Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 876
Filtrar
1.
Nature ; 613(7942): 96-102, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36517591

RESUMEN

Expansion of a single repetitive DNA sequence, termed a tandem repeat (TR), is known to cause more than 50 diseases1,2. However, repeat expansions are often not explored beyond neurological and neurodegenerative disorders. In some cancers, mutations accumulate in short tracts of TRs, a phenomenon termed microsatellite instability; however, larger repeat expansions have not been systematically analysed in cancer3-8. Here we identified TR expansions in 2,622 cancer genomes spanning 29 cancer types. In seven cancer types, we found 160 recurrent repeat expansions (rREs), most of which (155/160) were subtype specific. We found that rREs were non-uniformly distributed in the genome with enrichment near candidate cis-regulatory elements, suggesting a potential role in gene regulation. One rRE, a GAAA-repeat expansion, located near a regulatory element in the first intron of UGT2B7 was detected in 34% of renal cell carcinoma samples and was validated by long-read DNA sequencing. Moreover, in preliminary experiments, treating cells that harbour this rRE with a GAAA-targeting molecule led to a dose-dependent decrease in cell proliferation. Overall, our results suggest that rREs may be an important but unexplored source of genetic variation in human cancer, and we provide a comprehensive catalogue for further study.


Asunto(s)
Expansión de las Repeticiones de ADN , Genoma Humano , Neoplasias , Humanos , Secuencia de Bases , Expansión de las Repeticiones de ADN/genética , Genoma Humano/genética , Neoplasias/clasificación , Neoplasias/genética , Neoplasias/patología , Análisis de Secuencia de ADN , Regulación de la Expresión Génica , Elementos Reguladores de la Transcripción/genética , Intrones/genética , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Proliferación Celular/efectos de los fármacos , Reproducibilidad de los Resultados
2.
BMC Genomics ; 23(1): 714, 2022 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-36261804

RESUMEN

BACKGROUND: Mouse is probably the most important model organism to study mammal biology and human diseases. A better understanding of the mouse genome will help understand the human genome, biology and diseases. However, despite the recent progress, the characterization of the regulatory sequences in the mouse genome is still far from complete, limiting its use to understand the regulatory sequences in the human genome. RESULTS: Here, by integrating binding peaks in ~ 9,000 transcription factor (TF) ChIP-seq datasets that cover 79.9% of the mouse mappable genome using an efficient pipeline, we were able to partition these binding peak-covered genome regions into a cis-regulatory module (CRM) candidate (CRMC) set and a non-CRMC set. The CRMCs contain 912,197 putative CRMs and 38,554,729 TF binding sites (TFBSs) islands, covering 55.5% and 24.4% of the mappable genome, respectively. The CRMCs tend to be under strong evolutionary constraints, indicating that they are likely cis-regulatory; while the non-CRMCs are largely selectively neutral, indicating that they are unlikely cis-regulatory. Based on evolutionary profiles of the genome positions, we further estimated that 63.8% and 27.4% of the mouse genome might code for CRMs and TFBSs, respectively. CONCLUSIONS: Validation using experimental data suggests that at least most of the CRMCs are authentic. Thus, this unprecedentedly comprehensive map of CRMs and TFBSs can be a good resource to guide experimental studies of regulatory genomes in mice and humans.


Asunto(s)
Genoma Humano , Elementos Reguladores de la Transcripción , Humanos , Ratones , Animales , Elementos Reguladores de la Transcripción/genética , Sitios de Unión/genética , Unión Proteica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Mamíferos/genética
3.
Database (Oxford) ; 20222022 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-35452518

RESUMEN

More accurate and more complete predictions of cis-regulatory modules (CRMs) and constituent transcription factor (TF) binding sites (TFBSs) in genomes can facilitate characterizing functions of regulatory sequences. Here, we developed a database predicted cis-regulatory modules (PCRMS) (https://cci-bioinfo.uncc.edu) that stores highly accurate and unprecedentedly complete maps of predicted CRMs and TFBSs in the human and mouse genomes. The web interface allows the user to browse CRMs and TFBSs in an organism, find the closest CRMs to a gene, search CRMs around a gene and find all TFBSs of a TF. PCRMS can be a useful resource for the research community to characterize regulatory genomes. Database URL: https://cci-bioinfo.uncc.edu/.


Asunto(s)
Elementos Reguladores de la Transcripción , Factores de Transcripción , Animales , Sitios de Unión , Genoma/genética , Ratones , Unión Proteica , Elementos Reguladores de la Transcripción/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
Int J Mol Sci ; 22(21)2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34768856

RESUMEN

Endometriosis is a chronic gynecological disorder affecting the quality of life and fertility of many women around the world. Heterogeneous and non-specific symptoms may lead to a delay in diagnosis, with treatment options limited to surgery and hormonal therapy. Hence, there is a need to better understand the pathogenesis of the disease to improve diagnosis and treatment. Long non-coding RNAs (lncRNAs) have been increasingly shown to be involved in gene regulation but remain relatively under investigated in endometriosis. Mutational and transcriptomic studies have implicated lncRNAs in the pathogenesis of endometriosis. Single-nucleotide polymorphisms (SNPs) in lncRNAs or their regulatory regions have been associated with endometriosis. Genome-wide transcriptomic studies have identified lncRNAs that show deregulated expression in endometriosis, some of which have been subjected to further experiments, which support a role in endometriosis. Mechanistic studies indicate that lncRNAs may regulate genes involved in endometriosis by acting as a molecular sponge for miRNAs, by directly targeting regulatory elements via interactions with chromatin or transcription factors or by affecting signaling pathways. Future studies should concentrate on determining the role of uncharacterized lncRNAs revealed by endometriosis transcriptome studies and the relevance of lncRNAs implicated in the disease by in vitro and animal model studies.


Asunto(s)
Endometriosis/genética , Regulación de la Expresión Génica/genética , ARN Largo no Codificante/genética , Elementos Reguladores de la Transcripción/genética , Cromatina/genética , Endometriosis/patología , Femenino , Perfilación de la Expresión Génica , Humanos , Polimorfismo de Nucleótido Simple/genética , Transducción de Señal/genética , Transcriptoma/genética
5.
Nat Struct Mol Biol ; 28(12): 989-996, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34811519

RESUMEN

The SAGA complex is a regulatory hub involved in gene regulation, chromatin modification, DNA damage repair and signaling. While structures of yeast SAGA (ySAGA) have been reported, there are noteworthy functional and compositional differences for this complex in metazoans. Here we present the cryogenic-electron microscopy (cryo-EM) structure of human SAGA (hSAGA) and show how the arrangement of distinct structural elements results in a globally divergent organization from that of yeast, with a different interface tethering the core module to the TRRAP subunit, resulting in a dramatically altered geometry of functional elements and with the integration of a metazoan-specific splicing module. Our hSAGA structure reveals the presence of an inositol hexakisphosphate (InsP6) binding site in TRRAP and an unusual property of its pseudo-(Ψ)PIKK. Finally, we map human disease mutations, thus providing the needed framework for structure-guided drug design of this important therapeutic target for human developmental diseases and cancer.


Asunto(s)
Regulación de la Expresión Génica/genética , Histona Acetiltransferasas/metabolismo , Elementos Reguladores de la Transcripción/genética , Transcripción Genética/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Sitios de Unión , Línea Celular Tumoral , Cromatina/metabolismo , Microscopía por Crioelectrón , Células HeLa , Humanos , Proteínas Nucleares/metabolismo , Ácido Fítico/metabolismo , Regiones Promotoras Genéticas/genética , Conformación Proteica , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomycetales
6.
Nat Commun ; 12(1): 6749, 2021 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-34799566

RESUMEN

The hypothalamus regulates metabolic homeostasis by influencing behavior and endocrine systems. Given its role governing key traits, such as body weight and reproductive timing, understanding the genetic regulation of hypothalamic development and function could yield insights into disease pathogenesis. However, given its inaccessibility, studying human hypothalamic gene regulation has proven challenging. To address this gap, we generate a high-resolution chromatin architecture atlas of an established embryonic stem cell derived hypothalamic-like neuron model across three stages of in vitro differentiation. We profile accessible chromatin and identify physical contacts between gene promoters and putative cis-regulatory elements to characterize global regulatory landscape changes during hypothalamic differentiation. Next, we integrate these data with GWAS loci for various complex traits, identifying multiple candidate effector genes. Our results reveal common target genes for these traits, potentially affecting core developmental pathways. Our atlas will enable future efforts to determine hypothalamic mechanisms influencing disease susceptibility.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Células Madre Embrionarias Humanas/fisiología , Hipotálamo/embriología , Neuronas/fisiología , Diferenciación Celular/genética , Línea Celular , Mapeo Cromosómico , Estudio de Asociación del Genoma Completo , Humanos , Hipotálamo/citología , Herencia Multifactorial , RNA-Seq , Elementos Reguladores de la Transcripción/genética
7.
Mol Cell ; 81(16): 3368-3385.e9, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34375583

RESUMEN

The mechanistic understanding of nascent RNAs in transcriptional control remains limited. Here, by a high sensitivity method methylation-inscribed nascent transcripts sequencing (MINT-seq), we characterized the landscapes of N6-methyladenosine (m6A) on nascent RNAs. We uncover heavy but selective m6A deposition on nascent RNAs produced by transcription regulatory elements, including promoter upstream antisense RNAs and enhancer RNAs (eRNAs), which positively correlates with their length, inclusion of m6A motif, and RNA abundances. m6A-eRNAs mark highly active enhancers, where they recruit nuclear m6A reader YTHDC1 to phase separate into liquid-like condensates, in a manner dependent on its C terminus intrinsically disordered region and arginine residues. The m6A-eRNA/YTHDC1 condensate co-mixes with and facilitates the formation of BRD4 coactivator condensate. Consequently, YTHDC1 depletion diminished BRD4 condensate and its recruitment to enhancers, resulting in inhibited enhancer and gene activation. We propose that chemical modifications of eRNAs together with reader proteins play broad roles in enhancer activation and gene transcriptional control.


Asunto(s)
Adenosina/análogos & derivados , Proteínas de Ciclo Celular/genética , Proteínas del Tejido Nervioso/genética , Factores de Empalme de ARN/genética , ARN/genética , Factores de Transcripción/genética , Adenosina/genética , Elementos de Facilitación Genéticos/genética , Regulación de la Expresión Génica/genética , Humanos , Metilación , Elementos Reguladores de la Transcripción/genética , Activación Transcripcional/genética
8.
Nat Commun ; 12(1): 4897, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34385432

RESUMEN

Precise control of mammalian gene expression is facilitated through epigenetic mechanisms and nuclear organization. In particular, insulated chromosome structures are important for regulatory control, but the phenotypic consequences of their boundary disruption on developmental processes are complex and remain insufficiently understood. Here, we generated deeply sequenced Hi-C data for human pluripotent stem cells (hPSCs) that allowed us to identify CTCF loop domains that have highly conserved boundary CTCF sites and show a notable enrichment of individual developmental regulators. Importantly, perturbation of such a boundary in hPSCs interfered with proper differentiation through deregulated distal enhancer-promoter activity. Finally, we found that germline variations affecting such boundaries are subject to purifying selection and are underrepresented in the human population. Taken together, our findings highlight the importance of developmental gene isolation through chromosomal folding structures as a mechanism to ensure their proper expression.


Asunto(s)
Diferenciación Celular/genética , Perfilación de la Expresión Génica/métodos , Genoma Humano/genética , Células Madre Embrionarias Humanas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Elementos Reguladores de la Transcripción/genética , Sitios de Unión/genética , Western Blotting , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Línea Celular , Elementos de Facilitación Genéticos/genética , Células Madre Embrionarias Humanas/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Regiones Promotoras Genéticas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN/métodos
9.
Nat Commun ; 12(1): 3376, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099660

RESUMEN

Phenotypic plasticity is the variation in phenotype that a single genotype can produce in different environments and, as such, is an important component of individual fitness. However, whether the effect of new mutations, and hence evolution, depends on the direction of plasticity remains controversial. Here, we identify the cis-acting modifications that have reshaped gene expression in response to dehydration stress in three Arabidopsis species. Our study shows that the direction of effects of most cis-regulatory variants differentiating the response between A. thaliana and the sister species A. lyrata and A. halleri depends on the direction of pre-existing plasticity in gene expression. A comparison of the rate of cis-acting variant accumulation in each lineage indicates that the selective forces driving adaptive evolution in gene expression favors regulatory changes that magnify the stress response in A. lyrata. The evolutionary constraints measured on the amino-acid sequence of these genes support this interpretation. In contrast, regulatory changes that mitigate the plastic response to stress evolved more frequently in A. halleri. Our results demonstrate that pre-existing plasticity may be a stepping stone for adaptation, but its selective remodeling differs between lineages.


Asunto(s)
Adaptación Fisiológica/genética , Arabidopsis/genética , Evolución Molecular , Regulación de la Expresión Génica de las Plantas , Elementos Reguladores de la Transcripción/genética , Sequías , Genes de Plantas , Fitomejoramiento , Especificidad de la Especie
10.
Commun Biol ; 4(1): 675, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-34083716

RESUMEN

Elucidating transcription mediated by the glucocorticoid receptor (GR) is crucial for understanding the role of glucocorticoids (GCs) in the treatment of diseases. Podocyte is a useful model for studying GR regulation because GCs are the primary medication for podocytopathy. In this study, we integrated data from transcriptome, transcription factor binding, histone modification, and genome topology. Our data reveals that the GR binds and activates selective regulatory elements in podocyte. The 3D interactome captured by HiChIP facilitates the identification of remote targets of GR. We found that GR in podocyte is enriched at transcriptional interaction hubs and super-enhancers. We further demonstrate that the target gene of the top GR-associated super-enhancer is indispensable to the effective functioning of GC in podocyte. Our findings provided insights into the mechanisms underlying the protective effect of GCs on podocyte, and demonstrate the importance of considering transcriptional interactions in order to fine-map regulatory networks of GR.


Asunto(s)
Cromatina/metabolismo , Citoesqueleto/metabolismo , Podocitos/metabolismo , Receptores de Glucocorticoides/metabolismo , Transcripción Genética , Células A549 , Sitios de Unión/genética , Línea Celular , Células Cultivadas , Cromatina/genética , Secuenciación de Inmunoprecipitación de Cromatina/métodos , Glucocorticoides/farmacología , Células HeLa , Humanos , Células K562 , Células MCF-7 , Podocitos/citología , Podocitos/efectos de los fármacos , Unión Proteica , Receptores de Glucocorticoides/genética , Elementos Reguladores de la Transcripción/genética , Transcriptoma/genética
11.
Cell ; 184(11): 3041-3055.e21, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-33964211

RESUMEN

cis-regulatory elements (CREs) encode the genomic blueprints of spatiotemporal gene expression programs enabling highly specialized cell functions. Using single-cell genomics in six maize organs, we determined the cis- and trans-regulatory factors defining diverse cell identities and coordinating chromatin organization by profiling transcription factor (TF) combinatorics, identifying TFs with non-cell-autonomous activity, and uncovering TFs underlying higher-order chromatin interactions. Cell-type-specific CREs were enriched for enhancer activity and within unmethylated long terminal repeat retrotransposons. Moreover, we found cell-type-specific CREs are hotspots for phenotype-associated genetic variants and were targeted by selection during modern maize breeding, highlighting the biological implications of this CRE atlas. Through comparison of maize and Arabidopsis thaliana developmental trajectories, we identified TFs and CREs with conserved and divergent chromatin dynamics, showcasing extensive evolution of gene regulatory networks. In addition to this rich dataset, we developed single-cell analysis software, Socrates, which can be used to understand cis-regulatory variation in any species.


Asunto(s)
Regulación de la Expresión Génica de las Plantas/genética , Elementos Reguladores de la Transcripción/genética , Zea mays/genética , Arabidopsis/genética , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica de las Plantas/fisiología , Redes Reguladoras de Genes/genética , Genoma , Genómica , Elementos Reguladores de la Transcripción/fisiología , Análisis de la Célula Individual , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma/genética
12.
Cell Death Dis ; 12(6): 544, 2021 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-34039951

RESUMEN

Progesterone resistance can significantly restrict the efficacy of conservative treatment for patients with endometrial cancer who wish to preserve their fertility or those who suffer from advanced and recurrent cancer. SREBP1 is known to be involved in the occurrence and progression of endometrial cancer, although the precise mechanism involved remains unclear. In the present study, we carried out microarray analysis in progesterone-sensitive and progesterone-resistant cell lines and demonstrated that SREBP1 is related to progesterone resistance. Furthermore, we verified that SREBP1 is over-expressed in both drug-resistant tissues and cells. Functional studies further demonstrated that the inhibition of SREBP1 restored the sensitivity of endometrial cancer to progesterone both in vitro and in vivo, and that the over-expression of SREBP1 promoted resistance to progesterone. With regards to the mechanism involved, we found that SREBP1 promoted the proliferation of endometrial cancer cells and inhibited their apoptosis by activating the NF-κB pathway. To solve the problem of clinical application, we found that Fatostatin, an inhibitor of SREBP1, could increase the sensitivity of endometrial cancer to progesterone and reverse progesterone resistance by inhibiting SREBP1 both in vitro and in vivo. Our results highlight the important role of SREBP1 in progesterone resistance and suggest that the use of Fatostatin to target SREBP1 may represent a new method to solve progesterone resistance in patients with endometrial cancer.


Asunto(s)
Neoplasias Endometriales/tratamiento farmacológico , Endometrio/anomalías , FN-kappa B/efectos de los fármacos , Piridinas/uso terapéutico , Elementos Reguladores de la Transcripción/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/efectos de los fármacos , Tiazoles/uso terapéutico , Enfermedades Uterinas/dietoterapia , Femenino , Humanos , Piridinas/farmacología , Tiazoles/farmacología , Transfección
13.
Brain ; 144(10): 3005-3019, 2021 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-33950254

RESUMEN

MECP2 gene transfer has been shown to extend the survival of Mecp2-/y knockout mice modelling Rett syndrome, an X-linked neurodevelopmental disorder. However, controlling deleterious overexpression of MECP2 remains the critical unmet obstacle towards a safe and effective gene therapy approach for Rett syndrome. A recently developed truncated miniMECP2 gene has also been shown to be therapeutic after AAV9-mediated gene transfer in knockout neonates. We show that AAV9/miniMECP2 has a similar dose-dependent toxicity profile to that of a published second-generation AAV9/MECP2 vector after treatment in adolescent mice. To overcome that toxicity, we developed a risk-driven viral genome design strategy rooted in high-throughput profiling and genome mining to rationally develop a compact, synthetic microRNA target panel (miR-responsive auto-regulatory element, 'miRARE') to minimize the possibility of miniMECP2 transgene overexpression in the context of Rett syndrome gene therapy. The goal of miRARE is to have a built-in inhibitory element responsive to MECP2 overexpression. The data provided herein show that insertion of miRARE into the miniMECP2 gene expression cassette greatly improved the safety of miniMECP2 gene transfer without compromising efficacy. Importantly, this built-in regulation system does not require any additional exogenous drug application, and no miRNAs are expressed from the transgene cassette. Although broad applications of miRARE have yet to be determined, the design of miRARE suggests a potential use in gene therapy approaches for other dose-sensitive genes.


Asunto(s)
Terapia Genética/métodos , Proteína 2 de Unión a Metil-CpG/administración & dosificación , MicroARNs/administración & dosificación , Ingeniería de Proteínas/métodos , Elementos Reguladores de la Transcripción , Síndrome de Rett/terapia , Animales , Humanos , Inyecciones Espinales , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Noqueados , MicroARNs/genética , Elementos Reguladores de la Transcripción/genética , Síndrome de Rett/genética
14.
Nat Neurosci ; 24(7): 941-953, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34017130

RESUMEN

Common genetic risk for neuropsychiatric disorders is enriched in regulatory elements active during cortical neurogenesis. However, it remains poorly understood as to how these variants influence gene regulation. To model the functional impact of common genetic variation on the noncoding genome during human cortical development, we performed the assay for transposase accessible chromatin using sequencing (ATAC-seq) and analyzed chromatin accessibility quantitative trait loci (QTL) in cultured human neural progenitor cells and their differentiated neuronal progeny from 87 donors. We identified significant genetic effects on 988/1,839 neuron/progenitor regulatory elements, with highly cell-type and temporally specific effects. A subset (roughly 30%) of chromatin accessibility-QTL were also associated with changes in gene expression. Motif-disrupting alleles of transcriptional activators generally led to decreases in chromatin accessibility, whereas motif-disrupting alleles of repressors led to increases in chromatin accessibility. By integrating cell-type-specific chromatin accessibility-QTL and brain-relevant genome-wide association data, we were able to fine-map and identify regulatory mechanisms underlying noncoding neuropsychiatric disorder risk loci.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Variación Genética/genética , Trastornos Mentales/genética , Neuronas/fisiología , Sitios de Carácter Cuantitativo/genética , Diferenciación Celular/fisiología , Cromatina/genética , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo , Humanos , Células-Madre Neurales/fisiología , Neurogénesis/genética , Elementos Reguladores de la Transcripción/genética , Factores de Transcripción/genética
15.
Mol Biol Cell ; 32(9): 942-955, 2021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-33788585

RESUMEN

The histone locus body (HLB) is an evolutionarily conserved nuclear body that regulates the transcription and processing of replication-dependent (RD) histone mRNAs, which are the only eukaryotic mRNAs lacking a poly-A tail. Many nuclear bodies contain distinct domains, but how internal organization is related to nuclear body function is not fully understood. Here, we demonstrate using structured illumination microscopy that Drosophila HLBs have a "core-shell" organization in which the internal core contains transcriptionally active RD histone genes. The N-terminus of Mxc, which contains a domain required for Mxc oligomerization, HLB assembly, and RD histone gene expression, is enriched in the HLB core. In contrast, the C-terminus of Mxc is enriched in the HLB outer shell as is FLASH, a component of the active U7 snRNP that cotranscriptionally cleaves RD histone pre-mRNA. Consistent with these results, we show biochemically that FLASH binds directly to the Mxc C-terminal region. In the rapid S-M nuclear cycles of syncytial blastoderm Drosophila embryos, the HLB disassembles at mitosis and reassembles the core-shell arrangement as histone gene transcription is activated immediately after mitosis. Thus, the core-shell organization is coupled to zygotic histone gene transcription, revealing a link between HLB internal organization and RD histone gene expression.


Asunto(s)
Estructuras del Núcleo Celular/metabolismo , Histonas/metabolismo , Microscopía/métodos , Animales , Proteínas Portadoras/metabolismo , Núcleo Celular/metabolismo , Estructuras del Núcleo Celular/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Mitosis , Precursores del ARN/metabolismo , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , Elementos Reguladores de la Transcripción/genética , Ribonucleoproteína Nuclear Pequeña U7/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Cigoto/metabolismo
16.
Genes Dev ; 35(7-8): 489-494, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33737384

RESUMEN

While changes in MeCP2 dosage cause Rett syndrome (RTT) and MECP2 duplication syndrome (MDS), its transcriptional regulation is poorly understood. Here, we identified six putative noncoding regulatory elements of Mecp2, two of which are conserved in humans. Upon deletion in mice and human iPSC-derived neurons, these elements altered RNA and protein levels in opposite directions and resulted in a subset of RTT- and MDS-like behavioral deficits in mice. Our discovery provides insight into transcriptional regulation of Mecp2/MECP2 and highlights genomic sites that could serve as diagnostic and therapeutic targets in RTT or MDS.


Asunto(s)
Regulación de la Expresión Génica/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Proteína 2 de Unión a Metil-CpG/genética , Neuronas/patología , Elementos Reguladores de la Transcripción/genética , Síndrome de Rett/genética , Animales , Conducta Animal/fisiología , Secuencia Conservada/genética , Eliminación de Gen , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
17.
PLoS One ; 16(3): e0242949, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33750972

RESUMEN

LEAFY COTYLEDON1 (LEC1) is a HAP3 subunit of CCAAT-binding transcription factor, which controls several aspects of embryo and postembryo development, including embryo morphogenesis, storage reserve accumulation and skotomorphogenesis. Herein, using the method of chromosomal walking, a 2707bp upstream sequence from the ATG initiation codon site of AhLEC1A which is a homolog of Arabidopsis LEC1 was isolated in peanut. Its transcriptional start site confirmed by 5' RACE was located at 82 nt from 5' upstream of ATG. The bioinformatics analysis revealed that there existed many tissue-specific elements and light responsive motifs in its promoter. To identify the functional region of the AhLEC1A promoter, seven plant expression vectors expressing the GUS (ß-glucuronidase) gene, driven by 5' terminal series deleted fragments of AhLEC1A promoter, were constructed and transformed into Arabidopsis. Results of GUS histochemical staining showed that the regulatory region containing 82bp of 5' UTR and 2228bp promoter could facilitate GUS to express preferentially in the embryos at different development periods of Arabidopsis. Taken together, it was inferred that the expression of AhLEC1A during seed development of peanut might be controlled positively by several seed-specific regulatory elements, as well as negatively by some other regulatory elements inhibiting its expression in other organs. Moreover, the GUS expression pattern of transgenic seedlings in darkness and in light was relevant to the light-responsive elements scattered in AhLEC1A promoter segment, implying that these light-responsive elements harbored in the AhLEC1A promoter regulate skotomorphogenesis of peanut seeds, and AhLEC1A expression was inhibited after the germinated seedlings were transferred from darkness to light.


Asunto(s)
Arachis/genética , Factor de Unión a CCAAT/genética , Proteínas de Plantas/genética , Regiones no Traducidas 5' , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Arachis/crecimiento & desarrollo , Factor de Unión a CCAAT/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Regulación de la Expresión Génica de las Plantas/efectos de la radiación , Glucuronidasa/genética , Glucuronidasa/metabolismo , Luz , Proteínas de Plantas/metabolismo , Plantas Modificadas Genéticamente/metabolismo , Regiones Promotoras Genéticas , Subunidades de Proteína/genética , Elementos Reguladores de la Transcripción/genética , Semillas/genética
18.
Nat Commun ; 12(1): 1781, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33741908

RESUMEN

Prostate cancer (PCa) risk-associated SNPs are enriched in noncoding cis-regulatory elements (rCREs), yet their modi operandi and clinical impact remain elusive. Here, we perform CRISPRi screens of 260 rCREs in PCa cell lines. We find that rCREs harboring high risk SNPs are more essential for cell proliferation and H3K27ac occupancy is a strong indicator of essentiality. We also show that cell-line-specific essential rCREs are enriched in the 8q24.21 region, with the rs11986220-containing rCRE regulating MYC and PVT1 expression, cell proliferation and tumorigenesis in a cell-line-specific manner, depending on DNA methylation-orchestrated occupancy of a CTCF binding site in between this rCRE and the MYC promoter. We demonstrate that CTCF deposition at this site as measured by DNA methylation level is highly variable in prostate specimens, and observe the MYC eQTL in the 8q24.21 locus in individuals with low CTCF binding. Together our findings highlight a causal mechanism synergistically driven by a risk SNP and DNA methylation-mediated 3D genome architecture, advocating for the integration of genetics and epigenetics in assessing risks conferred by genetic predispositions.


Asunto(s)
Sistemas CRISPR-Cas , Metilación de ADN , Edición Génica/métodos , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo/métodos , Neoplasias de la Próstata/genética , Animales , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Carcinogénesis/genética , Línea Celular Tumoral , Humanos , Masculino , Ratones Endogámicos NOD , Ratones SCID , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-myc/genética , Sitios de Carácter Cuantitativo/genética , Elementos Reguladores de la Transcripción/genética , Factores de Riesgo
19.
Mol Cell ; 81(8): 1640-1650, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33689750

RESUMEN

Coordinated changes in gene expression allow a single fertilized oocyte to develop into a complex multi-cellular organism. These changes in expression are controlled by transcription factors that gain access to discrete cis-regulatory elements in the genome, allowing them to activate gene expression. Although nucleosomes present barriers to transcription factor occupancy, pioneer transcription factors have unique properties that allow them to bind DNA in the context of nucleosomes, define cis-regulatory elements, and facilitate the subsequent binding of additional factors that determine gene expression. In this capacity, pioneer factors act at the top of gene-regulatory networks to control developmental transitions. Developmental context also influences pioneer factor binding and activity. Here we discuss the interplay between pioneer factors and development, their role in driving developmental transitions, and the influence of the cellular environment on pioneer factor binding and activity.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Animales , ADN/genética , Genoma/genética , Humanos , Nucleosomas/genética , Unión Proteica/genética , Elementos Reguladores de la Transcripción/genética , Factores de Transcripción/genética
20.
Mol Cell ; 81(8): 1601-1616, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33770487

RESUMEN

The influence of genome organization on transcription is central to our understanding of cell type specification. Higher-order genome organization is established through short- and long-range DNA interactions. Coordination of these interactions, from single atoms to entire chromosomes, plays a fundamental role in transcriptional control of gene expression. Loss of this coupling can result in disease. Analysis of transcriptional regulation typically involves disparate experimental approaches, from structural studies that define angstrom-level interactions to cell-biological and genomic approaches that assess mesoscale relationships. Thus, to fully understand the mechanisms that regulate gene expression, it is critical to integrate the findings gained across these distinct size scales. In this review, I illustrate fundamental ways in which cells regulate transcription in the context of genome organization.


Asunto(s)
Emparejamiento Base/genética , Cromosomas/genética , Transcripción Genética/genética , Animales , Regulación de la Expresión Génica/genética , Humanos , Elementos Reguladores de la Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...