Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 151
Filtrar
1.
Cell ; 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38925112

RESUMEN

Most mammalian genes have multiple polyA sites, representing a substantial source of transcript diversity regulated by the cleavage and polyadenylation (CPA) machinery. To better understand how these proteins govern polyA site choice, we introduce CPA-Perturb-seq, a multiplexed perturbation screen dataset of 42 CPA regulators with a 3' scRNA-seq readout that enables transcriptome-wide inference of polyA site usage. We develop a framework to detect perturbation-dependent changes in polyadenylation and characterize modules of co-regulated polyA sites. We find groups of intronic polyA sites regulated by distinct components of the nuclear RNA life cycle, including elongation, splicing, termination, and surveillance. We train and validate a deep neural network (APARENT-Perturb) for tandem polyA site usage, delineating a cis-regulatory code that predicts perturbation response and reveals interactions between regulatory complexes. Our work highlights the potential for multiplexed single-cell perturbation screens to further our understanding of post-transcriptional regulation.

2.
Cell ; 186(24): 5254-5268.e26, 2023 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-37944513

RESUMEN

A fundamental feature of cellular growth is that total protein and RNA amounts increase with cell size to keep concentrations approximately constant. A key component of this is that global transcription rates increase in larger cells. Here, we identify RNA polymerase II (RNAPII) as the limiting factor scaling mRNA transcription with cell size in budding yeast, as transcription is highly sensitive to the dosage of RNAPII but not to other components of the transcriptional machinery. Our experiments support a dynamic equilibrium model where global RNAPII transcription at a given size is set by the mass action recruitment kinetics of unengaged nucleoplasmic RNAPII to the genome. However, this only drives a sub-linear increase in transcription with size, which is then partially compensated for by a decrease in mRNA decay rates as cells enlarge. Thus, limiting RNAPII and feedback on mRNA stability work in concert to scale mRNA amounts with cell size.


Asunto(s)
Tamaño de la Célula , ARN Polimerasa II , Transcripción Genética , Retroalimentación , ARN Polimerasa II/metabolismo , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo
3.
Cell ; 185(26): 4937-4953.e23, 2022 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-36563664

RESUMEN

To define the multi-cellular epigenomic and transcriptional landscape of cardiac cellular development, we generated single-cell chromatin accessibility maps of human fetal heart tissues. We identified eight major differentiation trajectories involving primary cardiac cell types, each associated with dynamic transcription factor (TF) activity signatures. We contrasted regulatory landscapes of iPSC-derived cardiac cell types and their in vivo counterparts, which enabled optimization of in vitro differentiation of epicardial cells. Further, we interpreted sequence based deep learning models of cell-type-resolved chromatin accessibility profiles to decipher underlying TF motif lexicons. De novo mutations predicted to affect chromatin accessibility in arterial endothelium were enriched in congenital heart disease (CHD) cases vs. controls. In vitro studies in iPSCs validated the functional impact of identified variation on the predicted developmental cell types. This work thus defines the cell-type-resolved cis-regulatory sequence determinants of heart development and identifies disruption of cell type-specific regulatory elements in CHD.


Asunto(s)
Cromatina , Cardiopatías Congénitas , Humanos , Cromatina/genética , Cardiopatías Congénitas/genética , Corazón , Mutación , Análisis de la Célula Individual
4.
Cell ; 184(19): 5053-5069.e23, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34390642

RESUMEN

Genetic perturbations of cortical development can lead to neurodevelopmental disease, including autism spectrum disorder (ASD). To identify genomic regions crucial to corticogenesis, we mapped the activity of gene-regulatory elements generating a single-cell atlas of gene expression and chromatin accessibility both independently and jointly. This revealed waves of gene regulation by key transcription factors (TFs) across a nearly continuous differentiation trajectory, distinguished the expression programs of glial lineages, and identified lineage-determining TFs that exhibited strong correlation between linked gene-regulatory elements and expression levels. These highly connected genes adopted an active chromatin state in early differentiating cells, consistent with lineage commitment. Base-pair-resolution neural network models identified strong cell-type-specific enrichment of noncoding mutations predicted to be disruptive in a cohort of ASD individuals and identified frequently disrupted TF binding sites. This approach illustrates how cell-type-specific mapping can provide insights into the programs governing human development and disease.


Asunto(s)
Corteza Cerebral/embriología , Cromatina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Análisis de la Célula Individual , Astrocitos/citología , Diferenciación Celular , Linaje de la Célula/genética , Análisis por Conglomerados , Aprendizaje Profundo , Epigénesis Genética , Lógica Difusa , Glutamatos/metabolismo , Humanos , Mutación/genética , Neuronas/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética
5.
Cell ; 183(7): 2020-2035.e16, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33326746

RESUMEN

Thousands of proteins localize to the nucleus; however, it remains unclear which contain transcriptional effectors. Here, we develop HT-recruit, a pooled assay where protein libraries are recruited to a reporter, and their transcriptional effects are measured by sequencing. Using this approach, we measure gene silencing and activation for thousands of domains. We find a relationship between repressor function and evolutionary age for the KRAB domains, discover that Homeodomain repressor strength is collinear with Hox genetic organization, and identify activities for several domains of unknown function. Deep mutational scanning of the CRISPRi KRAB maps the co-repressor binding surface and identifies substitutions that improve stability/silencing. By tiling 238 proteins, we find repressors as short as ten amino acids. Finally, we report new activator domains, including a divergent KRAB. These results provide a resource of 600 human proteins containing effectors and demonstrate a scalable strategy for assigning functions to protein domains.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Sistemas CRISPR-Cas/genética , Femenino , Silenciador del Gen , Genes Reporteros , Células HEK293 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células K562 , Lentivirus/fisiología , Anotación de Secuencia Molecular , Mutación/genética , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas/genética , Dominios Proteicos , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Reproducibilidad de los Resultados , Transcripción Genética , Dedos de Zinc
6.
Cell ; 166(2): 451-467, 2016 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-27419872

RESUMEN

Stem-cell differentiation to desired lineages requires navigating alternating developmental paths that often lead to unwanted cell types. Hence, comprehensive developmental roadmaps are crucial to channel stem-cell differentiation toward desired fates. To this end, here, we map bifurcating lineage choices leading from pluripotency to 12 human mesodermal lineages, including bone, muscle, and heart. We defined the extrinsic signals controlling each binary lineage decision, enabling us to logically block differentiation toward unwanted fates and rapidly steer pluripotent stem cells toward 80%-99% pure human mesodermal lineages at most branchpoints. This strategy enabled the generation of human bone and heart progenitors that could engraft in respective in vivo models. Mapping stepwise chromatin and single-cell gene expression changes in mesoderm development uncovered somite segmentation, a previously unobservable human embryonic event transiently marked by HOPX expression. Collectively, this roadmap enables navigation of mesodermal development to produce transplantable human tissue progenitors and uncover developmental processes. VIDEO ABSTRACT.


Asunto(s)
Mesodermo/citología , Transducción de Señal , Proteínas Morfogenéticas Óseas/metabolismo , Huesos/citología , Huesos/metabolismo , Corazón/crecimiento & desarrollo , Proteínas de Homeodominio/metabolismo , Humanos , Mesodermo/metabolismo , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes/metabolismo , Línea Primitiva/citología , Línea Primitiva/metabolismo , Análisis de la Célula Individual , Somitos/metabolismo , Células Madre , Proteínas Supresoras de Tumor/metabolismo , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/metabolismo
7.
Cell ; 162(5): 1051-65, 2015 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-26300125

RESUMEN

Deciphering the impact of genetic variants on gene regulation is fundamental to understanding human disease. Although gene regulation often involves long-range interactions, it is unknown to what extent non-coding genetic variants influence distal molecular phenotypes. Here, we integrate chromatin profiling for three histone marks in lymphoblastoid cell lines (LCLs) from 75 sequenced individuals with LCL-specific Hi-C and ChIA-PET-based chromatin contact maps to uncover one of the largest collections of local and distal histone quantitative trait loci (hQTLs). Distal QTLs are enriched within topologically associated domains and exhibit largely concordant variation of chromatin state coordinated by proximal and distal non-coding genetic variants. Histone QTLs are enriched for common variants associated with autoimmune diseases and enable identification of putative target genes of disease-associated variants from genome-wide association studies. These analyses provide insights into how genetic variation can affect human disease phenotypes by coordinated changes in chromatin at interacting regulatory elements.


Asunto(s)
Cromatina/metabolismo , Cromosomas Humanos/metabolismo , Proyecto Genoma Humano , Línea Celular , Cromosomas Humanos/química , Estudios de Cohortes , Femenino , Redes Reguladoras de Genes , Estudio de Asociación del Genoma Completo , Histonas/metabolismo , Humanos , Linfocitos/metabolismo , Masculino , Sitios de Carácter Cuantitativo , Elementos Reguladores de la Transcripción
8.
Cell ; 158(3): 673-88, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25083876

RESUMEN

Trimethylation of histone H3 at lysine 4 (H3K4me3) is a chromatin modification known to mark the transcription start sites of active genes. Here, we show that H3K4me3 domains that spread more broadly over genes in a given cell type preferentially mark genes that are essential for the identity and function of that cell type. Using the broadest H3K4me3 domains as a discovery tool in neural progenitor cells, we identify novel regulators of these cells. Machine learning models reveal that the broadest H3K4me3 domains represent a distinct entity, characterized by increased marks of elongation. The broadest H3K4me3 domains also have more paused polymerase at their promoters, suggesting a unique transcriptional output. Indeed, genes marked by the broadest H3K4me3 domains exhibit enhanced transcriptional consistency and [corrected] increased transcriptional levels, and perturbation of H3K4me3 breadth leads to changes in transcriptional consistency. Thus, H3K4me3 breadth contains information that could ensure transcriptional precision at key cell identity/function genes.


Asunto(s)
Células/metabolismo , Código de Histonas , Histonas/metabolismo , Transcripción Genética , Animales , Inteligencia Artificial , Genómica , Humanos , Lisina/metabolismo , Metilación , Ratones Endogámicos C57BL , Células-Madre Neurales/metabolismo , ARN Polimerasa II/metabolismo
9.
Nature ; 623(7987): 608-615, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37938768

RESUMEN

Cell therapies have yielded durable clinical benefits for patients with cancer, but the risks associated with the development of therapies from manipulated human cells are understudied. For example, we lack a comprehensive understanding of the mechanisms of toxicities observed in patients receiving T cell therapies, including recent reports of encephalitis caused by reactivation of human herpesvirus 6 (HHV-6)1. Here, through petabase-scale viral genomics mining, we examine the landscape of human latent viral reactivation and demonstrate that HHV-6B can become reactivated in cultures of human CD4+ T cells. Using single-cell sequencing, we identify a rare population of HHV-6 'super-expressors' (about 1 in 300-10,000 cells) that possess high viral transcriptional activity, among research-grade allogeneic chimeric antigen receptor (CAR) T cells. By analysing single-cell sequencing data from patients receiving cell therapy products that are approved by the US Food and Drug Administration2 or are in clinical studies3-5, we identify the presence of HHV-6-super-expressor CAR T cells in patients in vivo. Together, the findings of our study demonstrate the utility of comprehensive genomics analyses in implicating cell therapy products as a potential source contributing to the lytic HHV-6 infection that has been reported in clinical trials1,6-8 and may influence the design and production of autologous and allogeneic cell therapies.


Asunto(s)
Linfocitos T CD4-Positivos , Herpesvirus Humano 6 , Inmunoterapia Adoptiva , Receptores Quiméricos de Antígenos , Activación Viral , Latencia del Virus , Humanos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Ensayos Clínicos como Asunto , Regulación Viral de la Expresión Génica , Genómica , Herpesvirus Humano 6/genética , Herpesvirus Humano 6/aislamiento & purificación , Herpesvirus Humano 6/fisiología , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Encefalitis Infecciosa/complicaciones , Encefalitis Infecciosa/virología , Receptores Quiméricos de Antígenos/inmunología , Infecciones por Roseolovirus/complicaciones , Infecciones por Roseolovirus/virología , Análisis de Expresión Génica de una Sola Célula , Carga Viral
10.
Mol Cell ; 81(23): 4861-4875.e7, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34731644

RESUMEN

Biosynthesis scales with cell size such that protein concentrations generally remain constant as cells grow. As an exception, synthesis of the cell-cycle inhibitor Whi5 "sub-scales" with cell size so that its concentration is lower in larger cells to promote cell-cycle entry. Here, we find that transcriptional control uncouples Whi5 synthesis from cell size, and we identify histones as the major class of sub-scaling transcripts besides WHI5 by screening for similar genes. Histone synthesis is thereby matched to genome content rather than cell size. Such sub-scaling proteins are challenged by asymmetric cell division because proteins are typically partitioned in proportion to newborn cell volume. To avoid this fate, Whi5 uses chromatin-binding to partition similar protein amounts to each newborn cell regardless of cell size. Disrupting both Whi5 synthesis and chromatin-based partitioning weakens G1 size control. Thus, specific transcriptional and partitioning mechanisms determine protein sub-scaling to control cell size.


Asunto(s)
Cromatina/química , Regulación Fúngica de la Expresión Génica , Saccharomyces cerevisiae/metabolismo , Schizosaccharomyces/metabolismo , Transcripción Genética , Ciclo Celular , Cromatina/metabolismo , Biología Computacional , Histonas/química , Homeostasis , Hibridación Fluorescente in Situ , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Análisis de Regresión , Proteínas Represoras , Proteínas de Saccharomyces cerevisiae
11.
Nat Methods ; 21(4): 723-734, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38504114

RESUMEN

The ENCODE Consortium's efforts to annotate noncoding cis-regulatory elements (CREs) have advanced our understanding of gene regulatory landscapes. Pooled, noncoding CRISPR screens offer a systematic approach to investigate cis-regulatory mechanisms. The ENCODE4 Functional Characterization Centers conducted 108 screens in human cell lines, comprising >540,000 perturbations across 24.85 megabases of the genome. Using 332 functionally confirmed CRE-gene links in K562 cells, we established guidelines for screening endogenous noncoding elements with CRISPR interference (CRISPRi), including accurate detection of CREs that exhibit variable, often low, transcriptional effects. Benchmarking five screen analysis tools, we find that CASA produces the most conservative CRE calls and is robust to artifacts of low-specificity single guide RNAs. We uncover a subtle DNA strand bias for CRISPRi in transcribed regions with implications for screen design and analysis. Together, we provide an accessible data resource, predesigned single guide RNAs for targeting 3,275,697 ENCODE SCREEN candidate CREs with CRISPRi and screening guidelines to accelerate functional characterization of the noncoding genome.


Asunto(s)
Sistemas CRISPR-Cas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Humanos , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Sistemas CRISPR-Cas/genética , Genoma , Células K562 , ARN Guía de Sistemas CRISPR-Cas
12.
Nature ; 593(7858): 238-243, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33828297

RESUMEN

Genome-wide association studies (GWAS) have identified thousands of noncoding loci that are associated with human diseases and complex traits, each of which could reveal insights into the mechanisms of disease1. Many of the underlying causal variants may affect enhancers2,3, but we lack accurate maps of enhancers and their target genes to interpret such variants. We recently developed the activity-by-contact (ABC) model to predict which enhancers regulate which genes and validated the model using CRISPR perturbations in several cell types4. Here we apply this ABC model to create enhancer-gene maps in 131 human cell types and tissues, and use these maps to interpret the functions of GWAS variants. Across 72 diseases and complex traits, ABC links 5,036 GWAS signals to 2,249 unique genes, including a class of 577 genes that appear to influence multiple phenotypes through variants in enhancers that act in different cell types. In inflammatory bowel disease (IBD), causal variants are enriched in predicted enhancers by more than 20-fold in particular cell types such as dendritic cells, and ABC achieves higher precision than other regulatory methods at connecting noncoding variants to target genes. These variant-to-function maps reveal an enhancer that contains an IBD risk variant and that regulates the expression of PPIF to alter the membrane potential of mitochondria in macrophages. Our study reveals principles of genome regulation, identifies genes that affect IBD and provides a resource and generalizable strategy to connect risk variants of common diseases to their molecular and cellular functions.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Predisposición Genética a la Enfermedad , Variación Genética/genética , Genoma Humano/genética , Estudio de Asociación del Genoma Completo , Enfermedades Inflamatorias del Intestino/genética , Línea Celular , Cromosomas Humanos Par 10/genética , Ciclofilinas/genética , Células Dendríticas , Femenino , Humanos , Macrófagos/metabolismo , Masculino , Mitocondrias/metabolismo , Especificidad de Órganos/genética , Fenotipo
13.
Nature ; 583(7818): 737-743, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32728247

RESUMEN

Physical interactions between distal regulatory elements have a key role in regulating gene expression, but the extent to which these interactions vary between cell types and contribute to cell-type-specific gene expression remains unclear. Here, to address these questions as part of phase III of the Encyclopedia of DNA Elements (ENCODE), we mapped cohesin-mediated chromatin loops, using chromatin interaction analysis by paired-end tag sequencing (ChIA-PET), and analysed gene expression in 24 diverse human cell types, including core ENCODE cell lines. Twenty-eight per cent of all chromatin loops vary across cell types; these variations modestly correlate with changes in gene expression and are effective at grouping cell types according to their tissue of origin. The connectivity of genes corresponds to different functional classes, with housekeeping genes having few contacts, and dosage-sensitive genes being more connected to enhancer elements. This atlas of chromatin loops complements the diverse maps of regulatory architecture that comprise the ENCODE Encyclopedia, and will help to support emerging analyses of genome structure and function.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cromatina/química , Cromatina/genética , Proteínas Cromosómicas no Histona/metabolismo , Genoma Humano/genética , Anotación de Secuencia Molecular , Empalme Alternativo/genética , Diferenciación Celular/genética , Línea Celular , Células/metabolismo , Cromatina/metabolismo , Secuenciación de Inmunoprecipitación de Cromatina , Elementos de Facilitación Genéticos/genética , Regulación de la Expresión Génica , Humanos , Conformación Molecular , Regiones Promotoras Genéticas/genética , Cohesinas
14.
Nature ; 580(7801): 136-141, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32238925

RESUMEN

Cancer genomics studies have identified thousands of putative cancer driver genes1. Development of high-throughput and accurate models to define the functions of these genes is a major challenge. Here we devised a scalable cancer-spheroid model and performed genome-wide CRISPR screens in 2D monolayers and 3D lung-cancer spheroids. CRISPR phenotypes in 3D more accurately recapitulated those of in vivo tumours, and genes with differential sensitivities between 2D and 3D conditions were highly enriched for genes that are mutated in lung cancers. These analyses also revealed drivers that are essential for cancer growth in 3D and in vivo, but not in 2D. Notably, we found that carboxypeptidase D is responsible for removal of a C-terminal RKRR motif2 from the α-chain of the insulin-like growth factor 1 receptor that is critical for receptor activity. Carboxypeptidase D expression correlates with patient outcomes in patients with lung cancer, and loss of carboxypeptidase D reduced tumour growth. Our results reveal key differences between 2D and 3D cancer models, and establish a generalizable strategy for performing CRISPR screens in spheroids to reveal cancer vulnerabilities.


Asunto(s)
Sistemas CRISPR-Cas/genética , Técnicas de Cultivo de Célula/métodos , Proliferación Celular/genética , Genoma Humano/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Esferoides Celulares/patología , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Secuencias de Aminoácidos , Animales , Carboxipeptidasas/antagonistas & inhibidores , Carboxipeptidasas/deficiencia , Carboxipeptidasas/genética , Carboxipeptidasas/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , Terapia Molecular Dirigida , Mutación , Fenotipo , Receptor IGF Tipo 1/química , Receptor IGF Tipo 1/metabolismo , Transducción de Señal , Esferoides Celulares/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Nature ; 583(7818): 699-710, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32728249

RESUMEN

The human and mouse genomes contain instructions that specify RNAs and proteins and govern the timing, magnitude, and cellular context of their production. To better delineate these elements, phase III of the Encyclopedia of DNA Elements (ENCODE) Project has expanded analysis of the cell and tissue repertoires of RNA transcription, chromatin structure and modification, DNA methylation, chromatin looping, and occupancy by transcription factors and RNA-binding proteins. Here we summarize these efforts, which have produced 5,992 new experimental datasets, including systematic determinations across mouse fetal development. All data are available through the ENCODE data portal (https://www.encodeproject.org), including phase II ENCODE1 and Roadmap Epigenomics2 data. We have developed a registry of 926,535 human and 339,815 mouse candidate cis-regulatory elements, covering 7.9 and 3.4% of their respective genomes, by integrating selected datatypes associated with gene regulation, and constructed a web-based server (SCREEN; http://screen.encodeproject.org) to provide flexible, user-defined access to this resource. Collectively, the ENCODE data and registry provide an expansive resource for the scientific community to build a better understanding of the organization and function of the human and mouse genomes.


Asunto(s)
ADN/genética , Bases de Datos Genéticas , Genoma/genética , Genómica , Anotación de Secuencia Molecular , Sistema de Registros , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Cromatina/genética , Cromatina/metabolismo , ADN/química , Huella de ADN , Metilación de ADN/genética , Momento de Replicación del ADN , Desoxirribonucleasa I/metabolismo , Genoma Humano , Histonas/metabolismo , Humanos , Ratones , Ratones Transgénicos , Proteínas de Unión al ARN/genética , Transcripción Genética/genética , Transposasas/metabolismo
16.
Genome Res ; 32(3): 512-523, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35042722

RESUMEN

The intrinsic DNA sequence preferences and cell type-specific cooperative partners of transcription factors (TFs) are typically highly conserved. Hence, despite the rapid evolutionary turnover of individual TF binding sites, predictive sequence models of cell type-specific genomic occupancy of a TF in one species should generalize to closely matched cell types in a related species. To assess the viability of cross-species TF binding prediction, we train neural networks to discriminate ChIP-seq peak locations from genomic background and evaluate their performance within and across species. Cross-species predictive performance is consistently worse than within-species performance, which we show is caused in part by species-specific repeats. To account for this domain shift, we use an augmented network architecture to automatically discourage learning of training species-specific sequence features. This domain adaptation approach corrects for prediction errors on species-specific repeats and improves overall cross-species model performance. Our results show that cross-species TF binding prediction is feasible when models account for domain shifts driven by species-specific repeats.


Asunto(s)
Redes Neurales de la Computación , Factores de Transcripción , Sitios de Unión , Secuenciación de Inmunoprecipitación de Cromatina , Biología Computacional/métodos , Unión Proteica , Factores de Transcripción/metabolismo
18.
Br J Cancer ; 130(10): 1687-1696, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38561434

RESUMEN

BACKGROUND: Menopausal hormone therapy (MHT), a common treatment to relieve symptoms of menopause, is associated with a lower risk of colorectal cancer (CRC). To inform CRC risk prediction and MHT risk-benefit assessment, we aimed to evaluate the joint association of a polygenic risk score (PRS) for CRC and MHT on CRC risk. METHODS: We used data from 28,486 postmenopausal women (11,519 cases and 16,967 controls) of European descent. A PRS based on 141 CRC-associated genetic variants was modeled as a categorical variable in quartiles. Multiplicative interaction between PRS and MHT use was evaluated using logistic regression. Additive interaction was measured using the relative excess risk due to interaction (RERI). 30-year cumulative risks of CRC for 50-year-old women according to MHT use and PRS were calculated. RESULTS: The reduction in odds ratios by MHT use was larger in women within the highest quartile of PRS compared to that in women within the lowest quartile of PRS (p-value = 2.7 × 10-8). At the highest quartile of PRS, the 30-year CRC risk was statistically significantly lower for women taking any MHT than for women not taking any MHT, 3.7% (3.3%-4.0%) vs 6.1% (5.7%-6.5%) (difference 2.4%, P-value = 1.83 × 10-14); these differences were also statistically significant but smaller in magnitude in the lowest PRS quartile, 1.6% (1.4%-1.8%) vs 2.2% (1.9%-2.4%) (difference 0.6%, P-value = 1.01 × 10-3), indicating 4 times greater reduction in absolute risk associated with any MHT use in the highest compared to the lowest quartile of genetic CRC risk. CONCLUSIONS: MHT use has a greater impact on the reduction of CRC risk for women at higher genetic risk. These findings have implications for the development of risk prediction models for CRC and potentially for the consideration of genetic information in the risk-benefit assessment of MHT use.


Asunto(s)
Neoplasias Colorrectales , Predisposición Genética a la Enfermedad , Humanos , Femenino , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/epidemiología , Persona de Mediana Edad , Estudios de Casos y Controles , Factores de Riesgo , Anciano , Terapia de Reemplazo de Hormonas/efectos adversos , Medición de Riesgo , Menopausia , Posmenopausia , Terapia de Reemplazo de Estrógeno/efectos adversos
19.
Blood ; 139(16): 2534-2546, 2022 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-35030251

RESUMEN

Master regulators, such as the hematopoietic transcription factor (TF) GATA1, play an essential role in orchestrating lineage commitment and differentiation. However, the precise mechanisms by which such TFs regulate transcription through interactions with specific cis-regulatory elements remain incompletely understood. Here, we describe a form of congenital hemolytic anemia caused by missense mutations in an intrinsically disordered region of GATA1, with a poorly understood role in transcriptional regulation. Through integrative functional approaches, we demonstrate that these mutations perturb GATA1 transcriptional activity by partially impairing nuclear localization and selectively altering precise chromatin occupancy by GATA1. These alterations in chromatin occupancy and concordant chromatin accessibility changes alter faithful gene expression, with failure to both effectively silence and activate select genes necessary for effective terminal red cell production. We demonstrate how disease-causing mutations can reveal regulatory mechanisms that enable the faithful genomic targeting of master TFs during cellular differentiation.


Asunto(s)
Anemia , Factor de Transcripción GATA1 , Diferenciación Celular/genética , Cromatina/genética , Inmunoprecipitación de Cromatina , Eritropoyesis/genética , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Humanos
20.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34088849

RESUMEN

Somatic cell transcription factors are critical to maintaining cellular identity and constitute a barrier to human somatic cell reprogramming; yet a comprehensive understanding of the mechanism of action is lacking. To gain insight, we examined epigenome remodeling at the onset of human nuclear reprogramming by profiling human fibroblasts after fusion with murine embryonic stem cells (ESCs). By assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) and chromatin immunoprecipitation sequencing we identified enrichment for the activator protein 1 (AP-1) transcription factor c-Jun at regions of early transient accessibility at fibroblast-specific enhancers. Expression of a dominant negative AP-1 mutant (dnAP-1) reduced accessibility and expression of fibroblast genes, overcoming the barrier to reprogramming. Remarkably, efficient reprogramming of human fibroblasts to induced pluripotent stem cells was achieved by transduction with vectors expressing SOX2, KLF4, and inducible dnAP-1, demonstrating that dnAP-1 can substitute for exogenous human OCT4. Mechanistically, we show that the AP-1 component c-Jun has two unexpected temporally distinct functions in human reprogramming: 1) to potentiate fibroblast enhancer accessibility and fibroblast-specific gene expression, and 2) to bind to and repress OCT4 as a complex with MBD3. Our findings highlight AP-1 as a previously unrecognized potent dual gatekeeper of the somatic cell state.


Asunto(s)
Reprogramación Celular , Regulación de la Expresión Génica , Células Madre Embrionarias de Ratones/metabolismo , Factor de Transcripción AP-1/metabolismo , Animales , Línea Celular , Humanos , Factor 4 Similar a Kruppel , Ratones , Factor de Transcripción AP-1/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda