Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Nature ; 631(8021): 678-685, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38961301

ABSTRACT

Pericentric heterochromatin is a critical component of chromosomes marked by histone H3 K9 (H3K9) methylation1-3. However, what recruits H3K9-specific histone methyltransferases to pericentric regions in vertebrates remains unclear4, as does why pericentric regions in different species share the same H3K9 methylation mark despite lacking highly conserved DNA sequences2,5. Here we show that zinc-finger proteins ZNF512 and ZNF512B specifically localize at pericentric regions through direct DNA binding. Notably, both ZNF512 and ZNF512B are sufficient to initiate de novo heterochromatin formation at ectopically targeted repetitive regions and pericentric regions, as they directly recruit SUV39H1 and SUV39H2 (SUV39H) to catalyse H3K9 methylation. SUV39H2 makes a greater contribution to H3K9 trimethylation, whereas SUV39H1 seems to contribute more to silencing, probably owing to its preferential association with HP1 proteins. ZNF512 and ZNF512B from different species can specifically target pericentric regions of other vertebrates, because the atypical long linker residues between the zinc-fingers of ZNF512 and ZNF512B offer flexibility in recognition of non-consecutively organized three-nucleotide triplets targeted by each zinc-finger. This study addresses two long-standing questions: how constitutive heterochromatin is initiated and how seemingly variable pericentric sequences are targeted by the same set of conserved machinery in vertebrates.


Subject(s)
Centromere , Evolution, Molecular , Heterochromatin , Histone-Lysine N-Methyltransferase , Histones , Nucleotide Motifs , Animals , Humans , Mice , Centromere/genetics , Centromere/metabolism , Chickens , Chromobox Protein Homolog 5 , Gene Silencing , Heterochromatin/metabolism , Heterochromatin/chemistry , Heterochromatin/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/chemistry , Histones/metabolism , Histones/chemistry , Lancelets , Methylation , Petromyzon , Repressor Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Snakes , Xenopus laevis , Zebrafish , Zinc Fingers
2.
Immunity ; 49(4): 695-708.e4, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30291027

ABSTRACT

B cells can present antigens to CD4+ T cells, but it is thought that dendritic cells (DCs) are the primary initiators of naive CD4+ T cell responses. Nanoparticles, including virus-like particles (VLPs), are attractive candidates as carriers for vaccines and drug delivery. Using RNA phage Qß-derived VLP (Qß-VLP) as a model antigen, we found that antigen-specific B cells were the dominant antigen-presenting cells that initiated naive CD4+ T cell activation. B cells were sufficient to induce T follicular helper cell development in the absence of DCs. Qß-specific B cells promoted CD4+ T cell proliferation and differentiation via cognate interactions and through Toll-like receptor signaling-mediated cytokine production. Antigen-specific B cells were also involved in initiating CD4+ T cell responses during immunization with inactivated influenza virus. These findings have implications for the rational design of nanoparticles as vaccine candidates, particularly for therapeutic vaccines that aim to break immune tolerance.


Subject(s)
Antigen-Presenting Cells/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Immunization/methods , Influenza Vaccines/immunology , Animals , Antigen Presentation/immunology , Antigens, Viral/chemistry , Antigens, Viral/immunology , Cell Differentiation/immunology , Cytokines/immunology , Cytokines/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nanoparticles/chemistry , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Toll-Like Receptors/immunology , Vaccines, Inactivated/immunology
3.
Nature ; 564(7734): 136-140, 2018 12.
Article in English | MEDLINE | ID: mdl-30487604

ABSTRACT

Postnatal growth of mammalian oocytes is accompanied by a progressive gain of DNA methylation, which is predominantly mediated by DNMT3A, a de novo DNA methyltransferase1,2. Unlike the genome of sperm and most somatic cells, the oocyte genome is hypomethylated in transcriptionally inert regions2-4. However, how such a unique feature of the oocyte methylome is determined and its contribution to the developmental competence of the early embryo remains largely unknown. Here we demonstrate the importance of Stella, a factor essential for female fertility5-7, in shaping the oocyte methylome in mice. Oocytes that lack Stella acquire excessive DNA methylation at the genome-wide level, including in the promoters of inactive genes. Such aberrant hypermethylation is partially inherited by two-cell-stage embryos and impairs zygotic genome activation. Mechanistically, the loss of Stella leads to ectopic nuclear accumulation of the DNA methylation regulator UHRF18,9, which results in the mislocalization of maintenance DNA methyltransferase DNMT1 in the nucleus. Genetic analysis confirmed the primary role of UHRF1 and DNMT1 in generating the aberrant DNA methylome in Stella-deficient oocytes. Stella therefore safeguards the unique oocyte epigenome by preventing aberrant de novo DNA methylation mediated by DNMT1 and UHRF1.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation , Epigenesis, Genetic , Oocytes/metabolism , Repressor Proteins/metabolism , Animals , CCAAT-Enhancer-Binding Proteins , Cell Line , Cell Nucleus/metabolism , Chromosomal Proteins, Non-Histone , DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , Embryonic Development , Female , Genome/genetics , Humans , Mice , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Repressor Proteins/deficiency , Repressor Proteins/genetics , Ubiquitin-Protein Ligases , Zygote/metabolism
4.
Mol Cell ; 64(5): 913-925, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27840027

ABSTRACT

TET family enzymes successively oxidize 5-methylcytosine to 5-hydroxymethylcytosine, 5-formylcytosine, and 5-carboxylcytosine, leading to eventual demethylation. 5hmC and TET enzymes occupy distinct chromatin regions, suggesting unknown mechanisms controlling the fate of 5hmC within diverse chromatin environments. Here, we report that SALL4A preferentially associates with 5hmC in vitro and occupies enhancers in mouse embryonic stem cells in a largely TET1-dependent manner. Although most 5hmC at SALL4A peaks undergoes further oxidation, this process is abrogated upon deletion of Sall4 gene, with a concomitant reduction of TET2 at these regions. Thus, SALL4A facilitates further oxidation of 5hmC at its binding sites, which requires its 5hmC-binding activity and TET2, supporting a collaborative action between SALL4A and TET proteins in regulating stepwise oxidation of 5mC at enhancers. Our study identifies SALL4A as a 5hmC binder, which facilitates 5hmC oxidation by stabilizing TET2 association, thereby fine-tuning expression profiles of developmental genes in mouse embryonic stem cells.


Subject(s)
5-Methylcytosine/metabolism , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Transcription Factors/metabolism , Animals , DNA Methylation , Dioxygenases , Enhancer Elements, Genetic/physiology , Mice , Oxidation-Reduction , Proto-Oncogene Proteins/metabolism , Transcription, Genetic
5.
Genes Dev ; 29(4): 379-93, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25637356

ABSTRACT

GLP and G9a are major H3K9 dimethylases and are essential for mouse early embryonic development. GLP and G9a both harbor ankyrin repeat domains that are capable of binding H3K9 methylation. However, the functional significance of their recognition of H3K9 methylation is unknown. Here, we report that the histone methyltransferase activities of GLP and G9a are stimulated by neighboring nucleosomes that are premethylated at H3K9. These stimulation events function in cis and are dependent on the H3K9 methylation binding activities of ankyrin repeat domains of GLP and G9a. Disruption of the H3K9 methylation-binding activity of GLP in mice causes growth retardation of embryos, ossification defects of calvaria, and postnatal lethality due to starvation of the pups. In mouse embryonic stem cells (ESCs) harboring a mutant GLP that lacks H3K9me1-binding activity, critical pluripotent genes, including Oct4 and Nanog, display inefficient establishment of H3K9me2 and delayed gene silencing during differentiation. Collectively, our study reveals a new activation mechanism for GLP and G9a that plays an important role in ESC differentiation and mouse viability.


Subject(s)
Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Nucleosomes/metabolism , Animals , Cell Differentiation , Craniofacial Abnormalities/genetics , Embryonic Stem Cells/cytology , Female , Fetal Growth Retardation/genetics , Gene Silencing , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Homeodomain Proteins/genetics , Male , Methylation , Mice , Mice, Inbred C57BL , Mutation , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Protein Binding
6.
Proc Natl Acad Sci U S A ; 115(17): E4013-E4022, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29636421

ABSTRACT

Epigenetic silencing can be mediated by various mechanisms, and many regulators remain to be identified. Here, we report a genome-wide siRNA screening to identify regulators essential for maintaining gene repression of a CMV promoter silenced by DNA methylation. We identified CSE1L (chromosome segregation 1 like) as an essential factor for the silencing of the reporter gene and many endogenous methylated genes. CSE1L depletion did not cause DNA demethylation. On the other hand, the methylated genes derepressed by CSE1L depletion largely overlapped with methylated genes that were also reactivated by treatment with histone deacetylase inhibitors (HDACi). Gene silencing defects observed upon CSE1L depletion were linked to its nuclear import function for certain protein cargos because depletion of other factors involved in the same nuclear import pathway, including KPNAs and KPNB1 proteins, displayed similar derepression profiles at the genome-wide level. Therefore, CSE1L appears to be critical for the nuclear import of certain key repressive proteins. Indeed, NOVA1, HDAC1, HDAC2, and HDAC8, genes known as silencing factors, became delocalized into cytosol upon CSE1L depletion. This study suggests that the cargo specificity of the protein nuclear import system may impact the selectivity of gene silencing.


Subject(s)
Cell Nucleus/metabolism , Cellular Apoptosis Susceptibility Protein/metabolism , Gene Silencing/physiology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Cell Line , Cell Nucleus/genetics , Cellular Apoptosis Susceptibility Protein/genetics , DNA Methylation/drug effects , Gene Silencing/drug effects , Genome-Wide Association Study , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Neuro-Oncological Ventral Antigen , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , beta Karyopherins/genetics , beta Karyopherins/metabolism
7.
J Biol Chem ; 294(17): 6785-6795, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30824537

ABSTRACT

IL-32 is a cytokine involved in proinflammatory immune responses to bacterial and viral infections. However, the role of epigenetic events in the regulation of IL-32 gene expression is understudied. Here we show that IL-32 is repressed by DNA methylation in HEK293 cells. Using ChIP sequencing, locus-specific methylation analysis, CRISPR/Cas9-mediated genome editing, and RT-qPCR (quantitative RT-PCR) and immunoblot assays, we found that short-term treatment (a few hours) with the proinflammatory cytokine tumor necrosis factor α (TNFα) activates IL-32 in a DNA demethylation-independent manner. In contrast, prolonged TNFα treatment (several days) induced DNA demethylation at the promoter and a CpG island in the IL-32 gene in a TET (ten-eleven translocation) family enzyme- and NF-κB-dependent manner. Notably, the hypomethylation status of transcriptional regulatory elements in IL-32 was maintained for a long time (several weeks), causing elevated IL-32 expression even in the absence of TNFα. Considering that IL-32 can, in turn, induce TNFα expression, we speculate that such feedforward events may contribute to the transition from an acute inflammatory response to chronic inflammation.


Subject(s)
DNA Demethylation/drug effects , Inflammation Mediators/pharmacology , Interleukins/genetics , Tumor Necrosis Factor-alpha/pharmacology , CpG Islands , Cyclic AMP Response Element-Binding Protein/metabolism , Gene Silencing , HEK293 Cells , Humans , NF-kappa B/metabolism , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid , Transcription, Genetic , Up-Regulation
8.
J Biol Chem ; 294(22): 8907-8917, 2019 05 31.
Article in English | MEDLINE | ID: mdl-31018966

ABSTRACT

Stella is a maternal gene required for oogenesis and early embryogenesis. Stella overexpression in somatic cells causes global demethylation. As we have recently shown, Stella sequesters nuclear ubiquitin-like with PHD and RING finger domains 1 (UHRF1), a RING finger-type E3 ubiquitin ligase essential for DNA methylation mediated by DNA methyltransferase 1 and triggers global demethylation. Here, we report an overexpressed mutant Stella protein without nuclear export activity surprisingly retained its ability to cause global demethylation. By combining biochemical interaction assays, isothermal titration calorimetry, immunostaining, and live-cell imaging with fluorescence recovery after photobleaching, we found that Stella disrupts UHRF1's association with chromatin by directly binding to the plant homeodomain of UHRF1 and competing for the interaction between UHRF1 and the histone H3 tail. Consistently, overexpression of Stella mutants that do not directly interact with UHRF1 fails to cause genome-wide demethylation. In the presence of nuclear Stella, UHRF1 could not bind to chromatin and exhibited increased dynamics in the nucleus. Our results indicate that Stella employs a multilayered mechanism to achieve robust UHRF1 inhibition, which involves the dissociation from chromatin and cytoplasmic sequestration of UHRF1.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA Demethylation , Ubiquitin-Protein Ligases/metabolism , Active Transport, Cell Nucleus , CCAAT-Enhancer-Binding Proteins/chemistry , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , HEK293 Cells , Histones/metabolism , Humans , Mutagenesis , Protein Binding , Protein Domains , Ubiquitin-Protein Ligases/chemistry
9.
J Biol Chem ; 293(19): 7423-7436, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29559556

ABSTRACT

Regulation of gene expression by epigenetic modifications such as DNA methylation is crucial for developmental and disease processes, including cell differentiation and cancer development. Genes repressed by DNA methylation can be derepressed by various compounds that target DNA methyltransferases, histone deacetylases, and other regulatory factors. However, some additional, unknown mechanisms that promote DNA methylation-mediated gene silencing may exist. Chemical agents that can counteract the effects of epigenetic repression that is not regulated by DNA methyltransferases or histone deacetylases therefore may be of research interest. Here, we report the results of a high-throughput screen using a 308,251-member chemical library to identify potent small molecules that derepress an EGFP reporter gene silenced by DNA methylation. Seven hit compounds were identified that did not directly target bulk DNA methylation or histone acetylation. Analyzing the effect of these compounds on endogenous gene expression, we discovered that three of these compounds (compounds LX-3, LX-4, and LX-5) selectively activate the p38 mitogen-activated protein kinase (MAPK) pathway and derepress a subset of endogenous genes repressed by DNA methylation. Selective agonists of the p38 pathway have been lacking, and our study now provides critical compounds for studying this pathway and p38 MAPK-targeted genes repressed by DNA methylation.


Subject(s)
DNA Methylation/drug effects , Gene Expression Regulation/drug effects , Small Molecule Libraries/pharmacology , Acetylation , Animals , DNA Modification Methylases/antagonists & inhibitors , Enzyme Activation , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins/genetics , HEK293 Cells , Histone Deacetylases/metabolism , Histones/metabolism , Humans , MAP Kinase Signaling System , Mice , NIH 3T3 Cells , Phosphorylation , Small Molecule Libraries/chemistry , Structure-Activity Relationship , p38 Mitogen-Activated Protein Kinases/metabolism
10.
J Biol Chem ; 291(34): 17692-705, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27382058

ABSTRACT

Epigenetic systems are well known for the roles they play in regulating the differential expression of the same genome in different cell types. However, epigenetic systems can also directly impact genomic integrity by protecting genetic sequences. Using an experimental evolutionary approach, we studied rates of mutation in the fission yeast Schizosaccharomyces pombe strains that lacked genes encoding several epigenetic regulators or mismatch repair components. We report that loss of a functional mismatch repair pathway in S. pombe resulted in the preferential enrichment of mutations in euchromatin, indicating that the mismatch repair machinery preferentially protected genetic fidelity in euchromatin. This preference is probably determined by differences in the accessibility of chromatin at distinct chromatin regions, which is supported by our observations that chromatin accessibility positively correlated with mutation rates in S. pombe or human cancer samples with deficiencies in mismatch repair. Importantly, such positive correlation was not observed in S. pombe strains or human cancer samples with functional mismatch repair machinery.


Subject(s)
Chromatin/metabolism , DNA Mismatch Repair , Epigenesis, Genetic , Gene Expression Regulation, Fungal , Gene Expression Regulation, Neoplastic , Mutation , Neoplasms/metabolism , Schizosaccharomyces/metabolism , Chromatin/genetics , Chromatin/pathology , Humans , Neoplasms/genetics , Schizosaccharomyces/genetics
11.
RNA Biol ; 13(10): 1011-1024, 2016 Oct 02.
Article in English | MEDLINE | ID: mdl-27560004

ABSTRACT

Among all tissues of the metazoa, the transcritpome of testis displays the highest diversity and specificity. However, its composition and dynamics during spermatogenesis have not been fully understood. Here, we have identified 20,639 message RNAs (mRNAs), 7,168 long non-coding RNAs (lncRNAs) and 15,101 circular RNAs (circRNAs) in mouse spermatogenic cells, and found many of them were specifically expressed in testes. lncRNAs are significantly more testis-specific than mRNAs. At all stages, mRNAs are generally more abundant than lncRNAs, and linear transcripts are more abundant than circRNAs. We showed that the productions of circRNAs and piRNAs were highly regulated instead of random processes. Based on the results of a small-scale functional screening experiment using cultured mouse spermatogonial stem cells, many evolutionarily conserved lncRNAs are likely to play roles in spermatogenesis. Typical classes of transcription factor binding sites are enriched in the promoters of testis-specific m/lncRNA genes. Target genes of CREM and RFX2, 2 key TFs for spermatogenesis, were further validated by using ChIP-chip assays and RNA-seq on RFX2-knockout spermatogenic cells. Our results contribute to the current understanding of the transcriptomic complexity of spermatogenic cells and provide a valuable resource from which many candidate genes may be selected for further functional studies.

12.
PLoS Genet ; 9(6): e1003558, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23754967

ABSTRACT

Previously, we reported that little canonical (H3.1-H4)(2) tetramers split to form "hybrid" tetramers consisted of old and new H3.1-H(4) dimers, but approximately 10% of (H3.3-H4)2 tetramers split during each cell cycle. In this report, we mapped the H3.3 nucleosome occupancy, the H3.3 nucleosome turnover rate and H3.3 nucleosome splitting events at the genome-wide level. Interestingly, H3.3 nucleosome turnover rate at the transcription starting sites (TSS) of genes with different expression levels display a bimodal distribution rather than a linear correlation towards the transcriptional activity, suggesting genes are either active with high H3.3 nucleosome turnover or inactive with low H3.3 nucleosome turnover. H3.3 nucleosome splitting events are enriched at active genes, which are in fact better markers for active transcription than H3.3 nucleosome occupancy itself. Although both H3.3 nucleosome turnover and splitting events are enriched at active genes, these events only display a moderate positive correlation, suggesting H3.3 nucleosome splitting events are not the mere consequence of H3.3 nucleosome turnover. Surprisingly, H3.3 nucleosomes with high splitting index are remarkably enriched at enhancers in a cell-type specific manner. We propose that the H3.3 nucleosomes at enhancers may be split by an active mechanism to regulate cell-type specific transcription.


Subject(s)
DNA Replication/genetics , Enhancer Elements, Genetic , Histones/genetics , Nucleosomes/genetics , Cell Cycle/genetics , Chromatin/genetics , Chromatin/ultrastructure , Chromatin Assembly and Disassembly , HeLa Cells , Histones/chemistry , Humans , Nucleosomes/ultrastructure , Protein Multimerization/genetics , Regulatory Sequences, Nucleic Acid
13.
J Biol Chem ; 288(43): 30832-42, 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-24019522

ABSTRACT

Histone H3 lysine 27 (H3K27) methylation and H2A monoubiquitination (ubH2A) are two closely related histone modifications that regulate Polycomb silencing. Previous studies reported that H3K27 trimethylation (H3K27me3) rarely coexists with H3K36 di- or tri-methylation (H3K36me2/3) on the same histone H3 tails, which is partially controlled by the direct inhibition of the enzymatic activity of H3K27-specific methyltransferase PRC2. By contrast, H3K27 methylation does not affect the catalytic activity of H3K36-specific methyltransferases, suggesting other Polycomb mechanism(s) may negatively regulate the H3K36-specific methyltransferase(s). In this study, we established a simple protocol to purify milligram quantities of ubH2A from mammalian cells, which were used to reconstitute nucleosome substrates with fully ubiquitinated H2A. A number of histone methyltransferases were then tested on these nucleosome substrates. Notably, all of the H3K36-specific methyltransferases, including ASH1L, HYPB, NSD1, and NSD2 were inhibited by ubH2A, whereas the other histone methyltransferases, including PRC2, G9a, and Pr-Set7 were not affected by ubH2A. Together with previous reports, these findings collectively explain the mutual repulsion of H3K36me2/3 and Polycomb modifications.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Ubiquitination/physiology , Animals , Carrier Proteins/metabolism , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Methylation , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Nat Commun ; 15(1): 7310, 2024 Aug 25.
Article in English | MEDLINE | ID: mdl-39181896

ABSTRACT

In mammals, global passive demethylation contributes to epigenetic reprogramming during early embryonic development. At this stage, the majority of DNA-methyltransferase 1 (DNMT1) protein is excluded from nucleus, which is considered the primary cause. However, whether the remaining nuclear activity of DNMT1 is regulated by additional mechanisms is unclear. Here, we report that nuclear DNMT1 abundance is finetuned through proteasomal degradation in mouse zygotes. We identify a maternal factor, Pramel15, which targets DNMT1 for degradation via Cullin-RING E3 ligases. Loss of Pramel15 elevates DNMT1 levels in the zygote pronuclei, impairs zygotic DNA demethylation, and causes a stochastic gain of DNA methylation in early embryos. Thus, Pramel15 can modulate the residual level of DNMT1 in the nucleus during zygotic DNA replication, thereby ensuring efficient DNA methylation reprogramming in early embryos.


Subject(s)
Cell Nucleus , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Demethylation , Zygote , Animals , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Zygote/metabolism , Mice , Cell Nucleus/metabolism , Female , DNA Methylation , Proteolysis , Embryonic Development/genetics , Male , Embryo, Mammalian/metabolism , Mice, Knockout , Gene Expression Regulation, Developmental , DNA Replication
16.
Sci Adv ; 10(13): eadk0858, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38552009

ABSTRACT

The long-lasting humoral immunity induced by viral infections or vaccinations depends on memory B cells with greatly increased affinity to viral antigens, which are evolved from germinal center (GC) responses. However, it is unclear whether antiviral memory B cells represent a distinct subset among the highly heterogeneous memory B cell population. Here, we examined memory B cells induced by a virus-mimicking antigen at both transcriptome and epigenetic levels and found unexpectedly that antiviral memory B cells exhibit an enhanced innate immune response, which appeared to be facilitated by the epigenetic memory that is established through the memory B cell development. In addition, T-bet is associated with the altered chromatin architecture and is required for the formation of the antiviral memory B cells. Thus, antiviral memory B cells are distinct from other GC-derived memory B cells in both physiological functions and epigenetic landmarks.


Subject(s)
B-Lymphocytes , Memory B Cells , Epigenetic Memory , Immunity, Innate , Antiviral Agents
17.
RNA ; 17(7): 1191-203, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21602304

ABSTRACT

PIWI-interacting RNAs (piRNAs) are a class of small RNAs abundantly expressed in animal gonads. piRNAs that map to retrotransposons are generated by a "ping-pong" amplification loop to suppress the activity of retrotransposons. However, the biogenesis and function of other categories of piRNAs have yet to be investigated. In this study, we first profiled the expression of small RNAs in type A spermatogonia, pachytene spermatocytes, and round spermatids by deep sequencing. We then focused on the computational analysis of the potential piRNAs generated in the present study as well as other published sets. piRNAs mapping to retrotransposons, mRNAs, and intergenic regions had different length distributions and were differentially regulated in spermatogenesis. piRNA-generating mRNAs (PRMRs), whose expression positively correlated with their piRNA products, constituted one-third of the protein-coding genes and were evolutionarily conserved and enriched with splicing isoforms and antisense transcripts. PRMRs with piRNAs preferentially mapped to CDSs and 3' UTRs partitioned into three clusters differentially expressed during spermatogenesis and enriched with unique sets of functional annotation terms related to housekeeping activities as well as spermatogenesis-specific processes. Intergenic piRNAs were divided into 2992 clusters probably representing novel transcriptional units that have not been reported. The transcripts of a large number of genes involved in spermatogenesis are the precursors of piRNAs, and these genes are intricately regulated by alternative splicing and antisense transcripts. piRNAs, whose regulatory role in gene expression awaits to be identified, are clearly products of a novel regulatory process that needs to be defined.


Subject(s)
RNA, Small Interfering/genetics , Spermatogenesis/genetics , Animals , Animals, Newborn , Base Sequence , Cells, Cultured , Cluster Analysis , Gene Expression Profiling , Male , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Microarray Analysis , RNA, Small Interfering/metabolism , Spermatids/metabolism , Spermatocytes/metabolism , Spermatogenesis/physiology , Spermatogonia/metabolism , Time Factors
18.
Zhongguo Zhen Jiu ; 43(10): 1134-8, 2023 Oct 12.
Article in Zh | MEDLINE | ID: mdl-37802519

ABSTRACT

OBJECTIVE: To compare the clinical efficacy of acupuncture, Chinese medication and combination of acupuncture and medication in the treatment of dry eye complicated with computer vision syndrome (CVS). METHODS: A total of 152 patients with dry eye complicated with CVS were randomly divided into an acupuncture-medication group (38 cases, 1 case was removed), an acupuncture group (38 cases, 1 case dropped off), a Chinese medication group (38 cases, 1 case was removed), and a western medication group (38 cases, 1 case dropped off). In the western medication group, sodium hyaluronate eye drop combined with esculin and digitalis glycosides eye drop were used. In the acupuncture group, acupuncture was applied at bilateral Taiyang (EX-HN 5), Cuanzhu (BL 2), Fengchi (GB 20), Qimen (LR 14) , and Hegu (LI 4) etc., once a day. In the Chinese medication group, Yiqi Congming decoction formula ganule was given orally, one dose a day. In the acupuncture-medication group, acupuncture combined with Yiqi Congming decoction formula granule were used. All groups were treated for 14 d. The non-invasive first tear film break-up time (NIBUT f), non-invasive average tear film break-up time (NIBUT av), tear meniscus height (TMH), ocular surface disease index (OSDI) score, and CVS symptom score were compared between the patients of each group before and after treatment. RESULTS: After treatment, the NIBUT f, NIBUT av, and TMH were increased compared with those before treatment in the patients of the 4 groups (P<0.01); the NIBUT f and NIBUT av in the acupuncture-medication group and the acupuncture group were higher than those in the Chinese medication group and the western medication group (P<0.05), and the TMH in the acupuncture-medication group and the Chinese medication group were higher than those in the acupuncture group and the western medication group (P<0.05). After treatment, the OSDI scores, the various scores and total scores of CVS (except for head symptom score in the western medication group) were decreased compared with those before treatment in the patients of the 4 groups (P<0.01). The OSDI score, total score, eye symptom score, and body symptom score of CVS in the acupuncture-medication group were lower than those in the acupuncture group, the Chinese medication group, and the western medication group (P<0.01, P<0.05), the head symptom score of the acupuncture-medication group was lower than that in the western medication group (P<0.05), and the CVS physical symptom scores and mental cognitive symptom scores of the acupuncture-medication group and the acupuncture group were lower than those in the Chinese medication group and the western medication group (P<0.05). CONCLUSION: Acupuncture has advantages in improving NIBUT f, NIBUT av, and CVS physical symptoms and cognitive symptoms, and the Chinese medication has advantage in improving TMH. The combination of acupuncture and Chinese medication has better effects compared with monotherapy.


Subject(s)
Acupuncture Therapy , Dry Eye Syndromes , Humans , Dry Eye Syndromes/drug therapy , Dry Eye Syndromes/etiology , Computers , Treatment Outcome , Acupuncture Points , Ophthalmic Solutions
19.
Sci China Life Sci ; 66(7): 1600-1614, 2023 07.
Article in English | MEDLINE | ID: mdl-36914923

ABSTRACT

Epigenetic information carried by histone modifications not only reflects the state of gene expression, but also participates in the maintenance of chromatin states and the regulation of gene expression. Recycling of parental histones to daughter chromatin after DNA replication is vital to mitotic inheritance of epigenetic information and the maintenance of cell identity, because the locus-specific modifications of the parental histones need to be maintained. To assess the precision of parental histone recycling, we developed a synthetic local label-chasing system in budding yeast Saccharomyces cerevisiae. Using this system, we observed that parental histone H3 can be recycled to their original position, thereby recovering their position information after DNA replication at all tested loci, including heterochromatin boundary, non-transcribed region, and actively transcribed regions. Moreover, the recycling rate appears to be affected by local chromatin environment. We surveyed a number of potential regulatory factors and observed that histone H3-H4 chaperon Asf1 contributed to parental histone recycling, while the eukaryotic replisome-associated components Mcm2 and Dpb3 displayed compounding effects in this process. In addition, the FACT complex also plays a role in the recycling of parental histones and helps to stabilize the nucleosomes.


Subject(s)
Histones , Saccharomyces cerevisiae Proteins , Humans , Histones/genetics , Histones/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , DNA Replication , Chromatin/genetics , Chromatin/metabolism , Nucleosomes/genetics , Saccharomyces cerevisiae Proteins/genetics
20.
Cell Death Discov ; 9(1): 291, 2023 Aug 09.
Article in English | MEDLINE | ID: mdl-37558683

ABSTRACT

Radiotherapy resistance is an important and urgent challenge in the clinical management of esophageal squamous carcinoma (ESCC). However, the factors mediating the ESCC resistance to radiotherapy and its underlying molecular mechanisms are not fully clarified. Our previous studies have demonstrated the critical role of DNA polymerase iota (POLI) in ESCC development and progression, here, we aimed to investigate the involvement of POLI in ESCC radiotherapy resistance and elucidate the underlying molecular mechanism. We found that highly expressed POLI was correlated with shorter overall survival of ESCC patients received radiotherapy. Down-regulation of POLI sensitized ESCC to IR, prolonged γH2AX foci in nuclei and comet tails after IR. HR but not NHEJ repair is inhibited in POLI-deficient ESCC cells. POLI stabilizes RAD51 protein via competitively binding with and blocking the interaction between RAD51 and E3 ligase XIAP and XIAP-mediated ubiquitination. Furthermore, loss of POLI leads to the activation of GAS signaling. Our findings provide novel insight into the role of POLI in the development of radioresistance mediated by stabilizing RAD51 protein in ESCC.

SELECTION OF CITATIONS
SEARCH DETAIL