Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 16.686
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 176(4): 897-912.e20, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30686579

RESUMEN

A complete chart of cis-regulatory elements and their dynamic activity is necessary to understand the transcriptional basis of differentiation and function of an organ system. We generated matched epigenome and transcriptome measurements in 86 primary cell types that span the mouse immune system and its differentiation cascades. This breadth of data enable variance components analysis that suggests that genes fall into two distinct classes, controlled by either enhancer- or promoter-driven logic, and multiple regression that connects genes to the enhancers that regulate them. Relating transcription factor (TF) expression to the genome-wide accessibility of their binding motifs classifies them as predominantly openers or closers of local chromatin accessibility, pinpointing specific cis-regulatory elements where binding of given TFs is likely functionally relevant, validated by chromatin immunoprecipitation sequencing (ChIP-seq). Overall, this cis-regulatory atlas provides a trove of information on transcriptional regulation through immune differentiation and a foundational scaffold to define key regulatory events throughout the immunological genome.


Asunto(s)
Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Elementos Reguladores de la Transcripción/genética , Animales , Sitios de Unión/genética , Cromatina , Inmunoprecipitación de Cromatina/métodos , Elementos de Facilitación Genéticos/genética , Epigenómica/métodos , Regulación de la Expresión Génica/genética , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Factores de Transcripción/metabolismo , Transcriptoma/genética
2.
Cell ; 179(4): 923-936.e11, 2019 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-31675499

RESUMEN

Tight junctions are cell-adhesion complexes that seal tissues and are involved in cell polarity and signaling. Supra-molecular assembly and positioning of tight junctions as continuous networks of adhesion strands are dependent on the membrane-associated scaffolding proteins ZO1 and ZO2. To understand how zona occludens (ZO) proteins organize junction assembly, we performed quantitative cell biology and in vitro reconstitution experiments. We discovered that ZO proteins self-organize membrane-attached compartments via phase separation. We identified the multivalent interactions of the conserved PDZ-SH3-GuK supra-domain as the driver of phase separation. These interactions are regulated by phosphorylation and intra-molecular binding. Formation of condensed ZO protein compartments is sufficient to specifically enrich and localize tight-junction proteins, including adhesion receptors, cytoskeletal adapters, and transcription factors. Our results suggest that an active-phase transition of ZO proteins into a condensed membrane-bound compartment drives claudin polymerization and coalescence of a continuous tight-junction belt.


Asunto(s)
Uniones Estrechas/genética , Proteínas de la Zonula Occludens/genética , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-2/genética , Animales , Sitios de Unión/genética , Adhesión Celular/genética , Polaridad Celular/genética , Perros , Células HEK293 , Humanos , Células de Riñón Canino Madin Darby , Proteínas de la Membrana/genética , Dominios PDZ/genética , Fosfoproteínas/genética , Fosforilación/genética , Unión Proteica/genética , Transducción de Señal/genética , Uniones Estrechas/metabolismo , Proteínas de la Zonula Occludens/química , Proteínas de la Zonula Occludens/ultraestructura , Proteína de la Zonula Occludens-1/química , Proteína de la Zonula Occludens-1/ultraestructura , Proteína de la Zonula Occludens-2/química , Proteína de la Zonula Occludens-2/ultraestructura , Dominios Homologos src/genética
3.
Nat Immunol ; 22(12): 1503-1514, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34716452

RESUMEN

Prevention of viral escape and increased coverage against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern require therapeutic monoclonal antibodies (mAbs) targeting multiple sites of vulnerability on the coronavirus spike glycoprotein. Here we identify several potent neutralizing antibodies directed against either the N-terminal domain (NTD) or the receptor-binding domain (RBD) of the spike protein. Administered in combinations, these mAbs provided low-dose protection against SARS-CoV-2 infection in the K18-human angiotensin-converting enzyme 2 mouse model, using both neutralization and Fc effector antibody functions. The RBD mAb WRAIR-2125, which targets residue F486 through a unique heavy-chain and light-chain pairing, demonstrated potent neutralizing activity against all major SARS-CoV-2 variants of concern. In combination with NTD and other RBD mAbs, WRAIR-2125 also prevented viral escape. These data demonstrate that NTD/RBD mAb combinations confer potent protection, likely leveraging complementary mechanisms of viral inactivation and clearance.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , COVID-19/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/metabolismo , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/metabolismo , Sitios de Unión/genética , COVID-19/metabolismo , COVID-19/prevención & control , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Mapeo Epitopo , Epítopos/química , Epítopos/inmunología , Epítopos/metabolismo , Humanos , Ratones Transgénicos , Pruebas de Neutralización , Unión Proteica , Conformación Proteica , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Homología de Secuencia de Aminoácido , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Análisis de Supervivencia
4.
Mol Cell ; 83(14): 2595-2611.e11, 2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37421941

RESUMEN

RNA-binding proteins (RBPs) control RNA metabolism to orchestrate gene expression and, when dysfunctional, underlie human diseases. Proteome-wide discovery efforts predict thousands of RBP candidates, many of which lack canonical RNA-binding domains (RBDs). Here, we present a hybrid ensemble RBP classifier (HydRA), which leverages information from both intermolecular protein interactions and internal protein sequence patterns to predict RNA-binding capacity with unparalleled specificity and sensitivity using support vector machines (SVMs), convolutional neural networks (CNNs), and Transformer-based protein language models. Occlusion mapping by HydRA robustly detects known RBDs and predicts hundreds of uncharacterized RNA-binding associated domains. Enhanced CLIP (eCLIP) for HydRA-predicted RBP candidates reveals transcriptome-wide RNA targets and confirms RNA-binding activity for HydRA-predicted RNA-binding associated domains. HydRA accelerates construction of a comprehensive RBP catalog and expands the diversity of RNA-binding associated domains.


Asunto(s)
Aprendizaje Profundo , Hydra , Animales , Humanos , ARN/metabolismo , Unión Proteica , Sitios de Unión/genética , Hydra/genética , Hydra/metabolismo
5.
Mol Cell ; 82(9): 1708-1723.e10, 2022 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-35320755

RESUMEN

7SK is a conserved noncoding RNA that regulates transcription by sequestering the transcription factor P-TEFb. 7SK function entails complex changes in RNA structure, but characterizing RNA dynamics in cells remains an unsolved challenge. We developed a single-molecule chemical probing strategy, DANCE-MaP (deconvolution and annotation of ribonucleic conformational ensembles), that defines per-nucleotide reactivity, direct base pairing interactions, tertiary interactions, and thermodynamic populations for each state in RNA structural ensembles from a single experiment. DANCE-MaP reveals that 7SK RNA encodes a large-scale structural switch that couples dissolution of the P-TEFb binding site to structural remodeling at distal release factor binding sites. The 7SK structural equilibrium shifts in response to cell growth and stress and can be targeted to modulate expression of P-TEFbresponsive genes. Our study reveals that RNA structural dynamics underlie 7SK function as an integrator of diverse cellular signals to control transcription and establishes the power of DANCE-MaP to define RNA dynamics in cells.


Asunto(s)
Factor B de Elongación Transcripcional Positiva , Proteínas de Unión al ARN , Sitios de Unión/genética , Células HeLa , Humanos , Factor B de Elongación Transcripcional Positiva/genética , ARN Nuclear Pequeño/genética , ARN no Traducido , Proteínas de Unión al ARN/genética
6.
Nature ; 615(7953): 742-749, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36922591

RESUMEN

Our sense of smell enables us to navigate a vast space of chemically diverse odour molecules. This task is accomplished by the combinatorial activation of approximately 400 odorant G protein-coupled receptors encoded in the human genome1-3. How odorants are recognized by odorant receptors remains unclear. Here we provide mechanistic insight into how an odorant binds to a human odorant receptor. Using cryo-electron microscopy, we determined the structure of the active human odorant receptor OR51E2 bound to the fatty acid propionate. Propionate is bound within an occluded pocket in OR51E2 and makes specific contacts critical to receptor activation. Mutation of the odorant-binding pocket in OR51E2 alters the recognition spectrum for fatty acids of varying chain length, suggesting that odorant selectivity is controlled by tight packing interactions between an odorant and an odorant receptor. Molecular dynamics simulations demonstrate that propionate-induced conformational changes in extracellular loop 3 activate OR51E2. Together, our studies provide a high-resolution view of chemical recognition of an odorant by a vertebrate odorant receptor, providing insight into how this large family of G protein-coupled receptors enables our olfactory sense.


Asunto(s)
Microscopía por Crioelectrón , Odorantes , Propionatos , Receptores Odorantes , Humanos , Odorantes/análisis , Propionatos/química , Propionatos/metabolismo , Receptores Odorantes/química , Receptores Odorantes/genética , Receptores Odorantes/metabolismo , Receptores Odorantes/ultraestructura , Olfato/fisiología , Simulación de Dinámica Molecular , Mutación , Sitios de Unión/genética , Especificidad por Sustrato/genética
7.
Nature ; 615(7954): 913-919, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36922589

RESUMEN

Chromatin-binding proteins are critical regulators of cell state in haematopoiesis1,2. Acute leukaemias driven by rearrangement of the mixed lineage leukaemia 1 gene (KMT2Ar) or mutation of the nucleophosmin gene (NPM1) require the chromatin adapter protein menin, encoded by the MEN1 gene, to sustain aberrant leukaemogenic gene expression programs3-5. In a phase 1 first-in-human clinical trial, the menin inhibitor revumenib, which is designed to disrupt the menin-MLL1 interaction, induced clinical responses in patients with leukaemia with KMT2Ar or mutated NPM1 (ref. 6). Here we identified somatic mutations in MEN1 at the revumenib-menin interface in patients with acquired resistance to menin inhibition. Consistent with the genetic data in patients, inhibitor-menin interface mutations represent a conserved mechanism of therapeutic resistance in xenograft models and in an unbiased base-editor screen. These mutants attenuate drug-target binding by generating structural perturbations that impact small-molecule binding but not the interaction with the natural ligand MLL1, and prevent inhibitor-induced eviction of menin and MLL1 from chromatin. To our knowledge, this study is the first to demonstrate that a chromatin-targeting therapeutic drug exerts sufficient selection pressure in patients to drive the evolution of escape mutants that lead to sustained chromatin occupancy, suggesting a common mechanism of therapeutic resistance.


Asunto(s)
Resistencia a Antineoplásicos , Leucemia , Mutación , Proteínas Proto-Oncogénicas , Animales , Humanos , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Sitios de Unión/efectos de los fármacos , Sitios de Unión/genética , Cromatina/genética , Cromatina/metabolismo , Resistencia a Antineoplásicos/genética , Leucemia/tratamiento farmacológico , Leucemia/genética , Leucemia/metabolismo , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo
8.
Nature ; 622(7982): 376-382, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37696289

RESUMEN

Nirmatrelvir is a specific antiviral drug that targets the main protease (Mpro) of SARS-CoV-2 and has been approved to treat COVID-191,2. As an RNA virus characterized by high mutation rates, whether SARS-CoV-2 will develop resistance to nirmatrelvir is a question of concern. Our previous studies have shown that several mutational pathways confer resistance to nirmatrelvir, but some result in a loss of viral replicative fitness, which is then compensated for by additional alterations3. The molecular mechanisms for this observed resistance are unknown. Here we combined biochemical and structural methods to demonstrate that alterations at the substrate-binding pocket of Mpro can allow SARS-CoV-2 to develop resistance to nirmatrelvir in two distinct ways. Comprehensive studies of the structures of 14 Mpro mutants in complex with drugs or substrate revealed that alterations at the S1 and S4 subsites substantially decreased the level of inhibitor binding, whereas alterations at the S2 and S4' subsites unexpectedly increased protease activity. Both mechanisms contributed to nirmatrelvir resistance, with the latter compensating for the loss in enzymatic activity of the former, which in turn accounted for the restoration of viral replicative fitness, as observed previously3. Such a profile was also observed for ensitrelvir, another clinically relevant Mpro inhibitor. These results shed light on the mechanisms by which SARS-CoV-2 evolves to develop resistance to the current generation of protease inhibitors and provide the basis for the design of next-generation Mpro inhibitors.


Asunto(s)
Antivirales , Farmacorresistencia Viral , SARS-CoV-2 , Humanos , Antivirales/química , Antivirales/metabolismo , Antivirales/farmacología , COVID-19/virología , Lactamas , Leucina , Nitrilos , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/enzimología , SARS-CoV-2/genética , SARS-CoV-2/crecimiento & desarrollo , Farmacorresistencia Viral/efectos de los fármacos , Farmacorresistencia Viral/genética , Sitios de Unión/efectos de los fármacos , Sitios de Unión/genética , Mutación , Especificidad por Sustrato , Proteasas 3C de Coronavirus/antagonistas & inhibidores , Proteasas 3C de Coronavirus/genética , Proteasas 3C de Coronavirus/metabolismo , Replicación Viral/efectos de los fármacos , Diseño de Fármacos , Prolina
9.
Nature ; 622(7981): 180-187, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37648864

RESUMEN

Antibiotic binding sites are located in important domains of essential enzymes and have been extensively studied in the context of resistance mutations; however, their study is limited by positive selection. Using multiplex genome engineering1 to overcome this constraint, we generate and characterize a collection of 760 single-residue mutants encompassing the entire rifampicin binding site of Escherichia coli RNA polymerase (RNAP). By genetically mapping drug-enzyme interactions, we identify an alpha helix where mutations considerably enhance or disrupt rifampicin binding. We find mutations in this region that prolong antibiotic binding, converting rifampicin from a bacteriostatic to bactericidal drug by inducing lethal DNA breaks. The latter are replication dependent, indicating that rifampicin kills by causing detrimental transcription-replication conflicts at promoters. We also identify additional binding site mutations that greatly increase the speed of RNAP.Fast RNAP depletes the cell of nucleotides, alters cell sensitivity to different antibiotics and provides a cold growth advantage. Finally, by mapping natural rpoB sequence diversity, we discover that functional rifampicin binding site mutations that alter RNAP properties or confer drug resistance occur frequently in nature.


Asunto(s)
Antibacterianos , Sitios de Unión , ARN Polimerasas Dirigidas por ADN , Escherichia coli , Mutación , Rifampin , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacología , Sitios de Unión/efectos de los fármacos , Sitios de Unión/genética , Roturas del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , ARN Polimerasas Dirigidas por ADN/antagonistas & inhibidores , ARN Polimerasas Dirigidas por ADN/química , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Farmacorresistencia Bacteriana/genética , Escherichia coli/efectos de los fármacos , Escherichia coli/enzimología , Escherichia coli/genética , Nucleótidos/deficiencia , Nucleótidos/metabolismo , Regiones Promotoras Genéticas , Rifampin/química , Rifampin/metabolismo , Rifampin/farmacología , Factores de Tiempo , Transcripción Genética/efectos de los fármacos
10.
Mol Cell ; 81(16): 3422-3439.e11, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34320405

RESUMEN

Maturation of canonical microRNA (miRNA) is initiated by DROSHA that cleaves the primary transcript (pri-miRNA). More than 1,800 miRNA loci are annotated in humans, but it remains largely unknown whether and at which sites pri-miRNAs are cleaved by DROSHA. Here, we performed in vitro processing on a full set of human pri-miRNAs (miRBase version 21) followed by sequencing. This comprehensive profiling enabled us to classify miRNAs on the basis of DROSHA dependence and map their cleavage sites with respective processing efficiency measures. Only 758 pri-miRNAs are confidently processed by DROSHA, while the majority may be non-canonical or false entries. Analyses of the DROSHA-dependent pri-miRNAs show key cis-elements for processing. We observe widespread alternative processing and unproductive cleavage events such as "nick" or "inverse" processing. SRSF3 is a broad-acting auxiliary factor modulating alternative processing and suppressing unproductive processing. The profiling data and methods developed in this study will allow systematic analyses of miRNA regulation.


Asunto(s)
MicroARNs/genética , Procesamiento Postranscripcional del ARN/genética , Ribonucleasa III/genética , Factores de Empalme Serina-Arginina/genética , Sitios de Unión/genética , Genoma Humano/genética , Células HEK293 , Humanos , Interferencia de ARN
11.
Mol Cell ; 81(8): 1651-1665.e4, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33705711

RESUMEN

Enhancers harbor binding motifs that recruit transcription factors (TFs) for gene activation. While cooperative binding of TFs at enhancers is known to be critical for transcriptional activation of a handful of developmental enhancers, the extent of TF cooperativity genome-wide is unknown. Here, we couple high-resolution nuclease footprinting with single-molecule methylation profiling to characterize TF cooperativity at active enhancers in the Drosophila genome. Enrichment of short micrococcal nuclease (MNase)-protected DNA segments indicates that the majority of enhancers harbor two or more TF-binding sites, and we uncover protected fragments that correspond to co-bound sites in thousands of enhancers. From the analysis of co-binding, we find that cooperativity dominates TF binding in vivo at the majority of active enhancers. Cooperativity is highest between sites spaced 50 bp apart, indicating that cooperativity occurs without apparent protein-protein interactions. Our findings suggest nucleosomes promoting cooperativity because co-binding may effectively clear nucleosomes and promote enhancer function.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Unión Proteica/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Sitios de Unión/genética , Línea Celular , Drosophila/genética , Drosophila/metabolismo , Genoma/genética , Nucleasa Microcócica/genética , Nucleosomas/genética , Nucleosomas/metabolismo , Mapas de Interacción de Proteínas/genética , Activación Transcripcional/genética
12.
Genes Dev ; 35(21-22): 1401-1402, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34725128

RESUMEN

In this issue of Genes & Development, Amândio and colleagues (pp. 1490-1509) dissect the function of a cluster of several CTCF binding sites in the HoxD cluster by iterative deletions in mice. They found additive functions for some, and intriguingly found that some sites function as insulators, while others function as anchors for enhancer-promoter interactions. These functions vary depending on developmental context. The work provides new insights into the roles played by CTCF in regulating developmental patterns and 3D chromatin organization.


Asunto(s)
Cromatina , Elementos de Facilitación Genéticos , Animales , Sitios de Unión/genética , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Cromatina/genética , Elementos de Facilitación Genéticos/genética , Proteínas de Homeodominio , Ratones , Regiones Promotoras Genéticas/genética
13.
Genes Dev ; 35(9-10): 771-781, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33832988

RESUMEN

MicroRNAs (miRNAs) are short, noncoding RNAs that associate with Argonaute (AGO) to influence mRNA stability and translation, thereby regulating cellular determination and phenotype. While several individual miRNAs have been shown to control adipocyte function, including energy storage in white fat and energy dissipation in brown fat, a comprehensive analysis of miRNA activity in these tissues has not been performed. We used high-throughput sequencing of RNA isolated by cross-linking immunoprecipitation (HITS-CLIP) to comprehensively characterize the network of high-confidence, in vivo mRNA:miRNA interactions across white and brown fat, revealing >20,000 unique AGO binding sites. When coupled with miRNA and mRNA sequencing, we found an inverse correlation between depot-enriched miRNAs and their targets. To illustrate the functionality of our HITS-CLIP data set in identifying specific miRNA:mRNA interactions, we show that miR-29 is a novel regulator of leptin, an adipocyte-derived hormone that coordinates food intake and energy homeostasis. Two independent miR-29 binding sites in the leptin 3' UTR were validated using luciferase assays, and miR-29 gain and loss of function modulated leptin mRNA and protein secretion in primary adipocytes. This work represents the only experimentally generated miRNA targetome in adipose tissue and identifies multiple regulatory pathways that may specify the unique identities of white and brown fat.


Asunto(s)
Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Proteínas Argonautas/metabolismo , Secuenciación de Inmunoprecipitación de Cromatina , Regulación de la Expresión Génica , MicroARNs/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Animales , Sitios de Unión/genética , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo
14.
Genes Dev ; 35(5-6): 335-353, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33602870

RESUMEN

mSWI/SNF or BAF chromatin regulatory complexes are dosage-sensitive regulators of human neural development frequently mutated in autism spectrum disorders and intellectual disability. Cell cycle exit and differentiation of neural stem/progenitor cells is accompanied by BAF subunit switching to generate neuron-specific nBAF complexes. We manipulated the timing of BAF subunit exchange in vivo and found that early loss of the npBAF subunit BAF53a stalls the cell cycle to disrupt neurogenesis. Loss of BAF53a results in decreased chromatin accessibility at specific neural transcription factor binding sites, including the pioneer factors SOX2 and ASCL1, due to Polycomb accumulation. This results in repression of cell cycle genes, thereby blocking cell cycle progression and differentiation. Cell cycle block upon Baf53a deletion could be rescued by premature expression of the nBAF subunit BAF53b but not by other major drivers of proliferation or differentiation. WNT, EGF, bFGF, SOX2, c-MYC, or PAX6 all fail to maintain proliferation in the absence of BAF53a, highlighting a novel mechanism underlying neural progenitor cell cycle exit in the continued presence of extrinsic proliferative cues.


Asunto(s)
Actinas/metabolismo , Ciclo Celular/genética , Corteza Cerebelosa/embriología , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Actinas/genética , Animales , Sitios de Unión/genética , Células Cultivadas , Cromatina/genética , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/genética , Embrión de Mamíferos , Eliminación de Gen , Genes cdc/genética , Ratones , Neurogénesis/genética , Proteínas del Grupo Polycomb/metabolismo , Factores de Transcripción/metabolismo
15.
Nat Immunol ; 17(10): 1216-1225, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27500631

RESUMEN

The epigenetic processes that regulate antibody-secreting plasma cells are not well understood. Here, analysis of plasma cell differentiation revealed DNA hypomethylation of 10% of CpG loci that were overrepresented at enhancers. Inhibition of DNA methylation enhanced plasma cell commitment in a cell-division-dependent manner. Analysis of B cells differentiating in vivo stratified by cell division revealed a fivefold increase in mRNA transcription coupled to DNA hypomethylation. Demethylation occurred first at binding motifs for the transcription factors NF-κB and AP-1 and later at those for the transcription factors IRF and Oct-2 and was coincident with activation and differentiation gene-expression programs in a cell-division-dependent manner. These data provide mechanistic insight into cell-division-coupled transcriptional and epigenetic reprogramming and suggest that DNA hypomethylation reflects the cis-regulatory history of plasma cell differentiation.


Asunto(s)
Linfocitos B/fisiología , Metilación de ADN , FN-kappa B/metabolismo , Células Plasmáticas/fisiología , Factor de Transcripción AP-1/metabolismo , Animales , Sitios de Unión/genética , Diferenciación Celular/genética , División Celular/genética , Células Cultivadas , Islas de CpG/genética , Epigénesis Genética , Femenino , Regulación de la Expresión Génica , Inmunidad Humoral/genética , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , Factor 2 de Transcripción de Unión a Octámeros/genética , Factor 2 de Transcripción de Unión a Octámeros/metabolismo , Factor de Transcripción AP-1/genética
16.
Nat Rev Genet ; 23(12): 728-740, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35831531

RESUMEN

Cell type-specific gene expression relies on transcription factors (TFs) binding DNA sequence motifs embedded in chromatin. Understanding how motifs are accessed in chromatin is crucial to comprehend differential transcriptional responses and the phenotypic impact of sequence variation. Chromatin obstacles to TF binding range from DNA methylation to restriction of DNA access by nucleosomes depending on their position, composition and modification. In vivo and in vitro approaches now enable the study of TF binding in chromatin at unprecedented resolution. Emerging insights suggest that TFs vary in their ability to navigate chromatin states. However, it remains challenging to link binding and transcriptional outcomes to molecular characteristics of TFs or the local chromatin substrate. Here, we discuss our current understanding of how TFs access DNA in chromatin and novel techniques and directions towards a better understanding of this critical step in genome regulation.


Asunto(s)
Cromatina , Factores de Transcripción , Cromatina/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Sitios de Unión/genética , Nucleosomas/genética , Unión Proteica , ADN/genética
17.
Mol Cell ; 79(6): 1024-1036.e5, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32871103

RESUMEN

Bacterial ribosomal RNAs are synthesized by a dedicated, conserved transcription-elongation complex that transcribes at high rates, shields RNA polymerase from premature termination, and supports co-transcriptional RNA folding, modification, processing, and ribosomal subunit assembly by presently unknown mechanisms. We have determined cryo-electron microscopy structures of complete Escherichia coli ribosomal RNA transcription elongation complexes, comprising RNA polymerase; DNA; RNA bearing an N-utilization-site-like anti-termination element; Nus factors A, B, E, and G; inositol mono-phosphatase SuhB; and ribosomal protein S4. Our structures and structure-informed functional analyses show that fast transcription and anti-termination involve suppression of NusA-stabilized pausing, enhancement of NusG-mediated anti-backtracking, sequestration of the NusG C-terminal domain from termination factor ρ, and the ρ blockade. Strikingly, the factors form a composite RNA chaperone around the RNA polymerase RNA-exit tunnel, which supports co-transcriptional RNA folding and annealing of distal RNA regions. Our work reveals a polymerase/chaperone machine required for biosynthesis of functional ribosomes.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/genética , Chaperonas Moleculares/genética , Proteínas Ribosómicas/genética , Ribosomas/genética , Sitios de Unión/genética , Microscopía por Crioelectrón , Escherichia coli/genética , Escherichia coli/ultraestructura , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/ultraestructura , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/ultraestructura , Biosíntesis de Proteínas/genética , Pliegue del ARN/genética , ARN Ribosómico/genética , ARN Ribosómico/ultraestructura , Proteínas Ribosómicas/ultraestructura , Ribosomas/ultraestructura , Factores de Elongación Transcripcional/química , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/ultraestructura
18.
Genes Dev ; 34(17-18): 1227-1238, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32820039

RESUMEN

Identifying miRNA target genes is difficult, and delineating which targets are the most biologically important is even more difficult. We devised a novel strategy to test the phenotypic impact of individual microRNA-target interactions by disrupting each predicted miRNA-binding site by CRISPR-Cas9 genome editing in C. elegans We developed a multiplexed negative selection screening approach in which edited loci are deep sequenced, and candidate sites are prioritized based on apparent selection pressure against mutations that disrupt miRNA binding. Importantly, our screen was conducted in vivo on mutant animals, allowing us to interrogate organism-level phenotypes. We used this approach to screen for phenotypic targets of the essential mir-35-42 family. By generating 1130 novel 3'UTR alleles across all predicted targets, we identified egl-1 as a phenotypic target whose derepression partially phenocopies the mir-35-42 mutant phenotype by inducing embryonic lethality and low fecundity. These phenotypes can be rescued by compensatory CRISPR mutations that retarget mir-35 to the mutant egl-1 3'UTR. This study demonstrates that the application of in vivo whole organismal CRISPR screening has great potential to accelerate the discovery of phenotypic negative regulatory elements in the noncoding genome.


Asunto(s)
Caenorhabditis elegans/genética , MicroARNs/metabolismo , Regiones no Traducidas 3'/genética , Alelos , Animales , Sitios de Unión/genética , Sistemas CRISPR-Cas , Edición Génica , Pruebas Genéticas , MicroARNs/genética , Mutación , Fenotipo
19.
Genes Dev ; 34(7-8): 560-579, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32139421

RESUMEN

Mutations in the nuclear structural protein lamin A produce rare, tissue-specific diseases called laminopathies. The introduction of a human Emery-Dreifuss muscular dystrophy (EDMD)-inducing mutation into the C. elegans lamin (LMN-Y59C), recapitulates many muscular dystrophy phenotypes, and correlates with hyper-sequestration of a heterochromatic array at the nuclear periphery in muscle cells. Using muscle-specific emerin Dam-ID in worms, we monitored the effects of the mutation on endogenous chromatin. An increased contact with the nuclear periphery along chromosome arms, and an enhanced release of chromosomal centers, coincided with the disease phenotypes of reduced locomotion and compromised sarcomere integrity. The coupling of the LMN-Y59C mutation with the ablation of CEC-4, a chromodomain protein that anchors H3K9-methylated chromatin at the nuclear envelope (NE), suppressed the muscle-associated disease phenotypes. Deletion of cec-4 also rescued LMN-Y59C-linked alterations in chromatin organization and some changes in transcription. Sequences that changed position in the LMN-Y59C mutant, are enriched for E2F (EFL-2)-binding sites, consistent with previous studies suggesting that altered Rb-E2F interaction with lamin A may contribute to muscle dysfunction. In summary, we were able to counteract the dominant muscle-specific defects provoked by LMNA mutation by the ablation of a lamin-associated H3K9me anchor, suggesting a novel therapeutic pathway for EDMD.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Núcleo Celular/genética , Proteínas Cromosómicas no Histona/genética , Eliminación de Gen , Distrofia Muscular de Emery-Dreifuss/genética , Animales , Sitios de Unión/genética , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Núcleo Celular/patología , Cromatina/genética , Modelos Animales de Enfermedad , Genoma de los Helmintos/genética , Laminina/genética , Laminina/metabolismo , Músculos/fisiopatología , Distrofia Muscular de Emery-Dreifuss/fisiopatología , Mutación , Estructura Terciaria de Proteína/genética , Sarcómeros/química , Sarcómeros/genética , Transcripción Genética/genética
20.
Nat Methods ; 21(2): 247-258, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38200227

RESUMEN

RNA-binding proteins (RBPs) regulate diverse cellular processes by dynamically interacting with RNA targets. However, effective methods to capture both stable and transient interactions between RBPs and their RNA targets are still lacking, especially when the interaction is dynamic or samples are limited. Here we present an assay of reverse transcription-based RBP binding site sequencing (ARTR-seq), which relies on in situ reverse transcription of RBP-bound RNAs guided by antibodies to identify RBP binding sites. ARTR-seq avoids ultraviolet crosslinking and immunoprecipitation, allowing for efficient and specific identification of RBP binding sites from as few as 20 cells or a tissue section. Taking advantage of rapid formaldehyde fixation, ARTR-seq enables capturing the dynamic RNA binding by RBPs over a short period of time, as demonstrated by the profiling of dynamic RNA binding of G3BP1 during stress granule assembly on a timescale as short as 10 minutes.


Asunto(s)
ARN , Transcripción Reversa , ARN/genética , ARN/metabolismo , ADN Helicasas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , ARN Helicasas/genética , ARN Helicasas/metabolismo , Proteínas con Motivos de Reconocimiento de ARN/genética , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Sitios de Unión/genética , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA