Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 879
Filter
1.
Nucleic Acids Res ; 52(W1): W45-W53, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38749504

ABSTRACT

ChIP-Atlas (https://chip-atlas.org/) presents a suite of data-mining tools for analyzing epigenomic landscapes, powered by the comprehensive integration of over 376 000 public ChIP-seq, ATAC-seq, DNase-seq and Bisulfite-seq experiments from six representative model organisms. To unravel the intricacies of chromatin architecture that mediates the regulome-initiated generation of transcriptional and phenotypic diversity within cells, we report ChIP-Atlas 3.0 that enhances clarity by incorporating additional tracks for genomic and epigenomic features within a newly consolidated 'annotation track' section. The tracks include chromosomal conformation (Hi-C and eQTL datasets), transcriptional regulatory elements (ChromHMM and FANTOM5 enhancers), and genomic variants associated with diseases and phenotypes (GWAS SNPs and ClinVar variants). These annotation tracks are easily accessible alongside other experimental tracks, facilitating better elucidation of chromatin architecture underlying the diversification of transcriptional and phenotypic traits. Furthermore, 'Diff Analysis,' a new online tool, compares the query epigenome data to identify differentially bound, accessible, and methylated regions using ChIP-seq, ATAC-seq and DNase-seq, and Bisulfite-seq datasets, respectively. The integration of annotation tracks and the Diff Analysis tool, coupled with continuous data expansion, renders ChIP-Atlas 3.0 a robust resource for mining the landscape of transcriptional regulatory mechanisms, thereby offering valuable perspectives, particularly for genetic disease research and drug discovery.


Subject(s)
Chromatin Immunoprecipitation Sequencing , Data Mining , Software , Humans , Data Mining/methods , Chromatin Immunoprecipitation Sequencing/methods , Animals , Chromatin/genetics , Chromatin/metabolism , Chromosomes/genetics , Epigenomics/methods , Polymorphism, Single Nucleotide , Mice , Quantitative Trait Loci , Molecular Sequence Annotation , Regulatory Elements, Transcriptional/genetics , Genomics/methods
2.
Bioessays ; 46(7): e2300210, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38715516

ABSTRACT

Understanding the influence of cis-regulatory elements on gene regulation poses numerous challenges given complexities stemming from variations in transcription factor (TF) binding, chromatin accessibility, structural constraints, and cell-type differences. This review discusses the role of gene regulatory networks in enhancing understanding of transcriptional regulation and covers construction methods ranging from expression-based approaches to supervised machine learning. Additionally, key experimental methods, including MPRAs and CRISPR-Cas9-based screening, which have significantly contributed to understanding TF binding preferences and cis-regulatory element functions, are explored. Lastly, the potential of machine learning and artificial intelligence to unravel cis-regulatory logic is analyzed. These computational advances have far-reaching implications for precision medicine, therapeutic target discovery, and the study of genetic variations in health and disease.


Subject(s)
CRISPR-Cas Systems , Gene Regulatory Networks , Machine Learning , Humans , CRISPR-Cas Systems/genetics , Computational Biology/methods , Transcription Factors/metabolism , Transcription Factors/genetics , Gene Expression Regulation/genetics , Animals , Regulatory Elements, Transcriptional/genetics
3.
Sci Adv ; 10(21): eadj4452, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38781344

ABSTRACT

Most genetic variants associated with psychiatric disorders are located in noncoding regions of the genome. To investigate their functional implications, we integrate epigenetic data from the PsychENCODE Consortium and other published sources to construct a comprehensive atlas of candidate brain cis-regulatory elements. Using deep learning, we model these elements' sequence syntax and predict how binding sites for lineage-specific transcription factors contribute to cell type-specific gene regulation in various types of glia and neurons. The elements' evolutionary history suggests that new regulatory information in the brain emerges primarily via smaller sequence mutations within conserved mammalian elements rather than entirely new human- or primate-specific sequences. However, primate-specific candidate elements, particularly those active during fetal brain development and in excitatory neurons and astrocytes, are implicated in the heritability of brain-related human traits. Additionally, we introduce PsychSCREEN, a web-based platform offering interactive visualization of PsychENCODE-generated genetic and epigenetic data from diverse brain cell types in individuals with psychiatric disorders and healthy controls.


Subject(s)
Brain , Epigenesis, Genetic , Regulatory Sequences, Nucleic Acid , Humans , Brain/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Animals , Evolution, Molecular , Mental Disorders/genetics , Regulatory Elements, Transcriptional/genetics , Neurons/metabolism , Gene Expression Regulation , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Nature ; 613(7942): 96-102, 2023 01.
Article in English | MEDLINE | ID: mdl-36517591

ABSTRACT

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


Subject(s)
DNA Repeat Expansion , Genome, Human , Neoplasms , Humans , Base Sequence , DNA Repeat Expansion/genetics , Genome, Human/genetics , Neoplasms/classification , Neoplasms/genetics , Neoplasms/pathology , Sequence Analysis, DNA , Gene Expression Regulation , Regulatory Elements, Transcriptional/genetics , Introns/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Proliferation/drug effects , Reproducibility of Results
5.
BMC Genomics ; 23(1): 714, 2022 Oct 19.
Article in English | MEDLINE | ID: mdl-36261804

ABSTRACT

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


Subject(s)
Genome, Human , Regulatory Elements, Transcriptional , Humans , Mice , Animals , Regulatory Elements, Transcriptional/genetics , Binding Sites/genetics , Protein Binding , Transcription Factors/genetics , Transcription Factors/metabolism , Mammals/genetics
6.
Database (Oxford) ; 20222022 04 22.
Article in English | MEDLINE | ID: mdl-35452518

ABSTRACT

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


Subject(s)
Regulatory Elements, Transcriptional , Transcription Factors , Animals , Binding Sites , Genome/genetics , Mice , Protein Binding , Regulatory Elements, Transcriptional/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Int J Mol Sci ; 22(21)2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34768856

ABSTRACT

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


Subject(s)
Endometriosis/genetics , Gene Expression Regulation/genetics , RNA, Long Noncoding/genetics , Regulatory Elements, Transcriptional/genetics , Chromatin/genetics , Endometriosis/pathology , Female , Gene Expression Profiling , Humans , Polymorphism, Single Nucleotide/genetics , Signal Transduction/genetics , Transcriptome/genetics
8.
Nat Commun ; 12(1): 6749, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34799566

ABSTRACT

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


Subject(s)
Gene Expression Regulation, Developmental , Gene Regulatory Networks , Human Embryonic Stem Cells/physiology , Hypothalamus/embryology , Neurons/physiology , Cell Differentiation/genetics , Cell Line , Chromosome Mapping , Genome-Wide Association Study , Humans , Hypothalamus/cytology , Multifactorial Inheritance , RNA-Seq , Regulatory Elements, Transcriptional/genetics
9.
Nat Struct Mol Biol ; 28(12): 989-996, 2021 12.
Article in English | MEDLINE | ID: mdl-34811519

ABSTRACT

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


Subject(s)
Gene Expression Regulation/genetics , Histone Acetyltransferases/metabolism , Regulatory Elements, Transcriptional/genetics , Transcription, Genetic/genetics , Adaptor Proteins, Signal Transducing/metabolism , Binding Sites , Cell Line, Tumor , Chromatin/metabolism , Cryoelectron Microscopy , HeLa Cells , Humans , Nuclear Proteins/metabolism , Phytic Acid/metabolism , Promoter Regions, Genetic/genetics , Protein Conformation , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism , Saccharomycetales
10.
Mol Cell ; 81(16): 3368-3385.e9, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34375583

ABSTRACT

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


Subject(s)
Adenosine/analogs & derivatives , Cell Cycle Proteins/genetics , Nerve Tissue Proteins/genetics , RNA Splicing Factors/genetics , RNA/genetics , Transcription Factors/genetics , Adenosine/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation/genetics , Humans , Methylation , Regulatory Elements, Transcriptional/genetics , Transcriptional Activation/genetics
11.
Nat Commun ; 12(1): 4897, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34385432

ABSTRACT

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


Subject(s)
Cell Differentiation/genetics , Gene Expression Profiling/methods , Genome, Human/genetics , Human Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Regulatory Elements, Transcriptional/genetics , Binding Sites/genetics , Blotting, Western , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Cell Line , Enhancer Elements, Genetic/genetics , Human Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA/methods
12.
Commun Biol ; 4(1): 675, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34083716

ABSTRACT

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


Subject(s)
Chromatin/metabolism , Cytoskeleton/metabolism , Podocytes/metabolism , Receptors, Glucocorticoid/metabolism , Transcription, Genetic , A549 Cells , Binding Sites/genetics , Cell Line , Cells, Cultured , Chromatin/genetics , Chromatin Immunoprecipitation Sequencing/methods , Glucocorticoids/pharmacology , HeLa Cells , Humans , K562 Cells , MCF-7 Cells , Podocytes/cytology , Podocytes/drug effects , Protein Binding , Receptors, Glucocorticoid/genetics , Regulatory Elements, Transcriptional/genetics , Transcriptome/genetics
13.
Nat Commun ; 12(1): 3376, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099660

ABSTRACT

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


Subject(s)
Adaptation, Physiological/genetics , Arabidopsis/genetics , Evolution, Molecular , Gene Expression Regulation, Plant , Regulatory Elements, Transcriptional/genetics , Droughts , Genes, Plant , Plant Breeding , Species Specificity
14.
Cell Death Dis ; 12(6): 544, 2021 05 26.
Article in English | MEDLINE | ID: mdl-34039951

ABSTRACT

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


Subject(s)
Endometrial Neoplasms/drug therapy , Endometrium/abnormalities , NF-kappa B/drug effects , Pyridines/therapeutic use , Regulatory Elements, Transcriptional/genetics , Sterol Regulatory Element Binding Protein 1/drug effects , Thiazoles/therapeutic use , Uterine Diseases/diet therapy , Female , Humans , Pyridines/pharmacology , Thiazoles/pharmacology , Transfection
15.
Cell ; 184(11): 3041-3055.e21, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33964211

ABSTRACT

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


Subject(s)
Gene Expression Regulation, Plant/genetics , Regulatory Elements, Transcriptional/genetics , Zea mays/genetics , Arabidopsis/genetics , Gene Expression/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Plant/physiology , Gene Regulatory Networks/genetics , Genome , Genomics , Regulatory Elements, Transcriptional/physiology , Single-Cell Analysis , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome/genetics
16.
Nat Neurosci ; 24(7): 941-953, 2021 07.
Article in English | MEDLINE | ID: mdl-34017130

ABSTRACT

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


Subject(s)
Gene Expression Regulation, Developmental/genetics , Genetic Variation/genetics , Mental Disorders/genetics , Neurons/physiology , Quantitative Trait Loci/genetics , Cell Differentiation/physiology , Chromatin/genetics , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Humans , Neural Stem Cells/physiology , Neurogenesis/genetics , Regulatory Elements, Transcriptional/genetics , Transcription Factors/genetics
17.
Brain ; 144(10): 3005-3019, 2021 11 29.
Article in English | MEDLINE | ID: mdl-33950254

ABSTRACT

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


Subject(s)
Genetic Therapy/methods , Methyl-CpG-Binding Protein 2/administration & dosage , MicroRNAs/administration & dosage , Protein Engineering/methods , Regulatory Elements, Transcriptional , Rett Syndrome/therapy , Animals , Humans , Injections, Spinal , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Knockout , MicroRNAs/genetics , Regulatory Elements, Transcriptional/genetics , Rett Syndrome/genetics
18.
PLoS One ; 16(3): e0242949, 2021.
Article in English | MEDLINE | ID: mdl-33750972

ABSTRACT

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


Subject(s)
Arachis/genetics , CCAAT-Binding Factor/genetics , Plant Proteins/genetics , 5' Untranslated Regions , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Arachis/growth & development , CCAAT-Binding Factor/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Gene Expression Regulation, Plant/radiation effects , Glucuronidase/genetics , Glucuronidase/metabolism , Light , Plant Proteins/metabolism , Plants, Genetically Modified/metabolism , Promoter Regions, Genetic , Protein Subunits/genetics , Regulatory Elements, Transcriptional/genetics , Seeds/genetics
19.
Mol Cell ; 81(8): 1601-1616, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33770487

ABSTRACT

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


Subject(s)
Base Pairing/genetics , Chromosomes/genetics , Transcription, Genetic/genetics , Animals , Gene Expression Regulation/genetics , Humans , Regulatory Elements, Transcriptional/genetics
20.
Mol Biol Cell ; 32(9): 942-955, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33788585

ABSTRACT

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


Subject(s)
Cell Nucleus Structures/metabolism , Histones/metabolism , Microscopy/methods , Animals , Carrier Proteins/metabolism , Cell Nucleus/metabolism , Cell Nucleus Structures/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Mitosis , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Regulatory Elements, Transcriptional/genetics , Ribonucleoprotein, U7 Small Nuclear/metabolism , Tumor Suppressor Proteins/metabolism , Zygote/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...