Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Biomolecules ; 14(4)2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38672463

ABSTRACT

BACKGROUND: The scaffold protein tyrosine kinase substrate 4 (TKS4) undergoes tyrosine phosphorylation by the epidermal growth factor receptor (EGFR) pathway via Src kinase. The TKS4 deficiency in humans is responsible for the manifestation of a genetic disorder known as Frank-Ter Haar syndrome (FTHS). Based on our earlier investigation, the absence of TKS4 triggers migration, invasion, and epithelial-mesenchymal transition (EMT)-like phenomena while concurrently suppressing cell proliferation in HCT116 colorectal carcinoma cells. This indicates that TKS4 may play a unique role in the progression of cancer. In this study, we demonstrated that the enhancer of zeste homolog 2 (EZH2) and the histone methyltransferase of polycomb repressive complex 2 (PRC2) are involved in the migration, invasion, and EMT-like changes in TKS4-deficient cells (KO). EZH2 is responsible for the maintenance of the trimethylated lysine 27 on histone H3 (H3K27me3). METHODS: We performed transcriptome sequencing, chromatin immunoprecipitation, protein and RNA quantitative studies, cell mobility, invasion, and proliferation studies combined with/without the EZH2 activity inhibitor 3-deazanoplanocine (DZNep). RESULTS: We detected an elevation of global H3K27me3 levels in the TKS4 KO cells, which could be reduced with treatment with DZNep, an EZH2 inhibitor. Inhibition of EZH2 activity reversed the phenotypic effects of the knockout of TKS4, reducing the migration speed and wound healing capacity of the cells as well as decreasing the invasion capacity, while the decrease in cell proliferation became stronger. In addition, inhibition of EZH2 activity also reversed most epithelial and mesenchymal markers. We investigated the wider impact of TKS4 deletion on the gene expression profile of colorectal cancer cells using transcriptome sequencing of wild-type and TKS4 knockout cells, particularly before and after treatment with DZNep. Additionally, we observed changes in the expression of several protein-coding genes and long non-coding RNAs that showed a recovery in expression levels following EZH2 inhibition. CONCLUSIONS: Our results indicate that the removal of TKS4 causes a notable disruption in the gene expression pattern, leading to the disruption of several signal transduction pathways. Inhibiting the activity of EZH2 can restore most of these transcriptomics and phenotypic effects in colorectal carcinoma cells.


Subject(s)
Adenosine , Cell Movement , Cell Proliferation , Colorectal Neoplasms , Enhancer of Zeste Homolog 2 Protein , Epithelial-Mesenchymal Transition , Humans , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/deficiency , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Histones/metabolism
2.
J Cell Physiol ; 239(6): e31244, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38529784

ABSTRACT

Maternal histone methyltransferase is critical for epigenetic regulation and development of mammalian embryos by regulating histone and DNA modifications. Here, we reported a novel mechanism by revealing the critical effects of maternal Ezh1/2 deletion on mitochondria in MII oocytes and early embryos in mice. We found that Ezh1/2 knockout in mouse MII oocytes impaired the structure of mitochondria and decreased its number, but membrane potential and respiratory function of mitochondrion were increased. The similar effects of Ezh1/2 deletion have been observed in 2-cell and morula embryos, indicating that the effects of maternal Ezh1/2 deficiency on mitochondrion extend to early embryos. However, the loss of maternal Ezh1/2 resulted in a severe defect of morula: the number, membrane potential, respiratory function, and ATP production of mitochondrion dropped significantly. Content of reactive oxygen species was raised in both MII oocytes and early embryos, suggesting maternal Ezh1/2 knockout induced oxidative stress. In addition, maternal Ezh1/2 ablation interfered the autophagy in morula and blastocyst embryos. Finally, maternal Ezh1/2 deletion led to cell apoptosis in blastocyst embryos in mice. By analyzing the gene expression profile, we revealed that maternal Ezh1/2 knockout affected the expression of mitochondrial related genes in MII oocytes and early embryos. The chromatin immunoprecipitation-polymerase chain reaction assay demonstrated that Ezh1/2 directly regulated the expression of genes Fxyd6, Adpgk, Aurkb, Zfp521, Ehd3, Sgms2, Pygl, Slc1a1, and Chst12 by H3K27me3 modification. In conclusion, our study revealed the critical effect of maternal Ezh1/2 on the structure and function of mitochondria in oocytes and early embryos, and suggested a novel mechanism underlying maternal epigenetic regulation on early embryonic development through the modulation of mitochondrial status.


Subject(s)
Mitochondria , Oocytes , Polycomb Repressive Complex 2 , Animals , Female , Mice , Apoptosis/genetics , Autophagy/genetics , Blastocyst/metabolism , Embryonic Development/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/deficiency , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Membrane Potential, Mitochondrial , Mice, Knockout , Mitochondria/metabolism , Mitochondria/pathology , Mitochondria/genetics , Morula/metabolism , Oocytes/metabolism , Oxidative Stress/genetics , Polycomb Repressive Complex 2/metabolism , Polycomb Repressive Complex 2/genetics , Reactive Oxygen Species/metabolism , Histones/metabolism
3.
Nat Commun ; 12(1): 7019, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34857757

ABSTRACT

Yolk sac (YS) hematopoiesis is critical for the survival of the embryo and a major source of tissue-resident macrophages that persist into adulthood. Yet, the transcriptional and epigenetic regulation of YS hematopoiesis remains poorly characterized. Here we report that the epigenetic regulator Ezh2 is essential for YS hematopoiesis but dispensable for subsequent aorta-gonad-mesonephros (AGM) blood development. Loss of EZH2 activity in hemogenic endothelium (HE) leads to the generation of phenotypically intact but functionally deficient erythro-myeloid progenitors (EMPs), while the generation of primitive erythroid cells is not affected. EZH2 activity is critical for the generation of functional EMPs at the onset of the endothelial-to-hematopoietic transition but subsequently dispensable. We identify a lack of Wnt signaling downregulation as the primary reason for the production of non-functional EMPs. Together, our findings demonstrate a critical and stage-specific role of Ezh2 in modulating Wnt signaling during the generation of EMPs from YS HE.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Erythroid Cells/metabolism , Gene Expression Regulation, Developmental , Mouse Embryonic Stem Cells/metabolism , Myeloid Progenitor Cells/metabolism , Vesicular Transport Proteins/genetics , Yolk Sac/metabolism , Animals , Cell Differentiation , Embryo, Mammalian , Enhancer of Zeste Homolog 2 Protein/deficiency , Epigenesis, Genetic , Erythroid Cells/cytology , Female , Fetus , Genes, Reporter , Hematopoiesis/genetics , Liver/cytology , Liver/growth & development , Liver/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mouse Embryonic Stem Cells/cytology , Myeloid Progenitor Cells/pathology , Primary Cell Culture , Vesicular Transport Proteins/metabolism , Wnt Signaling Pathway , Yolk Sac/cytology , Yolk Sac/growth & development , Red Fluorescent Protein
4.
Nat Commun ; 12(1): 6323, 2021 11 03.
Article in English | MEDLINE | ID: mdl-34732703

ABSTRACT

Cancers develop from the accumulation of somatic mutations, yet it remains unclear how oncogenic lesions cooperate to drive cancer progression. Using a mouse model harboring NRasG12D and EZH2 mutations that recapitulates leukemic progression, we employ single-cell transcriptomic profiling to map cellular composition and gene expression alterations in healthy or diseased bone marrows during leukemogenesis. At cellular level, NRasG12D induces myeloid lineage-biased differentiation and EZH2-deficiency impairs myeloid cell maturation, whereas they cooperate to promote myeloid neoplasms with dysregulated transcriptional programs. At gene level, NRasG12D and EZH2-deficiency independently and synergistically deregulate gene expression. We integrate results from histopathology, leukemia repopulation, and leukemia-initiating cell assays to validate transcriptome-based cellular profiles. We use this resource to relate developmental hierarchies to leukemia phenotypes, evaluate oncogenic cooperation at single-cell and single-gene levels, and identify GEM as a regulator of leukemia-initiating cells. Our studies establish an integrative approach to deconvolute cancer evolution at single-cell resolution in vivo.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/metabolism , Leukemia/genetics , Leukemia/metabolism , Single-Cell Analysis , Animals , Apoptosis , Cell Cycle , Enhancer of Zeste Homolog 2 Protein/deficiency , Enhancer of Zeste Homolog 2 Protein/genetics , Epigenomics , GTP Phosphohydrolases , Gene Expression Regulation, Neoplastic , Genetic Heterogeneity , Humans , Leukemia, Myeloid, Acute , Membrane Proteins , Mice, Knockout , Mutation , Myeloid Cells , Oncogenes , Phenotype , Transcriptome
5.
Carcinogenesis ; 42(12): 1485-1495, 2021 12 31.
Article in English | MEDLINE | ID: mdl-34614148

ABSTRACT

Oral squamous cell carcinoma (oral SCC) is an aggressive disease and despite intensive treatments, 5-year survival rates for patients have remained low in the last 20 years. Enhancer of zeste homolog 2 (EZH2), part of polycomb repressive complex 2 (PRC2), is highly expressed in human oral SCC samples and cell lines and has been associated with greater epithelia-to-mesenchymal transition (EMT), invasion and metastasis. Here, we developed a tamoxifen-regulated, transgenic mouse line (KcEZH2) in which EZH2 is selectively knocked out (KO) in some tongue epithelial basal stem cells (SCs) in adult mice. EZH2 KO SCs do not show the H3K27me3 mark, as assessed by double-label immunofluorescence. We used this mouse line to assess EZH2 actions during oral tumorigenesis with our immunocompetent 4-nitroquinoline 1-oxide model of oral SCC. We report that higher percentages of mice with invasive SCCs and high-grade neoplastic lesions are observed in mice containing EZH2 KO SCs (KcEZH2-2TΔ and KcEZH2-5TΔ mice). Moreover, EZH2 expression does not correlate with the expression of markers of invasive SCCs. Finally, EZH2 KO cells that are E-cadherin+ are present at invasion fronts infiltrating underlying muscle tissue. Our findings indicate that the knockout of EZH2 in basal SCs of tongue epithelia results in more aggressive carcinomas, and this should be considered when targeting EZH2 as a therapeutic strategy.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/deficiency , Mouth Mucosa/metabolism , Mouth Mucosa/pathology , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Animals , Biomarkers, Tumor , Cell Line, Tumor , DNA Methylation , Disease Models, Animal , Disease Susceptibility , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Genetic Predisposition to Disease , Histones/metabolism , Humans , Hyaluronan Receptors/metabolism , Immunohistochemistry , Mice , Mice, Transgenic , Neoplasm Invasiveness , Phosphorylation , STAT3 Transcription Factor/metabolism , Squamous Cell Carcinoma of Head and Neck/drug therapy , Tamoxifen/pharmacology , Xenograft Model Antitumor Assays
6.
Breast Cancer ; 28(2): 355-367, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32990923

ABSTRACT

BACKGROUND: Acquired resistance to drug involves multilayered genetic and epigenetic regulation. Inhibition of EZH2 has proven to reverse the tamoxifen resistance back to the sensitive state in breast cancer. However, the molecular players involved in EZH2-mediated effects on tamoxifen-resistant MCF-7 cells are unknown. This study was conducted to understand the global change in proteome profile of tamoxifen-resistant MCF-7 breast cancer cells upon EZH2 knockdown. METHODS: Tamoxifen resistance MCF-7 breast cancer cells were established using increasing concentrations of 4-hydroxy tamoxifen. Using label free proteomics approach, we studied the alteration in total proteome in resistant cells as well as cells transfected with siEZH2 in comparison to sensitive and cells transfected with non-targeting siRNA. RESULTS: Here, we report list of proteins that were previously not recognized for their role in tamoxifen resistance and hold a close association with breast cancer patient survival. Proteins Annexin A2, CD44, nucleosome assembly protein 1, and lamin A/C were among the most upregulated protein in tamoxifen-resistant cells that were found to be abrogated upon EZH2 knockdown. The study suggests the involvement for various proteins in acquiring resistance towards tamoxifen and anticipates further research for investigating their therapeutic potentials. CONCLUSION: Overall, we propose that targeting EZH2 or the molecules down the cascade might be helpful in reacquiring sensitivity to tamoxifen in breast cancer.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Gene Knockdown Techniques/methods , Proteome/metabolism , Tamoxifen/pharmacology , Breast Neoplasms/pathology , Cell Survival/drug effects , Cell Survival/genetics , Enhancer of Zeste Homolog 2 Protein/deficiency , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Proteomics/methods , RNA, Small Interfering/genetics , Signal Transduction/genetics , Transfection , Up-Regulation/genetics
7.
Cell Cycle ; 19(5): 551-566, 2020 03.
Article in English | MEDLINE | ID: mdl-32054404

ABSTRACT

Recently, the role of microRNAs (miRs) in human diseases has been verified. This study was determined to explore the protective effects of microRNA-26a (miR-26a) in steroid-induced osteonecrosis of the femoral head (SONFH) with the involvement of enhancer of zeste homologue 2 (EZH2).Femoral head (FH) samples from SONFH patients and patients with femoral neck fracture were collected, and rat SONFH models were established by Escherichia coli endotoxin combining with large dose steroid pulse assay. The hemorheology, blood lipid, inflammatory factors, and pathologic changes were measured by a series of experiments. Moreover, the detection of osteoblasts, osteoclasts, miR-26a expression, EZH2 expression, osteoprotegerin (OPG) and osteoprotegerin ligand (OPGL), and the apoptosis of osteocytes were conducted. The target relation between miR-26a and EZH2 was clarified by bioinformatics and dual-luciferase reporter gene assay.MiR-26a was poorly expressed, while EZH2 was highly expressed in SONFH, and the elevation of miR-26a could repress EZH2 expression. Elevated miR-26a and reduced EZH2 were able to decelerate the apoptosis of osteocytes, increase osteoblasts, and decrease osteoclasts, resulting in a repression of SONFH progression. Additionally, EZH2 was a target gene of miR-26a. Furthermore, the elevation of EZH2 could reverse the repression of SONFH progression that is induced by elevated miR-26a.We found that up-regulation of miR-26a and knockdown of EZH2 could suppress the development of SONFH, which would contribute to the therapy of SONFH.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/deficiency , Femur Head/metabolism , MicroRNAs/genetics , Osteonecrosis/genetics , Osteonecrosis/therapy , Steroids/adverse effects , Animals , Apoptosis , Disease Models, Animal , Down-Regulation , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Femur Head/cytology , Femur Head/pathology , Humans , Inflammation/blood , Inflammation/chemically induced , Inflammation/genetics , Inflammation/therapy , Lipids/blood , Male , Middle Aged , Osteocytes/cytology , Osteocytes/metabolism , Osteocytes/pathology , Osteonecrosis/blood , Osteonecrosis/pathology , Rats , Rats, Sprague-Dawley , Up-Regulation
8.
J Autoimmun ; 108: 102404, 2020 03.
Article in English | MEDLINE | ID: mdl-31952907

ABSTRACT

The chromatin modifier enhancer of zeste homolog 2 (EZH2) methylates lysine 27 of histone H3 (H3K27) and regulates T cell differentiation. However, the potential role of EZH2 in the pathogenesis of rheumatoid arthritis (RA) remains elusive. We analyzed EZH2 expression in PBMC, CD4+ T cells, CD19+ B cell, and CD14+ monocytes from active treatment-naïve RA patients and healthy controls (HC). We also suppressed EZH2 expression using EZH2 inhibitor GSK126 and measured CD4+ T cell differentiation, proliferation and apoptosis. We further examined TGFß-SMAD and RUNX1 signaling pathways in EZH2-suppressed CD4+ T cells. Finally, we explored the regulation mechanism of EZH2 by RA synovial fluid and fibroblast-like synoviocyte (FLS) by neutralizing key proinflammatory cytokines. EZH2 expression is lower in PBMC and CD4+ T cells from RA patients than those from HC. EZH2 inhibition suppressed regulatory T cells (Tregs) differentiation and FOXP3 transcription, and downregulated RUNX1 and upregulated SMAD7 expression in CD4+ T cells. RA synovial fluid and fibroblast-like synoviocytes suppressed EZH2 expression in CD4+ T cells, which was partially neutralized by anti-IL17 antibody. Taken together, EZH2 in CD4+ T cells from RA patients was attenuated, which suppressed FOXP3 transcription through downregulating RUNX1 and upregulating SMAD7 in CD4+ T cells, and ultimately suppressed Tregs differentiation. IL17 in RA synovial fluid might promote downregulation of EZH2 in CD4+ T cells. Defective EZH2 in CD4+ T cells might contribute to Treg deficiency in RA.


Subject(s)
Arthritis, Rheumatoid/etiology , Cell Differentiation/genetics , Cell Differentiation/immunology , Enhancer of Zeste Homolog 2 Protein/deficiency , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers , Case-Control Studies , Cytokines/metabolism , Histones/metabolism , Humans , Inflammation Mediators/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Monocytes/immunology , Monocytes/metabolism , Synovial Fluid/immunology , Synovial Fluid/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology
9.
Front Immunol ; 11: 594603, 2020.
Article in English | MEDLINE | ID: mdl-33574814

ABSTRACT

Macrophages define a key component of immune cells present in atherosclerotic lesions and are central regulators of the disease. Since epigenetic processes are important in controlling macrophage function, interfering with epigenetic pathways in macrophages might be a novel approach to combat atherosclerosis. Histone H3K27 trimethylation is a repressive histone mark catalyzed by polycomb repressive complex with EZH2 as the catalytic subunit. EZH2 is described to increase macrophage inflammatory responses by supressing the suppressor of cytokine signaling, Socs3. We previously showed that myeloid deletion of Kdm6b, an enzymes that in contrast to EZH2 removes repressive histone H3K27me3 marks, results in advanced atherosclerosis. Because of its opposing function and importance of EZH2 in macrophage inflammatory responses, we here studied the role of myeloid EZH2 in atherosclerosis. A myeloid-specific Ezh2 deficient mouse strain (Ezh2del) was generated (LysM-cre+ x Ezh2fl/fl) and bone marrow from Ezh2del or Ezh2wt mice was transplanted to Ldlr-/- mice which were fed a high fat diet for 9 weeks to study atherosclerosis. Atherosclerotic lesion size was significantly decreased in Ezh2del transplanted mice compared to control. The percentage of macrophages in the atherosclerotic lesion was similar, however neutrophil numbers were lower in Ezh2del transplanted mice. Correspondingly, the migratory capacity of neutrophils was decreased in Ezh2del mice. Moreover, peritoneal Ezh2del foam cells showed a reduction in the inflammatory response with reduced production of nitric oxide, IL-6 and IL-12. In Conclusion, myeloid Ezh2 deficiency impairs neutrophil migration and reduces macrophage foam cell inflammatory responses, both contributing to reduced atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Enhancer of Zeste Homolog 2 Protein/deficiency , Foam Cells/immunology , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Enhancer of Zeste Homolog 2 Protein/immunology , Foam Cells/pathology , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Mice , Mice, Knockout , Organ Specificity
10.
Mol Cancer Res ; 18(2): 278-286, 2020 02.
Article in English | MEDLINE | ID: mdl-31704732

ABSTRACT

Mechanisms of intrinsic resistance of serous ovarian cancers to standard treatment with carboplatin and paclitaxel are poorly understood. Seventeen primary serous ovarian cancers classified as responders or nonresponders to standard treatment were screened with DigiWest protein array analysis for 279 analytes. Histone methyl transferase EZH2, an interaction partner of ataxia telangiectasia mutated (ATM), was found as one of the most significantly represented proteins in responsive tumors. Survival analysis of 616 patients confirmed a better outcome in patients with high EZH2 expression, but a worse outcome in patients with low EZH2 and high-ATM-expressing tumors compared with patients with low EZH2 and low-ATM-expressing tumors. A proximity ligation assay further confirmed an association between ATM and EZH2 in tumors of patients with an increased disease-free survival. Knockdown of EZH2 resulted in treatment-resistant cells, but suppression of both EZH2 and ATM, or ATM alone, had no effect. DigiWest protein analysis of EZH2-knockdown cells revealed a decrease in proteins involved in mitotic processes and checkpoint regulation, suggesting that deregulated ATM may induce treatment resistance. IMPLICATIONS: Ovarian cancer is a malignancy with high mortality rates, with to date, no successful molecular characterization strategies. Our study uncovers in a comprehensive approach the involvement of checkpoint regulation via ATM and EZH2, potentially providing a new therapeutic perspective for further investigations.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Carboplatin/pharmacology , Cystadenocarcinoma, Serous/drug therapy , Enhancer of Zeste Homolog 2 Protein/metabolism , Ovarian Neoplasms/drug therapy , Paclitaxel/pharmacology , Aged , Ataxia Telangiectasia Mutated Proteins/genetics , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , DNA Damage , DNA, Neoplasm/genetics , Drug Resistance, Neoplasm , Enhancer of Zeste Homolog 2 Protein/deficiency , Enhancer of Zeste Homolog 2 Protein/genetics , Female , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction
11.
Sci Rep ; 9(1): 4319, 2019 03 13.
Article in English | MEDLINE | ID: mdl-30867490

ABSTRACT

Trimethylation on H3K27 mediated by Polycomb Repressive Complex 2 (PRC2) is required to control gene repression programs involved in development, regulation of tissue homeostasis or maintenance and lineage specification of stem cells. In Drosophila, the PRC2 catalytic subunit is the single protein E(z), while in mammals this function is fulfilled by two proteins, Ezh1 and Ezh2. Based on database searches, we propose that Ezh1 arose from an Ezh2 gene duplication that has occurred in the common ancestor to elasmobranchs and bony vertebrates. Expression studies in zebrafish using in situ hybridization and RT-PCR followed by the sequencing of the amplicon revealed that ezh1 mRNAs are maternally deposited. Then, ezh1 transcripts are ubiquitously distributed in the entire embryo at 24 hpf and become more restricted to anterior part of the embryo at later developmental stages. To unveil the function of ezh1 in zebrafish, a mutant line was generated using the TALEN technology. Ezh1-deficient mutant fish are viable and fertile, but the loss of ezh1 function is responsible for the earlier death of ezh2 mutant larvae indicating that ezh1 contributes to zebrafish development in absence of zygotic ezh2 gene function. Furthermore, we show that presence of ezh1 transcripts from the maternal origin accounts for the delayed lethality of ezh2-deficient larvae.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Gene Duplication , Polycomb Repressive Complex 2/genetics , Zebrafish Proteins/genetics , Zebrafish/growth & development , Animals , Enhancer of Zeste Homolog 2 Protein/deficiency , Enhancer of Zeste Homolog 2 Protein/physiology , Longevity , Polycomb Repressive Complex 2/deficiency , Polycomb Repressive Complex 2/physiology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/physiology
12.
Int J Hematol ; 110(2): 170-178, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30706327

ABSTRACT

Polycomb proteins function in the maintenance of gene silencing via post-translational modifications of histones and chromatin compaction. Genetic and biochemical studies have revealed that the repressive function of Polycomb repressive complexes (PRCs) in transcription is counteracted by the activating function of Trithorax-group complexes; this balance fine-tunes the expression of genes critical for development and tissue homeostasis. The function of PRCs is frequently dysregulated in various cancer cells due to altered expression or recurrent somatic mutations in PRC genes. The tumor suppressive functions of EZH2-containing PRC2 and a PRC2-related protein ASXL1 have been investigated extensively in the pathogenesis of hematological malignancies, including myeloproliferative neoplasms (MPN). BCOR, a component of non-canonical PRC1, suppresses various hematological malignancies including MPN. In this review, we focus on recent findings on the role of PRCs in the pathogenesis of MPN and the therapeutic impact of targeting the pathological functions of PRCs in MPN.


Subject(s)
Cell Transformation, Neoplastic/genetics , Myeloproliferative Disorders/genetics , Neoplasm Proteins/physiology , Polycomb-Group Proteins/physiology , Cell Cycle Proteins/physiology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/deficiency , Enhancer of Zeste Homolog 2 Protein/physiology , Gain of Function Mutation , Gene Expression Regulation, Neoplastic , Hematopoiesis , Histone Code , Histone-Lysine N-Methyltransferase/physiology , Humans , Molecular Targeted Therapy , Myeloid-Lymphoid Leukemia Protein/physiology , Myeloproliferative Disorders/metabolism , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb-Group Proteins/deficiency , Polycomb-Group Proteins/genetics , Proto-Oncogene Proteins/physiology , Repressor Proteins/physiology , Transcription Factors/physiology
13.
J Clin Invest ; 128(9): 3872-3886, 2018 08 31.
Article in English | MEDLINE | ID: mdl-30080177

ABSTRACT

Early T cell precursor acute lymphoblastic leukemia (ETP-ALL) is a new pathological entity with poor outcomes in T cell ALL (T-ALL) that is characterized by a high incidence of loss-of-function mutations in polycomb repressive complex 2 (PRC2) genes. We generated a mouse model of ETP-ALL by deleting Ezh2, one of the PRC2 genes, in p53-null hematopoietic cells. The loss of Ezh2 in p53-null hematopoietic cells impeded the differentiation of ETPs and eventually induced ETP-ALL-like disease in mice, indicating that PRC2 functions as a bona fide tumor suppressor in ETPs. A large portion of PRC2 target genes acquired DNA hypermethylation of their promoters following reductions in H3K27me3 levels upon the loss of Ezh2, which included pivotal T cell differentiation-regulating genes. The reactivation of a set of regulators by a DNA-demethylating agent, but not the transduction of single regulator genes, effectively induced the differentiation of ETP-ALL cells. Thus, PRC2 protects key T cell developmental regulators from DNA hypermethylation in order to keep them primed for activation upon subsequent differentiation phases, while its insufficiency predisposes ETPs to leukemic transformation. These results revealed a previously unrecognized epigenetic switch in response to PRC2 dysfunction and provide the basis for specific rational epigenetic therapy for ETP-ALL with PRC2 insufficiency.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Enhancer of Zeste Homolog 2 Protein/deficiency , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Transformation, Neoplastic/pathology , DNA Methylation , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic , Genes, p53 , Mice , Mice, Inbred C57BL , Mice, Knockout , Polycomb Repressive Complex 2/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology
14.
Nat Commun ; 9(1): 2547, 2018 06 29.
Article in English | MEDLINE | ID: mdl-29959321

ABSTRACT

Emerging evidence has illustrated the importance of epigenomic reprogramming in cancer, with altered post-translational modifications of histones contributing to pathogenesis. However, the contributions of histone modifiers to breast cancer progression are unclear, and how these processes vary between molecular subtypes has yet to be adequately addressed. Here we report that genetic or pharmacological targeting of the epigenetic modifier Ezh2 dramatically hinders metastatic behaviour in both a mouse model of breast cancer and patient-derived xenografts reflective of the Luminal B subtype. We further define a subtype-specific molecular mechanism whereby EZH2 maintains H3K27me3-mediated repression of the FOXC1 gene, thereby inactivating a FOXC1-driven, anti-invasive transcriptional program. We demonstrate that higher FOXC1 is predictive of favourable outcome specifically in Luminal B breast cancer patients and establish the use of EZH2 methyltransferase inhibitors as a viable strategy to block metastasis in Luminal B breast cancer, where options for targeted therapy are limited.


Subject(s)
Breast Neoplasms/drug therapy , Enhancer of Zeste Homolog 2 Protein/genetics , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Histones/genetics , Indoles/pharmacology , Lung Neoplasms/drug therapy , Pyridones/pharmacology , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Doxycycline/pharmacology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/deficiency , Enzyme Inhibitors/pharmacology , Female , Forkhead Transcription Factors/agonists , Forkhead Transcription Factors/metabolism , Histones/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Mice , Mice, Knockout , Molecular Targeted Therapy , Protein Processing, Post-Translational , Signal Transduction , Transcription, Genetic , Xenograft Model Antitumor Assays
15.
Cell Rep ; 23(11): 3262-3274, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29898397

ABSTRACT

Regulatory T cells (Tregs) are critical for maintaining immune homeostasis, but their presence in tumor tissues impairs anti-tumor immunity and portends poor prognoses in cancer patients. Here, we reveal a mechanism to selectively target and reprogram the function of tumor-infiltrating Tregs (TI-Tregs) by exploiting their dependency on the histone H3K27 methyltransferase enhancer of zeste homolog 2 (EZH2) in tumors. Disruption of EZH2 activity in Tregs, either pharmacologically or genetically, drove the acquisition of pro-inflammatory functions in TI-Tregs, remodeling the tumor microenvironment and enhancing the recruitment and function of CD8+ and CD4+ effector T cells that eliminate tumors. Moreover, abolishing EZH2 function in Tregs was mechanistically distinct from, more potent than, and less toxic than a generalized Treg depletion approach. This study reveals a strategy to target Tregs in cancer that mitigates autoimmunity by reprogramming their function in tumors to enhance anti-cancer immunity.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , T-Lymphocytes, Regulatory/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein/deficiency , Enhancer of Zeste Homolog 2 Protein/genetics , Forkhead Transcription Factors/metabolism , Humans , Interferon-gamma/metabolism , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Neoplasms/pathology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment , Tumor Necrosis Factor-alpha/metabolism
16.
J Exp Med ; 215(5): 1365-1382, 2018 05 07.
Article in English | MEDLINE | ID: mdl-29626115

ABSTRACT

Histone 3 Lys27 (H3K27) trimethyltransferase Ezh2 is implicated in the pathogenesis of autoimmune inflammation. Nevertheless, the role of Ezh2 in macrophage/microglial activation remains to be defined. In this study, we identified that macrophage/microglial H3K27me3 or Ezh2, rather than functioning as a repressor, mediates toll-like receptor (TLR)-induced proinflammatory gene expression, and therefore Ezh2 depletion diminishes macrophage/microglial activation and attenuates the autoimmune inflammation in dextran sulfate sodium-induced colitis and experimental autoimmune encephalomyelitis. Mechanistic characterizations indicated that Ezh2 deficiency directly stimulates suppressor of cytokine signaling 3 (Socs3) expression and therefore enhances the Lys48-linked ubiquitination and degradation of tumor necrosis factor receptor-associated factor 6. As a consequence, TLR-induced MyD88-dependent nuclear factor κB activation and the expression of proinflammatory genes in macrophages/microglia are compromised in the absence of Ezh2. The functional dependence of Ezh2 for Socs3 is further illustrated by the rescue experiments in which silencing of Socs3 restores macrophage activation and rescues autoimmune inflammation in macrophage/microglial Ezh2-deficient mice. Together, these findings establish Ezh2 as a macrophage lineage-specific mediator of autoimmune inflammation and highlight a previously unknown mechanism of Ezh2 function.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Inflammation/pathology , Macrophages/metabolism , Microglia/metabolism , Suppressor of Cytokine Signaling 3 Protein/antagonists & inhibitors , Animals , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Dextran Sulfate , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Enhancer of Zeste Homolog 2 Protein/deficiency , Epigenesis, Genetic/drug effects , Histones/metabolism , Indoles/pharmacology , Lysine/metabolism , Macrophages/drug effects , Methylation , Mice , Microglia/drug effects , Models, Biological , Myeloid Differentiation Factor 88/metabolism , Proteolysis/drug effects , Pyridones/pharmacology , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 3 Protein/metabolism , TNF Receptor-Associated Factor 6/metabolism
17.
Nat Commun ; 8(1): 1312, 2017 11 03.
Article in English | MEDLINE | ID: mdl-29101351

ABSTRACT

Mesenchymal stem/progenitor cells (MSPCs) undergo rapid self-renewal and differentiation, contributing to fast skeletal growth during childhood and puberty. It remains unclear whether these cells change their properties during late puberty to young adulthood, when bone growth and accrual decelerate. Here we show that MSPCs in primary spongiosa of long bone in mice at late puberty undergo normal programmed senescence, characterized by loss of nestin expression. MSPC senescence is epigenetically controlled by the polycomb histone methyltransferase enhancer of zeste homolog 2 (Ezh2) and its trimethylation of histone H3 on Lysine 27 (H3K27me3) mark. Ezh2 maintains the repression of key cell senescence inducer genes through H3K27me3, and deletion of Ezh2 in early pubertal mice results in premature cellular senescence, depleted MSPCs pool, and impaired osteogenesis as well as osteoporosis in later life. Our data reveals a programmed cell fate change in postnatal skeleton and unravels a regulatory mechanism underlying this phenomenon.


Subject(s)
Bone Development/physiology , Cellular Senescence/physiology , Enhancer of Zeste Homolog 2 Protein/metabolism , Sexual Maturation/physiology , Adolescent , Animals , Bone Development/genetics , Cellular Senescence/genetics , Enhancer of Zeste Homolog 2 Protein/deficiency , Enhancer of Zeste Homolog 2 Protein/genetics , Epigenesis, Genetic , Female , Histones/metabolism , Humans , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Methylation , Mice , Mice, Knockout , Mice, Transgenic , Nestin/metabolism , Osteogenesis/genetics , Osteogenesis/physiology , Osteoporosis/etiology , Osteoporosis/genetics , Osteoporosis/metabolism , Puberty/genetics , Puberty/physiology , Sexual Maturation/genetics
18.
J Immunol ; 198(12): 4682-4691, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28490575

ABSTRACT

The histone methyltransferase EZH2 is required for B and T cell development; however, the molecular mechanisms underlying this requirement remain elusive. In a murine model of lymphoid-specific EZH2 deficiency we found that EZH2 was required for proper development of adaptive, but not innate, lymphoid cells. In adaptive lymphoid cells EZH2 prevented the premature expression of Cdkn2a and the consequent stabilization of p53, an effector of the pre-Ag receptor checkpoints. Deletion of Cdkn2a in EZH2-deficient lymphocytes prevented p53 stabilization, extended lymphocyte survival, and restored differentiation resulting in the generation of mature B and T lymphocytes. Our results uncover a crucial role for EZH2 in adaptive lymphocytes to control the developmental timing of effectors of the pre-Ag receptor checkpoints.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Receptors, Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Adaptive Immunity , Animals , B-Lymphocytes/immunology , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Enhancer of Zeste Homolog 2 Protein/deficiency , Enhancer of Zeste Homolog 2 Protein/genetics , Gene Expression Regulation , Genes, p53 , Killer Cells, Natural/immunology , Lymphopoiesis , Mice , Receptors, Antigen/genetics , Receptors, Antigen/immunology
19.
Int J Biochem Cell Biol ; 88: 226-235, 2017 07.
Article in English | MEDLINE | ID: mdl-28119131

ABSTRACT

In addition to ß-cells, pancreatic islets contain α- and δ-cells, which respectively produce glucagon and somatostatin. The reprogramming of these two endocrine cell types into insulin producers, as observed after a massive ß-cell ablation in mice, may help restoring a functional ß-cell mass in type 1 diabetes. Yet, the spontaneous α-to-ß and δ-to-ß conversion processes are relatively inefficient in adult animals and the underlying epigenetic mechanisms remain unclear. Several studies indicate that the conserved chromatin modifiers DNA methyltransferase 1 (Dnmt1) and Enhancer of zeste homolog 2 (Ezh2) are important for pancreas development and restrict islet cell plasticity. Here, to investigate the role of these two enzymes in α- and δ-cell development and fate maintenance, we genetically inactivated them in each of these two cell types. We found that loss of Dnmt1 does not enhance the conversion of α- or δ-cells toward a ß-like fate. In addition, while Dnmt1 was dispensable for the development of these two cell types, we noticed a gradual loss of α-, but not δ-cells in adult mice. Finally, we found that Ezh2 inactivation does not enhance α-cell plasticity, and, contrary to what is observed in ß-cells, does not impair α-cell proliferation. Our results indicate that both Dnmt1 and Ezh2 play distinct roles in the different islet cell types.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Glucagon-Secreting Cells/metabolism , Homeostasis , Somatostatin-Secreting Cells/metabolism , Animals , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/deficiency , Enhancer of Zeste Homolog 2 Protein/deficiency , Enzyme Activation , Glucagon-Secreting Cells/cytology , Mice , Somatostatin-Secreting Cells/cytology
20.
J Exp Med ; 213(8): 1479-96, 2016 07 25.
Article in English | MEDLINE | ID: mdl-27401344

ABSTRACT

Myeloproliferative neoplasm (MPN) patients frequently show co-occurrence of JAK2-V617F and mutations in epigenetic regulator genes, including EZH2 In this study, we show that JAK2-V617F and loss of Ezh2 in hematopoietic cells contribute synergistically to the development of MPN. The MPN phenotype induced by JAK2-V617F was accentuated in JAK2-V617F;Ezh2(-/-) mice, resulting in very high platelet and neutrophil counts, more advanced myelofibrosis, and reduced survival. These mice also displayed expansion of the stem cell and progenitor cell compartments and a shift of differentiation toward megakaryopoiesis at the expense of erythropoiesis. Single cell limiting dilution transplantation with bone marrow from JAK2-V617F;Ezh2(+/-) mice showed increased reconstitution and MPN disease initiation potential compared with JAK2-V617F alone. RNA sequencing in Ezh2-deficient hematopoietic stem cells (HSCs) and megakaryocytic erythroid progenitors identified highly up-regulated genes, including Lin28b and Hmga2, and chromatin immunoprecipitation (ChIP)-quantitative PCR (qPCR) analysis of their promoters revealed decreased H3K27me3 deposition. Forced expression of Hmga2 resulted in increased chimerism and platelet counts in recipients of retrovirally transduced HSCs. JAK2-V617F-expressing mice treated with an Ezh2 inhibitor showed higher platelet counts than vehicle controls. Our data support the proposed tumor suppressor function of EZH2 in patients with MPN and call for caution when considering using Ezh2 inhibitors in MPN.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/deficiency , Hematologic Neoplasms , Janus Kinase 2 , Mutation, Missense , Myeloproliferative Disorders , Tumor Suppressor Proteins/deficiency , Amino Acid Substitution , Animals , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Mice , Mice, Mutant Strains , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...