Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.016
Filter
1.
Sci Rep ; 14(1): 17786, 2024 08 01.
Article in English | MEDLINE | ID: mdl-39090226

ABSTRACT

A long-standing question concerns the role of Z-DNA in transcription. Here we use a deep learning approach DeepZ that predicts Z-flipons based on DNA sequence, structural properties of nucleotides and omics data. We examined Z-flipons that are conserved between human and mouse genomes after generating whole-genome Z-flipon maps and then validated them by orthogonal approaches based on high resolution chemical mapping of Z-DNA and the transformer algorithm Z-DNABERT. For human and mouse, we revealed similar pattern of transcription factors, chromatin remodelers, and histone marks associated with conserved Z-flipons. We found significant enrichment of Z-flipons in alternative and bidirectional promoters associated with neurogenesis genes. We show that conserved Z-flipons are associated with increased experimentally determined transcription reinitiation rates compared to promoters without Z-flipons, but without affecting elongation or pausing. Our findings support a model where Z-flipons engage Transcription Factor E and impact phenotype by enabling the reset of preinitiation complexes when active, and the suppression of gene expression when engaged by repressive chromatin complexes.


Subject(s)
DNA , Promoter Regions, Genetic , Animals , Humans , Mice , DNA/genetics , DNA/metabolism , Transcription, Genetic , Transcription Factors/metabolism , Transcription Factors/genetics , Chromatin Assembly and Disassembly , Transcription Initiation, Genetic , Chromatin/genetics , Chromatin/metabolism , Deep Learning , Conserved Sequence
2.
Med Oncol ; 41(9): 221, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117768

ABSTRACT

Cancer is characterized by metabolic reprogramming in cancer cells, which is crucial for tumorigenesis. The highly deregulated chromatin remodeler MORC2 contributes to cell proliferation, invasion, migration, DNA repair, and chemoresistance. MORC2 also plays a key role in metabolic reprogramming, including lipogenesis, glucose, and glutamine metabolism. A recent study showed that MORC2-regulated glucose metabolism affects the expression of E-cadherin, a crucial protein in the epithelial-to-mesenchymal transition. This review discusses recent developments in MORC2 regulated cancer cell metabolism and its role in cancer progression.


Subject(s)
Chromatin Assembly and Disassembly , Neoplasms , Transcription Factors , Humans , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/genetics , Transcription Factors/metabolism , Epithelial-Mesenchymal Transition , Animals
3.
Mol Cell ; 84(16): 3061-3079.e10, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39121853

ABSTRACT

Mouse FOXA1 and GATA4 are prototypes of pioneer factors, initiating liver cell development by binding to the N1 nucleosome in the enhancer of the ALB1 gene. Using cryoelectron microscopy (cryo-EM), we determined the structures of the free N1 nucleosome and its complexes with FOXA1 and GATA4, both individually and in combination. We found that the DNA-binding domains of FOXA1 and GATA4 mainly recognize the linker DNA and an internal site in the nucleosome, respectively, whereas their intrinsically disordered regions interact with the acidic patch on histone H2A-H2B. FOXA1 efficiently enhances GATA4 binding by repositioning the N1 nucleosome. In vivo DNA editing and bioinformatics analyses suggest that the co-binding mode of FOXA1 and GATA4 plays important roles in regulating genes involved in liver cell functions. Our results reveal the mechanism whereby FOXA1 and GATA4 cooperatively bind to the nucleosome through nucleosome repositioning, opening chromatin by bending linker DNA and obstructing nucleosome packing.


Subject(s)
Cryoelectron Microscopy , GATA4 Transcription Factor , Hepatocyte Nuclear Factor 3-alpha , Nucleosomes , Protein Binding , Hepatocyte Nuclear Factor 3-alpha/metabolism , Hepatocyte Nuclear Factor 3-alpha/genetics , Nucleosomes/metabolism , Nucleosomes/genetics , Nucleosomes/ultrastructure , Animals , GATA4 Transcription Factor/metabolism , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/chemistry , Mice , Chromatin/metabolism , Chromatin/genetics , Histones/metabolism , Histones/genetics , Histones/chemistry , Binding Sites , DNA/metabolism , DNA/genetics , DNA/chemistry , Chromatin Assembly and Disassembly , Humans
4.
Science ; 385(6711): eadl5816, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39088653

ABSTRACT

The human nucleosome acetyltransferase of histone H4 (NuA4)/Tat-interactive protein, 60 kilodalton (TIP60) coactivator complex, a fusion of the yeast switch/sucrose nonfermentable related 1 (SWR1) and NuA4 complexes, both incorporates the histone variant H2A.Z into nucleosomes and acetylates histones H4, H2A, and H2A.Z to regulate gene expression and maintain genome stability. Our cryo-electron microscopy studies show that, within the NuA4/TIP60 complex, the E1A binding protein P400 (EP400) subunit serves as a scaffold holding the different functional modules in specific positions, creating a distinct arrangement of the actin-related protein (ARP) module. EP400 interacts with the transformation/transcription domain-associated protein (TRRAP) subunit by using a footprint that overlaps with that of the Spt-Ada-Gcn5 acetyltransferase (SAGA) complex, preventing the formation of a hybrid complex. Loss of the TRRAP subunit leads to mislocalization of NuA4/TIP60, resulting in the redistribution of H2A.Z and its acetylation across the genome, emphasizing the dual functionality of NuA4/TIP60 as a single macromolecular assembly.


Subject(s)
Chromatin Assembly and Disassembly , Lysine Acetyltransferase 5 , Humans , Acetylation , Adaptor Proteins, Signal Transducing , Cryoelectron Microscopy , DNA-Binding Proteins/chemistry , Histones/chemistry , Lysine Acetyltransferase 5/chemistry , Nuclear Proteins/chemistry , Nucleosomes/chemistry , Nucleosomes/ultrastructure , Protein Domains , Transcription Factors/chemistry
5.
Mol Cell ; 84(16): 3011-3025.e7, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39116874

ABSTRACT

The histone variant macroH2A is generally linked to transcriptionally inactive chromatin, but how macroH2A regulates chromatin structure and functions in the transcriptional process remains elusive. This study reveals that while the integration of human macroH2A1.2 into nucleosomes does not affect their stability or folding dynamics, it notably hinders the maintenance of facilitates chromatin transcription's (FACT's) function. We show that FACT effectively diminishes the stability of macroH2A1.2-nucleosomes and expedites their depletion subsequent to the initial unfolding process. Furthermore, we identify the residue S139 in macroH2A1.2 as a critical switch to modulate FACT's function in nucleosome maintenance. Genome-wide analyses demonstrate that FACT-mediated depletion of macroH2A-nucleosomes allows the correct localization of macroH2A, while the S139 mutation reshapes macroH2A distribution and influences stimulation-induced transcription and cellular response in macrophages. Our findings provide mechanistic insights into the intricate interplay between macroH2A and FACT at the nucleosome level and elucidate their collective role in transcriptional regulation and immune response of macrophages.


Subject(s)
Histones , Nucleosomes , Transcription, Genetic , Transcriptional Elongation Factors , Humans , Nucleosomes/metabolism , Nucleosomes/genetics , Histones/metabolism , Histones/genetics , Transcriptional Elongation Factors/genetics , Transcriptional Elongation Factors/metabolism , High Mobility Group Proteins/metabolism , High Mobility Group Proteins/genetics , Animals , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Macrophages/metabolism , Mutation , Chromatin Assembly and Disassembly , Mice , Chromatin/metabolism , Chromatin/genetics , Gene Expression Regulation , RAW 264.7 Cells , Protein Binding , HEK293 Cells
7.
Nat Commun ; 15(1): 7303, 2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39181868

ABSTRACT

Genes encoding subunits of SWI/SNF (BAF) chromatin remodeling complexes are mutated in nearly 25% of cancers. To gain insight into the mechanisms by which SWI/SNF mutations drive cancer, we contributed ten rhabdoid tumor (RT) cell lines mutant for SWI/SNF subunit SMARCB1 to a genome-scale CRISPR-Cas9 depletion screen performed across 896 cell lines. We identify PHF6 as specifically essential for RT cell survival and demonstrate that dependency on Phf6 extends to Smarcb1-deficient cancers in vivo. As mutations in either SWI/SNF or PHF6 can cause the neurodevelopmental disorder Coffin-Siris syndrome, our findings of a dependency suggest a previously unrecognized functional link. We demonstrate that PHF6 co-localizes with SWI/SNF complexes at promoters, where it is essential for maintenance of an active chromatin state. We show that in the absence of SMARCB1, PHF6 loss disrupts the recruitment and stability of residual SWI/SNF complex members, collectively resulting in the loss of active chromatin at promoters and stalling of RNA Polymerase II progression. Our work establishes a mechanistic basis for the shared syndromic features of SWI/SNF and PHF6 mutations in CSS and the basis for selective dependency on PHF6 in SMARCB1-mutant cancers.


Subject(s)
Micrognathism , Promoter Regions, Genetic , Repressor Proteins , Rhabdoid Tumor , SMARCB1 Protein , Transcription Factors , Humans , SMARCB1 Protein/metabolism , SMARCB1 Protein/genetics , Rhabdoid Tumor/genetics , Rhabdoid Tumor/metabolism , Rhabdoid Tumor/pathology , Micrognathism/genetics , Micrognathism/metabolism , Cell Line, Tumor , Transcription Factors/metabolism , Transcription Factors/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Promoter Regions, Genetic/genetics , Face/abnormalities , Chromatin/metabolism , Intellectual Disability/genetics , Intellectual Disability/metabolism , Chromatin Assembly and Disassembly , Mutation , Foot Deformities, Congenital/genetics , Foot Deformities, Congenital/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , CRISPR-Cas Systems , Transcription, Genetic , Animals , Mice , Abnormalities, Multiple , Hand Deformities, Congenital , Neck/abnormalities
8.
Nat Commun ; 15(1): 6569, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39095374

ABSTRACT

Liquid-liquid phase separation (LLPS) facilitates the formation of membraneless organelles within cells, with implications in various biological processes and disease states. AT-rich interactive domain-containing protein 1A (ARID1A) is a chromatin remodeling factor frequently associated with cancer mutations, yet its functional mechanism remains largely unknown. Here, we find that ARID1A harbors a prion-like domain (PrLD), which facilitates the formation of liquid condensates through PrLD-mediated LLPS. The nuclear condensates formed by ARID1A LLPS are significantly elevated in Ewing's sarcoma patient specimen. Disruption of ARID1A LLPS results in diminished proliferative and invasive abilities in Ewing's sarcoma cells. Through genome-wide chromatin structure and transcription profiling, we identify that the ARID1A condensate localizes to EWS/FLI1 target enhancers and induces long-range chromatin architectural changes by forming functional chromatin remodeling hubs at oncogenic target genes. Collectively, our findings demonstrate that ARID1A promotes oncogenic potential through PrLD-mediated LLPS, offering a potential therapeutic approach for treating Ewing's sarcoma.


Subject(s)
Chromatin Assembly and Disassembly , DNA-Binding Proteins , RNA-Binding Protein EWS , Sarcoma, Ewing , Transcription Factors , Humans , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/pathology , Transcription Factors/metabolism , Transcription Factors/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Cell Line, Tumor , RNA-Binding Protein EWS/metabolism , RNA-Binding Protein EWS/genetics , Gene Expression Regulation, Neoplastic , Cell Proliferation , Oncogene Proteins, Fusion/metabolism , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Protein c-fli-1/metabolism , Proto-Oncogene Protein c-fli-1/genetics , Chromatin/metabolism , Carcinogenesis/genetics , Animals , Mice , Protein Domains , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Phase Separation
9.
Cell Death Dis ; 15(8): 589, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138148

ABSTRACT

It is still challenging to predict the efficacy of cisplatin-based therapy, particularly in relation to the activation of macroautophagy/autophagy in oral squamous cell carcinoma (OSCC). We studied the effect of selected chromatin remodeling genes on the cisplatin resistance and their interplay with autophagy in 3-dimensional tumor model and xenografts. We analyzed gene expression patterns in the cisplatin-sensitive UMSCC1, and a paired cisplatin-resistant UM-Cis cells. Many histone protein gene clusters involved in nucleosome assembly showed significant difference of expression. Gain- and loss-of-function analyses revealed an inverse correlation between cisplatin resistance and HIST1H3D expression, while a positive correlation was observed with HIST3H2A or HIST3H2B expression. In UM-Cis, HIST3H2A- and HIST3H2B-mediated chromatin remodeling upregulates autophagy status, which results in cisplatin resistance. Additionally, knockdown of HIST3H2A or HIST3H2B downregulated autophagy-activating genes via chromatin compaction of their promoter regions. MiTF, one of the key autophagy regulators upregulated in UM-Cis, negatively regulated transcription of HIST1H3D, suggesting an interplay between chromatin remodeling-dependent cisplatin resistance and autophagy. On comparing the staining intensity between cisplatin-sensitive and -insensitive tissues from OSCC patients, protein expression pattern of the selected histone protein genes were matched with the in vitro data. By examining the relationship between autophagy and chromatin remodeling genes, we identified a set of candidate genes with potential use as markers predicting chemoresistance in OSCC biopsy samples.


Subject(s)
Autophagy , Carcinoma, Squamous Cell , Chromatin Assembly and Disassembly , Cisplatin , Drug Resistance, Neoplasm , Mouth Neoplasms , Cisplatin/pharmacology , Cisplatin/therapeutic use , Humans , Autophagy/drug effects , Autophagy/genetics , Drug Resistance, Neoplasm/genetics , Chromatin Assembly and Disassembly/drug effects , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/drug therapy , Animals , Cell Line, Tumor , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Mice , Histones/metabolism , Mice, Nude , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Xenograft Model Antitumor Assays
10.
Methods Mol Biol ; 2842: 103-127, 2024.
Article in English | MEDLINE | ID: mdl-39012592

ABSTRACT

Epigenome editing applications are gaining broader use for targeted transcriptional control as more enzymes with diverse chromatin-modifying functions are being incorporated into fusion proteins. Development of these fusion proteins, called epigenome editors, has outpaced the study of proteins that interact with edited chromatin. One type of protein that acts downstream of chromatin editing is the reader-effector, which bridges epigenetic marks with biological effects like gene regulation. As the name suggests, a reader-effector protein is generally composed of a reader domain and an effector domain. Reader domains directly bind epigenetic marks, while effector domains often recruit protein complexes that mediate transcription, chromatin remodeling, and DNA repair. In this chapter, we discuss the role of reader-effectors in driving the outputs of epigenome editing and highlight instances where abnormal and context-specific reader-effectors might impair the effects of epigenome editing. Lastly, we discuss how engineered reader-effectors may complement the epigenome editing toolbox to achieve robust and reliable gene regulation.


Subject(s)
Epigenesis, Genetic , Epigenome , Gene Editing , Animals , Humans , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , CRISPR-Cas Systems , Epigenomics/methods , Gene Editing/methods , Gene Expression Regulation
11.
Nat Commun ; 15(1): 6226, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39043639

ABSTRACT

Fluctuations in the initiation rate of transcription, the first step in gene expression, ensue from the stochastic behavior of the molecular process that controls transcription. In steady state, the regulatory process is often assumed to operate reversibly, i.e., in equilibrium. However, reversibility imposes fundamental limits to information processing. For instance, the assumption of equilibrium is difficult to square with the precision with which the regulatory process executes its task in eukaryotes. Here we provide evidence - from microscopic analyses of the transcription dynamics at a single gene copy of yeast - that the regulatory process for transcription is cyclic and irreversible (out of equilibrium). The necessary coupling to reservoirs of free energy occurs via sequence-specific transcriptional activators and the recruitment, in part, of ATP-dependent chromatin remodelers. Our findings may help explain how eukaryotic cells reconcile the dual but opposing requirements for fast regulatory kinetics and high regulatory specificity.


Subject(s)
Gene Expression Regulation, Fungal , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Transcription, Genetic , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Chromatin Assembly and Disassembly , Transcription Factors/metabolism , Transcription Factors/genetics , Kinetics , Adenosine Triphosphate/metabolism
12.
Int J Mol Sci ; 25(14)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39063093

ABSTRACT

Higher plants have developed complex mechanisms to adapt to fluctuating environmental conditions with light playing a vital role in photosynthesis and influencing various developmental processes, including photomorphogenesis. Exposure to ultraviolet (UV) radiation can cause cellular damage, necessitating effective DNA repair mechanisms. Histone acetyltransferases (HATs) play a crucial role in regulating chromatin structure and gene expression, thereby contributing to the repair mechanisms. HATs facilitate chromatin relaxation, enabling transcriptional activation necessary for plant development and stress responses. The intricate relationship between HATs, light signaling pathways and chromatin dynamics has been increasingly understood, providing valuable insights into plant adaptability. This review explores the role of HATs in plant photomorphogenesis, chromatin remodeling and gene regulation, highlighting the importance of chromatin modifications in plant responses to light and various stressors. It emphasizes the need for further research on individual HAT family members and their interactions with other epigenetic factors. Advanced genomic approaches and genome-editing technologies offer promising avenues for enhancing crop resilience and productivity through targeted manipulation of HAT activities. Understanding these mechanisms is essential for developing strategies to improve plant growth and stress tolerance, contributing to sustainable agriculture in the face of a changing climate.


Subject(s)
Gene Expression Regulation, Plant , Histone Acetyltransferases , Plant Development , Ultraviolet Rays , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Plant Development/genetics , Plant Development/radiation effects , Plants/genetics , Plants/radiation effects , Plants/metabolism , Chromatin Assembly and Disassembly , Chromatin/metabolism , Chromatin/genetics , Morphogenesis/radiation effects , Morphogenesis/genetics
13.
Cell Rep ; 43(7): 114458, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38996070

ABSTRACT

Regulatory T (Treg) cells play a critical regulatory role in the immune system by suppressing excessive immune responses and maintaining immune balance. The effective migration of Treg cells is crucial for controlling the development and progression of inflammatory diseases. However, the mechanisms responsible for directing Treg cells into the inflammatory tissue remain incompletely elucidated. In this study, we identified BAF60b, a subunit of switch/sucrose nonfermentable (SWI/SNF) chromatin remodeling complexes, as a positive regulator of Treg cell migration that inhibits the progression of inflammation in experimental autoimmune encephalomyelitis (EAE) and colitis animal models. Mechanistically, transcriptome and genome-wide chromatin-landscaped analyses demonstrated that BAF60b interacts with the transcription factor RUNX1 to promote the expression of CCR9 on Treg cells, which in turn affects their ability to migrate to inflammatory tissues. Our work provides insights into the essential role of BAF60b in regulating Treg cell migration and its impact on inflammatory diseases.


Subject(s)
Cell Movement , Inflammation , Mice, Inbred C57BL , T-Lymphocytes, Regulatory , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Mice , Inflammation/pathology , Inflammation/metabolism , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Humans , Transcription Factors/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Colitis/metabolism , Colitis/pathology , Colitis/immunology , Colitis/genetics
14.
Proc Natl Acad Sci U S A ; 121(31): e2402944121, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39052837

ABSTRACT

In eukaryotes, repetitive DNA can become silenced de novo, either transcriptionally or post-transcriptionally, by processes independent of strong sequence-specific cues. The mechanistic nature of such processes remains poorly understood. We found that in the fungus Neurospora crassa, de novo initiation of both transcriptional and post-transcriptional silencing was linked to perturbed chromatin, which was produced experimentally by the aberrant activity of transcription factors at the tetO operator array. Transcriptional silencing was mediated by canonical constitutive heterochromatin. On the other hand, post-transcriptional silencing resembled repeat-induced quelling but occurred normally when homologous recombination was inactivated. All silencing of the tetO array was dependent on SAD-6, fungal ortholog of the SWI/SNF chromatin remodeler ATRX (Alpha Thalassemia/Mental Retardation Syndrome X-Linked), which was required to maintain nucleosome occupancy at the perturbed locus. In addition, we found that two other types of sequences (the lacO array and native AT-rich DNA) could also undergo recombination-independent quelling associated with perturbed chromatin. These results suggested a model in which the de novo initiation of transcriptional and post-transcriptional silencing is coupled to the remodeling of perturbed chromatin.


Subject(s)
Chromatin Assembly and Disassembly , Gene Silencing , Neurospora crassa , Transcription, Genetic , Neurospora crassa/genetics , Neurospora crassa/metabolism , Chromatin/metabolism , Chromatin/genetics , Heterochromatin/metabolism , Heterochromatin/genetics , Fungal Proteins/metabolism , Fungal Proteins/genetics , Gene Expression Regulation, Fungal , Transcription Factors/metabolism , Transcription Factors/genetics , Nucleosomes/metabolism , Nucleosomes/genetics
15.
JCI Insight ; 9(15)2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38954484

ABSTRACT

Upon infection, naive CD8+ T cells differentiate into cytotoxic effector cells to eliminate the pathogen-infected cells. Although many mechanisms underlying this process have been demonstrated, the regulatory role of chromatin remodeling system in this process remains largely unknown. Here we show that BRD7, a component of the polybromo-associated BAF complex (PBAF), was required for naive CD8+ T cells to differentiate into functional short-lived effector cells (SLECs) in response to acute infections caused by influenza virus or lymphocytic choriomeningitis virus (LCMV). BRD7 deficiency in CD8+ T cells resulted in profound defects in effector population and functions, thereby impairing viral clearance and host recovery. Further mechanical studies indicate that the expression of BRD7 significantly turned to high from naive CD8+ T cells to effector cells, which bridged BRG1 and PBRM1 to the core module of PBAF complex, consequently facilitating the assembly of PBAF complex rather than BAF complex in the effector cells. The PBAF complex changed the chromatin accessibility at the loci of Tbx21 gene and upregulated its expression, leading to the maturation of effector T cells. Our research demonstrates that BRD7 and the PBAF complex are key in CD8+ T cell development and present a significant target for advancing immune therapies.


Subject(s)
CD8-Positive T-Lymphocytes , Cell Differentiation , Chromosomal Proteins, Non-Histone , Lymphocytic choriomeningitis virus , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mice , Cell Differentiation/immunology , Cell Differentiation/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomal Proteins, Non-Histone/genetics , Lymphocytic choriomeningitis virus/immunology , Transcription Factors/genetics , Transcription Factors/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Chromatin Assembly and Disassembly , Bromodomain Containing Proteins
16.
Nat Neurosci ; 27(7): 1260-1273, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38956165

ABSTRACT

Direct neuronal reprogramming is a promising approach to regenerate neurons from local glial cells. However, mechanisms of epigenome remodeling and co-factors facilitating this process are unclear. In this study, we combined single-cell multiomics with genome-wide profiling of three-dimensional nuclear architecture and DNA methylation in mouse astrocyte-to-neuron reprogramming mediated by Neurogenin2 (Ngn2) and its phosphorylation-resistant form (PmutNgn2), respectively. We show that Ngn2 drives multilayered chromatin remodeling at dynamic enhancer-gene interaction sites. PmutNgn2 leads to higher reprogramming efficiency and enhances epigenetic remodeling associated with neuronal maturation. However, the differences in binding sites or downstream gene activation cannot fully explain this effect. Instead, we identified Yy1, a transcriptional co-factor recruited by direct interaction with Ngn2 to its target sites. Upon deletion of Yy1, activation of neuronal enhancers, genes and ultimately reprogramming are impaired without affecting Ngn2 binding. Thus, our work highlights the key role of interactors of proneural factors in direct neuronal reprogramming.


Subject(s)
Astrocytes , Basic Helix-Loop-Helix Transcription Factors , Cellular Reprogramming , Nerve Tissue Proteins , Neurons , YY1 Transcription Factor , Animals , YY1 Transcription Factor/metabolism , YY1 Transcription Factor/genetics , Astrocytes/metabolism , Mice , Cellular Reprogramming/physiology , Neurons/metabolism , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Epigenome , Chromatin Assembly and Disassembly , Epigenesis, Genetic , Cells, Cultured
17.
Nat Commun ; 15(1): 5187, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992002

ABSTRACT

The histone H2A variant H2A.W occupies transposons and thus prevents access to them in Arabidopsis thaliana. H2A.W is deposited by the chromatin remodeler DDM1, which also promotes the accessibility of chromatin writers to heterochromatin by an unknown mechanism. To shed light on this question, we solve the cryo-EM structures of nucleosomes containing H2A and H2A.W, and the DDM1-H2A.W nucleosome complex. These structures show that the DNA end flexibility of the H2A nucleosome is higher than that of the H2A.W nucleosome. In the DDM1-H2A.W nucleosome complex, DDM1 binds to the N-terminal tail of H4 and the nucleosomal DNA and increases the DNA end flexibility of H2A.W nucleosomes. Based on these biochemical and structural results, we propose that DDM1 counters the low accessibility caused by nucleosomes containing H2A.W to enable the maintenance of repressive epigenetic marks on transposons and prevent their activity.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Chromatin Assembly and Disassembly , Cryoelectron Microscopy , Histones , Nucleosomes , Arabidopsis/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/chemistry , Nucleosomes/metabolism , Nucleosomes/ultrastructure , Nucleosomes/chemistry , Histones/metabolism , Histones/genetics , Histones/chemistry , Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Factors/chemistry , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Protein Binding , Models, Molecular , DNA, Plant/metabolism , DNA, Plant/genetics
18.
PLoS Genet ; 20(7): e1011345, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38985845

ABSTRACT

The piRNA pathway is a conserved germline-specific small RNA pathway that ensures genomic integrity and continued fertility. In C. elegans and other nematodes, Type-I piRNAs are expressed from >10,000 independently transcribed genes clustered within two discrete domains of 1.5 and 3.5 MB on Chromosome IV. Clustering of piRNA genes contributes to their germline-specific expression, but the underlying mechanisms are unclear. We analyze isolated germ nuclei to demonstrate that the piRNA genomic domains are located in a heterochromatin-like environment. USTC (Upstream Sequence Transcription Complex) promotes strong association of nucleosomes throughout piRNA clusters, yet organizes the local nucleosome environment to direct the exposure of individual piRNA genes. Localization of USTC to the piRNA domains depends upon the ATPase chromatin remodeler ISW-1, which maintains high nucleosome density across piRNA clusters and ongoing production of piRNA precursors. Overall, this work provides insight into how chromatin states coordinate transcriptional regulation over large genomic domains, with implications for global genome organization.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Germ Cells , Nucleosomes , Promoter Regions, Genetic , RNA, Small Interfering , Animals , Caenorhabditis elegans/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Germ Cells/metabolism , Chromatin Assembly and Disassembly/genetics , Chromatin/genetics , Chromatin/metabolism , Transcription, Genetic , Gene Expression Regulation/genetics , Heterochromatin/genetics , Heterochromatin/metabolism , Piwi-Interacting RNA
19.
Curr Opin Cell Biol ; 89: 102398, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38991477

ABSTRACT

Eukaryotic genomes are organized into 3D structures, which range from small-scale nucleosome arrays to large-scale chromatin domains. These structures have an important role in the regulation of transcription and other nuclear processes. Despite advances in our understanding of the properties, functions, and underlying mechanisms of genome structures, there are many open questions about the interplay between these structures across scales. In particular, it is not well understood if and how 1D features of nucleosome arrays influence large-scale 3D genome folding patterns. In this review, we discuss recent studies that address these questions and summarize our current understanding of the relationship between nucleosome positioning and higher-order genome folding.


Subject(s)
Genome , Nucleosomes , Nucleosomes/metabolism , Nucleosomes/chemistry , Humans , Animals , Chromatin Assembly and Disassembly , Nucleic Acid Conformation , Chromatin/metabolism , Chromatin/chemistry
20.
Sci Adv ; 10(27): eadm9740, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38959309

ABSTRACT

Micrococcal nuclease sequencing is the state-of-the-art method for determining chromatin structure and nucleosome positioning. Data analysis is complex due to the AT-dependent sequence bias of the endonuclease and the requirement for high sequencing depth. Here, we present the nucleosome-based MNase accessibility (nucMACC) pipeline unveiling the regulatory chromatin landscape by measuring nucleosome accessibility and stability. The nucMACC pipeline represents a systematic and genome-wide approach for detecting unstable ("fragile") nucleosomes. We have characterized the regulatory nucleosome landscape in Drosophila melanogaster, Saccharomyces cerevisiae, and mammals. Two functionally distinct sets of promoters were characterized, one associated with an unstable nucleosome and the other being nucleosome depleted. We show that unstable nucleosomes present intermediate states of nucleosome remodeling, preparing inducible genes for transcriptional activation in response to stimuli or stress. The presence of unstable nucleosomes correlates with RNA polymerase II proximal pausing. The nucMACC pipeline offers unparalleled precision and depth in nucleosome research and is a valuable tool for future nucleosome studies.


Subject(s)
Drosophila melanogaster , Micrococcal Nuclease , Nucleosomes , Saccharomyces cerevisiae , Nucleosomes/metabolism , Nucleosomes/genetics , Animals , Micrococcal Nuclease/metabolism , Drosophila melanogaster/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Chromatin Assembly and Disassembly , Genome , Promoter Regions, Genetic , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Chromatin/genetics , Chromatin/metabolism , Sequence Analysis, DNA/methods
SELECTION OF CITATIONS
SEARCH DETAIL