Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 656
Filtrar
1.
Sci Rep ; 12(1): 2188, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-35140280

RESUMO

Although many studies have observed genome-wide host transposon expression alteration during viral infection, the mechanisms of induction and the impact on the host remain unclear. Utilizing recently published influenza A virus (IAV) time series data and ENCODE functional genomics data, we characterized virus induced host differentially expressed transposons (virus-induced-TE) by investigating genome-wide spatial and functional relevance between the virus-induced-TEs and epigenomic markers (e.g. histone modification and chromatin remodelers). We found that a significant fraction of virus-induced-TEs are derived from host enhancer regions, where CHD4 binding and/or H3K27ac occupancy is high or H3K9me3 occupancy is low. By overlapping virus-induced-TEs to human enhancer RNAs (eRNAs), we discovered that a proportion of virus-induced-TEs are either eRNAs or part of enhancer RNAs. Upon further analysis of the eRNA targeted genes, we found that the virus-induced-TE related eRNA targets are overrepresented in differentially expressed host genes of IAV infected samples. Our results suggest that changing chromatin accessibility from repressive to permissive in the transposon docked enhancer regions to regulate host downstream gene expression is potentially one of the virus and host cell interaction mechanisms, where transposons are likely important regulatory genomic elements. Our study provides a new insight into the mechanisms of virus-host interaction and may lead to novel strategies for prevention and therapeutics of IAV and other virus infectious diseases.


Assuntos
Elementos de DNA Transponíveis/fisiologia , Elementos Facilitadores Genéticos/fisiologia , Vírus da Influenza A/genética , RNA/fisiologia , Montagem e Desmontagem da Cromatina/fisiologia , Regulação da Expressão Gênica , Interações entre Hospedeiro e Microrganismos/genética , Humanos
2.
Sci Rep ; 12(1): 855, 2022 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-35039581

RESUMO

The proto-oncogene MYC is important for development and cell growth, however, its abnormal regulation causes cancer. Recent studies identified distinct enhancers of MYC in various cancers, but any MYC enhancer(s) in hepatocellular carcinoma (HCC) remain(s) elusive. By analyzing H3K27ac enrichment and enhancer RNA (eRNA) expression in cultured HCC cells, we identified six putative MYC enhancer regions. Amongst these, two highly active enhancers, located ~ 800 kb downstream of the MYC gene, were identified by qRT-PCR and reporter assays. We functionally confirmed these enhancers by demonstrating a significantly reduced MYC expression and cell proliferation upon CRISPR/Cas9-based deletion and/or antisense oligonucleotide (ASO)-mediated inhibition. In conclusion, we identified potential MYC enhancers of HCC and propose that the associated eRNAs may be suitable targets for HCC treatment.


Assuntos
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Proliferação de Células/genética , Elementos Facilitadores Genéticos/genética , Elementos Facilitadores Genéticos/fisiologia , Genes myc/efeitos dos fármacos , Genes myc/fisiologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , RNA não Traduzido , Linhagem Celular Tumoral , Humanos
3.
Plant J ; 109(3): 541-554, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34773305

RESUMO

Insulators characterized in Drosophila and mammals have been shown to play a key role in the restriction of promiscuous enhancer-promoter interactions, as well as reshaping the topological landscape of chromosomes. Yet the role of insulators in plants remains poorly understood, in large part because of a lack of well-characterized insulators and binding factor(s). In this study, we isolated a 1.2-kb RS2-9 insulator from the Oryza sativa (rice) genome that can, when interposed between an enhancer and promoter, efficiently block the activation function of both constitutive and floral organ-specific enhancers in transgenic Arabidopsis and Nicotiana tabacum (tobacco). In the rice genome, the genes flanking RS2-9 exhibit an absence of mutual transcriptional interactions, as well as a lack of histone modification spread. We further determined that O. sativa Homeobox 1 (OSH1) bound two regions of RS2-9, as well as over 50 000 additional sites in the rice genome, the majority of which resided in intergenic regions. Mutation of one of the two OSH1-binding sites in RS2-9 impaired insulation activity by up to 60%, whereas the mutation of both binding sites virtually abolished insulator function. We also demonstrated that OSH1 binding sites were associated with 72% of the boundaries of topologically associated domains (TADs) identified in the rice genome, which is comparable to the 77% of TAD boundaries bound by the insulator CCCTC-binding factor (CTCF) in mammals. Taken together, our findings indicate that OSH1-RS2-9 acts as a true insulator in plants, and highlight a potential role for OSH1 in gene insulation and topological organization in plant genomes.


Assuntos
Elementos Facilitadores Genéticos/fisiologia , Oryza/genética , Oryza/metabolismo , Regiões Promotoras Genéticas/fisiologia , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Produtos Agrícolas/genética , Produtos Agrícolas/metabolismo , Regulação da Expressão Gênica de Plantas , Genes de Plantas , Fatores de Transcrição/fisiologia
4.
Nat Commun ; 12(1): 6660, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34795220

RESUMO

Gene expression is controlled by the involvement of gene-proximal (promoters) and distal (enhancers) regulatory elements. Our previous results demonstrated that a subset of gene promoters, termed Epromoters, work as bona fide enhancers and regulate distal gene expression. Here, we hypothesized that Epromoters play a key role in the coordination of rapid gene induction during the inflammatory response. Using a high-throughput reporter assay we explored the function of Epromoters in response to type I interferon. We find that clusters of IFNa-induced genes are frequently associated with Epromoters and that these regulatory elements preferentially recruit the STAT1/2 and IRF transcription factors and distally regulate the activation of interferon-response genes. Consistently, we identified and validated the involvement of Epromoter-containing clusters in the regulation of LPS-stimulated macrophages. Our findings suggest that Epromoters function as a local hub recruiting the key TFs required for coordinated regulation of gene clusters during the inflammatory response.


Assuntos
Elementos Facilitadores Genéticos/fisiologia , Inflamação/genética , Fatores Reguladores de Interferon/metabolismo , Regiões Promotoras Genéticas/fisiologia , Animais , Elementos Facilitadores Genéticos/efeitos dos fármacos , Regulação da Expressão Gênica , Células HeLa , Humanos , Inflamação/metabolismo , Interferon Tipo I/metabolismo , Interferon-alfa/farmacologia , Células K562 , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Família Multigênica/efeitos dos fármacos , Família Multigênica/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT2/metabolismo
5.
Mol Cell Biol ; 41(7): e0059420, 2021 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-33875575

RESUMO

Vascular endothelial growth factors (VEGFs) are best known as key regulators of angiogenesis and lymphangiogenesis. Although VEGFs have been promising therapeutic targets for various cardiovascular diseases, their regulatory landscape in endothelial cells remains elusive. Several studies have highlighted the involvement of noncoding RNAs (ncRNAs) in the modulation of VEGF expression. In this study, we investigated the role of two classes of ncRNAs, long ncRNAs (lncRNAs) and enhancer RNAs (eRNAs), in the transcriptional regulation of VEGFA and VEGFC. By integrating genome-wide global run-on sequencing (GRO-Seq) and chromosome conformation capture (Hi-C) data, we identified putative lncRNAs and eRNAs associated with VEGFA and VEGFC genes in endothelial cells. A subset of the identified putative enhancers demonstrated regulatory activity in a reporter assay. Importantly, we demonstrate that deletion of enhancers and lncRNAs by CRISPR/Cas9 promoted significant changes in VEGFA and VEGFC expression. Transcriptome sequencing (RNA-Seq) data from lncRNA deletions showed downstream factors implicated in VEGFA- and VEGFC-linked pathways, such as angiogenesis and lymphangiogenesis, suggesting functional roles for these lncRNAs. Our study uncovers novel lncRNAs and eRNAs regulating VEGFA and VEGFC that can be targeted to modulate the expression of these important molecules in endothelial cells.


Assuntos
Células Endoteliais/metabolismo , RNA não Traduzido/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator C de Crescimento do Endotélio Vascular/metabolismo , Elementos Facilitadores Genéticos/genética , Elementos Facilitadores Genéticos/fisiologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Humanos , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , RNA Longo não Codificante/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator C de Crescimento do Endotélio Vascular/genética
6.
Endocrinology ; 162(7)2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33824966

RESUMO

FSH is critical for fertility. Transcription of FSHB, the gene encoding the beta subunit, is rate-limiting in FSH production and is regulated by both GnRH and activin. Activin signals through SMAD transcription factors. Although the mechanisms and importance of activin signaling in mouse Fshb transcription are well-established, activin regulation of human FSHB is less well understood. We previously reported a novel enhancer of FSHB that contains a fertility-associated single nucleotide polymorphism (rs10031006) and requires a region resembling a full (8 base-pair) SMAD binding element (SBE). Here, we investigated the role of the putative SBE within the enhancer in activin and GnRH regulation of FSHB. In mouse gonadotrope-derived LßT2 cells, the upstream enhancer potentiated activin induction of both the human and mouse FSHB proximal promoters and conferred activin responsiveness to a minimal promoter. Activin induction of the enhancer required the SBE and was blocked by the inhibitory SMAD7, confirming involvement of the classical SMAD signaling pathway. GnRH induction of FSHB was also potentiated by the enhancer and dependent on the SBE, consistent with known activin/GnRH synergy regulating FSHB transcription. In DNA pull-down, the enhancer SBE bound SMAD4, and chromatin immunoprecipitation demonstrated SMAD4 enrichment at the enhancer in native chromatin. Combined activin/GnRH treatment elevated levels of the active transcriptional histone marker, histone 3 lysine 27 acetylation, at the enhancer. Overall, this study indicates that the enhancer is directly targeted by activin signaling and identifies a novel, evolutionarily conserved mechanism by which activin and GnRH can regulate FSHB transcription.


Assuntos
Ativinas/farmacologia , Elementos Facilitadores Genéticos/fisiologia , Subunidade beta do Hormônio Folículoestimulante/genética , Hormônio Liberador de Gonadotropina/farmacologia , Transcrição Gênica/efeitos dos fármacos , Ativinas/metabolismo , Animais , Sinergismo Farmacológico , Elementos Facilitadores Genéticos/efeitos dos fármacos , Folistatina/farmacologia , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Camundongos , Regiões Promotoras Genéticas/efeitos dos fármacos , Transdução de Sinais , Proteínas Smad/fisiologia , Proteína Smad4/metabolismo
7.
BMC Cancer ; 21(1): 187, 2021 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-33622275

RESUMO

BACKGROUND: Super-enhancer-associated long noncoding RNAs (SE-lncRNAs) have been reported to play essential roles in tumorigenesis, but the fundamental mechanism of SE-lncRNAs in colorectal cancer (CRC) remains largely unknown. METHODS: A microarray was performed to identify the differentially expressed SE-lncRNAs between CRC tissues and peritumoral tissues. A novel SE-lncRNA, AC005592.2, was selected from these differentially expressed SE-lncRNAs to explore its effects on CRC development. Fluorescence quantitative real-time PCR (qRT-PCR) was used to assay the expression of AC005592.2 in CRC tissues and cell lines. Functional assays were applied to identify the biological effects of AC005592.2 in CRC cells. Furthermore, RNA-seq was employed to predict potential targets of AC005592.2. RESULTS: AC005592.2 was significantly increased in CRC tissues and cells. High expression of AC005592.2 was significantly associated with TNM stage and tumor differentiation in CRC patients. Knockdown of AC005592.2 suppressed CRC cell proliferation, invasion and migration but promoted apoptosis, while AC005592.2 overexpression exerted the opposite effects on CRC cells. In addition, AC005592.2 positively regulated the expression of olfactomedin 4 (OLFM4), which was also upregulated in CRC tissues. CONCLUSION: The findings suggested that AC005592.2 is a crucial promoter of CRC progression and may serve as an attractive therapeutic target for CRC.


Assuntos
Neoplasias Colorretais/patologia , Elementos Facilitadores Genéticos/fisiologia , Regulação Neoplásica da Expressão Gênica , Fator Estimulador de Colônias de Granulócitos/genética , RNA Longo não Codificante/fisiologia , Apoptose , Linhagem Celular Tumoral , Movimento Celular , Neoplasias Colorretais/genética , Progressão da Doença , Humanos , Invasividade Neoplásica , RNA Mensageiro/análise
8.
Genes Dev ; 35(1-2): 65-81, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33334824

RESUMO

During developmental progression the genomes of immune cells undergo large-scale changes in chromatin folding. However, insights into signaling pathways and epigenetic control of nuclear architecture remain rudimentary. Here, we found that in activated neutrophils calcium influx rapidly recruited the cohesin-loading factor NIPBL to thousands of active enhancers and promoters to dictate widespread changes in compartment segregation. NIPBL recruitment to enhancers and promoters occurred with distinct kinetics. The induction of NIPBL-binding was coordinate with increased P300, BRG1 and RNA polymerase II occupancy. NIPBL-bound enhancers were associated with NFAT, PU.1, and CEBP cis elements, whereas NIPBL-bound promoters were enriched for GC-rich DNA sequences. Using an acute degradation system, we found that the histone acetyltransferases P300 and CBP maintained H3K27ac abundance and facilitated NIPBL occupancy at enhancers and that active transcriptional elongation is essential to maintain H3K27ac abundance. Chromatin remodelers, containing either of the mutually exclusive BRG1 and BRM ATPases, promoted NIPBL recruitment at active enhancers. Conversely, at active promoters, depletion of BRG1 and BRM showed minimal effect on NIPBL occupancy. Finally, we found that calcium signaling in both primary innate and adaptive immune cells swiftly induced NIPBL occupancy. Collectively, these data reveal how transcriptional regulators, histone acetyltransferases, chromatin remodelers, and transcription elongation promote NIPBL occupancy at active enhancers while the induction of NIPLB occupancy at promoters is primarily associated with GC-rich DNA sequences.


Assuntos
Cálcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Elementos Facilitadores Genéticos/fisiologia , Genoma/fisiologia , Regiões Promotoras Genéticas/fisiologia , Transdução de Sinais/fisiologia , Animais , Proteínas de Ciclo Celular/imunologia , Células Cultivadas , Montagem e Desmontagem da Cromatina , Proteínas Cromossômicas não Histona/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Histona Acetiltransferases/metabolismo , Histonas/metabolismo , Camundongos , Fatores de Transcrição NFATC/metabolismo , Neutrófilos/citologia , Transporte Proteico , Elongação da Transcrição Genética
9.
Pharm Biol ; 58(1): 1252-1262, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33332210

RESUMO

CONTEXT: Dexamethasone (DXM) has an anti-immunoinflammatory effect, and is often used in acute kidney injury (AKI). However, the effects of DXM on albumin (ALB) have not been fully studied. OBJECTIVE: To investigate the effects of DXM on ALB production and renal function. MATERIALS AND METHODS: Male Wistar rats were divided into normal and DXM groups (0.25, 0.5, 1 mg/kg for 5 days) (n = 15) for a dose-dependent study. Rats were divided into normal group and DXM groups (0.5 mg/kg for 3, 5, 7 days) (n = 9) for a time-dependent study. In AKI experiment, rats were divided into normal (saline), cisplatin (CP, 5 mg/kg, i.v.), CP + DXM groups (0.25, 0.5 and 1 mg/kg, i.m.) (n = 16). The blood and the organs were isolated for analysis. RESULTS: In normal, serum ALB (sALB) and serum total protein (sTP) increased in DXM group with sALB increased 19.8-32.2% (from small to large dosages); and 30.2-32.5.6% (from 3 to 7 days of DXM); sTP 15.7-22.6% and 14.2-24.3%; urine ALB (uALB) 31.5-392.3%, and 1047.2-1390.8%; urine TP (uTP) 0.68-173.1% and 98.0-504.9%, compared with normal groups. DXM increased the mRNA expression of Cebp and Hnf, suppressing podocin. In AKI, DXM decreased serum BUN (53.7%), serum Cre (73.4%), sALB (30.0%), sTP (18.7%), uALB (74.5%), uTP (449.3%), rescuing the suppressed podocin in kidney. CONCLUSIONS: DXM acts on Cebp and Hnf and promotes ALB production. This finding helps to evaluate the rationale of DXM for kidney injury.


Assuntos
Injúria Renal Aguda/metabolismo , Dexametasona/farmacologia , Albumina Sérica/biossíntese , Animais , Proteínas Sanguíneas/análise , Cisplatino/toxicidade , Relação Dose-Resposta a Droga , Elementos Facilitadores Genéticos/fisiologia , Rim/efeitos dos fármacos , Masculino , Ratos , Ratos Wistar
10.
Front Endocrinol (Lausanne) ; 11: 572981, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33133019

RESUMO

Glucocorticoids (GCs) and the glucocorticoid receptor (GR) are important regulators of development, inflammation, stress response and metabolism, demonstrated in various diseases including Addison's disease, Cushing's syndrome and by the many side effects of prolonged clinical administration of GCs. These conditions include severe metabolic challenges in key metabolic organs like the liver. In the liver, GR is known to regulate the transcription of key enzymes in glucose and lipid metabolism and contribute to the regulation of circadian-expressed genes. Insights to the modes of GR regulation and the underlying functional mechanisms are key for understanding diseases and for the development of improved clinical uses of GCs. The activity and function of GR is regulated at numerous levels including ligand availability, interaction with heat shock protein (HSP) complexes, expression of GR isoforms and posttranslational modifications. Moreover, recent genomics studies show functional interaction with multiple transcription factors (TF) and coregulators in complex transcriptional networks controlling cell type-specific gene expression by GCs. In this review we describe the different regulatory steps important for GR activity and discuss how different TF interaction partners of GR selectively control hepatic gene transcription and metabolism.


Assuntos
Redes Reguladoras de Genes , Fígado/metabolismo , Receptores de Glucocorticoides/fisiologia , Fatores de Transcrição/fisiologia , Transporte Ativo do Núcleo Celular , Montagem e Desmontagem da Cromatina , Elementos Facilitadores Genéticos/fisiologia , Proteína Forkhead Box O1/fisiologia , Humanos , Receptores de Glucocorticoides/química , Receptores de Glucocorticoides/genética , Fator de Transcrição STAT5/fisiologia , Transdução de Sinais/fisiologia
11.
Sci Rep ; 10(1): 15491, 2020 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-32968144

RESUMO

Large-scale epigenomic projects have mapped hundreds of thousands of potential regulatory sites in the human genome, but only a small proportion of these elements are proximal to transcription start sites. It is believed that the majority of these sequences are remote promoter-activating genomic sites scattered within several hundreds of kilobases from their cognate promoters and referred to as enhancers. It is still unclear what principles, aside from relative closeness in the linear genome, determine which promoter(s) is controlled by a given enhancer; however, this understanding is of great fundamental and clinical relevance. In recent years, C-methods (chromosome conformation capture-based methods) have become a powerful tool for the identification of enhancer-promoter spatial contacts that, in most cases, reflect their functional link. Here, we describe a new hybridisation-based promoter Capture-C protocol that makes use of biotinylated dsDNA probes generated by PCR from a custom pool of long oligonucleotides. The described protocol allows high-resolution promoter interactome description, providing a flexible and cost-effective alternative to the existing promoter Capture-C modifications. Based on the obtained data, we propose several tips on probe design that could potentially improve the results of future experiments.


Assuntos
Epigenômica/métodos , Regiões Promotoras Genéticas , Biotinilação , Cromatina/genética , Cromatina/fisiologia , Cromatina/ultraestrutura , Cromossomos Humanos/genética , Cromossomos Humanos/fisiologia , Sondas de DNA/genética , Elementos Facilitadores Genéticos/genética , Elementos Facilitadores Genéticos/fisiologia , Genoma Humano/genética , Genoma Humano/fisiologia , Células HeLa , Humanos , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/fisiologia
12.
Science ; 369(6508)2020 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-32883834

RESUMO

Vertebrates vary in their ability to regenerate, and the genetic mechanisms underlying such disparity remain elusive. Comparative epigenomic profiling and single-cell sequencing of two related teleost fish uncovered species-specific and evolutionarily conserved genomic responses to regeneration. The conserved response revealed several regeneration-responsive enhancers (RREs), including an element upstream to inhibin beta A (inhba), a known effector of vertebrate regeneration. This element activated expression in regenerating transgenic fish, and its genomic deletion perturbed caudal fin regeneration and abrogated cardiac regeneration altogether. The enhancer is present in mammals, shares functionally essential activator protein 1 (AP-1)-binding motifs, and responds to injury, but it cannot rescue regeneration in fish. This work suggests that changes in AP-1-enriched RREs are likely a crucial source of loss of regenerative capacities in vertebrates.


Assuntos
Elementos Facilitadores Genéticos/fisiologia , Evolução Molecular , Peixes Listrados/genética , Peixes Listrados/fisiologia , Regeneração/genética , Motivos de Aminoácidos , Animais , Epigênese Genética , Perfilação da Expressão Gênica , Histonas/metabolismo , Subunidades beta de Inibinas/genética , RNA-Seq , Análise de Célula Única , Fator de Transcrição AP-1/química , Fator de Transcrição AP-1/metabolismo , Ativação Transcricional , Peixe-Zebra/genética , Peixe-Zebra/fisiologia
13.
Cells ; 9(7)2020 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-32635644

RESUMO

Transcriptional enhancers are major genomic elements that control gene activity in eukaryotes. Recent studies provided deeper insight into the temporal and spatial organization of transcription in the nucleus, the role of non-coding RNAs in the process, and the epigenetic control of gene expression. Thus, multiple molecular details of enhancer functioning were revealed. Here, we describe the recent data and models of molecular organization of enhancer-driven transcription.


Assuntos
Cromatina/metabolismo , Elementos Facilitadores Genéticos/fisiologia , Cromatina/genética , Elementos Facilitadores Genéticos/genética , Epigênese Genética/genética , Epigênese Genética/fisiologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/fisiologia , Humanos , Regiões Promotoras Genéticas/genética
14.
PLoS One ; 15(5): e0233191, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32453736

RESUMO

The Ikzf1 locus encodes the lymphoid specific transcription factor Ikaros, which plays an essential role in both T and B cell differentiation, while deregulation or mutation of IKZF1/Ikzf1 is involved in leukemia. Tissue-specific and cell identity genes are usually associated with clusters of enhancers, also called super-enhancers, which are believed to ensure proper regulation of gene expression throughout cell development and differentiation. Several potential regulatory regions have been identified in close proximity of Ikzf1, however, the full extent of the regulatory landscape of the Ikzf1 locus is not yet established. In this study, we combined epigenomics and transcription factor binding along with high-throughput enhancer assay and 4C-seq to prioritize an enhancer element located 120 kb upstream of the Ikzf1 gene. We found that deletion of the E120 enhancer resulted in a significant reduction of Ikzf1 mRNA. However, the epigenetic landscape and 3D topology of the locus were only slightly affected, highlighting the complexity of the regulatory landscape regulating the Ikzf1 locus.


Assuntos
Elementos Facilitadores Genéticos/fisiologia , Regulação da Expressão Gênica/fisiologia , Loci Gênicos/fisiologia , Fator de Transcrição Ikaros/biossíntese , Animais , Linhagem Celular , Epigenômica , Genes Reporter , Fator de Transcrição Ikaros/genética , Camundongos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
15.
Elife ; 92020 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-32342858

RESUMO

Can a single regulatory sequence be shared by two genes undergoing functional divergence? Here we describe a single promiscuous enhancer within the Drosophila Antennapedia Complex, EO053, that directs aspects of the expression of two adjacent genes, pb (a Hox2 ortholog) and zen2 (a divergent Hox3 paralog), with disparate spatial and temporal expression patterns. We were unable to separate the pb-like and zen2-like specificities within EO053, and we identify sequences affecting both expression patterns. Importantly, genomic deletion experiments demonstrate that EO053 cooperates with additional pb- and zen2-specific enhancers to regulate the mRNA expression of both genes. We examine sequence conservation of EO053 within the Schizophora, and show that patterns of synteny between the Hox2 and Hox3 orthologs in Arthropods are consistent with a shared regulatory relationship extending prior to the Hox3/zen divergence. Thus, EO053 represents an example of two genes having evolved disparate outputs while utilizing this shared regulatory region.Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).


Assuntos
Proteínas de Drosophila/genética , Elementos Facilitadores Genéticos/fisiologia , Proteínas de Homeodomínio/genética , Proteínas de Insetos/genética , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Animais , Drosophila , Evolução Molecular , Regulação da Expressão Gênica , RNA Mensageiro/análise
16.
Cancer Res ; 80(9): 1804-1818, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32127356

RESUMO

Although genome-wide association studies (GWAS) have identified more than 100 colorectal cancer risk loci, most of the biological mechanisms associated with these loci remain unclear. Here we first performed a comprehensive expression quantitative trait loci analysis in colorectal cancer tissues adjusted for multiple confounders to test the determinants of germline variants in established GWAS susceptibility loci on mRNA and long noncoding RNA (lncRNA) expression. Combining integrative functional genomic/epigenomic analyses and a large-scale population study consisting of 6,024 cases and 10,022 controls, we then prioritized rs174575 with a C>G change as a potential causal candidate for colorectal cancer at 11q12.2, as its G allele was associated with an increased risk of colorectal cancer (OR = 1.26; 95% confidence interval = 1.17-1.36; P = 2.57 × 10-9). rs174575 acted as an allele-specific enhancer to distally facilitate expression of both FADS2 and lncRNA AP002754.2 via long-range enhancer-promoter interaction loops, which were mediated by E2F1. AP002754.2 further activated a transcriptional activator that upregulated FADS2 expression. FADS2, in turn, was overexpressed in colorectal cancer tumor tissues and functioned as a potential oncogene that facilitated colorectal cancer cell proliferation and xenograft growth in vitro and in vivo by increasing the metabolism of PGE2, an oncogenic molecule involved in colorectal cancer tumorigenesis. Our findings represent a novel mechanism by which a noncoding variant can facilitate long-range genome interactions to modulate the expression of multiple genes including not only mRNA, but also lncRNA, which provides new insights into the understanding of colorectal cancer etiology. SIGNIFICANCE: This study provides an oncogenic regulatory circuit among several oncogenes including E2F1, FADS2, and AP002754.2 underlying the association of rs174575 with colorectal cancer risk, which is driven by long-range enhancer-promoter interaction loops. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/9/1804/F1.large.jpg.


Assuntos
Neoplasias Colorretais/genética , Fator de Transcrição E2F1/metabolismo , Elementos Facilitadores Genéticos/fisiologia , Ácidos Graxos Dessaturases/genética , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas/fisiologia , RNA Longo não Codificante/metabolismo , Alelos , Animais , Estudos de Casos e Controles , Linhagem Celular Tumoral , Proliferação de Células/genética , Cromossomos Humanos Par 11/genética , Neoplasias Colorretais/metabolismo , Dinoprostona/metabolismo , Dinoprostona/farmacologia , Fator de Transcrição E2F1/genética , Ácidos Graxos Dessaturases/metabolismo , Feminino , Estudo de Associação Genômica Ampla , Xenoenxertos/patologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas de Neoplasias/metabolismo , Oncogenes , Locos de Características Quantitativas , RNA Longo não Codificante/genética , Fatores de Transcrição
17.
Cell ; 180(6): 1262-1271.e15, 2020 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-32169219

RESUMO

Establishing causal links between non-coding variants and human phenotypes is an increasing challenge. Here, we introduce a high-throughput mouse reporter assay for assessing the pathogenic potential of human enhancer variants in vivo and examine nearly a thousand variants in an enhancer repeatedly linked to polydactyly. We show that 71% of all rare non-coding variants previously proposed as causal lead to reporter gene expression in a pattern consistent with their pathogenic role. Variants observed to alter enhancer activity were further confirmed to cause polydactyly in knockin mice. We also used combinatorial and single-nucleotide mutagenesis to evaluate the in vivo impact of mutations affecting all positions of the enhancer and identified additional functional substitutions, including potentially pathogenic variants hitherto not observed in humans. Our results uncover the functional consequences of hundreds of mutations in a phenotype-associated enhancer and establish a widely applicable strategy for systematic in vivo evaluation of human enhancer variants.


Assuntos
Elementos Facilitadores Genéticos/genética , Ensaios de Triagem em Larga Escala/métodos , Polidactilia/genética , Animais , Elementos Facilitadores Genéticos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Técnicas de Introdução de Genes/métodos , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Camundongos , Mutação , Fenótipo , Polidactilia/metabolismo , RNA não Traduzido/genética
18.
Development ; 147(6)2020 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-32094114

RESUMO

How temporal cues combine with spatial inputs to control gene expression during development is poorly understood. Here, we test the hypothesis that the Drosophila transcription factor E93 controls temporal gene expression by regulating chromatin accessibility. Precocious expression of E93 early in wing development reveals that it can simultaneously activate and deactivate different target enhancers. Notably, the precocious patterns of enhancer activity resemble the wild-type patterns that occur later in development, suggesting that expression of E93 alters the competence of enhancers to respond to spatial cues. Genomic profiling reveals that precocious E93 expression is sufficient to regulate chromatin accessibility at a subset of its targets. These accessibility changes mimic those that normally occur later in development, indicating that precocious E93 accelerates the wild-type developmental program. Further, we find that target enhancers that do not respond to precocious E93 in early wings become responsive after a developmental transition, suggesting that parallel temporal pathways work alongside E93. These findings support a model wherein E93 expression functions as an instructive cue that defines a broad window of developmental time through control of chromatin accessibility.


Assuntos
Cromatina/metabolismo , Proteínas de Drosophila/genética , Desenvolvimento Embrionário/genética , Elementos Facilitadores Genéticos/fisiologia , Fatores de Transcrição/metabolismo , Animais , Animais Geneticamente Modificados , Cromatina/química , Montagem e Desmontagem da Cromatina/fisiologia , Drosophila/embriologia , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Embrião não Mamífero , Regulação da Expressão Gênica no Desenvolvimento , Metamorfose Biológica/genética , Ligação Proteica , Fatores de Transcrição/genética , Asas de Animais/embriologia , Asas de Animais/metabolismo
19.
Nat Neurosci ; 23(1): 5-14, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31740812

RESUMO

Transcriptional enhancers are regulatory DNA elements that underlie the specificity and dynamic patterns of gene expression. Over the past decade, large-scale functional genomics projects have driven transformative progress in our understanding of enhancers. These data have relevance for identifying mechanisms of gene regulation in the CNS, elucidating the function of non-coding regulatory sequences in neurobiology and linking sequence variation within enhancers to genetic risk for neurological and psychiatric disorders. However, the sheer volume and complexity of genomic data presents a challenge to interpreting enhancer function in normal and pathogenic neurobiological processes. Here, to advance the application of genome-scale enhancer data, we offer a primer on current models of enhancer function in the CNS, we review how enhancers regulate gene expression across the neuronal lifespan, and we suggest how emerging findings regarding the role of non-coding sequence variation offer opportunities for understanding brain disorders and developing new technologies for neuroscience.


Assuntos
Encéfalo/fisiologia , Elementos Facilitadores Genéticos/fisiologia , Regulação da Expressão Gênica/fisiologia , Neurônios/fisiologia , Transcrição Gênica/fisiologia , Animais , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA