Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.509
Filter
Add more filters

Publication year range
1.
Annu Rev Biochem ; 87: 51-73, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29589958

ABSTRACT

Ribosome biogenesis is a complex and highly energy-demanding process that requires the concerted action of all three nuclear RNA polymerases (Pol I-III) in eukaryotes. The three largest ribosomal RNAs (rRNAs) originate from a precursor transcript (pre-rRNA) that is encoded by multicopy genes located in the nucleolus. Transcription of these rRNA genes (rDNA) by Pol I is the key regulation step in ribosome production and is tightly controlled by an intricate network of signaling pathways and epigenetic mechanisms. In this article, we give an overview of the composition of the basal Pol I machinery and rDNA chromatin. We discuss rRNA gene regulation in response to environmental signals and developmental cues and focus on perturbations occurring in diseases linked to either excessive or limited rRNA levels. Finally, we discuss the emerging view that rDNA integrity and activity may be involved in the aging process.


Subject(s)
RNA Polymerase I/genetics , RNA Polymerase I/metabolism , Aging/genetics , Aging/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Chromatin/genetics , Chromatin/metabolism , DNA, Ribosomal/genetics , DNA, Ribosomal/metabolism , Epigenesis, Genetic , Humans , Models, Biological , Multigene Family , Neoplasms/genetics , Neoplasms/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , Ribosomes/genetics , Ribosomes/metabolism , Signal Transduction , Transcription, Genetic
2.
Cell ; 172(1-2): 106-120.e21, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29249356

ABSTRACT

Cell fate transitions involve rapid gene expression changes and global chromatin remodeling, yet the underlying regulatory pathways remain incompletely understood. Here, we identified the RNA-processing factor Nudt21 as a novel regulator of cell fate change using transcription-factor-induced reprogramming as a screening assay. Suppression of Nudt21 enhanced the generation of induced pluripotent stem cells, facilitated transdifferentiation into trophoblast stem cells, and impaired differentiation of myeloid precursors and embryonic stem cells, suggesting a broader role for Nudt21 in cell fate change. We show that Nudt21 directs differential polyadenylation of over 1,500 transcripts in cells acquiring pluripotency, although only a fraction changed protein levels. Remarkably, these proteins were strongly enriched for chromatin regulators, and their suppression neutralized the effect of Nudt21 during reprogramming. Collectively, our data uncover Nudt21 as a novel post-transcriptional regulator of cell fate and establish a direct, previously unappreciated link between alternative polyadenylation and chromatin signaling.


Subject(s)
Cellular Reprogramming , Chromatin Assembly and Disassembly , Cleavage And Polyadenylation Specificity Factor/metabolism , Polyadenylation , Signal Transduction , Animals , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , Cleavage And Polyadenylation Specificity Factor/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , HEK293 Cells , Humans , Mice
3.
Annu Rev Biochem ; 85: 227-64, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27145843

ABSTRACT

The Cas9 protein (CRISPR-associated protein 9), derived from type II CRISPR (clustered regularly interspaced short palindromic repeats) bacterial immune systems, is emerging as a powerful tool for engineering the genome in diverse organisms. As an RNA-guided DNA endonuclease, Cas9 can be easily programmed to target new sites by altering its guide RNA sequence, and its development as a tool has made sequence-specific gene editing several magnitudes easier. The nuclease-deactivated form of Cas9 further provides a versatile RNA-guided DNA-targeting platform for regulating and imaging the genome, as well as for rewriting the epigenetic status, all in a sequence-specific manner. With all of these advances, we have just begun to explore the possible applications of Cas9 in biomedical research and therapeutics. In this review, we describe the current models of Cas9 function and the structural and biochemical studies that support it. We focus on the applications of Cas9 for genome editing, regulation, and imaging, discuss other possible applications and some technical considerations, and highlight the many advantages that CRISPR/Cas9 technology offers.


Subject(s)
Bacterial Proteins/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , DNA Repair , Endonucleases/genetics , Gene Editing/methods , RNA, Guide, Kinetoplastida/genetics , Bacterial Proteins/metabolism , Base Pairing , CRISPR-Associated Protein 9 , DNA Cleavage , Endonucleases/metabolism , Epigenesis, Genetic , Gene Targeting , Genome, Human , Humans , Molecular Imaging , Protein Engineering , Protein Structure, Secondary , RNA, Guide, Kinetoplastida/metabolism
4.
Annu Rev Biochem ; 85: 35-64, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27294438

ABSTRACT

The health of an organism is orchestrated by a multitude of molecular and biochemical networks responsible for ensuring homeostasis within cells and tissues. However, upon aging, a progressive failure in the maintenance of this homeostatic balance occurs in response to a variety of endogenous and environmental stresses, allowing the accumulation of damage, the physiological decline of individual tissues, and susceptibility to diseases. What are the molecular and cellular signaling events that control the aging process and how can this knowledge help design therapeutic strategies to combat age-associated diseases? Here we provide a comprehensive overview of the evolutionarily conserved biological processes that alter the rate of aging and discuss their link to disease prevention and the extension of healthy life span.


Subject(s)
DNA Damage , Longevity/genetics , Proteostasis Deficiencies/genetics , Signal Transduction , Telomere Shortening , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Caloric Restriction , Epigenesis, Genetic , Homeostasis/genetics , Humans , Inflammation , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Mitochondria/metabolism , Oxidative Stress , Proteostasis Deficiencies/metabolism , Proteostasis Deficiencies/pathology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
5.
Immunity ; 55(3): 557-574.e7, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35263570

ABSTRACT

The clinical benefit of T cell immunotherapies remains limited by incomplete understanding of T cell differentiation and dysfunction. We generated an epigenetic and transcriptional atlas of T cell differentiation from healthy humans that included exhausted CD8 T cells and applied this resource in three ways. First, we identified modules of gene expression and chromatin accessibility, revealing molecular coordination of differentiation after activation and between central memory and effector memory. Second, we applied this healthy molecular framework to three settings-a neoadjuvant anti-PD1 melanoma trial, a basal cell carcinoma scATAC-seq dataset, and autoimmune disease-associated SNPs-yielding insights into disease-specific biology. Third, we predicted genome-wide cis-regulatory elements and validated this approach for key effector genes using CRISPR interference, providing functional annotation and demonstrating the ability to identify targets for non-coding cellular engineering. These studies define epigenetic and transcriptional regulation of human T cells and illustrate the utility of interrogating disease in the context of a healthy T cell atlas.


Subject(s)
Epigenomics , Lymphocyte Activation , CD8-Positive T-Lymphocytes , Cell Differentiation/genetics , Chromatin/genetics , Chromatin/metabolism , Epigenesis, Genetic , Humans , Lymphocyte Activation/genetics
6.
Mol Cell ; 83(23): 4370-4385.e9, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-38016475

ABSTRACT

Targeting epigenetic regulators to potentiate anti-PD-1 immunotherapy converges on the activation of type I interferon (IFN-I) response, mimicking cellular response to viral infection, but how its strength and duration are regulated to impact combination therapy efficacy remains largely unknown. Here, we show that mitochondrial CPT1A downregulation following viral infection restrains, while its induction by epigenetic perturbations sustains, a double-stranded RNA-activated IFN-I response. Mechanistically, CPT1A recruits the endoplasmic reticulum-localized ZDHHC4 to catalyze MAVS Cys79-palmitoylation, which promotes MAVS stabilization and activation by inhibiting K48- but facilitating K63-linked ubiquitination. Further elevation of CPT1A incrementally increases MAVS palmitoylation and amplifies the IFN-I response, which enhances control of viral infection and epigenetic perturbation-induced antitumor immunity. Moreover, CPT1A chemical inducers augment the therapeutic effect of combined epigenetic treatment with PD-1 blockade in refractory tumors. Our study identifies CPT1A as a stabilizer of MAVS activation, and its link to epigenetic perturbation can be exploited for cancer immunotherapy.


Subject(s)
Interferon Type I , Virus Diseases , Humans , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Lipoylation , Epigenesis, Genetic , Immunity, Innate
7.
Mol Cell ; 82(5): 1053-1065.e8, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35245449

ABSTRACT

Enzymatic methylation of cytosine to 5-methylcytosine in DNA is a fundamental epigenetic mechanism involved in mammalian development and disease. DNA methylation is brought about by collective action of three AdoMet-dependent DNA methyltransferases, whose catalytic interactions and temporal interplay are poorly understood. We used structure-guided engineering of the Dnmt1 methyltransferase to enable catalytic transfer of azide tags onto DNA from a synthetic cofactor analog, Ado-6-azide, in vitro. We then CRISPR-edited the Dnmt1 locus in mouse embryonic stem cells to install the engineered codon, which, following pulse internalization of the Ado-6-azide cofactor by electroporation, permitted selective azide tagging of Dnmt1-specific genomic targets in cellulo. The deposited covalent tags were exploited as "click" handles for reading adjoining sequences and precise genomic mapping of the methylation sites. The proposed approach, Dnmt-TOP-seq, enables high-resolution temporal tracking of the Dnmt1 catalysis in mammalian cells, paving the way to selective studies of other methylation pathways in eukaryotic systems.


Subject(s)
Azides , DNA (Cytosine-5-)-Methyltransferases , 5-Methylcytosine , Animals , Azides/metabolism , DNA/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , DNA Modification Methylases/genetics , Mammals/metabolism , Mice
8.
Mol Cell ; 78(3): 445-458.e6, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32197065

ABSTRACT

Paternal dietary conditions may contribute to metabolic disorders in offspring. We have analyzed the role of the stress-dependent epigenetic regulator cyclic AMP-dependent transcription factor 7 (ATF7) in paternal low-protein diet (pLPD)-induced gene expression changes in mouse liver. Atf7+/- mutations cause an offspring phenotype similar to that caused by pLPD, and the effect of pLPD almost vanished when paternal Atf7+/- mice were used. ATF7 binds to the promoter regions of ∼2,300 genes, including cholesterol biosynthesis-related and tRNA genes in testicular germ cells (TGCs). LPD induces ATF7 phosphorylation by p38 via reactive oxygen species (ROS) in TGCs. This leads to the release of ATF7 and a decrease in histone H3K9 dimethylation (H3K9me2) on its target genes. These epigenetic changes are maintained and induce expression of some tRNA fragments in spermatozoa. These results indicate that LPD-induced and ATF7-dependent epigenetic changes in TGCs play an important role in paternal diet-induced metabolic reprograming in offspring.


Subject(s)
Activating Transcription Factors/genetics , Diet, Protein-Restricted , Epigenesis, Genetic , Liver/physiology , Spermatozoa/physiology , Activating Transcription Factors/metabolism , Animals , Female , Gene Expression Regulation , Histones/metabolism , Lysine/metabolism , Male , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Phosphorylation , Promoter Regions, Genetic
9.
Trends Biochem Sci ; 48(9): 788-800, 2023 09.
Article in English | MEDLINE | ID: mdl-37393166

ABSTRACT

Temperature is one of the main environmental cues affecting plant growth and development, and plants have evolved multiple mechanisms to sense and acclimate to high temperature. Emerging research has shown that transcription factors, epigenetic factors, and their coordination are essential for plant temperature responses and the resulting phenological adaptation. Here, we summarize recent advances in molecular and cellular mechanisms to understand how plants acclimate to high temperature and describe how plant meristems sense and integrate environmental signals. Furthermore, we lay out future directions for new technologies to reveal heterogeneous responses in different cell types thus improving plant environmental plasticity.


Subject(s)
Plant Development , Transcription Factors , Temperature , Transcription Factors/genetics , Plants/genetics , Epigenesis, Genetic
10.
Semin Immunol ; 70: 101841, 2023 11.
Article in English | MEDLINE | ID: mdl-37703611

ABSTRACT

Cells undergo an inflammatory programmed lytic cell death called 'pyroptosis' (with the Greek roots 'fiery'), often featuring morphological hallmarks such as large ballooning protrusions and subsequent bursting. Originally described as a caspase-1-dependent cell death in response to bacterial infection, pyroptosis has since been re-defined in 2018 as a cell death dependent on plasma membrane pores by a gasdermin (GSDM) family member [1,2]. GSDMs form pores in the plasma membrane as well as organelle membranes, thereby initiating membrane destruction and the rapid and lytic demise of a cell. The gasdermin family plays a profound role in the execution of pyroptosis in the context of infection, inflammation, tumor pathogenesis, and anti-tumor therapy. More recently, cell-death-independent functions for some of the GSDMs have been proposed. Therefore, a comprehensive understanding of gasdermin gene regulation, including mechanisms in both homeostatic conditions and during inflammation, is essential. In this review, we will summarize the role of gasdermins in pyroptosis and focus our discussion on the transcriptional and epigenetic mechanisms controlling the expression of GSDMs.


Subject(s)
Gasdermins , Neoplasm Proteins , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Apoptosis , Inflammation/metabolism , Epigenesis, Genetic , Inflammasomes/metabolism
11.
Trends Biochem Sci ; 47(8): 645-659, 2022 08.
Article in English | MEDLINE | ID: mdl-35397926

ABSTRACT

Age-associated changes in mitochondria are closely involved in aging. Apart from the established roles in bioenergetics and biosynthesis, mitochondria are signaling organelles that communicate their fitness to the nucleus, triggering transcriptional programs to adapt homeostasis stress that is essential for organismal health and aging. Emerging studies revealed that mitochondrial-to-nuclear (mito-nuclear) communication via altered levels of mitochondrial metabolites or stress signals causes various epigenetic changes, facilitating efforts to maintain homeostasis and affect aging. Here, we summarize recent studies on the mechanisms by which mito-nuclear communication modulates epigenomes and their effects on regulating the aging process. Insights into understanding how mitochondrial metabolites serve as prolongevity signals and how aging affects this communication will help us develop interventions to promote longevity and health.


Subject(s)
Longevity , Mitochondria , Cell Nucleus/metabolism , Epigenesis, Genetic , Longevity/physiology , Mitochondria/metabolism
12.
Development ; 150(11)2023 06 01.
Article in English | MEDLINE | ID: mdl-37260148

ABSTRACT

Evolutionarily conserved Notch signaling is highly sensitive to changes in Notch receptor dose caused by intrinsic and environmental fluctuations. It is well known that epigenetic regulation responds dynamically to genetic, cellular and environmental stresses. However, it is unclear whether the Notch receptor dose is directly regulated at the epigenetic level. Here, by studying the role of the upstream epigenetic regulator Stuxnet (Stx) in Drosophila developmental signaling, we find that Stx promotes Notch receptor mRNA expression by counteracting the activity of Polycomb repressive complex 1 (PRC1). In addition, we provide evidence that Notch is a direct PRC1 target by identifying and validating in vivo the only bona fide Polycomb response element (PRE) among the seven Polycomb group (PcG)-binding sites revealed by DamID-seq and ChIP-seq analysis. Importantly, in situ deletion of this PRE results in increased Notch expression and phenotypes resembling Notch hyperactivation in cell fate specification. These results not only underscore the importance of epigenetic regulation in fine-tuning the Notch activity dose, but also the need to assess the physiological significance of omics-based PcG binding in development.


Subject(s)
Drosophila Proteins , Epigenesis, Genetic , Animals , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Response Elements/genetics , Receptors, Notch/genetics , Receptors, Notch/metabolism
13.
Brief Bioinform ; 25(2)2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38329268

ABSTRACT

Nucleosomes represent hubs in chromatin organization and gene regulation and interact with a plethora of chromatin factors through different modes. In addition, alterations in histone proteins such as cancer mutations and post-translational modifications have profound effects on histone/nucleosome interactions. To elucidate the principles of histone interactions and the effects of those alterations, we developed histone interactomes for comprehensive mapping of histone-histone interactions (HHIs), histone-DNA interactions (HDIs), histone-partner interactions (HPIs) and DNA-partner interactions (DPIs) of 37 organisms, which contains a total of 3808 HPIs from 2544 binding proteins and 339 HHIs, 100 HDIs and 142 DPIs across 110 histone variants. With the developed networks, we explored histone interactions at different levels of granularities (protein-, domain- and residue-level) and performed systematic analysis on histone interactions at a large scale. Our analyses have characterized the preferred binding hotspots on both nucleosomal/linker DNA and histone octamer and unraveled diverse binding modes between nucleosome and different classes of binding partners. Last, to understand the impact of histone cancer-associated mutations on histone/nucleosome interactions, we complied one comprehensive cancer mutation dataset including 7940 cancer-associated histone mutations and further mapped those mutations onto 419,125 histone interactions at the residue level. Our quantitative analyses point to histone cancer-associated mutations' strongly disruptive effects on HHIs, HDIs and HPIs. We have further predicted 57 recurrent histone cancer mutations that have large effects on histone/nucleosome interactions and may have driver status in oncogenesis.


Subject(s)
Neoplasms , Nucleosomes , Humans , Nucleosomes/genetics , Histones/genetics , Histones/metabolism , DNA/chemistry , Mutation , Neoplasms/genetics
14.
Mol Cell ; 69(3): 398-411.e6, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29395062

ABSTRACT

The inflammatory response mediated by nuclear factor κB (NF-κB) signaling is essential for host defense against pathogens. Although the regulatory mechanism of NF-κB signaling has been well studied, the molecular basis for epigenetic regulation of the inflammatory response is poorly understood. Here we identify a new signaling axis of PKCα-LSD1-NF-κB, which is critical for activation and amplification of the inflammatory response. In response to excessive inflammatory stimuli, PKCα translocates to the nucleus and phosphorylates LSD1. LSD1 phosphorylation is required for p65 binding and facilitates p65 demethylation, leading to enhanced stability. In vivo genetic analysis using Lsd1SA/SA mice with ablation of LSD1 phosphorylation and chemical approaches in wild-type mice with inhibition of PKCα or LSD1 activity show attenuated sepsis-induced inflammatory lung injury and mortality. Together, we demonstrate that the PKCα-LSD1-NF-κB signaling cascade is crucial for epigenetic control of the inflammatory response, and targeting this signaling could be a powerful therapeutic strategy for systemic inflammatory diseases, including sepsis.


Subject(s)
Histone Demethylases/metabolism , Protein Kinase C/metabolism , Animals , Cell Nucleus/metabolism , Epigenesis, Genetic/genetics , Histone Demethylases/genetics , Inflammation/metabolism , Methylation , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphorylation , Protein Kinase C/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/metabolism
15.
Proc Natl Acad Sci U S A ; 120(49): e2315096120, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38011564

ABSTRACT

Hidradenitis suppurativa (HS) is a complex inflammatory skin disease with undefined mechanistic underpinnings. Here, we investigated HS epithelial cells and demonstrated that HS basal progenitors modulate their lineage restriction and give rise to pathogenic keratinocyte clones, resulting in epidermal hyperproliferation and dysregulated inflammation in HS. When comparing to healthy epithelial stem/progenitor cells, in HS, we identified changes in gene signatures that revolve around the mitotic cell cycle, DNA damage response and repair, as well as cell-cell adhesion and chromatin remodeling. By reconstructing cell differentiation trajectory and CellChat modeling, we identified a keratinocyte population specific to HS. This population is marked by S100A7/8/9 and KRT6 family members, triggering IL1, IL10, and complement inflammatory cascades. These signals, along with HS-specific proinflammatory cytokines and chemokines, contribute to the recruitment of certain immune cells during the disease progression. Furthermore, we revealed a previously uncharacterized role of S100A8 in regulating the local chromatin environment of target loci in HS keratinocytes. Through the integration of genomic and epigenomic datasets, we identified genome-wide chromatin rewiring alongside the switch of transcription factors (TFs), which mediated HS transcriptional profiles. Importantly, we identified numerous clinically relevant inflammatory enhancers and their coordinated TFs in HS basal CD49fhigh cells. The disruption of the S100A enhancer using the CRISPR/Cas9-mediated approach or the pharmacological inhibition of the interferon regulatory transcription factor 3 (IRF3) efficiently reduced the production of HS-associated inflammatory regulators. Our study not only uncovers the plasticity of epidermal progenitor cells in HS but also elucidates the epigenetic mechanisms underlying HS pathogenesis.


Subject(s)
Hidradenitis Suppurativa , Humans , Hidradenitis Suppurativa/genetics , Skin/metabolism , Epigenomics , Epigenesis, Genetic , Stem Cells/metabolism , Chromatin/metabolism
16.
Immunol Rev ; 305(1): 90-110, 2022 01.
Article in English | MEDLINE | ID: mdl-34908173

ABSTRACT

Immunological memory is the underlying mechanism by which the immune system remembers previous encounters with pathogens to produce an enhanced secondary response upon re-encounter. It stands as the hallmark feature of the adaptive immune system and the cornerstone of vaccine development. Classic recall responses are executed by conventional T and B cells, which undergo somatic recombination and modify their receptor repertoire to ensure recognition of a vast number of antigens. However, recent evidence has challenged the dogma that memory responses are restricted to the adaptive immune system, which has prompted a reevaluation of what delineates "immune memory." Natural killer (NK) cells of the innate immune system have been at the forefront of these pushed boundaries, and have proved to be more "adaptable" than previously thought. Like T cells, we now appreciate that their "natural" abilities actually require a myriad of signals for optimal responses. In this review, we discuss the many signals required for effector and memory NK cell responses and the epigenetic mechanisms that ultimately endow their enhanced features.


Subject(s)
Epigenesis, Genetic , Killer Cells, Natural , B-Lymphocytes , Humans , Immunity, Innate , Immunologic Memory , T-Lymphocytes
17.
Crit Rev Biochem Mol Biol ; 58(1): 1-18, 2023 02.
Article in English | MEDLINE | ID: mdl-36921088

ABSTRACT

In the human cell nucleus, dynamically organized chromatin is the substrate for gene regulation, DNA replication, and repair. A central mechanism of DNA loop formation is an ATPase motor cohesin-mediated loop extrusion. The cohesin complexes load and unload onto the chromosome under the control of other regulators that physically interact and affect motor activity. Regulation of the dynamic loading cycle of cohesin influences not only the chromatin structure but also genome-associated human disorders and aging. This review focuses on the recently spotlighted genome organizing factors and the mechanism by which their dynamic interactions shape the genome architecture in interphase.


Subject(s)
Chromatin , DNA Replication , Humans , Chromatin/genetics , Interphase/genetics , Gene Expression Regulation , Cell Nucleus
18.
J Biol Chem ; : 107788, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39303914

ABSTRACT

The incidence of germinal center B-cell-like type diffuse large B-cell lymphoma (GCB DLBCL) is steadily increasing, with a known hereditary component. Although some molecular mechanisms in GCB DLBCL have been elucidated, understanding remains incomplete, limiting the effectiveness of targeted therapies. In GCB DLBCL patients, abnormally high expression of zeste homologs 2 (EZH2) is noted, and the compensatory effect of EZH1 following EZH2 inhibition contributes to poor prognosis. This highlights the potential of dual targeting of EZH1/2 as a promising strategy. In this study, we developed a novel inhibitor, EZH-1-P2, targeting EZH1/2, and evaluated its anti-tumor effects on DLBCL cells. Mechanistically, inhibition of EZH1/2 affects the epigenetic regulation of gene expression related to p53, impacting cell cycle progression and GCB DLBCL cell growth. Additionally, while EZH1/2 inhibition impacts NOTCH signaling, the precise mechanism by which it affects M2-type tumor-associated macrophage (M2-TAM) polarization and germinal center expansion requires further investigation. Our research introduces EZH-1-P2 as a novel inhibitor with potential as a candidate for GCB DLBCL therapy, although further studies are needed to fully elucidate its mechanisms.

19.
Mol Biol Evol ; 41(2)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38266195

ABSTRACT

The cross-species characterization of evolutionary changes in the functional genome can facilitate the translation of genetic findings across species and the interpretation of the evolutionary basis underlying complex phenotypes. Yet, this has not been fully explored between cattle, sheep, goats, and other mammals. Here, we systematically characterized the evolutionary dynamics of DNA methylation and gene expression in 3 somatic tissues (i.e. brain, liver, and skeletal muscle) and sperm across 7 mammalian species, including 3 ruminant livestock species (cattle, sheep, and goats), humans, pigs, mice, and dogs, by generating and integrating 160 DNA methylation and transcriptomic data sets. We demonstrate dynamic changes of DNA hypomethylated regions and hypermethylated regions in tissue-type manner across cattle, sheep, and goats. Specifically, based on the phylo-epigenetic model of DNA methylome, we identified a total of 25,074 hypomethylated region extension events specific to cattle, which participated in rewiring tissue-specific regulatory network. Furthermore, by integrating genome-wide association studies of 50 cattle traits, we provided novel insights into the genetic and evolutionary basis of complex phenotypes in cattle. Overall, our study provides a valuable resource for exploring the evolutionary dynamics of the functional genome and highlights the importance of cross-species characterization of multiomics data sets for the evolutionary interpretation of complex phenotypes in cattle livestock.


Subject(s)
Cattle , DNA Methylation , Goats , Sheep , Animals , Cattle/genetics , Dogs , Humans , Male , Mice , Genome-Wide Association Study , Goats/genetics , Multifactorial Inheritance , Sheep/genetics , Swine
20.
EMBO J ; 40(10): e106632, 2021 05 17.
Article in English | MEDLINE | ID: mdl-33739466

ABSTRACT

HIV-1 latency is a major obstacle to achieving a functional cure for AIDS. Reactivation of HIV-1-infected cells followed by their elimination via immune surveillance is one proposed strategy for eradicating the viral reservoir. However, current latency-reversing agents (LRAs) show high toxicity and low efficiency, and new targets are needed to develop more promising LRAs. Here, we found that the histone chaperone CAF-1 (chromatin assembly factor 1) is enriched on the HIV-1 long terminal repeat (LTR) and forms nuclear bodies with liquid-liquid phase separation (LLPS) properties. CAF-1 recruits epigenetic modifiers and histone chaperones to the nuclear bodies to establish and maintain HIV-1 latency in different latency models and primary CD4+ T cells. Three disordered regions of the CHAF1A subunit are important for phase-separated CAF-1 nuclear body formation and play a key role in maintaining HIV-1 latency. Disruption of phase-separated CAF-1 bodies could be a potential strategy to reactivate latent HIV-1.


Subject(s)
HIV-1/metabolism , CD4-Positive T-Lymphocytes/metabolism , Chromatin Assembly Factor-1/genetics , Chromatin Assembly Factor-1/metabolism , Epigenesis, Genetic/genetics , Epigenesis, Genetic/physiology , HEK293 Cells , Humans , Promoter Regions, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL