Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 76(6): 885-895.e7, 2019 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-31629659

RESUMO

Hypoxia, which occurs during tumor growth, triggers complex adaptive responses in which peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) plays a critical role in mitochondrial biogenesis and oxidative metabolism. However, how PGC-1α is regulated in response to oxygen availability remains unclear. We demonstrated that lysine demethylase 3A (KDM3A) binds to PGC-1α and demethylates monomethylated lysine (K) 224 of PGC-1α under normoxic conditions. Hypoxic stimulation inhibits KDM3A, which has a high KM of oxygen for its activity, and enhances PGC-1α K224 monomethylation. This modification decreases PGC-1α's activity required for NRF1- and NRF2-dependent transcriptional regulation of TFAM, TFB1M, and TFB2M, resulting in reduced mitochondrial biogenesis. Expression of PGC-1α K224R mutant significantly increases mitochondrial biogenesis, reactive oxygen species (ROS) production, and tumor cell apoptosis under hypoxia and inhibits brain tumor growth in mice. This study revealed that PGC-1α monomethylation, which is dependent on oxygen availability-regulated KDM3A, plays a critical role in the regulation of mitochondrial biogenesis.


Assuntos
Neoplasias Encefálicas/enzimologia , Histona Desmetilases com o Domínio Jumonji/metabolismo , Mitocôndrias/enzimologia , Biogênese de Organelas , Oxigênio/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Apoptose , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Metilação , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitocôndrias/patologia , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Carga Tumoral , Hipóxia Tumoral , Microambiente Tumoral
2.
Mol Cell ; 74(6): 1250-1263.e6, 2019 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-31054974

RESUMO

Alternative pre-mRNA-splicing-induced post-transcriptional gene expression regulation is one of the pathways for tumors maintaining proliferation rates accompanying the malignant phenotype under stress. Here, we uncover a list of hyperacetylated proteins in the context of acutely reduced Acetyl-CoA levels under nutrient starvation. PHF5A, a component of U2 snRNPs, can be acetylated at lysine 29 in response to multiple cellular stresses, which is dependent on p300. PHF5A acetylation strengthens the interaction among U2 snRNPs and affects global pre-mRNA splicing pattern and extensive gene expression. PHF5A hyperacetylation-induced alternative splicing stabilizes KDM3A mRNA and promotes its protein expression. Pathologically, PHF5A K29 hyperacetylation and KDM3A upregulation axis are correlated with poor prognosis of colon cancer. Our findings uncover a mechanism of an anti-stress pathway through which acetylation on PHF5A promotes the cancer cells' capacity for stress resistance and consequently contributes to colon carcinogenesis.


Assuntos
Processamento Alternativo , Carcinogênese/genética , Neoplasias Colorretais/genética , Regulação Neoplásica da Expressão Gênica , Histona Desmetilases com o Domínio Jumonji/genética , Proteínas de Ligação a RNA/genética , Transativadores/genética , Acetilcoenzima A/deficiência , Acetilação , Animais , Carcinogênese/metabolismo , Carcinogênese/patologia , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Células HCT116 , Humanos , Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Histona Desmetilases com o Domínio Jumonji/metabolismo , Células MCF-7 , Masculino , Camundongos , Camundongos Nus , Prognóstico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteína Nuclear Pequena U2/genética , Ribonucleoproteína Nuclear Pequena U2/metabolismo , Transdução de Sinais , Análise de Sobrevida , Transativadores/antagonistas & inibidores , Transativadores/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Fatores de Transcrição de p300-CBP/genética , Fatores de Transcrição de p300-CBP/metabolismo
3.
Exp Cell Res ; 435(2): 113945, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38286256

