Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Mol Neurobiol ; 58(11): 5937-5953, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34435328

RESUMO

MiR-143-3p is aberrantly expressed in patients with ischemic stroke and associated with ischemic brain injury. However, the underlying mechanisms are largely unknown. Here, we confirmed circ_0025984 and TET1 as a sponge and target of miR-143-3p, respectively, by luciferase reporter assay. In astrocytes, OGD significantly decreased circ_0025984 and TET1 levels but increased miR-143-3p levels, which was also observed in brains of mice with MCAO. Treatment with miR-143-3p inhibitor or circ_0025984 significantly decreased astrocyte apoptosis and autophagy, as well as cerebral injury and neuron loss in mice with MCAO. Notably, TET1 overexpression decreased astrocyte apoptosis and autophagy and induced promoter hypomethylation and expression of ORP150. Our results demonstrated for the first time that circ_0025984 protects astrocytes from ischemia-induced autophagy and apoptosis by targeting the miR-143-3p/TET1 pathway and might inhibit cerebral injury induced by ischemic stroke. Furthermore, our data revealed the important positive regulation of ORP150 by TET1, which could be associated with its neuroprotective role. Graphical abstract Model for signaling pathway of circ_0025984/miR-143-3p/TET1 inastrocytes cultured under OGD. In astrocytes, circ_0025984 acts as a sponge of miR-143-3p, which directly targets TET1 and decreases its expression (A). After translocatinginto the nucleus, TET1 binds to the promoter of ORP150, converts 5mC into 5hmC,leading to DNA demethylation and increased expression of ORP150 (B). In astrocytescultured under OGD, ER stress is induced and eventually leads to apoptosis andautophagy mediated by ATG7, which is regulated by circ_0025984 via ORP150 andGRP78 (C).


Assuntos
Astrócitos/metabolismo , Dioxigenases/fisiologia , Proteínas de Choque Térmico HSP70/fisiologia , Infarto da Artéria Cerebral Média/fisiopatologia , MicroRNAs/fisiologia , Proteínas do Tecido Nervoso/fisiologia , RNA Circular/fisiologia , Animais , Apoptose , Astrócitos/patologia , Astrocitoma , Autofagia , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Genes Reporter , Humanos , Infarto da Artéria Cerebral Média/genética , Masculino , MicroRNAs/antagonistas & inibidores , Oxigenases de Função Mista/fisiologia , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Proteínas Proto-Oncogênicas/fisiologia , RNA Circular/biossíntese , RNA Circular/genética , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/metabolismo , Transdução de Sinais/fisiologia , Organismos Livres de Patógenos Específicos
2.
Int J Biol Macromol ; 182: 298-310, 2021 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-33811933

RESUMO

9-cis-epoxy carotenoid dioxygenase (NCED) is a fundamental enzyme, which plays an essential role in the process of organ development and stress resistance by regulating abscisic acid (ABA) synthesis in plant. In this study, a total of 7, 7, 14 and 14 NCED genes were identified from the genomes of G. arboreum, G. raimondii, G. barbadense and G. hirsutum, respectively. Phylogenetic tree showed that all forty-two NCED genes could be classified into three groups in cotton genus. Collinear analysis revealed that the NCED genes in G. hirsutum were not amplified by tandem repeats after polyploidy events. The function of NCED genes was evaluated between two accessions with contrasting plant height. The results showed that expression of the NCED genes in dwarf accession was higher than that in taller ones. GhNCED1-silenced cotton plants confirmed that suppression of NCED genes could increase the plant height, but reduce the resistance abilities to drought and salt stress. Our study systematically identified the homologs of NCED genes and their functions in cotton, which could provide new genetic resources for improving plant height and stress in future cotton breeding.


Assuntos
Dioxigenases/genética , Evolução Molecular , Gossypium/enzimologia , Proteínas de Plantas/genética , Ácido Abscísico/metabolismo , Dioxigenases/metabolismo , Dioxigenases/fisiologia , Secas , Regulação da Expressão Gênica de Plantas , Gossypium/genética , Gossypium/fisiologia , Família Multigênica , Filogenia , Proteínas de Plantas/metabolismo , Proteínas de Plantas/fisiologia , Análise de Sequência de DNA , Transdução de Sinais , Estresse Fisiológico
3.
Nat Rev Cancer ; 20(12): 710-726, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33087883

RESUMO

2-Oxoglutarate-dependent dioxygenases (2OGDDs) are a superfamily of enzymes that play diverse roles in many biological processes, including regulation of hypoxia-inducible factor-mediated adaptation to hypoxia, extracellular matrix formation, epigenetic regulation of gene transcription and the reprogramming of cellular metabolism. 2OGDDs all require oxygen, reduced iron and 2-oxoglutarate (also known as α-ketoglutarate) to function, although their affinities for each of these co-substrates, and hence their sensitivity to depletion of specific co-substrates, varies widely. Numerous 2OGDDs are recurrently dysregulated in cancer. Moreover, cancer-specific metabolic changes, such as those that occur subsequent to mutations in the genes encoding succinate dehydrogenase, fumarate hydratase or isocitrate dehydrogenase, can dysregulate specific 2OGDDs. This latter observation suggests that the role of 2OGDDs in cancer extends beyond cancers that harbour mutations in the genes encoding members of the 2OGDD superfamily. Herein, we review the regulation of 2OGDDs in normal cells and how that regulation is corrupted in cancer.


Assuntos
Dioxigenases/fisiologia , Ácidos Cetoglutáricos/metabolismo , Neoplasias/metabolismo , Hipóxia Celular , Ciclo do Ácido Cítrico , Dano ao DNA , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Prolina Dioxigenases do Fator Induzível por Hipóxia/fisiologia , Neoplasias/tratamento farmacológico , Neoplasias/etiologia , Espécies Reativas de Oxigênio/metabolismo
4.
Oncogene ; 38(25): 5038-5049, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30858546

RESUMO

Epigenetic changes that cause dysregulated gene expression during progression of androgen-independent prostate cancer (PCa) and metastatic skeletal lesions remain elusive. Here, we explored the role of histone demethylase NO66 in the pathogenesis of PCa and bone metastasis-related skeletal lesions. Tissue and cDNA microarrays of PCa were analyzed for NO66 mRNA and protein levels. We examined the effects of gain and loss of NO66 function on cell viability, colony formation, migration, invasion, and tumor-induced skeletal lesions in femoral bone. RNAseq and ChIPseq were performed to elucidate NO66-target genes in PCa. We report that NO66 levels were upregulated in advanced primary prostate tumors compared to normal tissue or tumors with low Gleason scores. Forced expression of NO66 promoted cell survival and invasion of PCa cells; whereas, knockdown of NO66 resulted in decreased cell survival and increased sensitivity to docetaxel. NO66-overexpressing PC3 cells implanted into the femoral bone of male SCID mice caused massive bone loss and stimulation of mouse osteoclast-promoting genes, including Dickkopf1, Cathepsin K, Nf-kß,; and Calcr, suggesting a role for NO66 in tumor growth in bone and osteoclast activity. Combined RNAseq and ChIP-seq revealed that NO66 activates the survival gene MCL1, the invasion-associated genes IGFBP5 and MMP3, the pro-oncogenic genes CTNNB1 and CCND1, and the epigenetic modifier gene KMT2A in androgen-independent PCa. Our findings uncover the role of NO66 as a key oncogenic driver in PCa, causing osteolytic lesions through upstream epigenetic regulation of key genes for survival, invasion and metastasis, and pro-osteoclastic factors.


Assuntos
Transformação Celular Neoplásica/genética , Dioxigenases/fisiologia , Histona Desmetilases/fisiologia , Osteólise/genética , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/patologia , Animais , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Linhagem Celular Tumoral , Transformação Celular Neoplásica/metabolismo , Dioxigenases/genética , Epigênese Genética/genética , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Histona Desmetilases/genética , Histonas/metabolismo , Humanos , Masculino , Camundongos , Camundongos SCID , Células NIH 3T3 , Osteólise/patologia , Células PC-3 , Neoplasias de Próstata Resistentes à Castração/metabolismo
5.
Nat Genet ; 50(1): 83-95, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29203910

RESUMO

TET enzymes oxidize 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), which can lead to DNA demethylation. However, direct connections between TET-mediated DNA demethylation and transcriptional output are difficult to establish owing to challenges in distinguishing global versus locus-specific effects. Here we show that TET1, TET2 and TET3 triple-knockout (TKO) human embryonic stem cells (hESCs) exhibit prominent bivalent promoter hypermethylation without an overall corresponding decrease in gene expression in the undifferentiated state. Focusing on the bivalent PAX6 locus, we find that increased DNMT3B binding is associated with promoter hypermethylation, which precipitates a neural differentiation defect and failure of PAX6 induction during differentiation. dCas9-mediated locus-specific demethylation and global inactivation of DNMT3B in TKO hESCs partially reverses the hypermethylation at the PAX6 promoter and improves differentiation to neuroectoderm. Taking these findings together with further genome-wide methylation and TET1 and DNMT3B ChIP-seq analyses, we conclude that TET proteins safeguard bivalent promoters from de novo methylation to ensure robust lineage-specific transcription upon differentiation.


Assuntos
Metilação de DNA , Proteínas de Ligação a DNA/fisiologia , Células-Tronco Embrionárias/metabolismo , Oxigenases de Função Mista/fisiologia , Regiões Promotoras Genéticas , Animais , Diferenciação Celular/genética , Células Cultivadas , Proteínas de Ligação a DNA/genética , Dioxigenases/genética , Dioxigenases/fisiologia , Células-Tronco Embrionárias/citologia , Humanos , Camundongos , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Mutação , Placa Neural/citologia , Fator de Transcrição PAX6/biossíntese , Fator de Transcrição PAX6/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/fisiologia
6.
Yi Chuan ; 39(12): 1138-1149, 2017 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-29258984

RESUMO

The development of the nervous system is coordinately regulated by multiple interacting factors. If a certain factor is altered or mutated, the coordinated developmental processes could be disrupted, resulting in neurological diseases. The 5-hydroxymethylcytosine (5hmC) is an intermediate product of the DNA demethylation processes. 5hmC and its metabolic enzymes, the ten-eleven translocation protein-TET family of dioxygenases, have recently been identified as new epigenetic players important in the regulation of the nervous system development, as well as in cognition, memory and other neurological functions. In various studies on neurodevelopment and neurodegeneration related diseases, the levels of 5hmC and TET proteins could be differentially regulated during development and/or disease pathogenesis, suggesting the potentially critical roles of 5hmC and TETs in these neural developmental and disease processes. In this review, we summarize the recent advances in research on 5hmC and TET dioxygenases in the regulation of neurodevelopment and neurological diseases, thereby providing significant insights on the involvements of 5hmC and TETs in neurodevelopment and on establishing new therapeutic strategies for human neurological diseases.


Assuntos
5-Metilcitosina/análogos & derivados , Dioxigenases/fisiologia , Oxigenases de Função Mista/fisiologia , Doenças do Sistema Nervoso/etiologia , Sistema Nervoso/crescimento & desenvolvimento , Proteínas Proto-Oncogênicas/fisiologia , 5-Metilcitosina/fisiologia , Animais , Humanos , Neurogênese
7.
Cancer Res ; 75(18): 3912-24, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26294212

RESUMO

Hypoxia is a hallmark of solid tumors that drives malignant progression by altering epigenetic controls. In breast tumors, aberrant DNA methylation is a prevalent epigenetic feature associated with increased risk of metastasis and poor prognosis. However, the mechanism by which hypoxia alters DNA methylation or other epigenetic controls that promote breast malignancy remains poorly understood. We discovered that hypoxia deregulates TET1 and TET3, the enzymes that catalyze conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), thereby leading to breast tumor-initiating cell (BTIC) properties. TET1/3 and 5hmC levels were closely associated with tumor hypoxia, tumor malignancy, and poor prognosis in breast cancer patients. Mechanistic investigations showed that hypoxia leads to genome-wide changes in DNA hydroxymethylation associated with upregulation of TNFα expression and activation of its downstream p38-MAPK effector pathway. Coordinate functions of TET1 and TET3 were also required to activate TNFα-p38-MAPK signaling as a response to hypoxia. Our results reveal how signal transduction through the TET-TNFα-p38-MAPK signaling axis is required for the acquisition of BTIC characteristics and tumorigenicity in vitro and in vivo, with potential implications for how to eradicate BTIC as a therapeutic strategy.


Assuntos
Neoplasias da Mama/genética , Hipóxia Celular/fisiologia , Metilação de DNA , Proteínas de Ligação a DNA/fisiologia , Dioxigenases/fisiologia , Proteínas de Neoplasias/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , 5-Metilcitosina/análogos & derivados , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Hipóxia Celular/genética , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Citosina/análogos & derivados , Citosina/biossíntese , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Dioxigenases/biossíntese , Dioxigenases/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos , Camundongos Nus , Oxigenases de Função Mista , Dados de Sequência Molecular , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/enzimologia , Prognóstico , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética , Proteínas Recombinantes de Fusão/biossíntese , Estudos Retrospectivos , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
8.
Cell Mol Life Sci ; 72(20): 3897-914, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26118662

RESUMO

Recent studies have revealed that the members of an ancient family of nonheme Fe(2+)/2-oxoglutarate-dependent dioxygenases (2-OGDO) are involved in the functions associated with the aging process. 2-Oxoglutarate and O2 are the obligatory substrates and Fe(2+) a cofactor in the activation of 2-OGDO enzymes, which can induce the hydroxylation of distinct proteins and the demethylation of DNA and histones. For instance, ten-eleven translocation 1-3 (TET1-3) are the demethylases of DNA, whereas Jumonji C domain-containing histone lysine demethylases (KDM2-7) are the major epigenetic regulators of chromatin landscape, known to be altered with aging. The functions of hypoxia-inducible factor (HIF) prolyl hydroxylases (PHD1-3) as well as those of collagen hydroxylases are associated with age-related degeneration. Moreover, the ribosomal hydroxylase OGFOD1 controls mRNA translation, which is known to decline with aging. 2-OGDO enzymes are the sensors of energy metabolism, since the Krebs cycle intermediate 2-oxoglutarate is an activator whereas succinate and fumarate are the potent inhibitors of 2-OGDO enzymes. In addition, O2 availability and iron redox homeostasis control the activities of 2-OGDO enzymes in tissues. We will briefly elucidate the catalytic mechanisms of 2-OGDO enzymes and then review the potential functions of the above-mentioned 2-OGDO enzymes in the control of the aging process.


