Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37.715
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.
Front Immunol ; 15: 1430187, 2024.
Article in English | MEDLINE | ID: mdl-39351229

ABSTRACT

Increased MMP-9 expression in the tumor microenvironment (TME) plays a crucial role in the extracellular matrix remodeling to facilitate cancer invasion and metastasis. However, the mechanism of MMP-9 upregulation in TME remains elusive. Since TGF-ß and TNF-α levels are elevated in TME, we asked whether these two agents interacted to induce/augment MMP-9 expression. Using a well-established MDA-MB-231 breast cancer model, we found that the synergy between TGF-ß and TNF-α led to MMP-9 upregulation at the transcriptional and translational levels, compared to treatments with each agent alone. Our in vitro findings are corroborated by co-expression of elevated MMP-9 with TGF-ß and TNF-α in human breast cancer tissues. Mechanistically, we found that the MMP-9 upregulation driven by TGF-ß/TNF-α cooperativity was attenuated by selective inhibition of the TGF-ßRI/Smad3 pathway. Comparable outcomes were observed upon inhibition of TGF-ß-induced phosphorylation of Smad2/3 and p38. As expected, the cells defective in Smad2/3 or p38-mediated signaling did not exhibit this synergistic induction of MMP-9. Importantly, the inhibition of histone methylation but not acetylation dampened the synergistic MMP-9 expression. Histone modification profiling further identified the H3K36me2 as an epigenetic regulatory mark of this synergy. Moreover, TGF-ß/TNF-α co-stimulation led to increased levels of the transcriptionally permissive dimethylation mark at H3K36 in the MMP-9 promoter. Comparable outcomes were noted in cells deficient in NSD2 histone methyltransferase. In conclusion, our findings support a cooperativity model in which TGF-ß could amplify the TNF-α-mediated MMP-9 production via chromatin remodeling and facilitate breast cancer invasion and metastasis.


Subject(s)
Breast Neoplasms , Gene Expression Regulation, Neoplastic , Matrix Metalloproteinase 9 , Neoplasm Metastasis , Transforming Growth Factor beta , Tumor Necrosis Factor-alpha , Humans , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Tumor Necrosis Factor-alpha/metabolism , Female , Transforming Growth Factor beta/metabolism , Cell Line, Tumor , Histones/metabolism , Methylation , Signal Transduction , Tumor Microenvironment
3.
Postepy Biochem ; 70(3): 336-347, 2024 09 30.
Article in Polish | MEDLINE | ID: mdl-39365566

ABSTRACT

Linker histones (H1) are basic proteins that are part of the nucleosome structure in the cell nucleus and are involved in the packaging of genetic material and the regulation of gene expression. As research progressed, it was discovered that linker histones constitute the largest group of histones in terms of variants found in humans. Even though the H1 variants differ slightly in the primary structure, they can perform different functions, undergo multiple post-translational modifications and differ in cellular localization. In addition to the nucleus, histones H1 can occur in the cytoplasm, on the cell surface and in the intercellular space. In these places, they play a supporting role for the immune system and act as signaling molecules. Changes in the levels of histones and their post-translational modifications have been associated with many human diseases and it is postulated that some of them may serve as biomarkers or therapeutic targets.


Subject(s)
Histones , Protein Processing, Post-Translational , Humans , Histones/metabolism
4.
Life Sci Alliance ; 7(12)2024 Dec.
Article in English | MEDLINE | ID: mdl-39353738

ABSTRACT

Wild soybean Glycine soja is the progenitor of cultivated soybean Glycine max Information on soybean functional centromeres is limited despite extensive genome analysis. These species are an ideal model for studying centromere dynamics for domestication and breeding. We performed a detailed chromatin immunoprecipitation analysis using centromere-specific histone H3 protein to delineate two distinct centromeric DNA sequences with unusual repeating units with monomer sizes of 90-92 bp (CentGm-1) and 413-bp (CentGm-4) shorter and longer than standard nucleosomes. These two unrelated DNA sequences with no sequence similarity are part of functional centromeres in both species. Our results provide a comparison of centromere properties between a cultivated and a wild species under the effect of the same kinetochore protein. Possible sequence homogenization specific to each chromosome could highlight the mechanism for evolutionary conservation of centromeric properties independent of domestication and breeding. Moreover, a unique barcode system to track each chromosome is developed using CentGm-4 units. Our results with a unifying centromere composition model using CentGm-1 and CentGm-4 superfamilies could have far-reaching implications for comparative and evolutionary genome research.


Subject(s)
Centromere , Chromosomes, Plant , Glycine max , Glycine max/genetics , Centromere/genetics , Chromosomes, Plant/genetics , DNA Barcoding, Taxonomic/methods , Domestication , Genome, Plant/genetics , Histones/genetics , Histones/metabolism , Plant Breeding/methods , DNA, Plant/genetics
5.
Sci Adv ; 10(40): eado8231, 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39356765

ABSTRACT

Despite novel therapeutic strategies, advanced-stage prostate cancer (PCa) remains highly lethal, pointing out the urgent need for effective therapeutic strategies. While dysregulation of the splicing process is considered a cancer hallmark, the role of certain splicing factors remains unknown in PCa. This study focuses on characterizing the levels and role of SRSF6 in this disease. Comprehensive analyses of SRSF6 alterations (copy number/mRNA/protein) were conducted across eight well-characterized PCa cohorts and the Hi-MYC transgenic model. SRSF6 was up-regulated in PCa samples, correlating with adverse clinical parameters. Functional assays, both in vitro (cell proliferation, migration, colony, and tumorsphere formation) and in vivo (xenograft tumors), demonstrated the impact of SRSF6 modulation on critical cancer hallmarks. Mechanistically, SRSF6 regulates the splicing pattern of the histone-chaperone HIRA, consequently affecting the activity of H3.3 in PCa and breast cancer cell models and disrupting pivotal oncogenic pathways (AR and E2F) in PCa cells. These findings underscore SRSF6 as a promising therapeutic target for PCa/advanced-stage PCa.


Subject(s)
Histone Chaperones , Prostatic Neoplasms , Serine-Arginine Splicing Factors , Humans , Serine-Arginine Splicing Factors/metabolism , Serine-Arginine Splicing Factors/genetics , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Animals , Cell Line, Tumor , Histone Chaperones/metabolism , Histone Chaperones/genetics , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , Gene Expression Regulation, Neoplastic , Mice , RNA Splicing , Cell Proliferation , Histones/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Phosphoproteins
6.
Proc Natl Acad Sci U S A ; 121(42): e2404058121, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39382996

ABSTRACT

The analysis of tissues of origin of cell-free DNA (cfDNA) is of research and diagnostic interest. Many studies focused on bisulfite treatment or immunoprecipitation protocols to assess the tissues of origin of cfDNA. DNA loss often occurs during such processes. Fragmentomics of cfDNA molecules has uncovered a wealth of information related to tissues of origin of cfDNA. There is still much room for the development of tools for assessing contributions from various tissues into plasma using fragmentomic features. Hence, we developed an approach to analyze the relative contributions of DNA from different tissues into plasma, by identifying characteristic fragmentation patterns associated with selected histone modifications. We named this technique as FRAGmentomics-based Histone modification Analysis (FRAGHA). Deduced placenta-specific histone H3 lysine 27 acetylation (H3K27ac)-associated signal correlated well with the fetal DNA fraction in maternal plasma (Pearson's r = 0.96). The deduced liver-specific H3K27ac-associated signal correlated with the donor-derived DNA fraction in liver transplantation recipients (Pearson's r = 0.92) and was significantly increased in patients with hepatocellular carcinoma (HCC) (P < 0.01, Wilcoxon rank-sum test). Significant elevations of erythroblasts-specific and colon-specific H3K27ac-associated signals were observed in patients with ß-thalassemia major and colorectal cancer, respectively. Furthermore, using the fragmentation patterns from tissue-specific H3K27ac regions, a machine learning algorithm was developed to enhance HCC detection, with an area under the curve (AUC) of up to 0.97. Finally, genomic regions with H3K27ac or histone H3 lysine 4 trimethylation (H3K4me3) were found to exhibit different fragmentomic patterns of cfDNA. This study has shed light on the relationship between cfDNA fragmentomics and histone modifications, thus expanding the armamentarium of liquid biopsy.


Subject(s)
Cell-Free Nucleic Acids , DNA Fragmentation , Histone Code , Histones , Nucleosomes , Humans , Nucleosomes/metabolism , Cell-Free Nucleic Acids/blood , Cell-Free Nucleic Acids/genetics , Histones/metabolism , Histones/blood , Female , Liver Neoplasms/blood , Liver Neoplasms/genetics , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/genetics , Pregnancy , Acetylation , Placenta/metabolism , Male
7.
Sci Rep ; 14(1): 23177, 2024 10 05.
Article in English | MEDLINE | ID: mdl-39369134

ABSTRACT

Although the role of low-density granulocytes (LDGs), neutrophils in the peripheral blood mononuclear cell (PBMC) fraction, and neutrophil extracellular traps (NETs) in assessing lupus disease severity is acknowledged, data specific to childhood-onset lupus remains scarce. This study analyzed 46 patients with childhood-onset systemic lupus erythematosus (82.6% females, mean age 14.5 ± 0.3 years), including 26 cases with normal complement levels and 20 with low complement levels, along with 20 healthy adult volunteers. Key parameters that distinguished healthy volunteers from lupus patients and differentiated between lupus patients with low and normal complement were serum interferon (IFN)-α, serum citrullinated histone 3 (CitH3), and extracellular traps (ETs) in LDGs. However, NETs (assessed by nuclear staining morphology), LDG abundance, and other parameters (such as endotoxemia, cytokines, and double-stranded (ds) DNA) did not show such differentiation. When lipopolysaccharide (LPS) was administered to LDGs in the PBMC fraction, it induced ETs in both low and normal complement groups, indicating the inducible nature of ETs. In adult healthy volunteers, activation by recombinant IFN-α or dsDNA in isolated neutrophils induced LDGs and NETs (identified using immunofluorescent staining for CitH3, myeloperoxidase, and neutrophil elastase) at 45 min and 3 h post-stimulation, respectively. Additionally, approximately half of the LDGs underwent late apoptosis at 3 h post-stimulation, as determined by flow cytometry analysis. Activation by IFN-α or dsDNA in LDGs also led to a more pronounced expression of CD66b, an adhesion molecule, compared to regular-density neutrophils, suggesting higher activity in LDGs. In conclusion, IFN-α and/or dsDNA in serum may transform regular-density neutrophils into LDGs before progressing to NETosis and apoptosis, potentially exacerbating lupus severity through cell death-induced self-antigens. Therefore, LDGs and ETs in LDGs could provide deeper insights into the pathophysiology of childhood-onset lupus.


Subject(s)
Extracellular Traps , Leukocytes, Mononuclear , Lupus Erythematosus, Systemic , Neutrophils , Humans , Extracellular Traps/metabolism , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/immunology , Female , Male , Adolescent , Leukocytes, Mononuclear/metabolism , Neutrophils/metabolism , Histones/metabolism , Adult , Interferon-alpha/blood , Interferon-alpha/metabolism , Child , Age of Onset , Granulocytes/metabolism , Complement System Proteins/metabolism , Lipopolysaccharides/pharmacology , Case-Control Studies
8.
Front Immunol ; 15: 1467774, 2024.
Article in English | MEDLINE | ID: mdl-39372408

ABSTRACT

Polycomb repressive complex 2 (PRC2) is an evolutionarily conserved epigenetic modifier responsible for tri-methylation of lysine 27 on histone H3 (H3K27me3). Previous studies have linked PRC2 to invariant natural killer T (iNKT) cell development, but its physiological and precise role remained unclear. To address this, we conditionally deleted Eed, a core subunit of PRC2, in mouse T cells. The results showed that Eed-deficient mice exhibited a severe reduction in iNKT cell numbers, particularly NKT1 and NKT17 cells, while conventional T cells and NKT2 cells remained intact. Deletion of Eed disrupted iNKT cell differentiation, leading to increased cell death, which was accompanied by a severe reduction in H3K27me3 levels and abnormal expression of Zbtb16, Cdkn2a, and Cdkn1a. Interestingly, Eed-deficient mice were highly susceptible to acetaminophen-induced liver injury and inflammation in an iNKT cell-dependent manner, highlighting the critical role of Eed-mediated H3K27me3 marks in liver-resident iNKT cells. These findings provide further insight into the epigenetic orchestration of iNKT cell-specific transcriptional programs.


Subject(s)
Histones , Mice, Knockout , Natural Killer T-Cells , Animals , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Mice , Histones/metabolism , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/metabolism , Cell Differentiation , Mice, Inbred C57BL , Histone Code , Epigenesis, Genetic , Acetaminophen/adverse effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Liver/metabolism , Liver/immunology , Liver/pathology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Promyelocytic Leukemia Zinc Finger Protein/metabolism , Promyelocytic Leukemia Zinc Finger Protein/genetics
9.
PLoS One ; 19(10): e0309689, 2024.
Article in English | MEDLINE | ID: mdl-39361575

ABSTRACT

Heavy water, containing the heavy hydrogen isotope, is toxic to cells, although the underlying mechanism remains incompletely understood. In addition, certain enzymatic proton transfer reactions exhibit kinetic isotope effects attributed to hydrogen isotopes and their temperature dependencies, indicative of quantum tunneling phenomena. However, the correlation between the biological effects of heavy water and the kinetic isotope effects mediated by hydrogen isotopes remains elusive. In this study, we elucidated the kinetic isotope effects arising from hydrogen isotopes of water and their temperature dependencies in vitro, focusing on deacetylation, DNA cleavage, and protein cleavage, which are crucial enzymatic reactions mediated by hydrolysis. Intriguingly, the intracellular isotope effects of heavy water, related to the in vitro kinetic isotope effects, significantly impeded multiple DNA double-strand break repair mechanisms crucial for cell survival. Additionally, heavy water exposure enhanced histone acetylation and associated transcriptional activation in cells, consistent with the in vitro kinetic isotope effects observed in histone deacetylation reactions. Moreover, as observed for the in vitro kinetic isotope effects, the cytotoxic effect on cell proliferation induced by heavy water exhibited temperature-dependency. These findings reveal the substantial impact of heavy water-induced isotope effects on cellular functions governed by hydrolytic enzymatic reactions, potentially mediated by quantum-level mechanisms underlying kinetic isotope effects.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Water , Kinetics , Hydrolysis , Humans , Water/chemistry , Water/metabolism , Histones/metabolism , Acetylation , Transcription, Genetic , Temperature , Cell Proliferation , DNA/metabolism
10.
Science ; 386(6718): eadl4492, 2024 Oct 11.
Article in English | MEDLINE | ID: mdl-39388542

ABSTRACT

Epigenetic reinforcement of T cell exhaustion is known to be a major barrier limiting T cell responses during immunotherapy. However, the core epigenetic regulators restricting antitumor immunity during prolonged antigen exposure are not clear. We investigated three commonly mutated epigenetic regulators that promote clonal hematopoiesis to determine whether they affect T cell stemness and response to checkpoint blockade immunotherapy. CD8 T cells lacking Dnmt3a, Tet2, or Asxl1 preserved a progenitor-exhausted (Tpex) population for more than 1 year during chronic antigen exposure without undergoing malignant transformation. Asxl1 controlled the self-renewal capacity of T cells and reduced CD8 T cell differentiation through H2AK119 ubiquitination and epigenetic modification of the polycomb group-repressive deubiquitinase pathway. Asxl1-deficient T cells synergized with anti-PD-L1 immunotherapy to improve tumor control in experimental models and conferred a survival advantage to mutated T cells from treated patients.


Subject(s)
CD8-Positive T-Lymphocytes , Clonal Hematopoiesis , DNA Methyltransferase 3A , DNA-Binding Proteins , Dioxygenases , Epigenesis, Genetic , Immunotherapy , Proto-Oncogene Proteins , Repressor Proteins , CD8-Positive T-Lymphocytes/immunology , Animals , Mice , Humans , Repressor Proteins/genetics , Repressor Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Cell Differentiation , Immune Checkpoint Inhibitors/therapeutic use , Histones/metabolism , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics
11.
Mol Med ; 30(1): 169, 2024 Oct 10.
Article in English | MEDLINE | ID: mdl-39390356

ABSTRACT

External constraints, such as development, disease, and environment, can induce changes in epigenomic patterns that may profoundly impact the health trajectory of fetuses and neonates into adulthood, influencing conditions like obesity. Epigenetic modifications encompass processes including DNA methylation, covalent histone modifications, and RNA-mediated regulation. Beyond forward cellular differentiation (cell programming), terminally differentiated cells are reverted to a pluripotent or even totipotent state, that is, cellular reprogramming. Epigenetic modulators facilitate or erase histone and DNA modifications both in vivo and in vitro during programming and reprogramming. Noticeably, obesity is a complex metabolic disorder driven by both genetic and environmental factors. Increasing evidence suggests that epigenetic modifications play a critical role in the regulation of gene expression involved in adipogenesis, energy homeostasis, and metabolic pathways. Hence, we discuss the mechanisms by which epigenetic interventions influence obesity, focusing on DNA methylation, histone modifications, and non-coding RNAs. We also analyze the methodologies that have been pivotal in uncovering these epigenetic regulations, i.e., Large-scale screening has been instrumental in identifying genes and pathways susceptible to epigenetic control, particularly in the context of adipogenesis and metabolic homeostasis; Single-cell RNA sequencing (scRNA-seq) provides a high-resolution view of gene expression patterns at the individual cell level, revealing the heterogeneity and dynamics of epigenetic regulation during cellular differentiation and reprogramming; Chromatin immunoprecipitation (ChIP) assays, focused on candidate genes, have been crucial for characterizing histone modifications and transcription factor binding at specific genomic loci, thereby elucidating the epigenetic mechanisms that govern cellular programming; Somatic cell nuclear transfer (SCNT) and cell fusion techniques have been employed to study the epigenetic reprogramming accompanying cloning and the generation of hybrid cells with pluripotent characteristics, etc. These approaches have been instrumental in identifying specific epigenetic marks and pathways implicated in obesity, providing a foundation for developing targeted therapeutic interventions. Understanding the dynamic interplay between epigenetic regulation and cellular programming is crucial for advancing mechanism and clinical management of obesity.


Subject(s)
Cellular Reprogramming , DNA Methylation , Epigenesis, Genetic , Obesity , Humans , Obesity/genetics , Obesity/metabolism , Animals , Cellular Reprogramming/genetics , Cell Differentiation/genetics , Adipogenesis/genetics , Histones/metabolism , Histone Code , Epigenomics/methods
12.
J Transl Med ; 22(1): 922, 2024 Oct 10.
Article in English | MEDLINE | ID: mdl-39390495

ABSTRACT

BACKGROUND: Recurrent spontaneous abortion (RSA) is defined as two or more consecutive spontaneous abortions before 20 weeks with the same spouse [1]. However, approximately 50% of RSA cases of unknown cause are classified as unexplained recurrent spontaneous abortion (URSA). Potential factors include decreased trophoblast cell migration and invasion, leading to impaired placental implantation and maintenance of the normal maternal-fetal interface. However, the mechanism of this pathogenesis remains unknown. In this study, we investigated the potential role and mechanism of KLF4 in regulating URSA by influencing the invasion and migration ability of trophoblast cells. METHODS: We firstly identified 817 differentially expressed genes by performing a difference analysis of the dataset GSE121950 [2] related to recurrent abortion, and intersected the top 10 genes obtained respectively by the three algorithms: DMNC, MNC, and EPC using Venn Diagram.To detect the expression levels of core genes, villi samples were obtained from normal pregnant women and patients with URSA. RT-qPCR analysis revealed a significant difference in KLF4 mRNA expression and KLF4 was then analyzed. Trophoblast cell lines HTR8 and JEG3 were used to investigate the effect of KLF4 on trophoblastic function. Wound healing and transwell assays was performed to detect the invasion and migration of trophoblast cells. The expression of epithelial-mesenchymal transition(EMT) molecules were detected by RT-qPCR and western blot. Promoter detection and epigenetic modification were detected by chromatin immunoprecipitation (ChIP) assay. Molecular nuclear localization was detected by immunofluorescence and subcellular fractionation. Miscarried mice model was used to study the effects of KLF4 on URSA induced by reduced trophoblast invasion and migration. RESULTS: KLF4 is highly expressed in the villi of patients with URSA. KLF4 inhibits the expression level of H3R2ME2a in trophoblast cells by regulating the transcriptional level and nuclear translocation of PRMT6, thereby inhibiting the possible regulatory mechanism of trophoblastic invasion and providing a potential treatment strategy for URSA in vivo. CONCLUSIONS: The KLF4/PRMT6/H3R2ME2a axis regulates mechanisms associated with unexplained recurrent spontaneous abortion by regulating trophoblast function.


Subject(s)
Abortion, Habitual , Cell Movement , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Trophoblasts , Trophoblasts/metabolism , Trophoblasts/pathology , Kruppel-Like Factor 4/metabolism , Female , Humans , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Pregnancy , Abortion, Habitual/metabolism , Abortion, Habitual/genetics , Abortion, Habitual/pathology , Cell Movement/genetics , Animals , Mice , Promoter Regions, Genetic/genetics , Adult , Epithelial-Mesenchymal Transition/genetics , Chorionic Villi/metabolism , Gene Expression Regulation , Cell Line , RNA, Messenger/metabolism , RNA, Messenger/genetics , DNA Methylation/genetics , Histones/metabolism
13.
Genome Biol ; 25(1): 262, 2024 Oct 10.
Article in English | MEDLINE | ID: mdl-39390531

ABSTRACT

BACKGROUND: In response to drought stress (DS), plants undergo complex processes that entail significant transcriptome reprogramming. However, the intricate relationship between the dynamic alterations in the three-dimensional (3D) genome and the modulation of gene co-expression in drought responses remains a relatively unexplored area. RESULTS: In this study, we reconstruct high-resolution 3D genome maps based on genomic regions marked by H3K9ac, an active histone modification that dynamically responds to soil water variations in rice. We discover a genome-wide disconnection of 3D genome contact upon DS with over 10,000 chromatin loops lost, which are partially recovered in the subsequent re-watering. Loops integrating promoter-promoter interactions (PPI) contribute to gene expression in addition to basal H3K9ac modifications. Moreover, H3K9ac-marked promoter regions with high affinities in mediating PPIs, termed as super-promoter regions (SPRs), integrate spatially clustered PPIs in a super-enhancer-like manner. Interestingly, the knockout mutation of OsbZIP23, a well-defined DS-responsive transcription factor, leads to the disassociation of over 80% DS-specific PPIs and decreased expression of the corresponding genes under DS. As a case study, we show how OsbZIP23 integrates the PPI cluster formation and the co-expression of four dehydrin genes, RAB16A-D, through targeting the RAB16C SPR in a stress signaling-dependent manner. CONCLUSIONS: Our high-resolution 3D genome maps unveil the principles and details of dynamic genome folding in response to water supply variations and illustrate OsbZIP23 as an indispensable integrator of the yet unique 3D genome organization that is essential for gene co-expression under DS in rice.


Subject(s)
Chromatin , Droughts , Gene Expression Regulation, Plant , Histones , Oryza , Plant Proteins , Promoter Regions, Genetic , Oryza/genetics , Oryza/metabolism , Chromatin/metabolism , Histones/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Stress, Physiological/genetics , Genome, Plant , Transcription Factors/metabolism , Transcription Factors/genetics
14.
Genome Biol ; 25(1): 263, 2024 Oct 10.
Article in English | MEDLINE | ID: mdl-39390582

ABSTRACT

BACKGROUND: Methylation of histone 3 lysine 36 (H3K36me) has emerged as an essential epigenetic component for the faithful regulation of gene expression. Despite its importance in development and disease, how the molecular agents collectively shape the H3K36me landscape is unclear. RESULTS: We use mouse mesenchymal stem cells to perturb the H3K36me methyltransferases (K36MTs) and infer the activities of the five most prominent enzymes: SETD2, NSD1, NSD2, NSD3, and ASH1L. We find that H3K36me2 is the most abundant of the three methylation states and is predominantly deposited at intergenic regions by NSD1, and partly by NSD2. In contrast, H3K36me1/3 are most abundant within exons and are positively correlated with gene expression. We demonstrate that while SETD2 deposits most H3K36me3, it may also deposit H3K36me2 within transcribed genes. Additionally, loss of SETD2 results in an increase of exonic H3K36me1, suggesting other (K36MTs) prime gene bodies with lower methylation states ahead of transcription. While NSD1/2 establish broad intergenic H3K36me2 domains, NSD3 deposits H3K36me2 peaks on active promoters and enhancers. Meanwhile, the activity of ASH1L is restricted to the regulatory elements of developmentally relevant genes, and our analyses implicate PBX2 as a potential recruitment factor. CONCLUSIONS: Within genes, SETD2 primarily deposits H3K36me3, while the other K36MTs deposit H3K36me1/2 independently of SETD2 activity. For the deposition of H3K36me1/2, we find a hierarchy of K36MT activities where NSD1 > NSD2 > NSD3 > ASH1L. While NSD1 and NSD2 are responsible for most genome-wide propagation of H3K36me2, the activities of NSD3 and ASH1L are confined to active regulatory elements.


Subject(s)
DNA-Binding Proteins , Histone-Lysine N-Methyltransferase , Histones , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Animals , Mice , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Histones/metabolism , Methylation , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Histone Methyltransferases/metabolism
15.
Stem Cell Res Ther ; 15(1): 350, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39380045

ABSTRACT

BACKGROUND: The histone-lysine N-methyltransferase SMYD1, which is specific to striated muscle, plays a crucial role in regulating early heart development. Its deficiency has been linked to the occurrence of congenital heart disease. Nevertheless, the precise mechanism by which SMYD1 deficiency contributes to congenital heart disease remains unclear. METHODS: We established a SMYD1 knockout pluripotent stem cell line and a doxycycline-inducible SMYD1 expression pluripotent stem cell line to investigate the functions of SMYD1 utilizing an in vitro-directed myocardial differentiation model. RESULTS: Cardiomyocytes lacking SMYD1 displayed drastically diminished differentiation efficiency, concomitant with heightened proliferation capacity of cardiac progenitor cells during the early cardiac differentiation stage. These cellular phenotypes were confirmed through experiments inducing the re-expression of SMYD1. Transcriptome sequencing and small molecule inhibitor intervention suggested that the GSK3ß/ß-catenin&ERK signaling pathway was involved in the proliferation of cardiac progenitor cells. Chromatin immunoprecipitation demonstrated that SMYD1 acted as a transcriptional activator of GSK3ß through histone H3 lysine 4 trimethylation. Additionally, dual-luciferase analyses indicated that SMYD1 could interact with the promoter region of GSK3ß, thereby augmenting its transcriptional activity. Moreover, administering insulin and Insulin-like growth factor 1 can enhance the efficacy of myocardial differentiation in SMYD1 knockout cells. CONCLUSIONS: Our research indicated that the participation of SMYD1 in the GSK3ß/ß-catenin&ERK signaling cascade modulated the proliferation of cardiac progenitor cells during myocardial differentiation. This process was partly reliant on the transcription of GSK3ß. Our research provided a novel insight into the genetic modification effect of SMYD1 during early myocardial differentiation. The findings were essential to the molecular mechanism and potential interventions for congenital heart disease.


Subject(s)
Cell Differentiation , Cell Proliferation , Glycogen Synthase Kinase 3 beta , Histone-Lysine N-Methyltransferase , Myocytes, Cardiac , beta Catenin , Humans , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , beta Catenin/metabolism , beta Catenin/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , MAP Kinase Signaling System , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Histones/metabolism , Muscle Proteins/metabolism , Muscle Proteins/genetics , Multipotent Stem Cells/metabolism , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Cell Line , DNA-Binding Proteins , Transcription Factors
16.
Elife ; 122024 Oct 07.
Article in English | MEDLINE | ID: mdl-39373720

ABSTRACT

Pediatric diffuse midline gliomas (pDMG) are an aggressive type of childhood cancer with a fatal outcome. Their major epigenetic determinism has become clear, notably with the identification of K27M mutations in histone H3. However, the synergistic oncogenic mechanisms that induce and maintain tumor cell phenotype have yet to be deciphered. In 20 to 30% of cases, these tumors have an altered BMP signaling pathway with an oncogenic mutation on the BMP type I receptor ALK2, encoded by ACVR1. However, the potential impact of the BMP pathway in tumors non-mutated for ACVR1 is less clear. By integrating bulk, single-cell, and spatial transcriptomic data, we show here that the BMP signaling pathway is activated at similar levels between ACVR1 wild-type and mutant tumors and identify BMP2 and BMP7 as putative activators of the pathway in a specific subpopulation of cells. By using both pediatric isogenic glioma lines genetically modified to overexpress H3.3K27M and patients-derived DIPG cell lines, we demonstrate that BMP2/7 synergizes with H3.3K27M to induce a transcriptomic rewiring associated with a quiescent but invasive cell state. These data suggest a generic oncogenic role for the BMP pathway in gliomagenesis of pDMG and pave the way for specific targeting of downstream effectors mediating the K27M/BMP crosstalk.


Subject(s)
Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 7 , Glioma , Histones , Humans , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 7/metabolism , Bone Morphogenetic Protein 7/genetics , Histones/metabolism , Histones/genetics , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Cell Line, Tumor , Signal Transduction , Activin Receptors, Type I/metabolism , Activin Receptors, Type I/genetics , Child , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Neoplasm Invasiveness , Mutation , Gene Expression Regulation, Neoplastic
17.
Sci Adv ; 10(40): eado3514, 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39365860

ABSTRACT

A hallmark of addiction is the ability of drugs of abuse to trigger relapse after periods of prolonged abstinence. Here, we describe an epigenetic mechanism whereby chronic cocaine exposure causes lasting chromatin and downstream transcriptional modifications in the nucleus accumbens (NAc), a critical brain region controlling motivation. We link prolonged withdrawal from cocaine to the depletion of the histone variant H2A.Z, coupled with increased genome accessibility and latent priming of gene transcription, in D1 dopamine receptor-expressing medium spiny neurons (D1 MSNs) that relate to aberrant gene expression upon drug relapse. The histone chaperone ANP32E removes H2A.Z from chromatin, and we demonstrate that D1 MSN-selective Anp32e knockdown prevents cocaine-induced H2A.Z depletion and blocks cocaine's rewarding actions. By contrast, very different effects of cocaine exposure, withdrawal, and relapse were found for D2 MSNs. These findings establish histone variant exchange as an important mechanism and clinical target engaged by drugs of abuse to corrupt brain function and behavior.


Subject(s)
Cocaine , Epigenesis, Genetic , Histones , Nucleus Accumbens , Nucleus Accumbens/metabolism , Nucleus Accumbens/drug effects , Cocaine/pharmacology , Animals , Epigenesis, Genetic/drug effects , Histones/metabolism , Mice , Male , Gene Expression Regulation/drug effects , Cocaine-Related Disorders/genetics , Cocaine-Related Disorders/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D1/genetics , Neurons/metabolism , Neurons/drug effects , Chromatin/metabolism , Chromatin/genetics
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL