Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Nanomedicine ; 19: 507-526, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38260240

RESUMEN

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.


Asunto(s)
Dioxigenasas , Inhibidores Enzimáticos , Neoplasias de la Mama Triple Negativas , Animales , Humanos , Ratones , Biomimética , Dioxigenasas/antagonistas & inhibidores , Doxorrubicina/farmacología , Inmunoterapia , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Microambiente Tumoral
2.
Biomed Pharmacother ; 166: 115324, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37598475

RESUMEN

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.


Asunto(s)
Dioxigenasas , Dioxigenasas/antagonistas & inhibidores
3.
Int J Mol Sci ; 23(23)2022 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-36499741

RESUMEN

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.


Asunto(s)
Neoplasias Óseas , Neoplasias de la Mama , Neoplasias Inflamatorias de la Mama , Femenino , Humanos , Neoplasias Óseas/genética , Neoplasias de la Mama/genética , Línea Celular Tumoral , Epigénesis Genética , Transición Epitelial-Mesenquimal/genética , Interleucina-1beta/farmacología , Células MCF-7 , Dioxigenasas/antagonistas & inhibidores , Proteínas de Unión al ADN/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología
4.
Mol Med Rep ; 25(4)2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35169856

RESUMEN

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.


Asunto(s)
Ciclohexilaminas/farmacología , Metilación de ADN/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Dioxigenasas/metabolismo , Ferroptosis/efectos de los fármacos , Células de la Granulosa/efectos de los fármacos , Oxigenasas de Función Mixta/metabolismo , Fenilendiaminas/farmacología , Proteínas Proto-Oncogénicas/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Proteínas de Unión al ADN/antagonistas & inhibidores , Dioxigenasas/antagonistas & inhibidores , Femenino , Glutatión/metabolismo , Homocisteína/toxicidad , Humanos , L-Lactato Deshidrogenasa/metabolismo , Malondialdehído/metabolismo , Oxigenasas de Función Mixta/antagonistas & inhibidores , Estrés Oxidativo/efectos de los fármacos , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Proteínas Proto-Oncogénicas/antagonistas & inhibidores
5.
J Clin Invest ; 132(4)2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35085104

RESUMEN

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.


Asunto(s)
Anemia Aplásica , Benzoatos/farmacología , Proliferación Celular , Proteínas de Unión al ADN , Dioxigenasas , Células Madre Hematopoyéticas/enzimología , Hidrazinas/farmacología , Pirazoles/farmacología , Anemia Aplásica/tratamiento farmacológico , Anemia Aplásica/genética , Anemia Aplásica/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dioxigenasas/antagonistas & inhibidores , Dioxigenasas/genética , Dioxigenasas/metabolismo , Humanos , Ratones , Ratones Noqueados
6.
J Med Chem ; 64(23): 17031-17050, 2021 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-34843649

RESUMEN

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.


Asunto(s)
Dioxigenasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Histona Demetilasas/antagonistas & inhibidores , Proteínas Nucleares/antagonistas & inhibidores , Ribosomas/enzimología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cristalización , Dioxigenasas/química , Dioxigenasas/metabolismo , Inhibidores Enzimáticos/metabolismo , Histona Demetilasas/química , Histona Demetilasas/metabolismo , Humanos , Modelos Moleculares , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Conformación Proteica , Especificidad por Sustrato
7.
Proc Natl Acad Sci U S A ; 118(34)2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34413196

RESUMEN

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.


Asunto(s)
Ácido Ascórbico/farmacología , Azacitidina/farmacología , Biomarcadores de Tumor/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/antagonistas & inhibidores , Dioxigenasas/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Antimetabolitos Antineoplásicos/farmacología , Antioxidantes/farmacología , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dioxigenasas/genética , Dioxigenasas/metabolismo , Quimioterapia Combinada , Humanos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Regiones Promotoras Genéticas , RNA-Seq , Células Tumorales Cultivadas
8.
Cancer Res ; 81(13): 3480-3494, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34127497

RESUMEN

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.


Asunto(s)
Ácido Ascórbico/farmacología , Homeostasis , Hierro/metabolismo , Mutación , Estrés Oxidativo , Succinato Deshidrogenasa/fisiología , Animales , Antioxidantes/farmacología , Dioxigenasas/antagonistas & inhibidores , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/patología , Fenotipo , Especies Reactivas de Oxígeno
9.
Cell Biol Int ; 45(8): 1654-1665, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33760331

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Nefropatías Diabéticas/metabolismo , Dioxigenasas/biosíntesis , Factor 2 Relacionado con NF-E2/biosíntesis , Transducción de Señal/fisiología , Factor de Transcripción AP-1/biosíntesis , Animales , Células Cultivadas , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/inducido químicamente , Nefropatías Diabéticas/patología , Dioxigenasas/antagonistas & inhibidores , Humanos , Masculino , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley
10.
Autophagy ; 17(11): 3444-3460, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33465003

RESUMEN

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.


Asunto(s)
Autofagia/genética , Daño del ADN , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Endodesoxirribonucleasas/metabolismo , Inestabilidad Genómica , Transporte Activo de Núcleo Celular , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dioxigenasas/antagonistas & inhibidores , Dioxigenasas/genética , Dioxigenasas/metabolismo , Endodesoxirribonucleasas/antagonistas & inhibidores , Endodesoxirribonucleasas/genética , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Neoplasias Hepáticas/metabolismo , Mitocondrias/enzimología , Mitocondrias/genética , Permeabilidad
11.
Trends Cancer ; 7(7): 635-646, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33468438

RESUMEN

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.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Proteínas de Unión al ADN/metabolismo , Dioxigenasas/metabolismo , Oxigenasas de Función Mixta/metabolismo , Neoplasias/genética , Proteínas Proto-Oncogénicas/metabolismo , 5-Metilcitosina/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinogénesis/genética , Carcinogénesis/inmunología , Carcinogénesis/patología , Ensayos Clínicos como Asunto , Metilación de ADN/efectos de los fármacos , Metilación de ADN/inmunología , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Dioxigenasas/antagonistas & inhibidores , Dioxigenasas/genética , Sinergismo Farmacológico , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/inmunología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Oxigenasas de Función Mixta/antagonistas & inhibidores , Oxigenasas de Función Mixta/genética , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/patología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Resultado del Tratamiento
12.
J Am Chem Soc ; 143(4): 1836-1845, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33373218

RESUMEN

Human indoleamine 2,3-dioxygenase 1 (hIDO1) and human tryptophan dioxygenase (hTDO) are two important heme proteins that degrade the essential amino acid, l-tryptophan (Trp), along the kynurenine pathway. The two enzymes share a similar active site structure and an analogous catalytic mechanism, but they exhibit a variety of distinct functional properties. Here we used carbon monoxide (CO) as a structural probe to interrogate how the functionalities of the two enzymes are encoded in their structures. With X-ray crystallography, we detected an unexpected photochemical intermediate trapped in a crystal of the hIDO1-CO-Trp complex, where CO is photolyzed from the heme iron by X-rays at cryogenic temperatures (100 K). The CO photolysis triggers a large-scale migration of the substrate Trp, as well as the photolyzed CO, from the active site to a temporary binding site, Sa*. It is accompanied by a large conformational change to an active site loop, JK-LoopC, despite the severely restricted protein motion under the frozen conditions, which highlights the remarkable conformational plasticity of the hIDO1 protein. Comparative studies of a crystal of the hTDO-CO-Trp complex show that CO and Trp remain bound in the active site under comparable X-ray illumination, indicating a much more rigid protein architecture. The data offer important new insights into the structure and function relationships of the heme-based dioxygenases and provide new guidelines for structure-based design of inhibitors targeting them.


Asunto(s)
Dioxigenasas/química , Hemo/química , Dominio Catalítico , Cristalografía por Rayos X , Dioxigenasas/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Procesos Fotoquímicos , Conformación Proteica , Relación Estructura-Actividad , Especificidad por Sustrato
13.
J Cereb Blood Flow Metab ; 41(3): 590-603, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32380888

RESUMEN

The 5-hydroxymethylcytosine (5hmC) epigenetic modification is highly enriched in the CNS and a critical modulator of neuronal function and development. We found that cortical 5hmC was enhanced from 5 min to three days of reperfusion following focal ischemia in adult mice. Blockade of the 5hmC-producing enzyme ten-eleven translocase 3 (TET3) increased edema, infarct volume, and motor function impairments. To determine the mechanism by which TET3 provides ischemic neuroprotection, we assessed the genomic regions where TET3 modulates 5hmC. Genome-wide sequencing analysis of differentially hydroxymethylated regions (DhMRs) revealed that focal ischemia robustly increased 5hmC at the promoters of thousands of genes in a TET3-dependent manner. TET3 inhibition reduced 5hmC at the promoters of neuroprotective genes involved in cell survival, angiogenesis, neurogenesis, antioxidant defense, DNA repair, and metabolism demonstrating a role for TET3 in endogenous protection against stroke. The mRNA expression of several genes with known involvement in ischemic neuroprotection were also reduced with TET3 knockdown in both male and female mice, establishing a correlation between decreased promoter 5hmC levels and decreased gene expression. Collectively, our results indicate that TET3 globally increases 5hmC at regulatory regions and overwhelmingly modulates 5hmC in several neuroprotective pathways that may improve outcome after ischemic injury.


Asunto(s)
Metilación de ADN , Dioxigenasas/metabolismo , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/química , 5-Metilcitosina/metabolismo , Animales , Dioxigenasas/antagonistas & inhibidores , Dioxigenasas/genética , Femenino , Regulación de la Expresión Génica/genética , Infarto de la Arteria Cerebral Media/inducido químicamente , Infarto de la Arteria Cerebral Media/complicaciones , Isquemia/etiología , Isquemia/genética , Isquemia/metabolismo , Isquemia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/química , ARN Interferente Pequeño/metabolismo
14.
Toxicology ; 447: 152631, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33188856

RESUMEN

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.


Asunto(s)
Cadmio/toxicidad , Dioxigenasas/antagonistas & inhibidores , Dioxigenasas/biosíntesis , Hígado/efectos de los fármacos , Hígado/metabolismo , Estrés Oxidativo/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Células Cultivadas , Metilación de ADN/efectos de los fármacos , Metilación de ADN/fisiología , Relación Dosis-Respuesta a Droga , Hígado/patología , Invasividad Neoplásica/patología , Estrés Oxidativo/fisiología , Ratas , Ratas Endogámicas F344
15.
Proc Natl Acad Sci U S A ; 117(7): 3621-3626, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32024762

RESUMEN

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.


Asunto(s)
Reprogramación Celular , Proteínas de Unión al ADN/antagonistas & inhibidores , Dioxigenasas/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Dominio Catalítico , Línea Celular , Reprogramación Celular/efectos de los fármacos , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dioxigenasas/química , Dioxigenasas/genética , Dioxigenasas/metabolismo , Humanos , Oxigenasas de Función Mixta/antagonistas & inhibidores , Oxigenasas de Función Mixta/química , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo
16.
Acta Biochim Biophys Sin (Shanghai) ; 52(2): 180-191, 2020 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-31990036

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/antagonistas & inhibidores , Células Endoteliales/patología , MicroARNs/farmacología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Antioxidantes/farmacología , Dioxigenasas/antagonistas & inhibidores , Células Endoteliales de la Vena Umbilical Humana , Humanos , MicroARNs/genética , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transfección
17.
J Cell Mol Med ; 23(11): 7785-7795, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31517438

RESUMEN

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.


Asunto(s)
Ciclina D1/metabolismo , Dioxigenasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Quelantes del Hierro/farmacología , Ácidos Cetoglutáricos/farmacología , Linfoma de Células del Manto/enzimología , Aminoácidos Dicarboxílicos/farmacología , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN , Deferoxamina/farmacología , Dioxigenasas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Proteína Forkhead Box O3/genética , Proteína Forkhead Box O3/metabolismo , Humanos , Hidroxilación , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Deficiencias de Hierro , ARN Mensajero/genética , ARN Mensajero/metabolismo
18.
Nucleic Acids Res ; 47(14): 7333-7347, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31165872

RESUMEN

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.


Asunto(s)
Sistemas CRISPR-Cas , Dioxigenasas/genética , Infecciones por VIH/genética , VIH-1/genética , Histona Demetilasas/genética , Proteínas Nucleares/genética , Latencia del Virus/genética , Aminopiridinas/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Línea Celular Tumoral , Células Cultivadas , Desmetilación/efectos de los fármacos , Dioxigenasas/antagonistas & inhibidores , Dioxigenasas/metabolismo , Regulación Viral de la Expresión Génica/efectos de los fármacos , Células HEK293 , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , VIH-1/fisiología , Inhibidores de Histona Desacetilasas/farmacología , Histona Demetilasas/antagonistas & inhibidores , Histona Demetilasas/metabolismo , Histonas/metabolismo , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/genética , Humanos , Hidrazonas/farmacología , Metilación/efectos de los fármacos , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Interferencia de ARN
19.
Bioorg Chem ; 88: 102809, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30999246

RESUMEN

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).


Asunto(s)
Dioxigenasas/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Hidrazonas/química , Quelantes del Hierro/química , Dioxigenasas/química , Pruebas de Enzimas , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/toxicidad , Epigénesis Genética/efectos de los fármacos , Hidrazonas/síntesis química , Hidrazonas/toxicidad , Hierro/química , Quelantes del Hierro/síntesis química , Quelantes del Hierro/toxicidad
20.
Molecules ; 24(6)2019 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-30889860

RESUMEN

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.


Asunto(s)
Productos Biológicos/farmacología , Dioxigenasas/antagonistas & inhibidores , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Naftoquinonas/farmacología , Dioxigenasas/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Simulación del Acoplamiento Molecular , Naftoquinonas/síntesis química , Naftoquinonas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...