Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.732
Filtrar
1.
Nat Commun ; 15(1): 6562, 2024 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-39095348

RESUMEN

Unraveling the regulatory mechanisms that govern complex traits is pivotal for advancing crop improvement. Here we present a comprehensive regulome atlas for rice (Oryza sativa), charting the chromatin accessibility across 23 distinct tissues from three representative varieties. Our study uncovers 117,176 unique open chromatin regions (OCRs), accounting for ~15% of the rice genome, a notably higher proportion compared to previous reports in plants. Integrating RNA-seq data from matched tissues, we confidently predict 59,075 OCR-to-gene links, with enhancers constituting 69.54% of these associations, including many known enhancer-to-gene links. Leveraging this resource, we re-evaluate genome-wide association study results and discover a previously unknown function of OsbZIP06 in seed germination, which we subsequently confirm through experimental validation. We optimize deep learning models to decode regulatory grammar, achieving robust modeling of tissue-specific chromatin accessibility. This approach allows to predict cross-variety regulatory dynamics from genomic sequences, shedding light on the genetic underpinnings of cis-regulatory divergence and morphological disparities between varieties. Overall, our study establishes a foundational resource for rice functional genomics and precision molecular breeding, providing valuable insights into regulatory mechanisms governing complex traits.


Asunto(s)
Cromatina , Regulación de la Expresión Génica de las Plantas , Genoma de Planta , Estudio de Asociación del Genoma Completo , Oryza , Oryza/genética , Oryza/crecimiento & desarrollo , Cromatina/metabolismo , Cromatina/genética , Mapeo Cromosómico/métodos , Sitios de Carácter Cuantitativo/genética , Germinación/genética , Elementos de Facilitación Genéticos/genética , Aprendizaje Profundo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo
2.
Mol Metab ; 86: 101978, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38950776

RESUMEN

OBJECTIVE: Aberrant glucolipid metabolism in the heart is a characteristic factor in diabetic cardiomyopathy (DbCM). Super-enhancers-driven noncoding RNAs (seRNAs) are emerging as powerful regulators in the progression of cardiac diseases. However, the functions of seRNAs in DbCM have not been fully elucidated. METHODS: Super enhancers and their associated seRNAs were screened and identified by H3K27ac ChIP-seq data in the Encyclopedia of DNA Elements (ENCODE) dataset. A dual-luciferase reporter assay was performed to analyze the function of super-enhancers on the transcription of peroxisome proliferator-activated receptor α-related seRNA (PPARα-seRNA). A DbCM mouse model was established using db/db leptin receptor-deficient mice. Adeno-associated virus serotype 9-seRNA (AAV9-seRNA) was injected via the tail vein to evaluate the role of seRNA in DbCM. The underlying mechanism was explored through RNA pull-down, RNA and chromatin immunoprecipitation, and chromatin isolation by RNA purification. RESULTS: PPARα-seRNA was regulated by super-enhancers and its levels were increased in response to high glucose and palmitic acid stimulation in cardiomyocytes. Functionally, PPARα-seRNA overexpression aggravated lipid deposition, reduced glucose uptake, and repressed energy production. In contrast, PPARα-seRNA knockdown ameliorated metabolic disorder in vitro. In vivo, overexpression of PPARα-seRNA exacerbated cardiac metabolic disorder and deteriorated cardiac dysfunction, myocardial fibrosis, and hypertrophy in DbCM. Mechanistically, PPARα-seRNA bound to the histone demethylase KDM4B (Lysine-specific demethylase 4B) and decreased H3K9me3 levels in the promoter region of PPARα, ultimately enhancing its transcription. CONCLUSIONS: Our study revealed the pivotal function of a super-enhancer-driven long noncoding RNA (lncRNA), PPARα-seRNA, in the deterioration of cardiac function and the exacerbation of metabolic abnormalities in diabetic cardiomyopathy, which recruited KDM4B to the promoter region of PPARα and repression of its transcription. This suggests a promising therapeutic strategy for the treatment of DbCM.


Asunto(s)
Cardiomiopatías Diabéticas , Metabolismo de los Lípidos , PPAR alfa , ARN Largo no Codificante , Animales , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/patología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Ratones , PPAR alfa/metabolismo , PPAR alfa/genética , Metabolismo de los Lípidos/genética , Ratones Endogámicos C57BL , Masculino , Miocitos Cardíacos/metabolismo , Elementos de Facilitación Genéticos/genética , Glucosa/metabolismo
3.
Development ; 151(13)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38994775

RESUMEN

Super-enhancers (SEs) are expansive regions of genomic DNA that regulate the expression of genes involved in cell identity and cell fate. We recently identified developmental stage- and cell type-specific modules within the murine Vsx2 SE. Here, we show that the human VSX2 SE modules have similar developmental stage- and cell type-specific activity in reporter gene assays. By inserting the human sequence of one VSX2 SE module into a mouse with microphthalmia, eye size was rescued. To understand the function of these SE modules during human retinal development, we deleted individual modules in human embryonic stem cells and generated retinal organoids. Deleting one module results in small organoids, recapitulating the small-eyed phenotype of mice with microphthalmia, while deletion of the other module led to disruptions in bipolar neuron development. This prototypical SE serves as a model for understanding developmental stage- and cell type-specific effects of neurogenic transcription factors with complex expression patterns. Moreover, by elucidating the gene regulatory mechanisms, we can begin to examine how dysregulation of these mechanisms contributes to phenotypic diversity and disease.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio , Retina , Factores de Transcripción , Animales , Humanos , Ratones , Elementos de Facilitación Genéticos/genética , Evolución Molecular , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/citología , Microftalmía/genética , Microftalmía/patología , Neurogénesis/genética , Organoides/metabolismo , Retina/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética
4.
Nat Commun ; 15(1): 6130, 2024 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-39033128

RESUMEN

Cancer genomes are composed of many complex structural alterations on chromosomes and extrachromosomal DNA (ecDNA), making it difficult to identify non-coding enhancer regions that are hijacked to activate oncogene expression. Here, we describe a 3D genomics-based analysis called HAPI (Highly Active Promoter Interactions) to characterize enhancer hijacking. HAPI analysis of HiChIP data from 34 cancer cell lines identified enhancer hijacking events that activate both known and potentially novel oncogenes such as MYC, CCND1, ETV1, CRKL, and ID4. Furthermore, we found enhancer hijacking among multiple oncogenes from different chromosomes, often including MYC, on the same complex amplicons such as ecDNA. We characterized a MYC-ERBB2 chimeric ecDNA, in which ERBB2 heavily hijacks MYC's enhancers. Notably, CRISPRi of the MYC promoter led to increased interaction of ERBB2 with MYC enhancers and elevated ERBB2 expression. Our HAPI analysis tool provides a robust strategy to detect enhancer hijacking and reveals novel insights into oncogene activation.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Genómica , Oncogenes , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myc , Receptor ErbB-2 , Humanos , Elementos de Facilitación Genéticos/genética , Línea Celular Tumoral , Regiones Promotoras Genéticas/genética , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Genómica/métodos , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología
5.
Adv Exp Med Biol ; 1459: 97-113, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39017841

RESUMEN

Helix-loop-helix (HLH) transcription factors (TFs) play a key role in various cellular differentiation and function through the regulation of enhancer activity. E2A, a member of the mammalian E-protein family (class I HLH protein), is well known to play an important role in hematopoiesis, especially in adaptive lymphocyte development. E2A instructs B- and T-cell lineage development through the regulation of enhancer activity for B- or T-cell signature gene expression, including Rag1 and Rag2 (Rag1/2) genes. In this chapter, we mainly focus on the function of E2A in B-cell development and on the roles of E2A in establishing the enhancer landscape through the recruitment of EP300/KAT3B, chromatin remodeling complex, mediator, cohesion, and TET proteins. Finally, we demonstrate how E2A orchestrates the assembly of the Rag1/2 gene super-enhancer (SE) formation by changing the chromatin conformation across the Rag gene locus.


Asunto(s)
Linfocitos B , Proteínas de Homeodominio , Humanos , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Elementos de Facilitación Genéticos/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Ensamble y Desensamble de Cromatina , Diferenciación Celular/genética , Cromatina/metabolismo , Cromatina/genética , Proteína p300 Asociada a E1A/metabolismo , Proteína p300 Asociada a E1A/genética , Proteínas de Unión al ADN , Proteínas Nucleares
6.
Clin Epigenetics ; 16(1): 86, 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38965562

RESUMEN

BACKGROUND: Presbycusis, also referred to as age-related hearing loss (ARHL), is a condition that results from the cumulative effects of aging on an individual's auditory capabilities. Given the limited understanding of epigenetic mechanisms in ARHL, our research focuses on alterations in chromatin-accessible regions. METHODS: We employed assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) in conjunction with unique identifier (UID) mRNA-seq between young and aging cochleae, and conducted integrated analysis as well as motif/TF-gene prediction. Additionally, the essential role of super-enhancers (SEs) in the development of ARHL was identified by comparative analysis to previous research. Meanwhile, an ARHL mouse model and an aging mimic hair cell (HC) model were established with a comprehensive identification of senescence phenotypes to access the role of SEs in ARHL progression. RESULTS: The control cochlear tissue exhibited greater chromatin accessibility than cochlear tissue affected by ARHL. Furthermore, the levels of histone 3 lysine 27 acetylation were significantly depressed in both aging cochlea and aging mimic HEI-OC1 cells, highlighting the essential role of SEs in the development of ARHL. The potential senescence-associated super-enhancers (SASEs) of ARHL were identified, most of which exhibited decreased chromatin accessibility. The majority of genes related to the SASEs showed obvious decreases in mRNA expression level in aging HCs and was noticeably altered following treatment with JQ1 (a commonly used SE inhibitor). CONCLUSION: The chromatin accessibility in control cochlear tissue was higher than that in cochlear tissue affected by ARHL. Potential SEs involved in ARHL were identified, which might provide a basis for future therapeutics targeting SASEs related to ARHL.


Asunto(s)
Envejecimiento , Cromatina , Cóclea , Elementos de Facilitación Genéticos , Presbiacusia , Animales , Ratones , Cóclea/metabolismo , Cóclea/efectos de los fármacos , Cromatina/genética , Cromatina/metabolismo , Envejecimiento/genética , Presbiacusia/genética , Presbiacusia/metabolismo , Elementos de Facilitación Genéticos/genética , Transcriptoma/genética , Modelos Animales de Enfermedad , Epigénesis Genética/genética , Histonas/metabolismo , Histonas/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Masculino
7.
Nat Commun ; 15(1): 5638, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38965263

RESUMEN

Triple Negative Breast Cancer (TNBC) is the most aggressive breast cancer subtype suffering from limited targeted treatment options. Following recent reports correlating Fibroblast growth factor-inducible 14 (Fn14) receptor overexpression in Estrogen Receptor (ER)-negative breast cancers with metastatic events, we show that Fn14 is specifically overexpressed in TNBC patients and associated with poor survival. We demonstrate that constitutive Fn14 signalling rewires the transcriptomic and epigenomic landscape of TNBC, leading to enhanced tumour growth and metastasis. We further illustrate that such mechanisms activate TNBC-specific super enhancers (SE) to drive the transcriptional activation of cancer dependency genes via chromatin looping. In particular, we uncover the SE-driven upregulation of Nicotinamide phosphoribosyltransferase (NAMPT), which promotes NAD+ and ATP metabolic reprogramming critical for filopodia formation and metastasis. Collectively, our study details the complex mechanistic link between TWEAK/Fn14 signalling and TNBC metastasis, which reveals several vulnerabilities which could be pursued for the targeted treatment of TNBC patients.


Asunto(s)
Citocina TWEAK , Regulación Neoplásica de la Expresión Génica , Nicotinamida Fosforribosiltransferasa , Transducción de Señal , Receptor de TWEAK , Neoplasias de la Mama Triple Negativas , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Humanos , Receptor de TWEAK/metabolismo , Receptor de TWEAK/genética , Femenino , Citocina TWEAK/metabolismo , Citocina TWEAK/genética , Nicotinamida Fosforribosiltransferasa/metabolismo , Nicotinamida Fosforribosiltransferasa/genética , Animales , Línea Celular Tumoral , Ratones , Metástasis de la Neoplasia , Citocinas/metabolismo , Elementos de Facilitación Genéticos/genética
8.
Genes (Basel) ; 15(7)2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-39062635

RESUMEN

In humans, the HS1.2 enhancer in the Ig heavy-chain locus is modular, with length polymorphism. Previous studies have shown the following features for this variation: (i) strong population structuring; (ii) association with autoimmune diseases; and (iii) association with developmental changes in Ig expression. The HS1.2 region could then be considered as a contributor to inter-individual diversity in humoral response in adaptive immunity. We experimentally determined the HS1.2-length class genotype in 72 of the 1000 Genomes CEU cell lines and assigned the HS1.2 alleles to haplotypes defined by 18 landmark SNPs. We also sequenced the variable portion and ~200 bp of the flanking DNA of 34 HS1.2 alleles. Furthermore, we computationally explored the ability of different allelic arrangements to bind transcription factors. Non-random association between HS1.2 and Gm allotypes in the European population clearly emerged. We show a wealth of variation in the modular composition of HS1.2, with five SNPs further contributing to diversity. Longer alleles offer more potential sites for binding but, for same-length alleles, SNP variation creates/destroys potential binding sites. Altogether, the arrangements of modules and SNP alleles both inside and outside HS1.2 denote an organization of diversity far from randomness. In the context of the strong divergence of human populations for this genomic region and the reported disease associations, our results suggest that selective forces shaped the pattern of its diversity.


Asunto(s)
Elementos de Facilitación Genéticos , Polimorfismo de Nucleótido Simple , Humanos , Elementos de Facilitación Genéticos/genética , Alelos , Haplotipos , Genoma Humano , Factores de Transcripción/genética , Sitios de Unión
9.
Front Immunol ; 15: 1387253, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38947339

RESUMEN

Type I diabetes is an autoimmune disease mediated by T-cell destruction of ß cells in pancreatic islets. Currently, there is no known cure, and treatment consists of daily insulin injections. Genome-wide association studies and twin studies have indicated a strong genetic heritability for type I diabetes and implicated several genes. As most strongly associated variants are noncoding, there is still a lack of identification of functional and, therefore, likely causal variants. Given that many of these genetic variants reside in enhancer elements, we have tested 121 CD4+ T-cell enhancer variants associated with T1D. We found four to be functional through massively parallel reporter assays. Three of the enhancer variants weaken activity, while the fourth strengthens activity. We link these to their cognate genes using 3D genome architecture or eQTL data and validate them using CRISPR editing. Validated target genes include CLEC16A and SOCS1. While these genes have been previously implicated in type 1 diabetes and other autoimmune diseases, we show that enhancers controlling their expression harbor functional variants. These variants, therefore, may act as causal type 1 diabetic variants.


Asunto(s)
Linfocitos T CD4-Positivos , Diabetes Mellitus Tipo 1 , Elementos de Facilitación Genéticos , Predisposición Genética a la Enfermedad , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Humanos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Elementos de Facilitación Genéticos/genética , Proteína 1 Supresora de la Señalización de Citocinas/genética , Estudio de Asociación del Genoma Completo , Lectinas Tipo C/genética , Variación Genética , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo
10.
Nat Commun ; 15(1): 5610, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38969652

RESUMEN

Group 2 innate lymphoid cells (ILC2s) are a subset of innate lymphocytes that produce type 2 cytokines, including IL-4, IL-5, and IL-13. GATA3 is a critical transcription factor for ILC2 development at multiple stages. However, when and how GATA3 is induced to the levels required for ILC2 development remains unclear. Herein, we identify ILC2-specific GATA3-related tandem super-enhancers (G3SE) that induce high GATA3 in ILC2-committed precursors. G3SE-deficient mice exhibit ILC2 deficiency in the bone marrow, lung, liver, and small intestine with minimal impact on other ILC lineages or Th2 cells. Single-cell RNA-sequencing and subsequent flow cytometry analysis show that GATA3 induction mechanism, which is required for entering the ILC2 stage, is lost in IL-17RB+PD-1- late ILC2-committed precursor stage in G3SE-deficient mice. Cnot6l, part of the CCR4-NOT deadenylase complex, is a possible GATA3 target during ILC2 development. Our findings implicate a stage-specific regulatory mechanism for GATA3 expression during ILC2 development.


Asunto(s)
Linaje de la Célula , Factor de Transcripción GATA3 , Inmunidad Innata , Linfocitos , Animales , Factor de Transcripción GATA3/metabolismo , Factor de Transcripción GATA3/genética , Ratones , Linfocitos/inmunología , Linfocitos/metabolismo , Linfocitos/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Elementos de Facilitación Genéticos/genética , Células Th2/inmunología , Diferenciación Celular/inmunología , Análisis de la Célula Individual
11.
Cell Rep ; 43(7): 114456, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-38990722

RESUMEN

The rearrangement and expression of the immunoglobulin µ heavy chain (Igh) gene require communication of the intragenic Eµ and 3' regulatory region (RR) enhancers with the variable (VH) gene promoter. Eµ binding of the transcription factor YY1 has been implicated in enhancer-promoter communication, but the YY1 protein network remains obscure. By analyzing the comprehensive proteome of the 1-kb Eµ wild-type enhancer and that of Eµ lacking the YY1 binding site, we identified the male-specific lethal (MSL)/MOF complex as a component of the YY1 protein network. We found that MSL2 recruitment depends on YY1 and that gene knockout of Msl2 in primary pre-B cells reduces µ gene expression and chromatin looping of Eµ to the 3' RR enhancer and VH promoter. Moreover, Mof heterozygosity in mice impaired µ expression and early B cell differentiation. Together, these data suggest that the MSL/MOF complex regulates Igh gene expression by augmenting YY1-mediated enhancer-promoter communication.


Asunto(s)
Elementos de Facilitación Genéticos , Regiones Promotoras Genéticas , Factor de Transcripción YY1 , Animales , Masculino , Ratones , Diferenciación Celular , Elementos de Facilitación Genéticos/genética , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/metabolismo , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Factor de Transcripción YY1/metabolismo , Factor de Transcripción YY1/genética , Femenino
12.
Nat Commun ; 15(1): 6464, 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39085231

RESUMEN

Gene regulatory elements drive complex biological phenomena and their mutations are associated with common human diseases. The impacts of human regulatory variants are often tested using model organisms such as mice. However, mapping human enhancers to conserved elements in mice remains a challenge, due to both rapid enhancer evolution and limitations of current computational methods. We analyze distal enhancers across 45 matched human/mouse cell/tissue pairs from a comprehensive dataset of DNase-seq experiments, and show that while cell-specific regulatory vocabulary is conserved, enhancers evolve more rapidly than promoters and CTCF binding sites. Enhancer conservation rates vary across cell types, in part explainable by tissue specific transposable element activity. We present an improved genome alignment algorithm using gapped-kmer features, called gkm-align, and make genome wide predictions for 1,401,803 orthologous regulatory elements. We show that gkm-align discovers 23,660 novel human/mouse conserved enhancers missed by previous algorithms, with strong evidence of conserved functional activity.


Asunto(s)
Algoritmos , Secuencia Conservada , Elementos de Facilitación Genéticos , Animales , Elementos de Facilitación Genéticos/genética , Humanos , Ratones , Evolución Molecular , Sitios de Unión/genética , Mamíferos/genética , Regiones Promotoras Genéticas/genética , Biología Computacional/métodos , Factor de Unión a CCCTC/metabolismo , Factor de Unión a CCCTC/genética
13.
Nat Commun ; 15(1): 4914, 2024 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-38851846

RESUMEN

FOXA family proteins act as pioneer factors by remodeling compact chromatin structures. FOXA1 is crucial for the chromatin binding of the androgen receptor (AR) in both normal prostate epithelial cells and the luminal subtype of prostate cancer (PCa). Recent studies have highlighted the emergence of FOXA2 as an adaptive response to AR signaling inhibition treatments. However, the role of the FOXA1 to FOXA2 transition in regulating cancer lineage plasticity remains unclear. Our study demonstrates that FOXA2 binds to distinct classes of developmental enhancers in multiple AR-independent PCa subtypes, with its binding depending on LSD1. Moreover, we reveal that FOXA2 collaborates with JUN at chromatin and promotes transcriptional reprogramming of AP-1 in lineage-plastic cancer cells, thereby facilitating cell state transitions to multiple lineages. Overall, our findings underscore the pivotal role of FOXA2 as a pan-plasticity driver that rewires AP-1 to induce the differential transcriptional reprogramming necessary for cancer cell lineage plasticity.


Asunto(s)
Linaje de la Célula , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 3-beta del Hepatocito , Neoplasias de la Próstata , Factor de Transcripción AP-1 , Masculino , Humanos , Factor Nuclear 3-beta del Hepatocito/metabolismo , Factor Nuclear 3-beta del Hepatocito/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Factor de Transcripción AP-1/metabolismo , Factor de Transcripción AP-1/genética , Línea Celular Tumoral , Linaje de la Célula/genética , Histona Demetilasas/metabolismo , Histona Demetilasas/genética , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Factor Nuclear 3-alfa del Hepatocito/genética , Receptores Androgénicos/metabolismo , Receptores Androgénicos/genética , Animales , Cromatina/metabolismo , Cromatina/genética , Plasticidad de la Célula/genética , Reprogramación Celular/genética , Ratones , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Elementos de Facilitación Genéticos/genética , Transcripción Genética
15.
J Transl Med ; 22(1): 547, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38849954

RESUMEN

BACKGROUND: Enhancers are important gene regulatory elements that promote the expression of critical genes in development and disease. Aberrant enhancer can modulate cancer risk and activate oncogenes that lead to the occurrence of various cancers. However, the underlying mechanism of most enhancers in cancer remains unclear. Here, we aim to explore the function and mechanism of a crucial enhancer in melanoma. METHODS: Multi-omics data were applied to identify an enhancer (enh17) involved in melanoma progression. To evaluate the function of enh17, CRISPR/Cas9 technology were applied to knockout enh17 in melanoma cell line A375. RNA-seq, ChIP-seq and Hi-C data analysis integrated with luciferase reporter assay were performed to identify the potential target gene of enh17. Functional experiments were conducted to further validate the function of the target gene ETV4. Multi-omics data integrated with CUT&Tag sequencing were performed to validate the binding profile of the inferred transcription factor STAT3. RESULTS: An enhancer, named enh17 here, was found to be aberrantly activated and involved in melanoma progression. CRISPR/Cas9-mediated deletion of enh17 inhibited cell proliferation, migration, and tumor growth of melanoma both in vitro and in vivo. Mechanistically, we identified ETV4 as a target gene regulated by enh17, and functional experiments further support ETV4 as a target gene that is involved in cancer-associated phenotypes. In addition, STAT3 acts as a transcription factor binding with enh17 to regulate the transcription of ETV4. CONCLUSIONS: Our findings revealed that enh17 plays an oncogenic role and promotes tumor progression in melanoma, and its transcriptional regulatory mechanisms were fully elucidated, which may open a promising window for melanoma prevention and treatment.


Asunto(s)
Proliferación Celular , Progresión de la Enfermedad , Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Melanoma , Humanos , Melanoma/genética , Melanoma/patología , Línea Celular Tumoral , Elementos de Facilitación Genéticos/genética , Proliferación Celular/genética , Movimiento Celular/genética , Animales , Oncogenes/genética , Sistemas CRISPR-Cas/genética , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Carcinogénesis/genética , Carcinogénesis/patología , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Proto-Oncogénicas c-ets/metabolismo , Secuencia de Bases , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/genética
16.
Nature ; 631(8021): 593-600, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38926583

RESUMEN

The current technologies to place new DNA into specific locations in plant genomes are low frequency and error-prone, and this inefficiency hampers genome-editing approaches to develop improved crops1,2. Often considered to be genome 'parasites', transposable elements (TEs) evolved to insert their DNA seamlessly into genomes3-5. Eukaryotic TEs select their site of insertion based on preferences for chromatin contexts, which differ for each TE type6-9. Here we developed a genome engineering tool that controls the TE insertion site and cargo delivered, taking advantage of the natural ability of the TE to precisely excise and insert into the genome. Inspired by CRISPR-associated transposases that target transposition in a programmable manner in bacteria10-12, we fused the rice Pong transposase protein to the Cas9 or Cas12a programmable nucleases. We demonstrated sequence-specific targeted insertion (guided by the CRISPR gRNA) of enhancer elements, an open reading frame and a gene expression cassette into the genome of the model plant Arabidopsis. We then translated this system into soybean-a major global crop in need of targeted insertion technology. We have engineered a TE 'parasite' into a usable and accessible toolkit that enables the sequence-specific targeting of custom DNA into plant genomes.


Asunto(s)
Arabidopsis , Sistemas CRISPR-Cas , Elementos Transponibles de ADN , Edición Génica , Genoma de Planta , Oryza , Transposasas , Transposasas/metabolismo , Transposasas/genética , Arabidopsis/genética , Oryza/genética , Genoma de Planta/genética , Elementos Transponibles de ADN/genética , Edición Génica/métodos , Sistemas CRISPR-Cas/genética , Sistemas de Lectura Abierta/genética , Elementos de Facilitación Genéticos/genética , ARN Guía de Sistemas CRISPR-Cas/genética , ARN Guía de Sistemas CRISPR-Cas/metabolismo , Mutagénesis Insercional/genética , Proteínas Asociadas a CRISPR/metabolismo , Proteínas Asociadas a CRISPR/genética , Proteína 9 Asociada a CRISPR/metabolismo , Proteína 9 Asociada a CRISPR/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Ingeniería Genética/métodos , Plantas Modificadas Genéticamente/genética , Endodesoxirribonucleasas
17.
Nat Commun ; 15(1): 4820, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38844479

RESUMEN

Chondrocyte differentiation controls skeleton development and stature. Here we provide a comprehensive map of chondrocyte-specific enhancers and show that they provide a mechanistic framework through which non-coding genetic variants can influence skeletal development and human stature. Working with fetal chondrocytes isolated from mice bearing a Col2a1 fluorescent regulatory sensor, we identify 780 genes and 2'704 putative enhancers specifically active in chondrocytes using a combination of RNA-seq, ATAC-seq and H3K27ac ChIP-seq. Most of these enhancers (74%) show pan-chondrogenic activity, with smaller populations being restricted to limb (18%) or trunk (8%) chondrocytes only. Notably, genetic variations overlapping these enhancers better explain height differences than those overlapping non-chondrogenic enhancers. Finally, targeted deletions of identified enhancers at the Fgfr3, Col2a1, Hhip and, Nkx3-2 loci confirm their role in regulating cognate genes. This enhancer map provides a framework for understanding how genes and non-coding variations influence bone development and diseases.


Asunto(s)
Condrocitos , Condrogénesis , Elementos de Facilitación Genéticos , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos , Animales , Elementos de Facilitación Genéticos/genética , Humanos , Condrocitos/metabolismo , Condrocitos/citología , Ratones , Condrogénesis/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Regulación del Desarrollo de la Expresión Génica , Desarrollo Óseo/genética , Extremidades/embriología , Masculino , Diferenciación Celular/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Femenino
18.
JCI Insight ; 9(11)2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38855864

RESUMEN

The transcription factor SRY-related HMG box 9 (Sox9) is essential for chondrogenesis. Mutations in and around SOX9 cause campomelic dysplasia (CD) characterized by skeletal malformations. Although the function of Sox9 in this context is well studied, the mechanisms that regulate Sox9 expression in chondrocytes remain to be elucidated. Here, we have used genome-wide profiling to identify 2 Sox9 enhancers located in a proximal breakpoint cluster responsible for CD. Enhancer activity of E308 (located 308 kb 5' upstream) and E160 (located 160 kb 5' upstream) correlated with Sox9 expression levels, and both enhancers showed a synergistic effect in vitro. While single deletions in mice had no apparent effect, simultaneous deletion of both E308 and E160 caused a dwarf phenotype, concomitant with a reduction of Sox9 expression in chondrocytes. Moreover, bone morphogenetic protein 2-dependent chondrocyte differentiation of limb bud mesenchymal cells was severely attenuated in E308/E160 deletion mice. Finally, we found that an open chromatin region upstream of the Sox9 gene was reorganized in the E308/E160 deletion mice to partially compensate for the loss of E308 and E160. In conclusion, our findings reveal a mechanism of Sox9 gene regulation in chondrocytes that might aid in our understanding of the pathophysiology of skeletal disorders.


Asunto(s)
Displasia Campomélica , Diferenciación Celular , Condrocitos , Condrogénesis , Factor de Transcripción SOX9 , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética , Animales , Condrocitos/metabolismo , Ratones , Displasia Campomélica/genética , Displasia Campomélica/patología , Displasia Campomélica/metabolismo , Condrogénesis/genética , Diferenciación Celular/genética , Elementos de Facilitación Genéticos/genética , Cromatina/metabolismo , Cromatina/genética , Regulación del Desarrollo de la Expresión Génica , Ratones Noqueados , Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 2/genética , Humanos , Desarrollo Óseo/genética
19.
Cell Rep ; 43(6): 114291, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38823017

RESUMEN

Atoh7 is transiently expressed in retinal progenitor cells (RPCs) and is required for retinal ganglion cell (RGC) differentiation. In humans, a deletion in a distal non-coding regulatory region upstream of ATOH7 is associated with optic nerve atrophy and blindness. Here, we functionally interrogate the significance of the Atoh7 regulatory landscape to retinogenesis in mice. Deletion of the Atoh7 enhancer structure leads to RGC deficiency, optic nerve hypoplasia, and retinal blood vascular abnormalities, phenocopying inactivation of Atoh7. Further, loss of the Atoh7 remote enhancer impacts ipsilaterally projecting RGCs and disrupts proper axonal projections to the visual thalamus. Deletion of the Atoh7 remote enhancer is also associated with the dysregulation of axonogenesis genes, including the derepression of the axon repulsive cue Robo3. Our data provide insights into how Atoh7 enhancer elements function to promote RGC development and optic nerve formation and highlight a key role of Atoh7 in the transcriptional control of axon guidance molecules.


Asunto(s)
Axones , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Elementos de Facilitación Genéticos , Células Ganglionares de la Retina , Animales , Células Ganglionares de la Retina/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Ratones , Axones/metabolismo , Elementos de Facilitación Genéticos/genética , Neurogénesis/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Nervio Óptico/metabolismo , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Retina/metabolismo , Ratones Endogámicos C57BL , Proteínas Roundabout , Receptores de Superficie Celular
20.
Nat Commun ; 15(1): 5233, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38898031

RESUMEN

Mutations in the FOXF1 gene, a key transcriptional regulator of pulmonary vascular development, cause Alveolar Capillary Dysplasia with Misalignment of Pulmonary Veins, a lethal lung disease affecting newborns and infants. Identification of new FOXF1 upstream regulatory elements is critical to explain why frequent non-coding FOXF1 deletions are linked to the disease. Herein, we use multiome single-nuclei RNA and ATAC sequencing of mouse and human patient lungs to identify four conserved endothelial and mesenchymal FOXF1 enhancers. We demonstrate that endothelial FOXF1 enhancers are autoactivated, whereas mesenchymal FOXF1 enhancers are regulated by EBF1 and GLI1. The cell-specificity of FOXF1 enhancers is validated by disrupting these enhancers in mouse embryonic stem cells using CRISPR/Cpf1 genome editing followed by lineage-tracing of mutant embryonic stem cells in mouse embryos using blastocyst complementation. This study resolves an important clinical question why frequent non-coding FOXF1 deletions that interfere with endothelial and mesenchymal enhancers can lead to the disease.


Asunto(s)
Elementos de Facilitación Genéticos , Factores de Transcripción Forkhead , Mesodermo , Síndrome de Circulación Fetal Persistente , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Animales , Humanos , Síndrome de Circulación Fetal Persistente/genética , Síndrome de Circulación Fetal Persistente/patología , Síndrome de Circulación Fetal Persistente/metabolismo , Ratones , Elementos de Facilitación Genéticos/genética , Mesodermo/metabolismo , Mesodermo/embriología , Pulmón/patología , Células Endoteliales/metabolismo , Proteína con Dedos de Zinc GLI1/genética , Proteína con Dedos de Zinc GLI1/metabolismo , Células Madre Embrionarias/metabolismo , Alveolos Pulmonares/anomalías
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA