Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 166
1.
Nucleic Acids Res ; 50(1): 244-258, 2022 01 11.
Article En | MEDLINE | ID: mdl-34904670

Loss of genome stability leads to reduced fitness, fertility and a high mutation rate. Therefore, the genome is guarded by the pathways monitoring its integrity and neutralizing DNA lesions. To analyze the mechanism of DNA damage induction by cytidine analog zebularine, we performed a forward-directed suppressor genetic screen in the background of Arabidopsis thaliana zebularine-hypersensitive structural maintenance of chromosomes 6b (smc6b) mutant. We show that smc6b hypersensitivity was suppressed by the mutations in EQUILIBRATIVE NUCLEOSIDE TRANSPORTER 3 (ENT3), DNA METHYLTRANSFERASE 1 (MET1) and DECREASE IN DNA METHYLATION 1 (DDM1). Superior resistance of ent3 plants to zebularine indicated that ENT3 is likely necessary for the import of the drug to the cells. Identification of MET1 and DDM1 suggested that zebularine induces DNA damage by interference with the maintenance of CG DNA methylation. The same holds for structurally similar compounds 5-azacytidine and 2-deoxy-5-azacytidine. Based on our genetic and biochemical data, we propose that zebularine induces enzymatic DNA-protein crosslinks (DPCs) of MET1 and zebularine-containing DNA in Arabidopsis, which was confirmed by native chromatin immunoprecipitation experiments. Moreover, zebularine-induced DPCs accumulate preferentially in 45S rDNA chromocenters in a DDM1-dependent manner. These findings open a new avenue for studying genome stability and DPC repair in plants.


Cytidine/analogs & derivatives , Heterochromatin/metabolism , Mutagens/toxicity , RNA, Ribosomal/genetics , Arabidopsis , Arabidopsis Proteins/genetics , Cell Cycle Proteins/genetics , Cytidine/toxicity , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA-Binding Proteins/genetics , Drug Resistance , Heterochromatin/drug effects , Membrane Transport Proteins/genetics , Mutation , RNA, Ribosomal/drug effects , Transcription Factors/genetics
2.
Oxid Med Cell Longev ; 2021: 5529518, 2021.
Article En | MEDLINE | ID: mdl-34603598

T-cell malignancies are still difficult to treat due to a paucity of plans that target critical dependencies. Drug-induced cellular senescence provides a permanent cell cycle arrest during tumorigenesis and cancer development, particularly when combined with senolytics to promote apoptosis of senescent cells, which is an innovation for cancer therapy. Here, our research found that wogonin, a well-known natural flavonoid compound, not only had a potential to inhibit cell growth and proliferation but also induced cellular senescence in T-cell malignancies with nonlethal concentration. Transcription activity of senescence-suppression human telomerase reverse transcriptase (hTERT) and oncogenic C-MYC was suppressed in wogonin-induced senescent cells, resulting in the inhibition of telomerase activity. We also substantiated the occurrence of DNA damage during the wogonin-induced aging process. Results showed that wogonin increased the activity of senescence-associated ß-galactosidase (SA-ß-Gal) and activated the DNA damage response pathway mediated by p53. In addition, we found the upregulated expression of BCL-2 in senescent T-cell malignancies because of the antiapoptotic properties of senescent cells. Following up this result, we identified a BCL-2 inhibitor Navitoclax (ABT-263), which was highly effective in decreasing cell viability and inducing apoptotic cell death in wogonin-induced senescent cells. Thus, the "one-two punch" approach increased the sensibility of T-cell malignancies with low expression of BCL-2 to Navitoclax. In conclusion, our research revealed that wogonin possesses potential antitumor effects based on senescence induction, offering a better insight into the development of novel therapeutic methods for T-cell malignancies.


Antineoplastic Agents/pharmacology , Cellular Senescence/drug effects , Tumor Suppressor Protein p53/metabolism , Aniline Compounds/pharmacology , Antineoplastic Agents/therapeutic use , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival/drug effects , DNA Damage/drug effects , Drugs, Chinese Herbal/therapeutic use , Flavanones/pharmacology , Flavanones/therapeutic use , Heterochromatin/drug effects , Heterochromatin/genetics , Heterochromatin/metabolism , Humans , Lymphoma, T-Cell/drug therapy , Lymphoma, T-Cell/pathology , RNA Interference , RNA, Small Interfering/metabolism , Sulfonamides/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
3.
J Enzyme Inhib Med Chem ; 36(1): 856-868, 2021 Dec.
Article En | MEDLINE | ID: mdl-33771089

The present study describes evaluation of epigenetic regulation by a small molecule as the therapeutic potential for treatment of Huntington's disease (HD). We identified 5-allyloxy-2-(pyrrolidin-1-yl)quinoline (APQ) as a novel SETDB1/ESET inhibitor using a combined in silico and in vitro cell based screening system. APQ reduced SETDB1 activity and H3K9me3 levels in a HD cell line model. In particular, not only APQ reduced H3K9me3 levels in the striatum but it also improved motor function and neuropathological symptoms such as neuronal size and activity in HD transgenic (YAC128) mice with minimal toxicity. Using H3K9me3-ChIP and genome-wide sequencing, we also confirmed that APQ modulates H3K9me3-landscaped epigenomes in YAC128 mice. These data provide that APQ, a novel small molecule SETDB1 inhibitor, coordinates H3K9me-dependent heterochromatin remodelling and can be an epigenetic drug for treating HD, leading with hope in clinical trials of HD.


Disease Models, Animal , Enzyme Inhibitors/pharmacology , Heterochromatin/drug effects , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Huntington Disease/drug therapy , Neurons/drug effects , Animals , Behavior, Animal/drug effects , Biosensing Techniques , Cell Survival/drug effects , Cells, Cultured , Enzyme Inhibitors/chemistry , Fluorescence Resonance Energy Transfer , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Huntington Disease/metabolism , Huntington Disease/pathology , Mice , Mice, Transgenic , Molecular Structure , Neurons/metabolism , Neurons/pathology
4.
J Ethnopharmacol ; 269: 113720, 2021 Apr 06.
Article En | MEDLINE | ID: mdl-33358858

ETHNOPHARMACOLOGICAL RELEVANCE: Jiajian Guishen Formula (JJGSF), which is a prescription of Traditional Chinese Medicine (TCM), has been reported to be useful in the treatment of premature ovarian insufficiency (POI). AIM OF THE STUDY: To investigate the therapeutic effects of JJGSF on the treatment of POI induced by 4-vinylcyclohexene diep-oxide (VCD), an endocrine-disrupting chemical (EDC), and to elucidate the potential mechanism. MATERIALS AND METHODS: Female 8-week-old ICR mice (N = 72) were randomized into six groups, containing the Model group, Control group, three JJGSF groups, and Progynova group which was served as a positive control. After model establishment by VCD, the Progynova group were given a daily intragastric administration of Progynova, and the three JJGSF groups (high dose group, medium dose group and low dose group) received a daily intragastric administration of JJGSF at doses of 9, 4.5 and 2.25 g/kg for four weeks. The general growth of the mice was observed and the estrous cycles were examined. The serum hormone concentrations were measured by enzyme-linked immunosorbent assay (ELISA). To explore the potential mechanism of effect, the protein expressions of H3K9me3, HP1, and HMGA1/HMGA2 related to senescence-associated heterochromatic foci (SAHF), were determined by Immunofluorescence and Western blot analysis, respectively. RESULTS: After treating with JJGSF, the estrous cycles were improved significantly. The level of estrogen (E2) and anti-müllerian hormone (AMH) was increased and the ratio of follicle-stimulating hormone (FSH) to luteinizing hormone (LH) in serum was decreased significantly. Furthermore, a significant down-regulation of HMGA1/HMGA2 on protein level, a reduction distribution of HP1 and H3K9me3 in ovarian, and a lower fraction of SAHF-positive cells were observed after the administration with JJGSF, additionally effects showed a positive correlation with dosages. CONCLUSIONS: JJGSF could treat POI by the mechanism of inhibiting SAHF.


Drugs, Chinese Herbal/pharmacology , Heterochromatin/drug effects , Primary Ovarian Insufficiency/drug therapy , Aging , Animals , Anti-Mullerian Hormone/metabolism , Cellular Senescence/drug effects , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , Cyclohexenes/toxicity , Cytokines/blood , Disease Models, Animal , Down-Regulation/drug effects , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/therapeutic use , Endocrine Disruptors/toxicity , Estradiol/pharmacology , Estradiol/therapeutic use , Estrogens/metabolism , Estrous Cycle/drug effects , Female , Follicle Stimulating Hormone/blood , HMGA1a Protein/genetics , HMGA1a Protein/metabolism , HMGA2 Protein/genetics , HMGA2 Protein/metabolism , Histones/metabolism , Luteinizing Hormone/blood , Medicine, Chinese Traditional , Mice, Inbred ICR , Ovary/drug effects , Ovary/metabolism , Ovary/pathology , Primary Ovarian Insufficiency/chemically induced , Primary Ovarian Insufficiency/genetics , Primary Ovarian Insufficiency/metabolism , Vinyl Compounds/toxicity
5.
Nature ; 585(7825): 453-458, 2020 09.
Article En | MEDLINE | ID: mdl-32908306

Heterochromatin that depends on histone H3 lysine 9 methylation (H3K9me) renders embedded genes transcriptionally silent1-3. In the fission yeast Schizosaccharomyces pombe, H3K9me heterochromatin can be transmitted through cell division provided the counteracting demethylase Epe1 is absent4,5. Heterochromatin heritability might allow wild-type cells under certain conditions to acquire epimutations, which could influence phenotype through unstable gene silencing rather than DNA change6,7. Here we show that heterochromatin-dependent epimutants resistant to caffeine arise in fission yeast grown with threshold levels of caffeine. Isolates with unstable resistance have distinct heterochromatin islands with reduced expression of embedded genes, including some whose mutation confers caffeine resistance. Forced heterochromatin formation at implicated loci confirms that resistance results from heterochromatin-mediated silencing. Our analyses reveal that epigenetic processes promote phenotypic plasticity, letting wild-type cells adapt to unfavourable environments without genetic alteration. In some isolates, subsequent or coincident gene-amplification events augment resistance. Caffeine affects two anti-silencing factors: Epe1 is downregulated, reducing its chromatin association, and a shortened isoform of Mst2 histone acetyltransferase is expressed. Thus, heterochromatin-dependent epimutation provides a bet-hedging strategy allowing cells to adapt transiently to insults while remaining genetically wild type. Isolates with unstable caffeine resistance show cross-resistance to antifungal agents, suggesting that related heterochromatin-dependent processes may contribute to resistance of plant and human fungal pathogens to such agents.


Drug Resistance, Fungal/genetics , Gene Silencing , Heterochromatin/genetics , Heterochromatin/metabolism , Schizosaccharomyces/genetics , Caffeine/pharmacology , Drug Resistance, Fungal/drug effects , Gene Silencing/drug effects , Heterochromatin/drug effects , Histone Acetyltransferases/metabolism , Nuclear Proteins/metabolism , Phenotype , Schizosaccharomyces/cytology , Schizosaccharomyces/drug effects , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism
6.
Sci Rep ; 10(1): 3478, 2020 02 26.
Article En | MEDLINE | ID: mdl-32103104

Heterochromatin is essential for regulating global gene transcription and protecting genome stability, and may play a role in tumor suppression. Drugs promoting heterochromatin are potential cancer therapeutics but very few are known. In order to identify drugs that can promote heterochromatin, we used a cell-based method and screened NCI drug libraries consisting of oncology drugs and natural compounds. Since heterochromatin is originally defined as intensely stained chromatin in the nucleus, we estimated heterochromatin contents of cells treated with different drugs by quantifying the fluorescence intensity of nuclei stained with Hoechst DNA dye. We used HeLa cells and screened 231 FDA-approved oncology and natural substance drugs included in two NCI drug libraries representing a variety of chemical structures. Among these drugs, streptonigrin most prominently caused an increase in Hoechst-stained nuclear fluorescence intensity. We further show that streptonigrin treated cells exhibit compacted DNA foci in the nucleus that co-localize with Heterochromatin Protein 1 alpha (HP1α), and exhibit an increase in total levels of the heterochromatin mark, H3K9me3. Interestingly, we found that streptonigrin promotes heterochromatin at a concentration as low as one nanomolar, and at this concentration there were no detectable effects on cell proliferation or viability. Finally, in line with a previous report, we found that streptonigrin inhibits STAT3 phosphorylation, raising the possibility that non-canonical STAT function may contribute to the effects of streptonigrin on heterochromatin. These results suggest that, at low concentrations, streptonigrin may primarily enhance heterochromatin formation with little toxic effects on cells, and therefore might be a good candidate for epigenetic cancer therapy.


Antibiotics, Antineoplastic/pharmacology , Chromatin Assembly and Disassembly/drug effects , Heterochromatin/physiology , Streptonigrin/pharmacology , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , HeLa Cells , Heterochromatin/drug effects , Histones/metabolism , Humans , Phosphorylation/drug effects , STAT3 Transcription Factor/metabolism
7.
Plant J ; 102(1): 68-84, 2020 04.
Article En | MEDLINE | ID: mdl-31733119

Repetitive DNA sequences and some genes are epigenetically repressed by transcriptional gene silencing (TGS). When genetic mutants are not available or problematic to use, TGS can be suppressed by chemical inhibitors. However, informed use of epigenetic inhibitors is partially hampered by the absence of any systematic comparison. In addition, there is emerging evidence that epigenetic inhibitors cause genomic instability, but the nature of this damage and its repair remain unclear. To bridge these gaps, we compared the effects of 5-azacytidine (AC), 2'-deoxy-5-azacytidine (DAC), zebularine and 3-deazaneplanocin A (DZNep) on TGS and DNA damage repair. The most effective inhibitor of TGS was DAC, followed by DZNep, zebularine and AC. We confirmed that all inhibitors induce DNA damage and suggest that this damage is repaired by multiple pathways with a critical role of homologous recombination and of the SMC5/6 complex. A strong positive link between the degree of cytidine analog-induced DNA demethylation and the amount of DNA damage suggests that DNA damage is an integral part of cytidine analog-induced DNA demethylation. This helps us to understand the function of DNA methylation in plants and opens the possibility of using epigenetic inhibitors in biotechnology.


DNA Damage , Epigenesis, Genetic , Gene Silencing , Adenosine/analogs & derivatives , Adenosine/pharmacology , Arabidopsis/genetics , Azacitidine/pharmacology , Chromosome Aberrations/drug effects , Cytidine/analogs & derivatives , Cytidine/pharmacology , DNA Damage/drug effects , DNA Methylation/drug effects , DNA Repair/drug effects , Decitabine/pharmacology , Epigenesis, Genetic/drug effects , Gene Silencing/drug effects , Heterochromatin/drug effects , RNA Interference/drug effects , Tandem Repeat Sequences/drug effects
8.
Sci Rep ; 9(1): 11673, 2019 08 12.
Article En | MEDLINE | ID: mdl-31406262

Heterochromatin is a tightly packed form of DNA involved in gene silencing, chromosome segregation, and protection of genome stability. Heterochromatin is becoming more recognized in tumor suppression and may thus serve as a potential target for cancer therapy. However, to date there are no drugs that are well established to specifically promote heterochromatin formation. Here, we describe a screening method using Drosophila to identify small molecule compounds that promote heterochromatin formation, with the purpose of developing epigenetic cancer therapeutics. We took advantage of a Drosophila strain with a variegated eye color phenotype that is sensitive to heterochromatin levels, and screened a library of 97 FDA approved oncology drugs. This screen identified methotrexate as the most potent small molecule drug, among the 97 oncology drugs screened, in promoting heterochromatin formation. Interestingly, methotrexate has been identified as a JAK/STAT inhibitor in a functional screen, causing reduced phosphorylation of STAT proteins. These findings are in line with our previous observation that unphosphorylated STAT (uSTAT) promotes heterochromatin formation in both Drosophila and human cells and suppresses tumor growth in mouse xenografts. Thus, Drosophila with variegated eye color phenotypes could be an effective tool for screening heterochromatin-promoting compounds that could be candidates as cancer therapeutics.


Antineoplastic Agents/pharmacology , Drosophila melanogaster/drug effects , Epigenesis, Genetic , Heterochromatin/drug effects , Methotrexate/pharmacology , Small Molecule Libraries/pharmacology , Animals , Animals, Genetically Modified , Chromatin Assembly and Disassembly/drug effects , Color , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Eye/anatomy & histology , Eye/cytology , Eye/drug effects , Eye/metabolism , Female , Genetic Variation , Genomic Instability , Heterochromatin/chemistry , High-Throughput Screening Assays , Histones/genetics , Histones/metabolism , Humans , Janus Kinases/antagonists & inhibitors , Janus Kinases/genetics , Janus Kinases/metabolism , Male , Models, Biological , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation/drug effects , Pigmentation/drug effects , Pigmentation/genetics , STAT Transcription Factors/antagonists & inhibitors , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism
9.
SLAS Discov ; 24(8): 802-816, 2019 09.
Article En | MEDLINE | ID: mdl-31145866

Heterochromatin protein 1 (HP1) facilitates the formation of repressive heterochromatin domains by recruiting histone lysine methyltransferase enzymes to chromatin, resulting in increased levels of histone H3K9me3. To identify chemical inhibitors of the HP1-heterochromatin gene repression pathway, we combined a cell-based assay that utilized chemical-mediated recruitment of HP1 to an endogenous active gene with high-throughput flow cytometry. Here we characterized small molecule inhibitors that block HP1-mediated heterochromatin formation. Our lead compounds demonstrated dose-dependent inhibition of HP1-stimulated gene repression and were validated in an orthogonal cell-based system. One lead inhibitor was improved by a change in stereochemistry, resulting in compound 2, which was further used to decouple the inverse relationship between H3K9 and H3K4 methylation states. We identified molecular components that bound compound 2, either directly or indirectly, by chemical affinity purification with a biotin-tagged derivative, followed by quantitative proteomic techniques. In summary, our pathway-based chemical screening approach resulted in the discovery of new inhibitors of HP1-mediated heterochromatin formation while identifying exciting new molecular interactions in the pathway to explore in the future. This modular platform can be expanded to test a wide range of chromatin modification pathways yielding inhibitors that are cell permeable and function in a physiologically relevant setting.


Chromatin Assembly and Disassembly/drug effects , Chromosomal Proteins, Non-Histone/metabolism , Drug Discovery , Heterochromatin/drug effects , Heterochromatin/metabolism , High-Throughput Screening Assays , Signal Transduction/drug effects , Animals , Cell Line , Chromatography, Liquid , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/chemistry , Drug Discovery/methods , Flow Cytometry , Heterochromatin/genetics , Histones/metabolism , Mice , Microscopy, Fluorescence , Models, Biological , Molecular Structure , Phosphoproteins/metabolism , Protein Binding , Proteomics/methods , Small Molecule Libraries , Structure-Activity Relationship , Tandem Mass Spectrometry
10.
Clin Exp Immunol ; 196(1): 67-75, 2019 04.
Article En | MEDLINE | ID: mdl-30714144

Epigenetic effects of anti-psychotic medications are poorly understood. We have appropriated a model whereby heterochromatin is established through 24- or 48-h lipopolysaccharide (LPS) treatment, and tested the epigenetic effects of risperidone along the adenylyl cyclase/protein kinase A (AC/PKA) pathway in human liposarcoma cells that express the LPS-sensitive Toll-like receptor (TLR)-4. Human SW872 cells were cultured with LPS and mRNA expression levels and epigenetic modifications of dimethylated lysine 9 of histone 2 (H3K9me2), geterochromatin protein 1γ (HP1γ) and phospho-H3S10 at promoters of interleukin (IL)-6, tumor necrosis factor (TNF)-α and IL1ß were measured. Pharmacological manipulation of the AC/PKA pathway was achieved through treatment with a PKA inhibitor (H89), mitogen- and stress-activated kinase 1 (MSK1) inhibitor (SB-747651A) or forskolin. Twenty-four and 48-h LPS treatment establishes heterochromatin at selected promoters, corresponding to decreased mRNA expression. Concurrent risperidone treatment with LPS treatment can both 'block' and 'reverse' heterochromatin formation. Forskolin treatment resulted in a similar disassembling effect on heterochromatin. Conversely, inhibition of PKA by H89 or MSK1 both blocked 'normalizing' effects of risperidone on LPS-induced heterochromatin. Our results demonstrate that risperidone can disassemble heterochromatin, exerting this effect along the G-protein/AC/PKA pathway. This approach can also be utilized to investigate functional outcomes of single or combined pharmacological treatments on chromatin assemblies in human cells.


Antipsychotic Agents/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Heterochromatin/drug effects , Liposarcoma/drug therapy , Risperidone/pharmacology , Adenylyl Cyclases/metabolism , Animals , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cytokines/genetics , Cytokines/metabolism , Epigenesis, Genetic/drug effects , GTP-Binding Proteins/metabolism , Histones/metabolism , Humans , Inflammation Mediators/metabolism , Isoquinolines/pharmacology , Lipopolysaccharides/immunology , Liposarcoma/genetics , Promoter Regions, Genetic/genetics , Signal Transduction , Sulfonamides/pharmacology , Toll-Like Receptor 4/metabolism
11.
J Cell Biol ; 218(1): 134-149, 2019 01 07.
Article En | MEDLINE | ID: mdl-30396998

The centromere is an important genomic locus for chromosomal segregation. Although the centromere is specified by sequence-independent epigenetic mechanisms in most organisms, it is usually composed of highly repetitive sequences, which associate with heterochromatin. We have previously generated various chicken DT40 cell lines containing differently positioned neocentromeres, which do not contain repetitive sequences and do not associate with heterochromatin. In this study, we performed systematic 4C analysis using three cell lines containing differently positioned neocentromeres to identify neocentromere-associated regions at the 3D level. This analysis reveals that these neocentromeres commonly associate with specific heterochromatin-rich regions, which were distantly located from neocentromeres. In addition, we demonstrate that centromeric chromatin adopts a compact structure, and centromere clustering also occurs in vertebrate interphase nuclei. Interestingly, the occurrence of centromere-heterochromatin associations depend on CENP-H, but not CENP-C. Our analyses provide an insight into understanding the 3D architecture of the genome, including the centromeres.


Centromere/ultrastructure , Chromosomal Proteins, Non-Histone/genetics , Epigenesis, Genetic , Genome , Heterochromatin/ultrastructure , Animals , Cell Line, Tumor , Centromere/drug effects , Centromere/metabolism , Chickens , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation/drug effects , Flow Cytometry , Heterochromatin/drug effects , Heterochromatin/metabolism , Indoleacetic Acids/pharmacology , Lymphocytes/drug effects , Lymphocytes/metabolism , Lymphocytes/ultrastructure , Methyltransferases/genetics , Methyltransferases/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism
12.
Article En | MEDLINE | ID: mdl-30249477

Many DNA damaging agents also react with RNA and protein, and could thus cause epigenetic as well as genotoxic changes. To investigate which DNA damaging agents alter epigenetic states, we studied the chemical-induced changes in expression of the yeast silent mating type locus HMLα, which can be triggered by inhibiting yeast Sir2. We observed that the alkylating agent methyl methane sulfonate (MMS) can result in HMLα expression, using a colony sector assay that results from expression of a HML-positioned cre gene. Using single-cell imaging we also observed that alkylating agents, including MMS and methyl-3-nitro-1-nitrosoguanidine (MNNG), as well as short-wave UV, also decreased HML silencing. We suggest that chemical-induced alterations in heterochromatin structure could confer transient phenotypic changes that affect the cellular responses to DNA damaging agents.


DNA, Fungal/genetics , Gene Expression Regulation, Fungal/drug effects , Gene Silencing , Genes, Mating Type, Fungal , Heterochromatin/pathology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Sulfinic Acids/pharmacology , Heterochromatin/drug effects , Heterochromatin/genetics , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/genetics
13.
Int J Radiat Oncol Biol Phys ; 100(1): 174-187, 2018 01 01.
Article En | MEDLINE | ID: mdl-29107335

PURPOSE: We previously reported that sphere-forming non-small cell lung cancer (NSCLC) tumor-initiating cells (TICs) have an altered activation of DNA damage response- and repair proteins and are refractory to DNA-damaging treatments. We analyzed whether chromatin organization plays a role in the observed refractoriness. METHODS AND MATERIALS: Bulk cells and TICs from the NSCLC H23 and H1299 cell lines were examined using cell viability, clonogenic survival, Western blot, short interfering RNA analysis, and micronucleus assay. RESULTS: NSCLC TICs displayed elevated heterochromatin markers trimethylated lysine 9 of histone H3 and heterochromatin protein 1γ relative to bulk cells and reduced cell viability upon histone deacetylase inhibition (HDACi). Vorinostat and trichostatin A increased the euchromatin markers acetylated lysine 9/14 of histone H3 and lysine 8 of histone H4, and HDACi pretreatment increased the phosphorylation of the DNA damage response proteins ataxia telangiectasia mutated and DNA-dependent protein kinase, catalytic subunit, upon irradiation in TICs. HDACi sensitized TICs to cisplatin and to some extent to ionizing irradiation. The protectiveness of a dense chromatin structure was indicated by an enhanced frequency of micronuclei in TICs following irradiation, after knockdown of heterochromatin protein 1γ. CONCLUSIONS: Although confirmatory studies in additional NSCLC model systems and with respect to analyses of other DNA damage response proteins are needed, our data point toward a heterochromatic structure of NSCLC TICs, such that HDACi can sensitize TICs to DNA damage.


Carcinoma, Non-Small-Cell Lung/pathology , Chromatin/drug effects , DNA Damage , Histone Deacetylase Inhibitors/pharmacology , Lung Neoplasms/pathology , Neoplastic Stem Cells/drug effects , AC133 Antigen/metabolism , Antineoplastic Agents/pharmacology , Blotting, Western , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Cell Line, Tumor , Cell Survival , Chromatin/chemistry , Chromosomal Proteins, Non-Histone/genetics , Cisplatin/pharmacology , Heterochromatin/chemistry , Heterochromatin/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Micronucleus Tests , Nanog Homeobox Protein/metabolism , Neoplastic Stem Cells/chemistry , Neoplastic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Phosphorylation , RNA, Small Interfering/analysis , SOXB1 Transcription Factors/metabolism
14.
Mol Biol Cell ; 29(2): 220-233, 2018 01 15.
Article En | MEDLINE | ID: mdl-29142071

Nuclear shape and architecture influence gene localization, mechanotransduction, transcription, and cell function. Abnormal nuclear morphology and protrusions termed "blebs" are diagnostic markers for many human afflictions including heart disease, aging, progeria, and cancer. Nuclear blebs are associated with both lamin and chromatin alterations. A number of prior studies suggest that lamins dictate nuclear morphology, but the contributions of altered chromatin compaction remain unclear. We show that chromatin histone modification state dictates nuclear rigidity, and modulating it is sufficient to both induce and suppress nuclear blebs. Treatment of mammalian cells with histone deacetylase inhibitors to increase euchromatin or histone methyltransferase inhibitors to decrease heterochromatin results in a softer nucleus and nuclear blebbing, without perturbing lamins. Conversely, treatment with histone demethylase inhibitors increases heterochromatin and chromatin nuclear rigidity, which results in reduced nuclear blebbing in lamin B1 null nuclei. Notably, increased heterochromatin also rescues nuclear morphology in a model cell line for the accelerated aging disease Hutchinson-Gilford progeria syndrome caused by mutant lamin A, as well as cells from patients with the disease. Thus, chromatin histone modification state is a major determinant of nuclear blebbing and morphology via its contribution to nuclear rigidity.


Heterochromatin/metabolism , Histones/metabolism , Lamins/metabolism , Nuclear Envelope/ultrastructure , Animals , Cells, Cultured , HeLa Cells , Heterochromatin/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Lamins/genetics , Mechanotransduction, Cellular/drug effects , Mice , Nuclear Envelope/drug effects , Progeria/genetics , Protein Processing, Post-Translational
15.
Inorg Chem ; 56(21): 13491-13499, 2017 Nov 06.
Article En | MEDLINE | ID: mdl-28990789

Octahedral rhenium cluster complexes have recently emerged as relevant building blocks for the design of singlet oxygen photosensitizing materials toward biological applications such as blue-light photodynamic therapy. However, their singlet oxygen generation ability as well as biological properties have been studied only superficially. Herein we investigate in detail the singlet oxygen photogeneration, dark and photoinduced cytotoxicity, cellular uptake kinetics, cellular localization and in vitro photoinduced oxidative stress, and photodynamic cytotoxicity of the series of octahedral rhenium cluster complexes [{Re6Q8}(CN)6]4-, where Q = S, Se, Te. Our results demonstrate that the selenium-containing complex possesses optimal properties in terms of absorption and singlet oxygen productivity. These features coupled with the cellular internalization and low dark toxicity lead to the first photoinduced cytotoxic effect observed for a molecular [{M6Q8}L6] complex, making it a promising object for further study in terms of blue-light PDT.


Contrast Media/pharmacology , Coordination Complexes/pharmacology , Photosensitizing Agents/pharmacology , Rhenium/chemistry , Singlet Oxygen/chemistry , Cell Line, Tumor , Contrast Media/chemical synthesis , Contrast Media/radiation effects , Contrast Media/toxicity , Coordination Complexes/chemical synthesis , Coordination Complexes/radiation effects , Coordination Complexes/toxicity , Heterochromatin/drug effects , Humans , Ligands , Light , Luminescence , Mitochondria/drug effects , Oxidative Stress/drug effects , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/radiation effects , Photosensitizing Agents/toxicity
16.
PLoS Genet ; 13(7): e1006900, 2017 Jul.
Article En | MEDLINE | ID: mdl-28749973

The fidelity of epigenetic inheritance or, the precision by which epigenetic information is passed along, is an essential parameter for measuring the effectiveness of the process. How the precision of the process is achieved or modulated, however, remains largely elusive. We have performed quantitative measurement of epigenetic fidelity, using position effect variegation (PEV) in Schizosaccharomyces pombe as readout, to explore whether replication perturbation affects nucleosome-mediated epigenetic inheritance. We show that replication stresses, due to either hydroxyurea treatment or various forms of genetic lesions of the replication machinery, reduce the inheritance accuracy of CENP-A/Cnp1 nucleosome positioning within centromere. Mechanistically, we demonstrate that excessive formation of single-stranded DNA, a common molecular abnormality under these conditions, might have correlation with the reduction in fidelity of centromeric chromatin duplication. Furthermore, we show that replication stress broadly changes chromatin structure at various loci in the genome, such as telomere heterochromatin expanding and mating type locus heterochromatin spreading out of the boundaries. Interestingly, the levels of inheritable expanding at sub-telomeric heterochromatin regions are highly variable among independent cell populations. Finally, we show that HU treatment of the multi-cellular organisms C. elegans and D. melanogaster affects epigenetically programmed development and PEV, illustrating the evolutionary conservation of the phenomenon. Replication stress, in addition to its demonstrated role in genetic instability, promotes variable epigenetic instability throughout the epigenome.


Chromosomal Position Effects/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA Replication/genetics , Epigenesis, Genetic/genetics , Schizosaccharomyces pombe Proteins/genetics , Animals , Caenorhabditis elegans/genetics , Centromere/genetics , Chromatin/drug effects , Chromatin/genetics , DNA, Single-Stranded/drug effects , Drosophila melanogaster/genetics , Epigenesis, Genetic/drug effects , Heterochromatin/drug effects , Heterochromatin/genetics , Histones/genetics , Hydroxyurea/pharmacology , Nucleosomes/genetics , Schizosaccharomyces/genetics
17.
Nucleus ; 8(2): 205-221, 2017 03 04.
Article En | MEDLINE | ID: mdl-28068183

The role of the nucleolus and autophagy in maintenance of nuclear integrity is poorly understood. In addition, the mechanisms of nuclear destruction in cancer cells senesced after conventional chemotherapy are unclear. In an attempt to elucidate these issues, we studied teratocarcinoma PA1 cells treated with Etoposide (ETO), focusing on the nucleolus. Following treatment, most cells enter G2 arrest, display persistent DNA damage and activate p53, senescence, and macroautophagy markers. 2-5 µm sized nucleolar aggresomes (NoA) containing fibrillarin (FIB) and damaged rDNA, colocalized with ubiquitin, pAMPK, and LC3-II emerge, accompanied by heterochromatin fragments, when translocated perinuclearly. Microscopic counts following application of specific inhibitors revealed that formation of FIB-NoA is dependent on deficiency of the ubiquitin proteasome system coupled to functional autophagy. In contrast, the accompanying NoAs release of pericentric heterochromatin, which exceeds their frequency, is favored by debilitation of autophagic flux. Potential survivors release NoA in the cytoplasm during rare mitoses, while exit of pericentric fragments often depleted of H3K9Me3, with or without encompassing by NoA, occurs through the nucleolar protrusions and defects of the nuclear envelope. Foci of LC3-II are accumulated in the nucleoli undergoing cessation of rDNA transcription. As an origin of heterochromatin fragmentation, the unscheduled DNA synthesis and circular DNAs were found in the perinucleolar heterochromatin shell, along with activation and retrotransposition of ALU elements, colocalized with 45S rDNA in NoAs. The data indicate coordination of the basic nucleolar function with autophagy regulation in maintenance of the integrity of the nucleolus associated domains secured by inactivity of retrotransposons.


Cell Nucleolus/drug effects , Cell Nucleolus/metabolism , Heterochromatin/drug effects , Heterochromatin/metabolism , Mutagens/toxicity , Autophagy/drug effects , Autophagy/genetics , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Nucleolus/genetics , Cellular Senescence/drug effects , Cellular Senescence/genetics , Chromosomal Proteins, Non-Histone/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Damage , DNA, Ribosomal/genetics , DNA, Ribosomal/metabolism , Etoposide/toxicity , Humans , Retroelements/drug effects , Retroelements/genetics
18.
Acta Histochem ; 119(1): 18-25, 2017 Jan.
Article En | MEDLINE | ID: mdl-27863708

The nucleus of mammalian embryos differs by transcriptional activity at different stages of early development. Here, we studied nuclear distribution of the chromatin-remodeling protein ATRX in pre-implantation mouse embryos. Immunofluorescent staining revealed the changes of ATRX nuclear distribution at the initial stages of early mouse development. At the stage of early zygote, a diffuse ATRX distribution pattern was prevalent. During the course of zygotic genome activation (ZGA), zones of increased ATRX concentration are observed, and they are most expressed in the nuclei of late 2-cell embryos. In the morula stage, the ATRX distribution becomes diffuse again. In zygotes, the patterns of ATRX distribution differ between male and female pronuclei. At all the stages, ATRX concentrates in the DAPI-positive areas of condensed chromatin. The level of colocalization between ATRX and heterochromatin was found the highest at the late 2-cell stage. When transcription was artificially suppressed, the pattern of intranuclear ATRX distribution was mostly determined by the mechanism of inhibitor action rather than the decreased level of transcriptional activity. Thus, the obvious changes of ATRX distribution occur and partially correlate with the main stages of ZGA during mouse early development, but these changes seem to be determined by other processes of structural and functional rearrangements of blastomere nuclei.


Chromatin Assembly and Disassembly , DNA Helicases/genetics , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Nuclear Proteins/genetics , Oocytes/metabolism , Zygote/metabolism , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , DNA Helicases/antagonists & inhibitors , DNA Helicases/metabolism , Dichlororibofuranosylbenzimidazole/pharmacology , Embryo, Mammalian , Embryonic Development/drug effects , Female , Heterochromatin/chemistry , Heterochromatin/drug effects , Heterochromatin/metabolism , Male , Mice , Mice, Inbred BALB C , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Nucleic Acid Synthesis Inhibitors/pharmacology , Oocytes/drug effects , Oocytes/ultrastructure , Transcriptional Activation , X-linked Nuclear Protein , Zygote/drug effects , Zygote/ultrastructure
19.
Biochem Pharmacol ; 121: 18-32, 2016 Dec 01.
Article En | MEDLINE | ID: mdl-27659811

Posttranslational modifications of histone tails can alter chromatin structure and regulate gene transcription. While recent studies implicate the lysine/arginine protein methyltransferases in the regulation of genes for endothelial metabolism, the role of AMI-1 and AMI-5 compounds in angiogenesis remains unknown. Here, we show that global inhibition of arginine and lysine histone methyltransferases (HMTs) by AMI-5 induced an angiostatic profile in human microvascular endothelial cells and human umbilical vein endothelial cells. Based on FACS analysis, we found that inhibition of HMTs significantly affects proliferation of endothelial cells, by suppressing cell cycle progression in the G0/G1 phase. Immunofluorescent studies of the endothelial cells replication pattern by 5-ethynyl-2'-deoxyuridine incorporation disclosed that AMI-5, and the arginine methyltransferase inhibitor AMI-1, induced heterochromatin formation and a number of nuclear abnormalities, such as formation of micronuclei (MNs) and nucleoplasmic bridges (NPBs), which are markers of chromosomal instability. In addition to the modification of the cell cycle machinery in response to AMIs treatment, also endothelial cells migration and capillary-like tube formation processes were significantly inhibited, implicating a stimulatory role of HMTs in angiogenesis.


Angiogenesis Inhibitors/pharmacology , Benzoates/pharmacology , Endothelial Cells/drug effects , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Micronuclei, Chromosome-Defective/drug effects , Naphthalenesulfonates/pharmacology , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Urea/analogs & derivatives , Xanthenes/pharmacology , Cell Cycle/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Endothelial Cells/enzymology , Heterochromatin/drug effects , Heterochromatin/pathology , Humans , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/prevention & control , Urea/pharmacology
20.
Cell Death Dis ; 7: e2220, 2016 05 12.
Article En | MEDLINE | ID: mdl-27171262

Expansive growth of neural progenitor cells (NPCs) is a prerequisite to the temporal waves of neuronal differentiation that generate the six-layered neocortex, while also placing a heavy burden on proteins that regulate chromatin packaging and genome integrity. This problem is further reflected by the growing number of developmental disorders caused by mutations in chromatin regulators. ATRX gene mutations cause a severe intellectual disability disorder (α-thalassemia mental retardation X-linked (ATRX) syndrome; OMIM no. 301040), characterized by microcephaly, urogenital abnormalities and α-thalassemia. Although the ATRX protein is required for the maintenance of repetitive DNA within heterochromatin, how this translates to disease pathogenesis remain poorly understood and was a focus of this study. We demonstrate that Atrx(FoxG1Cre) forebrain-specific conditional knockout mice display poly(ADP-ribose) polymerase-1 (Parp-1) hyperactivation during neurogenesis and generate fewer late-born Cux1- and Brn2-positive neurons that accounts for the reduced cortical size. Moreover, DNA damage, induced Parp-1 and Atm activation is elevated in progenitor cells and contributes to their increased level of cell death. ATRX-null HeLa cells are similarly sensitive to hydroxyurea-induced replication stress, accumulate DNA damage and proliferate poorly. Impaired BRCA1-RAD51 colocalization and PARP-1 hyperactivation indicated that stalled replication forks are not efficiently protected. DNA fiber assays confirmed that MRE11 degradation of stalled replication forks was rampant in the absence of ATRX or DAXX. Indeed, fork degradation in ATRX-null cells could be attenuated by treatment with the MRE11 inhibitor mirin, or exacerbated by inhibiting PARP-1 activity. Taken together, these results suggest that ATRX is required to limit replication stress during cellular proliferation, whereas upregulation of PARP-1 activity functions as a compensatory mechanism to protect stalled forks, limiting genomic damage, and facilitating late-born neuron production.


DNA Helicases/genetics , DNA Replication , Heterochromatin/chemistry , Neurons/metabolism , Nuclear Proteins/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA1 Protein , Carrier Proteins/genetics , Cell Proliferation/drug effects , Co-Repressor Proteins , DNA/genetics , DNA/metabolism , DNA Damage , DNA Helicases/deficiency , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , HeLa Cells , Heterochromatin/drug effects , Heterochromatin/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hydroxyurea/pharmacology , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , MRE11 Homologue Protein , Mice , Mice, Knockout , Molecular Chaperones , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurogenesis/genetics , Neurons/cytology , Neurons/drug effects , Nuclear Proteins/deficiency , Nuclear Proteins/metabolism , POU Domain Factors/genetics , POU Domain Factors/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Prosencephalon/cytology , Prosencephalon/drug effects , Prosencephalon/metabolism , Rad51 Recombinase/genetics , Rad51 Recombinase/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , X-linked Nuclear Protein
...