RESUMO

Bronchopulmonary dysplasia (BPD) is the most common chronic lung disease among neonates, with increasing morbidity and mortality. This study aims to investigate the effect and mechanism of lysine demethylase 3A (KDM3A) on hyperoxia-induced BPD. Hyperoxia-induced BPD mouse and alveolar epithelial cell models were constructed. The effects of hyperoxia on lung development were evaluated by histological and morphological analysis. The levels of KDM3A, E26 transformation specific-1 (ETS1), H3 lysine 9 dimethylation (H3K9me2), and endoplasmic reticulum (ER) stress-related indexes were quantified by RT-qPCR, Western blot, and IF staining. Cell apoptosis was assessed by flow cytometry and TUNEL staining. Transfection of oe-ETS1, oe-KDM3A, and sh-ETS1 was applied in hyperoxia-induced alveolar epithelial cells to explore the mechanism of the KDM3A/ETS1 axis in hyperoxia-induced apoptosis. KDM3A inhibitor IOX1 was applied to validate the in vivo effect of KDM3A in hyperoxia-induced BPD mice. The results displayed that hyperoxia-induced BPD mice showed reduced body weight, severe destruction of alveolar structure, decreased radial alveolar count (RAC), and increased mean linear intercept (MLI) and mean alveolar diameter (MAD). Further, hyperoxia induction down-regulated ETS1 expression, raised ER stress levels, and increased apoptosis rate in BPD mice and alveolar epithelial cells. However, transfection of oe-ETS1 improved the above changes in hyperoxia-induced alveolar epithelial cells. Moreover, transfection of oe-KDM3A up-regulated ETS1 expression, down-regulated H3K9me2 expression, inhibited ER stress, and reduced apoptosis rate in hyperoxia-induced alveolar epithelial cells. In addition, transfection of sh-ETS1 reversed the inhibitory effect of KDM3A on hyperoxia-induced apoptosis by regulating ER stress. In vivo experiments, KDM3A inhibitor IOX1 intervention further aggravated BPD in newborn mice. In a word, KDM3A alleviated hyperoxia-induced BPD in mice by promoting ETS1 expression.


Assuntos
Displasia Broncopulmonar , Hiperóxia , Animais , Camundongos , Animais Recém-Nascidos , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/metabolismo , Modelos Animais de Doenças , Hiperóxia/complicações , Hiperóxia/metabolismo , Hiperóxia/patologia , Pulmão/metabolismo , Lisina/metabolismo , Fatores de Transcrição/metabolismo
4.
Mol Cell Neurosci ; 122: 103759, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35901929

RESUMO

Microglia activation has been suggested as the key factor in neuro-inflammation and thus participates in neurological diseases. Although taurine exhibits anti-inflammatory and neuro-protective effects, its underlying epigenetic mechanism is unknown. In this study, taurine was administered to lipopolysaccharide (LPS)-treated mice and BV-2 cells. Behavioral test, morphological analyze, detection of microglia activation, and lysine demethylase 3a (KDM3a) measurements were performed to investigate the mechanism by which taurine regulates KDM3a and subsequently antagonizes microglia activation. Taurine improved the sociability of LPS-treated mice, inhibited microglia activation in the hippocampus, and reduced generation of brain inflammatory factors, such as interleukin-6, tumor necrosis factor-α, inducible nitric oxide synthase, and cyclooxygenase-2. Meanwhile, taurine suppressed the LPS-induced increase in microglial KDM3a, and increased the level of mono-, di- or tri-methylation of lysine 9 on histone H3 (H3K9me1/2/3). Furthermore, taurine inhibited the LPS-induced increase in KDM3a, elevated the H3K9me1/2/3 level, and reduced inflammatory factors and reactive oxygen species in a concentration-dependent manner in LPS-stimulated BV-2 cells. In conclusion, taurine inhibited KDM3a and microglia activation, thereby playing an anti-inflammatory role in LPS-treated mice and BV-2 cells.


Assuntos
Lipopolissacarídeos , Microglia , Animais , Anti-Inflamatórios/farmacologia , Linhagem Celular , Ciclo-Oxigenase 2/metabolismo , Ciclo-Oxigenase 2/farmacologia , Lipopolissacarídeos/toxicidade , Lisina , Camundongos , Microglia/metabolismo , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Taurina/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
5.
Adv Exp Med Biol ; 1433: 69-86, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37751136

RESUMO

Methylation of histone H3 lysine 9 (H3K9) is a repressive histone mark and associated with inhibition of gene expression. KDM3 is a subfamily of the JmjC histone demethylases. It specifically removes the mono- or di-methyl marks from H3K9 and thus contributes to activation of gene expression. KDM3 subfamily includes three members: KDM3A, KDM3B and KDM3C. As KDM3A (also known as JMJD1A or JHDM2A) is the best studied, this chapter will mainly focus on the role of KDM3A-mediated gene regulation in the biology of normal and cancer cells. Knockout mouse studies have revealed that KDM3A plays a role in the physiological processes such as spermatogenesis, metabolism and sex determination. KDM3A is upregulated in several types of cancers and has been shown to promote cancer development, progression and metastasis. KDM3A can enhance the expression or activity of transcription factors through its histone demethylase activity, thereby altering the transcriptional program and promoting cancer cell proliferation and survival. We conclude that KDM3A may serve as a promising target for anti-cancer therapies.


Assuntos
Histona Desmetilases com o Domínio Jumonji , Neoplasias , Animais , Masculino , Camundongos , Regulação da Expressão Gênica , Camundongos Knockout , Neoplasias/genética , Fatores de Transcrição , Histona Desmetilases com o Domínio Jumonji/genética
6.
Balkan J Med Genet ; 25(2): 23-27, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37265967

RESUMO

Colorectal cancer (CRC) is one of the leading causes of cancer-linked deaths globally. The determination of biomarkers is important in the prognosis and treatment of CRC. Previous studies emphasized the relationship between hypoxia and CRC in humans, and there is strong evidence that this process is strongly related to HIF-1. KDM3A is a histone demethylase that could directly bind to HIF-1α, a subunit of HIF-1. This study aimed to reveal whether the expression level of the KDM3A gene could be used as a predictor of CRC. The expression levels of HIF-1α, KDM3A, and Epithelial-Mesenchymal Transition (EMT) genes were evaluated by qRT-PCR in leukocyte samples of 50 CRC patients in different stages and 50 healthy controls. HIF-1α and KDM3A expression levels were significantly higher in the CRC group, compared to the controls. Slug and ZEB-1 genes, the mesenchymal markers, showed the same significance pattern between groups. We acquired 0.664 AUC with 54% sensitivity and 85.4% specificity for separating controls from CRC patients by using the KDM3A expression levels in ROC analysis. This data support that KDM3A could be a novel supplementary biomarker in diagnosis of CRC, which could be noninvasively detected in circulation.

7.
Mol Carcinog ; 61(4): 392-396, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35020958

RESUMO

JMJD1A (also called lysine demethylase 3A [KDM3A]) belongs to the Jumonji C family of histone demethylases. It specifically removes the repressive mono- or di-methyl marks from histone H3 at lysine 9 and thus contributes to the activation of gene transcription. JMJD1A plays a key role in a variety of biological processes such as spermatogenesis, metabolism, sex determination, and stem cell activity. JMJD1A is upregulated in various types of cancers and can promote cancer development, progression, and therapeutic resistance. JMJD1A can epigenetically regulate the expression or activity of transcription factors such as c-Myc, androgen receptor (AR), estrogen receptor (ER), ß-catenin, and so on. Expression and activity of JMJD1A in cancer cells can be regulated at transcriptional, post-transcriptional, and post-translational levels. Targeting JMJD1A may repress the oncogenic transcription factors as a potential anticancer therapy.


Assuntos
Histona Desmetilases , Neoplasias , Resistencia a Medicamentos Antineoplásicos/genética , Histona Desmetilases/metabolismo , Histonas/metabolismo , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Lisina , Masculino , Neoplasias/tratamento farmacológico , Neoplasias/genética , Fatores de Transcrição/metabolismo
8.
BMC Cancer ; 22(1): 562, 2022 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-35590288

RESUMO

BACKGROUND: Lysine-specific histone demethylase 3A (KDM3A) is a potent histone modifier that is frequently implicated in the progression of several malignancies. However, its role in aerobic glycolysis of osteosarcoma (OS) remains unclear. METHODS: KDM3A expression in OS tissues was determined by immunohistochemistry, and that in acquired OS cells was determined by RT-qPCR and western blot assays. KDM3A was silenced in OS cells to examine cellular behaviors and the aerobic glycolysis. Stably transfected cells were injected into nude mice for in vivo experiments. The downstream targets of KDM3A were predicted by bioinformatics systems and validated by ChIP-qPCR. Rescue experiments of SP1 and PFKFB4 were performed to examine their roles in the KDM3A-mediated events. RESULTS: KDM3A was highly expressed in OS tissues and cells. Knockdown of KDM3A weakened OS cell growth and metastasis in vivo and in vitro, and it suppressed the aerobic glycolysis in OS cells. KDM3A enhanced the transcription of SP1 by demethylating H3K9me2 on its promoter. Restoration of SP1 rescued growth and metastasis of OS cells and recovered the glycolytic flux in cells suppressed by knockdown of KDM3A. SP1 bound to the PFKFB4 promoter to activate its transcription and expression. PFKFB4 expression in OS cells was suppressed by KDM3A silencing but increased after SP1 restoration. Overexpression of PFKFB4 significantly promoted OS cell growth and metastasis as well as the glycolytic flux in cells. CONCLUSION: This paper elucidates that upregulation of PFKFB4 mediated by the KDM3A-SP1 axis promotes aerobic glycolysis in OS and augments tumor development.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Animais , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Glicólise/genética , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Camundongos , Camundongos Nus , Osteossarcoma/patologia , Fosfofrutoquinase-2/genética , Fosfofrutoquinase-2/metabolismo , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo
9.
FASEB J ; 35(6): e21664, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34042215

RESUMO

The pluripotency gene regulatory network of porcine induced pluripotent stem cells(piPSCs), especially in epigenetics, remains elusive. To determine the biological function of epigenetics, we cultured piPSCs in different culture conditions. We found that activation of pluripotent gene- and pluripotency-related pathways requires the erasure of H3K9 methylation modification which was further influenced by mouse embryonic fibroblast (MEF) served feeder. By dissecting the dynamic change of H3K9 methylation during loss of pluripotency, we demonstrated that the H3K9 demethylases KDM3A and KDM3B regulated global H3K9me2/me3 level and that their co-depletion led to the collapse of the pluripotency gene regulatory network. Immunoprecipitation-mass spectrometry (IP-MS) provided evidence that KDM3A and KDM3B formed a complex to perform H3K9 demethylation. The genome-wide regulation analysis revealed that OCT4 (O) and SOX2 (S), the core pluripotency transcriptional activators, maintained the pluripotent state of piPSCs depending on the H3K9 hypomethylation. Further investigation revealed that O/S cooperating with histone demethylase complex containing KDM3A and KDM3B promoted pluripotency genes expression to maintain the pluripotent state of piPSCs. Together, these data offer a unique insight into the epigenetic pluripotency network of piPSCs.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Células-Tronco Pluripotentes Induzidas/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Animais , Metilação de DNA , Epigênese Genética , Células-Tronco Pluripotentes Induzidas/citologia , Histona Desmetilases com o Domínio Jumonji/genética , Fator 3 de Transcrição de Octâmero/genética , Fatores de Transcrição SOXB1/genética , Suínos
10.
Mol Biol Rep ; 49(4): 3177-3185, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35113304

RESUMO

BACKGROUND: The epigenetic modifier molecule lysine demethylase 3A (KDM3A) has been shown to help ameliorate cardiovascular diseases, but its effect on cardiac fibroblasts (CFs) remains unclear. METHODS AND RESULTS: We designed gain- and loss-of-function experiments to investigate the biological functions of KDM3A in CFs. Moreover, we used SIS3-HCl (a specific inhibitor of p-Smad3) to explore the underlying mechanism. Cell viability and migration were verified by CCK-8 and cell migration experiments, respectively, and the degree of fibrosis was measured by Western blot analysis. Our data revealed that KDM3A enhanced the proliferation and migration of CFs and increased the fibroblast-to-myofibroblast transition while enabling the Smad3 phosphorylation response to transforming growth factor beta1 (TGFß1) stimulation. However, these effects were abolished by SIS3-HCl. Furthermore, KDM3A inhibition obviously protected against cardiac myofibroblast transdifferentiation under TGFß1 stimulation. CONCLUSIONS: KDM3A may act as a novel regulator of cardiac myofibroblast transdifferentiation through its ability to modulate the phosphorylation of Smad3 following TGFß1 stimulation.


Assuntos
Miofibroblastos , Fator de Crescimento Transformador beta1 , Transdiferenciação Celular , Fibroblastos/metabolismo , Lisina/metabolismo , Miofibroblastos/metabolismo , Fosforilação , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/farmacologia
11.
J Cell Mol Med ; 25(17): 8127-8139, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34350711

RESUMO

Pre-eclampsia (PE) is the leading cause of maternal death; however, the causative molecular basis remains largely unknown. Recent studies have revealed the important role microRNAs (miRNAs) play in PE. We aimed to explore the effects of let-7d on trophoblast proliferation, migration, invasion and apoptosis in PE and its underlying mechanism. Placental tissues were collected from PE patients and healthy pregnant women, and it was found that let-7d expression was increased, while KDM3A and ENO2 expression was decreased in PE tissues and cells. Bioinformatics analysis indicated the interaction among let-7d, KDM3A and ENO2, confirmed by dual luciferase reporter gene assay; ChIP experiment identified methylated modification to ENO2 by KDM3A. With gain- and loss-function method, silencing of let-7d increased KDM3A expression and enhanced the binding between KDM3A and ENO2. Furthermore, overexpression of let-7d suppressed cell proliferation, migration and invasion of trophoblasts, and induced apoptosis of trophoblasts, while these capacities were restored upon additional treatment of overexpressed ENO2. PE rat models were established to explore the effects of let-7d and ENO2 on PE in vivo. The results established that the silencing of let-7d alleviated the tissue injury and PE-related symptoms when reducing urine protein, TUNEL-positive cells and increasing ENO2, and KDM3A expression in rats. Cumulatively, let-7d suppressed cell progression of trophoblasts, and induced apoptosis through the down-regulation of KDM3A to promote ENO2 methylation, thereby promoting progression of PE. Such an epigenetic network of let-7d, KDM3A and ENO2 in the pathogenesis of PE might provide novel insight into targeted therapy against this disorder.


Assuntos
Histona Desmetilases com o Domínio Jumonji/metabolismo , MicroRNAs/fisiologia , Fosfopiruvato Hidratase/metabolismo , Pré-Eclâmpsia/metabolismo , Trofoblastos/metabolismo , Animais , Movimento Celular , Proliferação de Células , Feminino , Humanos , Gravidez , Ratos , Ratos Sprague-Dawley , Trofoblastos/patologia
12.
Proc Natl Acad Sci U S A ; 115(46): 11766-11771, 2018 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-30377265

RESUMO

Janus tyrosine kinase 2 (JAK2)-signal transducer and activator of transcription 3 (STAT3) signaling pathway is essential for modulating cellular development, differentiation, and homeostasis. Thus, dysregulation of JAK2-STAT3 signaling pathway is frequently associated with human malignancies. Here, we provide evidence that lysine-specific demethylase 3A (KDM3A) functions as an essential epigenetic enzyme for the activation of JAK2-STAT3 signaling pathway. KDM3A is tyrosine-phosphorylated by JAK2 in the nucleus and functions as a STAT3-dependent transcriptional coactivator. JAK2-KDM3A signaling cascade induced by IL-6 leads to alteration of histone H3K9 methylation as a predominant epigenetic event, thereby providing the functional and mechanistic link between activation of JAK2-STAT3 signaling pathway and its epigenetic control. Together, our findings demonstrate that inhibition of KDM3A phosphorylation could be a potent therapeutic strategy to control oncogenic effect of JAK2-STAT3 signaling pathway.


Assuntos
Histona Desmetilases com o Domínio Jumonji/metabolismo , Epigênese Genética , Células HEK293/metabolismo , Células HeLa , Histonas/metabolismo , Humanos , Interleucina-6/metabolismo , Janus Quinase 2/metabolismo , Histona Desmetilases com o Domínio Jumonji/genética , Fosforilação , Proteínas Tirosina Quinases/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Ativação Transcricional
13.
J Cell Mol Med ; 24(1): 1099-1115, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31755219

RESUMO

Myocardial infarction (MI) remains the leading cause of morbidity and mortality worldwide, and novel therapeutic targets still need to be investigated to alleviate myocardial injury and the ensuing maladaptive cardiac remodelling. Accumulating studies have indicated that lncRNA H19 might exert a crucial regulatory effect on cardiovascular disease. In this study, we aimed to explore the biological function and molecular mechanism of H19 in MI. To investigate the biological functions of H19, miRNA-22-3p and KDM3A, gain- and loss-of-function experiments were performed. In addition, bioinformatics analysis, dual-luciferase reporter assays, RNA immunoprecipitation (RIP) assays, RNA pull-down assays, quantitative RT-PCR and Western blot analyses as well as rescue experiments were conducted to reveal an underlying competitive endogenous RNA (ceRNA) mechanism. We found that H19 was significantly down-regulated after MI. Functionally, enforced H19 expression dramatically reduced infarct size, improved cardiac performance and alleviated cardiac fibrosis by mitigating myocardial apoptosis and decreasing inflammation. However, H19 knockdown resulted in the opposite effects. Bioinformatics analysis and dual-luciferase assays revealed that, mechanistically, miR-22-3p was a direct target of H19, which was also confirmed by RIP and RNA pull-down assays in primary cardiomyocytes. In addition, bioinformatics analysis and dual-luciferase reporter assays also demonstrated that miRNA-22-3p directly targeted the KDM3A gene. Moreover, subsequent rescue experiments further verified that H19 regulated the expression of KDM3A to ameliorate MI-induced myocardial injury in a miR-22-3p-dependent manner. The present study revealed the critical role of the lncRNAH19/miR-22-3p/KDM3A pathway in MI. These findings suggest that H19 may act as a potential biomarker and therapeutic target for MI.


Assuntos
Regulação da Expressão Gênica , Traumatismos Cardíacos/prevenção & controle , Histona Desmetilases/metabolismo , Inflamação/prevenção & controle , Infarto do Miocárdio/complicações , RNA Longo não Codificante/genética , Remodelação Ventricular , Animais , Apoptose , Movimento Celular , Proliferação de Células , Transição Epitelial-Mesenquimal , Traumatismos Cardíacos/etiologia , Traumatismos Cardíacos/metabolismo , Traumatismos Cardíacos/patologia , Histona Desmetilases/genética , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Masculino , MicroRNAs/genética , Infarto do Miocárdio/patologia , Ratos , Ratos Sprague-Dawley
14.
Biochem Biophys Res Commun ; 526(1): 176-183, 2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32201075

RESUMO

Autophagy is an essential process to maintain cell survival and homeostasis under various stress conditions. Here, we report that lysine-specific demethylase 3A (KDM3A) plays an important role in starvation-induced autophagy. Using Kdm3a knockout mice, we demonstrate that KDM3A is crucial for proper hepatic autophagy in vivo. Hepatic mRNA expression analysis and ChIP assay in WT and Kdm3a knockout mouse livers reveal that KDM3A activates autophagy genes by reducing histone H3K9me2 levels upon fasting. Together, our finding represents previously unidentified function of KDM3A as a key regulator of autophagy, implicating potential therapeutic approaches for autophagy-related diseases.


Assuntos
Autofagia , Histona Desmetilases com o Domínio Jumonji/metabolismo , Animais , Autofagossomos/metabolismo , Jejum , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Células Hep G2 , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Fígado/citologia , Fígado/metabolismo , Lisossomos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
15.
Development ; 144(20): 3674-3685, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-29042477

RESUMO

Neurog2 is a crucial regulator of neuronal fate specification and differentiation in vivo and in vitro However, it remains unclear how Neurog2 transactivates neuronal genes that are silenced by repressive chromatin. Here, we provide evidence that the histone H3 lysine 9 demethylase KDM3A facilitates the Xenopus Neurog2 (formerly known as Xngnr1) chromatin accessibility during neuronal transcription. Loss-of-function analyses reveal that KDM3A is not required for the transition of naive ectoderm to neural progenitor cells but is essential for primary neuron formation. ChIP series followed by qPCR analyses reveal that Neurog2 promotes the removal of the repressive H3K9me2 marks and addition of active histone marks, including H3K27ac and H3K4me3, at the NeuroD1 and Tubb2b promoters; this activity depends on the presence of KDM3A because Neurog2, via its C-terminal domain, interacts with KDM3A. Interestingly, KDM3A is dispensable for the neuronal transcription initiated by Ascl1, a proneural factor related to neurogenin in the bHLH family. In summary, our findings uncover a crucial role for histone H3K9 demethylation during Neurog2-mediated neuronal transcription and help in the understanding of the different activities of Neurog2 and Ascl1 in initiating neuronal development.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Cromatina/metabolismo , Histonas/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Ectoderma/metabolismo , Feminino , Lisina/química , Neurogênese , Neurônios/metabolismo , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Ativação Transcricional , Xenopus laevis
16.
Cell Mol Life Sci ; 74(18): 3305-3315, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28364192

RESUMO

Arginine methylation of histones is one mechanism of epigenetic regulation in eukaryotic cells. Methylarginines can also be found in non-histone proteins involved in various different processes in a cell. An enzyme family of nine protein arginine methyltransferases catalyses the addition of methyl groups on arginines of histone and non-histone proteins, resulting in either mono- or dimethylated-arginine residues. The reversibility of histone modifications is an essential feature of epigenetic regulation to respond to changes in environmental factors, signalling events, or metabolic alterations. Prominent histone modifications like lysine acetylation and lysine methylation are reversible. Enzyme family pairs have been identified, with each pair of lysine acetyltransferases/deacetylases and lysine methyltransferases/demethylases operating complementarily to generate or erase lysine modifications. Several analyses also indicate a reversible nature of arginine methylation, but the enzymes facilitating direct removal of methyl moieties from arginine residues in proteins have been discussed controversially. Differing reports have been seen for initially characterized putative candidates, like peptidyl arginine deiminase 4 or Jumonji-domain containing protein 6. Here, we review the most recent cellular, biochemical, and mass spectrometry work on arginine methylation and its reversible nature with a special focus on putative arginine demethylases, including the enzyme superfamily of Fe(II) and 2-oxoglutarate-dependent oxygenases.


Assuntos
Arginina/metabolismo , Histonas/metabolismo , Animais , Arginina/análogos & derivados , Arginina/análise , Histona Desmetilases/metabolismo , Humanos , Hidrolases/metabolismo , Metilação , Proteína-Arginina Desiminase do Tipo 4 , Desiminases de Arginina em Proteínas , Proteína-Arginina N-Metiltransferases/metabolismo
17.
Biochim Biophys Acta ; 1859(6): 792-804, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26877236

RESUMO

The metazoan-specific POU domain transcription factor family comprises activities underpinning developmental processes such as embryonic pluripotency and neuronal specification. Some POU family proteins efficiently bind an 8-bp DNA element known as the octamer motif. These proteins are known as Oct transcription factors. Oct1/POU2F1 is the only widely expressed POU factor. Unlike other POU factors it controls no specific developmental or organ system. Oct1 was originally described to operate at target genes associated with proliferation and immune modulation, but more recent results additionally identify targets associated with oxidative and cytotoxic stress resistance, metabolic regulation, stem cell function and other unexpected processes. Oct1 is pro-oncogenic in multiple contexts, and several recent reports provide broad evidence that Oct1 has prognostic and therapeutic value in multiple epithelial tumor settings. This review focuses on established and emerging roles of Oct1 in epithelial tumors, with an emphasis on mechanisms of transcription regulation by Oct1 that may underpin these findings. This article is part of a Special Issue entitled: The Oct Transcription Factor Family, edited by Dr. Dean Tantin.


Assuntos
Carcinogênese/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Epiteliais e Glandulares/genética , Fator 1 de Transcrição de Octâmero/genética , Sequência de Aminoácidos , Sítios de Ligação , Carcinogênese/metabolismo , Carcinogênese/patologia , Histonas/genética , Histonas/metabolismo , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Dados de Sequência Molecular , Neoplasias Epiteliais e Glandulares/metabolismo , Neoplasias Epiteliais e Glandulares/patologia , Fator 1 de Transcrição de Octâmero/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Transdução de Sinais
18.
Am J Med Genet A ; 170(11): 2916-2926, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27748065

RESUMO

The disruption of genes involved in epigenetic regulation is well known to cause Intellectual Disability (ID). We reported a custom microarray study that interrogated among others, the epigenetic regulatory gene-class, at single exon resolution. Here we elaborate on identified intragenic CNVs involving epigenetic regulatory genes; specifically discussing those in three genes previously unreported in ID etiology-ARID2, KDM3A, and ARID4B. The changes in ARID2 and KDM3A are likely pathogenic while the ARID4B variant is uncertain. Previously, we found a CNV involving only exon 6 of the JARID2 gene occurred apparently de novo in seven patients. JARID2 is known to cause ID and other neurodevelopmental conditions. However, exon 6 of this gene encodes one of a series of repeated motifs. We therefore, investigated the impact of this variant in two cohorts and present a genotype-phenotype assessment. We find the JARID2 exon 6 CNV is benign, with a high population frequency (>14%), but nevertheless could have a contributory effect. We also present results from an interrogation of the exomes of 2,044 patients with neurocognitive phenotypes for the incidence of potentially damaging mutation in the epigenetic regulatory gene-class. This paper provides a survey of the fine-scale CNV landscape for epigenetic regulatory genes in the context of ID, describing likely pathogenic as well as benign single exon imbalances. © 2016 Wiley Periodicals, Inc.


Assuntos
Variações do Número de Cópias de DNA , Epigênese Genética , Éxons , Regulação da Expressão Gênica , Deficiência Intelectual/genética , Adolescente , Criança , Pré-Escolar , Metilação de DNA , Feminino , Deleção de Genes , Duplicação Gênica , Estudos de Associação Genética , Genótipo , Humanos , Deficiência Intelectual/epidemiologia , Histona Desmetilases com o Domínio Jumonji/genética , Masculino , Mutação , Fenótipo , Complexo Repressor Polycomb 2/genética , Vigilância da População
19.
Biochem Biophys Res Commun ; 467(3): 556-61, 2015 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-26431874

RESUMO

Chemoresistance remains a major problem in the treatment of gastric cancer patients, leading to the serious limitation of efficacy of chemotherapeutic regime. However, the underlying mechanism remains largely unknown. In our present study, we for the first time found that knock down of KDM3A can promote apoptosis induced by chemoreagent Cisplatin and Paclitaxel through p53. Mechanistically, through promoting p53 binding to the promoter of PUMA. However, knock down of KDM3A as such doesn't affect p53 level. In addition, KDM3A can interact with p53K372me1 in protein-protein interaction fashion, leading to the inactivation of p53, may eventually leading to chemoresistance of gastric cancer.

20.
Anal Biochem ; 463: 54-60, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25010373

RESUMO

Covalent modifications, such as methylation and demethylation of lysine residues in histones, play important roles in chromatin dynamics and the regulation of gene expression. The lysine demethylases (KDMs) catalyze the demethylation of lysine residues on histone tails and are associated with diverse human diseases, including cancer, and are therefore proposed as targets for the therapeutic modulation of gene transcription. High-throughput assays have been developed to find inhibitors of KDMs, most of which are fluorescence-based assays. Here we report the development of a coupled scintillation proximity assay (SPA) for 3 KDMs: KDM1A (LSD1), KDM3A (JMJD1A), and KDM4A (JMJD2A). In this assay methylated peptides are first demethylated by a KDM, and a protein methyltransferase (PMT) is added to methylate the resulting peptide with tritiated S-(5'-adenosyl)-l-methionine. The enzyme activities were optimized and kinetic parameters were determined. These robust coupled assays are suitable for screening KDMs in 384-well format (Z' factors of 0.70-0.80), facilitating discovery of inhibitors in the quest for cancer therapeutics.


Assuntos
Ensaios Enzimáticos , Histona Desmetilases/metabolismo , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Histona Desmetilases/química , Humanos , Histona Desmetilases com o Domínio Jumonji/química , Histona Desmetilases com o Domínio Jumonji/metabolismo , Cinética , Lisina/metabolismo , Metilação , Ligação Proteica , Proteínas Metiltransferases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA