Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54.508
Filter
1.
Methods Mol Biol ; 2856: 341-356, 2025.
Article in English | MEDLINE | ID: mdl-39283463

ABSTRACT

To reveal gene regulation mechanisms, it is essential to understand the role of regulatory elements, which are possibly distant from gene promoters. Integrative analysis of epigenetic and transcriptomic data can be used to gain insights into gene-expression regulation in specific phenotypes. Here, we discuss STITCHIT, an approach to dissect epigenetic variation in a gene-specific manner across many samples for the identification of regulatory elements without relying on peak calling algorithms. The obtained genomic regions are then further refined using a regularized linear model approach, which can also be used to predict gene expression. We illustrate the use of STITCHIT using H3k27ac ChIP-seq and RNA-seq data from the International Human Epigenome Consortium (IHEC).


Subject(s)
Epigenesis, Genetic , Epigenomics , Transcriptome , Humans , Epigenomics/methods , Transcriptome/genetics , Enhancer Elements, Genetic , Software , Computational Biology/methods , Chromatin Immunoprecipitation Sequencing/methods , Gene Expression Regulation , Algorithms , Histones/genetics , Histones/metabolism , Gene Expression Profiling/methods
2.
BMC Cancer ; 24(1): 1189, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333927

ABSTRACT

BACKGROUND: Histone acetylation plays a critical role in the progression of acute myeloid leukemia (AML). This study aimed to explore the prognostic significance and biological implications of histone acetylation-related genes in AML and to identify potential oncoproteins and therapeutic compounds. METHODS: Genes associated with AML and histone acetylation were identified using the TCGA-LAML and IMEx Interactome databases. A histone acetylation-related risk model was developed using the least absolute shrinkage and selection operator method. The prognostic value of the model was evaluated through Kaplan-Meier survival analysis, time-dependent receiver operating characteristic curve, univariate and multivariate Cox regression, and nomogram calibration. Key genes were identified using random forest, support vector machine, and multivariate Cox analysis. Molecular docking was employed to assess the binding affinity between ribosomal protein S6 kinase A1 (RPS6KA1) and potential compounds. Furthermore, the effects of RPS6KA1 and afzelin on the malignant behaviors and downstream pathways of AML cells were validated through in vitro experiments. RESULTS: A risk model composed of 6 genes, including HDAC6, CREB3, KLF13, GOLGA2, RPS6KA1 and ZMIZ2, was established, demonstrating strong prognostic predictive capability. Among these, RPS6KA1 emerged as a key risk factor linked to histone acetylation status in AML. Elevated RPS6KA1 expression was observed in AML samples and was associated with poor prognosis. RPS6KA1 knockdown suppressed AML cell proliferation, migration, and invasion, induced G0/G1 phase arrest, and promoted apoptosis. Additionally, RPS6KA1 was identified as a potential target for afzelin, which exhibited anti-AML activity by inactivating RPS6KA1. CONCLUSION: Histone acetylation status is closely associated with AML patient prognosis. RPS6KA1 acts as an oncoprotein in AML, facilitating disease progression. Afzelin may represent a novel therapeutic agent for AML by targeting RPS6KA1, which requires validation by clinical trials.


Subject(s)
Histones , Leukemia, Myeloid, Acute , Ribosomal Protein S6 Kinases, 90-kDa , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Acetylation , Histones/metabolism , Prognosis , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Cell Proliferation , Cell Line, Tumor , Molecular Docking Simulation , Male , Female , Apoptosis , Oncogene Proteins/metabolism , Oncogene Proteins/genetics , Middle Aged , Ribosomal Protein S6 Kinases, 70-kDa
3.
Zool Res ; 45(6): 1175-1187, 2024 Nov 18.
Article in English | MEDLINE | ID: mdl-39318125

ABSTRACT

Isocitrate dehydrogenase 2 (IDH2) and glutamate dehydrogenase 1 (GLUD1) are key enzymes involved in the production of α-ketoglutarate (α-KG), a metabolite central to the tricarboxylic acid cycle and glutamine metabolism. In this study, we investigated the impact of IDH2 and GLUD1 on early porcine embryonic development following IDH2 and GLUD1 knockdown (KD) via double-stranded RNA (dsRNA) microinjection. Results showed that KD reduced α-KG levels, leading to delayed embryonic development, decreased blastocyst formation, increased apoptosis, reduced blastomere proliferation, and pluripotency. Additionally, IDH2 and GLUD1 KD induced abnormally high levels of trimethylation of lysine 20 of histone H4 (H4K20me3) at the 4-cell stage, likely resulting in transcriptional repression of embryonic genome activation (EGA)-related genes. Notably, KD of lysine methyltransferase 5C ( KMT5C) and supplementation with exogenous α-KG reduced H4K20me3 expression and partially rescued these defects, suggesting a critical role of IDH2 and GLUD1 in the epigenetic regulation and proper development of porcine embryos. Overall, this study highlights the significance of IDH2 and GLUD1 in maintaining normal embryonic development through their influence on α-KG production and subsequent epigenetic modifications.


Subject(s)
Embryonic Development , Epigenesis, Genetic , Glutamate Dehydrogenase , Isocitrate Dehydrogenase , Parthenogenesis , Animals , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Swine/embryology , Glutamate Dehydrogenase/metabolism , Glutamate Dehydrogenase/genetics , Histones/metabolism , Histones/genetics , Gene Expression Regulation, Developmental , Gene Knockdown Techniques
4.
Biomolecules ; 14(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39334865

ABSTRACT

N-alpha-acetyltransferase 40 (NAA40) is an evolutionarily conserved N-terminal acetyltransferase (NAT) linked to oncogenesis and chemoresistance. A recent study reported the generation of a second, shorter NAA40 isoform (NAA40S) through alternative translation, which we proceeded to further characterise. Notably, recombinant NAA40S had a greater in vitro enzymatic activity and affinity towards its histone H2A/H4 substrates compared to full-length NAA40 (NAA40L). Within cells, NAA40S was enzymatically active, based on its ability to suppress the H2A/H4S1Ph antagonistic mark in CRISPR-generated NAA40 knockout cells. Finally, we show that in addition to alternative translation, the NAA40S isoform could be derived from a primate and testis-specific transcript, which may align with the "out-of-testis" origin of recently evolved genes and isoforms. To summarise, our data reveal an even greater functional divergence between the two NAA40 isoforms than had been previously recognised.


Subject(s)
Histones , Humans , Histones/metabolism , Histones/genetics , Animals , N-Terminal Acetyltransferase E/metabolism , N-Terminal Acetyltransferase E/genetics , Isoenzymes/metabolism , Isoenzymes/genetics , Isoenzymes/chemistry , Protein Isoforms/metabolism , Protein Isoforms/genetics , HEK293 Cells
5.
Genes (Basel) ; 15(9)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39336747

ABSTRACT

The African clawed frog (Xenopus laevis) endures prolonged periods of dehydration while estivating underground during the dry season. Epigenetic modifications play crucial roles in regulating gene expression in response to environmental changes. The elucidation of epigenetic changes relevant to survival could serve as a basis for further studies on organ preservation under extreme stress. The current study examined the relative protein levels of key enzymes involved in the arginine methylation of histones in the liver and kidney tissues of control versus dehydrated (35 ± 1%) X. laevis through immunoblotting. Protein arginine methyltransferases (PRMT) 4, 5, and 6 showed significant protein level decreases of 35 ± 3%, 71 ± 7%, and 25 ± 5%, respectively, in the liver tissues of the dehydrated frogs relative to controls. In contrast, PRMT7 exhibited an increase of 36 ± 4%. Similarly, the methylated histone markers H3R2m2a, H3R8m2a, and H3R8m2s were downregulated by 34 ± 11%, 15 ± 4%, and 42 ± 12%, respectively, in the livers of dehydrated frogs compared to controls. By contrast, the kidneys of dehydrated frogs showed an upregulation of histone markers. H3R2m2a, H3R8m2a, H3R8m2s, and H4R3m2a were significantly increased by 126 ± 12%, 112 ± 7%, 47 ± 13%, and 13 ± 3%, respectively. These changes can play vital roles in the metabolic reorganization of X. laevis during dehydration, and are likely to increase the chances of survival. In turn, the tissue-specific regulation of the histone arginine methylation mechanism suggests the importance of epigenetic regulation in the adaptation of X. laevis for whole-body dehydration.


Subject(s)
Arginine , Histones , Liver , Xenopus laevis , Animals , Xenopus laevis/genetics , Histones/metabolism , Histones/genetics , Methylation , Arginine/metabolism , Liver/metabolism , Dehydration/genetics , Dehydration/metabolism , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism , Epigenesis, Genetic , Kidney/metabolism , Gene Expression Regulation , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
6.
Genes (Basel) ; 15(9)2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39336823

ABSTRACT

BACKGROUND/OBJECTIVES: Transgene applications, ranging from gene therapy to the development of stable cell lines and organisms, rely on maintaining the expression of transgenes. To date, the use of plasmid-based transgenes has been limited by the loss of their expression shortly after their delivery into the target cells. The short-lived expression of plasmid-based transgenes has been largely attributed to host-cell-mediated degradation and/or silencing of transgenes. The development of chromatin-based strategies for gene delivery has the potential to facilitate defining the requirements for establishing epigenetic states and to enhance transgene expression for numerous applications. METHODS: To assess the impact of "priming" plasmid-based transgenes to adopt accessible chromatin states to promote gene expression, nucleosome positioning elements were introduced at promoters of transgenes, and vectors were pre-assembled into nucleosomes containing unmodified histones or mutants mimicking constitutively acetylated states at residues 9 and 14 of histone H3 or residue 16 of histone H4 prior to their introduction into cells, then the transgene expression was monitored over time. RESULTS: DNA sequences capable of positioning nucleosomes could positively impact the expression of adjacent transgenes in a distance-dependent manner in the absence of their pre-assembly into chromatin. Intriguingly, the pre-assembly of plasmids into chromatin facilitated the prolonged expression of transgenes relative to plasmids that were not pre-packaged into chromatin. Interactions between pre-assembled chromatin states and nucleosome positioning-derived effects on expression were also assessed and, generally, nucleosome positioning played the predominant role in influencing gene expression relative to priming with hyperacetylated chromatin states. CONCLUSIONS: Strategies incorporating nucleosome positioning elements and the pre-assembly of plasmids into chromatin prior to nuclear delivery can modulate the expression of plasmid-based transgenes.


Subject(s)
Chromatin Assembly and Disassembly , Histones , Nucleosomes , Transgenes , Nucleosomes/genetics , Nucleosomes/metabolism , Histones/genetics , Histones/metabolism , Chromatin Assembly and Disassembly/genetics , Humans , Chromatin/genetics , Chromatin/metabolism , Plasmids/genetics , Promoter Regions, Genetic , Animals
7.
Cell Commun Signal ; 22(1): 453, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39327549

ABSTRACT

BACKGROUND: A growing body of evidence indicates that histone variants play an oncogenic role in cancer progression. However, the role and mechanism of histone variant H2AZ1 in lung cancer remain poorly understood. In this study, we aim to identify novel functions and molecular mechanisms of H2AZ1 in lung cancer. METHODS: We analyzed H2AZ1 expression in lung adenocarcinoma using several RNA-seq and microarray datasets. Immunohistochemistry staining for H2AZ1 was performed on two sets of lung cancer tissue microarrays. To study the function of H2AZ1, we conducted assays for cell proliferation, colony formation, invasion, and migration. We employed CUT&Tag-seq, ATAC-seq, RNA-seq, and Western blotting to explore the regulatory patterns and potential mechanisms of H2AZ1 in lung adenocarcinoma. RESULTS: Our findings reveal that H2AZ1 is highly expressed in lung cancer and high levels of H2AZ1 mRNA are associated with poor patient survival. Silencing H2AZ1 impaired cell proliferation, colony formation, migration, and invasion. Mechanistically, our CUT&Tag-seq, ATAC-seq, and RNA-seq results showed that H2AZ1 is primarily deposited around TSS and affects multiple oncogenic signaling pathways. Importantly, we uncovered that H2AZ1 may drive lung cancer progression through the RELA-HIF1A-EGFR signaling pathway. CONCLUSION: H2AZ1 plays an oncogenic role via several cancer-related pathways, including the RELA-HIF1A-EGFR axis in lung cancer. Intervention targeting H2AZ1 and its related signaling genes may have translational potential for precision therapy.


Subject(s)
Cell Proliferation , Disease Progression , ErbB Receptors , Histones , Hypoxia-Inducible Factor 1, alpha Subunit , Lung Neoplasms , Signal Transduction , Transcription Factor RelA , Humans , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , ErbB Receptors/metabolism , ErbB Receptors/genetics , Signal Transduction/genetics , Transcription Factor RelA/metabolism , Transcription Factor RelA/genetics , Histones/metabolism , Histones/genetics , Cell Proliferation/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism
8.
Viruses ; 16(9)2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39339966

ABSTRACT

Primary Effusion Lymphoma (PEL) cells carry Kaposi's sarcoma-associated herpesvirus (KSHV) in a latent state, except for a small number of cells in which the virus replicates to ensure its persistence into the infected host. However, the lytic cycle can be reactivated in vitro by exposing these lymphoma cells to various treatments, leading to cell lysis. To restrict viral antigen expression, KSHV induces repressive epigenetic changes, including DNA methylation and histone modifications. Among the latter, histone deacetylation and tri-methylation of Histone H3 lisyne-27 (H3K27me3) have been reported to play a role. Here, we found that the inhibition of H3K27 tri-methylation by valemetostat DS3201 (DS), a small molecule that inhibits Enhancer of Zeste Homolog 2 (EZH2) methyltransferase, induced the KSHV lytic cycle in PEL cells, and that this effect involved the activation of the wtp53-p21 axis and autophagic dysregulation. DS also potentiated the lytic cycle activation mediated by the Histone deacetylases (HDAC) inhibitor Suberoylanilide hydroxamic acid (SAHA) and reinforced its cytotoxic effect, suggesting that such a combination could be used to unbalance the latent/lytic cycle and further impair the survival of PEL cells.


Subject(s)
Enhancer of Zeste Homolog 2 Protein , Herpesvirus 8, Human , Histone Deacetylase Inhibitors , Lymphoma, Primary Effusion , Vorinostat , Herpesvirus 8, Human/drug effects , Herpesvirus 8, Human/physiology , Herpesvirus 8, Human/genetics , Humans , Lymphoma, Primary Effusion/virology , Lymphoma, Primary Effusion/drug therapy , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/metabolism , Histone Deacetylase Inhibitors/pharmacology , Vorinostat/pharmacology , Cell Line, Tumor , Virus Latency/drug effects , Virus Activation/drug effects , Virus Replication/drug effects , Autophagy/drug effects , Histones/metabolism
9.
Nat Commun ; 15(1): 8334, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333072

ABSTRACT

Autophagy, a highly conserved self-digestion process crucial for cellular homeostasis, is triggered by various environmental signals, including nutrient scarcity. The regulation of lysosomal and autophagy-related processes is pivotal to maintaining cellular homeostasis and basal metabolism. The consequences of disrupting or diminishing lysosomal and autophagy systems have been investigated; however, information on the implications of hyperactivating lysosomal and autophagy genes on homeostasis is limited. Here, we present a mechanism of transcriptional repression involving upstream stimulatory factor 2 (USF2), which inhibits lysosomal and autophagy genes under nutrient-rich conditions. We find that USF2, together with HDAC1, binds to the CLEAR motif within lysosomal genes, thereby diminishing histone H3K27 acetylation, restricting chromatin accessibility, and downregulating lysosomal gene expression. Under starvation, USF2 competes with transcription factor EB (TFEB), a master transcriptional activator of lysosomal and autophagy genes, to bind to target gene promoters in a phosphorylation-dependent manner. The GSK3ß-mediated phosphorylation of the USF2 S155 site governs USF2 DNA-binding activity, which is involved in lysosomal gene repression. These findings have potential applications in the treatment of protein aggregation-associated diseases, including α1-antitrypsin deficiency. Notably, USF2 repression is a promising therapeutic strategy for lysosomal and autophagy-related diseases.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Lysosomes , Upstream Stimulatory Factors , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Lysosomes/metabolism , Autophagy/genetics , Humans , Upstream Stimulatory Factors/metabolism , Upstream Stimulatory Factors/genetics , Phosphorylation , Histone Deacetylase 1/metabolism , Histone Deacetylase 1/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Gene Expression Regulation , Promoter Regions, Genetic , HEK293 Cells , Animals , Histones/metabolism , HeLa Cells , Mice , Acetylation
10.
Nat Commun ; 15(1): 8309, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333103

ABSTRACT

Chimeric antigen receptor-modified T cell (CAR-T) immunotherapy has revolutionised blood cancer treatment. Parsing the genetic underpinnings of T cell quality and CAR-T efficacy is challenging. Transcriptomics inform CAR-T state, but the nature of dynamic transcription during activation hinders identification of transiently or minimally expressed genes, such as transcription factors, and over-emphasises effector and metabolism genes. Here we explore whether analyses of transcriptionally repressive and permissive histone methylation marks describe CAR-T cell functional states and therapeutic potential beyond transcriptomic analyses. Histone mark analyses improve identification of differences between naïve, central memory, and effector memory CD8 + T cell subsets of human origin, and CAR-T derived from these subsets. We find important differences between CAR-T manufactured from central memory cells of healthy donors and of patients. By examining CAR-T products from a clinical trial in lymphoma (NCT01865617), we find a novel association between the activity of the transcription factor KLF7 with in vivo CAR-T accumulation in patients and demonstrate that over-expression of KLF7 increases in vitro CAR-T proliferation and IL-2 production. In conclusion, histone marks provide a rich dataset for identification of functionally relevant genes not apparent by transcriptomics.


Subject(s)
CD8-Positive T-Lymphocytes , Histone Code , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Immunotherapy, Adoptive/methods , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Histones/metabolism , Lymphoma/genetics , Lymphoma/metabolism , Lymphoma/therapy , Cell Proliferation/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Immunologic Memory
11.
Nat Commun ; 15(1): 8283, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333491

ABSTRACT

The nucleosome is one of the hallmarks of eukaryotes, a dynamic platform that supports many critical functions in eukaryotic cells. Here, we engineer the in vivo assembly of the nucleosome core in the model bacterium Escherichia coli. We show that bacterial chromosome DNA and eukaryotic histones can assemble in vivo to form nucleosome complexes with many features resembling those found in eukaryotes. The formation of nucleosomes in E. coli was visualized with atomic force microscopy and using tripartite split green fluorescent protein. Under a condition that moderate histones expression was induced at 1 µM IPTG, the nucleosome-forming bacterium is viable and has sustained growth for at least 110 divisions in longer-term growth experiments. It exhibits stable nucleosome formation, a consistent transcriptome across passages, and reduced growth fitness under stress conditions. In particular, the nucleosome arrays in E. coli genic regions have profiles resembling those in eukaryotic cells. The observed compatibility between the eukaryotic nucleosome and the bacterial chromosome machinery may reflect a prerequisite for bacteria-archaea union, providing insight into eukaryogenesis and the origin of the nucleosome.


Subject(s)
Escherichia coli , Histones , Microscopy, Atomic Force , Nucleosomes , Nucleosomes/metabolism , Nucleosomes/ultrastructure , Escherichia coli/metabolism , Escherichia coli/genetics , Histones/metabolism , Histones/genetics , DNA, Bacterial/metabolism , DNA, Bacterial/genetics , Chromosomes, Bacterial/metabolism , Chromosomes, Bacterial/genetics , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/genetics , Eukaryotic Cells/metabolism
12.
Nat Commun ; 15(1): 8385, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333493

ABSTRACT

The spatial co-presence of aberrant long non-coding RNAs (lncRNAs) and abnormal coding genes contributes to malignancy development in various tumors. However, precise coordinated mechanisms underlying this phenomenon in tumorigenesis remains incompletely understood. Here, we show that Prohibitin 2 (PHB2) orchestrates the transcription of an oncogenic CASC15-New-Isoform 2 (CANT2) lncRNA and the coding tumor-suppressor gene CCBE1, thereby accelerating melanoma tumorigenesis. In melanoma cells, PHB2 initially accesses the open chromatin sites at the CANT2 promoter, recruiting MLL2 to augment H3K4 trimethylation and activate CANT2 transcription. Intriguingly, PHB2 further binds the activated CANT2 transcript, targeting the promoter of the tumor-suppressor gene CCBE1. This interaction recruits histone deacetylase HDAC1 to decrease H3K27 acetylation at the CCBE1 promoter and inhibit its transcription, significantly promoting tumor cell growth and metastasis both in vitro and in vivo. Our study elucidates a PHB2-mediated mechanism that orchestrates the aberrant transcription of lncRNAs and coding genes, providing an intriguing epigenetic regulatory model in tumorigenesis.


Subject(s)
Carcinogenesis , Gene Expression Regulation, Neoplastic , Prohibitins , Promoter Regions, Genetic , RNA, Long Noncoding , Repressor Proteins , Transcription, Genetic , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Humans , Repressor Proteins/metabolism , Repressor Proteins/genetics , Carcinogenesis/genetics , Animals , Cell Line, Tumor , Promoter Regions, Genetic/genetics , Melanoma/genetics , Melanoma/pathology , Melanoma/metabolism , Mice , Mice, Nude , Cell Proliferation/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 1/genetics , Histones/metabolism
13.
Nat Commun ; 15(1): 7769, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39237515

ABSTRACT

Histone H3-mutant gliomas are deadly brain tumors characterized by a dysregulated epigenome and stalled differentiation. In contrast to the extensive datasets available on tumor cells, limited information exists on their tumor microenvironment (TME), particularly the immune infiltrate. Here, we characterize the immune TME of H3.3K27M and G34R/V-mutant gliomas, and multiple H3.3K27M mouse models, using transcriptomic, proteomic and spatial single-cell approaches. Resolution of immune lineages indicates high infiltration of H3-mutant gliomas with diverse myeloid populations, high-level expression of immune checkpoint markers, and scarce lymphoid cells, findings uniformly reproduced in all H3.3K27M mouse models tested. We show these myeloid populations communicate with H3-mutant cells, mediating immunosuppression and sustaining tumor formation and maintenance. Dual inhibition of myeloid cells and immune checkpoint pathways show significant therapeutic benefits in pre-clinical syngeneic mouse models. Our findings provide a valuable characterization of the TME of oncohistone-mutant gliomas, and insight into the means for modulating the myeloid infiltrate for the benefit of patients.


Subject(s)
Brain Neoplasms , Glioma , Histones , Mutation , Myeloid Cells , Tumor Microenvironment , Animals , Glioma/genetics , Glioma/immunology , Glioma/pathology , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Myeloid Cells/metabolism , Myeloid Cells/immunology , Histones/metabolism , Histones/genetics , Mice , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Humans , Cell Line, Tumor , Disease Models, Animal , Mice, Inbred C57BL , Gene Expression Regulation, Neoplastic , Single-Cell Analysis
14.
Nat Commun ; 15(1): 7758, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39237615

ABSTRACT

Lysine-specific histone demethylase 1 (LSD1), which demethylates mono- or di- methylated histone H3 on lysine 4 (H3K4me1/2), is essential for early embryogenesis and development. Here we show that LSD1 is dispensable for mouse embryonic stem cell (ESC) self-renewal but is required for mouse ESC growth and differentiation. Reintroduction of a catalytically-impaired LSD1 (LSD1MUT) recovers the proliferation capability of mouse ESCs, yet the enzymatic activity of LSD1 is essential to ensure proper differentiation. Indeed, increased H3K4me1 in Lsd1 knockout (KO) mouse ESCs does not lead to major changes in global gene expression programs related to stemness. However, ablation of LSD1 but not LSD1MUT results in decreased DNMT1 and UHRF1 proteins coupled to global hypomethylation. We show that both LSD1 and LSD1MUT control protein stability of UHRF1 and DNMT1 through interaction with HDAC1 and the ubiquitin-specific peptidase 7 (USP7), consequently, facilitating the deacetylation and deubiquitination of DNMT1 and UHRF1. Our studies elucidate a mechanism by which LSD1 controls DNA methylation in mouse ESCs, independently of its lysine demethylase activity.


Subject(s)
CCAAT-Enhancer-Binding Proteins , Cell Differentiation , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation , Histone Demethylases , Mice, Knockout , Mouse Embryonic Stem Cells , Ubiquitin-Protein Ligases , Animals , Histone Demethylases/metabolism , Histone Demethylases/genetics , Mice , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Mouse Embryonic Stem Cells/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 1/genetics , Histones/metabolism , Cell Proliferation , Ubiquitination
15.
Sci Rep ; 14(1): 20771, 2024 09 05.
Article in English | MEDLINE | ID: mdl-39237663

ABSTRACT

G9a is a histone methyltransferase that catalyzes the methylation of histone 3 lysine 9 (H3K9), which is involved in the regulation of gene expression. We had previously reported that G9a is expressed in developing tendons in vivo and in vitro and that G9a-deficient tenocytes show impaired proliferation and differentiation in vitro. In this study, we investigated the functions of G9a in tendon development in vivo by using G9a conditional knockout (G9a cKO) mice. We crossed Sox9Cre/+ mice with G9afl/fl mice to generate G9afl/fl; Sox9Cre/+ mice. The G9a cKO mice showed hypoplastic tendon formation at 3 weeks of age. Bromodeoxyuridine labeling on embryonic day 16.5 (E16.5) revealed decreased cell proliferation in the tenocytes of G9a cKO mice. Immunohistochemical analysis revealed decreased expression levels of G9a and its substrate, H3K9me2, in the vertebral tendons of G9a cKO mice. The tendon tissue of the vertebrae and limbs of G9a cKO mice showed reduced expression of a tendon marker, tenomodulin (Tnmd), and col1a1 genes, suggesting that tenocyte differentiation was suppressed. Overexpression of G9a resulted in enhancement of Tnmd and col1a1 expression in tenocytes in vitro. These results suggest that G9a regulates the proliferation and differentiation of tendon progenitor cells during tendon development. Thus, our results suggest that G9a plays an essential role in tendon development.


Subject(s)
Cell Differentiation , Cell Proliferation , Histone-Lysine N-Methyltransferase , Mice, Knockout , Tendons , Animals , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Tendons/metabolism , Tendons/embryology , Mice , Tenocytes/metabolism , Histones/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Collagen Type I, alpha 1 Chain/metabolism , Collagen Type I/metabolism , Collagen Type I/genetics , Gene Expression Regulation, Developmental , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics
16.
Neurosci Lett ; 839: 137957, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39218294

ABSTRACT

Post-traumatic stress disorder (PTSD) presents distinct sex-specific differences in both symptom expression and treatment outcomes, with the underlying biological mechanisms still remain unclear. Epigenetic modifications, particularly histone acetylation, have been increasingly recognized as critical factors in the pathophysiology of PTSD. Valproic acid (VPA), a potent histone deacetylase (HDAC) inhibitor, has shown promise in modulating epigenetic responses and improving therapeutic outcomes is PTSD, though its effect may differ between sexes. This study aimed to explore the sex-specific epigenetic changes in response to trauma and the impact of VPA treatment in a rat model of PTSD induced by predator scent stress. Sprague-Dawley rats of both sexes were randomly assigned to stressed and non-stressed groups and treated with either VPA (100 mg/kg) or vehicle. Anxiety levels were assessed using the elevated plus maze, followed by analysis of histone H3 and H4 acetylation, HDAC activity, and c-fos expression in the hippocampus. Our findings revealed that traumatic stress led to increased freezing time and anxiety levels, with more pronounced effects observed in females. Additionally, we have identified sex-specific differences in hippocampal epigenetic modifications; stressed females exhibited higher H3 acetylation, and VPA-treated stressed males showed increased H4 acetylation. These results highlight the importance of considering sex differences in the epigenetic mechanism underlying PTSD and suggest that personalized therapeutic approaches may be necessary to address these complexities.


Subject(s)
Epigenesis, Genetic , Histone Deacetylase Inhibitors , Rats, Sprague-Dawley , Stress Disorders, Post-Traumatic , Valproic Acid , Animals , Valproic Acid/pharmacology , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/genetics , Stress Disorders, Post-Traumatic/metabolism , Male , Female , Epigenesis, Genetic/drug effects , Rats , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Disease Models, Animal , Histones/metabolism , Sex Characteristics , Hippocampus/metabolism , Hippocampus/drug effects , Acetylation/drug effects , Anxiety/drug therapy
17.
Cell Mol Life Sci ; 81(1): 381, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39222083

ABSTRACT

Epigenetic modifications (methylation, acetylation, etc.) of core histones play a key role in regulation of gene expression. Thus, the epigenome changes strongly during various biological processes such as cell differentiation and dedifferentiation. Classical methods of analysis of epigenetic modifications such as mass-spectrometry and chromatin immuno-precipitation, work with fixed cells only. Here we present a genetically encoded fluorescent probe, MPP8-Green, for detecting H3K9me3, a histone modification associated with inactive chromatin. This probe, based on the chromodomain of MPP8, allows for visualization of H3K9me3 epigenetic landscapes in single living cells. We used this probe to track changes in H3K9me3 landscapes during the differentiation of induced pluripotent stem cells (iPSCs) into induced neurons. Our findings revealed two major waves of global H3K9me3 reorganization during 4-day differentiation, namely on the first and third days, whereas nearly no changes occurred on the second and fourth days. The proposed method LiveMIEL (Live-cell Microscopic Imaging of Epigenetic Landscapes), which combines genetically encoded epigenetic probes and machine learning approaches, enables classification of multiparametric epigenetic signatures of single cells during stem cell differentiation and potentially in other biological models.


Subject(s)
Cell Differentiation , Epigenesis, Genetic , Fluorescent Dyes , Histones , Induced Pluripotent Stem Cells , Cell Differentiation/genetics , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Histones/metabolism , Histones/genetics , Humans , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Neurons/metabolism , Neurons/cytology , Animals , Mice
18.
Radiat Oncol ; 19(1): 116, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223539

ABSTRACT

BACKGROUND: A predictive assay for late radiation toxicity would allow more personalized treatment planning, reducing the burden of toxicity for the more sensitive minority, and improving the therapeutic index for the majority. In a previous study in prostate cancer patients, the γ-H2AX foci decay ratio (γ-FDR) was the strongest predictor of late radiation toxicity. The current study aimed to validate this finding in a more varied group of patients with pelvic cancer. Additionally, the potential correlation between the γ-FDR and patient-reported outcomes was investigated. METHODS: Prostate and gynecological cancer patients with ≥ 24 months of follow-up were included in the current analysis. Toxicity was evaluated by physician (CTCAE version 4) and patient (EORTC questionnaires). γ-FDRs were determined in ex vivo irradiated lymphocytes. Correlation between γ-FDR and toxicity was assessed using both linear and logistic regression analyses. The highest toxicity grade recorded during follow-up was used. The association between global quality of life and γ-FDR was tested by comparing the change in quality of life over time in patients with γ-FDR < or ≥ 3.41, a previously established threshold. RESULTS: Eighty-eight patients were included. Physician-assessed and patient-reported cumulative grade ≥ 2 toxicity was 25% and 29%, respectively; which is much lower than in the previous cohort (i.e., 51% CTCAE grade ≥ 2). Patients with toxicity exhibited less favorable dose-volume parameters. In men, these parameters showed significant improvement compared to the previous cohort. The proportion of patients with a low γ-FDR increased with severity of toxicity, but this trend was not statistically significant. In addition, a γ-FDR < 3.41 was not correlated with the development of moderate to severe toxicity. Post-treatment decline in global quality of life was minimal, and similar for patients with γ-FDR < or ≥ 3.41. CONCLUSIONS: In the present study, the γ-H2AX foci decay ratio could not be validated as a predictor of late radiation toxicity in patients with pelvic cancer. Improved radiotherapy techniques with smaller irradiated bladder and bowel volumes have probably resulted in less toxicities. Future studies on genetic markers of toxicity should be powered on these lower incidences. We further recommend taking persistency, next to severity, into consideration.


Subject(s)
Histones , Prostatic Neoplasms , Quality of Life , Radiation Injuries , Radiotherapy, Image-Guided , Humans , Male , Female , Aged , Radiotherapy, Image-Guided/methods , Radiotherapy, Image-Guided/adverse effects , Middle Aged , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms/pathology , Histones/genetics , Histones/analysis , Radiation Injuries/etiology , Aged, 80 and over , Genital Neoplasms, Female/radiotherapy , Adult , Follow-Up Studies , Pelvic Neoplasms/radiotherapy , Biomarkers, Tumor/genetics , Prognosis
19.
BMC Biol ; 22(1): 188, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39218869

ABSTRACT

BACKGROUND: The histone variant macroH2A (mH2A), the most deviant variant, is about threefold larger than the conventional histone H2A and consists of a histone H2A-like domain fused to a large Non-Histone Region responsible for recruiting PARP-1 to chromatin. The available data suggest that the histone variant mH2A participates in the regulation of transcription, maintenance of heterochromatin, NAD+ metabolism, and double-strand DNA repair. RESULTS: Here, we describe a novel function of mH2A, namely its implication in DNA oxidative damage repair through PARP-1. The depletion of mH2A affected both repair and cell survival after the induction of oxidative lesions in DNA. PARP-1 formed a specific complex with mH2A nucleosomes in vivo. The mH2A nucleosome-associated PARP-1 is inactive. Upon oxidative damage, mH2A is ubiquitinated, PARP-1 is released from the mH2A nucleosomal complex, and is activated. The in vivo-induced ubiquitination of mH2A, in the absence of any oxidative damage, was sufficient for the release of PARP-1. However, no release of PARP-1 was observed upon treatment of the cells with either the DNA alkylating agent MMS or doxorubicin. CONCLUSIONS: Our data identify a novel pathway for the repair of DNA oxidative lesions, requiring the ubiquitination of mH2A for the release of PARP-1 from chromatin and its activation.


Subject(s)
DNA Damage , DNA Repair , Histones , Poly (ADP-Ribose) Polymerase-1 , Ubiquitination , Histones/metabolism , Histones/genetics , Humans , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly (ADP-Ribose) Polymerase-1/genetics , Oxidative Stress , Nucleosomes/metabolism
20.
Chem Res Toxicol ; 37(9): 1588-1597, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39237351

ABSTRACT

Histones and their posttranslational modifications (PTMs) are critical regulators of gene expression. Differentiation, environmental stressors, xenobiotics, and major human diseases cause significant changes in histone variants and PTMs. Western blotting is the mainstay methodology for detection of histones and their PTMs in the majority of studies. Surprisingly, despite their high abundance in cells, immunoblotting of histones typically involves loading of large protein amounts that are normally used for detection of sparse cellular proteins. We systematically examined technical factors in the Western-blotting-based detection of human histones with >30 antibodies. We found that under multiple protein transfer conditions, many histone epitopes on polyvinylidene fluoride (PVDF) membranes had a very low antibody accessibility, which was dramatically increased by the addition of a simple denaturation step. Denaturation of membrane-bound proteins also enhanced the specificity of some histone antibodies. In comparison to standard PVDF membranes, the sensitivity of histone detection on standard nitrocellulose membranes was typically much higher, which was further increased by the inclusion of the same denaturation step. Optimized protocols increased by >100-times detection sensitivity for the genotoxic marker γ-H2AX with two monoclonal antibodies. The impact of denaturation and nitrocellulose use varied for different histones, but for each histone, it was generally similar for antibodies targeting N-terminal and C-terminal regions. In summary, denaturation of membrane-bound histones strongly improves their detection by Westerns, resulting in more accurate measurements and permitting analyses with small biological samples.


Subject(s)
Histones , Histones/chemistry , Histones/metabolism , Histones/analysis , Humans , Blotting, Western , Polyvinyls/chemistry , Fluorocarbon Polymers
SELECTION OF CITATIONS
SEARCH DETAIL