Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 61(6): 859-73, 2016 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-26990989

RESUMO

Dysregulation of MLL complex-mediated histone methylation plays a pivotal role in gene expression associated with diseases, but little is known about cellular factors modulating MLL complex activity. Here, we report that SON, previously known as an RNA splicing factor, controls MLL complex-mediated transcriptional initiation. SON binds to DNA near transcription start sites, interacts with menin, and inhibits MLL complex assembly, resulting in decreased H3K4me3 and transcriptional repression. Importantly, alternatively spliced short isoforms of SON are markedly upregulated in acute myeloid leukemia. The short isoforms compete with full-length SON for chromatin occupancy but lack the menin-binding ability, thereby antagonizing full-length SON function in transcriptional repression while not impairing full-length SON-mediated RNA splicing. Furthermore, overexpression of a short isoform of SON enhances replating potential of hematopoietic progenitors. Our findings define SON as a fine-tuner of the MLL-menin interaction and reveal short SON overexpression as a marker indicating aberrant transcriptional initiation in leukemia.


Assuntos
Proteínas de Ligação a DNA/genética , Histona-Lisina N-Metiltransferase/biossíntese , Leucemia Mieloide Aguda/genética , Proteína de Leucina Linfoide-Mieloide/biossíntese , Proteínas Proto-Oncogênicas/genética , Transcrição Gênica , Processamento Alternativo/genética , Linhagem Celular Tumoral , Cromatina/genética , Proteínas de Ligação a DNA/biossíntese , Regulação Leucêmica da Expressão Gênica , Histona-Lisina N-Metiltransferase/genética , Humanos , Leucemia Mieloide Aguda/patologia , Metilação , Antígenos de Histocompatibilidade Menor , Proteína de Leucina Linfoide-Mieloide/genética , Ligação Proteica , Isoformas de Proteínas/genética , Proteínas Proto-Oncogênicas/metabolismo
2.
J Biol Chem ; 297(5): 101250, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34592314

RESUMO

Loss of functional pancreatic ß-cell mass leads to type 2 diabetes (T2D), attributable to modified ß-cell-dependent adaptive gene expression patterns. SetD7 is a histone methyltransferase enriched in pancreatic islets that mono- and dimethylates histone-3-lysine-4 (H3K4), promoting euchromatin modifications, and also maintains the regulation of key ß-cell function and survival genes. However, the transcriptional regulation of this important epigenetic modifier is unresolved. Here we identified the nuclear hormone receptor peroxisome proliferator-activated receptor-gamma (PPARγ) as a major transcriptional regulator of SetD7 and provide evidence for direct binding and functionality of PPARγ in the SetD7 promoter region. Furthermore, constitutive shRNA-mediated PPARγ knockdown in INS-1 ß-cells or pancreas-specific PPARγ deletion in mice led to downregulation of SetD7 expression as well as its nuclear enrichment. The relevance of the SetD7-PPARγ interaction in ß-cell adaptation was tested in normoglycemic 60% partial pancreatectomy (Px) and hyperglycemic 90% Px rat models. Whereas a synergistic increase in islet PPARγ and SetD7 expression was observed upon glycemic adaptation post-60% Px, in hyperglycemic 90% Px rats, islet PPARγ, and PPARγ targets SetD7 and Pdx1 were downregulated. PPARγ agonist pioglitazone treatment in 90% Px rats partially restored glucose homeostasis and ß-cell mass and enhanced expression of SetD7 and Pdx1. Collectively, these data provide evidence that the SetD7-PPARγ interaction serves as an important element of the adaptive ß-cell response.


Assuntos
Regulação Enzimológica da Expressão Gênica , Histona-Lisina N-Metiltransferase/biossíntese , Hiperglicemia/metabolismo , Células Secretoras de Insulina/metabolismo , PPAR gama/metabolismo , Elementos de Resposta , Animais , Linhagem Celular , Histona-Lisina N-Metiltransferase/genética , Hiperglicemia/genética , Camundongos , Camundongos Transgênicos , PPAR gama/genética , Ratos
3.
Blood ; 135(25): 2271-2285, 2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32202636

RESUMO

SETD2, the histone H3 lysine 36 methyltransferase, previously identified by us, plays an important role in the pathogenesis of hematologic malignancies, but its role in myelodysplastic syndromes (MDSs) has been unclear. In this study, low expression of SETD2 correlated with shortened survival in patients with MDS, and the SETD2 levels in CD34+ bone marrow cells of those patients were increased by decitabine. We knocked out Setd2 in NUP98-HOXD13 (NHD13) transgenic mice, which phenocopies human MDS, and found that loss of Setd2 accelerated the transformation of MDS into acute myeloid leukemia (AML). Loss of Setd2 enhanced the ability of NHD13+ hematopoietic stem and progenitor cells (HSPCs) to self-renew, with increased symmetric self-renewal division and decreased differentiation and cell death. The growth of MDS-associated leukemia cells was inhibited though increasing the H3K36me3 level by using epigenetic modifying drugs. Furthermore, Setd2 deficiency upregulated hematopoietic stem cell signaling and downregulated myeloid differentiation pathways in the NHD13+ HSPCs. Our RNA-seq and chromatin immunoprecipitation-seq analysis indicated that S100a9, the S100 calcium-binding protein, is a target gene of Setd2 and that the addition of recombinant S100a9 weakens the effect of Setd2 deficiency in the NHD13+ HSPCs. In contrast, downregulation of S100a9 leads to decreases of its downstream targets, including Ikba and Jnk, which influence the self-renewal and differentiation of HSPCs. Therefore, our results demonstrated that SETD2 deficiency predicts poor prognosis in MDS and promotes the transformation of MDS into AML, which provides a potential therapeutic target for MDS-associated acute leukemia.


Assuntos
Anemia Refratária com Excesso de Blastos/patologia , Calgranulina B/fisiologia , Histona-Lisina N-Metiltransferase/deficiência , Histona-Lisina N-Metiltransferase/fisiologia , Leucemia Mieloide Aguda/etiologia , Anemia Refratária com Excesso de Blastos/genética , Anemia Refratária com Excesso de Blastos/metabolismo , Animais , Calgranulina B/biossíntese , Calgranulina B/genética , Transformação Celular Neoplásica , Células Cultivadas , Decitabina/farmacologia , Regulação para Baixo , Regulação Leucêmica da Expressão Gênica , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/patologia , Código das Histonas/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/biossíntese , Histona-Lisina N-Metiltransferase/genética , Proteínas de Homeodomínio/genética , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/mortalidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Síndromes Mielodisplásicas/patologia , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Proteínas de Fusão Oncogênica/genética , Prognóstico , Proteínas Recombinantes/uso terapêutico , Fatores de Tempo , Análise Serial de Tecidos , Transcriptoma
4.
Biomarkers ; 25(3): 268-273, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32091270

RESUMO

Purpose: To assess the prognostic significance of the nuclear receptor binding SET protein 2 (NSD2), a co-activator of the NFkB-pathway, on tumour progression in patients with advanced prostate cancer (PCa).Methods: We retrospectively assessed NSD2 expression in 53 patients with metastatic and castration-resistant PCa. Immunohistochemical staining for NSD2 was carried out on specimen obtained from palliative resection of the prostate. Univariable and multivariable analyses were performed to assess the association between NSD2 expression and PCa progression.Results: Of the 53 patients, 41 had castration-resistant PCa and 48 men had metastases at time of tissue acquisition. NSD2 expression was increased in tumour specimen from 42 patients (79.2%). In univariable Cox regression analyses, NSD2 expression was associated with PSA progression, progression on imaging and overall survival (p = 0.04, respectively). In multivariable analyses, NSD2 expression did not retain its association with these endpoints.Conclusions: NSD2 expression is abnormal in almost 80% of patients with advanced PCa. Expression levels of this epigenetic regulator are easily detected by immunohistochemistry while this biomarker exhibited prognostic value for PCa progression and death in univariable analysis. Further studies on NSD2 involvement in PCa proliferation, progression, metastasis and resistance mechanisms are needed.


Assuntos
Biomarcadores Tumorais/biossíntese , Histona-Lisina N-Metiltransferase/biossíntese , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , Proteínas Repressoras/biossíntese , Idoso , Idoso de 80 Anos ou mais , Progressão da Doença , Humanos , Imuno-Histoquímica/estatística & dados numéricos , Masculino , Prognóstico , Modelos de Riscos Proporcionais , Próstata/patologia , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/terapia , Estudos Retrospectivos , Análise de Sobrevida
5.
Acta Biochim Biophys Sin (Shanghai) ; 52(5): 506-516, 2020 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-32369110

RESUMO

Hyperglycemia-mediated reactive oxygen species (ROS) accumulation plays an important role in hyperglycemia-induced endothelial injury. Kelch-like ECH-associated protein 1 (Keap1)/nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway inhibition participates in hyperglycemia-induced ROS accumulation. Our previous study indicated that SET8 overexpression inhibits high glucose-mediated ROS accumulation in human umbilical vein endothelial cells (HUVECs). In the present study, we hypothesize that SET8 may play a major role in high glucose-induced ROS accumulation via modulation of Keap1/Nrf2/ARE pathway. Our data indicated that high glucose mediated cell viability reduction, ROS accumulation, and Nrf2/ARE signal pathway inhibition via upregulation of Keap1 expression in HUVECs. Moreover, high glucose inhibited the expressions of SET8 and H4K20me1 (a downstream target of SET8). SET8 overexpression improved high glucose-mediated Keap1/Nrf2/ARE pathway inhibition and endothelial oxidation. Consistently, the effects of sh-SET8 were similar to that of high glucose treatment and were reversed by si-Keap1. A mechanistic study found that H4K20me1 was enriched at the Keap1 promoter region. SET8 overexpression attenuated Keap1 promoter activity and its expression, while mutant SET8 R259G did not affect Keap1 promoter activity and expression. The results of this study demonstrated that SET8 negatively regulates Keap1 expression, thus participating in high glucose-mediated Nrf2/ARE signal pathway inhibition and oxidative injury in HUVECs.


Assuntos
Elementos de Resposta Antioxidante , Regulação para Baixo/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Histona-Lisina N-Metiltransferase/biossíntese , Células Endoteliais da Veia Umbilical Humana/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/genética , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Hiperglicemia/genética , Hiperglicemia/metabolismo , Hiperglicemia/patologia , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Fator 2 Relacionado a NF-E2/genética , Transdução de Sinais/genética
6.
J Neurosci ; 38(4): 803-813, 2018 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-29217682

RESUMO

Repeated exposure to cocaine induces lasting epigenetic changes in neurons that promote the development and persistence of addiction. One epigenetic alteration involves reductions in levels of the histone dimethyltransferase G9a in nucleus accumbens (NAc) after chronic cocaine administration. This reduction in G9a may enhance cocaine reward because overexpressing G9a in the NAc decreases cocaine-conditioned place preference. Therefore, we hypothesized that HSV-mediated G9a overexpression in the NAc shell (NAcSh) would attenuate cocaine self-administration (SA) and cocaine-seeking behavior. Instead, we found that G9a overexpression, and the resulting increase in histone 3 lysine 9 dimethylation (H3K9me2), increases sensitivity to cocaine reinforcement and enhances motivation for cocaine in self-administering male rats. Moreover, when G9a overexpression is limited to the initial 15 d of cocaine SA training, it produces an enduring postexpression enhancement in cocaine SA and prolonged (over 5 weeks) increases in reinstatement of cocaine seeking induced by foot-shock stress, but in the absence of continued global elevations in H3K9me2. The increase in stress-induced reinstatement is paralleled by heightened anxiety measures, suggesting that countering the cocaine-induced decreases in endogenous G9a with ectopic G9a overexpression leads to lasting anxiogenic effects. Finally, we found an enduring reduction in phosphorylated cAMP-response element binding protein levels in the NAcSh that could account for the increased anxiety. These data demonstrate a novel role for G9a in promoting comorbid cocaine addiction and anxiety and suggest that increased epigenetic repression of transcription through H3K9 during cocaine use can have long-lasting and unexpected negative consequences on behavior.SIGNIFICANCE STATEMENT Cocaine addiction is a neuropsychiatric disorder that is detrimental to society and currently has no effective treatments. The difficulty in treating drug addiction is compounded by the high comorbidity with other psychiatric illnesses, including anxiety disorders. Here, we demonstrate that G9a, an epigenetic repressor of gene expression, acting in the nucleus accumbens, a brain reward region, is capable of increasing both addiction- and anxiety-like behaviors in rats. These findings are intriguing because repeated cocaine exposure decreases G9a in this region and thereby enhances expression of certain addiction-promoting genes. However, our results suggest that countering this cocaine-induced decrease in G9a activity actually exacerbates addiction and sensitivity to relapse under stressful situations.


Assuntos
Transtornos Relacionados ao Uso de Cocaína/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/biossíntese , Núcleo Accumbens/metabolismo , Animais , Ansiedade/etiologia , Ansiedade/metabolismo , Cocaína/farmacologia , Condicionamento Operante , Inibidores da Captação de Dopamina/farmacologia , Comportamento de Procura de Droga/fisiologia , Epigênese Genética/fisiologia , Extinção Psicológica , Regulação da Expressão Gênica/fisiologia , Histonas/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Autoadministração
7.
Dev Biol ; 434(1): 149-163, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29258872

RESUMO

Amacrine interneurons play a critical role in the processing of visual signals within the retina. They are highly diverse, representing 30 or more distinct subtypes. Little is known about how amacrine subtypes acquire their unique gene expression and morphological features. We characterized the gene expression pattern of the zinc-finger transcription factor Prdm13 in the mouse. Consistent with a developmental role, Prdm13 was expressed by Ptf1a+ amacrine and horizontal precursors. Over time, Prdm13 expression diverged from the transiently expressed Ptf1a and marked just a subset of amacrine cells in the adult retina. While heterogeneous, we show that most of these Prdm13+ amacrine cells express the transcription factor Ebf3 and the calcium binding protein calretinin. Loss of Prdm13 did not affect the number of amacrine cells formed during development. However, we observed a modest loss of amacrine cells and increased apoptosis that correlated with the onset timing of Ebf3 expression. Adult Prdm13 loss-of-function mice had 25% fewer amacrine cells, altered calretinin expression, and a lack of Ebf3+ amacrines. Forcing Prdm13 expression in retinal progenitor cells did not significantly increase amacrine cell formation, Ebf3 or calretinin expression, and appeared detrimental to the survival of photoreceptors. Our data show that Prdm13 is not required for amacrine fate as a class, but is essential for the formation of Ebf3+ amacrine cell subtypes. Rather than driving subtype identity, Prdm13 may act by restricting competing fate programs to maintain identity and survival.


Assuntos
Células Amácrinas/metabolismo , Apoptose/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Histona-Lisina N-Metiltransferase/biossíntese , Células-Tronco/metabolismo , Fatores de Transcrição/biossíntese , Células Amácrinas/citologia , Animais , Calbindina 2/biossíntese , Calbindina 2/genética , Sobrevivência Celular/fisiologia , Histona-Lisina N-Metiltransferase/genética , Camundongos , Camundongos Transgênicos , Células-Tronco/citologia , Fatores de Transcrição/genética
8.
Genome Res ; 26(8): 1057-68, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27225844

RESUMO

The three-dimensional packing of the genome plays an important role in regulating gene expression. We have used Hi-C, a genome-wide chromatin conformation capture (3C) method, to analyze Arabidopsis thaliana chromosomes dissected into subkilobase segments, which is required for gene-level resolution in this species with a gene-dense genome. We found that the repressive H3K27me3 histone mark is overrepresented in the promoter regions of genes that are in conformational linkage over long distances. In line with the globally dispersed distribution of RNA polymerase II in A. thaliana nuclear space, actively transcribed genes do not show a strong tendency to associate with each other. In general, there are often contacts between 5' and 3' ends of genes, forming local chromatin loops. Such self-loop structures of genes are more likely to occur in more highly expressed genes, although they can also be found in silent genes. Silent genes with local chromatin loops are highly enriched for the histone variant H3.3 at their 5' and 3' ends but depleted of repressive marks such as heterochromatic histone modifications and DNA methylation in flanking regions. Our results suggest that, different from animals, a major theme of genome folding in A. thaliana is the formation of structural units that correspond to gene bodies.


Assuntos
Arabidopsis/genética , Cromatina/genética , Metilação de DNA/genética , Histona-Lisina N-Metiltransferase/genética , Animais , Arabidopsis/crescimento & desenvolvimento , Cromossomos de Plantas , Regulação da Expressão Gênica de Plantas , Genoma de Planta , Histona-Lisina N-Metiltransferase/biossíntese , Proteínas de Plantas/genética
9.
Genome Res ; 26(8): 1034-46, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27034506

RESUMO

For a long time, it has been assumed that the only role of sperm at fertilization is to introduce the male genome into the egg. Recently, ideas have emerged that the epigenetic state of the sperm nucleus could influence transcription in the embryo. However, conflicting reports have challenged the existence of epigenetic marks on sperm genes, and there are no functional tests supporting the role of sperm epigenetic marking on embryonic gene expression. Here, we show that sperm is epigenetically programmed to regulate embryonic gene expression. By comparing the development of sperm- and spermatid-derived frog embryos, we show that the programming of sperm for successful development relates to its ability to regulate transcription of a set of developmentally important genes. During spermatid maturation into sperm, these genes lose H3K4me2/3 and retain H3K27me3 marks. Experimental removal of these epigenetic marks at fertilization de-regulates gene expression in the resulting embryos in a paternal chromatin-dependent manner. This demonstrates that epigenetic instructions delivered by the sperm at fertilization are required for correct regulation of gene expression in the future embryos. The epigenetic mechanisms of developmental programming revealed here are likely to relate to the mechanisms involved in transgenerational transmission of acquired traits. Understanding how parental experience can influence development of the progeny has broad potential for improving human health.


Assuntos
Metilação de DNA/genética , Epigênese Genética , Histona-Lisina N-Metiltransferase/genética , Espermatozoides/metabolismo , Animais , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Histona-Lisina N-Metiltransferase/biossíntese , Histonas , Humanos , Masculino , Ranidae/genética , Ranidae/crescimento & desenvolvimento , Espermátides/crescimento & desenvolvimento , Espermátides/metabolismo , Espermatozoides/crescimento & desenvolvimento
10.
Semin Cancer Biol ; 42: 70-80, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27554136

RESUMO

SMYD3 is a member of the SET and MYND-domain family of methyl-transferases, the increased expression of which correlates with poor prognosis in various types of cancer. In liver and colon tumors, SMYD3 is localized in the nucleus, where it interacts with RNA Pol II and H3K4me3 and functions as a selective transcriptional amplifier of oncogenes and genes that control cell proliferation and metastatic spread. Smyd3 expression has a high discriminative power for the characterization of liver tumors and positively correlates with poor prognosis. In lung and pancreatic cancer, SMYD3 acts in the cytoplasm, potentiating oncogenic Ras/ERK signaling through the methylation of the MAP3K2 kinase and the subsequent release from its inhibitor. A clinico-pathological analysis of lung cancer patients uncovers prognostic significance of SMYD3 only for first progression survival. However, stratification of patients according to their smoking history significantly expands the prognostic value of SMYD3 to overall survival and other features, suggesting that smoking-related effects saturate the clinical analysis and mask the function of SMYD3 as an oncogenic potentiator.


Assuntos
Histona-Lisina N-Metiltransferase/genética , Neoplasias/genética , Prognóstico , Carcinogênese/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Histona-Lisina N-Metiltransferase/biossíntese , Humanos , Neoplasias/patologia , RNA Polimerase II/genética
11.
Br J Haematol ; 182(4): 542-553, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29978456

RESUMO

Histone deacetylase inhibitors (HDACi) had emerged as promising drugs in leukaemia, but their toxicity due to lack of specificity limited their use. Therefore, there is a need to elucidate the role of HDACs in specific settings. The study of HDAC expression in childhood leukaemia could help to choose more specific HDACi for selected candidates in a personalized approach. We analysed HDAC1-11, SIRT1, SIRT7, MEF2C and MEF2D mRNA expression in 211 paediatric patients diagnosed with acute leukaemia. There was a global overexpression of HDACs, while specific HDACs correlated with clinical and biological features, and some even predicted outcome. Thus, some HDAC and MEF2C profiles probably reflected the lineage and the maturation of the blasts and some profiles identified specific oncogenic pathways active in the leukaemic cells. Specifically, we identified a distinctive signature for patients with KMT2A (MLL) rearrangement, with high HDAC9 and MEF2D expression, regardless of age, KMT2A partner and lineage. Moreover, we observed an adverse prognostic value of HDAC9 overexpression, regardless of KMT2A rearrangement. Our results provide useful knowledge on the complex picture of HDAC expression in childhood leukaemia and support the directed use of specific HDACi to selected paediatric patients with acute leukaemia.


Assuntos
Regulação Enzimológica da Expressão Gênica , Regulação Leucêmica da Expressão Gênica , Rearranjo Gênico , Histona Desacetilases/biossíntese , Histona-Lisina N-Metiltransferase , Leucemia/enzimologia , Leucemia/genética , Proteína de Leucina Linfoide-Mieloide , Doença Aguda , Adolescente , Criança , Pré-Escolar , Feminino , Inibidores de Histona Desacetilases/administração & dosagem , Histona-Lisina N-Metiltransferase/biossíntese , Histona-Lisina N-Metiltransferase/genética , Humanos , Lactente , Recém-Nascido , Leucemia/tratamento farmacológico , Masculino , Proteína de Leucina Linfoide-Mieloide/biossíntese , Proteína de Leucina Linfoide-Mieloide/genética , Estudos Retrospectivos
12.
Toxicol Appl Pharmacol ; 342: 22-30, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29391238

RESUMO

While hexavalent chromium [Cr(VI)] is generally considered as a genotoxic environmental carcinogen, studies showed that Cr(VI) exposure also causes epigenetic changes. However, whether Cr(VI)-caused epigenetic dysregulations plays an important role in Cr(VI) carcinogenicity remain largely unknown. The aim of this study was to determine if chronic low dose Cr(VI) exposure causes epigenetic changes, the underlying mechanism and whether chronic low dose Cr(VI) exposure-caused epigenetic dysregulation contributes causally to Cr(VI)-induced cancer stem cell (CSC)-like property and cell transformation. Two immortalized human bronchial epithelial cell lines (BEAS-2B and 16HBE) were exposed to 0.25 µM of K2Cr2O7 for 20 and 40 weeks to induce cell transformation, respectively. Cr(VI)-induced epigenetic changes were examined in Cr(VI)-transformed cells and Cr(VI) exposure-caused human lung cancer tissues. Pharmacological inhibitors and gene knockdown experiments were used to determine the role of epigenetic dysregulation in Cr(VI) carcinogenicity. We found that chronic Cr(VI) exposure causes epigenetic dysregulation as evidenced by the increased levels of histone H3 repressive methylation marks (H3K9me2 and H3K27me3) and the related histone-lysing methyltransferases (HMTases). Pharmacological inhibition or knockdown of HMTases reduces H3 repressive methylation marks and malignant phenotypes of Cr(VI)-transformed cells. Moreover, knockdown of HMTases in parental cells significantly reduces chronic Cr(VI) exposure-induced CSC-like property and cell transformation. Further mechanistic study revealed that knockdown of HMTases decreases Cr(VI) exposure-caused DNA damage. Our findings indicate that chronic Cr(VI) exposure increases H3 repressive methylation marks by increasing the related HMTases expression; and that increased expression of HMTases plays a causal role in Cr(VI)-induced CSC-like property and cell transformation.


Assuntos
Transformação Celular Neoplásica/metabolismo , Cromo/toxicidade , Histona-Lisina N-Metiltransferase/biossíntese , Células-Tronco Neoplásicas/metabolismo , Regulação para Cima/fisiologia , Animais , Linhagem Celular Transformada , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/patologia , Humanos , Camundongos , Camundongos Nus , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Regulação para Cima/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
13.
Med Sci Monit ; 24: 7357-7365, 2018 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-30319138

RESUMO

BACKGROUND SET and MYND domain-containing protein 2 (SMYD2), which is identified as a protein-lysine methyltransferase, plays a crucial role in the progression of some tumors such as bladder carcinoma. However, the clinical significance of SMYD2 in patients with papillary thyroid carcinoma (PTC) has not been elucidated. In the present study, we aimed to investigate the expression and role of SMYD2 in human PTC. MATERIAL AND METHODS Clinicopathological analysis was performed in 107 patients with PTC. Expression of SMYD2 was determined by immunohistochemistry staining, quantitative RT-PCR, or Western blotting in PTC tissues, adjacent normal tissues, and PTC cells (K1 and B-CPAP). The prognostic value of SMYD2 in PTC patients was assessed by univariate and multivariate analysis. Clinical outcomes were evaluated by Kaplan-Meier log-rank tests. Cell proliferation was examined in PTC cells following overexpression or knockdown of SMYD2. RESULTS SMYD2 was highly expressed in PTC tissues compared to adjacent thyroid tissues. Additionally, high expression of SMYD2 was significantly related to tumor size, lymph node metastasis, and TNM stage. Moreover, SMYD2 was identified as an independent prognosis factor by multivariate analysis. Using 2 PTC cell lines, K1 and B-CPAP, we demonstrated that high expression of SMYD2 can promote tumor cell proliferation. CONCLUSIONS SMYD2 expression was upregulated in PTC tissues and significantly related to the poorer prognosis of PTC patients. Our studies suggested the potential role of SMYD2 as a new therapeutic target and prognostic biomarker in human PTC.


Assuntos
Histona-Lisina N-Metiltransferase/biossíntese , Câncer Papilífero da Tireoide/enzimologia , Adulto , Idoso , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Progressão da Doença , Feminino , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Prognóstico , Câncer Papilífero da Tireoide/genética , Transcriptoma , Regulação para Cima
14.
Biochem Biophys Res Commun ; 489(2): 96-102, 2017 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-28528974

RESUMO

Endometrial cancer (EC) is the estrogen-dependent gynecologic malignancy, however the molecular mechanism involved in the development and progression of EC remain unclear. The aim of this study was to investigate the role of RIZ1 in EC. Immunohistochemical analysis revealed that RIZ1was decreased in EC than in normal endometrium. Lower RIZ1 level was correlated with high-grade carcinoma (p = 0.048) and positive expression of ERα (p = 0.004). In EC cells, estrogen could down regulated the expression of RIZ1, however, ICI182,780 could up regulated the expression of RIZ1. Besides, in vitro and in vivo, RIZ1 could remarkably suppress tumor proliferation, metastasis and invasion. Our data support that RIZ1 was a novel tumor suppressor and could provide a potential therapeutic target in human EC.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Progressão da Doença , Regulação para Baixo , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Estrogênios/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Ensaios de Seleção de Medicamentos Antitumorais , Neoplasias do Endométrio/tratamento farmacológico , Estradiol/análogos & derivados , Estradiol/farmacologia , Feminino , Fulvestranto , Histona-Lisina N-Metiltransferase/biossíntese , Histona-Lisina N-Metiltransferase/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Relação Estrutura-Atividade , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Células Tumorais Cultivadas
15.
J Neuroinflammation ; 14(1): 132, 2017 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-28662718

RESUMO

BACKGROUND: Controlling thrombin-driven microglial activation may serve as a therapeutic target for intracerebral hemorrhage (ICH). Here, we investigated microRNA (miRNA)-based regulation of thrombin-driven microglial activation using an in vitro thrombin toxicity model applied to primary human microglia. METHODS: A miRNA array identified 22 differential miRNA candidates. Quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) identified miR-181c as the most significantly downregulated miRNA. TargetScan analysis identified mixed lineage leukemia-1 (MLL1) as a putative gene target for miR-181c. qRT-PCR was applied to assess tumor necrosis factor-alpha (TNF-α), miR-181c, and MLL1 levels following thrombin or proteinase-activated receptor-4-specific activating peptide (PAR4AP) exposure. Anti-TNF-α antibodies and tumor necrosis factor receptor (TNFR) silencing were employed to test TNF-α/TNFR dependence. A dual-luciferase reporter system and miR-181c mimic transfection assessed whether mir-181c directly binds to and negatively regulates MLL1. Nuclear factor kappa-B (NF-κB)-dependent luciferase reporter assays and NF-κB target gene expression were assessed in wild-type (MLL1+) and MLL1-silenced cells. RESULTS: Thrombin or PAR4AP-induced miR-181c downregulation (p < 0.05) and MLL1 upregulation (p < 0.05) that were dependent upon TNF-α/TNFR. miR-181c decreased wild-type MLL1 3'-UTR luciferase reporter activity (p < 0.05), and a miR-181c mimic suppressed MLL1 expression (p < 0.05). Thrombin treatment increased, while miR-181c reduced, NF-κB activity and NF-κB target gene expression in both wild-type (MLL1+) and MLL1-silenced cells (p < 0.05). CONCLUSIONS: Thrombin-induced, TNF-α/TNFR-dependent miR-181c downregulation promotes MLL1 expression, increases NF-κB activity, and upregulates NF-κB target gene expression. As miR-181c opposes thrombin's stimulation of pro-inflammatory NF-κB activity, miR-181c mimic therapy may show promise in controlling thrombin-driven microglial activation following ICH.


Assuntos
Histona-Lisina N-Metiltransferase/biossíntese , MicroRNAs/metabolismo , Microglia/metabolismo , Proteína de Leucina Linfoide-Mieloide/biossíntese , NF-kappa B/biossíntese , Receptores do Fator de Necrose Tumoral/metabolismo , Trombina/farmacologia , Adulto , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Feminino , Expressão Gênica , Células HEK293 , Histona-Lisina N-Metiltransferase/genética , Humanos , Masculino , Microglia/efeitos dos fármacos , Pessoa de Meia-Idade , Proteína de Leucina Linfoide-Mieloide/genética , NF-kappa B/genética
16.
PLoS Genet ; 10(2): e1004201, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24586211

RESUMO

Plasma cholesterol lowering (PCL) slows and sometimes prevents progression of atherosclerosis and may even lead to regression. Little is known about how molecular processes in the atherosclerotic arterial wall respond to PCL and modify responses to atherosclerosis regression. We studied atherosclerosis regression and global gene expression responses to PCL (≥80%) and to atherosclerosis regression itself in early, mature, and advanced lesions. In atherosclerotic aortic wall from Ldlr(-/-)Apob (100/100) Mttp (flox/flox)Mx1-Cre mice, atherosclerosis regressed after PCL regardless of lesion stage. However, near-complete regression was observed only in mice with early lesions; mice with mature and advanced lesions were left with regression-resistant, relatively unstable plaque remnants. Atherosclerosis genes responding to PCL before regression, unlike those responding to the regression itself, were enriched in inherited risk for coronary artery disease and myocardial infarction, indicating causality. Inference of transcription factor (TF) regulatory networks of these PCL-responsive gene sets revealed largely different networks in early, mature, and advanced lesions. In early lesions, PPARG was identified as a specific master regulator of the PCL-responsive atherosclerosis TF-regulatory network, whereas in mature and advanced lesions, the specific master regulators were MLL5 and SRSF10/XRN2, respectively. In a THP-1 foam cell model of atherosclerosis regression, siRNA targeting of these master regulators activated the time-point-specific TF-regulatory networks and altered the accumulation of cholesterol esters. We conclude that PCL leads to complete atherosclerosis regression only in mice with early lesions. Identified master regulators and related PCL-responsive TF-regulatory networks will be interesting targets to enhance PCL-mediated regression of mature and advanced atherosclerotic lesions.


Assuntos
Aorta/metabolismo , Aterosclerose/sangue , Colesterol/sangue , Receptores de LDL/genética , Animais , Aorta/efeitos dos fármacos , Apolipoproteínas B/genética , Aterosclerose/tratamento farmacológico , Aterosclerose/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/biossíntese , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Camundongos , Camundongos Transgênicos , Proteínas Nucleares/biossíntese , Ribonucleoproteínas/biossíntese , Fatores de Processamento de Serina-Arginina
17.
Microbiol Immunol ; 60(6): 407-17, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27080060

RESUMO

Hepatitis C virus (HCV) non-structural protein 5A (NS5A) is a multifunctional protein that is involved in the HCV life cycle and pathogenesis. In this study, a host protein(s) interacting with NS5A by tandem affinity purification were searched for with the aim of elucidating the role of NS5A. An NS5A-interacting protein, SET and MYND domain-containing 3 (SMYD3), a lysine methyltransferase reportedly involved in the development of cancer, was identified. The interaction between NS5A and SMYD3 was confirmed in ectopically expressing, HCV RNA replicon-harboring and HCV-infected cells. The other HCV proteins did not bind to SMYD3. SMYD3 bound to NS5A of HCV genotypes 1b and 2a. Deletion mutational analysis revealed that domains II and III of NS5A (amino acids [aa] 250 to 447) and the MYND and N-SET domains of SMYD3 (aa 1 to 87) are involved in the full extent of NS5A-SMYD3 interaction. NS5A co-localized with SMYD3 exclusively in the cytoplasm, thereby inhibiting nuclear localization of SMYD3. Moreover, NS5A formed a complex with SMYD3 and heat shock protein 90 (HSP90), which is a positive regulator of SMYD3. The intensity of binding between SMYD3 and HSP90 was enhanced by NS5A. Luciferase reporter assay demonstrated that NS5A significantly induces activator protein 1 (AP-1) activity, this being potentiated by co-expression of SMYD3 with NS5A. Taken together, the present results suggest that NS5A interacts with SMYD3 and induces AP-1 activation, possibly by facilitating binding between HSP90 and SMYD3. This may be a novel mechanism of AP-1 activation in HCV-infected cells.


Assuntos
Hepacivirus/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Fator de Transcrição AP-1/biossíntese , Fator de Transcrição AP-1/metabolismo , Proteínas não Estruturais Virais/metabolismo , Linhagem Celular , Citoplasma/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Hepacivirus/genética , Hepatite C/virologia , Histona-Lisina N-Metiltransferase/biossíntese , Interações Hospedeiro-Patógeno , Humanos , Mapeamento de Interação de Proteínas/métodos , Mapas de Interação de Proteínas , Replicon/fisiologia , Análise de Sequência de Proteína , Deleção de Sequência , Proteínas não Estruturais Virais/biossíntese , Proteínas não Estruturais Virais/genética , Replicação Viral/fisiologia
18.
Med Sci Monit ; 22: 3009-17, 2016 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-27561414

RESUMO

BACKGROUND The overall prognosis of acute myeloid leukemia (AML) patients with mixed-lineage leukemia (MLL) gene-positivity is unfavorable. In this study, we evaluated the expression levels of the MLL gene in AML patients. MATERIAL AND METHODS We enrolled 68 MLL gene-positive patients out of 433 newly diagnosed AML patients, and 216 bone marrow samples were collected. Real-time fluorescence quantitative PCR (RQ-PCR) was used to precisely detect the expression levels of the MLL gene. RESULTS We divided 41 patients into 2 groups according to the variation of MRD (minimal residual disease) level of the MLL gene. Group 1 (n=22) had a rapid reduction of MRD level to ≤10^-4 in all samples collected in the first 3 chemotherapy cycles, while group 2 (n=19) had MRD levels constantly >10^-4 in all samples collected in the first 3 chemotherapy cycles. Group 1 had a significantly better overall survival (p=0.001) and event-free survival (p=0.001) compared to group 2. Moreover, the patients with >10^-4 MRD level before the start of HSCT (hematopoietic stem cell transplantation) had worse prognosis and higher risk of relapse compared to patients with ≤10^-4 before the start of HSCT. CONCLUSIONS We found that a rapid reduction of MRD level to ≤10^-4 appears to be a prerequisite for better overall survival and event-free survival during the treatment of AML. The MRD levels detected by RQ-PCR were basically in line with the clinical outcome and may be of great importance in guiding early allogeneic HSCT (allo-HSCT) treatment.


Assuntos
Histona-Lisina N-Metiltransferase/genética , Leucemia Mieloide Aguda/genética , Proteína de Leucina Linfoide-Mieloide/genética , Adolescente , Adulto , Idoso , Intervalo Livre de Doença , Feminino , Fluorescência , Transplante de Células-Tronco Hematopoéticas/métodos , Histona-Lisina N-Metiltransferase/biossíntese , Humanos , Leucemia Mieloide Aguda/enzimologia , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/terapia , Masculino , Pessoa de Meia-Idade , Proteína de Leucina Linfoide-Mieloide/biossíntese , Neoplasia Residual , Prognóstico , Reação em Cadeia da Polimerase em Tempo Real/métodos , Recidiva , Transcriptoma
19.
Biotechnol Lett ; 38(3): 395-402, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26588904

RESUMO

OBJECTIVES: We have examined dynamic changes of histone H3 lysine 9 following trimethylation (H3K9me3), the mRNA expression levels of SUV39H1 and SUV39H2 in bovine oocytes and the role in the development of in vitro fertilization (IVF) pre-implantation embryos. RESULTS: There were strong H3K9me3 signals in germinal vesicle (GV) oocytes but no signals in MII oocytes. H3K9me3 signals were maintained during IVF pre-implantation embryo development. SUV39H1 and SUV39H2 showed significantly higher mRNA expression levels in GV oocytes than MII oocytes (P < 0.01). SUV39H1 showed high mRNA expression level in two-cell embryos, however, SUV39H2 showed high mRNA expression level in four-cell embryos. In other development stage, SUV39H1 and SUV39H2 showed low expression levels. CONCLUSION: Bovine IVF pre-implantation embryos maintain strong H3K9me3 signals and SUV39H1 and SUV39H2 are highly expressed at the early development stage of pre-implantation embryos.


Assuntos
Embrião de Mamíferos , Desenvolvimento Embrionário , Histona-Lisina N-Metiltransferase/biossíntese , Histonas/biossíntese , Animais , Bovinos , Fertilização in vitro , Perfilação da Expressão Gênica , Histonas/metabolismo , Metilação , Oócitos , Processamento de Proteína Pós-Traducional
20.
Mol Biol (Mosk) ; 50(6): 968-977, 2016.
Artigo em Russo | MEDLINE | ID: mdl-28064313

RESUMO

MLL is involved in fusion genes with more than 100 partner genes, approximately 80 of which have been characterized at the molecular level. MLL fusion genes are often found in infants (60-80% of acute lymphoblastic leukemia (ALL) cases and 40-50% of acute myeloblastic leukemia (AML) cases) and are appreciably rarer (8-10%) in children older than 1 year of age. MLL rearrangements are important markers in diagnosis and treatment choice. To identify the partner gene is of primary importance for prognosis and minimal residual disease monitoring. The structure of the fusion gene, including localization of the MLL breakpoints, is also informative. A method was developed to examine the fusion transcripts in order to identify the partner gene among the six most common ones and to establish the exon structure of the rearranged MLL. The method includes a multiplex reverse transcriptase-polymerase chain reaction (RT-PCR) to amplify and to fluorescently label a fusion transcript fragment and subsequent hybridization of the product on a biological microchip with immobilized oligonucleotides complementary to exons of MLL and its partner genes AFF1, MLLT1, MLLT3, MLLT4, MLLT10, and ELL. Hybridization results were verified by sequencing the RT-PCR products and, in some cases, performing long-distance inverse PCR (LDI-PCR). The study involved 38 bone marrow samples from ALL patients (including 33 children younger than 1 year of age) and 15 samples from AML patients (including 10 from children younger than 1 year of age). The main partner genes were AFF1 (49%), MLLT1 (27%), MLLT3 (12%), and MLLT10 (12%) in ALL and MLLT3 (80%), MLLT10 (10%), and MLLT4 (10%) in AML. Fusion gene transcripts most commonly included MLL exon 11 (58% of ALL cases and 50% of AML cases), suggesting a breakpoint in MLL intron 11.


Assuntos
Perfilação da Expressão Gênica/instrumentação , Regulação Leucêmica da Expressão Gênica , Histona-Lisina N-Metiltransferase/biossíntese , Leucemia Mieloide Aguda/metabolismo , Proteína de Leucina Linfoide-Mieloide/biossíntese , Análise de Sequência com Séries de Oligonucleotídeos/instrumentação , Proteínas de Fusão Oncogênica/biossíntese , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Transcrição Gênica , Criança , Pré-Escolar , Feminino , Perfilação da Expressão Gênica/métodos , Histona-Lisina N-Metiltransferase/genética , Humanos , Lactente , Recém-Nascido , Leucemia Mieloide Aguda/genética , Masculino , Proteína de Leucina Linfoide-Mieloide/genética , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Proteínas de Fusão Oncogênica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA