (Myo1), Sla2/End4 or Arp2, proteins involved in the early steps of endocytosis, led to increased proliferation in low-phosphate medium compared to controls. We show that once cells are deprived of phosphate they undergo a quiescence response that is dependent on the endocytic function of Myo1. Transcriptomic analysis revealed a wide perturbation of gene expression with induction of stress-regulated genes upon phosphate starvation in wild-type but not Δmyo1 cells. Thus, endocytosis plays a pivotal role in mediating the cellular response to nutrients, bridging the external environment and internal molecular functions of the cell.


Subject(s)
Endocytosis/physiology , Gene Expression Regulation, Fungal/physiology , Myosin Heavy Chains/metabolism , Phosphates/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/metabolism , Signal Transduction/physiology , Actin-Related Protein 2/genetics , Actin-Related Protein 2/metabolism , Gene Deletion , Myosin Heavy Chains/genetics , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics , Transcriptome
11.
Proc Natl Acad Sci U S A ; 111(6): 2235-40, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24469834

ABSTRACT

In female mammals, one of the two X chromosomes is transcriptionally silenced to equalize X-linked gene dosage relative to XY males, a process termed X chromosome inactivation. Mechanistically, this is thought to occur via directed recruitment of chromatin modifying factors by the master regulator, X-inactive specific transcript (Xist) RNA, which localizes in cis along the entire length of the chromosome. A well-studied example is the recruitment of polycomb repressive complex 2 (PRC2), for which there is evidence of a direct interaction involving the PRC2 proteins Enhancer of zeste 2 (Ezh2) and Supressor of zeste 12 (Suz12) and the A-repeat region located at the 5' end of Xist RNA. In this study, we have analyzed Xist-mediated recruitment of PRC2 using two approaches, microarray-based epigenomic mapping and superresolution 3D structured illumination microscopy. Making use of an ES cell line carrying an inducible Xist transgene located on mouse chromosome 17, we show that 24 h after synchronous induction of Xist expression, acquired PRC2 binding sites map predominantly to gene-rich regions, notably within gene bodies. Paradoxically, these new sites of PRC2 deposition do not correlate with Xist-mediated gene silencing. The 3D structured illumination microscopy was performed to assess the relative localization of PRC2 proteins and Xist RNA. Unexpectedly, we observed significant spatial separation and absence of colocalization both in the inducible Xist transgene ES cell line and in normal XX somatic cells. Our observations argue against direct interaction between Xist RNA and PRC2 proteins and, as such, prompt a reappraisal of the mechanism for PRC2 recruitment in X chromosome inactivation.


Subject(s)
Polycomb-Group Proteins/isolation & purification , RNA, Long Noncoding/isolation & purification , RNA/genetics , Animals , Cell Line , Gene Silencing , Mice , Microscopy, Electron , Oligonucleotide Array Sequence Analysis , RNA, Long Noncoding/genetics , Transcription, Genetic
12.
Bioessays ; 36(8): 798-806, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24888900

ABSTRACT

Transcription factor binding sites (TFBSs) on the DNA are generally accepted as the key nodes of gene control. However, the multitudes of TFBSs identified in genome-wide studies, some of them seemingly unconstrained in evolution, have prompted the view that in many cases TF binding may serve no biological function. Yet, insights from transcriptional biochemistry, population genetics and functional genomics suggest that rather than segregating into 'functional' or 'non-functional', TFBS inputs to their target genes may be generally cumulative, with varying degrees of potency and redundancy. As TFBS redundancy can be diminished by mutations and environmental stress, some of the apparently 'spurious' sites may turn out to be important for maintaining adequate transcriptional regulation under these conditions. This has significant implications for interpreting the phenotypic effects of TFBS mutations, particularly in the context of genome-wide association studies for complex traits.


Subject(s)
Transcription Factors/physiology , Transcriptional Activation , Animals , Binding Sites , Evolution, Molecular , Humans , Promoter Regions, Genetic , Protein Binding , Transcription, Genetic
13.
Blood ; 121(10): 1769-82, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23303821

ABSTRACT

Ikaros family DNA-binding proteins are critical regulators of B-cell development. Because the current knowledge of Ikaros targets in B-cell progenitors is limited, we have identified genes that are bound and regulated by Ikaros in pre-B cells. To elucidate the role of Ikaros in B-cell lineage specification and differentiation, we analyzed the differential expression of Ikaros targets during the progression of multipotent to lymphoid-restricted progenitors, B- and T-cell lineage specification, and progression along the B-cell lineage. Ikaros targets accounted for one-half of all genes up-regulated during B-cell lineage specification in vivo, explaining the essential role of Ikaros in this process. Expression of the Ikaros paralogs Ikzf1 and Ikzf3 increases incrementally during B-cell progenitor differentiation, and, remarkably, inducible Ikaros expression in cycling pre-B cells was sufficient to drive transcriptional changes resembling the differentiation of cycling to resting pre-Bcells in vivo. The data suggest that Ikaros transcription factor dosage drives the progression of progenitors along a predetermined lineage by regulating multiple targets in key pathways, including pre-B­cell receptor signaling, cell cycle progression, and lymphocyte receptor rearrangement.Our approachmay be of general use to map the contribution of transcription factors to cell lineage commitment and differentiation.


Subject(s)
B-Lymphocytes/cytology , Cell Differentiation , Cell Lineage , Genome , Ikaros Transcription Factor/metabolism , Precursor Cells, B-Lymphoid/cytology , Transcription Factors/metabolism , Animals , B-Lymphocytes/metabolism , Binding Sites , Cell Cycle , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation , Ikaros Transcription Factor/genetics , Lymphocyte Activation , Mice , Precursor Cells, B-Lymphoid/metabolism , Signal Transduction , Transcription Factors/genetics
14.
Nat Cardiovasc Res ; 3(6): 714-733, 2024.
Article in English | MEDLINE | ID: mdl-38898928

ABSTRACT

Aberrant vascular smooth muscle cell (VSMC) homeostasis and proliferation characterize vascular diseases causing heart attack and stroke. Here we elucidate molecular determinants governing VSMC proliferation by reconstructing gene regulatory networks from single-cell transcriptomics and epigenetic profiling. We detect widespread activation of enhancers at disease-relevant loci in proliferation-predisposed VSMCs. We compared gene regulatory network rewiring between injury-responsive and nonresponsive VSMCs, which suggested shared transcription factors but differing target loci between VSMC states. Through in silico perturbation analysis, we identified and prioritized previously unrecognized regulators of proliferation, including RUNX1 and TIMP1. Moreover, we showed that the pioneer transcription factor RUNX1 increased VSMC responsiveness and that TIMP1 feeds back to promote VSMC proliferation through CD74-mediated STAT3 signaling. Both RUNX1 and the TIMP1-CD74 axis were expressed in human VSMCs, showing low levels in normal arteries and increased expression in disease, suggesting clinical relevance and potential as vascular disease targets.

15.
Nat Cell Biol ; 8(5): 532-8, 2006 May.
Article in English | MEDLINE | ID: mdl-16570078

ABSTRACT

Epigenetic genome modifications are thought to be important for specifying the lineage and developmental stage of cells within a multicellular organism. Here, we show that the epigenetic profile of pluripotent embryonic stem cells (ES) is distinct from that of embryonic carcinoma cells, haematopoietic stem cells (HSC) and their differentiated progeny. Silent, lineage-specific genes replicated earlier in pluripotent cells than in tissue-specific stem cells or differentiated cells and had unexpectedly high levels of acetylated H3K9 and methylated H3K4. Unusually, in ES cells these markers of open chromatin were also combined with H3K27 trimethylation at some non-expressed genes. Thus, pluripotency of ES cells is characterized by a specific epigenetic profile where lineage-specific genes may be accessible but, if so, carry repressive H3K27 trimethylation modifications. H3K27 methylation is functionally important for preventing expression of these genes in ES cells as premature expression occurs in embryonic ectoderm development (Eed)-deficient ES cells. Our data suggest that lineage-specific genes are primed for expression in ES cells but are held in check by opposing chromatin modifications.


Subject(s)
Chromatin/genetics , Pluripotent Stem Cells/metabolism , Animals , Carcinoma/genetics , Cell Line , Cells, Cultured , DNA Replication Timing/genetics , Down-Regulation/genetics , Epigenesis, Genetic , Gene Expression Profiling , Genetic Markers , Hematopoietic Stem Cells/metabolism , Mice , Multipotent Stem Cells/metabolism , Polycomb Repressive Complex 2 , Repressor Proteins/metabolism , T-Lymphocytes/metabolism
16.
Cardiovasc Res ; 119(5): 1279-1294, 2023 05 22.
Article in English | MEDLINE | ID: mdl-35994249

ABSTRACT

AIMS: Quiescent, differentiated adult vascular smooth muscle cells (VSMCs) can be induced to proliferate and switch phenotype. Such plasticity underlies blood vessel homeostasis and contributes to vascular disease development. Oligoclonal VSMC contribution is a hallmark of end-stage vascular disease. Here, we aim to understand cellular mechanisms underpinning generation of this VSMC oligoclonality. METHODS AND RESULTS: We investigate the dynamics of VSMC clone formation using confocal microscopy and single-cell transcriptomics in VSMC-lineage-traced animal models. We find that activation of medial VSMC proliferation occurs at low frequency after vascular injury and that only a subset of expanding clones migrate, which together drives formation of oligoclonal neointimal lesions. VSMC contribution in small atherosclerotic lesions is typically from one or two clones, similar to observations in mature lesions. Low frequency (<0.1%) of clonal VSMC proliferation is also observed in vitro. Single-cell RNA-sequencing revealed progressive cell state changes across a contiguous VSMC population at onset of injury-induced proliferation. Proliferating VSMCs mapped selectively to one of two distinct trajectories and were associated with cells showing extensive phenotypic switching. A proliferation-associated transitory state shared pronounced similarities with atypical SCA1+ VSMCs from uninjured mouse arteries and VSMCs in healthy human aorta. We show functionally that clonal expansion of SCA1+ VSMCs from healthy arteries occurs at higher rate and frequency compared with SCA1- cells. CONCLUSION: Our data suggest that activation of proliferation at low frequency is a general, cell-intrinsic feature of VSMCs. We show that rare VSMCs in healthy arteries display VSMC phenotypic switching akin to that observed in pathological vessel remodelling and that this is a conserved feature of mouse and human healthy arteries. The increased proliferation of modulated VSMCs from healthy arteries suggests that these cells respond more readily to disease-inducing cues and could drive oligoclonal VSMC expansion.


Subject(s)
Atherosclerosis , Cardiovascular Diseases , Spinocerebellar Ataxias , Adult , Animals , Humans , Muscle, Smooth, Vascular/pathology , Cardiovascular Diseases/pathology , Cell Proliferation , Atherosclerosis/pathology , Phenotype , Spinocerebellar Ataxias/pathology , Myocytes, Smooth Muscle/pathology , Cells, Cultured
17.
Dev Cell ; 57(12): 1439-1441, 2022 06 20.
Article in English | MEDLINE | ID: mdl-35728521

ABSTRACT

In a recent issue of Nature, Zuin et al. (2022) integrate a transcriptional enhancer into various genomic locations surrounding a reporter gene promoter in order to determine how enhancer-promoter proximity affects gene expression. Their findings suggest a non-linear relationship, which the authors explain via a quantitative biophysical model with intriguing biological implications.


Subject(s)
Enhancer Elements, Genetic , Enhancer Elements, Genetic/genetics , Gene Expression , Genes, Reporter , Promoter Regions, Genetic/genetics
18.
Proc Natl Acad Sci U S A ; 105(22): 7797-802, 2008 Jun 03.
Article in English | MEDLINE | ID: mdl-18509048

ABSTRACT

Regulatory T (Treg) cells safeguard against autoimmunity and immune pathology. Because determinants of the Treg cell fate are not completely understood, we have delineated signaling events that control the de novo expression of Foxp3 in naive peripheral CD4 T cells and in thymocytes. We report that premature termination of TCR signaling and inibition of phosphatidyl inositol 3-kinase (PI3K) p110alpha, p110delta, protein kinase B (Akt), or mammalian target of rapamycin (mTOR) conferred Foxp3 expression and Treg-like gene expression profiles. Conversely, continued TCR signaling and constitutive PI3K/Akt/mTOR activity antagonised Foxp3 induction. At the chromatin level, di- and trimethylation of lysine 4 of histone H3 (H3K4me2 and -3) near the Foxp3 transcription start site (TSS) and within the 5' untranslated region (UTR) preceded active Foxp3 expression and, like Foxp3 inducibility, was lost upon continued TCR stimulation. These data demonstrate that the PI3K/Akt/mTOR signaling network regulates Foxp3 expression.


Subject(s)
Forkhead Transcription Factors/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Antigen, T-Cell/metabolism , 5' Untranslated Regions/metabolism , Animals , Forkhead Transcription Factors/genetics , Histones/metabolism , Isoenzymes/metabolism , Methylation , Mice , Mice, Inbred Strains , MicroRNAs/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Receptors, Antigen, T-Cell/agonists , Signal Transduction , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases , Transcription Initiation Site , Transforming Growth Factor beta/metabolism
19.
Front Genet ; 12: 745672, 2021.
Article in English | MEDLINE | ID: mdl-34759959

ABSTRACT

Genetic variants showing associations with specific biological traits and diseases detected by genome-wide association studies (GWAS) commonly map to non-coding DNA regulatory regions. Many of these regions are located considerable distances away from the genes they regulate and come into their proximity through 3D chromosomal interactions. We previously developed COGS, a statistical pipeline for linking GWAS variants with their putative target genes based on 3D chromosomal interaction data arising from high-resolution assays such as Promoter Capture Hi-C (PCHi-C). Here, we applied COGS to COVID-19 Host Genetic Consortium (HGI) GWAS meta-analysis data on COVID-19 susceptibility and severity using our previously generated PCHi-C results in 17 human primary cell types and SARS-CoV-2-infected lung carcinoma cells. We prioritise 251 genes putatively associated with these traits, including 16 out of 47 genes highlighted by the GWAS meta-analysis authors. The prioritised genes are expressed in a broad array of tissues, including, but not limited to, blood and brain cells, and are enriched for genes involved in the inflammatory response to viral infection. Our prioritised genes and pathways, in conjunction with results from other prioritisation approaches and targeted validation experiments, will aid in the understanding of COVID-19 pathology, paving the way for novel treatments.

20.
Nat Protoc ; 16(9): 4144-4176, 2021 09.
Article in English | MEDLINE | ID: mdl-34373652

ABSTRACT

Capture Hi-C is widely used to obtain high-resolution profiles of chromosomal interactions involving, at least on one end, regions of interest such as gene promoters. Signal detection in Capture Hi-C data is challenging and cannot be adequately accomplished with tools developed for other chromosome conformation capture methods, including standard Hi-C. Capture Hi-C Analysis of Genomic Organization (CHiCAGO) is a computational pipeline developed specifically for Capture Hi-C analysis. It implements a statistical model accounting for biological and technical background components, as well as bespoke normalization and multiple testing procedures for this data type. Here we provide a step-by-step guide to the CHiCAGO workflow that is aimed at users with basic experience of the command line and R. We also describe more advanced strategies for tuning the key parameters for custom experiments and provide guidance on data preprocessing and downstream analysis using companion tools. In a typical experiment, CHiCAGO takes ~2-3 h to run, although pre- and postprocessing steps may take much longer.


Subject(s)
Chromosomes/physiology , Models, Statistical , Software , Chromatin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL