Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 114
Filtrer
1.
Genome Biol ; 25(1): 146, 2024 06 06.
Article de Anglais | MEDLINE | ID: mdl-38844976

RÉSUMÉ

BACKGROUND: DNA methylation is an important epigenetic modification which has numerous roles in modulating genome function. Its levels are spatially correlated across the genome, typically high in repressed regions but low in transcription factor (TF) binding sites and active regulatory regions. However, the mechanisms establishing genome-wide and TF binding site methylation patterns are still unclear. RESULTS: Here we use a comparative approach to investigate the association of DNA methylation to TF binding evolution in mammals. Specifically, we experimentally profile DNA methylation and combine this with published occupancy profiles of five distinct TFs (CTCF, CEBPA, HNF4A, ONECUT1, FOXA1) in the liver of five mammalian species (human, macaque, mouse, rat, dog). TF binding sites are lowly methylated, but they often also have intermediate methylation levels. Furthermore, biding sites are influenced by the methylation status of CpGs in their wider binding regions even when CpGs are absent from the core binding motif. Employing a classification and clustering approach, we extract distinct and species-conserved patterns of DNA methylation levels at TF binding regions. CEBPA, HNF4A, ONECUT1, and FOXA1 share the same methylation patterns, while CTCF's differ. These patterns characterize alternative functions and chromatin landscapes of TF-bound regions. Leveraging our phylogenetic framework, we find DNA methylation gain upon evolutionary loss of TF occupancy, indicating coordinated evolution. Furthermore, each methylation pattern has its own evolutionary trajectory reflecting its genomic contexts. CONCLUSIONS: Our epigenomic analyses indicate a role for DNA methylation in TF binding changes across species including that specific DNA methylation profiles characterize TF binding and are associated with their regulatory activity, chromatin contexts, and evolutionary trajectories.


Sujet(s)
Méthylation de l'ADN , Évolution moléculaire , Facteurs de transcription , Animaux , Sites de fixation , Humains , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Souris , Rats , Ilots CpG , Chiens , Facteur nucléaire hépatocytaire HNF-3 alpha/métabolisme , Facteur nucléaire hépatocytaire HNF-3 alpha/génétique , Liaison aux protéines , Foie/métabolisme , Facteur nucléaire hépatocytaire HNF-4/métabolisme , Facteur nucléaire hépatocytaire HNF-4/génétique , Protéines liant les séquences stimulatrices de type CCAAT/métabolisme , Protéines liant les séquences stimulatrices de type CCAAT/génétique
3.
Cell ; 187(3): 692-711.e26, 2024 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-38262408

RÉSUMÉ

Transcription factors (TFs) can define distinct cellular identities despite nearly identical DNA-binding specificities. One mechanism for achieving regulatory specificity is DNA-guided TF cooperativity. Although in vitro studies suggest that it may be common, examples of such cooperativity remain scarce in cellular contexts. Here, we demonstrate how "Coordinator," a long DNA motif composed of common motifs bound by many basic helix-loop-helix (bHLH) and homeodomain (HD) TFs, uniquely defines the regulatory regions of embryonic face and limb mesenchyme. Coordinator guides cooperative and selective binding between the bHLH family mesenchymal regulator TWIST1 and a collective of HD factors associated with regional identities in the face and limb. TWIST1 is required for HD binding and open chromatin at Coordinator sites, whereas HD factors stabilize TWIST1 occupancy at Coordinator and titrate it away from HD-independent sites. This cooperativity results in the shared regulation of genes involved in cell-type and positional identities and ultimately shapes facial morphology and evolution.


Sujet(s)
Protéines de liaison à l'ADN , Développement embryonnaire , Facteurs de transcription , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Sites de fixation , ADN/métabolisme , Protéines de liaison à l'ADN/métabolisme , Régulation de l'expression des gènes , Mésoderme/métabolisme , Facteurs de transcription/métabolisme , Humains , Animaux , Souris , Membres/croissance et développement
4.
Nature ; 625(7993): 41-50, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38093018

RÉSUMÉ

Gene expression is regulated by transcription factors that work together to read cis-regulatory DNA sequences. The 'cis-regulatory code' - how cells interpret DNA sequences to determine when, where and how much genes should be expressed - has proven to be exceedingly complex. Recently, advances in the scale and resolution of functional genomics assays and machine learning have enabled substantial progress towards deciphering this code. However, the cis-regulatory code will probably never be solved if models are trained only on genomic sequences; regions of homology can easily lead to overestimation of predictive performance, and our genome is too short and has insufficient sequence diversity to learn all relevant parameters. Fortunately, randomly synthesized DNA sequences enable testing a far larger sequence space than exists in our genomes, and designed DNA sequences enable targeted queries to maximally improve the models. As the same biochemical principles are used to interpret DNA regardless of its source, models trained on these synthetic data can predict genomic activity, often better than genome-trained models. Here we provide an outlook on the field, and propose a roadmap towards solving the cis-regulatory code by a combination of machine learning and massively parallel assays using synthetic DNA.


Sujet(s)
Génomique , Apprentissage machine , Modèles génétiques , Séquences d'acides nucléiques régulatrices , ADN/synthèse chimique , ADN/génétique , ADN/métabolisme , Séquences d'acides nucléiques régulatrices/génétique , Facteurs de transcription/métabolisme
6.
bioRxiv ; 2023 May 29.
Article de Anglais | MEDLINE | ID: mdl-37398193

RÉSUMÉ

Transcription factors (TFs) can define distinct cellular identities despite nearly identical DNA-binding specificities. One mechanism for achieving regulatory specificity is DNA-guided TF cooperativity. Although in vitro studies suggest it may be common, examples of such cooperativity remain scarce in cellular contexts. Here, we demonstrate how 'Coordinator', a long DNA motif comprised of common motifs bound by many basic helix-loop-helix (bHLH) and homeodomain (HD) TFs, uniquely defines regulatory regions of embryonic face and limb mesenchyme. Coordinator guides cooperative and selective binding between the bHLH family mesenchymal regulator TWIST1 and a collective of HD factors associated with regional identities in the face and limb. TWIST1 is required for HD binding and open chromatin at Coordinator sites, while HD factors stabilize TWIST1 occupancy at Coordinator and titrate it away from HD-independent sites. This cooperativity results in shared regulation of genes involved in cell-type and positional identities, and ultimately shapes facial morphology and evolution.

7.
Nat Biotechnol ; 41(2): 197-203, 2023 02.
Article de Anglais | MEDLINE | ID: mdl-36163549

RÉSUMÉ

Here we describe a competitive genome editing method that measures the effect of mutations on molecular functions, based on precision CRISPR editing using template libraries with either the original or altered sequence, and a sequence tag, enabling direct comparison between original and mutated cells. Using the example of the MYC oncogene, we identify important transcriptional targets and show that E-box mutations at MYC target gene promoters reduce cellular fitness.


Sujet(s)
Édition de gène , Facteurs de transcription , Sites de fixation/génétique , Clustered regularly interspaced short palindromic repeats/génétique , Systèmes CRISPR-Cas/génétique , Édition de gène/méthodes , Régions promotrices (génétique)/génétique , Facteurs de transcription/composition chimique , Facteurs de transcription/génétique
8.
Dev Cell ; 57(8): 1024-1036.e5, 2022 04 25.
Article de Anglais | MEDLINE | ID: mdl-35472319

RÉSUMÉ

The transcription factor Myc drives cell growth across animal phyla and is activated in most forms of human cancer. However, it is unclear which Myc target genes need to be regulated to induce growth and whether multiple targets act additively or if induction of each target is individually necessary. Here, we identified Myc target genes whose regulation is conserved between humans and flies and deleted Myc-binding sites (E-boxes) in the promoters of fourteen of these genes in Drosophila. E-box mutants of essential genes were homozygous viable, indicating that the E-boxes are not required for basal expression. Eight E-box mutations led to Myc-like phenotypes; the strongest mutant, ppanEbox-/-, also made the flies resistant to Myc-induced cell growth without affecting Myc-induced apoptosis. The ppanEbox-/- flies are healthy and display only a minor developmental delay, suggesting that it may be possible to treat or prevent tumorigenesis by targeting individual downstream targets of Myc.


Sujet(s)
Protéines proto-oncogènes c-myc , Ribosomes , Animaux , Prolifération cellulaire/génétique , Protéines proto-oncogènes c-myc/métabolisme , Ribosomes/métabolisme , Activation de la transcription , Régulation positive
9.
Nat Genet ; 54(3): 283-294, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-35190730

RÉSUMÉ

DNA can determine where and when genes are expressed, but the full set of sequence determinants that control gene expression is unknown. Here, we measured the transcriptional activity of DNA sequences that represent an ~100 times larger sequence space than the human genome using massively parallel reporter assays (MPRAs). Machine learning models revealed that transcription factors (TFs) generally act in an additive manner with weak grammar and that most enhancers increase expression from a promoter by a mechanism that does not appear to involve specific TF-TF interactions. The enhancers themselves can be classified into three types: classical, closed chromatin and chromatin dependent. We also show that few TFs are strongly active in a cell, with most activities being similar between cell types. Individual TFs can have multiple gene regulatory activities, including chromatin opening and enhancing, promoting and determining transcription start site (TSS) activity, consistent with the view that the TF binding motif is the key atomic unit of gene expression.


Sujet(s)
Séquences d'acides nucléiques régulatrices , Facteurs de transcription , Sites de fixation/génétique , Génome humain/génétique , Humains , Liaison aux protéines , Séquences d'acides nucléiques régulatrices/génétique , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme
10.
Cell Genom ; 2(12): 100214, 2022 Dec 14.
Article de Anglais | MEDLINE | ID: mdl-36778047

RÉSUMÉ

We combined functional genomics and human genetics to investigate processes that affect type 1 diabetes (T1D) risk by mediating beta cell survival in response to proinflammatory cytokines. We mapped 38,931 cytokine-responsive candidate cis-regulatory elements (cCREs) in beta cells using ATAC-seq and snATAC-seq and linked them to target genes using co-accessibility and HiChIP. Using a genome-wide CRISPR screen in EndoC-ßH1 cells, we identified 867 genes affecting cytokine-induced survival, and genes promoting survival and up-regulated in cytokines were enriched at T1D risk loci. Using SNP-SELEX, we identified 2,229 variants in cytokine-responsive cCREs altering transcription factor (TF) binding, and variants altering binding of TFs regulating stress, inflammation, and apoptosis were enriched for T1D risk. At the 16p13 locus, a fine-mapped T1D variant altering TF binding in a cytokine-induced cCRE interacted with SOCS1, which promoted survival in cytokine exposure. Our findings reveal processes and genes acting in beta cells during inflammation that modulate T1D risk.

11.
Elife ; 102021 12 13.
Article de Anglais | MEDLINE | ID: mdl-34898428

RÉSUMÉ

Precision CRISPR gene editing relies on the cellular homology-directed DNA repair (HDR) to introduce custom DNA sequences to target sites. The HDR editing efficiency varies between cell types and genomic sites, and the sources of this variation are incompletely understood. Here, we have studied the effect of 450 DNA repair protein-Cas9 fusions on CRISPR genome editing outcomes. We find the majority of fusions to improve precision genome editing only modestly in a locus- and cell-type specific manner. We identify Cas9-POLD3 fusion that enhances editing by speeding up the initiation of DNA repair. We conclude that while DNA repair protein fusions to Cas9 can improve HDR CRISPR editing, most need to be optimized to the cell type and genomic site, highlighting the diversity of factors contributing to locus-specific genome editing outcomes.


Sujet(s)
Protéine-9 associée à CRISPR/génétique , Protéine-9 associée à CRISPR/métabolisme , Systèmes CRISPR-Cas/génétique , Cellules cultivées/physiologie , DNA polymerase III/génétique , DNA polymerase III/métabolisme , Édition de gène/méthodes , Réparation de l'ADN/génétique , Réparation de l'ADN/physiologie , Humains
12.
Cancers (Basel) ; 13(18)2021 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-34572796

RÉSUMÉ

Studies have indicated that some genes involved in carcinogenesis are highly methylated in their promoter regions but nevertheless strongly transcribed. It has been proposed that transcription factors could bind specifically to methylated promoters and trigger transcription. We looked at this rather comprehensively for pancreatic ductal adenocarcinoma (PDAC) and studied some cases in more detail. Some 2% of regulated genes in PDAC exhibited higher transcription coupled to promoter hypermethylation in comparison to healthy tissue. Screening 661 transcription factors, several were found to bind specifically to methylated promoters, in particular molecules of the NFAT family. One of them-NFATc1-was substantially more strongly expressed in PDAC than control tissue and exhibited a strong oncogenic role. Functional studies combined with computational analyses allowed determining affected genes. A prominent one was gene ALDH1A3, which accelerates PDAC metastasis and correlates with a bad prognosis. Further studies confirmed the direct up-regulation of ALDH1A3 transcription by NFATc1 promoter binding in a methylation-dependent process, providing insights into the oncogenic role of transcription activation in PDAC that is promoted by DNA methylation.

13.
Nat Commun ; 12(1): 5448, 2021 09 14.
Article de Anglais | MEDLINE | ID: mdl-34521855

RÉSUMÉ

Mechanical forces in a constrained cellular environment were recently established as a facilitator of chromosomal damage. Whether this could contribute to tumorigenesis is not known. Uterine leiomyomas are common neoplasms that display relatively few chromosomal aberrations. We hypothesized that if mechanical forces contribute to chromosomal damage, signs of this could be seen in uterine leiomyomas from parous women. We examined the karyotypes of 1946 tumors, and found a striking overrepresentation of chromosomal damage associated with parity. We then subjected myometrial cells to physiological forces similar to those encountered during pregnancy, and found this to cause DNA breaks and a DNA repair response. While mechanical forces acting in constrained cellular environments may thus contribute to neoplastic degeneration, and genesis of uterine leiomyoma, further studies are needed to prove possible causality of the observed association. No evidence for progression to malignancy was found.


Sujet(s)
Aberrations des chromosomes , Réparation de l'ADN , Léiomyome/génétique , Complexe médiateur/génétique , Parité , Tumeurs de l'utérus/génétique , Adulte , Phénomènes biomécaniques , Cassures double-brin de l'ADN , Femelle , Expression des gènes , Humains , Hystérectomie , Caryotype , Léiomyome/étiologie , Léiomyome/anatomopathologie , Léiomyome/chirurgie , Mutation , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Myomètre/métabolisme , Myomètre/anatomopathologie , Grossesse , Culture de cellules primaires , Études prospectives , Tumeurs de l'utérus/étiologie , Tumeurs de l'utérus/anatomopathologie , Tumeurs de l'utérus/chirurgie
14.
Nature ; 596(7872): 398-403, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-34349258

RÉSUMÉ

One in four women suffers from uterine leiomyomas (ULs)-benign tumours of the uterine wall, also known as uterine fibroids-at some point in premenopausal life. ULs can cause excessive bleeding, pain and infertility1, and are a common cause of hysterectomy2. They emerge through at least three distinct genetic drivers: mutations in MED12 or FH, or genomic rearrangement of HMGA23. Here we created genome-wide datasets, using DNA, RNA, assay for transposase-accessible chromatin (ATAC), chromatin immunoprecipitation (ChIP) and HiC chromatin immunoprecipitation (HiChIP) sequencing of primary tissues to profoundly understand the genesis of UL. We identified somatic mutations in genes encoding six members of the SRCAP histone-loading complex4, and found that germline mutations in the SRCAP members YEATS4 and ZNHIT1 predispose women to UL. Tumours bearing these mutations showed defective deposition of the histone variant H2A.Z. In ULs, H2A.Z occupancy correlated positively with chromatin accessibility and gene expression, and negatively with DNA methylation, but these correlations were weak in tumours bearing SRCAP complex mutations. In these tumours, open chromatin emerged at transcription start sites where H2A.Z was lost, which was associated with upregulation of genes. Furthermore, YEATS4 defects were associated with abnormal upregulation of bivalent embryonic stem cell genes, as previously shown in mice5. Our work describes a potential mechanism of tumorigenesis-epigenetic instability caused by deficient H2A.Z deposition-and suggests that ULs arise through an aberrant differentiation program driven by deranged chromatin, emanating from a small number of mutually exclusive driver mutations.


Sujet(s)
Assemblage et désassemblage de la chromatine , Chromatine/génétique , Chromatine/métabolisme , Histone/déficit , Léiomyome/génétique , Mutation , Tumeurs de l'utérus/génétique , Carcinogenèse/génétique , Lignée cellulaire , Chromatine/composition chimique , Cellules souches embryonnaires/métabolisme , Épigenèse génétique , Femelle , Régulation de l'expression des gènes tumoraux , Histone/génétique , Histone/métabolisme , Humains , Léiomyome/métabolisme , Léiomyome/anatomopathologie , Ligases/génétique , Complexe répresseur Polycomb-1/génétique , Protéines du groupe Polycomb/génétique , Facteurs de transcription/génétique , Tumeurs de l'utérus/métabolisme , Tumeurs de l'utérus/anatomopathologie
15.
Curr Opin Struct Biol ; 71: 171-179, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34364091

RÉSUMÉ

In eukaryotic cells, DNA interacts with two main types of binding proteins: transcription factors and histones. Histones form the core of nucleosomes and display weak sequence preference owing to differences in bendability of different DNA sequences. By contrast, the affinity of transcription factors is highly dependent on DNA sequence - all sequences are bound with moderate affinity, but only few specific sequences are bound more tightly via molecular recognition of the DNA bases. Transcription factors can interact with nucleosomes directly by recognizing nucleosome-associated DNA and also indirectly by recruiting histone-modifying enzymes and nucleosome remodelers. These interactions result in sequence-dependent formation of a pattern of open and closed chromatin, where specific positions are occupied by transcription factors, histone-modifying enzymes, and modified histones. These patterns are then recognized by large DNA-associated macromolecular complexes such as cohesin and RNA polymerase II, which are involved in regulation of higher-order chromatin structure and transcription, respectively. Here, we review recent work that has provided structural and mechanistic insight into the interactions between all these classes of DNA-associated proteins.


Sujet(s)
Nucléosomes , Facteurs de transcription , Chromatine , Assemblage et désassemblage de la chromatine , Histone/métabolisme , Facteurs de transcription/métabolisme
16.
Hum Mol Genet ; 30(24): 2429-2440, 2021 11 30.
Article de Anglais | MEDLINE | ID: mdl-34274970

RÉSUMÉ

Many hereditary cancer syndromes are associated with an increased risk of small and large intestinal adenocarcinomas. However, conditions bearing a high risk to both adenocarcinomas and neuroendocrine tumors are yet to be described. We studied a family with 16 individuals in four generations affected by a wide spectrum of intestinal tumors, including hyperplastic polyps, adenomas, small intestinal neuroendocrine tumors, and colorectal and small intestinal adenocarcinomas. To assess the genetic susceptibility and understand the novel phenotype, we utilized multiple molecular methods, including whole genome sequencing, RNA sequencing, single cell sequencing, RNA in situ hybridization and organoid culture. We detected a heterozygous deletion at the cystic fibrosis locus (7q31.2) perfectly segregating with the intestinal tumor predisposition in the family. The deletion removes a topologically associating domain border between CFTR and WNT2, aberrantly activating WNT2 in the intestinal epithelium. These consequences suggest that the deletion predisposes to small intestinal neuroendocrine tumors and small and large intestinal adenocarcinomas, and reveals the broad tumorigenic effects of aberrant WNT activation in the human intestine.


Sujet(s)
Adénocarcinome , Adénomes , Tumeurs colorectales , Tumeurs neuroendocrines , Adénocarcinome/génétique , Adénocarcinome/anatomopathologie , Adénomes/génétique , Adénomes/anatomopathologie , Tumeurs colorectales/génétique , Humains , Muqueuse intestinale/anatomopathologie , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/anatomopathologie , Protéine Wnt2
17.
Oncogene ; 40(36): 5533-5547, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34302118

RÉSUMÉ

Cancer is the most complex genetic disease known, with mutations implicated in more than 250 genes. However, it is still elusive which specific mutations found in human patients lead to tumorigenesis. Here we show that a combination of oncogenes that is characteristic of liver cancer (CTNNB1, TERT, MYC) induces senescence in human fibroblasts and primary hepatocytes. However, reprogramming fibroblasts to a liver progenitor fate, induced hepatocytes (iHeps), makes them sensitive to transformation by the same oncogenes. The transformed iHeps are highly proliferative, tumorigenic in nude mice, and bear gene expression signatures of liver cancer. These results show that tumorigenesis is triggered by a combination of three elements: the set of driver mutations, the cellular lineage, and the state of differentiation of the cells along the lineage. Our results provide direct support for the role of cell identity as a key determinant in transformation and establish a paradigm for studying the dynamic role of oncogenic drivers in human tumorigenesis.


Sujet(s)
Transformation cellulaire néoplasique , Proto-oncogènes , Animaux , Différenciation cellulaire , Humains , Souris , Translocation génétique
18.
Gastroenterology ; 161(2): 592-607, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-33930428

RÉSUMÉ

BACKGROUND & AIMS: Inflammatory bowel disease (IBD) is a chronic, relapsing inflammatory disorder associated with an elevated risk of colorectal cancer (CRC). IBD-associated CRC (IBD-CRC) may represent a distinct pathway of tumorigenesis compared to sporadic CRC (sCRC). Our aim was to comprehensively characterize IBD-associated tumorigenesis integrating multiple high-throughput approaches, and to compare the results with in-house data sets from sCRCs. METHODS: Whole-genome sequencing, single nucleotide polymorphism arrays, RNA sequencing, genome-wide methylation analysis, and immunohistochemistry were performed using fresh-frozen and formalin-fixed tissue samples of tumor and corresponding normal tissues from 31 patients with IBD-CRC. RESULTS: Transcriptome-based tumor subtyping revealed the complete absence of canonical epithelial tumor subtype associated with WNT signaling in IBD-CRCs, dominated instead by mesenchymal stroma-rich subtype. Negative WNT regulators AXIN2 and RNF43 were strongly down-regulated in IBD-CRCs and chromosomal gains at HNF4A, a negative regulator of WNT-induced epithelial-mesenchymal transition (EMT), were less frequent compared to sCRCs. Enrichment of hypomethylation at HNF4α binding sites was detected solely in sCRC genomes. PIGR and OSMR involved in mucosal immunity were dysregulated via epigenetic modifications in IBD-CRCs. Genome-wide analysis showed significant enrichment of noncoding mutations to 5'untranslated region of TP53 in IBD-CRCs. As reported previously, somatic mutations in APC and KRAS were less frequent in IBD-CRCs compared to sCRCs. CONCLUSIONS: Distinct mechanisms of WNT pathway dysregulation skew IBD-CRCs toward mesenchymal tumor subtype, which may affect prognosis and treatment options. Increased OSMR signaling may favor the establishment of mesenchymal tumors in patients with IBD.


Sujet(s)
Marqueurs biologiques tumoraux/génétique , Transformation cellulaire néoplasique/génétique , Néoplasmes associés aux colites/génétique , Méthylation de l'ADN , Épigenèse génétique , Maladies inflammatoires intestinales/génétique , Transcriptome , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Transformation cellulaire néoplasique/immunologie , Transformation cellulaire néoplasique/anatomopathologie , Néoplasmes associés aux colites/immunologie , Néoplasmes associés aux colites/anatomopathologie , Analyse de mutations d'ADN , Épigénomique , Femelle , Finlande , Analyse de profil d'expression de gènes , Séquençage nucléotidique à haut débit , Humains , Maladies inflammatoires intestinales/immunologie , Maladies inflammatoires intestinales/anatomopathologie , Mâle , Adulte d'âge moyen , Mutation , Grading des tumeurs , Stadification tumorale , Séquençage par oligonucléotides en batterie , Polymorphisme de nucléotide simple , Analyse de séquence d'ARN , Microenvironnement tumoral/génétique , Microenvironnement tumoral/immunologie , Séquençage du génome entier
19.
Nature ; 591(7848): 147-151, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-33505025

RÉSUMÉ

Many sequence variants have been linked to complex human traits and diseases1, but deciphering their biological functions remains challenging, as most of them reside in noncoding DNA. Here we have systematically assessed the binding of 270 human transcription factors to 95,886 noncoding variants in the human genome using an ultra-high-throughput multiplex protein-DNA binding assay, termed single-nucleotide polymorphism evaluation by systematic evolution of ligands by exponential enrichment (SNP-SELEX). The resulting 828 million measurements of transcription factor-DNA interactions enable estimation of the relative affinity of these transcription factors to each variant in vitro and evaluation of the current methods to predict the effects of noncoding variants on transcription factor binding. We show that the position weight matrices of most transcription factors lack sufficient predictive power, whereas the support vector machine combined with the gapped k-mer representation show much improved performance, when assessed on results from independent SNP-SELEX experiments involving a new set of 61,020 sequence variants. We report highly predictive models for 94 human transcription factors and demonstrate their utility in genome-wide association studies and understanding of the molecular pathways involved in diverse human traits and diseases.


Sujet(s)
Polymorphisme de nucléotide simple/génétique , Technique SELEX , Machine à vecteur de support , Facteurs de transcription/métabolisme , Sites de fixation/génétique , Maladie/génétique , Génome humain/génétique , Humains , Ligands , Liaison aux protéines
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...