Assuntos
Envelhecimento/metabolismo , Dioxigenases/fisiologia , Metabolismo Energético , Ferro/metabolismo , Oxigênio/metabolismo , Animais , Metilação de DNA , Dioxigenases/metabolismo , Epigênese Genética , Homeostase , Hipóxia/metabolismo , Biossíntese de Proteínas
9.
Med Sci (Paris) ; 31(3): 268-74, 2015 Mar.
Artigo em Francês | MEDLINE | ID: mdl-25855280

RESUMO

DNA methylation is associated with a large number of biological processes and mainly concerns the cytosine methylation at position 5 (5-mC). An active demethylation mechanism was highlighted in 2009 following the discovery that TET proteins were enzymes implicated in the hydroxylation of 5-mC to 5-hydroxymethylcytosine. Simultaneously, other studies showed frequent acquired TET2 mutations in hematological malignancies and have depicted their role in their pathogenesis. An entire field of research has developed rapidly showing that these proteins are involved in many biological processes.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Dioxigenases/fisiologia , Desenvolvimento Embrionário/genética , Hematopoese/genética , Proteínas Proto-Oncogênicas/fisiologia , Animais , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Dioxigenases/química , Dioxigenases/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Oxigenases de Função Mista , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/genética
10.
Oncogene ; 34(50): 6079-91, 2015 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-25745994

RESUMO

Epstein-Barr virus (EBV) causes human lymphoid malignancies, and the EBV product latent membrane protein 1 (LMP1) has been identified as an oncogene in epithelial carcinomas such as nasopharyngeal carcinoma (NPC). EBV can epigenetically reprogram lymphocyte-specific processes and induce cell immortalization. However, the interplay between LMP1 and the NPC host cell remains largely unknown. Here, we report that LMP1 is important to establish the Hox gene expression signature in NPC cell lines and tumor biopsies. LMP1 induces repression of several Hox genes in part via stalling of RNA polymerase II (RNA Pol II). Pol II stalling can be overcome by irradiation involving the epigenetic regulator TET3. Furthermore, we report that HoxC8, one of the genes silenced by LMP1, has a role in tumor growth. Ectopic expression of HoxC8 inhibits NPC cell growth in vitro and in vivo, modulates glycolysis and regulates the expression of tricarboxylic acid (TCA) cycle-related genes. We propose that viral latency products may repress via stalling key mediators that in turn modulate glycolysis.


Assuntos
Genes Homeobox/fisiologia , Proteínas de Homeodomínio/fisiologia , Neoplasias Nasofaríngeas/genética , Proteínas da Matriz Viral/fisiologia , Células Cultivadas , Ciclo do Ácido Cítrico , Metilação de DNA , Dioxigenases/fisiologia , Glicólise , Humanos , Neoplasias Nasofaríngeas/metabolismo , RNA Polimerase II/metabolismo
11.
Mol Cell Biol ; 35(5): 789-804, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25512612

RESUMO

The ten-eleven translocation 2 gene (TET2) encodes a member of the TET family of DNA methylcytosine oxidases that converts 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) to initiate the demethylation of DNA within genomic CpG islands. Somatic loss-of-function mutations of TET2 are frequently observed in human myelodysplastic syndrome (MDS), which is a clonal malignancy characterized by dysplastic changes of developing blood cell progenitors, leading to ineffective hematopoiesis. We used genome-editing technology to disrupt the zebrafish Tet2 catalytic domain. tet2(m/m) (homozygous for the mutation) zebrafish exhibited normal embryonic and larval hematopoiesis but developed progressive clonal myelodysplasia as they aged, culminating in myelodysplastic syndromes (MDS) at 24 months of age, with dysplasia of myeloid progenitor cells and anemia with abnormal circulating erythrocytes. The resultant tet2(m/m) mutant zebrafish lines show decreased levels of 5hmC in hematopoietic cells of the kidney marrow but not in other cell types, most likely reflecting the ability of other Tet family members to provide this enzymatic activity in nonhematopoietic tissues but not in hematopoietic cells. tet2(m/m) zebrafish are viable and fertile, providing an ideal model to dissect altered pathways in hematopoietic cells and, for small-molecule screens in embryos, to identify compounds with specific activity against tet2 mutant cells.


Assuntos
Dioxigenases/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica , Síndromes Mielodisplásicas/metabolismo , Proteínas de Peixe-Zebra/genética , Animais , Domínio Catalítico , Diferenciação Celular , Ilhas de CpG , Proteínas de Ligação a DNA/metabolismo , Dioxigenases/fisiologia , Citometria de Fluxo , Genótipo , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Humanos , Hibridização In Situ , Rim/metabolismo , Mutação , Células-Tronco/citologia , Peixe-Zebra , Proteínas de Peixe-Zebra/fisiologia
12.
Cancer Prev Res (Phila) ; 7(12): 1219-27, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25293877

RESUMO

Obesity is associated with increased liver cancer risks and mortality. We recently showed that apo-10'-lycopenoic acid, a lycopene metabolite generated by beta-carotene-9',10'-oxygenase (BCO2), inhibited carcinogen-initiated, high-fat diet (HFD)-promoted liver inflammation, and hepatic tumorigenesis development. The present investigation examined the outstanding question of whether lycopene could suppress HFD-promoted hepatocellular carcinoma (HCC) progression, and if BCO2 expression is important using BCO2-knockout (BCO2-KO) and wild-type male mice. Results showed that lycopene supplementation (100 mg/kg diet) for 24 weeks resulted in comparable accumulation of hepatic lycopene (19.4 vs. 18.2 nmol/g) and had similar effects on suppressing HFD-promoted HCC incidence (19% vs. 20%) and multiplicity (58% vs. 62%) in wild-type and BCO2-KO mice, respectively. Intriguingly, lycopene chemopreventive effects in wild-type mice were associated with reduced hepatic proinflammatory signaling (phosphorylation of NK-κB p65 and STAT3; IL6 protein) and inflammatory foci. In contrast, the protective effects of lycopene in BCO2-KO but not in wild-type mice were associated with reduced hepatic endoplasmic reticulum stress-mediated unfolded protein response (ER(UPR)), through decreasing ER(UPR)-mediated protein kinase RNA-activated like kinase-eukaryotic initiation factor 2α activation, and inositol requiring 1α-X-box-binding protein 1 signaling. Lycopene supplementation in BCO2-KO mice suppressed oncogenic signals, including Met mRNA, ß-catenin protein, and mTOR complex 1 activation, which was associated with increased hepatic microRNA (miR)-199a/b and miR214 levels. These results provided novel experimental evidence that dietary lycopene can prevent HFD-promoted HCC incidence and multiplicity in mice, and may elicit different mechanisms depending on BCO2 expression.


Assuntos
Anticarcinógenos/administração & dosagem , Carcinoma Hepatocelular/prevenção & controle , Carotenoides/administração & dosagem , Dieta Hiperlipídica , Dioxigenases/fisiologia , Neoplasias Hepáticas/prevenção & controle , Animais , Western Blotting , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Transformação Celular Neoplásica , Suplementos Nutricionais , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Humanos , Técnicas Imunoenzimáticas , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Licopeno , Masculino , Camundongos , Camundongos Knockout , Fosforilação/efeitos dos fármacos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Células Tumorais Cultivadas
13.
Genome Biol ; 15(6): R81, 2014 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-24958354

RESUMO

BACKGROUND: The TET family of dioxygenases catalyze conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), but their involvement in establishing normal 5mC patterns during mammalian development and their contributions to aberrant control of 5mC during cellular transformation remain largely unknown. We depleted TET1, TET2, and TET3 in a pluripotent embryonic carcinoma cell model and examined the impact on genome-wide 5mC, 5hmC, and transcriptional patterns. RESULTS: TET1 depletion yields widespread reduction of 5hmC, while depletion of TET2 and TET3 reduces 5hmC at a subset of TET1 targets suggesting functional co-dependence. TET2 or TET3 depletion also causes increased 5hmC, suggesting these proteins play a major role in 5hmC removal. All TETs prevent hypermethylation throughout the genome, a finding dramatically illustrated in CpG island shores, where TET depletion results in prolific hypermethylation. Surprisingly, TETs also promote methylation, as hypomethylation was associated with 5hmC reduction. TET function is highly specific to chromatin environment: 5hmC maintenance by all TETs occurs at polycomb-marked chromatin and genes expressed at moderate levels; 5hmC removal by TET2 is associated with highly transcribed genes enriched for H3K4me3 and H3K36me3. Importantly, genes prone to hypermethylation in cancer become depleted of 5hmC with TET deficiency, suggesting that TETs normally promote 5hmC at these loci. Finally, all three TETs, but especially TET2, are required for 5hmC enrichment at enhancers, a condition necessary for expression of adjacent genes. CONCLUSIONS: These results provide novel insight into the division of labor among TET proteins and reveal important connections between TET activity, the chromatin landscape, and gene expression.


Assuntos
5-Metilcitosina/metabolismo , Metilação de DNA , Proteínas de Ligação a DNA/fisiologia , Dioxigenases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Diferenciação Celular , Linhagem Celular Tumoral , Citosina/análogos & derivados , Citosina/metabolismo , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Oxigenases de Função Mista , Regiões Promotoras Genéticas , Análise de Sequência de DNA
14.
EMBO Rep ; 14(4): 337-46, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23492828

RESUMO

Vitamin C is an antioxidant that maintains the activity of iron and α-ketoglutarate-dependent dioxygenases. Despite these enzymes being implicated in a wide range of biological pathways, vitamin C is rarely included in common cell culture media. Recent studies show that reprogramming of pluripotent stem cells is enhanced when vitamin C is present, thereby illustrating previous limitations in reprogramming cultures. Here, we summarize understanding of dioxygenase function in reprogramming and epigenetic regulation. The available data suggest a link between dioxygenase function and stem cell differentiation, which is exposed to environmental influence and is relevant for human disease.


Assuntos
Antioxidantes/fisiologia , Ácido Ascórbico/fisiologia , Epigênese Genética , Animais , Diferenciação Celular , Metilação de DNA , Proteínas de Ligação a DNA/fisiologia , Dioxigenases/fisiologia , Histona Desmetilases/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/enzimologia , Oxirredução , Proteínas Proto-Oncogênicas/fisiologia
15.
PLoS One ; 7(8): e40053, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22905089

RESUMO

Grade IV astrocytoma or glioblastoma has a poor clinical outcome that can be linked to hypoxia, invasiveness and active vascular remodeling. It has recently been suggested that hypoxia-inducible factors, Hifs, increase glioma growth and aggressiveness [1], [2], [3]. Here, we tested the hypothesis that Egl 9 homolog 3 (Egln3), a prolyl-hydroxylase that promotes Hif degradation, suppresses tumor progression of human and rodent glioma models. Through intracranial tumorigenesis and in vitro assays, we demonstrate for the first time that Egln3 was sufficient to decrease the kinetics of tumor progression and increase survival. We also find that Klf5, a transcription factor important to vascular remodeling, was regulated by hypoxia in glioma. An analysis of the tumor vasculature revealed that elevated Egln3 normalized glioma capillary architecture, consistent with a role for Egln3 in eliciting decreases in the production of Hif-regulated, angiogenic factors. We also find that the hydroxylase-deficient mutant, Egln3(H196A) partially maintained tumor suppressive activity. These results highlight a bifurcation of Egln3 signaling and suggest that Egln3 has a non-hydroxylase-dependent function in glioma. We conclude that Egln3 is a critical determinant of glioma formation and tumor vascular functionality.


Assuntos
Dioxigenases/fisiologia , Regulação Neoplásica da Expressão Gênica , Glioma/metabolismo , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Proteínas de Ligação a DNA/metabolismo , Dioxigenases/metabolismo , Progressão da Doença , Glioma/patologia , Glioma/terapia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia , Proteínas Imediatamente Precoces/metabolismo , Cinética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Transplante de Neoplasias , Neovascularização Patológica , Fator 3 de Transcrição de Octâmero/metabolismo , Ratos , Resultado do Tratamento
16.
Langenbecks Arch Surg ; 397(4): 603-10, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22395314

RESUMO

BACKGROUND: Since mammalian cells rely on the availability of oxygen, they have devised mechanisms to sense environmental oxygen tension, and to efficiently counteract oxygen deprivation (hypoxia). These adaptive responses to hypoxia are essentially mediated by hypoxia inducible transcription factors (HIFs). Three HIF prolyl hydroxylase enzymes (PHD1, PHD2 and PHD3) function as oxygen sensing enzymes, which regulate the activity of HIFs in normoxic and hypoxic conditions. Many of the compensatory functions exerted by the PHD-HIF system are of immediate surgical relevance since they regulate the biological response of ischemic tissues following ligation of blood vessels, of oxygen-deprived inflamed tissues, and of tumors outgrowing their vascular supply. PURPOSE: Here, we outline specific functions of PHD enzymes in surgically relevant pathological conditions, and discuss how these functions might be exploited in order to support the treatment of surgically relevant diseases.


Assuntos
Hipóxia Celular/genética , Hipóxia Celular/fisiologia , Dioxigenases/genética , Dioxigenases/fisiologia , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Pró-Colágeno-Prolina Dioxigenase/genética , Pró-Colágeno-Prolina Dioxigenase/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Vísceras/irrigação sanguínea , Vísceras/cirurgia , Neoplasias Abdominais/irrigação sanguínea , Neoplasias Abdominais/genética , Trifosfato de Adenosina/metabolismo , Animais , Morte Celular/genética , Morte Celular/fisiologia , Metabolismo Energético/genética , Metabolismo Energético/fisiologia , Homeostase/genética , Homeostase/fisiologia , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia , Imunidade Inata/genética , Imunidade Inata/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/fisiopatologia
17.
Med Oncol ; 29(4): 2710-5, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22290580

RESUMO

Hypoxia-induced factors (HIFs) play a central role in the adaptive mechanisms of cancer cells to survive under conditions of hypoxia. HIFs are regulated by prolyl hydroxylases (PHDs) among which PHD3 is implicated as a tumor suppressor. We aimed to correlate PHD3 expression with clinicopathologic parameters and to evaluate its prognostic significance in gastric cancer. The 101 tissue samples were collected from 83 resected stages I­IV gastric cancer patients, which were grouped as non-cancerous mucosa (n=18) and primary carcinoma (n=83). PHD3 expression was evaluated by immunohistochemistry. We adopted Pearson chi-square test, univariate analysis, multivariate analysis and Kaplan­Meier method. The positive frequency of PHD3 in cancer cells was 42.2%, whereas non-cancerous mucosa had no detectable PHD3. The expression of PHD3 increased significantly from non-cancerous mucosa to cancer. A significant difference was observed between PHD3 expression and tumor differentiation (P=0.007). The overexpression of PHD3 was associated with well differentiation. In univariate analyses, American Joint Committee on Cancer (AJCC) stage (P<0.0001), pT classification (P<0.0001), pN classification (P<0.0001), differentiation (P=0.0121), peritoneal metastasis (P=0.0006) and gross features (P=0.0104) were significantly associated with survival except PHD3 (P=0.2228) (Table 3). In multivariate analysis, AJCC stage was prognostically independent [hazard ratio (HR), 3.078; 95% confidence interval (CI), 2.228­4.252; P<0.0001]. Overexpression of PHD3 is a favorable prognosticator for gastric cancer. AJCC stage is an independent prognostic factor of gastric cancer.


Assuntos
Dioxigenases/fisiologia , Neoplasias Gástricas/mortalidade , Adulto , Idoso , Idoso de 80 Anos ou mais , Dioxigenases/análise , Feminino , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Estadiamento de Neoplasias , Prognóstico , Neoplasias Gástricas/enzimologia , Neoplasias Gástricas/patologia
18.
PLoS One ; 7(1): e30588, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22291995

RESUMO

BACKGROUND: ALKBH proteins, the homologs of Escherichia coli AlkB dioxygenase, constitute a direct, single-protein repair system, protecting cellular DNA and RNA against the cytotoxic and mutagenic activity of alkylating agents, chemicals significantly contributing to tumor formation and used in cancer therapy. In silico analysis and in vivo studies have shown the existence of AlkB homologs in almost all organisms. Nine AlkB homologs (ALKBH1-8 and FTO) have been identified in humans. High ALKBH levels have been found to encourage tumor development, questioning the use of alkylating agents in chemotherapy. The aim of this work was to assign biological significance to multiple AlkB homologs by characterizing their activity in the repair of nucleic acids in prokaryotes and their subcellular localization in eukaryotes. METHODOLOGY AND FINDINGS: Bioinformatic analysis of protein sequence databases identified 1943 AlkB sequences with eight new AlkB subfamilies. Since Cyanobacteria and Arabidopsis thaliana contain multiple AlkB homologs, they were selected as model organisms for in vivo research. Using E. coli alkB(-) mutant and plasmids expressing cyanobacterial AlkBs, we studied the repair of methyl methanesulfonate (MMS) and chloroacetaldehyde (CAA) induced lesions in ssDNA, ssRNA, and genomic DNA. On the basis of GFP fusions, we investigated the subcellular localization of ALKBHs in A. thaliana and established its mostly nucleo-cytoplasmic distribution. Some of the ALKBH proteins were found to change their localization upon MMS treatment. CONCLUSIONS: Our in vivo studies showed highly specific activity of cyanobacterial AlkB proteins towards lesions and nucleic acid type. Subcellular localization and translocation of ALKBHs in A. thaliana indicates a possible role for these proteins in the repair of alkyl lesions. We hypothesize that the multiplicity of ALKBHs is due to their involvement in the metabolism of nucleo-protein complexes; we find their repair by ALKBH proteins to be economical and effective alternative to degradation and de novo synthesis.


Assuntos
Dioxigenases/fisiologia , Modelos Biológicos , Homólogo AlkB 1 da Histona H2a Dioxigenase , Animais , Arabidopsis/enzimologia , Arabidopsis/genética , Arabidopsis/metabolismo , Arabidopsis/fisiologia , Células Cultivadas , Análise por Conglomerados , Biologia Computacional , Simulação por Computador , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Enzimas Reparadoras do DNA/fisiologia , Dioxigenases/genética , Dioxigenases/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/fisiologia , Teste de Complementação Genética , Humanos , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Oxigenases de Função Mista/fisiologia , Modelos Animais , Mutagênese Sítio-Dirigida , Organismos Geneticamente Modificados , Filogenia
19.
PLoS One ; 6(11): e27112, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22087251

RESUMO

Hypoxia restricts cell proliferation and cell cycle progression at the G1/S interface but at least a subpopulation of carcinoma cells can escape the restriction. In carcinoma hypoxia may in fact select for cells with enhanced hypoxic survival and increased aggressiveness. The cellular oxygen sensors HIF proline hydroxylases (PHDs) adapt the cellular functions to lowered environmental oxygen tension. PHD3 isoform has shown the strongest hypoxic upregulation among the family members. We detected a strong PHD3 mRNA expression in tumors of head and neck squamous cell carcinoma (HNSCC). The PHD3 expression associated with expression of hypoxic marker gene. Using siRNA in cell lines derived from HNSCC we show that specific inhibition of PHD3 expression in carcinoma cells caused reduced cell survival in hypoxia. The loss of PHD3, but not that of PHD2, led to marked cell number reduction. Although caspase-3 was activated at early hypoxia no induction of apoptosis was detected. However, hypoxic PHD3 inhibition caused a block in cell cycle progression. Cell population in G1 phase was increased and the population in S phase reduced demonstrating a block in G1 to S transition under PHD3 inhibition. In line with this, the level of hyperphosphorylated retinoblastoma protein Rb was reduced by PHD3 knock-down in hypoxia. PHD3 loss led to increase in cyclin-dependent kinase inhibitor p27 expression but not that of p21 or p16. The data demonstrated that increased PHD3 expression under hypoxia enhances cell cycle progression and survival of carcinoma cells.


Assuntos
Carcinoma de Células Escamosas/patologia , Dioxigenases/fisiologia , Fase G1 , Neoplasias de Cabeça e Pescoço/patologia , Hipóxia/genética , Fase S , Carcinoma de Células Escamosas/tratamento farmacológico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Dioxigenases/antagonistas & inibidores , Dioxigenases/genética , Fase G1/efeitos dos fármacos , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia , Pró-Colágeno-Prolina Dioxigenase/genética , Pró-Colágeno-Prolina Dioxigenase/fisiologia , RNA Interferente Pequeno/farmacologia , Fase S/efeitos dos fármacos , Carcinoma de Células Escamosas de Cabeça e Pescoço
20.
Br J Cancer ; 104(4): 700-6, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21285982

RESUMO

BACKGROUND: We have demonstrated for the first time that a novel human AlkB homologue, ALKBH3, contributes to prostate cancer development, but its clinical and biological roles in lung cancer remain unclear. METHODS: Expression of both mRNA and protein of PCA-1 was examined by RT-PCR and western blotting. We also assessed association with senescence and in vivo ALKBH3 treatment on orthotopic tumour cell inoculation, and analysed it clinicopathologically. RESULTS: We have since found novel biological roles for ALKBH3 in human lung cancers, particularly in adenocarcinoma. Our immunohistochemical analysis of human adenocarcinomas and squamous cell carcinomas of the lung not only showed overexpression of ALKBH3 in these tumours but the percentage of cells positive for ALKBH3 also correlated statistically to recurrence-free survival in adenocarcinoma. Knockdown of ALKBH3 by siRNA transfection induced expression of p21(WAF1/Cip1) and p27(Kip1) in the human lung adenocarcinoma cell line A549, resulting in cell cycle arrest, senescence and strong suppression of cell growth in vitro. In vivo, peritoneal tumour growth and dissemination was inhibited in nude mice, previously inoculated with the A549 cell line, by intraperitoneal injection of ALKBH3 siRNA + atelocollagen, as demonstrated by the reduction in both number and diameter of tumours developing in the peritoneum. CONCLUSION: We suggest that ALKBH3 contributes significantly to cancer cell survival and may be a therapeutic target for human adenocarcinoma of the lung.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Enzimas Reparadoras do DNA/fisiologia , Dioxigenases/fisiologia , Neoplasias Pulmonares/genética , Idoso , Idoso de 80 Anos ou mais , Homólogo AlkB 1 da Histona H2a Dioxigenase , Homólogo AlkB 3 da Dioxigenase Dependente de alfa-Cetoglutarato , Animais , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Enzimas Reparadoras do DNA/genética , Dioxigenases/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica/fisiologia , Humanos , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , RNA Interferente Pequeno/farmacologia , Homologia de Sequência , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA