Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.747
Filter
1.
Nat Commun ; 15(1): 6562, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39095348

ABSTRACT

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.


Subject(s)
Chromatin , Gene Expression Regulation, Plant , Genome, Plant , Genome-Wide Association Study , Oryza , Oryza/genetics , Oryza/growth & development , Chromatin/metabolism , Chromatin/genetics , Chromosome Mapping/methods , Quantitative Trait Loci/genetics , Germination/genetics , Enhancer Elements, Genetic/genetics , Deep Learning , Plant Proteins/genetics , Plant Proteins/metabolism
2.
Cell Mol Life Sci ; 81(1): 361, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39158745

ABSTRACT

Genome-wide studies have demonstrated regulatory roles for diverse non-coding elements, but their precise and interrelated functions have often remained enigmatic. Addressing the need for mechanistic insight, we studied their roles in expression of Lhb which encodes the pituitary gonadotropic hormone that controls reproduction. We identified a bi-directional enhancer in gonadotrope-specific open chromatin, whose functional eRNA (eRNA2) supports permissive chromatin at the Lhb locus. The central untranscribed region of the enhancer contains an iMotif (iM), and is bound by Hmgb2 which stabilizes the iM and directs transcription specifically towards the functional eRNA2. A distinct downstream lncRNA, associated with an inducible G-quadruplex (G4) and iM, also facilitates Lhb expression, following its splicing in situ. GnRH activates Lhb transcription and increased levels of all three RNAs, eRNA2 showing the highest response, while estradiol, which inhibits Lhb, repressed levels of eRNA2 and the lncRNA. The levels of these regulatory RNAs and Lhb mRNA correlate highly in female mice, though strikingly not in males, suggesting a female-specific function. Our findings, which shed new light on the workings of non-coding elements and non-canonical DNA structures, reveal novel mechanisms regulating transcription which have implications not only in the central control of reproduction but also for other inducible genes.


Subject(s)
Enhancer Elements, Genetic , RNA, Long Noncoding , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Animals , Enhancer Elements, Genetic/genetics , Female , Male , Mice , Gene Expression Regulation , Mice, Inbred C57BL , Chromatin/metabolism , Chromatin/genetics , Humans , G-Quadruplexes
3.
Nat Commun ; 15(1): 7258, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39179577

ABSTRACT

The specificity of gene expression during development requires the insulation of regulatory domains to avoid inappropriate enhancer-gene interactions. In vertebrates, this insulator function is mostly attributed to clusters of CTCF sites located at topologically associating domain (TAD) boundaries. However, TAD boundaries allow some physical crosstalk across regulatory domains, which is at odds with the specific and precise expression of developmental genes. Here we show that developmental genes and nearby clusters of CTCF sites cooperatively foster the robust insulation of regulatory domains. By genetically dissecting a couple of representative loci in mouse embryonic stem cells, we show that CTCF sites prevent undesirable enhancer-gene contacts (i.e. physical insulation), while developmental genes preferentially contribute to regulatory insulation through non-structural mechanisms involving promoter competition rather than enhancer blocking. Overall, our work provides important insights into the insulation of regulatory domains, which in turn might help interpreting the pathological consequences of certain structural variants.


Subject(s)
CCCTC-Binding Factor , Enhancer Elements, Genetic , Mouse Embryonic Stem Cells , Promoter Regions, Genetic , Animals , CCCTC-Binding Factor/metabolism , CCCTC-Binding Factor/genetics , Mice , Promoter Regions, Genetic/genetics , Enhancer Elements, Genetic/genetics , Mouse Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Insulator Elements/genetics
4.
Nat Commun ; 15(1): 7324, 2024 Aug 25.
Article in English | MEDLINE | ID: mdl-39183203

ABSTRACT

During the progression of proliferative vitreoretinopathy (PVR) following ocular trauma, previously quiescent retinal pigment epithelial (RPE) cells transition into a state of rapid proliferation, migration, and secretion. The elusive molecular mechanisms behind these changes have hindered the development of effective pharmacological treatments, presenting a pressing clinical challenge. In this study, by monitoring the dynamic changes in chromatin accessibility and various histone modifications, we chart the comprehensive epigenetic landscape of RPE cells in male mice subjected to traumatic PVR. Coupled with transcriptomic analysis, we reveal a robust correlation between enhancer activation and the upregulation of the PVR-associated gene programs. Furthermore, by constructing transcription factor regulatory networks, we identify the aberrant activation of enhancer-driven RANK-NFATc1 pathway as PVR advanced. Importantly, we demonstrate that intraocular interventions, including nanomedicines inhibiting enhancer activity, gene therapies targeting NFATc1 and antibody therapeutics against RANK pathway, effectively mitigate PVR progression. Together, our findings elucidate the epigenetic basis underlying the activation of PVR-associated genes during RPE cell fate transitions and offer promising therapeutic avenues targeting epigenetic modulation and the RANK-NFATc1 axis for PVR management.


Subject(s)
NFATC Transcription Factors , Retinal Pigment Epithelium , Signal Transduction , Vitreoretinopathy, Proliferative , Animals , Vitreoretinopathy, Proliferative/metabolism , Vitreoretinopathy, Proliferative/genetics , Vitreoretinopathy, Proliferative/pathology , Retinal Pigment Epithelium/metabolism , NFATC Transcription Factors/metabolism , NFATC Transcription Factors/genetics , Mice , Male , Mice, Inbred C57BL , Humans , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Disease Models, Animal , Eye Injuries/metabolism , Eye Injuries/genetics , Eye Injuries/pathology , Gene Expression Profiling , Multiomics
5.
Nat Commun ; 15(1): 6852, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39127768

ABSTRACT

Cis-regulatory elements (CREs) are pivotal in orchestrating gene expression throughout diverse biological systems. Accurate identification and in-depth characterization of functional CREs are crucial for decoding gene regulation networks during cellular processes. In this study, we develop Kethoxal-Assisted Single-stranded DNA Assay for Transposase-Accessible Chromatin with Sequencing (KAS-ATAC-seq) to quantitatively analyze the transcriptional activity of CREs. A main advantage of KAS-ATAC-seq lies in its precise measurement of ssDNA levels within both proximal and distal ATAC-seq peaks, enabling the identification of transcriptional regulatory sequences. This feature is particularly adept at defining Single-Stranded Transcribing Enhancers (SSTEs). SSTEs are highly enriched with nascent RNAs and specific transcription factors (TFs) binding sites that define cellular identity. Moreover, KAS-ATAC-seq provides a detailed characterization and functional implications of various SSTE subtypes. Our analysis of CREs during mouse neural differentiation demonstrates that KAS-ATAC-seq can effectively identify immediate-early activated CREs in response to retinoic acid (RA) treatment. Our findings indicate that KAS-ATAC-seq provides more precise annotation of functional CREs in transcription. Future applications of KAS-ATAC-seq would help elucidate the intricate dynamics of gene regulation in diverse biological processes.


Subject(s)
Transcription Factors , Animals , Mice , Transcription Factors/metabolism , Transcription Factors/genetics , Transcription, Genetic , Enhancer Elements, Genetic/genetics , Chromatin/metabolism , Chromatin/genetics , Binding Sites , Humans , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Chromatin Immunoprecipitation Sequencing/methods , Transposases/metabolism , Transposases/genetics , Regulatory Elements, Transcriptional , Tretinoin/pharmacology , Tretinoin/metabolism , Gene Expression Regulation , Cell Differentiation/genetics , Sequence Analysis, DNA/methods , Regulatory Sequences, Nucleic Acid/genetics
6.
Int J Mol Sci ; 25(15)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39125583

ABSTRACT

Coronaviruses constitute a global threat to human and animal health. It is essential to investigate the long-distance RNA-RNA interactions that approximate remote regulatory elements in strategies, including genome circularization, discontinuous transcription, and transcriptional enhancers, aimed at the rapid replication of their large genomes, pathogenicity, and immune evasion. Based on the primary sequences and modeled RNA-RNA interactions of two experimentally defined coronaviral enhancers, we detected via an in silico primary and secondary structural analysis potential enhancers in various coronaviruses, from the phylogenetically ancient avian infectious bronchitis virus (IBV) to the recently emerged SARS-CoV-2. These potential enhancers possess a core duplex-forming region that could transition between closed and open states, as molecular switches directed by viral or host factors. The duplex open state would pair with remote sequences in the viral genome and modulate the expression of downstream crucial genes involved in viral replication and host immune evasion. Consistently, variations in the predicted IBV enhancer region or its distant targets coincide with cases of viral attenuation, possibly driven by decreased open reading frame (ORF)3a immune evasion protein expression. If validated experimentally, the annotated enhancer sequences could inform structural prediction tools and antiviral interventions.


Subject(s)
Enhancer Elements, Genetic , Genome, Viral , Infectious bronchitis virus , SARS-CoV-2 , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Infectious bronchitis virus/genetics , Humans , Enhancer Elements, Genetic/genetics , Animals , RNA, Viral/genetics , RNA, Viral/metabolism , COVID-19/virology , COVID-19/genetics , Betacoronavirus/genetics , Virus Replication/genetics , Coronavirus Infections/virology , Transcription, Genetic , Gene Expression Regulation, Viral , Pneumonia, Viral/virology
7.
Cell Metab ; 36(8): 1639-1641, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39111283

ABSTRACT

During aging, transcriptional programs of cell identity are partially eroded, reducing cellular fitness and resilience. Patrick et al.1 unveil a general mechanism for this process that consists of the progressive loss of transcription factor AP-1 from cell identity enhancers and its relocation by competition to stress-response elements.


Subject(s)
Chromatin , Transcription Factor AP-1 , Transcription Factor AP-1/metabolism , Chromatin/metabolism , Animals , Aging/metabolism , Humans , Enhancer Elements, Genetic/genetics , Cellular Senescence
8.
Article in English | MEDLINE | ID: mdl-39167800

ABSTRACT

Enhancers are DNA sequences that can strengthen transcription initiation. However, the global identification of plant enhancers is complicated due to uncertainty in the distance and orientation of enhancers, especially in species with large genomes. In this study, we performed self-transcribing active regulatory region sequencing (STARR-seq) for the first time to identify enhancers across the barley genome. A total of 7323 enhancers were successfully identified, and among 45 randomly selected enhancers, over 75% were effective as validated by a dual-luciferase reporter assay system in the lower epidermis of tobacco leaves. Interestingly, up to 53.5% of the barley enhancers were repetitive sequences, especially transposable elements (TEs), thus reinforcing the vital role of repetitive enhancers in gene expression. Both the common active mark H3K4me3 and repressive mark H3K27me3 were abundant among the barley STARR-seq enhancers. In addition, the functional range of barley STARR-seq enhancers seemed much broader than that of rice or maize and extended to ±100 kb of the gene body, and this finding was consistent with the high expression levels of genes in the genome. This study specifically depicts the unique features of barley enhancers and provides available barley enhancers for further utilization.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Plant , Hordeum , Hordeum/genetics , Hordeum/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Plant/genetics , Histones/metabolism , Histones/genetics , DNA Transposable Elements/genetics , Genome, Plant/genetics , Repetitive Sequences, Nucleic Acid/genetics , Sequence Analysis, DNA/methods
9.
J Immunol ; 213(3): 283-295, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39140825

ABSTRACT

The IL-7R regulates the homeostasis, activation, and distribution of T cells in peripheral tissues. Although several transcriptional enhancers that regulate IL-7Rα expression in αß T cells have been identified, enhancers active in γδ T cells remain unknown. In this article, we discovered an evolutionarily conserved noncoding sequence (CNS) in intron 2 of the IL-7Rα-chain (IL-7Rα) locus and named this region CNS9. CNS9 contained a conserved retinoic acid receptor-related orphan receptor (ROR)-responsive element (RORE) and exerted RORγt-dependent enhancer activity in vitro. Mice harboring point mutations in the RORE in CNS9 (CNS9-RORmut) showed reduced IL-7Rα expression in IL-17-producing Vγ4+ γδ T cells. In addition, the cell number and IL-17A production of Vγ4+ γδ T cells were reduced in the adipose tissue of CNS9-RORmut mice. Consistent with the reduction in IL-17A, CNS9-RORmut mice exhibited decreased IL-33 expression in the adipose tissue, resulting in fewer regulatory T cells and glucose intolerance. The CNS9-ROR motif was partially responsible for IL-7Rα expression in RORγt+ regulatory T cells, whereas IL-7Rα expression was unaffected in RORγt-expressing Vγ2+ γδ T cells, Th17 cells, type 3 innate lymphoid cells, and invariant NKT cells. Our results indicate that CNS9 is a RORΕ-dependent, Vγ4+ γδ T cell-specific IL-7Rα enhancer that plays a critical role in adipose tissue homeostasis via regulatory T cells, suggesting that the evolutionarily conserved RORΕ in IL-7Rα intron 2 may influence the incidence of type 2 diabetes.


Subject(s)
Enhancer Elements, Genetic , Introns , Nuclear Receptor Subfamily 1, Group F, Member 3 , Receptors, Antigen, T-Cell, gamma-delta , Animals , Mice , Introns/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Enhancer Elements, Genetic/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Glucose/metabolism , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/metabolism , Mice, Inbred C57BL , Th17 Cells/immunology , Interleukin-17/metabolism , Interleukin-17/genetics , Humans , Adipose Tissue/metabolism , Adipose Tissue/immunology
10.
Cell Death Dis ; 15(8): 566, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39107271

ABSTRACT

Super-enhancers are a class of DNA cis-regulatory elements that can regulate cell identity, cell fate, stem cell pluripotency, and even tumorigenesis. Increasing evidence shows that epigenetic modifications play an important role in the pathogenesis of various types of cancer. However, the current research is far from enough to reveal the complex mechanism behind it. This study found a super-enhancer enriched with abnormally active histone modifications in pancreatic ductal adenocarcinoma (PDAC), called DKK1-super-enhancer (DKK1-SE). The major active component of DKK1-SE is component enhancer e1. Mechanistically, AP1 induces chromatin remodeling in component enhancer e1 and activates the transcriptional activity of DKK1. Moreover, DKK1 was closely related to the malignant clinical features of PDAC. Deletion or knockdown of DKK1-SE significantly inhibited the proliferation, colony formation, motility, migration, and invasion of PDAC cells in vitro, and these phenomena were partly mitigated upon rescuing DKK1 expression. In vivo, DKK1-SE deficiency not only inhibited tumor proliferation but also reduced the complexity of the tumor microenvironment. This study identifies that DKK1-SE drives DKK1 expression by recruiting AP1 transcription factors, exerting oncogenic effects in PDAC, and enhancing the complexity of the tumor microenvironment.


Subject(s)
Cell Proliferation , Disease Progression , Intercellular Signaling Peptides and Proteins , Pancreatic Neoplasms , Transcription Factor AP-1 , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Animals , Transcription Factor AP-1/metabolism , Cell Line, Tumor , Mice , Gene Expression Regulation, Neoplastic , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , Cell Movement/genetics , Tumor Microenvironment , Male , Mice, Nude , Enhancer Elements, Genetic/genetics , Female
11.
Nat Commun ; 15(1): 5610, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38969652

ABSTRACT

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.


Subject(s)
Cell Lineage , GATA3 Transcription Factor , Immunity, Innate , Lymphocytes , Animals , GATA3 Transcription Factor/metabolism , GATA3 Transcription Factor/genetics , Mice , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/cytology , Mice, Inbred C57BL , Mice, Knockout , Enhancer Elements, Genetic/genetics , Th2 Cells/immunology , Cell Differentiation/immunology , Single-Cell Analysis
12.
Development ; 151(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38994775

ABSTRACT

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.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Homeodomain Proteins , Retina , Transcription Factors , Animals , Humans , Mice , Enhancer Elements, Genetic/genetics , Evolution, Molecular , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Microphthalmos/genetics , Microphthalmos/pathology , Neurogenesis/genetics , Organoids/metabolism , Retina/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics
13.
Genes (Basel) ; 15(7)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-39062635

ABSTRACT

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.


Subject(s)
Enhancer Elements, Genetic , Polymorphism, Single Nucleotide , Humans , Enhancer Elements, Genetic/genetics , Alleles , Haplotypes , Genome, Human , Transcription Factors/genetics , Binding Sites
14.
Nat Commun ; 15(1): 6130, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39033128

ABSTRACT

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.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Genomics , Oncogenes , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc , Receptor, ErbB-2 , Humans , Enhancer Elements, Genetic/genetics , Cell Line, Tumor , Promoter Regions, Genetic/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Genomics/methods , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology
15.
Nat Commun ; 15(1): 6464, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39085231

ABSTRACT

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.


Subject(s)
Algorithms , Conserved Sequence , Enhancer Elements, Genetic , Animals , Enhancer Elements, Genetic/genetics , Humans , Mice , Evolution, Molecular , Binding Sites/genetics , Mammals/genetics , Promoter Regions, Genetic/genetics , Computational Biology/methods , CCCTC-Binding Factor/metabolism , CCCTC-Binding Factor/genetics
16.
Cell Rep ; 43(7): 114456, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38990722

ABSTRACT

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.


Subject(s)
Enhancer Elements, Genetic , Promoter Regions, Genetic , YY1 Transcription Factor , Animals , Male , Mice , Cell Differentiation , Enhancer Elements, Genetic/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/metabolism , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , YY1 Transcription Factor/metabolism , YY1 Transcription Factor/genetics , Female
17.
Front Immunol ; 15: 1387253, 2024.
Article in English | MEDLINE | ID: mdl-38947339

ABSTRACT

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.


Subject(s)
CD4-Positive T-Lymphocytes , Diabetes Mellitus, Type 1 , Enhancer Elements, Genetic , Genetic Predisposition to Disease , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Humans , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Enhancer Elements, Genetic/genetics , Suppressor of Cytokine Signaling 1 Protein/genetics , Genome-Wide Association Study , Lectins, C-Type/genetics , Genetic Variation , Polymorphism, Single Nucleotide , Quantitative Trait Loci
18.
Clin Epigenetics ; 16(1): 86, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965562

ABSTRACT

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.


Subject(s)
Aging , Chromatin , Cochlea , Enhancer Elements, Genetic , Presbycusis , Animals , Mice , Cochlea/metabolism , Cochlea/drug effects , Chromatin/genetics , Chromatin/metabolism , Aging/genetics , Presbycusis/genetics , Presbycusis/metabolism , Enhancer Elements, Genetic/genetics , Transcriptome/genetics , Disease Models, Animal , Epigenesis, Genetic/genetics , Histones/metabolism , Histones/genetics , High-Throughput Nucleotide Sequencing/methods , Male
19.
Nat Plants ; 10(8): 1246-1257, 2024 08.
Article in English | MEDLINE | ID: mdl-39080503

ABSTRACT

Unstable transcripts have emerged as markers of active enhancers in vertebrates and shown to be involved in many cellular processes and medical disorders. However, their prevalence and role in plants is largely unexplored. Here, we comprehensively captured all actively initiating (nascent) transcripts across diverse crops and other plants using capped small (cs)RNA sequencing. We discovered that unstable transcripts are rare in plants, unlike in vertebrates, and when present, often originate from promoters. In addition, many 'distal' elements in plants initiate tissue-specific stable transcripts and are likely bona fide promoters of as-yet-unannotated genes or non-coding RNAs, cautioning against using reference genome annotations to infer putative enhancer sites. To investigate enhancer function, we integrated data from self-transcribing active regulatory region (STARR) sequencing. We found that annotated promoters and other regions that initiate stable transcripts, but not those marked by unstable or bidirectional unstable transcripts, showed stronger enhancer activity in this assay. Our findings underscore the blurred line between promoters and enhancers and suggest that cis-regulatory elements can encompass diverse structures and mechanisms in eukaryotes, including humans.


Subject(s)
Enhancer Elements, Genetic , Promoter Regions, Genetic , RNA, Plant , Enhancer Elements, Genetic/genetics , RNA, Plant/genetics , RNA, Plant/metabolism , Gene Expression Regulation, Plant , RNA Stability/genetics , Plants/genetics , Sequence Analysis, RNA
20.
Adv Exp Med Biol ; 1459: 97-113, 2024.
Article in English | MEDLINE | ID: mdl-39017841

ABSTRACT

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.


Subject(s)
B-Lymphocytes , Homeodomain Proteins , Humans , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Enhancer Elements, Genetic/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chromatin Assembly and Disassembly , Cell Differentiation/genetics , Chromatin/metabolism , Chromatin/genetics , E1A-Associated p300 Protein/metabolism , E1A-Associated p300 Protein/genetics , DNA-Binding Proteins , Nuclear Proteins
SELECTION OF CITATIONS
SEARCH DETAIL