Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mol Med Rep ; 24(5)2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34542160

RESUMO

Acute respiratory distress syndrome (ARDS) is a deadly illness which presents with severe hypoxemia as well as diffuse alveolar damage. Jumonji domain­containing 3 (JMJD3), which belongs to the UTX/UTY JmjC­domain protein subfamily, is involved in infection, development, aging and immune disorders. However, the role of JMJD3 in acute lung injury (ALI) is still unclear. The present study explored the roles and potential mechanisms of JMJD3 in ALI. Alveolar epithelial cell­specific knockout of JMJD3 mice and A549 alveolar epithelial cells were used to investigate the function of JMJD3 in ALI. Lipopolysaccharide (LPS) was used to establish an in vivo and in vitro ALI model. The expression of JMJD3 in murine lung tissue and alveolar epithelial cells was detected. Pathological injury of lung tissue and alveolar epithelial cells was also investigated following inhibition of JMJD3. The results showed that JMJD3 expression was significantly increased in murine lung tissues and in A549 cells following LPS stimulation. JMJD3­deficient mice in alveolar epithelial cells exhibited alleviated lung pathological injury and ferroptosis following h stimulation. Mechanistically, it was found that JMJD3 knockout could increase the expression of nuclear factor erythroid­2­related factor­2 (Nrf2) in lung tissues challenged with h. However, Nrf2 overexpression by adenovirus could further enhance the anti­ferroptotic effect from JMJD3 silence in h­treated A549 cells. Taken together, the present study revealed that JMJD3 deficiency may relieve LPS­induced ALI by blocking alveolar epithelial ferroptosis in a Nrf2­dependent manner, which may serve as a novel therapeutic target against ALI.


Assuntos
Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/metabolismo , Ferroptose/genética , Histona Desmetilases com o Domínio Jumonji/deficiência , Lipopolissacarídeos/efeitos adversos , Fator 2 Relacionado a NF-E2/metabolismo , Alvéolos Pulmonares/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Biomarcadores , Modelos Animais de Doenças , Suscetibilidade a Doenças , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Alvéolos Pulmonares/patologia
2.
Biochem Biophys Res Commun ; 550: 77-83, 2021 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-33689883

RESUMO

Osteosarcoma (OS) is the most common type of bone tumor that seriously affects limb function and induces great pain in patients. Lung metastasis and chemotherapy resistance are two key issues leading to the poor prognosis of OS patients, therefore new treatment targets and strategies are urgently needed. In our study, we uncovered the role of histone demethylase KDM4A in regulating OS cell ferroptosis and tumor progression. KDM4A was significantly upregulated in OS specimens and high KDM4A expression was associated with poorer prognosis in OS patients. Our data indicated that targeting KDM4A significantly increased OS cell death, enhanced cisplatin response, and attenuated migration ability in vitro. KDM4A depletion dramatically inhibited tumor progression and lung metastasis of OS in vivo Further experiments confirmed that KDM4A knockdown promoted OS cell ferroptosis, a special non-apoptotic form of cell death. KDM4A regulates SLC7A11 transcription and OS cell ferroptosis by controlling H3K9me3 demethylation in the promoter region of SLC7A11. Our findings deepened the recognition of epigenetic regulatory mechanism in OS tumorigenesis, chemoresistance, and metastasis, suggesting that KDM4A activity may be a potential therapeutic target for future OS treatment.


Assuntos
Sistema y+ de Transporte de Aminoácidos/genética , Desmetilação , Ferroptose , Histonas/química , Histonas/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Osteossarcoma/metabolismo , Animais , Carcinogênese , Morte Celular , Linhagem Celular Tumoral , Movimento Celular , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Feminino , Ferroptose/genética , Técnicas de Silenciamento de Genes , Humanos , Histona Desmetilases com o Domínio Jumonji/deficiência , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundário , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Metástase Neoplásica/genética , Osteossarcoma/genética , Osteossarcoma/patologia , Prognóstico , Regulação para Cima
3.
Cell Death Differ ; 28(6): 1880-1899, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33462408

RESUMO

Kdm2a catalyzes H3K36me2 demethylation to play an intriguing epigenetic regulatory role in cell proliferation, differentiation, and apoptosis. Herein we found that myeloid-specific knockout of Kdm2a (LysM-Cre-Kdm2af/f, Kdm2a-/-) promoted macrophage M2 program by reprograming metabolic homeostasis through enhancing fatty acid uptake and lipolysis. Kdm2a-/- increased H3K36me2 levels at the Pparg locus along with augmented chromatin accessibility and Stat6 recruitment, which rendered macrophages with preferential M2 polarization. Therefore, the Kdm2a-/- mice were highly protected from high-fat diet (HFD)-induced obesity, insulin resistance, and hepatic steatosis, and featured by the reduced accumulation of adipose tissue macrophages and repressed chronic inflammation following HFD challenge. Particularly, Kdm2a-/- macrophages provided a microenvironment in favor of thermogenesis. Upon HFD or cold challenge, the Kdm2a-/- mice manifested higher capacity for inducing adipose browning and beiging to promote energy expenditure. Collectively, our findings demonstrate the importance of Kdm2a-mediated H3K36 demethylation in orchestrating macrophage polarization, providing novel insight that targeting Kdm2a in macrophages could be a viable therapeutic approach against obesity and insulin resistance.


Assuntos
Histona Desmetilases com o Domínio Jumonji/deficiência , Macrófagos/metabolismo , Obesidade/genética , PPAR gama/metabolismo , Animais , Dieta Hiperlipídica , Humanos , Masculino , Camundongos , Obesidade/patologia , Termogênese , Transfecção
4.
Cell Rep ; 31(8): 107668, 2020 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-32460011

RESUMO

The liver is a key regulator of systemic energy homeostasis whose proper function is dependent on the circadian clock. Here, we show that livers deficient in the oscillator component JARID1a exhibit a dysregulation of genes involved in energy metabolism. Importantly, we find that mice that lack hepatic JARID1a have decreased lean body mass, decreased respiratory exchange ratios, faster production of ketones, and increased glucose production in response to fasting. Finally, we find that JARID1a loss compromises the response of the hepatic transcriptome to nutrient availability. In all, ablation of hepatic JARID1a disrupts the coordination of hepatic metabolic programs with whole-body consequences.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Comportamento Alimentar/fisiologia , Histona Desmetilases com o Domínio Jumonji/metabolismo , Fígado/metabolismo , Adaptação Fisiológica , Animais , Ritmo Circadiano/fisiologia , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Humanos , Histona Desmetilases com o Domínio Jumonji/deficiência , Histona Desmetilases com o Domínio Jumonji/genética , Camundongos , Camundongos Knockout
5.
Gut ; 69(2): 243-251, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31085554

RESUMO

OBJECTIVE: Cancer-associated fibroblasts (CAFs), a major component of cancer stroma, can confer aggressive properties to cancer cells by secreting multiple factors. Their phenotypes are stably maintained, but the mechanisms are not fully understood. We aimed to show the critical role of epigenetic changes in CAFs in maintaining their tumour-promoting capacity and to show the validity of the epigenomic approach in identifying therapeutic targets from CAFs to starve cancer cells. DESIGN: Twelve pairs of primary gastric CAFs and their corresponding non-CAFs (NCAFs) were established from surgical specimens. Genome-wide DNA methylation and H3K27me3 analyses were conducted by BeadArray 450K and ChIP-on-Chip, respectively. Functions of potential a therapeutic target were analysed by inhibiting it, and prognostic impact was assessed in a database. RESULTS: CAFs had diverse and distinct DNA methylation and H3K27me3 patterns compared with NCAFs. Loss of H3K27me3, but not DNA methylation, in CAFs was enriched for genes involved in stem cell niche, cell growth, tissue development and stromal-epithelial interactions, such as WNT5A, GREM1, NOG and IGF2. Among these, we revealed that WNT5A, which had been considered to be derived from cancer cells, was highly expressed in cancer stromal fibroblasts, and was associated with poor prognosis. Inhibition of secreted WNT5A from CAFs suppressed cancer cell growth and migration. CONCLUSIONS: H3K27me3 plays a crucial role in defining tumour-promoting capacities of CAFs, and multiple stem cell niche factors were secreted from CAFs due to loss of H3K27me3. The validity of the epigenetic approach to uncover therapeutic targets for cancer-starving therapy was demonstrated.


Assuntos
Fibroblastos Associados a Câncer/metabolismo , Histona Desmetilases com o Domínio Jumonji/genética , Neoplasias Gástricas/genética , Meios de Cultivo Condicionados , Metilação de DNA , DNA de Neoplasias/genética , Epigenômica/métodos , Ontologia Genética , Estudo de Associação Genômica Ampla/métodos , Humanos , Histona Desmetilases com o Domínio Jumonji/deficiência , Mutação , Nicho de Células-Tronco , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Células Tumorais Cultivadas
6.
Cell Mol Immunol ; 17(8): 834-842, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-31197256

RESUMO

IL-6 plays important and pleiotropic roles in infection and inflammatory diseases, and its production needs to be tightly regulated. However, the epigenetic mechanism underlying Il6 gene transcription remains to be fully elucidated. Here, we report that lysine-specific demethylase 2b (KDM2B), which demethylates H3K4me3 and H3K36me2, is required in macrophages and dendritic cells for the induction of IL-6 but not TNF-α, IL-1, and IFN-ß. Compared to wild-type mice, KDM2B-deficient mice were more resistant to endotoxin shock and colitis, with a less severe inflammatory pathogenesis phenotype and decreased IL-6 production in sera. KDM2B selectively bound the Il6 promoter but did not alter histone demethylation; instead, KDM2B interacted with Brahma-related gene 1 (Brg1), the core ATPase subunit of SWI/SNF chromatin remodeling complexes, to facilitate chromatin accessibility of the Il6 promoter. Furthermore, KDM2B directly recruited RNA Polymerase II to further initiate and promote Il6 transcription. Thus, our finding identifies a novel nonclassical function of KDM2B in gene-specific transcription initiation and enhancement of Il6 independent of its demethylase activity and adds new insight into the specific epigenetic modification mechanism of inflammatory immune responses.


Assuntos
Montagem e Desmontagem da Cromatina , DNA Helicases , Proteínas F-Box , Inflamação , Interleucina-6 , Histona Desmetilases com o Domínio Jumonji , Proteínas Nucleares , Fatores de Transcrição , Humanos , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina/genética , DNA Helicases/metabolismo , Metilação de DNA/genética , Proteínas F-Box/metabolismo , Células HEK293 , Imunidade Inata , Inflamação/genética , Inflamação/imunologia , Interleucina-6/biossíntese , Histona Desmetilases com o Domínio Jumonji/deficiência , Histona Desmetilases com o Domínio Jumonji/metabolismo , Proteínas Nucleares/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica/genética , RNA Polimerase II/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional/genética , Animais , Camundongos
7.
Blood Adv ; 3(17): 2537-2549, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31471323

RESUMO

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.


Assuntos
Carcinogênese/química , Proteínas F-Box/fisiologia , Histona Desmetilases com o Domínio Jumonji/fisiologia , Leucemia de Células T/etiologia , Complexo Repressor Polycomb 1/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Ilhas de CpG , Proteínas F-Box/metabolismo , Histonas , Humanos , Histona Desmetilases com o Domínio Jumonji/deficiência , Histona Desmetilases com o Domínio Jumonji/metabolismo , Camundongos , Mutação , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , Domínios Proteicos , Receptor Notch1/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Ativação Transcricional , Dedos de Zinco
8.
Biochim Biophys Acta Mol Cell Res ; 1866(12): 118541, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31473257

RESUMO

Jumonji C (JmjC) domain-containing proteins have been shown to regulate cellular processes by hydroxylating or demethylating histone and non-histone targets. JMJD8 is a Jumonji C domain-containing protein localized in the lumen of the endoplasmic reticulum and was recently shown to be involved in endothelial differentiation and cellular inflammation response. However, other physiological functions of JMJD8 remain to be elucidated. In this research, we found that knockdown of JMJD8 in cancer cells significantly increased cell proliferation, and attenuated ionizing irradiation or etoposide treatment-induced DNA double-strand breaks (DSBs) level through enhancing the expression of Ku70 and Ku80 which are key participants in the non-homologous end-joining repair of DSBs. We also provided evidence to show that knockdown of JMJD8 up-regulated cyclooxygenase-2 (COX-2) expression which contributed to the enhanced expression of Ku70/Ku80 as shown by the results that pre-treatment of JMJD8 knockdown cells with COX-2 selective inhibitor NS-398 inhibited the induction of Ku70/Ku80. Furthermore, we identified that the up-regulation of COX-2 in JMJD8 knockdown cells was partially due to the increased activation of AKT/NF-κB signaling, and LY294002 (an inhibitor of the PI3K/AKT signaling pathway) repressed the induction of COX-2 and Ku70/Ku80. In conclusion, our research provided data to establish the role of JMJD8 in regulating tumor cell proliferation and their sensitivity to ionizing irradiation or chemo-therapy drug, and the AKT/NF-κB/COX-2 signaling mediated expression of Ku70/Ku80 was involved. The results of this research indicated that JMJD8 is a potential target for enhancing the efficacy of tumor radio- and chemo-therapies.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Autoantígeno Ku/biossíntese , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proliferação de Células , Humanos , Histona Desmetilases com o Domínio Jumonji/deficiência , Transdução de Sinais , Células Tumorais Cultivadas , Regulação para Cima
9.
Epigenomics ; 11(7): 751-766, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31172793

RESUMO

Aim: To investigate the function of Kdm2a in embryonic stem cells (ESCs). Materials & methods: Expression profile analysis after Kdm2a knockout. Analysis of Kdm2a, H3K4me3 and H3K27me3 ChIP-seq data in ESCs. qPCR analysis and ChIP-qPCR analysis of epigenetic changes after Kdm2a loss. Results:Kdm2a was dispensable for pluripotency maintenance in ESCs. Kdm2a genomic binding profile was positively correlated with that of H3K4me3, Zfx and Tet1. Kdm2a directly regulated germ cell genes in primordial germ cell-like cells. Kdm2a loss led to the reduced expression of endogenous retrovirus IAPEy and resulted in the gain of H3K36me2 and loss of H3K4me3 on IAPEy. Conclusion: Kdm2a regulates germ cell genes and endogenous retroviruses in ESCs possibly through demethylating H3K36me2 and influencing H3K4me3 deposition.


Assuntos
Retrovirus Endógenos/fisiologia , Histona Desmetilases com o Domínio Jumonji/metabolismo , Animais , Sítios de Ligação , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Corpos Embrioides/patologia , Regulação da Expressão Gênica , Histonas/metabolismo , Histona Desmetilases com o Domínio Jumonji/deficiência , Histona Desmetilases com o Domínio Jumonji/genética , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Embrionárias Murinas/virologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , RNA Guia de Cinetoplastídeos/metabolismo
10.
Nucleus ; 9(1): 431-441, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30059280

RESUMO

Aberrant levels of histone modifications lead to chromatin malfunctioning and consequently to various developmental defects and human diseases. Therefore, the proteins bearing the ability to modify histones have been extensively studied and the molecular mechanisms of their action are now fairly well understood. However, little attention has been paid to naturally occurring alternative isoforms of chromatin modifying proteins and to their biological roles. In this review, we focus on mammalian KDM2A and KDM2B, the only two lysine demethylases whose genes have been described to produce also an alternative isoform lacking the N-terminal demethylase domain. These short KDM2A/B-SF isoforms arise through alternative promoter usage and seem to play important roles in development and disease. We hypothesise about the biological significance of these alternative isoforms, which might represent a more common evolutionarily conserved regulatory mechanism.


Assuntos
Histona Desmetilases com o Domínio Jumonji/metabolismo , Neoplasias/enzimologia , Animais , Humanos , Isoenzimas/deficiência , Isoenzimas/genética , Isoenzimas/metabolismo , Histona Desmetilases com o Domínio Jumonji/deficiência , Histona Desmetilases com o Domínio Jumonji/genética , Neoplasias/metabolismo
11.
J Clin Invest ; 128(7): 3144-3159, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29911994

RESUMO

Jumonji D3 (JMJD3) histone demethylase epigenetically regulates development and differentiation, immunity, and tumorigenesis by demethylating a gene repression histone mark, H3K27-me3, but a role for JMJD3 in metabolic regulation has not been described. SIRT1 deacetylase maintains energy balance during fasting by directly activating both hepatic gluconeogenic and mitochondrial fatty acid ß-oxidation genes, but the underlying epigenetic and gene-specific mechanisms remain unclear. In this study, JMJD3 was identified unexpectedly as a gene-specific transcriptional partner of SIRT1 and epigenetically activated mitochondrial ß-oxidation, but not gluconeogenic, genes during fasting. Mechanistically, JMJD3, together with SIRT1 and the nuclear receptor PPARα, formed a positive autoregulatory loop upon fasting-activated PKA signaling and epigenetically activated ß-oxidation-promoting genes, including Fgf21, Cpt1a, and Mcad. Liver-specific downregulation of JMJD3 resulted in intrinsic defects in ß-oxidation, which contributed to hepatosteatosis as well as glucose and insulin intolerance. Remarkably, the lipid-lowering effects by JMJD3 or SIRT1 in diet-induced obese mice were mutually interdependent. JMJD3 histone demethylase may serve as an epigenetic drug target for obesity, hepatosteatosis, and type 2 diabetes that allows selective lowering of lipid levels without increasing glucose levels.


Assuntos
Jejum/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Mitocôndrias Hepáticas/metabolismo , Animais , Regulação para Baixo , Epigênese Genética , Ácidos Graxos/metabolismo , Fígado Gorduroso/etiologia , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Células Hep G2 , Hepatócitos/metabolismo , Código das Histonas , Humanos , Histona Desmetilases com o Domínio Jumonji/deficiência , Histona Desmetilases com o Domínio Jumonji/genética , Masculino , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Obesidade/genética , Obesidade/metabolismo , Oxirredução , PPAR alfa/metabolismo , Transdução de Sinais , Sirtuína 1/metabolismo
12.
Atherosclerosis ; 275: 156-165, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29908485

RESUMO

BACKGROUND AND AIMS: Atherosclerosis is a lipid-driven chronic inflammatory disorder of the arteries, and monocytes and macrophages play a central role in this process. Within the atherosclerotic lesion, macrophages can scavenge modified lipids and become the so-called foam cells. We previously reported that the epigenetic enzyme Kdm6b (also known as Jmjd3) controls the pro-fibrotic transcriptional profile of peritoneal foam cells. Given the importance of these cells in atherosclerosis, we now studied the effect of myeloid Kdm6b on disease progression. METHODS: Bone marrow of myeloid Kdm6b deficient (Kdm6bdel) mice or wild type littermates (Kdm6bwt) was transplanted to lethally irradiated Ldlr-/- mice fed a high fat diet for 9 weeks to induce atherosclerosis. RESULTS: Lesion size was similar in Kdm6bwt and Kdm6bdel transplanted mice. However, lesions of Kdm6bdel mice contained more collagen and were more necrotic. Pathway analysis on peritoneal foam cells showed that the pathway involved in leukocyte chemotaxis was most significantly upregulated. Although macrophage and neutrophil content was similar after 9 weeks of high fat diet feeding, the relative increase in collagen content and necrosis revealed that atherosclerotic lesions in Kdm6bdel mice progress faster. CONCLUSION: Myeloid Kdm6b deficiency results in more advanced atherosclerosis.


Assuntos
Aorta/enzimologia , Doenças da Aorta/enzimologia , Aterosclerose/enzimologia , Células Espumosas/enzimologia , Histona Desmetilases com o Domínio Jumonji/deficiência , Macrófagos Peritoneais/enzimologia , Placa Aterosclerótica , Animais , Aorta/patologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Aterosclerose/genética , Aterosclerose/patologia , Células Cultivadas , Quimiotaxia de Leucócito , Colágeno/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Progressão da Doença , Feminino , Fibrose , Células Espumosas/patologia , Histona Desmetilases com o Domínio Jumonji/genética , Macrófagos Peritoneais/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Necrose , Infiltração de Neutrófilos , Receptores de LDL/deficiência , Receptores de LDL/genética , Fatores de Tempo
13.
Cell Rep ; 23(2): 389-403, 2018 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-29641999

RESUMO

The arginine methylation status of histones dynamically changes during many cellular processes, including hematopoietic stem/progenitor cell (HSPC) development. The arginine methyltransferases and the readers that transduce the histone codes have been defined. However, whether arginine demethylation actively occurs in cells and what enzyme demethylates the methylarginine residues during various cellular processes are unknown. We report that JMJD1B, previously identified as a lysine demethylase for H3K9me2, mediates arginine demethylation of H4R3me2s and its intermediate, H4R3me1. We show that demethylation of H4R3me2s and H3K9me2s in promoter regions is correlated with active gene expression. Furthermore, knockout of JMJD1B blocks demethylation of H4R3me2s and/or H3K9me2 at distinct clusters of genes and impairs the activation of genes important for HSPC differentiation and development. Consequently, JMJD1B-/- mice show defects in hematopoiesis. Altogether, our study demonstrates that demethylase-mediated active arginine demethylation process exists in eukaryotes and that JMJD1B demethylates both H4R3me2s and H3K9me2 for epigenetic programming during hematopoiesis.


Assuntos
Histonas/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Animais , Desmetilação , Epigênese Genética , Feminino , Células HEK293 , Hematopoese , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Histonas/química , Humanos , Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Histona Desmetilases com o Domínio Jumonji/deficiência , Histona Desmetilases com o Domínio Jumonji/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas , Proteína-Arginina N-Metiltransferases/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Especificidade por Substrato
14.
Proc Natl Acad Sci U S A ; 115(16): E3741-E3748, 2018 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-29610306

RESUMO

Inactivation of the retinoblastoma gene (RB1) product, pRB, is common in many human cancers. Targeting downstream effectors of pRB that are central to tumorigenesis is a promising strategy to block the growth of tumors harboring loss-of-function RB1 mutations. One such effector is retinoblastoma-binding protein 2 (RBP2, also called JARID1A or KDM5A), which encodes an H3K4 demethylase. Binding of pRB to RBP2 has been linked to the ability of pRB to promote senescence and differentiation. Importantly, genetic ablation of RBP2 is sufficient to phenocopy pRB's ability to induce these cellular changes in cell culture experiments. Moreover, germline Rbp2 deletion significantly impedes tumorigenesis in Rb1+/- mice. The value of RBP2 as a therapeutic target in cancer, however, hinges on whether loss of RBP2 could block the growth of established tumors as opposed to simply delaying their onset. Here we show that conditional, systemic ablation of RBP2 in tumor-bearing Rb1+/- mice is sufficient to slow tumor growth and significantly extend survival without causing obvious toxicity to the host. These findings show that established Rb1-null tumors require RBP2 for growth and further credential RBP2 as a therapeutic target in human cancers driven by RB1 inactivation.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Código das Histonas/fisiologia , Histona Desmetilases com o Domínio Jumonji/fisiologia , Terapia de Alvo Molecular/métodos , Proteínas de Neoplasias/fisiologia , Neoplasias Hipofisárias/enzimologia , Proteína do Retinoblastoma/deficiência , Neoplasias da Glândula Tireoide/enzimologia , Alelos , Animais , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Ecocardiografia , Ativação Enzimática/efeitos dos fármacos , Fibroblastos , Genes do Retinoblastoma , Defeitos dos Septos Cardíacos/genética , Código das Histonas/efeitos dos fármacos , Integrases/efeitos dos fármacos , Histona Desmetilases com o Domínio Jumonji/deficiência , Histona Desmetilases com o Domínio Jumonji/genética , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/terapia , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Tamoxifeno/farmacologia , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/terapia , Transgenes/efeitos dos fármacos
15.
Stem Cell Reports ; 10(4): 1340-1354, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29526734

RESUMO

Histone H3 lysine 9 (H3K9) methylation is unevenly distributed in mammalian chromosomes. However, the molecular mechanism controlling the uneven distribution and its biological significance remain to be elucidated. Here, we show that JMJD1A and JMJD1B preferentially target H3K9 demethylation of gene-dense regions of chromosomes, thereby establishing an H3K9 hypomethylation state in euchromatin. JMJD1A/JMJD1B-deficient embryos died soon after implantation accompanying epiblast cell death. Furthermore, combined loss of JMJD1A and JMJD1B caused perturbed expression of metabolic genes and rapid cell death in embryonic stem cells (ESCs). These results indicate that JMJD1A/JMJD1B-meditated H3K9 demethylation has critical roles for early embryogenesis and ESC maintenance. Finally, genetic rescue experiments clarified that H3K9 overmethylation by G9A was the cause of the cell death and perturbed gene expression of JMJD1A/JMJD1B-depleted ESCs. We summarized that JMJD1A and JMJD1B, in combination, ensure early embryogenesis and ESC viability by establishing the correct H3K9 methylated epigenome.


Assuntos
Desmetilação , Desenvolvimento Embrionário , Histonas/metabolismo , Histona Desmetilases com o Domínio Jumonji/deficiência , Lisina/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Animais , Pontos de Checagem do Ciclo Celular , Linhagem Celular , Sobrevivência Celular , Cromossomos de Mamíferos/metabolismo , Regulação para Baixo/genética , Regulação da Expressão Gênica no Desenvolvimento , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Metilação , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Mutação/genética
16.
PLoS Genet ; 13(9): e1007034, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28949961

RESUMO

Histone H3 lysine 9 (H3K9) methylation is a hallmark of heterochromatin. H3K9 demethylation is crucial in mouse sex determination; The H3K9 demethylase Jmjd1a deficiency leads to increased H3K9 methylation at the Sry locus in embryonic gonads, thereby compromising Sry expression and causing male-to-female sex reversal. We hypothesized that the H3K9 methylation level at the Sry locus is finely tuned by the balance in activities between the H3K9 demethylase Jmjd1a and an unidentified H3K9 methyltransferase to ensure correct Sry expression. Here we identified the GLP/G9a H3K9 methyltransferase complex as the enzyme catalyzing H3K9 methylation at the Sry locus. Based on this finding, we tried to rescue the sex-reversal phenotype of Jmjd1a-deficient mice by modulating GLP/G9a complex activity. A heterozygous GLP mutation rescued the sex-reversal phenotype of Jmjd1a-deficient mice by restoring Sry expression. The administration of a chemical inhibitor of GLP/G9a enzyme into Jmjd1a-deficient embryos also successfully rescued sex reversal. Our study not only reveals the molecular mechanism underlying the tuning of Sry expression but also provides proof on the principle of therapeutic strategies based on the pharmacological modulation of epigenetic balance.


Assuntos
Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Histona Desmetilases com o Domínio Jumonji/genética , Proteína da Região Y Determinante do Sexo/metabolismo , Desenvolvimento Sexual/genética , Animais , Feminino , Regulação da Expressão Gênica , Loci Gênicos , Gônadas/embriologia , Gônadas/metabolismo , Histona-Lisina N-Metiltransferase/genética , Histonas/genética , Histona Desmetilases com o Domínio Jumonji/deficiência , Histona Desmetilases com o Domínio Jumonji/metabolismo , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Análise de Sequência de DNA , Proteína da Região Y Determinante do Sexo/genética
17.
Biochim Biophys Acta Mol Basis Dis ; 1863(7): 1709-1717, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28501567

RESUMO

Differentiation of adipocytes is a highly regulated process modulated by multiple transcriptional co-activators and co-repressors. JMJD1C belongs to the family of jumonji C (jmjC) domain-containing histone demethylases and was originally described as a ligand-dependent co-activator of thyroid hormone and androgen receptors. Here, we explored the potential role of Jmjd1c in white adipocyte differentiation. To investigate the relevance of Jmjd1c in adipogenesis, murine 3T3-L1 preadipocyte cells with transient knock-down of Jmjd1c (3T3_Jmjd1c) were generated. Depletion of Jmjd1c led to the formation of smaller lipid droplets, reduced accumulation of triglycerides and maintenance of a more fibroblast-like morphology after adipocyte differentiation. Concomitantly, insulin stimulated uptake of glucose and fatty acids was significantly reduced in 3T3_Jmjd1c adipocytes. In line with these observations we detected lower expression of key genes associated with lipid droplet formation (Plin1, Plin4, Cidea) and uptake of glucose and fatty acids (Glut4, Fatp1, Fatp4, Aqp7) respectively. Finally, we demonstrate that depletion of Jmjd1c interferes with mitotic clonal expansion (MCE), increases levels of H3K9me2 (dimethylation of lysine 9 of histone H3) at promotor regions of adipogenic transcription factors (C/EBPs and PPARγ) and leads to reduced induction of these key regulators. In conclusion, we have identified Jmjd1c as a modulator of adipogenesis. Our data suggest that Jmjd1c may participate in MCE and the activation of the adipogenic transcription program during the induction phase of adipocyte differentiation in 3T3-L1 cells.


Assuntos
Adipócitos/metabolismo , Adipogenia , Diferenciação Celular , Fibroblastos/metabolismo , Histona Desmetilases com o Domínio Jumonji/deficiência , Gotículas Lipídicas/metabolismo , Células 3T3-L1 , Adipócitos/citologia , Animais , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Ácidos Graxos/genética , Ácidos Graxos/metabolismo , Fibroblastos/citologia , Glucose/genética , Glucose/metabolismo , Histonas/genética , Histonas/metabolismo , Camundongos , Mitose , PPAR gama/genética , PPAR gama/metabolismo , Regiões Promotoras Genéticas
18.
Oncogene ; 35(19): 2441-52, 2016 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-26279298

RESUMO

The histone demethylase JMJD1A, which controls gene expression by epigenetic regulation of H3K9 methylation marks, functions in diverse activities, including spermatogenesis, metabolism and stem cell self-renewal and differentiation. Here, we found that JMJD1A knockdown in prostate cancer cells antagonizes their proliferation and survival. Profiling array analyses revealed that JMJD1A-dependent genes function in cellular growth, proliferation and survival, and implicated that the c-Myc transcriptional network is deregulated following JMJD1A inhibition. Biochemical analyses confirmed that JMJD1A enhances c-Myc transcriptional activity by upregulating c-Myc expression levels. Mechanistically, JMJD1A activity promoted recruitment of androgen receptor (AR) to the c-Myc gene enhancer and induced H3K9 demethylation, increasing AR-dependent transcription of c-Myc mRNA. In parallel, we found that JMJD1A regulated c-Myc stability, likely by inhibiting HUWE1, an E3 ubiquitin ligase known to target degradation of several substrates including c-Myc. JMJD1A (wild type or mutant lacking histone demethylase activity) bound to HUWE1, attenuated HUWE1-dependent ubiquitination and subsequent degradation of c-Myc, increasing c-Myc protein levels. Furthermore, c-Myc knockdown in prostate cancer cells phenocopied effects of JMJD1A knockdown, and c-Myc re-expression in JMJD1A-knockdown cells partially rescued prostate cancer cell growth in vitro and in vivo. c-Myc protein levels were positively correlated with those of JMJD1A in a subset of human prostate cancer specimens. Collectively, our findings identify a critical role for JMJD1A in regulating proliferation and survival of prostate cancer cells by controlling c-Myc expression at transcriptional and post-translational levels.


Assuntos
Histona Desmetilases com o Domínio Jumonji/metabolismo , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Carcinogênese , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Elementos Facilitadores Genéticos/genética , Técnicas de Silenciamento de Genes , Humanos , Histona Desmetilases com o Domínio Jumonji/deficiência , Histona Desmetilases com o Domínio Jumonji/genética , Masculino , Estabilidade Proteica , Transporte Proteico , Proteólise , Proteínas Proto-Oncogênicas c-myc/deficiência , Proteínas Proto-Oncogênicas c-myc/genética , Receptores Androgênicos/metabolismo , Transcrição Gênica , Proteínas Supressoras de Tumor , Ubiquitina-Proteína Ligases/antagonistas & inibidores
19.
Cell Tissue Res ; 363(3): 723-33, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26334721

RESUMO

Genetic studies have shown that aberrant activation of p53 signaling leads to embryonic lethality. Maintenance of a fine balance of the p53 protein level is critical for normal development. Previously, we have reported that Jmjd5, a member of the Jumonji C (JmjC) family, regulates embryonic cell proliferation through the control of Cdkn1a expression. Since Cdkn1a is the representative p53-regulated gene, we have examined whether the expression of other p53 target genes is coincidentally upregulated with Cdkn1a in Jmjd5-deficient embryos. The expression of a subset of p53-regulated genes was increased in both Jmjd5 hypomorphic mouse embryonic fibroblasts (MEFs) and Jmjd5-deficient embryos at embryonic day 8.25 without the induced expression of Trp53. Intercrossing of Jmjd5-deficient mice with Trp53 knockout mice showed that the growth defect of Jmjd5 mutant cells was significantly recovered under a Trp53 null genetic background. Chromatin immunoprecipitation analysis in Jmjd5 hypomorphic MEFs indicated the increased recruitment of p53 at several p53 target gene loci, such as Cdkn1a, Pmaip1, and Mdm2. These results suggest that Jmjd5 is involved in the transcriptional regulation of a subset of p53-regulated genes, possibly through the control of p53 recruitment at the gene loci. In Jmjd5-deficient embryos, the enhanced recruitment of p53 might result in the abnormal activation of p53 signaling leading to embryonic lethality.


Assuntos
Desenvolvimento Embrionário , Histona Desmetilases com o Domínio Jumonji/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Animais , Proliferação de Células , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário/genética , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Loci Gênicos , Humanos , Histona Desmetilases com o Domínio Jumonji/deficiência , Histona Desmetilases com o Domínio Jumonji/genética , Camundongos , Fenótipo , Transdução de Sinais/genética , Regulação para Cima/genética
20.
Mech Dev ; 133: 36-53, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25016215

RESUMO

Post-translational modification of histones plays essential roles in the transcriptional regulation of genes in eukaryotes. Methylation on basic residues of histones is regulated by histone methyltransferases and histone demethylases, and misregulation of these enzymes has been linked to a range of diseases such as cancer. Histone lysine demethylase 2 (KDM2) family proteins have been shown to either promote or suppress tumorigenesis in different human malignancies. However, the roles and regulation of KDM2 in development are poorly understood, and the exact roles of KDM2 in regulating demethylation remain controversial. Since KDM2 proteins are highly conserved in multicellular animals, we analyzed the KDM2 ortholog in Drosophila. We have observed that dKDM2 is a nuclear protein and its level fluctuates during fly development. We generated three deficiency lines that disrupt the dKdm2 locus, and together with 10 transposon insertion lines within the dKdm2 locus, we characterized the developmental defects of these alleles. The alleles of dKdm2 define three phenotypic classes, and the intragenic complementation observed among these alleles and our subsequent analyses suggest that dKDM2 is not required for viability. In addition, loss of dKDM2 appears to have rather weak effects on histone H3 lysine 36 and 4 methylation (H3K36me and H3K4me) in the third instar wandering larvae, and we observed no effect on methylation of H3K9me2, H3K27me2 and H3K27me3 in dKdm2 mutants. Taken together, these genetic, molecular and biochemical analyses suggest that dKDM2 is not required for viability of flies, indicating that dKdm2 is likely redundant with other histone lysine demethylases in regulating normal development in Drosophila.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/enzimologia , Drosophila melanogaster/genética , Histona Desmetilases com o Domínio Jumonji/genética , Animais , Animais Geneticamente Modificados , Deleção Cromossômica , Sequência Conservada , Elementos de DNA Transponíveis , Proteínas de Drosophila/química , Proteínas de Drosophila/deficiência , Drosophila melanogaster/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Técnicas de Inativação de Genes , Genes de Insetos , Teste de Complementação Genética , Humanos , Histona Desmetilases com o Domínio Jumonji/química , Histona Desmetilases com o Domínio Jumonji/deficiência , Mutação , Filogenia , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA