Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Int J Nanomedicine ; 19: 507-526, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38260240

RESUMO

Introduction: Chemotherapy is still the treatment of choice for advanced triple-negative breast cancer. Chemotherapy combined with immunotherapy is being tried in patients with triple-negative breast cancer. As a kind of "cold tumor", triple-negative breast cancer has a bottleneck in immunotherapy. Indoleamine 2, 3-dioxygenase-1 inhibitors can reverse the immunosuppressive state and enhance the immune response. Methods: In this study, mesoporous silica nanoparticles were coated with the chemotherapeutic drug doxorubicin and indoleamine 2, 3-dioxygenase 1 inhibitor 1-Methyl-DL-tryptophan (1-MT), and then encapsulate the surfaces of a triple-negative breast cancer cell membrane to construct the tumor dual-targeted delivery system CDIMSN for chemotherapy and immunotherapy, and to investigate the immunogenic death effect of CDIMSN. Results and discussion: The CDIMSN could target the tumor microenvironment. Doxorubicin induced tumor immunogenic death, while 1-MT reversed immunosuppression. In vivo findings showed that the tumor size in the CDIMSN group was 2.66-fold and 1.56-fold smaller than that in DOX and DIMSN groups, respectively. CDIMSN group was better than naked DIMSN in stimulating CD8+T cells, CD4+T cells and promoting Dendritic Cells(DC) maturation. In addition, blood analysis, biochemical analysis and Hematoxylin staining analysis of mice showed that the bionic nanoparticles had good biological safety.


Assuntos
Dioxigenases , Inibidores Enzimáticos , Neoplasias de Mama Triplo Negativas , Animais , Humanos , Camundongos , Biomimética , Dioxigenases/antagonistas & inibidores , Doxorrubicina/farmacologia , Imunoterapia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Microambiente Tumoral
2.
Biomed Pharmacother ; 166: 115324, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37598475

RESUMO

TET proteins (methylcytosine dioxygenases) play an important role in the regulation of gene expression. Dysregulation of their activity is associated with many serious pathogenic states such as oncological diseases. Regulation of their activity by specific inhibitors could represent a promising therapeutic strategy. Therefore, this review describes various types of TET protein inhibitors in terms of their inhibitory mechanism and possible applicability. The potential and possible limitations of this approach are thoroughly discussed in the context of TET protein functionality in living systems. Furthermore, possible therapeutic strategies based on the inhibition of TET proteins are presented and evaluated, especially in the field of oncological diseases.


Assuntos
Dioxigenases , Dioxigenases/antagonistas & inibidores
3.
Int J Mol Sci ; 23(23)2022 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-36499741

RESUMO

The existence of a tight relationship between inflammation and epigenetics that in primary breast tumor cells can lead to tumor progression and the formation of bone metastases was investigated. It was highlighted how the induction of tumor progression and bone metastasis by Interleukin-1 beta, in a non-metastatic breast cancer cell line, MCF-7, was dependent on the de-methylating actions of ten-eleven translocation proteins (TETs). In fact, the inhibition of their activity by the Bobcat339 molecule, an inhibitor of TET enzymes, determined on the one hand, the modulation of the epithelial-mesenchymal transition process, and on the other hand, the reduction in the expression of markers of bone metastasis, indicating that the epigenetic action of TETs is a prerequisite for IL-1ß-dependent tumor progression and bone metastasis formation.


Assuntos
Neoplasias Ósseas , Neoplasias da Mama , Neoplasias Inflamatórias Mamárias , Feminino , Humanos , Neoplasias Ósseas/genética , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Epigênese Genética , Transição Epitelial-Mesenquimal/genética , Interleucina-1beta/farmacologia , Células MCF-7 , Dioxigenases/antagonistas & inibidores , Proteínas de Ligação a DNA/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia
4.
Mol Med Rep ; 25(4)2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35169856

RESUMO

Polycystic ovary syndrome is one of the most common endocrine and metabolic gynecological disorders, of which dysfunction of ovarian granulosa cells is a key contributing factor. The aim of the present study was to explore the role of ferrostatin­1 (Fer­1), a ferroptosis inhibitor, in a cell injury model established by homocysteine (Hcy)­induced ovarian granulosa KGN cell line and the potential underlying mechanism. Cell viability was measured using Cell Counting Kit­8 assay in the presence or absence of Hcy and Fer­1. Cell apoptosis was assessed using TUNEL staining and the expression levels of apoptosis­related proteins were measured using western blotting. To explore the effects of Fer­1 on oxidative stress in Hcy­treated ovarian granulosa cells, the levels of reactive oxygen species (ROS), malondialdehyde (MDA), lactate dehydrogenase (LDH) and glutathione (GSH) were measured using their corresponding kits. Furthermore, Fe2+ levels were assessed using Phen Green™ SK labeling and western blotting was performed to measure the protein expression levels of ferroptosis­associated proteins GPX4, SLC7A11, ASCL4 and DMT1. Subsequently, DNA methylation and ten­eleven translocation (TET) 1/2 demethylase levels were also detected to evaluate the extent of overall DNA methylation in ovarian granulosa cells after Hcy treatment. The TET1/2 inhibitor Bobcat339 hydrochloride was applied to treat ovarian granulosa cells before evaluating the possible effects of Fer­1 on TET1/2 and DNA methylation. Fer­1 was found to markedly elevate ovarian granulosa cell viability following Hcy treatment. The apoptosis rate in Fer­1­treated groups was also markedly decreased, which was accompanied by downregulated Bax and cleaved caspase­3 expression and upregulated Bcl­2 protein expression. In addition, Fer­1 treatment reduced the levels of ROS, MDA and LDH whilst enhancing the levels of GSH. Fe2+ levels were significantly decreased following Fer­1 treatment, which also elevated glutathione peroxidase 4 expression whilst reducing solute carrier family 7 member 11, achaete­scute family BHLH transcription factor 4 and divalent metal transporter 1 protein expression. Fer­1 significantly inhibited DNA methylation and enhanced TET1/2 levels, which were reversed by treatment with Bobcat339 hydrochloride. Subsequent experiments on cell viability, oxidative stress, Fe2+ content, ferroptosis­ and apoptosis­related proteins levels revealed that Bobcat339 hydrochloride reversed the effects of Fer­1 on ovarian granulosa Hcy­induced cell injury. These results suggest that Fer­1 may potentially protect ovarian granulosa cells against Hcy­induced injury by increasing TET levels and reducing DNA methylation.


Assuntos
Cicloexilaminas/farmacologia , Metilação de DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Dioxigenases/metabolismo , Ferroptose/efeitos dos fármacos , Células da Granulosa/efeitos dos fármacos , Oxigenases de Função Mista/metabolismo , Fenilenodiaminas/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/antagonistas & inibidores , Dioxigenases/antagonistas & inibidores , Feminino , Glutationa/metabolismo , Homocisteína/toxicidade , Humanos , L-Lactato Desidrogenase/metabolismo , Malondialdeído/metabolismo , Oxigenases de Função Mista/antagonistas & inibidores , Estresse Oxidativo/efeitos dos fármacos , Síndrome do Ovário Policístico/tratamento farmacológico , Proteínas Proto-Oncogênicas/antagonistas & inibidores
5.
J Clin Invest ; 132(4)2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35085104

RESUMO

Eltrombopag, an FDA-approved non-peptidyl thrombopoietin receptor agonist, is clinically used for the treatment of aplastic anemia, a disease characterized by hematopoietic stem cell failure and pancytopenia, to improve platelet counts and stem cell function. Eltrombopag treatment results in a durable trilineage hematopoietic expansion in patients. Some of the eltrombopag hematopoietic activity has been attributed to its off-target effects, including iron chelation properties. However, the mechanism of action for its full spectrum of clinical effects is still poorly understood. Here, we report that eltrombopag bound to the TET2 catalytic domain and inhibited its dioxygenase activity, which was independent of its role as an iron chelator. The DNA demethylating enzyme TET2, essential for hematopoietic stem cell differentiation and lineage commitment, is frequently mutated in myeloid malignancies. Eltrombopag treatment expanded TET2-proficient normal hematopoietic stem and progenitor cells, in part because of its ability to mimic loss of TET2 with simultaneous thrombopoietin receptor activation. On the contrary, TET inhibition in TET2 mutant malignant myeloid cells prevented neoplastic clonal evolution in vitro and in vivo. This mechanism of action may offer a restorative therapeutic index and provide a scientific rationale to treat selected patients with TET2 mutant-associated or TET deficiency-associated myeloid malignancies.


Assuntos
Anemia Aplástica , Benzoatos/farmacologia , Proliferação de Células , Proteínas de Ligação a DNA , Dioxigenases , Células-Tronco Hematopoéticas/enzimologia , Hidrazinas/farmacologia , Pirazóis/farmacologia , Anemia Aplástica/tratamento farmacológico , Anemia Aplástica/genética , Anemia Aplástica/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dioxigenases/antagonistas & inibidores , Dioxigenases/genética , Dioxigenases/metabolismo , Humanos , Camundongos , Camundongos Knockout
6.
J Med Chem ; 64(23): 17031-17050, 2021 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-34843649

RESUMO

MINA53 is a JmjC domain 2-oxoglutarate-dependent oxygenase that catalyzes ribosomal hydroxylation and is a target of the oncogenic transcription factor c-MYC. Despite its anticancer target potential, no small-molecule MINA53 inhibitors are reported. Using ribosomal substrate fragments, we developed mass spectrometry assays for MINA53 and the related oxygenase NO66. These assays enabled the identification of 2-(aryl)alkylthio-3,4-dihydro-4-oxoypyrimidine-5-carboxylic acids as potent MINA53 inhibitors, with selectivity over NO66 and other JmjC oxygenases. Crystallographic studies with the JmjC demethylase KDM5B revealed active site binding but without direct metal chelation; however, molecular modeling investigations indicated that the inhibitors bind to MINA53 by directly interacting with the iron cofactor. The MINA53 inhibitors manifest evidence for target engagement and selectivity for MINA53 over KDM4-6. The MINA53 inhibitors show antiproliferative activity with solid cancer lines and sensitize cancer cells to conventional chemotherapy, suggesting that further work investigating their potential in combination therapies is warranted.


Assuntos
Dioxigenases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Histona Desmetilases/antagonistas & inibidores , Proteínas Nucleares/antagonistas & inibidores , Ribossomos/enzimologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cristalização , Dioxigenases/química , Dioxigenases/metabolismo , Inibidores Enzimáticos/metabolismo , Histona Desmetilases/química , Histona Desmetilases/metabolismo , Humanos , Modelos Moleculares , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Conformação Proteica , Especificidade por Substrato
7.
Proc Natl Acad Sci U S A ; 118(34)2021 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-34413196

RESUMO

Pediatric T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy resulting from overproduction of immature T-cells in the thymus and is typified by widespread alterations in DNA methylation. As survival rates for relapsed T-ALL remain dismal (10 to 25%), development of targeted therapies to prevent relapse is key to improving prognosis. Whereas mutations in the DNA demethylating enzyme TET2 are frequent in adult T-cell malignancies, TET2 mutations in T-ALL are rare. Here, we analyzed RNA-sequencing data of 321 primary T-ALLs, 20 T-ALL cell lines, and 25 normal human tissues, revealing that TET2 is transcriptionally repressed or silenced in 71% and 17% of T-ALL, respectively. Furthermore, we show that TET2 silencing is often associated with hypermethylation of the TET2 promoter in primary T-ALL. Importantly, treatment with the DNA demethylating agent, 5-azacytidine (5-aza), was significantly more toxic to TET2-silenced T-ALL cells and resulted in stable re-expression of the TET2 gene. Additionally, 5-aza led to up-regulation of methylated genes and human endogenous retroviruses (HERVs), which was further enhanced by the addition of physiological levels of vitamin C, a potent enhancer of TET activity. Together, our results clearly identify 5-aza as a potential targeted therapy for TET2-silenced T-ALL.


Assuntos
Ácido Ascórbico/farmacologia , Azacitidina/farmacologia , Biomarcadores Tumorais/metabolismo , Metilação de DNA , Proteínas de Ligação a DNA/antagonistas & inibidores , Dioxigenases/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , Antimetabólitos Antineoplásicos/farmacologia , Antioxidantes/farmacologia , Apoptose , Biomarcadores Tumorais/genética , Proliferação de Células , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dioxigenases/genética , Dioxigenases/metabolismo , Quimioterapia Combinada , Humanos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Regiões Promotoras Genéticas , RNA-Seq , Células Tumorais Cultivadas
8.
Cancer Res ; 81(13): 3480-3494, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34127497

RESUMO

Succinate dehydrogenase is a key enzyme in the tricarboxylic acid cycle and the electron transport chain. All four subunits of succinate dehydrogenase are tumor suppressor genes predisposing to paraganglioma, but only mutations in the SDHB subunit are associated with increased risk of metastasis. Here we generated an Sdhd knockout chromaffin cell line and compared it with Sdhb-deficient cells. Both cell types exhibited similar SDH loss of function, metabolic adaptation, and succinate accumulation. In contrast, Sdhb-/- cells showed hallmarks of mesenchymal transition associated with increased DNA hypermethylation and a stronger pseudo-hypoxic phenotype compared with Sdhd-/- cells. Loss of SDHB specifically led to increased oxidative stress associated with dysregulated iron and copper homeostasis in the absence of NRF2 activation. High-dose ascorbate exacerbated the increase in mitochondrial reactive oxygen species, leading to cell death in Sdhb-/- cells. These data establish a mechanism linking oxidative stress to iron homeostasis that specifically occurs in Sdhb-deficient cells and may promote metastasis. They also highlight high-dose ascorbate as a promising therapeutic strategy for SDHB-related cancers. SIGNIFICANCE: Loss of different succinate dehydrogenase subunits can lead to different cell and tumor phenotypes, linking stronger 2-OG-dependent dioxygenases inhibition, iron overload, and ROS accumulation following SDHB mutation.


Assuntos
Ácido Ascórbico/farmacologia , Homeostase , Ferro/metabolismo , Mutação , Estresse Oxidativo , Succinato Desidrogenase/fisiologia , Animais , Antioxidantes/farmacologia , Dioxigenases/antagonistas & inibidores , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Fenótipo , Espécies Reativas de Oxigênio
9.
Cell Biol Int ; 45(8): 1654-1665, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33760331

RESUMO

TET1 mediates demethylation in tumors, but its role in diabetic nephropathy (DN), a prevalent diabetic complication, is unclear. We attempted to probe the possible mechanism of TET1 in DN. A DN rat model was established and verified by marker detection and histopathological observation. The in vitro model was established on human mesangial cells (HMCs) induced by high glucose (HG), and verified by evaluation of fibrosis and inflammation. The differentially expressed mRNA was screened out by microarray analysis. The most differentially expressed mRNA (TET1) was reduced in DN rats and HG-HMCs. The upstream and downstream factors of TET1 were verified, and their roles in DN were analyzed by gain- and loss-function assays. TET1 was decreased in DN rats and HG-HMCs. High expression of TET1 decreased biochemical indexes and renal injury of DN rats and hampered the activity, fibrosis, and inflammation of HG-HMCs. Ap1 lowered TET1 expression, and enhanced inflammation in HG-HMCs, and accentuated renal injury in DN rats. TET1 overexpression inhibited the effect of Ap1 on DN. TET1 promoted the transcription of Nrf2. The Ap1/TET1 axis mediated the Nrf2/ARE pathway activity. Overall, TET1 overexpression weakened the inhibitory effect of Ap1 on the Nrf2/ARE pathway, thus alleviating inflammation and renal injury in DN.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/metabolismo , Dioxigenases/biossíntese , Fator 2 Relacionado a NF-E2/biossíntese , Transdução de Sinais/fisiologia , Fator de Transcrição AP-1/biossíntese , Animais , Células Cultivadas , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/patologia , Nefropatias Diabéticas/induzido quimicamente , Nefropatias Diabéticas/patologia , Dioxigenases/antagonistas & inibidores , Humanos , Masculino , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley
10.
Autophagy ; 17(11): 3444-3460, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33465003

RESUMO

Genotoxic insult causes nuclear and mitochondrial DNA damages with macroautophagy/autophagy induction. The role of mitochondrial DNA (mtDNA) damage in the requirement of autophagy for nuclear DNA (nDNA) stability is unclear. Using site-specific DNA damage approaches, we show that specific nDNA damage alone does not require autophagy for repair unless in the presence of mtDNA damage. We provide evidence that after IR exposure-induced mtDNA and nDNA damages, autophagy suppression causes non-apoptotic mitochondrial permeability, by which mitochondrial ENDOG (endonuclease G) is released and translocated to nuclei to sustain nDNA damage in a TET (tet methylcytosine dioxygenase)-dependent manner. Furthermore, blocking lysosome function is sufficient to increase the amount of mtDNA leakage to the cytosol, accompanied by ENDOG-free mitochondrial puncta formation with concurrent ENDOG nuclear accumulation. We proposed that autophagy eliminates the mitochondria specified by mtDNA damage-driven mitochondrial permeability to prevent ENDOG-mediated genome instability. Finally, we showed that HBx, a hepatitis B viral protein capable of suppressing autophagy, also causes mitochondrial permeability-dependent ENDOG mis-localization in nuclei and is linked to hepatitis B virus (HBV)-mediated hepatocellular carcinoma development.Abbreviations: 3-MA: 3-methyladenine; 5-hmC: 5-hydroxymethylcytosine; ACTB: actin beta; ATG5: autophagy related 5; ATM: ATM serine/threonine kinase; DFFB/CAD: DNA fragmentation factor subunit beta; cmtDNA: cytosolic mitochondrial DNA; ConA: concanamycin A; CQ: chloroquine; CsA: cyclosporin A; Dox: doxycycline; DSB: double-strand break; ENDOG: endonuclease G; GFP: green fluorescent protein; Gy: gray; H2AX: H2A.X variant histone; HBV: hepatitis B virus; HBx: hepatitis B virus X protein; HCC: hepatocellular carcinoma; I-PpoI: intron-encoded endonuclease; IR: ionizing radiation; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MOMP: mitochondrial outer membrane permeability; mPTP: mitochondrial permeability transition pore; mtDNA: mitochondrial DNA; nDNA: nuclear DNA; 4-OHT: 4-hydroxytamoxifen; rDNA: ribosomal DNA; ROS: reactive oxygen species; SQSTM1/p62: sequestosome 1; TET: tet methylcytosine dioxygenase; TFAM: transcription factor A, mitochondrial; TOMM20: translocase of outer mitochondrial membrane 20; VDAC: voltage dependent anion channel.


Assuntos
Autofagia/genética , Dano ao DNA , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Endodesoxirribonucleases/metabolismo , Instabilidade Genômica , Transporte Ativo do Núcleo Celular , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dioxigenases/antagonistas & inibidores , Dioxigenases/genética , Dioxigenases/metabolismo , Endodesoxirribonucleases/antagonistas & inibidores , Endodesoxirribonucleases/genética , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Neoplasias Hepáticas/metabolismo , Mitocôndrias/enzimologia , Mitocôndrias/genética , Permeabilidade
11.
Trends Cancer ; 7(7): 635-646, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33468438

RESUMO

The mechanisms governing the methylome profile of tumor suppressors and oncogenes have expanded with the discovery of oxidized states of 5-methylcytosine (5mC). Ten-eleven translocation (TET) enzymes are a family of dioxygenases that iteratively catalyze 5mC oxidation and promote cytosine demethylation, thereby creating a dynamic global and local methylation landscape. While the catalytic function of TET enzymes during stem cell differentiation and development have been well studied, less is known about the multifaceted roles of TET enzymes during carcinogenesis. This review outlines several tiers of TET regulation and overviews how TET deregulation promotes a cancer phenotype. Defining the tissue-specific and context-dependent roles of TET enzymes will deepen our understanding of the epigenetic perturbations that promote or inhibit carcinogenesis.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Proteínas de Ligação a DNA/metabolismo , Dioxigenases/metabolismo , Oxigenases de Função Mista/metabolismo , Neoplasias/genética , Proteínas Proto-Oncogênicas/metabolismo , 5-Metilcitosina/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Carcinogênese/genética , Carcinogênese/imunologia , Carcinogênese/patologia , Ensaios Clínicos como Assunto , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/imunologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Dioxigenases/antagonistas & inibidores , Dioxigenases/genética , Sinergismo Farmacológico , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/imunologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Oxigenases de Função Mista/antagonistas & inibidores , Oxigenases de Função Mista/genética , Mutação , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/patologia , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Resultado do Tratamento
12.
Toxicology ; 447: 152631, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33188856

RESUMO

Cadmium (Cd) is recognized as a highly toxic heavy metal for humans in part because it is a multi-organ carcinogen. To clarify the mechanism of Cd carcinogenicity, we have established an experimental system using rat liver TRL1215 cells exposed to 2.5 µM Cd for 10 weeks and then cultured in Cd-free medium for an additional 4 weeks (total 14 weeks). Recently, we demonstrated, by using this experimental system, that 1) Cd stimulates cell invasion by suppression of apolipoprotein E (ApoE) expression, and 2) Cd induces DNA hypermethylation of the regulatory region of the ApoE gene. However, the underlying mechanism(s) as well as other potential genetic participants in the Cd-stimulated invasion are undefined. In the present work, we found that concurrent with enhanced invasion, Cd induced oxidative stress, coupled with the production of oxidative stress-sensitive metallothionein 2A (MT2A), which lead to down-modulation of ten-eleven translocation methylcytosine dioxygenase 1 (TET1: DNA demethylation) in addition to ApoE, without impacting DNA methyltransferases (DNMTs: DNA methylation) levels. Furthermore, the expression of tissue inhibitor of metalloproteinase 2 and 3 (TIMP2 and TIMP3) that are positively regulated by TET1, were decreased by Cd. The genes (ApoE/TET1/TIMP2/TIMP3) suppressed by Cd were further suppressed by hydroquinone (HQ; a reactive oxygen species [ROS] producer), whereas N-acetyl-l-cysteine (NAC; a ROS scavenger) prevented the suppression of their expression by HQ. In addition, NAC reversed their expression suppressed by Cd. Cd-stimulated cell invasion was clearly dampened by NAC in a concentration-dependent manner. Overall these findings suggest that 1) altered TET1 expression and activity together with ApoE are likely involved in the enhanced invasiveness due to Cd exposure, and 2) Cd down-regulation of TET1 likely evokes a reduction in ApoE expression (possible by DNA hypermethylation), and 3) anti-oxidants are effective in abrogation of the enhanced invasiveness that occurs concurrently with Cd-induced malignant transformation.


Assuntos
Cádmio/toxicidade , Dioxigenases/antagonistas & inibidores , Dioxigenases/biossíntese , Fígado/efeitos dos fármacos , Fígado/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Células Cultivadas , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/fisiologia , Relação Dose-Resposta a Droga , Fígado/patologia , Invasividade Neoplásica/patologia , Estresse Oxidativo/fisiologia , Ratos , Ratos Endogâmicos F344
13.
Proc Natl Acad Sci U S A ; 117(7): 3621-3626, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32024762

RESUMO

Ten-eleven translocation (TET) family enzymes (TET1, TET2, and TET3) oxidize 5-methylcytosine (5mC) and generate 5-hydroxymethylcytosine (5hmC) marks on the genome. Each TET protein also interacts with specific binding partners and partly plays their role independent of catalytic activity. Although the basic role of TET enzymes is well established now, the molecular mechanism and specific contribution of their catalytic and noncatalytic domains remain elusive. Here, by combining in silico and biochemical screening strategy, we have identified a small molecule compound, C35, as a first-in-class TET inhibitor that specifically blocks their catalytic activities. Using this inhibitor, we explored the enzymatic function of TET proteins during somatic cell reprogramming. Interestingly, we found that C35-mediated TET inactivation increased the efficiency of somatic cell programming without affecting TET complexes. Using high-throughput mRNA sequencing, we found that by targeting 5hmC repressive marks in the promoter regions, C35-mediated TET inhibition activates the transcription of the BMP-SMAD-ID signaling pathway, which may be responsible for promoting somatic cell reprogramming. These results suggest that C35 is an important tool for inducing somatic cell reprogramming, as well as for dissecting the other biological functions of TET enzymatic activities without affecting their other nonenzymatic roles.


Assuntos
Reprogramação Celular , Proteínas de Ligação a DNA/antagonistas & inibidores , Dioxigenases/antagonistas & inibidores , Inibidores Enzimáticos/química , Proteínas Proto-Oncogênicas/antagonistas & inibidores , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Domínio Catalítico , Linhagem Celular , Reprogramação Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dioxigenases/química , Dioxigenases/genética , Dioxigenases/metabolismo , Humanos , Oxigenases de Função Mista/antagonistas & inibidores , Oxigenases de Função Mista/química , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo
14.
Acta Biochim Biophys Sin (Shanghai) ; 52(2): 180-191, 2020 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-31990036

RESUMO

Endothelial cell (EC) dysfunction represents an early key event in atherosclerosis. Recently, MicroRNAs have been demonstrated to regulate EC function. miR-101-3p has been discovered to regulate cell apoptosis and proliferation in cardiovascular diseases. Therefore, the aim of the current study was to clarify whether miR-101-3p regulates the dysfunction of vascular endothelial cells. In this study, the transfection of human umbilical vein endothelial cells (HUVECs) with miR-101-3p mimic induced reactive oxygen species (ROS) production, EC dysfunction, and activated nuclear factor-κB (NF-κB), whereas transfection with miR-101-3p inhibitor alleviated these events. The antioxidant N-acetylcysteine alleviated miR-101-3p-induced EC dysfunction. Moreover, we observed that miR-101-3p inhibited the expression of tet methylcytosine dioxygenase 2 (TET2) at the posttranscriptional level, resulting in increased ROS production and activated NF-κB. TET2 overexpression inhibited ROS production, EC dysfunction, and NF-κB activation in miR-101-3p-transfected HUVECs. These results indicate that miR-101-3p induces EC dysfunction by targeting TET2, which regulates ROS production, EC dysfunction, and NF-κB activation. Taken together, our current study reveals a novel pathway associated with EC dysfunction. The modulation of miR-101-3p and TET2 expression levels may serve as a potential target for therapeutic strategies for atherosclerosis.


Assuntos
Proteínas de Ligação a DNA/antagonistas & inibidores , Células Endoteliais/patologia , MicroRNAs/farmacologia , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Antioxidantes/farmacologia , Dioxigenases/antagonistas & inibidores , Células Endoteliais da Veia Umbilical Humana , Humanos , MicroRNAs/genética , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transfecção
15.
J Cell Mol Med ; 23(11): 7785-7795, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31517438

RESUMO

The patients with mantle cell lymphoma (MCL) have translocation t(11;14) associated with cyclin D1 overexpression. We observed that iron (an essential cofactor of dioxygenases including prolyl hydroxylases [PHDs]) depletion by deferoxamine blocked MCL cells' proliferation, increased expression of DNA damage marker γH2AX, induced cell cycle arrest and decreased cyclin D1 level. Treatment of MCL cell lines with dimethyloxalylglycine, which blocks dioxygenases involving PHDs by competing with their substrate 2-oxoglutarate, leads to their decreased proliferation and the decrease of cyclin D1 level. We then postulated that loss of EGLN2/PHD1 in MCL cells may lead to down-regulation of cyclin D1 by blocking the degradation of FOXO3A, a cyclin D1 suppressor. However, the CRISPR/Cas9-based loss-of-function of EGLN2/PHD1 did not affect cyclin D1 expression and the loss of FOXO3A did not restore cyclin D1 levels after iron chelation. These data suggest that expression of cyclin D1 in MCL is not controlled by ENGL2/PHD1-FOXO3A pathway and that chelation- and 2-oxoglutarate competition-mediated down-regulation of cyclin D1 in MCL cells is driven by yet unknown mechanism involving iron- and 2-oxoglutarate-dependent dioxygenases other than PHD1. These data support further exploration of the use of iron chelation and 2-oxoglutarate-dependent dioxygenase inhibitors as a novel therapy of MCL.


Assuntos
Ciclina D1/metabolismo , Dioxigenases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Quelantes de Ferro/farmacologia , Ácidos Cetoglutáricos/farmacologia , Linfoma de Célula do Manto/enzimologia , Aminoácidos Dicarboxílicos/farmacologia , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Dano ao DNA , Desferroxamina/farmacologia , Dioxigenases/metabolismo , Regulação para Baixo/efeitos dos fármacos , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Humanos , Hidroxilação , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Deficiências de Ferro , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
16.
Nucleic Acids Res ; 47(14): 7333-7347, 2019 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-31165872

RESUMO

Although combination antiretroviral therapy is potent to block active replication of HIV-1 in AIDS patients, HIV-1 persists as transcriptionally inactive proviruses in infected cells. These HIV-1 latent reservoirs remain a major obstacle for clearance of HIV-1. Investigation of host factors regulating HIV-1 latency is critical for developing novel antiretroviral reagents to eliminate HIV-1 latent reservoirs. From our recently accomplished CRISPR/Cas9 sgRNA screens, we identified that the histone demethylase, MINA53, is potentially a novel HIV-1 latency-promoting gene (LPG). We next validated MINA53's function in maintenance of HIV-1 latency by depleting MINA53 using the alternative RNAi approach. We further identified that in vitro MINA53 preferentially demethylates the histone substrate, H3K36me3 and that in cells MINA53 depletion by RNAi also increases the local level of H3K36me3 at LTR. The effort to map the downstream effectors unraveled that H3K36me3 has the cross-talk with another epigenetic mark H4K16ac, mediated by KAT8 that recognizes the methylated H3K36 and acetylated H4K16. Removing the MINA53-mediated latency mechanisms could benefit the reversal of post-integrated latent HIV-1 proviruses for purging of reservoir cells. We further demonstrated that a pan jumonji histone demethylase inhibitor, JIB-04, inhibits MINA53-mediated demethylation of H3K36me3, and JIB-04 synergizes with other latency-reversing agents (LRAs) to reactivate latent HIV-1.


Assuntos
Sistemas CRISPR-Cas , Dioxigenases/genética , Infecções por HIV/genética , HIV-1/genética , Histona Desmetilases/genética , Proteínas Nucleares/genética , Latência Viral/genética , Aminopiridinas/farmacologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Linhagem Celular Tumoral , Células Cultivadas , Desmetilação/efeitos dos fármacos , Dioxigenases/antagonistas & inibidores , Dioxigenases/metabolismo , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Infecções por HIV/metabolismo , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , HIV-1/fisiologia , Inibidores de Histona Desacetilases/farmacologia , Histona Desmetilases/antagonistas & inibidores , Histona Desmetilases/metabolismo , Histonas/metabolismo , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/genética , Humanos , Hidrazonas/farmacologia , Metilação/efeitos dos fármacos , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Interferência de RNA
17.
Bioorg Chem ; 88: 102809, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30999246

RESUMO

Ten-eleven translocation protein (TET) 1 plays a key role in control of DNA demethylation and thereby of gene expression. Dysregulation of these processes leads to serious pathological states such as oncological and neurodegenerative ones and thus TET 1 targeting is highly requested. Therefore, in this work, we examined the ability of hydrazones (acyl-, aroyl- and heterocyclic hydrazones) to inhibit the TET 1 protein and its mechanism of action. Inhibitory activity of hydrazones 1-7 towards TET 1 was measured. The results showed a high affinity of the tested chelators for iron(II). The study clearly showed a significant correlation between the chelator's affinity for iron(II) ions (represented by the binding constant) and TET 1 protein inhibitory activity (represented by IC50 values).


Assuntos
Dioxigenases/antagonistas & inibidores , Inibidores Enzimáticos/química , Hidrazonas/química , Quelantes de Ferro/química , Dioxigenases/química , Ensaios Enzimáticos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/toxicidade , Epigênese Genética/efeitos dos fármacos , Hidrazonas/síntese química , Hidrazonas/toxicidade , Ferro/química , Quelantes de Ferro/síntese química , Quelantes de Ferro/toxicidade
18.
Molecules ; 24(6)2019 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-30889860

RESUMO

There is great interest in developing small molecules agents capable of reversing tumor immune escape to restore the body's immune system. As an immunosuppressive enzyme, indoleamine 2,3-dioxygenase 1 (IDO-1) is considered a promising target for oncology immunotherapy. Currently, none of IDO-1 inhibitors have been launched for clinical practice yet. Thus, the discovery of new IDO-1 inhibitors is still in great demand. Herein, a series of diverse ortho-naphthaquinone containing natural product derivatives were synthesized as novel IDO-1 inhibitors. Among them, 1-ene-3-ketone-17-hydroxyl derivative 12 exhibited significantly improved enzymatic and cellular inhibitory activity against IDO-1 when compared to initial lead compounds. Besides, the molecular docking study disclosed that the two most potent compounds 11 and 12 have more interactions within the binding pocket of IDO-1 via hydrogen-bonding, which may account for their higher IDO-1 inhibitory activity.


Assuntos
Produtos Biológicos/farmacologia , Dioxigenases/antagonistas & inibidores , Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Naftoquinonas/farmacologia , Dioxigenases/metabolismo , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Simulação de Acoplamento Molecular , Naftoquinonas/síntese química , Naftoquinonas/química
19.
J Cell Biol ; 218(1): 317-332, 2019 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-30487181

RESUMO

The process by which tumor cells mechanically invade through surrounding stroma into peripheral tissues is an essential component of metastatic dissemination. The directed recruitment of the metalloproteinase MT1-MMP to invadopodia plays a critical role in this invasive process. Here, we provide mechanistic insight into MT1-MMP cytoplasmic tail binding protein 1 (MTCBP-1) with respect to invadopodia formation, matrix remodeling, and invasion by pancreatic tumor cells. MTCBP-1 localizes to invadopodia and interacts with MT1-MMP. We find that this interaction displaces MT1-MMP from invadopodia, thereby attenuating their number and function and reducing the capacity of tumor cells to degrade matrix. Further, we observe an inverse correlation between MTCBP-1 and MT1-MMP expression both in cultured cell lines and human pancreatic tumors. Consistently, MTCBP-1-expressing cells show decreased ability to invade in vitro and metastasize in vivo. These findings implicate MTCBP-1 as an inhibitor of the metastatic process.


Assuntos
Dioxigenases/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Intestinais/genética , Metaloproteinase 14 da Matriz/genética , Neoplasias Pancreáticas/genética , Podossomos/genética , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Dioxigenases/antagonistas & inibidores , Dioxigenases/metabolismo , Humanos , Neoplasias Intestinais/metabolismo , Neoplasias Intestinais/mortalidade , Neoplasias Intestinais/secundário , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Metástase Linfática , Metaloproteinase 14 da Matriz/metabolismo , Camundongos , Camundongos Nus , Invasividade Neoplásica , Pâncreas/metabolismo , Pâncreas/patologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/patologia , Podossomos/metabolismo , Podossomos/patologia , Ligação Proteica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Análise de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Cell Death Dis ; 9(11): 1062, 2018 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-30333481

RESUMO

MYC-induced nuclear antigen (MINA53) is a JmjC (jumonji C domain)-containing protein, which is highly expressed in many cancers including glioblastoma. We have revealed in our previous report that MINA53 is a poor prognostic indicator for glioblastoma patients, and knockdown of MINA53 could reduce glioblastoma malignancy. In this study, we found that MINA53 knockdown could decrease the DNA replication initiation in glioblastoma cells. Through further investigations, we revealed that MINA53 could regulate the expression of the CDC45-MCM-GINS (CMG) complex genes, which are vital for DNA replication initiation. Knockdown of MINA53 reduced the CMG genes expression and thus induced DNA replication stress and DNA damage. Furthermore, MINA53 knockdown diminished DNA damage response (DDR) by reducing the ATM/ATR-H2AX pathway activity and finally led glioblastoma cells to apoptosis and death. We further applied a genotoxic drug Doxorubicin and found that MINA53 deficiency sensitized glioblastoma cells to Doxorubicin. Our study reveals that MINA53 is involved in DNA replication initiation and DNA damage response, and provides support for MINA53 as a novel and potential therapeutic target for glioblastoma treatment.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Reparo do DNA , DNA de Neoplasias/genética , Dioxigenases/genética , Doxorrubicina/farmacologia , Regulação Neoplásica da Expressão Gênica , Histona Desmetilases/genética , Proteínas Nucleares/genética , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Dano ao DNA , Replicação do DNA , DNA de Neoplasias/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dioxigenases/antagonistas & inibidores , Dioxigenases/metabolismo , Células HEK293 , Histona Desmetilases/antagonistas & inibidores , Histona Desmetilases/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Proteínas de Manutenção de Minicromossomo/genética , Proteínas de Manutenção de Minicromossomo/metabolismo , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neuroglia/patologia , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA