Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Genes Dev ; 38(1-2): 46-69, 2024 02 13.
Article in English | MEDLINE | ID: mdl-38286657

ABSTRACT

Approximately 20% of head and neck squamous cell carcinomas (HNSCCs) exhibit reduced methylation on lysine 36 of histone H3 (H3K36me) due to mutations in histone methylase NSD1 or a lysine-to-methionine mutation in histone H3 (H3K36M). Whether such alterations of H3K36me can be exploited for therapeutic interventions is still unknown. Here, we show that HNSCC models expressing H3K36M can be divided into two groups: those that display aberrant accumulation of H3K27me3 and those that maintain steady levels of H3K27me3. The former group exhibits reduced proliferation, genome instability, and heightened sensitivity to genotoxic agents like PARP1/2 inhibitors. Conversely, H3K36M HNSCC models with constant H3K27me3 levels lack these characteristics unless H3K27me3 is elevated by DNA hypomethylating agents or inhibiting H3K27me3 demethylases KDM6A/B. Mechanistically, H3K36M reduces H3K36me by directly impeding the activities of the histone methyltransferase NSD3 and the histone demethylase LSD2. Notably, aberrant H3K27me3 levels induced by H3K36M expression are not a bona fide epigenetic mark because they require continuous expression of H3K36M to be inherited. Moreover, increased sensitivity to PARP1/2 inhibitors in H3K36M HNSCC models depends solely on elevated H3K27me3 levels and diminishing BRCA1- and FANCD2-dependent DNA repair. Finally, a PARP1/2 inhibitor alone reduces tumor burden in a H3K36M HNSCC xenograft model with elevated H3K27me3, whereas in a model with consistent H3K27me3, a combination of PARP1/2 inhibitors and agents that up-regulate H3K27me3 proves to be successful. These findings underscore the crucial balance between H3K36 and H3K27 methylation in maintaining genome instability, offering new therapeutic options for patients with H3K36me-deficient tumors.


Subject(s)
Head and Neck Neoplasms , Histones , Humans , Histones/metabolism , Lysine/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Methylation , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Genomic Instability/genetics
2.
Biochem Soc Trans ; 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39248209

ABSTRACT

Histones are essential for maintaining chromatin structure and function. Histone mutations lead to changes in chromatin compaction, gene expression, and the recruitment of DNA repair proteins to the DNA lesion. These disruptions can impair critical DNA repair pathways, such as homologous recombination and non-homologous end joining, resulting in increased genomic instability, which promotes an environment favorable to tumor development and progression. Understanding these mechanisms underscores the potential of targeting DNA repair pathways in cancers harboring mutated histones, offering novel therapeutic strategies to exploit their inherent genomic instability for better treatment outcomes. Here, we examine how mutations in histone H3 disrupt normal chromatin function and DNA damage repair processes and how these mechanisms can be exploited for therapeutic interventions.

3.
Trends Biochem Sci ; 44(8): 688-700, 2019 08.
Article in English | MEDLINE | ID: mdl-31085088

ABSTRACT

Polycomb-group (PcG) complexes are multiprotein, evolutionarily conserved epigenetic machineries that regulate stem cell fate decisions and development, and are also implicated in cancer and other maladies. The PcG machinery can be divided into two major complexes: Polycomb repressive complex 1 and 2 (PRC1 and PRC2). Traditionally, PcG complexes have been associated with maintenance of gene repression mainly via histone-modifying activities. However, during the last years, increasing evidence indicates that the PcG complexes can also positively regulate gene transcription and modify non-histone substrates in multiple biological processes, cellular stages, and cancers. In this review, we will illustrate recent findings in PcG-mediated gene regulation, with special focus on the recently described non-classical functions of PcG complexes in stem cells and cancer.


Subject(s)
Polycomb-Group Proteins/chemistry , Polycomb-Group Proteins/metabolism , Amino Acid Sequence , Binding Sites , Gene Expression Regulation , Histones/chemistry , Humans , Neoplasms/genetics , Neoplasms/metabolism , Polycomb-Group Proteins/genetics , Protein Conformation , Protein Processing, Post-Translational , Stem Cells/drug effects , Stem Cells/metabolism
4.
Nature ; 544(7648): 59-64, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28289288

ABSTRACT

The folding of genomic DNA from the beads-on-a-string-like structure of nucleosomes into higher-order assemblies is crucially linked to nuclear processes. Here we calculate 3D structures of entire mammalian genomes using data from a new chromosome conformation capture procedure that allows us to first image and then process single cells. The technique enables genome folding to be examined at a scale of less than 100 kb, and chromosome structures to be validated. The structures of individual topological-associated domains and loops vary substantially from cell to cell. By contrast, A and B compartments, lamina-associated domains and active enhancers and promoters are organized in a consistent way on a genome-wide basis in every cell, suggesting that they could drive chromosome and genome folding. By studying genes regulated by pluripotency factor and nucleosome remodelling deacetylase (NuRD), we illustrate how the determination of single-cell genome structure provides a new approach for investigating biological processes.


Subject(s)
Chromatin Assembly and Disassembly , Genome , Molecular Imaging/methods , Nucleosomes/chemistry , Single-Cell Analysis/methods , Animals , CCCTC-Binding Factor , Cell Cycle Proteins/metabolism , Chromatin Assembly and Disassembly/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosomes, Mammalian/chemistry , Chromosomes, Mammalian/genetics , Chromosomes, Mammalian/metabolism , DNA/chemistry , DNA/genetics , DNA/metabolism , Enhancer Elements, Genetic , G1 Phase , Gene Expression Regulation , Gene Regulatory Networks , Genome/genetics , Haploidy , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Mice , Models, Molecular , Molecular Conformation , Molecular Imaging/standards , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , Promoter Regions, Genetic , Repressor Proteins/metabolism , Reproducibility of Results , Single-Cell Analysis/standards , Cohesins
5.
Nucleic Acids Res ; 49(17): 9768-9782, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34428304

ABSTRACT

Polycomb complexes have traditionally been prescribed roles as transcriptional repressors, though increasing evidence demonstrate they can also activate gene expression. However, the mechanisms underlying positive gene regulation mediated by Polycomb proteins are poorly understood. Here, we show that RING1B, a core component of Polycomb Repressive Complex 1, regulates enhancer-promoter interaction of the bona fide estrogen-activated GREB1 gene. Systematic characterization of RNA:DNA hybrid formation (R-loops), nascent transcription and RNA Pol II activity upon estrogen administration revealed a key role of RING1B in gene activation by regulating R-loop formation and RNA Pol II elongation. We also found that the estrogen receptor alpha (ERα) and RNA are both necessary for full RING1B recruitment to estrogen-activated genes. Notably, RING1B recruitment was mostly unaffected upon RNA Pol II depletion. Our findings delineate the functional interplay between RING1B, RNA and ERα to safeguard chromatin architecture perturbations required for estrogen-mediated gene regulation and highlight the crosstalk between steroid hormones and Polycomb proteins to regulate oncogenic programs.


Subject(s)
Enhancer Elements, Genetic , Estradiol/physiology , Polycomb Repressive Complex 1/metabolism , Promoter Regions, Genetic , R-Loop Structures , Transcriptional Activation , Cell Line , Chromatin/metabolism , Estrogen Receptor alpha/metabolism , Humans , RNA/metabolism
6.
Proc Natl Acad Sci U S A ; 116(14): 7005-7014, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30877256

ABSTRACT

p27 shifts from CDK inhibitor to oncogene when phosphorylated by PI3K effector kinases. Here, we show that p27 is a cJun coregulator, whose assembly and chromatin association is governed by p27 phosphorylation. In breast and bladder cancer cells with high p27pT157pT198 or expressing a CDK-binding defective p27pT157pT198 phosphomimetic (p27CK-DD), cJun is activated and interacts with p27, and p27/cJun complexes localize to the nucleus. p27/cJun up-regulates TGFB2 to drive metastasis in vivo. Global analysis of p27 and cJun chromatin binding and gene expression shows that cJun recruitment to many target genes is p27 dependent, increased by p27 phosphorylation, and activates programs of epithelial-mesenchymal transformation and metastasis. Finally, human breast cancers with high p27pT157 differentially express p27/cJun-regulated genes of prognostic relevance, supporting the biological significance of the work.


Subject(s)
Cell Movement , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Cell Adhesion , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p27/genetics , Humans , Neoplasms/genetics , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-jun/genetics
7.
Genes Dev ; 28(2): 182-97, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24449271

ABSTRACT

The molecular mechanisms underlying specification from embryonic stem cells (ESCs) and maintenance of neural progenitor cells (NPCs) are largely unknown. Recently, we reported that the Zuotin-related factor 1 (Zrf1) is necessary for chromatin displacement of the Polycomb-repressive complex 1 (PRC1). We found that Zrf1 is required for NPC specification from ESCs and that it promotes the expression of NPC markers, including the key regulator Pax6. Moreover, Zrf1 is essential to establish and maintain Wnt ligand expression levels, which are necessary for NPC self-renewal. Reactivation of proper Wnt signaling in Zrf1-depleted NPCs restores Pax6 expression and the self-renewal capacity. ESC-derived NPCs in vitro resemble most of the characteristics of the self-renewing NPCs located in the developing embryonic cortex, which are termed radial glial cells (RGCs). Depletion of Zrf1 in vivo impairs the expression of key self-renewal regulators and Wnt ligand genes in RGCs. Thus, we demonstrate that Zrf1 plays an essential role in NPC generation and maintenance.


Subject(s)
Cell Differentiation , DNA-Binding Proteins/metabolism , Neural Plate/cytology , Neural Plate/metabolism , Oncogene Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Line , DNA-Binding Proteins/genetics , Embryonic Stem Cells/cytology , Eye Proteins/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Humans , Ligands , Mice , Molecular Chaperones , Neurogenesis/genetics , Oncogene Proteins/genetics , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , RNA-Binding Proteins , Repressor Proteins/genetics , Signal Transduction , Wnt Proteins/metabolism
8.
PLoS Pathog ; 15(12): e1008221, 2019 12.
Article in English | MEDLINE | ID: mdl-31881074

ABSTRACT

Kaposi's sarcoma (KS) is an AIDS-defining cancer caused by the KS-associated herpesvirus (KSHV). Unanswered questions regarding KS are its cellular ontology and the conditions conducive to viral oncogenesis. We identify PDGFRA(+)/SCA-1(+) bone marrow-derived mesenchymal stem cells (Pα(+)S MSCs) as KS spindle-cell progenitors and found that pro-angiogenic environmental conditions typical of KS are critical for KSHV sarcomagenesis. This is because growth in KS-like conditions generates a de-repressed KSHV epigenome allowing oncogenic KSHV gene expression in infected Pα(+)S MSCs. Furthermore, these growth conditions allow KSHV-infected Pα(+)S MSCs to overcome KSHV-driven oncogene-induced senescence and cell cycle arrest via a PDGFRA-signaling mechanism; thus identifying PDGFRA not only as a phenotypic determinant for KS-progenitors but also as a critical enabler for viral oncogenesis.


Subject(s)
Mesenchymal Stem Cells/virology , Neovascularization, Pathologic/virology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Sarcoma, Kaposi/virology , Animals , Carcinogenesis/metabolism , Gene Expression/physiology , Herpesvirus 8, Human/genetics , Mesenchymal Stem Cells/cytology , Mice , Signal Transduction/physiology
9.
Mol Cell ; 46(3): 369-76, 2012 05 11.
Article in English | MEDLINE | ID: mdl-22483618

ABSTRACT

Methylation of lysine 4 (K4) within histone H3 has been linked to active transcription and is removed by LSD1 and the JmjC domain-containing proteins by amino-oxidation or hydroxylation, respectively. Here, we describe the deamination catalyzed by Lysyl oxidase-like 2 protein (LOXL2) as an unconventional chemical mechanism for H3K4 modification. Infrared spectroscopy and mass spectrometry analyses demonstrated that recombinant LOXL2 specifically deaminates trimethylated H3K4. Moreover, LOXL2 activity is linked with the transcriptional control of CDH1 gene by regulating H3K4me3 deamination. These results reveal another H3 modification and provide a different mechanism for H3K4 modification.


Subject(s)
Amino Acid Oxidoreductases/physiology , Histones/metabolism , Antigens, CD , Cadherins/genetics , Cell Line, Tumor , Deamination , Gene Expression Regulation , Humans , Lysine/metabolism , Methylation
11.
Appl Microbiol Biotechnol ; 100(23): 10137-10146, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27687996

ABSTRACT

Methanogenic archaea enrichment of a granular sludge was undertaken in an upflow anaerobic sludge blanket (UASB) reactor fed with methanol in order to enrich methylotrophic and hydrogenotrophic methanogenic populations. A microbial community assessment, in terms of microbial composition and activity-throughout the different stages of the feeding process with methanol and acetate-was performed using specific methanogenic activity (SMA) assays, quantitative real-time polymerase chain reaction (qPCR), and high-throughput sequencing of 16S ribosomal RNA (rRNA) genes from DNA and complementary DNA (cDNA). Distinct methanogenic enrichment was revealed by qPCR of mcrA gene in the methanol-fed community, being two orders of magnitude higher with respect to the initial inoculum, achieving a final mcrA/16S rRNA ratio of 0.25. High-throughput sequencing analysis revealed that the resulting methanogenic population was mainly composed by methylotrophic archaea (Methanomethylovorans and Methanolobus genus), being also highly active according to the RNA-based assessment. SMA confirmed that the methylotrophic pathway, with a direct conversion of methanol to CH4, was the main step of methanol degradation in the UASB. The biomass from the UASB, enriched in methanogenic archaea, may bear great potential as additional inoculum for bioreactors to carry out biogas production and other related processes.


Subject(s)
Archaea/classification , Archaea/metabolism , Bioreactors/microbiology , Methane/metabolism , Methanol/metabolism , Sewage/microbiology , Acetates/metabolism , Anaerobiosis , Archaea/genetics , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , High-Throughput Nucleotide Sequencing , Hydrogen/metabolism , Oxidoreductases/genetics , RNA, Ribosomal, 16S/genetics , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA
12.
EMBO J ; 30(22): 4586-600, 2011 Nov 16.
Article in English | MEDLINE | ID: mdl-22020125

ABSTRACT

H3K4 methylation is associated with active transcription and in combination with H3K27me3 thought to keep genes regulating development in a poised state. The contribution of enzymes regulating trimethylation of lysine 4 at histone 3 (H3K4me3) levels to embryonic stem cell (ESC) self-renewal and differentiation is just starting to emerge. Here, we show that the H3K4me2/3 histone demethylase Jarid1b (Kdm5b/Plu1) is dispensable for ESC self-renewal, but essential for ESC differentiation along the neural lineage. By genome-wide location analysis, we demonstrate that Jarid1b localizes predominantly to transcription start sites of genes encoding developmental regulators, of which more than half are also bound by Polycomb group proteins. Virtually all Jarid1b target genes are associated with H3K4me3 and depletion of Jarid1b in ESCs leads to a global increase of H3K4me3 levels. During neural differentiation, Jarid1b-depleted ESCs fail to efficiently silence lineage-inappropriate genes, specifically stem and germ cell genes. Our results delineate an essential role for Jarid1b-mediated transcriptional control during ESC differentiation.


Subject(s)
Embryonic Stem Cells/physiology , Histones/metabolism , Neurogenesis , Neurons/physiology , Transcription, Genetic , Animals , Antibodies, Monoclonal , Cell Line , Central Nervous System/embryology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Gene Expression Profiling , Gene Knockout Techniques/methods , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/immunology , Jumonji Domain-Containing Histone Demethylases/metabolism , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Polycomb-Group Proteins , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering , Repressor Proteins/metabolism
13.
EMBO J ; 30(17): 3635-46, 2011 Aug 02.
Article in English | MEDLINE | ID: mdl-21811233

ABSTRACT

Jarid2 is required for the genomic recruitment of the polycomb repressive complex-2 (PRC2) in embryonic stem cells. However, its specific role during late development and adult tissues remains largely uncharacterized. Here, we show that deletion of Jarid2 in mouse epidermis reduces the proliferation and potentiates the differentiation of postnatal epidermal progenitors, without affecting epidermal development. In neonatal epidermis, Jarid2 deficiency reduces H3K27 trimethylation, a chromatin repressive mark, in epidermal differentiation genes previously shown to be targets of the PRC2. However, in adult epidermis Jarid2 depletion does not affect interfollicular epidermal differentiation but results in delayed hair follicle (HF) cycling as a consequence of decreased proliferation of HF stem cells and their progeny. We conclude that Jarid2 is required for the scheduled proliferation of epidermal stem and progenitor cells necessary to maintain epidermal homeostasis.


Subject(s)
Cell Differentiation , Epidermal Cells , Keratinocytes/cytology , Nerve Tissue Proteins/metabolism , Stem Cells/cytology , Animals , Cells, Cultured , Epidermis/metabolism , Hair Follicle/metabolism , Histones/metabolism , Humans , Keratinocytes/metabolism , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Polycomb Repressive Complex 2 , Stem Cells/metabolism
14.
Cancer Cell ; 11(6): 513-25, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17560333

ABSTRACT

Epigenetic changes are common alterations in cancer cells. Here, we have investigated the role of Polycomb group proteins in the establishment and maintenance of the aberrant silencing of tumor suppressor genes during transformation induced by the leukemia-associated PML-RARalpha fusion protein. We show that in leukemic cells knockdown of SUZ12, a key component of Polycomb repressive complex 2 (PRC2), reverts not only histone modification but also induces DNA demethylation of PML-RARalpha target genes. This results in promoter reactivation and granulocytic differentiation. Importantly, the epigenetic alterations caused by PML-RARalpha can be reverted by retinoic acid treatment of primary blasts from leukemic patients. Our results demonstrate that the direct targeting of Polycomb group proteins by an oncogene plays a key role during carcinogenesis.


Subject(s)
Carrier Proteins/physiology , Leukemia, Promyelocytic, Acute/metabolism , Nuclear Proteins/physiology , Oncogene Proteins, Fusion/physiology , Repressor Proteins/metabolism , Cell Differentiation , DNA Methylation , Epigenesis, Genetic , Gene Silencing , Granulocytes/physiology , Histones , Humans , Neoplasm Proteins , Oncogene Proteins, Fusion/genetics , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Transcription Factors , Tretinoin/pharmacology , Tumor Cells, Cultured
15.
Trends Biochem Sci ; 35(6): 323-32, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20346678

ABSTRACT

The polycomb group (PcG) proteins are essential for the normal development of multicellular organisms. They form multi-protein complexes that work as transcriptional repressors of several thousand genes controlling differentiation pathways during development. How the PcG proteins work as transcriptional repressors is incompletely understood, but involves post-translational modifications of histones by two major PcG protein complexes: polycomb repressive complex 1 and polycomb repressive complex 2.


Subject(s)
Repressor Proteins/metabolism , Transcription, Genetic , Animals , Histones/metabolism , Humans , Polycomb-Group Proteins , Response Elements , Transcription Factors/metabolism
16.
Nat Commun ; 15(1): 5152, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38886396

ABSTRACT

In many cancers, a stem-like cell subpopulation mediates tumor initiation, dissemination and drug resistance. Here, we report that cancer stem cell (CSC) abundance is transcriptionally regulated by C-terminally phosphorylated p27 (p27pT157pT198). Mechanistically, this arises through p27 co-recruitment with STAT3/CBP to gene regulators of CSC self-renewal including MYC, the Notch ligand JAG1, and ANGPTL4. p27pTpT/STAT3 also recruits a SIN3A/HDAC1 complex to co-repress the Pyk2 inhibitor, PTPN12. Pyk2, in turn, activates STAT3, creating a feed-forward loop increasing stem-like properties in vitro and tumor-initiating stem cells in vivo. The p27-activated gene profile is over-represented in STAT3 activated human breast cancers. Furthermore, mammary transgenic expression of phosphomimetic, cyclin-CDK-binding defective p27 (p27CK-DD) increases mammary duct branching morphogenesis, yielding hyperplasia and microinvasive cancers that can metastasize to liver, further supporting a role for p27pTpT in CSC expansion. Thus, p27pTpT interacts with STAT3, driving transcriptional programs governing stem cell expansion or maintenance in normal and cancer tissues.


Subject(s)
Breast Neoplasms , Cyclin-Dependent Kinase Inhibitor p27 , Hyperplasia , Neoplastic Stem Cells , STAT3 Transcription Factor , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Humans , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Animals , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Female , Phosphorylation , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Hyperplasia/metabolism , Mice , Gene Expression Regulation, Neoplastic , Cell Self Renewal/genetics , Cell Line, Tumor , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Glands, Animal/cytology , Jagged-1 Protein/metabolism , Jagged-1 Protein/genetics
17.
Stem Cell Reports ; 18(11): 2283-2296, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37832538

ABSTRACT

Adult neurogenesis occurs in the mammalian olfactory epithelium to maintain populations of neurons that are vulnerable to injury yet essential for olfaction. Multipotent olfactory basal stem cells are activated by damage, although mechanisms regulating lineage decisions are not understood. Using mouse lesion models, we focused on defining the role of Polycomb repressive complexes (PRCs) in olfactory neurogenesis. PRC2 has a well-established role in developing tissues, orchestrating transcriptional programs via chromatin modification. PRC2 proteins are expressed in olfactory globose basal cells (GBCs) and nascent neurons. Conditional PRC2 loss perturbs lesion-induced neuron production, accompanied by altered histone modifications and misexpression of lineage-specific transcription factors in GBCs. De-repression of Sox9 in PRC2-mutant GBCs is accompanied by increased Bowman's gland production, defining an unrecognized role for PRC2 in regulating gland versus neuron cell fate. Our findings support a model for PRC2-dependent mechanisms promoting sensory neuronal differentiation in an adult neurogenic niche.


Subject(s)
Polycomb Repressive Complex 2 , Smell , Mice , Animals , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Neurogenesis/physiology , Cell Differentiation/physiology , Olfactory Mucosa , Polycomb Repressive Complex 1 , Mammals/metabolism
18.
bioRxiv ; 2023 Nov 06.
Article in English | MEDLINE | ID: mdl-38076924

ABSTRACT

Approximately 20% of head and neck squamous cell carcinomas (HNSCC) exhibit reduced methylation on lysine 36 of histone H3 (H3K36me) due to mutations in histone methylase NSD1 or a lysine-to-methionine mutation in histone H3 (H3K36M). Whether such alterations of H3K36me can be exploited for therapeutic interventions is still unknown. Here, we show that HNSCC models expressing H3K36M can be divided into two groups: those that display aberrant accumulation of H3K27me3 and those that maintain steady levels of H3K27me3. The first group shows decreased proliferation, genome instability, and increased sensitivity to genotoxic agents, such as PARP1/2 inhibitors. In contrast, the H3K36M HNSCC models with steady H3K27me3 levels do not exhibit these characteristics unless H3K27me3 levels are elevated, either by DNA hypomethylating agents or by inhibiting the H3K27me3 demethylases KDM6A/B. Mechanistically, we found that H3K36M reduces H3K36me by directly impeding the activities of the histone methyltransferase NSD3 and the histone demethylase LSD2. Notably, we found that aberrant H3K27me3 levels induced by H3K36M expression is not a bona fide epigenetic mark in HNSCC since it requires continuous expression of H3K36M to be inherited. Moreover, increased sensitivity of H3K36M HNSCC models to PARP1/2 inhibitors solely depends on the increased H3K27me3 levels. Indeed, aberrantly high H3K27me3 levels decrease BRCA1 and FANCD2-dependent DNA repair, resulting in higher sensitivity to DNA breaks and replication stress. Finally, in support of our in vitro findings, a PARP1/2 inhibitor alone reduce tumor burden in a H3K36M HNSCC xenograft model with elevated H3K27me3, whereas in a H3K36M HNSCC xenograft model with consistent H3K27me3 levels, a combination of PARP1/2 inhibitors and agents that upregulate H3K27me3 proves to be successful. In conclusion, our findings underscore a delicate balance between H3K36 and H3K27 methylation, essential for maintaining genome stability. This equilibrium presents promising therapeutic opportunities for patients with H3K36me-deficient tumors.

19.
J Clin Invest ; 133(13)2023 07 03.
Article in English | MEDLINE | ID: mdl-37200093

ABSTRACT

During emergency hematopoiesis, hematopoietic stem cells (HSCs) rapidly proliferate to produce myeloid and lymphoid effector cells, a response that is critical against infection or tissue injury. If unresolved, this process leads to sustained inflammation, which can cause life-threatening diseases and cancer. Here, we identify a role of double PHD fingers 2 (DPF2) in modulating inflammation. DPF2 is a defining subunit of the hematopoiesis-specific BAF (SWI/SNF) chromatin-remodeling complex, and it is mutated in multiple cancers and neurological disorders. We uncovered that hematopoiesis-specific Dpf2-KO mice developed leukopenia, severe anemia, and lethal systemic inflammation characterized by histiocytic and fibrotic tissue infiltration resembling a clinical hyperinflammatory state. Dpf2 loss impaired the polarization of macrophages responsible for tissue repair, induced the unrestrained activation of Th cells, and generated an emergency-like state of HSC hyperproliferation and myeloid cell-biased differentiation. Mechanistically, Dpf2 deficiency resulted in the loss of the BAF catalytic subunit BRG1 from nuclear factor erythroid 2-like 2-controlled (NRF2-controlled) enhancers, impairing the antioxidant and antiinflammatory transcriptional response needed to modulate inflammation. Finally, pharmacological reactivation of NRF2 suppressed the inflammation-mediated phenotypes and lethality of Dpf2Δ/Δ mice. Our work establishes an essential role of the DPF2-BAF complex in licensing NRF2-dependent gene expression in HSCs and immune effector cells to prevent chronic inflammation.


Subject(s)
Chromatin , Neoplasms , Mice , Animals , Antioxidants , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Chromatin Assembly and Disassembly , Inflammation/genetics , Gene Expression , DNA-Binding Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Nature ; 439(7078): 871-4, 2006 Feb 16.
Article in English | MEDLINE | ID: mdl-16357870

ABSTRACT

The establishment and maintenance of epigenetic gene silencing is fundamental to cell determination and function. The essential epigenetic systems involved in heritable repression of gene activity are the Polycomb group (PcG) proteins and the DNA methylation systems. Here we show that the corresponding silencing pathways are mechanistically linked. We find that the PcG protein EZH2 (Enhancer of Zeste homolog 2) interacts-within the context of the Polycomb repressive complexes 2 and 3 (PRC2/3)-with DNA methyltransferases (DNMTs) and associates with DNMT activity in vivo. Chromatin immunoprecipitations indicate that binding of DNMTs to several EZH2-repressed genes depends on the presence of EZH2. Furthermore, we show by bisulphite genomic sequencing that EZH2 is required for DNA methylation of EZH2-target promoters. Our results suggest that EZH2 serves as a recruitment platform for DNA methyltransferases, thus highlighting a previously unrecognized direct connection between two key epigenetic repression systems.


Subject(s)
DNA Methylation , DNA-Binding Proteins/classification , DNA-Binding Proteins/metabolism , Gene Silencing , Repressor Proteins/classification , Repressor Proteins/metabolism , Transcription Factors/classification , Transcription Factors/metabolism , Chromatin Immunoprecipitation , CpG Islands/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , HeLa Cells , Humans , Polycomb Repressive Complex 2 , Polycomb-Group Proteins , Promoter Regions, Genetic/genetics , Protein Binding , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL