Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Int J Mol Sci ; 25(19)2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39409021

ABSTRACT

Genome instability relies on preserving the chromatin structure, with any histone imbalances threating DNA integrity. Histone synthesis occurs in the cytoplasm, followed by a maturation process before their nuclear translocation. This maturation involves protein folding and the establishment of post-translational modifications. Disruptions in this pathway hinder chromatin assembly and contribute to genome instability. JMJD1B, a histone demethylase, not only regulates gene expression but also ensures a proper supply of histones H3 and H4 for the chromatin assembly. Reduced JMJD1B levels lead to the cytoplasmic accumulation of histones, causing defects in the chromatin assembly and resulting in DNA damage. To investigate the role of JMJD1B in regulating genome stability and the malignancy of melanoma tumors, we used a JMJD1B/KDM3B knockout in B16F10 mouse melanoma cells to perform tumorigenic and genome instability assays. Additionally, we analyzed the transcriptomic data of human cutaneous melanoma tumors. Our results show the enhanced tumorigenic properties of JMJD1B knockout melanoma cells both in vitro and in vivo. The γH2AX staining, Micrococcal Nuclease sensitivity, and comet assays demonstrated increased DNA damage and genome instability. The JMJD1B expression in human melanoma tumors correlates with a lower mutational burden and fewer oncogenic driver mutations. Our findings highlight JMJD1B's role in maintaining genome integrity by ensuring a proper histone supply to the nucleus, expanding its function beyond gene expression regulation. JMJD1B emerges as a crucial player in preserving genome stability and the development of melanoma, with a potential role as a safeguard against oncogenic mutations.


Subject(s)
DNA Damage , Genomic Instability , Histones , Jumonji Domain-Containing Histone Demethylases , Melanoma , Skin Neoplasms , Animals , Humans , Mice , Cell Line, Tumor , DNA Damage/genetics , Gene Expression Regulation, Neoplastic , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Melanoma/genetics , Melanoma/pathology , Melanoma/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Melanoma, Experimental/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/metabolism
2.
Sci Rep ; 14(1): 20416, 2024 09 02.
Article in English | MEDLINE | ID: mdl-39223259

ABSTRACT

Autism spectrum disorders (ASD) are complex neurodevelopmental conditions characterized by impairments in social communication, repetitive behaviors, and restricted interests. Epigenetic modifications serve as critical regulators of gene expression playing a crucial role in controlling brain function and behavior. Lysine (K)-specific demethylase 6B (KDM6B), a stress-inducible H3K27me3 demethylase, has emerged as one of the highest ASD risk genes, but the precise effects of KDM6B mutations on neuronal activity and behavioral function remain elusive. Here we show the impact of KDM6B mosaic brain knockout on the manifestation of different autistic-like phenotypes including repetitive behaviors, social interaction, and significant cognitive deficits. Moreover, KDM6B mosaic knockout display abnormalities in hippocampal excitatory synaptic transmission decreasing NMDA receptor mediated synaptic transmission and plasticity. Understanding the intricate interplay between epigenetic modifications and neuronal function may provide novel insights into the pathophysiology of ASD and potentially inform the development of targeted therapeutic interventions.


Subject(s)
Autism Spectrum Disorder , Jumonji Domain-Containing Histone Demethylases , Mice, Knockout , Synaptic Transmission , Animals , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Synaptic Transmission/genetics , Autism Spectrum Disorder/genetics , Mice , Brain/metabolism , Neuronal Plasticity/genetics , Behavior, Animal , Hippocampus/metabolism , Epigenesis, Genetic , Male , Synapses/metabolism
3.
Biol Res ; 57(1): 27, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38745315

ABSTRACT

BACKGROUND: Aberrant gluconeogenesis is considered among primary drivers of hyperglycemia under insulin resistant conditions, with multiple studies pointing towards epigenetic dysregulation. Here we examine the role of miR-721 and effect of epigenetic modulator laccaic acid on the regulation of gluconeogenesis under high fat diet induced insulin resistance. RESULTS: Reanalysis of miRNA profiling data of high-fat diet-induced insulin-resistant mice model, GEO dataset (GSE94799) revealed a significant upregulation of miR-721, which was further validated in invivo insulin resistance in mice and invitro insulin resistance in Hepa 1-6 cells. Interestingly, miR-721 mimic increased glucose production in Hepa 1-6 cells via activation of FOXO1 regulated gluconeogenic program. Concomitantly, inhibition of miR-721 reduced glucose production in palmitate induced insulin resistant Hepa 1-6 cells by blunting the FOXO1 induced gluconeogenesis. Intriguingly, at epigenetic level, enrichment of the transcriptional activation mark H3K36me2 got decreased around the FOXO1 promoter. Additionally, identifying targets of miR-721 using miRDB.org showed H3K36me2 demethylase KDM2A as a potential target. Notably, miR-721 inhibitor enhanced KDM2A expression which correlated with H3K36me2 enrichment around FOXO1 promoter and the downstream activation of the gluconeogenic pathway. Furthermore, inhibition of miR-721 in high-fat diet-induced insulin-resistant mice resulted in restoration of KDM2A levels, concomitantly reducing FOXO1, PCK1, and G6PC expression, attenuating gluconeogenesis, hyperglycemia, and improving glucose tolerance. Interestingly, the epigenetic modulator laccaic acid also reduced the hepatic miR-721 expression and improved KDM2A expression, supporting our earlier report that laccaic acid attenuates insulin resistance by reducing gluconeogenesis. CONCLUSION: Our study unveils the role of miR-721 in regulating gluconeogenesis through KDM2A and FOXO1 under insulin resistance, pointing towards significant clinical and therapeutic implications for metabolic disorders. Moreover, the promising impact of laccaic acid highlights its potential as a valuable intervention in managing insulin resistance-associated metabolic diseases.


Subject(s)
Gluconeogenesis , Insulin Resistance , Jumonji Domain-Containing Histone Demethylases , MicroRNAs , Animals , Male , Mice , Diet, High-Fat , Epigenesis, Genetic , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , Gluconeogenesis/genetics , Gluconeogenesis/physiology , Insulin Resistance/physiology , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Mice, Inbred C57BL , MicroRNAs/metabolism , MicroRNAs/genetics
4.
Curr Mol Pharmacol ; 16(2): 197-210, 2023.
Article in English | MEDLINE | ID: mdl-35297358

ABSTRACT

BACKGROUND: Valproic acid/sodium valproate (VPA), a well-known anti-epileptic agent, inhibits histone deacetylases, induces histone hyperacetylation, promotes DNA demethylation, and affects the histone methylation status in some cell models. Histone methylation profiles have been described as potential markers for cervical cancer prognosis. However, histone methylation markers that can be studied in a cervical cancer cell line, like HeLa cells, have not been investigated following treatment with VPA. METHODS: In this study, the effect of 0.5 mM and 2.0 mM VPA for 24 h on H3K4me2/me3, H3K9me/me2 and H3K27me/me3 signals as well as on KMT2D, EZH2, and KDM3A gene expression was investigated using confocal microscopy, Western blotting, and RT-PCR. Histone methylation changes were also investigated by Fourier-transform infrared spectroscopy (FTIR). RESULTS: We found that VPA induces increased levels of H3K4me2/me3 and H3K9me, which are indicative of chromatin activation. Particularly, H3K4me2 markers appeared intensified close to the nuclear periphery, which may suggest their implication in increased transcriptional memory. The abundance of H3K4me2/me3 in the presence of VPA was associated with increased methyltransferase KMT2D gene expression. VPA induced hypomethylation of H3K9me2, which is associated with gene silencing, and concomitant with the demethylase KDM3A, it increased gene expression. Although VPA induces increased H3K27me/me3 levels, it is suggested that the role of the methyltransferase EZH2 in this context could be affected by interactions with this drug. CONCLUSION: Histone FTIR spectra were not affected by VPA under present experimental conditions. Whether our epigenetic results are consistent with VPA affecting the aggressive tumorous state of HeLa cells, further investigation is required.


Subject(s)
DNA Methylation , Histones , Uterine Cervical Neoplasms , Valproic Acid , Female , Humans , HeLa Cells , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Lysine/metabolism , Methyltransferases , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , Valproic Acid/pharmacology , Valproic Acid/therapeutic use
5.
Int J Mol Sci ; 23(19)2022 Oct 07.
Article in English | MEDLINE | ID: mdl-36233212

ABSTRACT

Despite advances in diagnostic and therapeutic approaches for lung cancer, new therapies targeting metastasis by the specific regulation of cancer genes are needed. In this study, we screened a small library of epigenetic inhibitors in non-small-cell lung cancer (NSCLC) cell lines and evaluated 38 epigenetic targets for their potential role in metastatic NSCLC. The potential candidates were ranked by a streamlined approach using in silico and in vitro experiments based on publicly available databases and evaluated by real-time qPCR target gene expression, cell viability and invasion assays, and transcriptomic analysis. The survival rate of patients with lung adenocarcinoma is inversely correlated with the gene expression of eight epigenetic targets, and a systematic review of the literature confirmed that four of them have already been identified as targets for the treatment of NSCLC. Using nontoxic doses of the remaining inhibitors, KDM6B and PADI4 were identified as potential targets affecting the invasion and migration of metastatic lung cancer cell lines. Transcriptomic analysis of KDM6B and PADI4 treated cells showed altered expression of important genes related to the metastatic process. In conclusion, we showed that KDM6B and PADI4 are promising targets for inhibiting the metastasis of lung adenocarcinoma cancer cells.


Subject(s)
Adenocarcinoma of Lung , Carcinoma, Non-Small-Cell Lung , Jumonji Domain-Containing Histone Demethylases , Lung Neoplasms , Protein-Arginine Deiminase Type 4 , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Early Detection of Cancer , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Protein-Arginine Deiminase Type 4/genetics
6.
Clin Transl Oncol ; 24(7): 1238-1249, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35239138

ABSTRACT

Histone lysine methylation plays a key role in gene activation and repression. The trimethylation of histone H3 on lysine-27 (H3K27me3) is a critical epigenetic event that is controlled by Jumonji domain-containing protein-3 (JMJD3). JMJD3 is a histone demethylase that specifically removes methyl groups. Previous studies have suggested that JMJD3 has a dual role in cancer cells. JMJD3 stimulates the expression of proliferative-related genes and increases tumor cell growth, propagation, and migration in various cancers, including neural, prostate, ovary, skin, esophagus, leukemia, hepatic, head and neck, renal, lymphoma, and lung. In contrast, JMJD3 can suppress the propagation of tumor cells, and enhance their apoptosis in colorectal, breast, and pancreatic cancers. In this review, we summarized the recent advances of JMJD3 function in cancer cells.


Subject(s)
Lysine , Neoplasms , Apoptosis , Female , Histones/genetics , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Lysine/metabolism , Male , Methylation , Neoplasms/genetics
7.
Aging (Albany NY) ; 13(18): 21914-21940, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34528900

ABSTRACT

Metformin has been tested as an anti-cancer therapy with potential to improve conventional chemotherapy. However, in some cases, metformin fails to sensitize tumors to chemotherapy. Here we test if the presence of P53 could predict the activity of metformin as an adjuvant for cisplatin-based therapy in non-small cell lung cancer (NSCLC). A549, HCC 827 (TP53 WT), H1299, and H358 (TP53 null) cell lines were used in this study. A549 cells were pre-treated with a sub-lethal dose of cisplatin to induce chemoresistance. The effects of metformin were tested both in vitro and in vivo and related to the ability of cells to accumulate Jarid1b, a histone demethylase involved in cisplatin resistance in different cancers. Metformin sensitized A549 and HCC 827 cells (but not H1299 and H358 cells) to cisplatin in a P53-dependent manner, changing its subcellular localization to the mitochondria. Treatment with a sub-lethal dose of cisplatin increased Jarid1b expression, yet downregulated P53 levels, protecting A549Res cells from metformin-induced chemosensitization to cisplatin and favored a glycolytic phenotype. Treatment with FL3, a synthetic flavagline, sensitized A549Res cells to cisplatin. In conclusion, metformin could potentially be used as an adjuvant for cisplatin-based therapy in NSCLC cells if wild type P53 is present.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Cisplatin/pharmacology , Jumonji Domain-Containing Histone Demethylases/genetics , Metformin/pharmacology , Nuclear Proteins/genetics , Repressor Proteins/genetics , Tumor Suppressor Protein p53/genetics , A549 Cells , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Drug Synergism , Gene Expression Regulation, Neoplastic , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Male , Mice , Mice, Inbred NOD , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Protein p53/metabolism
8.
Parasit Vectors ; 13(1): 140, 2020 Mar 17.
Article in English | MEDLINE | ID: mdl-32178714

ABSTRACT

BACKGROUND: Schistosomiasis chemotherapy is largely based on praziquantel (PZQ). Although PZQ is very safe and tolerable, it does not prevent reinfection and emerging resistance is a primary concern. Recent studies have shown that the targeting of epigenetic machinery in Schistosoma mansoni may result in severe alterations in parasite development, leading to death. This new route for drug discovery in schistosomiasis has focused on classes of histone deacetylases (HDACs) and histone acetyltransferases (HATs) as epigenetic drug targets. Schistosoma histone demethylases also seem to be important in the transition of cercariae into schistosomula, as well as sexual differentiation in adult worms. METHODS: The Target-Pathogen database and molecular docking assays were used to prioritize the druggability of S. mansoni histone demethylases. The transcription profile of Smp_03400 was re-analyzed using available databases. The effect of GSK-J4 inhibitor in schistosomula and adult worms' motility/viability/oviposition was assessed by in vitro assays. Ultrastructural analysis was performed on adult worms exposed to GSK-J4 by scanning electron microscopy, while internal structures and muscle fiber integrity was investigated by confocal microscopy after Langeron's carmine or phalloidin staining. RESULTS: The present evaluation of the potential druggability of 14 annotated S. mansoni demethylase enzymes identified the S. mansoni ortholog of human KDM6A/UTX (Smp_034000) as the most suitable druggable target. In silico analysis and molecular modeling indicated the potential for cofactor displacement by the chemical probe GSK-J4. Our re-analysis of transcriptomic data revealed that Smp_034000 expression peaks at 24 h in newly transformed schistosomula and 5-week-old adult worms. Moreover, this gene was highly expressed in the testes of mature male worms compared to the rest of the parasite body. In in vitro schistosome cultures, treatment with GSK-J4 produced striking effects on schistosomula mortality and adult worm motility and mortality, as well as egg oviposition, in a dose- and time-dependent manner. Unexpectedly, western blot assays did not demonstrate overall modulation of H3K27me3 levels in response to GSK-J4. Confocal and scanning electron microscopy revealed the loss of original features in muscle fibers and alterations in cell-cell contact following GSK-J4 treatment. CONCLUSIONS: GSK-J4 presents promising potential for antischistosomal control; however, the underlying mechanisms warrant further investigation.


Subject(s)
Anthelmintics/pharmacology , Benzazepines/pharmacology , Drug Discovery/methods , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Models, Molecular , Pyrimidines/pharmacology , Schistosoma mansoni/drug effects , Animals , Computational Biology , Epigenesis, Genetic/drug effects , Female , Jumonji Domain-Containing Histone Demethylases/genetics , Male , Microscopy, Electron, Scanning , Molecular Docking Simulation , Schistosoma mansoni/genetics , Schistosoma mansoni/ultrastructure , Schistosomiasis mansoni/drug therapy , Transcriptome
9.
Epigenetics Chromatin ; 13(1): 6, 2020 02 18.
Article in English | MEDLINE | ID: mdl-32070414

ABSTRACT

BACKGROUND: Maintaining a proper supply of soluble histones throughout the cell cycle is important to ensure chromatin and genome stability. Following their synthesis, histones undergo a series of maturation steps to prepare them for deposition onto chromatin. RESULTS: Here, we identify the lysine demethylase JMJD1B as a novel player in the maturation cascade that contributes to regulate histone provision. We find that depletion of JMJD1B increases the protein levels of the histone chaperone tNASP leading to an accumulation of newly synthesized histones H3 and H4 at early steps of the histone maturation cascade, which perturbs chromatin assembly. Furthermore, we find a high rate of JMJD1B mutations in cancer patients, and a correlation with genomic instability. CONCLUSIONS: Our data support a role for JMJD1B in fine-tuning histone supply to maintain genome integrity, opening novel avenues for cancer therapeutics.


Subject(s)
Genomic Instability , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Protein Processing, Post-Translational , HeLa Cells , Histone Code , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Mutation
11.
PLoS One ; 14(1): e0210207, 2019.
Article in English | MEDLINE | ID: mdl-30629659

ABSTRACT

The process of neuronal differentiation is associated with neurite elongation and membrane biogenesis, and phosphatidylcholine (PtdCho) is the major membrane phospholipid in mammalian cells. During neuroblast differentiation, the transcription of two genes involved in PtdCho biosynthesis are stimulated: Chka gene for choline kinase (CK) alpha isoform and Pcyt1a gene for CTP:phosphocholine cytidylyltransferase (CCT) alpha isoform. Here we show that CKα is essential for neuronal differentiation. In addition, we demonstrated that KDM2B regulates CKα expression and, as a consequence, neuronal differentiation. This factor is up-regulated in the course of the neuroblasts proliferative and undifferentiated state and down-regulated during differentiation induced by retinoic acid (RA). During proliferation, KDM2B binds to the Box2 located in the Chka promoter repressing its transcription. Interestingly, KDM2B knockdown enhances the levels of CKα expression in neuroblast cells and induces neuronal differentiation even in the absence of RA. These results suggest that KDM2B is required for the appropriate regulation of CKα during neuronal differentiation and to the maintaining of the undifferentiated stage of neuroblast cells.


Subject(s)
Choline Kinase/genetics , F-Box Proteins/metabolism , Gene Expression Regulation, Neoplastic , Jumonji Domain-Containing Histone Demethylases/metabolism , Neuroblastoma/genetics , Tretinoin/metabolism , Animals , Cell Differentiation/genetics , Cell Line, Tumor , Choline Kinase/metabolism , Epigenesis, Genetic , F-Box Proteins/genetics , Follow-Up Studies , Gene Knockdown Techniques , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Neural Stem Cells/physiology , Neuroblastoma/mortality , Neuroblastoma/pathology , Prognosis , Promoter Regions, Genetic/genetics , RNA, Small Interfering/metabolism , Up-Regulation
12.
Stem Cells ; 35(12): 2430-2441, 2017 12.
Article in English | MEDLINE | ID: mdl-28895234

ABSTRACT

Novel bone regeneration approaches aim to obtain immature osteoblasts from somatic stem cells. Umbilical cord Wharton's jelly mesenchymal stem cells (WJ-MSCs) are an ideal source for cell therapy. Hence, the study of mechanisms involved in WJ-MSC osteoblastic differentiation is crucial to exploit their developmental capacity. Here, we have assessed epigenetic control of the Runt-related transcription factor 2 (RUNX2) osteogenic master regulator gene in WJ-MSC. We present evidence indicating that modulation of RUNX2 expression through preventing Jumonji AT-rich interactive domain 1B (JARID1B) histone demethylase activity is relevant to enhance WJ-MSC osteoblastic potential. Hence, JARID1B loss of function in WJ-MSC results in increased RUNX2/p57 expression. Our data highlight JARID1B activity as a novel target to modulate WJ-MSC osteoblastic differentiation with potential applications in bone tissue engineering. Stem Cells 2017;35:2430-2441.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/metabolism , Mesenchymal Stem Cells/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Epigenomics , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Osteoblasts/metabolism , Umbilical Cord/cytology , Wharton Jelly/cytology
13.
Genet Mol Res ; 14(2): 5417-26, 2015 May 22.
Article in English | MEDLINE | ID: mdl-26125737

ABSTRACT

The aims of this study were to investigate the expression of the H3K4 demethylase, jumonji AT-rich interactive domain 1B (JARID1B/KDM5B) and of p16 (multiple tumor suppressor gene MTS1) in breast cancer tissue and determine its clinicopathological significance. JARID1B/KDM5B and P16 protein expression in 176 resected breast cancer specimens and adjacent normal breast tissue was detected by the streptavidin-peroxidase (S-P) immunohistochemical method. The TNM staging grade was assigned according to the World Health Organization (2012) breast classification system. The positive staining rate of JARID1B/KDM5B and p16 protein in cancer tissue was 74.43 and 35.8%, respectively. JARID1B/KDM5B protein expression was positively associated with T grade, Bloom and Richardson (B&R) score and axillary lymph node metastasis (P < 0.05). p16 protein expression was negatively associated with T grade, B&R score, and axillary lymph node metastasis (P < 0.05). JARID1B/KDM5B and p16 protein expression in breast cancer and adjacent normal breast tissue were negatively correlated (r = -0.303, P < 0.001). The data demonstrated that protein expression of p16 and JARID1B/KDM5B is negatively correlated in invasive ductal carcinoma of the breast.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Ductal/genetics , Cyclin-Dependent Kinase Inhibitor p16/biosynthesis , Jumonji Domain-Containing Histone Demethylases/biosynthesis , Nuclear Proteins/biosynthesis , Repressor Proteins/biosynthesis , Adult , Aged , Breast Neoplasms/pathology , Carcinoma, Ductal/pathology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Lymphatic Metastasis , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Staging , Nuclear Proteins/genetics , Repressor Proteins/genetics
14.
Cancer Genet ; 208(5): 215-24, 2015 May.
Article in English | MEDLINE | ID: mdl-25633974

ABSTRACT

Histone posttranslational modifications are important components of epigenetic regulation. One extensively studied modification is the methylation of lysine residues. These modifications were thought to be irreversible. However, several proteins with histone lysine demethylase functions have been discovered and characterized. Among these proteins, KDM4A is the first histone lysine demethylase shown to demethylate trimethylated residues. This enzyme plays an important role in gene expression, cellular differentiation, and animal development. Recently, it has also been shown to be involved in cancer. In this review, we focus on describing the structure, mechanisms, and function of KDM4A. We primarily discuss the role of KDM4A in cancer development and the importance of KDM4A as a potential therapeutic target.


Subject(s)
Cell Transformation, Neoplastic/genetics , Histones/chemistry , Jumonji Domain-Containing Histone Demethylases/physiology , Neoplasms/enzymology , Epigenesis, Genetic , Histones/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/genetics , Neoplasms/genetics , Transcription, Genetic/genetics
15.
Genet Mol Res ; 14(4): 18249-58, 2015 Dec 28.
Article in English | MEDLINE | ID: mdl-26782472

ABSTRACT

Cell reprogramming mediated by histone methylation and demethylation is crucial for the activation of the embryonic genome in early embryonic development. In this study, we employed quantitative real-time polymerase chain reaction (qRT-PCR) to detect mRNA levels and expression patterns of all known histone demethylases in early germinal vesicle stage and in vitro-matured metaphase II (MII) oocytes (which are commonly used as donor cells for nuclear transfer). On screening, the Jumonji domain containing 1C (JMJD1C) gene had the highest level of expression and hence was used for subsequent experiments. We also found that JMJD1C was primarily expressed in the nucleus and showed relatively high levels of expression at the 2-cell, 4-cell, 8-cell, 16-cell, morula, and blastocyst stages of embryos developed from MII oocytes fertilized in vitro. Further, we knocked down the JMJD1C gene in MII oocytes using siRNA and monitored the cleavage of zygotes and development of early embryos after in vitro fertilization. The results showed that the zygote cleavage and blastocyst rates of the transfection group were reduced by 57.1 ± 0.07 and 50 ± 0.01% respectively, which were significantly lower than those of the negative control group (P < 0.05). These data suggest that JMJD1C plays a key role in the normal development of early bovine embryos. Our results also provide a theoretical basis for the investigation of the role and molecular mechanism of histone demethylation in the early development of bovine embryos.


Subject(s)
Cell Nucleus/genetics , Embryo, Mammalian , Embryonic Development/genetics , Jumonji Domain-Containing Histone Demethylases/biosynthesis , Animals , Blastocyst/metabolism , Cattle , Cell Nucleus/metabolism , Female , Fertilization in Vitro , Jumonji Domain-Containing Histone Demethylases/genetics , Methylation , Morula/metabolism , Nuclear Transfer Techniques , Oocytes/enzymology , Oocytes/growth & development , Pregnancy , RNA, Messenger/genetics , Zygote/growth & development
16.
Clin Oral Investig ; 17(4): 1279-85, 2013 May.
Article in English | MEDLINE | ID: mdl-22875665

ABSTRACT

OBJECTIVE: The role of epigenetic regulation in inflammatory diseases such as periodontitis is poorly known. The aim of this study was to assess whether Porphyromonas gingivalis lipopolysaccharide (LPS) can modulate gene expression levels of the some enzymes that promote epigenetic events in cultures of the human keratinocytes and gingival fibroblasts. In addition, the same enzymes were evaluated in gingival samples from healthy and periodontitis-affected individuals. MATERIALS AND METHODS: Primary gingival fibroblast and keratinocyte (HaCaT) cultures were treated with medium containing P. gingivalis LPS or P. gingivalis LPS vehicle for 24 h. After this period, cell viability was assessed by MTT test and total RNA extracted to evaluate gene expression levels of the following enzymes by qRT-PCR: DNA methyltransferase 1 (DNMT1), DNA methyltransferase 3a (DNMT3a), histone demethylases Jumonji domain containing 3 (JMJD3) and ubiquitously transcribed tetratricopeptide repeat, X chromosome (UTX). To evaluate gene expression in healthy and periodontitis-affected individuals, total RNA was extracted from biopsies of gingival tissue from healthy and periodontitis sites, and gene expression of DNMT1, DNAMT3a, JMJD3, and UTX was evaluated by qRT-PCR. RESULTS: No significant differences were found in the gene expression analysis between healthy and periodontitis-affected gingival samples. The results showed that LPS downregulated DNMT1 (p < 0.05), DNMT3a (p < 0.05), and JMJD3 (p < 0.01) gene expression in HaCaT cells, but no modulation was observed in gingival fibroblasts. CONCLUSION: P. gingivalis LPS exposure to human HaCaT keratinocytes downregulates gene expression of the enzymes that promote epigenetic events. CLINICAL RELEVANCE: The advance knowledge about epigenetic modifications caused by periodontopathogens may to possibly led to the development of new periodontal therapies.


Subject(s)
Chronic Periodontitis/genetics , Epigenesis, Genetic/drug effects , Gene Expression Regulation/drug effects , Gingiva/cytology , Lipopolysaccharides/pharmacology , Porphyromonas gingivalis/pathogenicity , Adult , Aged , Case-Control Studies , Cell Survival/drug effects , Cells, Cultured , Chronic Periodontitis/enzymology , Chronic Periodontitis/microbiology , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/genetics , DNA Methyltransferase 3A , Down-Regulation , Female , Fibroblasts , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Keratinocytes , Male , Middle Aged , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL