Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 15.920
Filtrar
1.
Methods Mol Biol ; 2856: 357-400, 2025.
Artículo en Inglés | MEDLINE | ID: mdl-39283464

RESUMEN

Three-dimensional (3D) chromatin interactions, such as enhancer-promoter interactions (EPIs), loops, topologically associating domains (TADs), and A/B compartments, play critical roles in a wide range of cellular processes by regulating gene expression. Recent development of chromatin conformation capture technologies has enabled genome-wide profiling of various 3D structures, even with single cells. However, current catalogs of 3D structures remain incomplete and unreliable due to differences in technology, tools, and low data resolution. Machine learning methods have emerged as an alternative to obtain missing 3D interactions and/or improve resolution. Such methods frequently use genome annotation data (ChIP-seq, DNAse-seq, etc.), DNA sequencing information (k-mers and transcription factor binding site (TFBS) motifs), and other genomic properties to learn the associations between genomic features and chromatin interactions. In this review, we discuss computational tools for predicting three types of 3D interactions (EPIs, chromatin interactions, and TAD boundaries) and analyze their pros and cons. We also point out obstacles to the computational prediction of 3D interactions and suggest future research directions.


Asunto(s)
Cromatina , Aprendizaje Profundo , Cromatina/genética , Cromatina/metabolismo , Humanos , Biología Computacional/métodos , Aprendizaje Automático , Genómica/métodos , Elementos de Facilitación Genéticos , Regiones Promotoras Genéticas , Sitios de Unión , Genoma , Programas Informáticos
2.
Methods Mol Biol ; 2856: 327-339, 2025.
Artículo en Inglés | MEDLINE | ID: mdl-39283462

RESUMEN

Disentangling the relationship of enhancers and genes is an ongoing challenge in epigenomics. We present STARE, our software to quantify the strength of enhancer-gene interactions based on enhancer activity and chromatin contact data. It implements the generalized Activity-by-Contact (gABC) score, which allows predicting putative target genes of candidate enhancers over any desired genomic distance. The only requirement for its application is a measurement of enhancer activity. In addition to regulatory interactions, STARE calculates transcription factor (TF) affinities on gene level. We illustrate its usage on a public single-cell data set of the human heart by predicting regulatory interactions on cell type level, by giving examples on how to integrate them with other data modalities, and by constructing TF affinity matrices.


Asunto(s)
Cromatina , Elementos de Facilitación Genéticos , Epigenómica , Programas Informáticos , Humanos , Cromatina/genética , Cromatina/metabolismo , Epigenómica/métodos , Epigenoma , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Biología Computacional/métodos
3.
Methods Mol Biol ; 2856: 341-356, 2025.
Artículo en Inglés | MEDLINE | ID: mdl-39283463

RESUMEN

To reveal gene regulation mechanisms, it is essential to understand the role of regulatory elements, which are possibly distant from gene promoters. Integrative analysis of epigenetic and transcriptomic data can be used to gain insights into gene-expression regulation in specific phenotypes. Here, we discuss STITCHIT, an approach to dissect epigenetic variation in a gene-specific manner across many samples for the identification of regulatory elements without relying on peak calling algorithms. The obtained genomic regions are then further refined using a regularized linear model approach, which can also be used to predict gene expression. We illustrate the use of STITCHIT using H3k27ac ChIP-seq and RNA-seq data from the International Human Epigenome Consortium (IHEC).


Asunto(s)
Epigénesis Genética , Epigenómica , Transcriptoma , Humanos , Epigenómica/métodos , Transcriptoma/genética , Elementos de Facilitación Genéticos , Programas Informáticos , Biología Computacional/métodos , Secuenciación de Inmunoprecipitación de Cromatina/métodos , Regulación de la Expresión Génica , Algoritmos , Histonas/genética , Histonas/metabolismo , Perfilación de la Expresión Génica/métodos
4.
Proc Natl Acad Sci U S A ; 121(41): e2405001121, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39361648

RESUMEN

Well-differentiated low-grade lung neuroendocrine tumors (lung carcinoids or LNETs) are histopathologically classified as typical and atypical LNETs, but each subtype is still heterogeneous at both the molecular level and its clinical manifestation. Here, we report genome-wide profiles of primary LNETs' cis-regulatory elements by H3K27ac ChIP-seq with matching RNA-seq profiles. Analysis of these regulatory landscapes revealed three regulatory subtypes, independent of the typical/atypical classification. We identified unique differentiation signals that delineate each subtype. The "proneuronal" subtype emerges under the influence of ASCL1, SOX4, and TCF4 transcription factors, embodying a pronounced proneuronal signature. The "luminal-like" subtype is characterized by gain of acetylation at markers of luminal cells and GATA2 activation and loss of LRP5 and OTP. The "HNF+" subtype is characterized by a robust enhancer landscape driven by HNF1A, HNF4A, and FOXA3, with notable acetylation and expression of FGF signaling genes, especially FGFR3 and FGFR4, pivotal components of the FGF pathway. Our findings not only deepen the understanding of LNETs' regulatory and developmental diversity but also spotlight the HNF+ subtype's reliance on FGFR signaling. We demonstrate that targeting this pathway with FGF inhibitors curtails tumor growth both in vitro and in xenograft models, unveiling a potential vulnerability and paving the way for targeted therapies. Overall, our work provides an important resource for studying LNETs to reveal regulatory networks, differentiation signals, and therapeutically relevant dependencies.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares , Tumores Neuroendocrinos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/patología , Tumores Neuroendocrinos/metabolismo , Elementos de Facilitación Genéticos/genética , Animales , Ratones , Línea Celular Tumoral
5.
Nat Commun ; 15(1): 8584, 2024 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-39362902

RESUMEN

Genomic enhancers are key transcriptional regulators which, upon the binding of sequence-specific transcription factors, activate their cognate target promoters. Although enhancers have been extensively studied in isolation, a substantial number of genes have more than one simultaneously active enhancer, and it remains unclear how these cooperate to regulate transcription. Using Drosophila melanogaster S2 cells as a model, we assay the activities of more than a thousand individual enhancers and about a million enhancer pairs toward housekeeping and developmental core promoters with STARR-seq. We report that housekeeping and developmental enhancers show distinct modes of enhancer-enhancer cooperativity: while housekeeping enhancers are additive such that their combined activity mirrors the sum of their individual activities, developmental enhancers are super-additive and combine multiplicatively. Super-additivity between developmental enhancers is promiscuous and neither depends on the enhancers' endogenous genomic contexts nor on specific transcription factor motif signatures. However, it can be further boosted by Twist and Trl motifs and saturates for the highest levels of enhancer activity. These results have important implications for our understanding of gene regulation in complex multi-enhancer developmental loci and genomically clustered housekeeping genes, providing a rationale to interpret the transcriptional impact of non-coding mutations at different loci.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Regiones Promotoras Genéticas , Factores de Transcripción , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Genes Esenciales , Transcripción Genética , Línea Celular , Proteínas de Unión al ADN , Proteína 1 Relacionada con Twist
6.
Elife ; 132024 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-39392676

RESUMEN

Annotation of newly sequenced genomes frequently includes genes, but rarely covers important non-coding genomic features such as the cis-regulatory modules-e.g., enhancers and silencers-that regulate gene expression. Here, we begin to remedy this situation by developing a workflow for rapid initial annotation of insect regulatory sequences, and provide a searchable database resource with enhancer predictions for 33 genomes. Using our previously developed SCRMshaw computational enhancer prediction method, we predict over 2.8 million regulatory sequences along with the tissues where they are expected to be active, in a set of insect species ranging over 360 million years of evolution. Extensive analysis and validation of the data provides several lines of evidence suggesting that we achieve a high true-positive rate for enhancer prediction. One, we show that our predictions target specific loci, rather than random genomic locations. Two, we predict enhancers in orthologous loci across a diverged set of species to a significantly higher degree than random expectation would allow. Three, we demonstrate that our predictions are highly enriched for regions of accessible chromatin. Four, we achieve a validation rate in excess of 70% using in vivo reporter gene assays. As we continue to annotate both new tissues and new species, our regulatory annotation resource will provide a rich source of data for the research community and will have utility for both small-scale (single gene, single species) and large-scale (many genes, many species) studies of gene regulation. In particular, the ability to search for functionally related regulatory elements in orthologous loci should greatly facilitate studies of enhancer evolution even among distantly related species.


Asunto(s)
Genoma de los Insectos , Insectos , Anotación de Secuencia Molecular , Animales , Insectos/genética , Insectos/clasificación , Genoma de los Insectos/genética , Elementos de Facilitación Genéticos/genética , Biología Computacional/métodos , Bases de Datos Genéticas
7.
Nat Commun ; 15(1): 8793, 2024 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-39389973

RESUMEN

Approximately a quarter of the human genome consists of gene deserts, large regions devoid of genes often located adjacent to developmental genes and thought to contribute to their regulation. However, defining the regulatory functions embedded within these deserts is challenging due to their large size. Here, we explore the cis-regulatory architecture of a gene desert flanking the Shox2 gene, which encodes a transcription factor indispensable for proximal limb, craniofacial, and cardiac pacemaker development. We identify the gene desert as a regulatory hub containing more than 15 distinct enhancers recapitulating anatomical subdomains of Shox2 expression. Ablation of the gene desert leads to embryonic lethality due to Shox2 depletion in the cardiac sinus venosus, caused in part by the loss of a specific distal enhancer. The gene desert is also required for stylopod morphogenesis, mediated via distributed proximal limb enhancers. In summary, our study establishes a multi-layered role of the Shox2 gene desert in orchestrating pleiotropic developmental expression through modular arrangement and coordinated dynamics of tissue-specific enhancers.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Animales , Humanos , Ratones , Morfogénesis/genética
8.
Epigenetics Chromatin ; 17(1): 30, 2024 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-39385277

RESUMEN

BACKGROUND: It is generally accepted that methylation status of CpG sites spaced up to 50 bp apart is correlated, and accumulation of locally disordered methylation at adjacent CpG sites is involved in neoplastic transformation, acting in similar way as stochastic accumulation of mutations. RESULTS: We used EPIC microarray data from 596 samples, representing 12 healthy tissue and cell types, as well as 572 blood cancer specimens to analyze methylation status of adjacent CpG sites across human genome, and subsequently validated our findings with NGS and Sanger sequencing. Our analysis showed that there is a subset of the adjacent CpG sites in human genome, with cytosine at one CpG site methylated and the other devoid of methyl group. These loci map to enhancers that are targeted by families of transcription factors involved in cell differentiation. Moreover, our results suggest that the methylation at these loci differ between alleles within a cell, what allows for remarkable level of heterogeneity of methylation patterns. However, different types of specialized cells acquire only one specific and stable pattern of methylation at each of these loci and that pattern is to a large extent lost during neoplastic transformation. CONCLUSIONS: We identified a substantial number of adjacent CpG loci in human genome that display remarkably stable and cell type specific methylation pattern. The methylation pattern at these loci appears to reflect different methylation of alleles in cells. Furthermore, we showed that changes of methylation status at those loci are likely to be involved in regulation of the activity of enhancers and contribute to neoplastic transformation.


Asunto(s)
Islas de CpG , Metilación de ADN , Elementos de Facilitación Genéticos , Humanos , Genoma Humano , Diferenciación Celular
9.
Elife ; 132024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-39373630

RESUMEN

Gastric cancer (GC) is a major cause of cancer-related mortality worldwide. Despite the widespread recognition of tumor immunotherapy in treating unresectable GC, challenges, including ineffective immunotherapy and drug resistance, persist. Therefore, understanding the regulatory mechanisms of PD-L1, particularly in the context of super-enhancers (SEs) and zinc finger protein 36 ring finger protein-like 1 (ZFP36L1) RNA-binding protein, is crucial. In this study, we performed H3K27ac Cleavage Under Targets and Tagmentation (CUT&Tag) sequencing, investigated the heterogeneity of SEs between two GC subtypes with differential growth patterns, and revealed the immune escape signatures driven by ZFP36L1-SE in infiltrative GC through SEs inhibitors treatment. The regulation of ZFP36L1 to PD-L1 was evaluated by quantitative PCR, western blot, flow cytometry, and immunohistochemistry. Furthermore, we explored its regulatory mechanisms using a combination of molecular biology techniques, including luciferase reporter assay, GST/RNA pull-down, chromatin immunoprecipitation (ChIP)/RIP experiments, and in vivo functional assays. We demonstrated that ZFP36L1, driven by an SE, enhances IFN-γ-induced PD-L1 expression, with SPI1 identified as the specific transcription factor binding to ZFP36L1-SE. Mechanistically, ZFP36L1 binds to the adenylate uridylate-rich element in the 3' untranslated region (3'UTR) of HDAC3 mRNA, exacerbating its mRNA decay, and thereby facilitating PD-L1 abnormal transcriptional activation. Collectively, our findings provide mechanistic insights into the role of the SPI1-ZFP36L1-HDAC3-PD-L1 signaling axis in orchestrating immune escape mechanisms in GC, thereby offering valuable insights into the potential targets for immune checkpoint therapy in GC management.


Asunto(s)
Antígeno B7-H1 , Factor 1 de Respuesta al Butirato , Regulación Neoplásica de la Expresión Génica , Neoplasias Gástricas , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Humanos , Factor 1 de Respuesta al Butirato/metabolismo , Factor 1 de Respuesta al Butirato/genética , Línea Celular Tumoral , Ratones , Animales , Elementos de Facilitación Genéticos/genética
10.
Nat Commun ; 15(1): 8539, 2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39358345

RESUMEN

The primed epiblast acts as a transitional stage between the relatively homogeneous naïve epiblast and the gastrulating embryo. Its formation entails coordinated changes in regulatory circuits driven by transcription factors and epigenetic modifications. Using a multi-omic approach in human embryonic stem cell models across the spectrum of peri-implantation development, we demonstrate that the transcription factors ZIC2 and ZIC3 have overlapping but essential roles in opening primed-specific enhancers. Together, they are essential to facilitate progression to and maintain primed pluripotency. ZIC2/3 accomplish this by recruiting SWI/SNF to chromatin and loss of ZIC2/3 or degradation of SWI/SNF both prevent enhancer activation. Loss of ZIC2/3 also results in transcriptome changes consistent with perturbed Polycomb activity and a shift towards the expression of genes linked to differentiation towards the mesendoderm. Additionally, we find an intriguing dependency on the transcriptional machinery for sustained recruitment of ZIC2/3 over a subset of primed-hESC specific enhancers. Taken together, ZIC2 and ZIC3 regulate highly dynamic lineage-specific enhancers and collectively act as key regulators of human primed pluripotency.


Asunto(s)
Proteínas Cromosómicas no Histona , Proteínas de Homeodominio , Células Madre Embrionarias Humanas , Proteínas Nucleares , Células Madre Pluripotentes , Factores de Transcripción , Estratos Germinativos/citología , Estratos Germinativos/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Cromatina/metabolismo , ADN Helicasas/metabolismo , Elementos de Facilitación Genéticos , Plasticidad de la Célula , Ensamble y Desensamble de Cromatina , Transcripción Genética , Desarrollo Embrionario
11.
Nat Commun ; 15(1): 8350, 2024 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-39333469

RESUMEN

A key question in regulatory genomics is whether cis-regulatory elements (CREs) are modular elements that can function anywhere in the genome, or whether they are adapted to certain genomic locations. To distinguish between these possibilities we develop MPIRE (Massively Parallel Integrated Regulatory Elements), a technology for recurrently assaying CREs at thousands of defined locations across the genome in parallel. MPIRE allows us to separate the intrinsic activity of CREs from the effects of their genomic environments. We apply MPIRE to assay three insulator sequences at thousands of genomic locations and find that each insulator functions in locations with distinguishable properties. All three insulators can block enhancers, but each insulator blocks specific enhancers at specific locations. However, only ALOXE3 appears to block heterochromatin silencing. We conclude that insulator function is highly context dependent and that MPIRE is a robust method for revealing the context dependencies of CREs.


Asunto(s)
Elementos de Facilitación Genéticos , Elementos Aisladores , Elementos Aisladores/genética , Elementos de Facilitación Genéticos/genética , Animales , Genoma/genética , Heterocromatina/genética , Heterocromatina/metabolismo , Genómica/métodos , Ratones , Secuenciación de Nucleótidos de Alto Rendimiento/métodos
12.
Elife ; 132024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39302339

RESUMEN

Genome-wide association studies (GWAS) have identified hundreds of genetic signals associated with autoimmune disease. The majority of these signals are located in non-coding regions and likely impact cis-regulatory elements (cRE). Because cRE function is dynamic across cell types and states, profiling the epigenetic status of cRE across physiological processes is necessary to characterize the molecular mechanisms by which autoimmune variants contribute to disease risk. We localized risk variants from 15 autoimmune GWAS to cRE active during TCR-CD28 co-stimulation of naïve human CD4+ T cells. To characterize how dynamic changes in gene expression correlate with cRE activity, we measured transcript levels, chromatin accessibility, and promoter-cRE contacts across three phases of naive CD4+ T cell activation using RNA-seq, ATAC-seq, and HiC. We identified ~1200 protein-coding genes physically connected to accessible disease-associated variants at 423 GWAS signals, at least one-third of which are dynamically regulated by activation. From these maps, we functionally validated a novel stretch of evolutionarily conserved intergenic enhancers whose activity is required for activation-induced IL2 gene expression in human and mouse, and is influenced by autoimmune-associated genetic variation. The set of genes implicated by this approach are enriched for genes controlling CD4+ T cell function and genes involved in human inborn errors of immunity, and we pharmacologically validated eight implicated genes as novel regulators of T cell activation. These studies directly show how autoimmune variants and the genes they regulate influence processes involved in CD4+ T cell proliferation and activation.


Asunto(s)
Linfocitos T CD4-Positivos , Cromatina , Estudio de Asociación del Genoma Completo , Interleucina-2 , Activación de Linfocitos , Humanos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Cromatina/metabolismo , Cromatina/genética , Activación de Linfocitos/genética , Interleucina-2/genética , Interleucina-2/metabolismo , Animales , Ratones , Elementos de Facilitación Genéticos/genética , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Regulación de la Expresión Génica , Autoinmunidad/genética
13.
Int J Mol Sci ; 25(18)2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39337329

RESUMEN

Gene expression is orchestrated by a complex array of gene regulatory elements that govern transcription in a cell-type-specific manner. Though previously studied, the ability to utilize regulatory elements to identify disrupting variants remains largely elusive. To identify important factors within these regions, we generated enhancer-promoter interaction (EPI) networks and investigated the presence of disease-associated variants that fall within these regions. Our study analyzed six neuronal cell types across neural differentiation, allowing us to examine closely related cell types and across differentiation stages. Our results expand upon previous findings of cell-type specificity of enhancer, promoter, and transcription factor binding sites. Notably, we find that regulatory regions within EPI networks can identify the enrichment of variants associated with neuropsychiatric disorders within specific cell types and network sub-structures. This enrichment within sub-structures can allow for a better understanding of potential mechanisms by which variants may disrupt transcription. Together, our findings suggest that EPIs can be leveraged to better understand cell-type-specific regulatory architecture and used as a selection method for disease-associated variants to be tested in future functional assays. Combined with these future functional characterization assays, EPIs can be used to better identify and characterize regulatory variants' effects on such networks and model their mechanisms of gene regulation disruption across different disorders. Such findings can be applied in practical settings, such as diagnostic tools and drug development.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Regiones Promotoras Genéticas , Humanos , Transcripción Genética , Neuronas/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Diferenciación Celular/genética , Sitios de Unión
14.
Nat Commun ; 15(1): 8070, 2024 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-39277592

RESUMEN

Recent advances in high-resolution mapping of spatial interactions among regulatory elements support the existence of complex topological assemblies of enhancers and promoters known as enhancer-promoter hubs or cliques. Yet, organization principles of these multi-interacting enhancer-promoter hubs and their potential role in regulating gene expression in cancer remain unclear. Here, we systematically identify enhancer-promoter hubs in breast cancer, lymphoma, and leukemia. We find that highly interacting enhancer-promoter hubs form at key oncogenes and lineage-associated transcription factors potentially promoting oncogenesis of these diverse cancer types. Genomic and optical mapping of interactions among enhancer and promoter elements further show that topological alterations in hubs coincide with transcriptional changes underlying acquired resistance to targeted therapy in T cell leukemia and B cell lymphoma. Together, our findings suggest that enhancer-promoter hubs are dynamic and heterogeneous topological assemblies with the potential to control gene expression circuits promoting oncogenesis and drug resistance.


Asunto(s)
Carcinogénesis , Resistencia a Antineoplásicos , Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Regiones Promotoras Genéticas , Humanos , Regiones Promotoras Genéticas/genética , Elementos de Facilitación Genéticos/genética , Resistencia a Antineoplásicos/genética , Carcinogénesis/genética , Femenino , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Oncogenes/genética , Línea Celular Tumoral , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Leucemia/genética , Leucemia/metabolismo , Linfoma/genética , Linfoma/metabolismo
15.
Commun Biol ; 7(1): 1142, 2024 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-39277686

RESUMEN

Transcription enhancers are genomic sequences regulating common and tissue-specific genes and their disruption can contribute to human disease development and progression. Klotho, a sexually dimorphic gene specifically expressed in kidney, is well-linked to kidney dysfunction and its deletion from the mouse genome leads to premature aging and death. However, the sexually dimorphic regulation of Klotho is not understood. Here, we characterize two candidate Klotho enhancers using H3K27ac epigenetic marks and transcription factor binding and investigate their functions, individually and combined, through CRISPR-Cas9 genome engineering. We discovered that only the distal (E1), but not the proximal (E2) candidate region constitutes a functional enhancer, with the double deletion not causing Klotho expression to further decrease. E1 activity is dependent on HNF1b transcription factor binding site within the enhancer. Further, E1 controls the sexual dimorphism of Klotho as evidenced by qPCR and RNA-seq. Despite the sharp reduction of Klotho mRNA, unlike germline Klotho knockouts, mutant mice present normal phenotype, including weight, lifespan, and serum biochemistry. Lastly, only males lacking E1 display more prominent acute, but not chronic kidney injury responses, indicating a remarkable range of potential adaptation to isolated Klotho loss, especially in female E1 knockouts, retaining renoprotection despite over 80% Klotho reduction.


Asunto(s)
Elementos de Facilitación Genéticos , Glucuronidasa , Factor Nuclear 1-beta del Hepatocito , Riñón , Proteínas Klotho , Caracteres Sexuales , Animales , Proteínas Klotho/metabolismo , Ratones , Masculino , Femenino , Riñón/metabolismo , Glucuronidasa/genética , Glucuronidasa/metabolismo , Factor Nuclear 1-beta del Hepatocito/genética , Factor Nuclear 1-beta del Hepatocito/metabolismo , Ratones Noqueados , Regulación de la Expresión Génica , Ratones Endogámicos C57BL
16.
Nat Commun ; 15(1): 8209, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39294132

RESUMEN

CRISPR-based gene activation (CRISPRa) is a strategy for upregulating gene expression by targeting promoters or enhancers in a tissue/cell-type specific manner. Here, we describe an experimental framework that combines highly multiplexed perturbations with single-cell RNA sequencing (sc-RNA-seq) to identify cell-type-specific, CRISPRa-responsive cis-regulatory elements and the gene(s) they regulate. Random combinations of many gRNAs are introduced to each of many cells, which are then profiled and partitioned into test and control groups to test for effect(s) of CRISPRa perturbations of both enhancers and promoters on the expression of neighboring genes. Applying this method to a library of 493 gRNAs targeting candidate cis-regulatory elements in both K562 cells and iPSC-derived excitatory neurons, we identify gRNAs capable of specifically upregulating intended target genes and no other neighboring genes within 1 Mb, including gRNAs yielding upregulation of six autism spectrum disorder (ASD) and neurodevelopmental disorder (NDD) risk genes in neurons. A consistent pattern is that the responsiveness of individual enhancers to CRISPRa is restricted by cell type, implying a dependency on either chromatin landscape and/or additional trans-acting factors for successful gene activation. The approach outlined here may facilitate large-scale screens for gRNAs that activate genes in a cell type-specific manner.


Asunto(s)
Sistemas CRISPR-Cas , Elementos de Facilitación Genéticos , Análisis de la Célula Individual , Humanos , Análisis de la Célula Individual/métodos , Células K562 , Elementos de Facilitación Genéticos/genética , Regiones Promotoras Genéticas/genética , ARN Guía de Sistemas CRISPR-Cas/genética , Trastorno del Espectro Autista/genética , Neuronas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética
17.
Sci Rep ; 14(1): 21912, 2024 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-39300145

RESUMEN

The Notch receptor is a pleiotropic signaling protein that translates intercellular ligand interactions into changes in gene expression via the nuclear localization of the Notch intracellular Domain (NICD). Using a combination of immunohistochemistry, RNA in situ, Optogenetics and super-resolution live imaging of transcription in human cells, we show that the N1ICD can form condensates that positively facilitate Notch target gene expression. We determined that N1ICD undergoes Phase Separation Coupled Percolation (PSCP) into transcriptional condensates, which recruit, enrich, and encapsulate a broad set of core transcriptional proteins. We show that the capacity for condensation is due to the intrinsically disordered transcriptional activation domain of the N1ICD. In addition, the formation of such transcriptional condensates acts to promote Notch-mediated super enhancer-looping and concomitant activation of the MYC protooncogene expression. Overall, we introduce a novel mechanism of Notch1 activity in which discrete changes in nuclear N1ICD abundance are translated into the assembly of transcriptional condensates that facilitate gene expression by enriching essential transcriptional machineries at target genomic loci.


Asunto(s)
Elementos de Facilitación Genéticos , Receptor Notch1 , Receptor Notch1/metabolismo , Receptor Notch1/genética , Humanos , Regulación de la Expresión Génica , Núcleo Celular/metabolismo , Separación de Fases
18.
Adv Immunol ; 163: 51-96, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39271259

RESUMEN

Interferon regulatory factor-8 (IRF8) is the lineage determining transcription factor for the type one classical dendritic cell (cDC1) subset, a terminal selector for plasmacytoid dendritic cells and important for the function of monocytes. Studies of Irf8 gene regulation have identified several enhancers controlling its activity during development of progenitors in the bone marrow that precisely regulate expression at distinct developmental stages. Each enhancer responds to distinct transcription factors that are expressed at each stage. IRF8 is first expressed in early progenitors that form the monocyte dendritic cell progenitor (MDP) in response to induction of the transcription factor CCAAT/enhancer-binding protein alpha (C/EBPα) acting at the Irf8 +56 kb enhancer. IRF8 levels increase further as the MDP transits into the common dendritic cell progenitor (CDP) in response to E protein activity at the Irf8 +41 kb enhancer. Upon Nfil3-induction in CDPs leading to specification of the cDC1 progenitor, abrupt induction of BATF3 forms the JUN/BATF3/IRF8 heterotrimer that activates the Irf8 +32 kb enhancer that sustains Irf8 autoactivation throughout the cDC1 lifetime. Deletions of each of these enhancers has revealed their stage dependent activation. Surprisingly, studies of compound heterozygotes for each combination of enhancer deletions revealed that activation of each subsequent enhancer requires the successful activation of the previous enhancer in strictly cis-dependent mechanism. Successful progression of enhancer activation is finely tuned to alter the functional accessibility of subsequent enhancers to factors active in the next stage of development. The molecular basis for these phenomenon is still obscure but could have implications for genomic regulation in a broader developmental context.


Asunto(s)
Células Dendríticas , Elementos de Facilitación Genéticos , Factores Reguladores del Interferón , Humanos , Factores Reguladores del Interferón/metabolismo , Factores Reguladores del Interferón/genética , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Elementos de Facilitación Genéticos/genética , Diferenciación Celular , Regulación de la Expresión Génica , Súper Potenciadores
19.
PLoS Biol ; 22(9): e3002735, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39241209

RESUMEN

Transitions between the fed and fasted state are common in mammals. The liver orchestrates adaptive responses to feeding/fasting by transcriptionally regulating metabolic pathways of energy usage and storage. Transcriptional and enhancer dynamics following cessation of fasting (refeeding) have not been explored. We examined the transcriptional and chromatin events occurring upon refeeding in mice, including kinetic behavior and molecular drivers. We found that the refeeding response is temporally organized with the early response focused on ramping up protein translation while the later stages of refeeding drive a bifurcated lipid synthesis program. While both the cholesterol biosynthesis and lipogenesis pathways were inhibited during fasting, most cholesterol biosynthesis genes returned to their basal levels upon refeeding while most lipogenesis genes markedly overshoot above pre-fasting levels. Gene knockout, enhancer dynamics, and ChIP-seq analyses revealed that lipogenic gene overshoot is dictated by LXRα. These findings from unbiased analyses unravel the mechanism behind the long-known phenomenon of refeeding fat overshoot.


Asunto(s)
Elementos de Facilitación Genéticos , Ayuno , Receptores X del Hígado , Hígado , Animales , Masculino , Ratones , Colesterol/metabolismo , Elementos de Facilitación Genéticos/genética , Ayuno/metabolismo , Regulación de la Expresión Génica , Lipogénesis , Hígado/metabolismo , Receptores X del Hígado/metabolismo , Receptores X del Hígado/genética , Ratones Endogámicos C57BL , Ratones Noqueados
20.
Yi Chuan ; 46(9): 677-689, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39275868

RESUMEN

The sex determination in mammals refers to the development of an initial bipotential organ, termed the bipotential gonad/genital ridge, into either a testis or an ovary at the early stages of embryonic development, under the precise regulation of transcription factors. SOX9 (SRY-box transcription factor 9) is a multifunctional transcription factor in mammalian development and plays a critical role in sex determination and subsequent male reproductive organs development. Recent studies have shown that several enhancers upstream of SOX9 also play an important role in the process of sex determination. In this review, we summarize the progress on the role of SOX9 and its gonadal enhancers in sex determination. This review will facilitate to understand the regulatory mechanism of sex determination of SOX9 and provides a theoretical basis for the further development of animal sex manipulation technologies.


Asunto(s)
Mamíferos , Factor de Transcripción SOX9 , Procesos de Determinación del Sexo , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Animales , Procesos de Determinación del Sexo/genética , Humanos , Mamíferos/genética , Masculino , Femenino , Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA