Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.018
Filter
1.
Biol Res ; 57(1): 22, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38704609

ABSTRACT

BACKGROUND: Chromatin dynamics is deeply involved in processes that require access to DNA, such as transcriptional regulation. Among the factors involved in chromatin dynamics at gene regulatory regions are general regulatory factors (GRFs). These factors contribute to establishment and maintenance of nucleosome-depleted regions (NDRs). These regions are populated by nucleosomes through histone deposition and nucleosome sliding, the latter catalyzed by a number of ATP-dependent chromatin remodeling complexes, including ISW1a. It has been observed that GRFs can act as barriers against nucleosome sliding towards NDRs. However, the relative ability of the different GRFs to hinder sliding activity is currently unknown. RESULTS: Considering this, we performed a comparative analysis for the main GRFs, with focus in their ability to modulate nucleosome sliding mediated by ISW1a. Among the GRFs tested in nucleosome remodeling assays, Rap1 was the only factor displaying the ability to hinder the activity of ISW1a. This effect requires location of the Rap1 cognate sequence on linker that becomes entry DNA in the nucleosome remodeling process. In addition, Rap1 was able to hinder nucleosome assembly in octamer transfer assays. Concurrently, Rap1 displayed the highest affinity for and longest dwell time from its target sequence, compared to the other GRFs tested. Consistently, through bioinformatics analyses of publicly available genome-wide data, we found that nucleosome occupancy and histone deposition in vivo are inversely correlated with the affinity of Rap1 for its target sequences in the genome. CONCLUSIONS: Our findings point to DNA binding affinity, residence time and location at particular translational positions relative to the nucleosome core as the key features of GRFs underlying their roles played in nucleosome sliding and assembly.


Subject(s)
Chromatin Assembly and Disassembly , DNA-Binding Proteins , Nucleosomes , Nucleosomes/metabolism , Nucleosomes/genetics , Chromatin Assembly and Disassembly/physiology , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Histones/metabolism
2.
Cell Mol Life Sci ; 80(2): 52, 2023 Jan 25.
Article in English | MEDLINE | ID: mdl-36695883

ABSTRACT

One of the major mysteries in science is how it is possible to pack the cellular chromatin with a total length of over 1 m, into a small sphere with a diameter of 5 mm "the nucleus", and even more difficult to envisage how to make it functional. Although we know that compaction is achieved through the histones, however, the DNA needs to be accessible to the transcription machinery and this is allowed thanks to a variety of very complex epigenetic mechanisms. Either DNA (methylation) or post-translational modifications of histone proteins (acetylation, methylation, ubiquitination and sumoylation) play a crucial role in chromatin remodelling and consequently on gene expression. Recently the serotonylation and dopaminylation of the histone 3, catalyzed by the Transglutaminase type 2 (TG2), has been reported. These novel post-translational modifications catalyzed by a predominantly cytoplasmic enzyme opens a new avenue for future investigations on the enzyme function itself and for the possibility that other biological amines, substrate of TG2, can influence the genome regulation under peculiar cellular conditions. In this review we analyzed the nuclear TG2's biology by discussing both its post-translational modification of various transcription factors and the implications of its epigenetic new face. Finally, we will focus on the potential impact of these events in human diseases.


Subject(s)
Chromatin Assembly and Disassembly , Cytoplasm , Epigenesis, Genetic , Histones , Transglutaminases , Humans , Acetylation , Chromatin , DNA/genetics , DNA Methylation , Histones/metabolism , Protein Processing, Post-Translational , Transglutaminases/genetics , Transglutaminases/metabolism , Cytoplasm/enzymology , Cytoplasm/genetics , Cytoplasm/metabolism , Cell Nucleus/enzymology , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/physiology
3.
Epigenetics Chromatin ; 15(1): 41, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36544209

ABSTRACT

BACKGROUND: Regulatory elements such as promoters, enhancers, and insulators interact each other to mediate molecular processes. To capture chromatin interactions of regulatory elements, 3C-derived methods such as Hi-C and Micro-C are developed. Here, we generated and analyzed Hi-C, Micro-C, and promoter capture Micro-C datasets with different sequencing depths to study chromatin interactions of regulatory elements and nucleosome positions in human prostate cancer cells. RESULTS: Compared to Hi-C, Micro-C identifies more high-resolution loops, including ones around structural variants. By evaluating the effect of sequencing depth, we revealed that more than 2 billion reads of Micro-C are needed to detect chromatin interactions at 1 kb resolution. Moreover, we found that deep-sequencing identifies additional long-range loops that are longer than 1 Mb in distance. Furthermore, we found that more than 50% of the loops are involved in insulators while less than 10% of the loops are promoter-enhancer loops. To comprehensively capture chromatin interactions that promoters are involved in, we performed promoter capture Micro-C. Promoter capture Micro-C identifies loops near promoters with a lower amount of sequencing reads. Sequencing of 160 million reads of promoter capture Micro-C resulted in reaching a plateau of identifying loops. However, there was still a subset of promoters that are not involved in loops even after deep-sequencing. By integrating Micro-C with NOMe-seq and ChIP-seq, we found that active promoters involved in loops have a more accessible region with lower levels of DNA methylation and more highly phased nucleosomes, compared to active promoters that are not involved in loops. CONCLUSION: We determined the required sequencing depth for Micro-C and promoter capture Micro-C to generate high-resolution chromatin interaction maps and loops. We also investigated the effect of sequencing coverage of Hi-C, Micro-C, and promoter capture Micro-C on detecting chromatin loops. Our analyses suggest the presence of distinct regulatory element groups, which are differently involved in nucleosome positions and chromatin interactions. This study does not only provide valuable insights on understanding chromatin interactions of regulatory elements, but also present guidelines for designing research projects on chromatin interactions among regulatory elements.


Subject(s)
Chromatin Assembly and Disassembly , Nucleosomes , Regulatory Sequences, Nucleic Acid , Humans , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/physiology , Enhancer Elements, Genetic , Nucleosomes/genetics , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid/genetics
4.
Sci Rep ; 12(1): 15636, 2022 09 17.
Article in English | MEDLINE | ID: mdl-36115870

ABSTRACT

Mutations in the chromodomain helicase DNA binding protein 2 (CHD2) gene are associated with neurodevelopmental disorders. However, mechanisms by which CHD2 regulates human brain development remain largely uncharacterized. Here, we used a human embryonic stem cell model of cortical interneuron (hcIN) development to elucidate its roles in this process. We identified genome-wide CHD2 binding profiles during hcIN differentiation, defining direct CHD2 targets related to neurogenesis in hcIN progenitors and to neuronal function in hcINs. CHD2 bound sites were frequently coenriched with histone H3 lysine 27 acetylation (H3K27ac) and associated with high gene expression, indicating roles for CHD2 in promoting gene expression during hcIN development. Binding sites for different classes of transcription factors were enriched at CHD2 bound regions during differentiation, suggesting transcription factors that may cooperatively regulate stage-specific gene expression with CHD2. We also demonstrated that CHD2 haploinsufficiency altered CHD2 and H3K27ac coenrichment on chromatin and expression of associated genes, decreasing acetylation and expression of cell cycle genes while increasing acetylation and expression of neuronal genes, to cause precocious differentiation. Together, these data describe CHD2 direct targets and mechanisms by which CHD2 prevents precocious hcIN differentiation, which are likely to be disrupted by pathogenic CHD2 mutation to cause neurodevelopmental disorders.


Subject(s)
Cerebral Cortex , Chromatin Assembly and Disassembly , DNA-Binding Proteins , Interneurons , Neurogenesis , Cerebral Cortex/metabolism , Cerebral Cortex/physiology , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histones/genetics , Histones/metabolism , Humans , Interneurons/metabolism , Interneurons/physiology , Lysine/metabolism , Neurogenesis/genetics , Neurogenesis/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Nat Struct Mol Biol ; 29(2): 121-129, 2022 02.
Article in English | MEDLINE | ID: mdl-35173352

ABSTRACT

Chromatin remodelers are ATP-dependent enzymes that reorganize nucleosomes within all eukaryotic genomes. Here we report a complex of the Chd1 remodeler bound to a nucleosome in a nucleotide-free state, determined by cryo-EM to 2.3 Å resolution. The remodeler stimulates the nucleosome to absorb an additional nucleotide on each strand at two different locations: on the tracking strand within the ATPase binding site and on the guide strand one helical turn from the ATPase motor. Remarkably, the additional nucleotide on the tracking strand is associated with a local transformation toward an A-form geometry, explaining how sequential ratcheting of each DNA strand occurs. The structure also reveals a histone-binding motif, ChEx, which can block opposing remodelers on the nucleosome and may allow Chd1 to participate in histone reorganization during transcription.


Subject(s)
DNA, Fungal/chemistry , DNA, Fungal/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Nucleosomes/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Binding Sites , Chromatin Assembly and Disassembly/physiology , Cryoelectron Microscopy , DNA-Binding Proteins/genetics , Models, Biological , Models, Molecular , Molecular Motor Proteins/chemistry , Molecular Motor Proteins/metabolism , Nucleosomes/chemistry , Nucleotides/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics
6.
Sci Rep ; 12(1): 2188, 2022 02 09.
Article in English | MEDLINE | ID: mdl-35140280

ABSTRACT

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


Subject(s)
DNA Transposable Elements/physiology , Enhancer Elements, Genetic/physiology , Influenza A virus/genetics , RNA/physiology , Chromatin Assembly and Disassembly/physiology , Gene Expression Regulation , Host Microbial Interactions/genetics , Humans
7.
Clin Transl Med ; 12(2): e746, 2022 02.
Article in English | MEDLINE | ID: mdl-35220680

ABSTRACT

BACKGROUND: Aging-associated osteoporosis is frequently seen in the elderly in clinic, but efficient managements are limited because of unclear nosogenesis. The current study aims to investigate the role of melatonin on senescent bone marrow stromal cells (BMSCs) and the underlying regulating mechanism. METHODS: Melatonin levels were tested by ELISA. Gene expression profiles were performed by RNA-sequencing, enrichment of H3K36me2 on gene promoters was analyzed by Chromatin Immunoprecipitation Sequencing (ChIP-seq), and chromatin accessibility was determined by Assay for Transposase-Accessible Chromatin with high-throughput sequencing (ATAC-seq). Osteogenesis of BMSCs in vitro was measured by Alizarin Red and Alkaline Phosphatase staining, and in vivo effects of melatonin was assessed by histological staining and micro computed tomography (micro-CT) scan. Correlation of NSD2 expression and severity of senile osteoporosis patients were analyzed by Pearson correlation. RESULTS: Melatonin levels were decreased during aging in human bone marrow, accompanied by downregulation of the histone methyltransferase nuclear receptor binding SET domain protein 2 (NSD2) expression in the senescent BMSCs. Melatonin stimulated the expression of NSD2 through MT1/2-mediated signaling pathways, resulting in the rebalancing of H3K36me2 and H3K27me3 modifications to increase chromatin accessibility of the osteogenic genes, runt-related transcription factor 2 (RUNX2) and bone gamma-carboxyglutamate protein (BGLAP). Melatonin promoted osteogenesis of BMSCs in vitro, and alleviates osteoporosis progression in the aging mice. In clinic, severity of senile osteoporosis (SOP) was negatively correlated with melatonin level in bone marrow, as well as NSD2 expression in BMSCs. Similarly, melatonin remarkably enhanced osteogenic differentiation of BMSCs derived from SOP patients in vitro. CONCLUSIONS: Collectively, our study dissects previously unreported mechanistic insights into the epigenetic regulating machinery of melatonin in meliorating osteogenic differentiation of senescent BMSC, and provides evidence for application of melatonin in preventing aging-associated bone loss.


Subject(s)
Chromatin Assembly and Disassembly/drug effects , Histone-Lysine N-Methyltransferase/pharmacology , Melatonin/metabolism , Mesenchymal Stem Cells/drug effects , Osteoblasts/drug effects , Repressor Proteins/pharmacology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Cell Differentiation/drug effects , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/physiology , Disease Models, Animal , Female , Histone-Lysine N-Methyltransferase/metabolism , Humans , Male , Melatonin/therapeutic use , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL/metabolism , Middle Aged , Osteoblasts/physiology , Repressor Proteins/metabolism
8.
Mol Biol Cell ; 33(2): rt1, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35076260

ABSTRACT

Formation of a bipolar spindle is required for the faithful segregation of chromosomes during cell division. Twenty-five years ago, a transformative insight into how bipolarity is achieved was provided by Rebecca Heald, Eric Karsenti, and colleagues in their landmark publication characterizing a chromatin-mediated spindle assembly pathway in which centrosomes and kinetochores were dispensable. The discovery revealed that bipolar spindle assembly is a self-organizing process where microtubules, which possess an intrinsic polarity, polymerize around chromatin and become sorted by mitotic motors into a bipolar structure. On the 25th anniversary of this seminal paper, we discuss what was known before, what we have learned since, and what may lie ahead in understanding the bipolar spindle.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Chromatin/metabolism , Spindle Apparatus/physiology , Animals , Anniversaries and Special Events , Cell Cycle , Centrosome , Humans , Kinetochores , Microtubules/metabolism , Mitosis
9.
Nat Rev Genet ; 23(3): 137-153, 2022 03.
Article in English | MEDLINE | ID: mdl-34608297

ABSTRACT

DNA methylation (DNAme) and histone post-translational modifications (PTMs) have important roles in transcriptional regulation. Although many reports have characterized the functions of such chromatin marks in isolation, recent genome-wide studies reveal surprisingly complex interactions between them. Here, we focus on the interplay between DNAme and methylation of specific lysine residues on the histone H3 tail. We describe the impact of genetic perturbation of the relevant methyltransferases in the mouse on the landscape of chromatin marks as well as the transcriptome. In addition, we discuss the specific neurodevelopmental growth syndromes and cancers resulting from pathogenic mutations in the human orthologues of these genes. Integrating these observations underscores the fundamental importance of crosstalk between DNA and histone H3 methylation in development and disease.


Subject(s)
Chromatin/metabolism , DNA Methylation/genetics , Disease/genetics , Growth and Development/genetics , Animals , Chromatin Assembly and Disassembly/physiology , Histones/metabolism , Humans , Mice , Protein Processing, Post-Translational
10.
Mol Cell Biol ; 42(2): e0066920, 2022 02 17.
Article in English | MEDLINE | ID: mdl-34898280

ABSTRACT

Nucleophosmin (NPM1) is a multifunctional histone chaperone that can activate acetylation-dependent transcription from chromatin templates in vitro. p300-mediated acetylation of NPM1 has been shown to further enhance its transcription activation potential. Acetylated and total NPM1 pools are increased in oral squamous cell carcinoma. However, the role of NPM1 or its acetylated form (AcNPM1) in transcriptional regulation in cells and oral tumorigenesis is not fully elucidated. Using ChIP-seq analyses, we provide the first genome-wide profile of AcNPM1 and show that AcNPM1 is enriched at transcriptional regulatory elements. AcNPM1 co-occupies marks of active transcription at promoters and DNase I hypersensitive sites at enhancers. In addition, using a high-throughput protein interaction profiling approach, we show that NPM1 interacts with RNA Pol II, general transcription factors, mediator subunits, histone acetyltransferase complexes, and chromatin remodelers. NPM1 histone chaperone activity also contributes to its transcription activation potential. Further, NPM1 depletion leads to decreased AcNPM1 occupancy and reduced expression of genes required for proliferative, migratory and invasive potential of oral cancer cells. NPM1 depletion also abrogates the growth of orthotopic tumors in mice. Collectively, these results establish that AcNPM1 functions as a coactivator during during RNA polymerase II-driven transcription and regulates the expression of genes that promote oral tumorigenesis.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Gene Expression Regulation/physiology , Histone Chaperones/metabolism , Mouth Neoplasms/genetics , Nucleophosmin/metabolism , Animals , Carcinogenesis/metabolism , Carcinoma, Squamous Cell/genetics , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/physiology , Gene Expression Regulation/genetics , Histones/metabolism , Humans , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics
11.
Life Sci Alliance ; 4(11)2021 11.
Article in English | MEDLINE | ID: mdl-34493579

ABSTRACT

Telomeres, highly ordered DNA-protein complexes at eukaryotic linear chromosome ends, are specialized heterochromatin loci conserved among eukaryotes. In Schizosaccharomyces pombe, the shelterin complex is important for subtelomeric heterochromatin establishment. Despite shelterin has been demonstrated to mediate the recruitment of the Snf2/histone deacetylase-containing repressor complex (SHREC) and the Clr4 methyltransferase complex (CLRC) to telomeres, the mechanism involved in telomeric heterochromatin assembly remains elusive due to the multiple functions of the shelterin complex. Here, we found that CLRC plays a dominant role in heterochromatin establishment at telomeres. In addition, we identified a series of amino acids in the shelterin subunit Ccq1 that are important for the specific interaction between Ccq1 and the CLRC subunit Raf2. Finally, we demonstrated that the Ccq1-Raf2 interaction is essential for the recruitment of CLRC to telomeres, that contributes to histone H3 lysine 9 methylation, nucleosome stability and the shelterin-chromatin association, promoting a positive feedback mechanism for the nucleation and spreading of heterochromatin at subtelomeres. Together, our findings provide a mechanistic understanding of subtelomeric heterochromatin assembly by shelterin-dependent CLRC recruitment to chromosomal ends.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Schizosaccharomyces pombe Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromatin Assembly and Disassembly/physiology , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Methylation , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/genetics , Shelterin Complex/metabolism , Shelterin Complex/physiology , Telomere/metabolism
12.
Nat Commun ; 12(1): 5240, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34475390

ABSTRACT

ß-actin is a crucial component of several chromatin remodeling complexes that control chromatin structure and accessibility. The mammalian Brahma-associated factor (BAF) is one such complex that plays essential roles in development and differentiation by regulating the chromatin state of critical genes and opposing the repressive activity of polycomb repressive complexes (PRCs). While previous work has shown that ß-actin loss can lead to extensive changes in gene expression and heterochromatin organization, it is not known if changes in ß-actin levels can directly influence chromatin remodeling activities of BAF and polycomb proteins. Here we conduct a comprehensive genomic analysis of ß-actin knockout mouse embryonic fibroblasts (MEFs) using ATAC-Seq, HiC-seq, RNA-Seq and ChIP-Seq of various epigenetic marks. We demonstrate that ß-actin levels can induce changes in chromatin structure by affecting the complex interplay between chromatin remodelers such as BAF/BRG1 and EZH2. Our results show that changes in ß-actin levels and associated chromatin remodeling activities can not only impact local chromatin accessibility but also induce reversible changes in 3D genome architecture. Our findings reveal that ß-actin-dependent chromatin remodeling plays a role in shaping the chromatin landscape and influences the regulation of genes involved in development and differentiation.


Subject(s)
Actins/metabolism , Chromatin Assembly and Disassembly/physiology , Actins/genetics , Animals , Chromatin/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic , Fibroblasts , Gene Dosage , Gene Knockout Techniques , Histones/metabolism , Mice , Nuclear Proteins/metabolism , Polycomb-Group Proteins/metabolism , Protein Binding , Transcription Factors/metabolism
13.
Mol Cell ; 81(17): 3560-3575.e6, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34375585

ABSTRACT

Transcription initiation by RNA polymerase II (RNA Pol II) requires preinitiation complex (PIC) assembly at gene promoters. In the dynamic nucleus, where thousands of promoters are broadly distributed in chromatin, it is unclear how multiple individual components converge on any target to establish the PIC. Here we use live-cell, single-molecule tracking in S. cerevisiae to visualize constrained exploration of the nucleoplasm by PIC components and Mediator's key role in guiding this process. On chromatin, TFIID/TATA-binding protein (TBP), Mediator, and RNA Pol II instruct assembly of a short-lived PIC, which occurs infrequently but efficiently within a few seconds on average. Moreover, PIC exclusion by nucleosome encroachment underscores regulated promoter accessibility by chromatin remodeling. Thus, coordinated nuclear exploration and recruitment to accessible targets underlies dynamic PIC establishment in yeast. Our study provides a global spatiotemporal model for transcription initiation in live cells.


Subject(s)
Mediator Complex/metabolism , RNA Polymerase II/metabolism , Transcription Initiation, Genetic/physiology , Chromatin/metabolism , Chromatin Assembly and Disassembly/physiology , Mediator Complex/genetics , Nucleosomes/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Spatio-Temporal Analysis , TATA-Box Binding Protein/genetics , Transcription Factor TFIID/genetics , Transcription, Genetic/genetics
14.
Nat Plants ; 7(9): 1213-1219, 2021 09.
Article in English | MEDLINE | ID: mdl-34354260

ABSTRACT

To compensate for a sessile nature, plants have developed sophisticated mechanisms to sense varying environmental conditions. Phytochromes (phys) are light and temperature sensors that regulate downstream genes to render plants responsive to environmental stimuli1-4. Here, we show that phyB directly triggers the formation of a repressive chromatin loop by physically interacting with VERNALIZATION INSENSITIVE 3-LIKE1/VERNALIZATION 5 (VIL1/VRN5), a component of Polycomb Repressive Complex 2 (PRC2)5,6, in a light-dependent manner. VIL1 and phyB cooperatively contribute to the repression of growth-promoting genes through the enrichment of Histone H3 Lys27 trimethylation (H3K27me3), a repressive histone modification. In addition, phyB and VIL1 mediate the formation of a chromatin loop to facilitate the repression of ATHB2. Our findings show that phyB directly utilizes chromatin remodelling to regulate the expression of target genes in a light-dependent manner.


Subject(s)
Acclimatization/genetics , Adaptation, Ocular/genetics , Chromatin Assembly and Disassembly/genetics , Homeodomain Proteins/metabolism , Phytochrome B/metabolism , Polycomb-Group Proteins/metabolism , Stress, Physiological/genetics , Arabidopsis/genetics , Chromatin Assembly and Disassembly/physiology , Gene Expression Regulation, Plant , Genes, Plant , Genetic Variation , Genotype , Homeodomain Proteins/genetics , Mutation , PHD Zinc Fingers/genetics , PHD Zinc Fingers/physiology , Phytochrome B/genetics , Polycomb-Group Proteins/genetics , Stress, Physiological/physiology
15.
STAR Protoc ; 2(3): 100706, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34401775

ABSTRACT

We describe a fluorescence recovery after photobleaching (FRAP) protocol for assessing the dynamics of heterochromatin/euchromatin and identifying chromatin relaxers for cell fate transition. Here, we developed a system to track heterochromatin foci with HP1α-cherry and performed FRAP assay of H1-GFP to analyze the dynamics of heterochromatin and euchromatin during somatic cell reprogramming. This protocol is used to screen factors that impact chromatin structure, which could also be used to identify chromatin relaxers and repressors in various cell fate transitions. For complete details on the use and execution of this protocol, please refer to Chen et al. (2016) and Chen et al. (2020).


Subject(s)
Chromatin Assembly and Disassembly/drug effects , Drug Evaluation, Preclinical/methods , Fluorescence Recovery After Photobleaching/methods , Animals , Cell Line , Chromatin , Chromatin Assembly and Disassembly/physiology , Chromosomal Proteins, Non-Histone/metabolism , Euchromatin , Fibroblasts/metabolism , Heterochromatin , Histones/genetics , Mice , NIH 3T3 Cells
17.
Nat Commun ; 12(1): 4057, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34210977

ABSTRACT

Chromatin remodeler ALC1 (amplification in liver cancer 1) is crucial for repairing damaged DNA. It is autoinhibited and activated by nucleosomal epitopes. However, the mechanisms by which ALC1 is regulated remain unclear. Here we report the crystal structure of human ALC1 and the cryoEM structure bound to the nucleosome. The structure shows the macro domain of ALC1 binds to lobe 2 of the ATPase motor, sequestering two elements for nucleosome recognition, explaining the autoinhibition mechanism of the enzyme. The H4 tail competes with the macro domain for lobe 2-binding, explaining the requirement for this nucleosomal epitope for ALC1 activation. A dual-arginine-anchor motif of ALC1 recognizes the acidic pocket of the nucleosome, which is critical for chromatin remodeling in vitro. Together, our findings illustrate the structures of ALC1 and shed light on its regulation mechanisms, paving the way for the discovery of drugs targeting ALC1 for the treatment of cancer.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Crystallography, X-Ray/methods , DNA Helicases/metabolism , DNA-Binding Proteins/chemistry , Liver Neoplasms/pathology , Nucleosomes/metabolism , Recombinant Proteins/chemistry , Cells, Cultured , DNA Helicases/chemistry , DNA Repair , DNA-Binding Proteins/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Protein Domains , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Structure-Activity Relationship
18.
Nat Genet ; 53(8): 1196-1206, 2021 08.
Article in English | MEDLINE | ID: mdl-34253920

ABSTRACT

To systematically define molecular features in human tumor cells that determine their degree of sensitivity to human allogeneic natural killer (NK) cells, we quantified the NK cell responsiveness of hundreds of molecularly annotated 'DNA-barcoded' solid tumor cell lines in multiplexed format and applied genome-scale CRISPR-based gene-editing screens in several solid tumor cell lines, to functionally interrogate which genes in tumor cells regulate the response to NK cells. In these orthogonal studies, NK cell-sensitive tumor cells tend to exhibit 'mesenchymal-like' transcriptional programs; high transcriptional signature for chromatin remodeling complexes; high levels of B7-H6 (NCR3LG1); and low levels of HLA-E/antigen presentation genes. Importantly, transcriptional signatures of NK cell-sensitive tumor cells correlate with immune checkpoint inhibitor (ICI) resistance in clinical samples. This study provides a comprehensive map of mechanisms regulating tumor cell responses to NK cells, with implications for future biomarker-driven applications of NK cell immunotherapies.


Subject(s)
Cytotoxicity, Immunologic/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Immune Checkpoint Inhibitors/pharmacology , Killer Cells, Natural/physiology , Allogeneic Cells/physiology , Animals , B7 Antigens/genetics , Cell Line, Tumor , Chromatin Assembly and Disassembly/physiology , Cytotoxicity Tests, Immunologic/methods , Cytotoxicity, Immunologic/physiology , Drug Resistance, Neoplasm/drug effects , Female , Genome, Human , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Mice, Inbred NOD , Xenograft Model Antitumor Assays , HLA-E Antigens
19.
PLoS Comput Biol ; 17(7): e1009253, 2021 07.
Article in English | MEDLINE | ID: mdl-34314440

ABSTRACT

The organization of nucleosomes along the Eukaryotic genome is maintained over time despite disruptive events such as replication. During this complex process, histones and DNA can form a variety of non-canonical nucleosome conformations, but their precise molecular details and roles during nucleosome assembly remain unclear. In this study, employing coarse-grained molecular dynamics simulations and Markov state modeling, we characterized the complete kinetics of nucleosome assembly. On the nucleosome-positioning 601 DNA sequence, we observe a rich transition network among various canonical and non-canonical tetrasome, hexasome, and nucleosome conformations. A low salt environment makes nucleosomes stable, but the kinetic landscape becomes more rugged, so that the system is more likely to be trapped in off-pathway partially assembled intermediates. Finally, we find that the co-operativity between DNA bending and histone association enables positioning sequence motifs to direct the assembly process, with potential implications for the dynamic organization of nucleosomes on real genomic sequences.


Subject(s)
Nucleosomes/metabolism , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/physiology , Computational Biology , Kinetics , Markov Chains , Models, Biological , Molecular Dynamics Simulation , Nucleic Acid Conformation , Nucleosomes/chemistry , Nucleosomes/genetics , Protein Conformation , Sodium Chloride/metabolism
20.
Mutat Res Rev Mutat Res ; 787: 108346, 2021.
Article in English | MEDLINE | ID: mdl-34083038

ABSTRACT

DNA replication stress is a major source of DNA damage, including double-stranded breaks that promote DNA damage response (DDR) signaling. Inefficient repair of such lesions can affect genome integrity. During DNA replication different factors act on chromatin remodeling in a coordinated way. While recent studies have highlighted individual molecular mechanisms of interaction, less is known about the orchestration of chromatin changes under replication stress. In this review we attempt to explore the complex relationship between DNA replication stress, DDR and genome integrity in mammalian cells, taking into account the role of chromatin disposition as an important modulator of DNA repair. Recent data on chromatin restoration and epigenetic re-establishment after DNA replication stress are reviewed.


Subject(s)
DNA Damage/physiology , DNA Replication/physiology , Genomic Instability/physiology , Animals , Chromatin/metabolism , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/physiology , DNA Damage/genetics , DNA Replication/genetics , Genomic Instability/genetics , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...