Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
1.
J Mol Biol ; 434(7): 167500, 2022 04 15.
Article in English | MEDLINE | ID: mdl-35183557

ABSTRACT

Histone lysine methylation is a key epigenetic modification that regulates eukaryotic transcription. In Saccharomyces cerevisiae, it is controlled by a reduced but evolutionarily conserved suite of methyltransferase (Set1p, Set2p, Dot1p, and Set5p) and demethylase (Jhd1p, Jhd2p, Rph1p, and Gis1p) enzymes. Many of these enzymes are extensively phosphorylated in vivo; however, the functions of almost all phosphosites remain unknown. Here, we comprehensively analyse the phosphoregulation of the yeast histone methylation network by functionally investigating 40 phosphosites on six enzymes. A total of 82 genomically-edited S. cerevisiae strains were generated through mutagenesis of sites to aspartate as a phosphomimetic or alanine as a phosphonull. These phosphosite mutants were screened for changes in native H3K4, H3K36, and H3K79 methylation levels, and for sensitivity to environmental stress conditions. For methyltransferase Set2p, we found that phosphorylation at threonine 127 significantly decreased H3K36 methylation in vivo, and that an N-terminal phosphorylation cluster at serine residues 6, 8, and 10 is required for the diamide stress response. Proteomic analysis of Set2p phosphosite mutants revealed a specific downregulation of membrane-associated proteins and processes, consistent with changes brought about by SET2 deletion and the sensitivity of mutants to diamide. For demethylase Jhd1p, we found that its sole phosphorylation site at serine 44 is required for the cold stress response. This study represents the first systematic investigation into the phosphoregulation of the epigenetic network in any eukaryote, and shows that phosphosites on histone methylation enzymes are required for a normal cellular response to stress in S.cerevisiae.


Subject(s)
Histone Methyltransferases , Jumonji Domain-Containing Histone Demethylases , Methyltransferases , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Stress, Physiological , Diamide/pharmacology , Histone Methyltransferases/genetics , Histone Methyltransferases/physiology , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/physiology , Methyltransferases/genetics , Methyltransferases/physiology , Phosphorylation , Proteomics , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/physiology , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/physiology , Serine/metabolism
2.
Theranostics ; 11(16): 7779-7796, 2021.
Article in English | MEDLINE | ID: mdl-34335964

ABSTRACT

Rationale: The progression of prostate cancer (PCa) to castration-resistant PCa (CRPC) despite continuous androgen deprivation therapy is a major clinical challenge. Over 90% of patients with CRPC exhibit sustained androgen receptor (AR) signaling. KDM4B that removes the repressive mark H3K9me3/2 is a transcriptional activator of AR and has been implicated in the development of CRPC. However, the mechanisms of KDM4B involvement in CRPC remain largely unknown. Here, we sought to demonstrate the molecular pathway mediated by KDM4B in CRPC and to provide proof-of-concept evidence that KDM4B is a potential CRPC target. Methods: CRPC cells (C4-2B or CWR22Rv1) depleted with KDM4B followed by cell proliferation (in vitro and xenograft), microarray, qRT-PCR, Seahorse Flux, and metabolomic analyses were employed to identify the expression and metabolic profiles mediated by KDM4B. Immunoprecipitation was used to determine the KDM4B-c-Myc interaction region. Reporter activity assay and ChIP analysis were used to characterize the KDM4B-c-Myc complex-mediated mechanistic actions. The clinical relevance between KDM4B and c-Myc was determined using UCSC Xena analysis and immunohistochemistry. Results: We showed that KDM4B knockdown impaired CRPC proliferation, switched Warburg to OXPHOS metabolism, and suppressed gene expressions including those targeted by c-Myc. We further demonstrated that KDM4B physically interacted with c-Myc and they were co-recruited to the c-Myc-binding sequence on the promoters of metabolic genes (LDHA, ENO1, and PFK). Importantly, KDM4B and c-Myc synergistically promoted the transactivation of the LDHA promoter in a demethylase-dependent manner. We also provided evidence that KDM4B and c-Myc are co-expressed in PCa tissue and that high expression of both is associated with worse clinical outcome. Conclusions: KDM4B partners with c-Myc and serves as a coactivator of c-Myc to directly enhance c-Myc-mediated metabolism, hence promoting CRPC progression. Targeting KDM4B is thus an alternative therapeutic strategy for advanced prostate cancers driven by c-Myc and AR.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Androgen Antagonists , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Jumonji Domain-Containing Histone Demethylases/physiology , Male , Mice, Inbred BALB C , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/metabolism , Signal Transduction/genetics , Transcription Factors/metabolism
3.
Cancer Res ; 81(4): 1087-1100, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33822745

ABSTRACT

Endocrine resistance (EnR) in advanced prostate cancer is fatal. EnR can be mediated by androgen receptor (AR) splice variants, with AR splice variant 7 (AR-V7) arguably the most clinically important variant. In this study, we determined proteins key to generating AR-V7, validated our findings using clinical samples, and studied splicing regulatory mechanisms in prostate cancer models. Triangulation studies identified JMJD6 as a key regulator of AR-V7, as evidenced by its upregulation with in vitro EnR, its downregulation alongside AR-V7 by bromodomain inhibition, and its identification as a top hit of a targeted siRNA screen of spliceosome-related genes. JMJD6 protein levels increased (P < 0.001) with castration resistance and were associated with higher AR-V7 levels and shorter survival (P = 0.048). JMJD6 knockdown reduced prostate cancer cell growth, AR-V7 levels, and recruitment of U2AF65 to AR pre-mRNA. Mutagenesis studies suggested that JMJD6 activity is key to the generation of AR-V7, with the catalytic machinery residing within a druggable pocket. Taken together, these data highlight the relationship between JMJD6 and AR-V7 in advanced prostate cancer and support further evaluation of JMJD6 as a therapeutic target in this disease. SIGNIFICANCE: This study identifies JMJD6 as being critical for the generation of AR-V7 in prostate cancer, where it may serve as a tractable target for therapeutic intervention.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/physiology , Prostatic Neoplasms, Castration-Resistant/genetics , Receptors, Androgen/genetics , Alternative Splicing , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cohort Studies , Enzyme Inhibitors/therapeutic use , Gene Expression Regulation, Neoplastic , Humans , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/genetics , Male , Molecular Targeted Therapy , Oxygenases/genetics , Oxygenases/physiology , Prognosis , Prostatic Neoplasms, Castration-Resistant/diagnosis , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/mortality , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Retrospective Studies
4.
Blood ; 137(5): 610-623, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33538795

ABSTRACT

This study was conducted to determine the dosage effect of c-Myc on hematopoiesis and its distinct role in mediating the Wnt/ß-catenin pathway in hematopoietic stem cell (HSC) and bone marrow niche cells. c-Myc haploinsufficiency led to ineffective hematopoiesis by inhibiting HSC self-renewal and quiescence and by promoting apoptosis. We have identified Nr4a1, Nr4a2, and Jmjd3, which are critical for the maintenance of HSC functions, as previously unrecognized downstream targets of c-Myc in HSCs. c-Myc directly binds to the promoter regions of Nr4a1, Nr4a2, and Jmjd3 and regulates their expression. Our results revealed that Nr4a1 and Nr4a2 mediates the function of c-Myc in regulating HSC quiescence, whereas all 3 genes contribute to the function of c-Myc in the maintenance of HSC survival. Adenomatous polyposis coli (Apc) is a negative regulator of the Wnt/ß-catenin pathway. We have provided the first evidence that Apc haploinsufficiency induces a blockage of erythroid lineage differentiation through promoting secretion of IL6 in bone marrow endothelial cells. We found that c-Myc haploinsufficiency failed to rescue defective function of Apc-deficient HSCs in vivo but it was sufficient to prevent the development of severe anemia in Apc-heterozygous mice and to significantly prolong the survival of those mice. Furthermore, we showed that c-Myc-mediated Apc loss induced IL6 secretion in endothelial cells, and c-Myc haploinsufficiency reversed the negative effect of Apc-deficient endothelial cells on erythroid cell differentiation. Our studies indicate that c-Myc has a context-dependent role in mediating the function of Apc in hematopoiesis.


Subject(s)
Genes, myc , Hematopoiesis/physiology , Proto-Oncogene Proteins c-myb/physiology , Adenomatous Polyposis Coli Protein/physiology , Anemia/genetics , Anemia/prevention & control , Animals , Apoptosis/physiology , Bone Marrow Transplantation , Cell Self Renewal/physiology , Colony-Forming Units Assay , Endothelial Cells/pathology , Erythroid Cells/pathology , Gene Deletion , Genes, APC , Haploinsufficiency , Hematopoiesis/genetics , Hematopoietic Stem Cells , Interleukin-6/physiology , Jumonji Domain-Containing Histone Demethylases/physiology , Mice, Mutant Strains , Nuclear Receptor Subfamily 4, Group A, Member 1/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/physiology , Poly I-C/pharmacology , Radiation Chimera , Wnt Signaling Pathway/physiology
5.
Int J Biol Sci ; 17(2): 527-538, 2021.
Article in English | MEDLINE | ID: mdl-33613110

ABSTRACT

Germline specification is a fundamental step for human reproduction and this biological phenomenon possesses technical challenges to study in vivo as it occurs immediately after blastocyst implantation. The establishment of in vitro human primordial germ cell-like cells (hPGCLCs) induction system allows sophisticated characterization of human primordial germ cells (hPGCs) development. However, the underlying molecular mechanisms of hPGCLC specification are not fully elucidated. Here, we observed particularly high expression of the histone demethylase KDM2B in male fetal germ cells (FGCs) but not in male somatic cells. Besides, KDM2B shared similar expression pattern with hPGC marker genes in hPGCLCs, suggesting an important role of KDM2B in germ cell development. Although deletion of KDM2B had no significant effects on human embryonic stem cell (hESC)'s pluripotency, loss of KDM2B dramatically impaired hPGCLCs differentiation whereas ectopically expressed KDM2B could efficiently rescue such defect, indicating this defect was due to KDM2B's loss in hPGCLC specification. Mechanistically, as revealed by the transcriptional profiling, KDM2B suppressed the expression of somatic genes thus inhibited somatic differentiation during hPGCLC specification. These data collectively indicate that KDM2B is an indispensable epigenetic regulator for hPGCLC specification, shedding lights on how epigenetic regulations orchestrate transcriptional events in hPGC development for future investigation.


Subject(s)
Cell Differentiation/physiology , Cell Lineage , F-Box Proteins/physiology , Germ Cells/cytology , Jumonji Domain-Containing Histone Demethylases/physiology , Cells, Cultured , Embryonic Stem Cells/cytology , F-Box Proteins/genetics , Gene Knockdown Techniques , Humans , Jumonji Domain-Containing Histone Demethylases/genetics
6.
Sci Rep ; 11(1): 884, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33441614

ABSTRACT

Trophoblast stem cell (TSC) is crucial to the formation of placenta in mammals. Histone demethylase JMJD2 (also known as KDM4) family proteins have been previously shown to support self-renewal and differentiation of stem cells. However, their roles in the context of the trophoblast lineage remain unclear. Here, we find that knockdown of Jmjd2b resulted in differentiation of TSCs, suggesting an indispensable role of JMJD2B/KDM4B in maintaining the stemness. Through the integration of transcriptome and ChIP-seq profiling data, we show that JMJD2B is associated with a loss of H3K36me3 in a subset of embryonic lineage genes which are marked by H3K9me3 for stable repression. By characterizing the JMJD2B binding motifs and other transcription factor binding datasets, we discover that JMJD2B forms a protein complex with AP-2 family transcription factor TFAP2C and histone demethylase LSD1. The JMJD2B-TFAP2C-LSD1 complex predominantly occupies active gene promoters, whereas the TFAP2C-LSD1 complex is located at putative enhancers, suggesting that these proteins mediate enhancer-promoter interaction for gene regulation. We conclude that JMJD2B is vital to the TSC transcriptional program and safeguards the trophoblast cell fate via distinctive protein interactors and epigenetic targets.


Subject(s)
Epigenesis, Genetic/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Trophoblasts/metabolism , Adult Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Chromatin Immunoprecipitation Sequencing/methods , Epigenomics/methods , Gene Expression , Gene Expression Profiling/methods , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histone Demethylases/physiology , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/physiology , Mice , Mice, 129 Strain , Promoter Regions, Genetic , Stem Cells/metabolism , Transcription Factor AP-2/metabolism , Transcription, Genetic/genetics , Trophoblasts/physiology
7.
Int J Mol Sci ; 22(2)2021 Jan 19.
Article in English | MEDLINE | ID: mdl-33478063

ABSTRACT

Cancer is a major cause of death worldwide. Epigenetic changes in response to external (diet, sports activities, etc.) and internal events are increasingly implicated in tumor initiation and progression. In this review, we focused on post-translational changes in histones and, more particularly, the tri methylation of lysine from histone 3 (H3K27me3) mark, a repressive epigenetic mark often under- or overexpressed in a wide range of cancers. Two actors regulate H3K27 methylation: Jumonji Domain-Containing Protein 3 demethylase (JMJD3) and Enhancer of zeste homolog 2 (EZH2) methyltransferase. A number of studies have highlighted the deregulation of these actors, which is why this scientific review will focus on the role of JMJD3 and, consequently, H3K27me3 in cancer development. Data on JMJD3's involvement in cancer are classified by cancer type: nervous system, prostate, blood, colorectal, breast, lung, liver, ovarian, and gastric cancers.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/physiology , Neoplasms/genetics , Animals , DNA Methylation/genetics , Enhancer of Zeste Homolog 2 Protein/physiology , Epigenesis, Genetic/genetics , Female , Histone Demethylases/physiology , Histones/metabolism , Humans , Male , Neoplasms/metabolism , Neoplasms/pathology
8.
Biochem Soc Trans ; 49(1): 93-105, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33492339

ABSTRACT

Alterations in global epigenetic signatures on chromatin are well established to contribute to tumor initiation and progression. Chromatin methylation status modulates several key cellular processes that maintain the integrity of the genome. KDM4A, a demethylase that belongs to the Fe-II dependent dioxygenase family that uses α-ketoglutarate and molecular oxygen as cofactors, is overexpressed in several cancers and is associated with an overall poor prognosis. KDM4A demethylates lysine 9 (H3K9me2/3) and lysine 36 (H3K36me3) methyl marks on histone H3. Given the complexity that exists with these marks on chromatin and their effects on transcription and proliferation, it naturally follows that demethylation serves an equally important role in these cellular processes. In this review, we highlight the role of KDM4A in transcriptional modulation, either dependent or independent of its enzymatic activity, arising from the amplification of this demethylase in cancer. KDM4A modulates re-replication of distinct genomic loci, activates cell cycle inducers, and represses proteins involved in checkpoint control giving rise to proliferative damage, mitotic disturbances and chromosomal breaks, ultimately resulting in genomic instability. In parallel, emerging evidence of non-nuclear substrates of epigenetic modulators emphasize the need to investigate the role of KDM4A in regulating non-nuclear substrates and evaluate their contribution to genomic instability in this context. The existence of promising KDM-specific inhibitors makes these demethylases an attractive target for therapeutic intervention in cancers.


Subject(s)
Genomic Instability/genetics , Jumonji Domain-Containing Histone Demethylases/physiology , Animals , Cell Transformation, Neoplastic/genetics , Histones/metabolism , Humans , Methylation , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Protein Processing, Post-Translational/genetics , Signal Transduction/genetics
9.
Cell Death Dis ; 11(10): 893, 2020 10 22.
Article in English | MEDLINE | ID: mdl-33093461

ABSTRACT

Pancreatic adenocarcinoma (PDAC) is an extremely malignant tumor that is associated with low survival rates. Fisetin is a natural flavonoid that shows diverse antitumor effects, including DNA damage, in various cancers. Increasing studies have demonstrated that epigenetic modifications play critical roles in DNA-damage response. However, the epigenetic regulation mechanism of fisetin in cancers is hardly studied. RFXAP is a critical transcription factor for MHC II molecules, however, its transcriptional role in PDAC is poorly understood. The anti-PDAC effect of fisetin was measured by CCK-8, flow cytometry, xenograft tumor nude mice model. DNA-damage levels were examined by immunofluorescence. Bioinformatics analysis was used to examine the expression of RFXAP and other genes involved in DNA-damage response. ChIP sequencing was used to explore the transcriptional role of RFXAP. The expression of target gene KDM4A was measured by qRT-PCR and western blots. KDM4A promoter activity was analyzed using dual-luciferase reporter assay. RFXAP overexpressing or silencing of PDAC cells was used to explore the effect of RFXAP in DNA damage induced by fisetin. We found that fisetin inhibited cell proliferation and induced DNA damage and S-phase arrest in PDAC. Expression of RFXAP and other DNA-damage response genes were upregulated by fisetin. We revealed that RFXAP expression was relatively low in PDAC and correlated with tumor stage and poor prognosis. Then we explored the transcriptional role of RFXAP and found that RFXAP targeted KDM4A, a special demethylase specific for tri- and dimethylated histone H3K36. We found that overexpression of RFXAP upregulated KDM4A and attenuated methylation of H3K36, thereby impairing DNA repair and enhancing the DNA damage induced by fisetin, while RFXAP silencing showed the opposite effect. We also found the function of fisetin in enhancing the effect of chemotherapy on pancreatic cancer cells. Our findings revealed that fisetin induced DNA damage via RFXAP/KDM4A-dependent histone H3K36 demethylation, thus causing inhibition of proliferation in PDAC.


Subject(s)
Adenocarcinoma/pathology , Flavonols/pharmacology , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/physiology , Pancreatic Neoplasms/pathology , Transcription Factors/physiology , Adenocarcinoma/genetics , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Line , Cell Proliferation/drug effects , Cell Survival , DNA Damage , Demethylation , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Nude , Middle Aged , Pancreatic Neoplasms/genetics , S Phase/drug effects , Xenograft Model Antitumor Assays
10.
Cells ; 9(10)2020 10 10.
Article in English | MEDLINE | ID: mdl-33050392

ABSTRACT

Metformin, which is suggested to have anti-cancer effects, activates KDM2A to reduce rRNA transcription and proliferation of cancer cells. Thus, the specific activation of KDM2A may be applicable to the treatment of cancers. In this study, we screened a food-additive compound library to identify compounds that control cell proliferation. We found that gallic acid activated KDM2A to reduce rRNA transcription and cell proliferation in breast cancer MCF-7 cells. Gallic acid accelerated ROS production and activated AMPK. When ROS production or AMPK activity was inhibited, gallic acid did not activate KDM2A. These results suggest that both ROS production and AMPK activation are required for activation of KDM2A by gallic acid. Gallic acid did not reduce the succinate level, which was required for KDM2A activation by metformin. Metformin did not elevate ROS production. These results suggest that the activation of KDM2A by gallic acid includes mechanisms distinct from those by metformin. Therefore, signals from multiple intracellular conditions converge in KDM2A to control rRNA transcription. Gallic acid did not induce KDM2A-dependent anti-proliferation activity in non-tumorigenic MCF10A cells. These results suggest that the mechanism of KDM2A activation by gallic acid may be applicable to the treatment of breast cancers.


Subject(s)
F-Box Proteins/metabolism , Gallic Acid/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Transcription, Genetic/drug effects , Adenylate Kinase/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/genetics , DNA Methylation/genetics , F-Box Proteins/physiology , Female , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/physiology , MCF-7 Cells , Metformin/pharmacology , Promoter Regions, Genetic/genetics , Reactive Oxygen Species/metabolism , Transcription, Genetic/genetics
11.
Zhonghua Bing Li Xue Za Zhi ; 49(10): 1041-1045, 2020 Oct 08.
Article in Chinese | MEDLINE | ID: mdl-32992420

ABSTRACT

Objective: To observe the expression level of histone demethyltransferase Jmjd3 in patients with pre-eclampsia (PE), and to investigate the possible mechanism of its epigenetic modification in regulating Th1/Th2 imbalance in PE patients. Methods: The mRNA levels of histone demethyltransferase Jmjd3 from peripheral blood mononuclear cells (PBMC) of PE patients and normal pregnant women were detected by RT-PCR. Peripheral serum IFN-γ and IL-4  were detected by ELISA. RT-PCR was used to detect the mRNA levels of Jmjd3, Tbx21 and Cxcr3 in the spleen of PE and control mice. Immunomagnetic beads were used to sort out the initial CD4+ T cells in the spleen of control and PE mice. Western blot was used to detect H3K27me1 and H3K27me3 levels. ChIP analysis was used for H3K27me3 demethylation modification in spleens of PE mice. Results: Compared with normal pregnant women, the mRNA level of Jmjd3 in PBMC of PE patients was significantly increased, the level of IFN-γ in serum was significantly increased, and the level of IL-4 was significantly reduced (P<0.01). Compared with normal control mice, the mRNA level of Jmjd3 in the spleen of PE mice was significantly increased, and the expression of Tbx21 and Cxcr3 was significantly increased in PE mice (P<0.01); the H3K27me3 level of CD4+ T cells in PE mice was significantly reduced (P<0.05), but H3K27me1 was not changed. ChIP analysis showed that CD4+ T cells H3K27me3 in PE group mice were in the Ifng promoter region, compared with control mice. Recruitment was significantly reduced, while recruitment in the promoter region of Il4 was significantly increased (P<0.01). Conclusions: In both PE patients and mice with PE model, the relative expression level of histone demethyltransferase Jmjd3 is significantly up-regulated, which further induces the demethylation of H3K27me3 in the Ifng promoter region and promotes the initial CD4+ T cells to Th1 cell differentiation and development, leading to an imbalance of Th1/Th2, which may be one of the important reasons for the development of preeclampsia.


Subject(s)
Histones , Jumonji Domain-Containing Histone Demethylases/physiology , Pre-Eclampsia/genetics , Animals , Epigenesis, Genetic , Female , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Leukocytes, Mononuclear , Mice , Pregnancy
12.
Inflamm Res ; 69(7): 657-666, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32394143

ABSTRACT

OBJECTIVES: This study aimed to explore the effects and relative mechanism of JMJD3 on knee osteoarthritis (OA). METHODS: In this study, we first analyzed the expression of JMJD3 in OA cartilage using western blot and immunohistochemistry. In an in vitro study, the effects of GSK-J4, JMJD3 inhibitor, on ATDC-5 chondrocytes were evaluated by CCK-8 assay. Real-time PCR and western blot were used to examine the inhibitory effect of GSK-J4 on the inflammation and ECM degradation of chondrocytes. NF-κB p65 phosphorylation and nuclear translocation were measured by western blot and immunofluorescence. In the animal study, twenty mice were randomized into four experimental groups: sham group, DMM-induced OA + DMSO group, OA + low-dose GSK-J4 group, and OA + high-dose GSK-J4 group. After the treatment, hematoxylin-eosin and safranin O/fast green staining were used to evaluate cartilage degradation of knee joint, with OARSI scores for quantitative assessment of cartilage damage. RESULTS: Our results revealed that JMJD3 was overexpressed in OA cartilage and GSK-J4 could suppress the IL-1ß-induced production of pro-inflammatory cytokines and catabolic enzymes, including IL-6, IL-8, MMP-9 and ADAMTS-5. Consistent with these findings, GSK-J4 could inhibit IL-1ß-induced degradation of collagen II and aggrecan. Mechanistically, GSK-J4 dramatically suppressed IL-1ß-stimulated NF-κB signal pathway activation. In vivo, GSK-J4 prevented cartilage damage in mouse DMM-induced OA model. CONCLUSIONS: This study elucidates the important role of JMJD3 in cartilage degeneration in OA, and our results indicate that JDJM3 may become a novel therapeutic target in OA therapy.


Subject(s)
Benzazepines/pharmacology , Cartilage/drug effects , Chondrocytes/drug effects , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Osteoarthritis/prevention & control , Pyrimidines/pharmacology , Aggrecans/metabolism , Animals , Cartilage/physiopathology , Cell Line , Chondrocytes/physiology , Collagen/metabolism , Gene Expression , Humans , Inflammation/prevention & control , Interleukin-1beta/pharmacology , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/physiology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Osteoarthritis/physiopathology , Rats , Signal Transduction/physiology
13.
Biol Reprod ; 103(1): 114-125, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32318688

ABSTRACT

Correct reprogramming of epigenetic marks in the donor nucleus is a prerequisite for successful cloning by somatic cell transfer (SCT). In several mammalian species, repressive histone (H) lysine (K) trimethylation (me3) marks, in particular H3K9me3, form a major barrier to somatic cell reprogramming into pluripotency and totipotency. We engineered bovine embryonic fibroblasts (BEFs) for the doxycycline-inducible expression of a biologically active, truncated form of murine Kdm4b, a demethylase that removes H3K9me3 and H3K36me3 marks. Upon inducing Kdm4b, H3K9me3 and H3K36me3 levels were reduced about 3-fold and 5-fold, respectively, compared with noninduced controls. Donor cell quiescence has been previously associated with reduced somatic trimethylation levels and increased cloning efficiency in cattle. Simultaneously inducing Kdm4b expression (via doxycycline) and quiescence (via serum starvation) further reduced global H3K9me3 and H3K36me3 levels by a total of 18-fold and 35-fold, respectively, compared with noninduced, nonstarved control fibroblasts. Following SCT, Kdm4b-BEFs reprogrammed significantly better into cloned blastocysts than noninduced donor cells. However, detrimethylated donors and sustained Kdm4b-induction during embryo culture did not increase the rates of postblastocyst development from implantation to survival into adulthood. In summary, overexpressing Kdm4b in donor cells only improved their reprogramming into early preimplantation stages, highlighting the need for alternative experimental approaches to reliably improve somatic cloning efficiency in cattle.


Subject(s)
Blastocyst/physiology , Cattle/embryology , Cellular Reprogramming/physiology , Cloning, Organism , Histones/metabolism , Nuclear Transfer Techniques , Animals , Cellular Reprogramming/genetics , Demethylation , Embryonic Development/physiology , Epigenesis, Genetic , Female , Gene Expression , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/physiology , Mice , Up-Regulation
14.
Cell Death Differ ; 27(10): 2843-2855, 2020 10.
Article in English | MEDLINE | ID: mdl-32346138

ABSTRACT

Medullary thymic epithelial cells (mTECs) play a central role in the establishment of T cell central immunological tolerance by promiscuously expressing tissue-restricted antigens (TRAs) and presenting them to developing T cells, leading to deletion of T cells responding to self-antigens. However, molecular mechanisms especially epigenetic regulation of mTEC homeostasis and TRA expression remain elusive. Here we show that the H3K27 demethylase Kdm6b is essential to maintain the postnatal thymic medulla by promoting mTEC survival and regulating the expression of TRA genes. Moreover, mice lacking Kdm6b developed pathological autoimmune disorders. Mechanically, Kdm6b exerted its function by reducing repressive H3K27 trimethylation (H3K27me3) at the promoters of anti-apoptotic gene Bcl2 and a set of Aire-dependent TRA genes. Thus, our findings reveal a dual role of Kdm6b in the regulation of mTEC-mediated T cell central tolerance.


Subject(s)
Epithelial Cells , Jumonji Domain-Containing Histone Demethylases/physiology , T-Lymphocytes, Regulatory , Thymus Gland , Animals , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/cytology , Thymus Gland/metabolism
15.
Respir Med ; 161: 105852, 2020 01.
Article in English | MEDLINE | ID: mdl-32056726

ABSTRACT

OBJECTIVE: The aim of the study was to investigate the mechanism and effect of FBXL10 in myocardial ischemia reperfusion injury in vivo and in vitro. METHODS: The myocardial ischemia reperfusion (I/R) model was established by 30 min of coronary occlusion followed by 2 h of reperfusion in rats. Western blot and TUNEL assay were used to measure the apoptosis during I/R. The expression levels of endoplasmic reticulum related proteins in myocardial tissues and H9c2 cells were detected by immunohistochemistry staining and immunofluorescence staining. Flow cytometry and CCK-8 were used to detect the apoptosis and viability of H9c2 cells. RESULTS: The results revealed that FBXL10 significantly reduced myocardial infarction, improved the pathological morphology of myocardium, markedly reduced inflammatory response in the myocardial ischemia reperfusion rats. Moreover the expressions of endoplasmic reticulum stress key proteins were caused by I/R were suppressed significantly by FBXL10 treatment, including CHOP, GRP78, ATF4 and p-PERK. Additionally FBXL10 inhibited the expression of endoplasmic reticulum stress key proteins in H/R H9c2 cells. Furthermore, FBXL10 reduced the levels of apoptotic cells and inflammatory response compared with I/R and H/R group. CONCLUSION: Taken together, we found that FBXL10 could attenuate I/R injury through inhibiting endoplasmic reticulum stress (ERs).


Subject(s)
Endoplasmic Reticulum Stress/genetics , F-Box Proteins/physiology , Jumonji Domain-Containing Histone Demethylases/physiology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/prevention & control , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Apoptosis/genetics , Cells, Cultured , Endoplasmic Reticulum Chaperone BiP , Gene Expression/genetics , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Male , Myocardial Reperfusion Injury/pathology , Rats , Rats, Sprague-Dawley , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
16.
Plant Mol Biol ; 102(4-5): 417-430, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31898146

ABSTRACT

KEY MESSAGE: We genome-wide identified 28 JmjC domain-containing genes, further spatio-temporal expression profiling and genetic analysis defined them as epigenetic regulators in flowering initiation of Rosa chinensis. The JmjC domain-containing histone demethylases play critical roles in maintaining homeostasis of histone methylations, thus are vital for plant growth and development. Genome-wide identification of the JmjC domain-containing genes have been reported in several species, however, no systematic study has been performed in rose plants. In this paper, we identified 28 JmjC domain-containing genes from the newly published genome database of Rosa chinensis. The JmjC domain-containing proteins in R. chinensis were divided into seven groups, KDM3 was the largest group with 13 members, and JmjC domain-only A and KDM5B were the smallest clades both with only one member. Although all the JmjC domain proteins having a conserved JmjC domain, the gene and protein structure experienced differentiation and specification during the evolution, especially in KDM3 clade, one gene (RcJMJ40) was found carrying site deletions for cofactors Fe (II) and α-KG binding which were crucial for demethylase activities, three genes (RcJMJ41, RcJMJ43 and RcJMJ44) had no intron while two of them had tandem JmjC domains. Spatial expression pattern analysis of these JmjC domain-containing genes in different tissues showed most of them were highly expressed in reproductive tissues such as floral meristem and closed flowers than vegetative tissues, demonstrating their important functions in developmental switch from vegetative to reproductive growth of roses. Temporal expression profiling indicated majority of JmjC domain-containing genes from R. chinensis fluctuated along with floral bud differentiation and development, further proving their essential roles in flower organogenesis. VIGS induced silencing of RcJMJ12 led to delayed flowering time, and decreased the expression levels of flowering integrator such as RcFT, RcSOC1, RcFUL, RcLFY and RcAP1, therefore providing the genetic evidence of RcJMJ12 in flowering initiation. Collectively, spatio-temporal expression profiling and genetic analysis defined the JmjC domain-containing genes as important epigenetic regulators in flower development of R. chinensis.


Subject(s)
Epigenesis, Genetic , Flowers/genetics , Gene Expression Regulation, Plant , Jumonji Domain-Containing Histone Demethylases/genetics , Meristem/genetics , Rosa/genetics , Gene Deletion , Gene Expression Profiling , Gene Silencing , Genome, Plant , Jumonji Domain-Containing Histone Demethylases/physiology , Methylation , Phylogeny , Plant Proteins/genetics , Protein Domains , Transcriptome
17.
Protein Cell ; 11(3): 161-186, 2020 03.
Article in English | MEDLINE | ID: mdl-31317506

ABSTRACT

The mechanisms underlying spatial and temporal control of cortical neurogenesis of the brain are largely elusive. Long non-coding RNAs (lncRNAs) have emerged as essential cell fate regulators. Here we found LncKdm2b (also known as Kancr), a lncRNA divergently transcribed from a bidirectional promoter of Kdm2b, is transiently expressed during early differentiation of cortical projection neurons. Interestingly, Kdm2b's transcription is positively regulated in cis by LncKdm2b, which has intrinsic-activating function and facilitates a permissive chromatin environment at the Kdm2b's promoter by associating with hnRNPAB. Lineage tracing experiments and phenotypic analyses indicated LncKdm2b and Kdm2b are crucial in proper differentiation and migration of cortical projection neurons. These observations unveiled a lncRNA-dependent machinery in regulating cortical neuronal differentiation.


Subject(s)
Cerebral Cortex/cytology , F-Box Proteins/physiology , Jumonji Domain-Containing Histone Demethylases/physiology , Neurogenesis , Neurons/metabolism , RNA, Long Noncoding/physiology , Animals , Cell Lineage , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Neurons/cytology
18.
Protein Cell ; 10(12): 864-882, 2019 12.
Article in English | MEDLINE | ID: mdl-31701394

ABSTRACT

In recent years, many studies have shown that histone methylation plays an important role in maintaining the active and silent state of gene expression in human diseases. The Jumonji domain-containing protein D3 (JMJD3), specifically demethylate di- and trimethyl-lysine 27 on histone H3 (H3K27me2/3), has been widely studied in immune diseases, infectious diseases, cancer, developmental diseases, and aging related diseases. We will focus on the recent advances of JMJD3 function in human diseases, and looks ahead to the future of JMJD3 gene research in this review.


Subject(s)
Aging/metabolism , Communicable Diseases/metabolism , Immune System Diseases/metabolism , Jumonji Domain-Containing Histone Demethylases/physiology , Neoplasms/metabolism , Animals , Cell Line , Embryonic Development , Humans
19.
EMBO J ; 38(22): e101681, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31583744

ABSTRACT

Epigenetic modifications operate in concert to maintain cell identity, yet how these interconnected networks suppress alternative cell fates remains unknown. Here, we uncover a link between the removal of repressive histone H3K9 methylation and DNA methylation during the reprogramming of somatic cells to pluripotency. The H3K9me2 demethylase, Kdm3b, transcriptionally controls DNA hydroxymethylase Tet1 expression. Unexpectedly, in the absence of Kdm3b, loci that must be DNA demethylated are trapped in an intermediate hydroxymethylated (5hmC) state and do not resolve to unmethylated cytosine. Ectopic 5hmC trapping precludes the chromatin association of master pluripotency factor, POU5F1, and pluripotent gene activation. Increased Tet1 expression is important for the later intermediates of the reprogramming process. Taken together, coordinated removal of distinct chromatin modifications appears to be an important mechanism for altering cell identity.


Subject(s)
Cell Lineage/genetics , Cellular Reprogramming , Chromatin/genetics , DNA Methylation , Epigenesis, Genetic , Histones/genetics , Induced Pluripotent Stem Cells/cytology , Animals , Cells, Cultured , DNA-Binding Proteins/physiology , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Induced Pluripotent Stem Cells/metabolism , Jumonji Domain-Containing Histone Demethylases/physiology , Mice , Mice, Knockout , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins/physiology
20.
Blood Adv ; 3(17): 2537-2549, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31471323

ABSTRACT

KDM2B together with RING1B, PCGF1, and BCOR or BCORL1 comprise polycomb repressive complex 1.1 (PRC1.1), a noncanonical PRC1 that catalyzes H2AK119ub1. It binds to nonmethylated CpG islands through its zinc finger-CxxC DNA binding domain and recruits the complex to target gene loci. Recent studies identified the loss of function mutations in the PRC1.1 gene, BCOR and BCORL1 in human T-cell acute lymphoblastic leukemia (T-ALL). We previously reported that Bcor insufficiency induces T-ALL in mice, supporting a tumor suppressor role for BCOR. However, the function of BCOR responsible for tumor suppression, either its corepressor function for BCL6 or that as a component of PRC1.1, remains unclear. We herein examined mice specifically lacking the zinc finger-CxxC domain of KDM2B in hematopoietic cells. Similar to Bcor-deficient mice, Kdm2b-deficient mice developed lethal T-ALL mostly in a NOTCH1-dependent manner. A chromatin immunoprecipitation sequence analysis of thymocytes revealed the binding of KDM2B at promoter regions, at which BCOR and EZH2 colocalized. KDM2B target genes markedly overlapped with those of NOTCH1 in human T-ALL cells, suggesting that noncanonical PRC1.1 antagonizes NOTCH1-mediated gene activation. KDM2B target genes were expressed at higher levels than the others and were marked with high levels of H2AK119ub1 and H3K4me3, but low levels of H3K27me3, suggesting that KDM2B target genes are transcriptionally active or primed for activation. These results indicate that PRC1.1 plays a key role in restricting excessive transcriptional activation by active NOTCH1, thereby acting as a tumor suppressor in the initiation of T-cell leukemogenesis.


Subject(s)
Carcinogenesis/chemistry , F-Box Proteins/physiology , Jumonji Domain-Containing Histone Demethylases/physiology , Leukemia, T-Cell/etiology , Polycomb Repressive Complex 1/physiology , Tumor Suppressor Proteins/physiology , Animals , CpG Islands , F-Box Proteins/metabolism , Histones , Humans , Jumonji Domain-Containing Histone Demethylases/deficiency , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Mutation , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Protein Domains , Receptor, Notch1/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcriptional Activation , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL
...