Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.893
Filtrar
1.
Int J Mol Sci ; 23(6)2022 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-35328634

RESUMO

Approximately one-third of the human population is infected with the intracellular cosmopolitan protozoan Toxoplasma gondii (Tg), and a specific treatment for this parasite is still needed. Additionally, the increasing resistance of Tg to drugs has become a challenge for numerous research centers. The high selectivity of a compound toward the protozoan, along with low cytotoxicity toward the host cells, form the basis for further research, which aims at determining the molecular targets of the active compounds. Thiosemicarbazide derivatives are biologically active organic compounds. Previous studies on the initial preselection of 58 new 4-arylthiosemicarbazide derivatives in terms of their anti-Tg activity and selectivity made it possible to select two promising derivatives for further research. One of the important amino acids involved in the proliferation of Tg and the formation of parasitophorous vacuoles is tyrosine, which is converted by two unique aromatic amino acid hydroxylases to levodopa. Enzymatic studies with two derivatives (R: para-nitro and meta-iodo) and recombinant aromatic amino acid hydroxylase (AAHs) obtained in the E. coli expression system were performed, and the results indicated that toxoplasmic AAHs are a molecular target for 4-arylthiosemicarbazide derivatives. Moreover, the drug affinity responsive target stability assay also confirmed that the selected compounds bind to AAHs. Additionally, the anti-inflammatory activity of these derivatives was tested using THP1-Blue™ NF-κB reporter cells due to the similarity of the thiosemicarbazide scaffold to thiosemicarbazone, both of which are known NF-κB pathway inhibitors.


Assuntos
Anti-Inflamatórios , Antiprotozoários , Oxigenases de Função Mista , Semicarbazidas , Toxoplasma , Anti-Inflamatórios/farmacologia , Antiprotozoários/farmacologia , Escherichia coli , Humanos , Oxigenases de Função Mista/antagonistas & inibidores , NF-kappa B , Semicarbazidas/farmacologia , Toxoplasma/efeitos dos fármacos , Tirosina
2.
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
3.
Biomolecules ; 11(12)2021 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-34944533

RESUMO

Fermented persimmon juice, Kakishibu, has traditionally been used for wood and paper protection. This protective effect stems at least partially from inhibition of microbial cellulose degrading enzymes. The inhibitory effect of Kakishibu on lytic polysaccharide monooxygenases (LPMOs) and on a cocktail of cellulose hydrolases was studied, using three different cellulosic substrates. Dose dependent inhibition of LPMO activity by a commercial Kakishibu product was assessed for the well-characterized LPMO from Thermoascus aurantiacus TaAA9A, and the inhibitory effect was confirmed on five additional microbial LPMOs. The model tannin compound, tannic acid exhibited a similar inhibitory effect on TaAA9A as Kakishibu. It was further shown that both polyethylene glycol and tannase can alleviate the inhibitory effect of Kakishibu and tannic acid, indicating a likely mechanism of inhibition caused by unspecific tannin-protein interactions.


Assuntos
Diospyros/química , Inibidores Enzimáticos/farmacologia , Sucos de Frutas e Vegetais/microbiologia , Oxigenases de Função Mista/antagonistas & inibidores , Thermoascus/enzimologia , Hidrolases de Éster Carboxílico/efeitos adversos , Diospyros/microbiologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/química , Fermentação , Sucos de Frutas e Vegetais/análise , Proteínas Fúngicas/antagonistas & inibidores , Regulação Fúngica da Expressão Gênica/efeitos dos fármacos , Hidrolases/antagonistas & inibidores , Polietilenoglicóis/efeitos adversos , Taninos/farmacologia , Thermoascus/efeitos dos fármacos
4.
Cells ; 10(5)2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-34062826

RESUMO

Alkylglycerol monooxygenase (AGMO) is a tetrahydrobiopterin (BH4)-dependent enzyme with major expression in the liver and white adipose tissue that cleaves alkyl ether glycerolipids. The present study describes the disclosure and biological characterization of a candidate compound (Cp6), which inhibits AGMO with an IC50 of 30-100 µM and 5-20-fold preference of AGMO relative to other BH4-dependent enzymes, i.e., phenylalanine-hydroxylase and nitric oxide synthase. The viability and metabolic activity of mouse 3T3-L1 fibroblasts, HepG2 human hepatocytes and mouse RAW264.7 macrophages were not affected up to 10-fold of the IC50. However, Cp6 reversibly inhibited the differentiation of 3T3-L1 cells towards adipocytes, in which AGMO expression was upregulated upon differentiation. Cp6 reduced the accumulation of lipid droplets in adipocytes upon differentiation and in HepG2 cells exposed to free fatty acids. Cp6 also inhibited IL-4-driven differentiation of RAW264.7 macrophages towards M2-like macrophages, which serve as adipocyte progenitors in adipose tissue. Collectively, the data suggest that pharmacologic AGMO inhibition may affect lipid storage.


Assuntos
Adipogenia/efeitos dos fármacos , Tecido Adiposo/efeitos dos fármacos , Oxigenases de Função Mista/antagonistas & inibidores , Células 3T3-L1 , Adipócitos/metabolismo , Animais , Diferenciação Celular , Fibroblastos/metabolismo , Células Hep G2 , Humanos , Concentração Inibidora 50 , Metabolismo dos Lipídeos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase/metabolismo , Células RAW 264.7 , Ratos , Ratos Sprague-Dawley
5.
Cell Death Differ ; 28(9): 2818-2836, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33953349

RESUMO

The biological function of PRMT5 remains poorly understood in cervical cancer metastasis. Here, we report that PRMT5 physically associates with the transcription factor Snail and the NuRD(MTA1) complex to form a transcriptional-repressive complex that catalyzes the symmetrical histone dimethylation and deacetylation. This study shows that the Snail/PRMT5/NuRD(MTA1) complex targets genes, such as TET1 and E-cadherin, which are critical for epithelial-mesenchymal transition (EMT). This complex also affects the conversion of 5mC to 5hmC. This study demonstrates that the Snail/PRMT5/NuRD(MTA1) complex promotes the invasion and metastasis of cervical cancer in vitro and in vivo. This study also shows that PRMT5 expression is upregulated in cervical cancer and various human cancers, and the PRMT5 inhibitor EPZ015666 suppresses EMT and the invasion potential of cervical cancer cells by disinhibiting the expression of TET1 and increasing 5hmC, suggesting that PRMT5 is a potential target for cancer therapy.


Assuntos
Metilação de DNA/genética , Epigenômica/métodos , Oxigenases de Função Mista/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Fatores de Transcrição da Família Snail/genética , Transfecção/métodos , Neoplasias do Colo do Útero/genética , Animais , Feminino , Humanos , Camundongos , Camundongos Nus , Transdução de Sinais
6.
J Med Chem ; 64(11): 7189-7209, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-34029087

RESUMO

Factor inhibiting hypoxia-inducible factor (FIH) is a JmjC domain 2-oxogluarate and Fe(II)-dependent oxygenase that catalyzes hydroxylation of specific asparagines in the C-terminal transcriptional activation domain of hypoxia-inducible factor alpha (HIF-α) isoforms. This modification suppresses the transcriptional activity of HIF by reducing its interaction with the transcriptional coactivators p300/CBP. By contrast with inhibition of the HIF prolyl hydroxylases (PHDs), inhibitors of FIH, which accepts multiple non-HIF substrates, are less studied; they are of interest due to their potential ability to alter metabolism (either in a HIF-dependent and/or -independent manner) and, provided HIF is upregulated, to modulate the course of the HIF-mediated hypoxic response. Here we review studies on the mechanism and inhibition of FIH. We discuss proposed biological roles of FIH including its regulation of HIF activity and potential roles of FIH-catalyzed oxidation of non-HIF substrates. We highlight potential therapeutic applications of FIH inhibitors.


Assuntos
Oxigenases de Função Mista/metabolismo , Oxigênio/metabolismo , Proteínas Repressoras/metabolismo , Asparagina/metabolismo , Humanos , Hidroxilação , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/química , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Proteínas I-kappa B/metabolismo , Oxigenases de Função Mista/antagonistas & inibidores , Oxigênio/química , Proteínas Repressoras/antagonistas & inibidores , Especificidade por Substrato , Fatores de Transcrição de p300-CBP/química , Fatores de Transcrição de p300-CBP/metabolismo
7.
Methods Mol Biol ; 2272: 181-194, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34009614

RESUMO

Methylation of DNA at cytosine bases is an important DNA modification underlying normal development and disease states. Despite decades of research into the biological function of DNA methylation, most of the observations so far have relied primarily on associative data between observed changes in DNA methylation states and local changes in transcriptional activity or chromatin state processes. This is primarily due to the lack of molecular tools to precisely modify DNA methylation in the genome. Recent advances in genome editing technologies have allowed repurposing the CRISPR-Cas9 system for epigenome editing by fusing the catalytically dead Cas9 (dCas9) to epigenome modifying enzymes. Moreover, methods of recruiting multiple protein domains, including the SunTag system, have increased the efficacy of epigenome editing at target sites. Here, we describe an end-to-end protocol for efficient targeted removal of DNA methylation by recruiting multiple catalytic domain of TET1 enzymes to the target sites with the dCas9-SunTag system, including sgRNA design, molecular cloning, delivery of plasmid into mammalian cells, and targeted DNA methylation analysis.


Assuntos
Sistemas CRISPR-Cas , Desmetilação do DNA , DNA/análise , Edição de Genes , Genoma Humano , Oxigenases de Função Mista/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Sulfitos/química , Cromatina , Biologia Computacional/métodos , DNA/química , DNA/genética , Epigênese Genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Oxigenases de Função Mista/antagonistas & inibidores , Oxigenases de Função Mista/genética , Oxirredução , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética
8.
Methods Mol Biol ; 2272: 195-206, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34009615

RESUMO

Aberrant promoter hypermethylation leads to gene silencing and is associated with various pathologies including cancer and organ fibrosis. Active DNA demethylation is mediated by TET enzymes: TET1, TET2, and TET3, which convert 5-methylcytosine to 5-hydroxymethylcytosine. Induction of gene-specific hydroxymethylation via CRISPR/Cas9 gene technology provides an opportunity to reactivate a single target gene silenced in pathological conditions. We utilized a spCas9 variant fused with TET3 catalytic domain to mediate gene-specific hydroxymethylation with subsequent gene reactivation which holds promise for gene therapy. Here, we present guidelines for gene-specific hydroxymethylation targeting with a specific focus on designing sgRNA and functional assessments in vitro.


Assuntos
5-Metilcitosina/análogos & derivados , 5-Metilcitosina/química , Sistemas CRISPR-Cas , Metilação de DNA , DNA/análise , Edição de Genes , Oxigenases de Função Mista/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Cromatina , Biologia Computacional/métodos , DNA/química , DNA/genética , Epigênese Genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Oxigenases de Função Mista/antagonistas & inibidores , Oxigenases de Função Mista/genética , Oxirredução , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Sulfitos/química
9.
Int J Mol Sci ; 22(5)2021 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-33668187

RESUMO

Xenoantigens cause hyperacute rejection and limit the success of interspecific xenografts. Therefore, genes involved in xenoantigen biosynthesis, such as GGTA1, CMAH, and B4GALNT2, are key targets to improve the outcomes of xenotransplantation. In this study, we introduced a CRISPR/Cas9 system simultaneously targeting GGTA1, CMAH, and B4GALNT2 into in vitro-fertilized zygotes using electroporation for the one-step generation of multiple gene-edited pigs without xenoantigens. First, we optimized the combination of guide RNAs (gRNAs) targeting GGTA1 and CMAH with respect to gene editing efficiency in zygotes, and transferred electroporated embryos with the optimized gRNAs and Cas9 into recipient gilts. Next, we optimized the Cas9 protein concentration with respect to the gene editing efficiency when GGTA1, CMAH, and B4GALNT2 were targeted simultaneously, and generated gene-edited pigs using the optimized conditions. We achieved the one-step generation of GGTA1/CMAH double-edited pigs and GGTA1/CMAH/B4GALNT2 triple-edited pigs. Immunohistological analyses demonstrated the downregulation of xenoantigens; however, these multiple gene-edited pigs were genetic mosaics that failed to knock out some xenoantigens. Although mosaicism should be resolved, the electroporation technique could become a primary method for the one-step generation of multiple gene modifications in pigs aimed at improving pig-to-human xenotransplantation.


Assuntos
Animais Geneticamente Modificados/genética , Antígenos Heterófilos/biossíntese , Sistemas CRISPR-Cas , Galactosiltransferases/antagonistas & inibidores , Oxigenases de Função Mista/antagonistas & inibidores , N-Acetilgalactosaminiltransferases/antagonistas & inibidores , Zigoto/fisiologia , Animais , Feminino , Edição de Genes , Suínos
10.
Chem Biol Drug Des ; 97(6): 1170-1184, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33764683

RESUMO

DNA alkylation damage, emanating from the exposure to environmental alkylating agents or produced by certain endogenous metabolic processes, affects cell viability and genomic stability. Fe(II)/2-oxoglutarate-dependent dioxygenase enzymes, such as Escherichia coli AlkB, are involved in protecting DNA from alkylation damage. Inspired by the natural product indenone derivatives reported to inhibit this class of enzymes, and a set of 2-chloro-3-amino indenone derivatives was synthesized and screened for their inhibitory properties against AlkB. The synthesis of 2-chloro-3-amino indenone derivatives was achieved from 2,3-dichloro indenones through addition-elimination method using alkyl/aryl amines under catalyst-free conditions. Using an in vitro reconstituted DNA repair assay, we have identified a 2-chloro-3-amino indenone compound 3o to be an inhibitor of AlkB. We have determined the binding affinity, mode of interaction, and kinetic parameters of inhibition of 3o and tested its ability to sensitize cells to methyl methanesulfonate that mainly produce DNA alkylation damage. This study established the potential of indenone-derived compounds as inhibitors of Fe(II)/2-oxoglutarate-dependent dioxygenase AlkB.


Assuntos
Alquilantes/síntese química , Reparo do DNA , Indenos/química , Alquilantes/farmacologia , Sítios de Ligação , Dano ao DNA , Desmetilação do DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Escherichia coli/enzimologia , Proteínas de Escherichia coli/antagonistas & inibidores , Proteínas de Escherichia coli/metabolismo , Humanos , Indenos/metabolismo , Indenos/farmacologia , Cinética , Oxigenases de Função Mista/antagonistas & inibidores , Oxigenases de Função Mista/metabolismo , Simulação de Acoplamento Molecular , Ligação Proteica
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.
Arch Biochem Biophys ; 699: 108765, 2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33460580

RESUMO

Flavin-dependent monooxygenases catalyze a wide variety of redox reactions in important biological processes and are responsible for the synthesis of highly complex natural products. Although much has been learned about FMO chemistry in the last ~80 years of research, several aspects of the reactions catalyzed by these enzymes remain unknown. In this review, we summarize recent advancements in the flavin-dependent monooxygenase field including aspects of flavin dynamics, formation and stabilization of reactive species, and the hydroxylation mechanism. Novel catalysis of flavin-dependent N-oxidases involving consecutive oxidations of amines to generate oximes or nitrones is presented and the biological relevance of the products is discussed. In addition, the activity of some FMOs have been shown to be essential for the virulence of several human pathogens. We also discuss the biomedical relevance of FMOs in antibiotic resistance and the efforts to identify inhibitors against some members of this important and growing family enzymes.


Assuntos
Flavoproteínas/química , Oxigenases de Função Mista/química , Animais , Bactérias/enzimologia , Biocatálise , Descoberta de Drogas , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Flavinas/química , Flavinas/metabolismo , Flavoproteínas/antagonistas & inibidores , Flavoproteínas/metabolismo , Humanos , Hidroxilação , Oxigenases de Função Mista/antagonistas & inibidores , Oxigenases de Função Mista/metabolismo , Ligação Proteica , Conformação Proteica
13.
Dig Dis Sci ; 66(4): 1080-1089, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32445050

RESUMO

BACKGROUND: Cholangiocarcinoma is a devastating disease with a 2% 5-year survival if the disease has spread outside the liver. The enzyme aspartate beta-hydroxylase (ASPH) has been demonstrated to be highly expressed in cholangiocarcinoma but not in normal bile ducts and found to stimulate tumor cell migration. In addition, it was found that targeting ASPH inhibits cholangiocarcinoma malignant progression. However, it is not clear whether targeting ASPH with the small molecule inhibitor MO-I-1182 suppresses cholangiocarcinoma metastasis. The current study aims to study the efficacy of MO-I-1182 in suppressing cholangiocarcinoma metastasis. METHODS: The analysis was performed in vitro and in vivo with a preclinical animal model by using molecular and biochemical strategies to regulate ASPH expression and function. RESULTS: Knockdown of ASPH substantially inhibited cell migration and invasion in two human cholangiocarcinoma cell lines. Targeting ASPH with a small molecule inhibitor suppressed cholangiocarcinoma progression. Molecular mechanism studies demonstrated that knockdown of ASPH subsequently suppressed protein levels of the matrix metalloproteinases. The ASPH knockdown experiments suggest that this enzyme may modulate cholangiocarcinoma metastasis by regulating matrix metalloproteinases expression. Furthermore, using an ASPH inhibitor in a rat cholangiocarcinoma intrahepatic model established with BED-Neu-CL#24 cholangiocarcinoma cells, it was found that targeting ASPH inhibited intrahepatic cholangiocarcinoma metastasis and downstream expression of the matrix metalloproteinases. CONCLUSION: ASPH may modulate cholangiocarcinoma metastasis via matrix metalloproteinases expression. Taken together, targeting ASPH function may inhibit intrahepatic cholangiocarcinoma metastasis and improve survival.


Assuntos
Proteínas de Ligação ao Cálcio , Colangiocarcinoma , Inibidores Enzimáticos/farmacologia , Neoplasias Hepáticas , Proteínas de Membrana , Oxigenases de Função Mista , Proteínas Musculares , Metástase Neoplásica/prevenção & controle , Animais , Ductos Biliares Intra-Hepáticos/metabolismo , Ductos Biliares Intra-Hepáticos/patologia , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Proteínas de Ligação ao Cálcio/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Colangiocarcinoma/tratamento farmacológico , Colangiocarcinoma/metabolismo , Colangiocarcinoma/patologia , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Metaloproteinases da Matriz/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Oxigenases de Função Mista/antagonistas & inibidores , Oxigenases de Função Mista/metabolismo , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/metabolismo , Ratos
14.
J Atheroscler Thromb ; 28(7): 716-729, 2021 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-32981917

RESUMO

AIM: During the development of atherosclerosis, the vascular smooth muscle cells (SMCs) undergo phenotypic switching from contractile phenotype to synthetic phenotype. This study aimed at examining the role of DNA modification mediated by the oxidative stress dependent ten eleven translocation enzymes (TETs) expression at early stage of phenotypic switching. METHODS: Based on the in vitro SMCs calcification model, DNA damage, phenotypic switching and 5-hydroxymethylcytosine (5hmC) were examined by comet assay, alkaline DNA unwinding assay, immunofluorescence staining, Dot blotting and Western blotting. Then Western blotting and qRT-PCR were performed to analyze the TETs expression and the relationship between the activity of poly(ADP-ribose) polymerase 1 (PARP1) and TETs expression. We further alter 5hmC modification by inhibition of TET1 or PARP1 to rescue the phenotypic switching of SMCs using immunofluorescence staining, Dot blotting and qRT-PCR. We performed immunochemistry staining to examine the activated PARP1-TET1 pathway in vivo. RESULTS: The phenotypic switching was observed in the SMCs cultured with calcification medium as the expression of the cell markers of contractile SMCs decreased and cell proliferation increased. In contrast, PAR and 5hmC were markedly increased in SMCs with calcification due to DNA damage. Our study further demonstrated that oxidative stress-activated PARP1, promotes TET1 expression and 5hmC increase during the phenotypic switching. Inhibition of TET1 or PARP1 can rescue the phenotypic switching of SMCs with calcification. CONCLUSION: Our study demonstrated the important role of PARylation dependent 5hmC, in SMCs phenotypic switching. It raises the possibility to target TET1 and PARP1 for atherosclerosis treatment.


Assuntos
Aterosclerose , Oxigenases de Função Mista , Músculo Liso Vascular , Poli(ADP-Ribose) Polimerase-1 , Proteínas Proto-Oncogênicas , Calcificação Vascular , Aterosclerose/metabolismo , Aterosclerose/fisiopatologia , Aterosclerose/prevenção & controle , Plasticidade Celular , Proliferação de Células , Transdiferenciação Celular , Células Cultivadas , Descoberta de Drogas , Humanos , Oxigenases de Função Mista/antagonistas & inibidores , Oxigenases de Função Mista/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Poli(ADP-Ribose) Polimerase-1/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Calcificação Vascular/tratamento farmacológico , Calcificação Vascular/metabolismo , Calcificação Vascular/fisiopatologia
15.
J Biol Chem ; 296: 100038, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33158989

RESUMO

Microbial metabolism of carnitine to trimethylamine (TMA) in the gut can accelerate atherosclerosis and heart disease, and these TMA-producing enzymes are therefore important drug targets. Here, we report the first structures of the carnitine oxygenase CntA, an enzyme of the Rieske oxygenase family. CntA exists in a head-to-tail α3 trimeric structure. The two functional domains (the Rieske and the catalytic mononuclear iron domains) are located >40 Å apart in the same monomer but adjacent in two neighboring monomers. Structural determination of CntA and subsequent electron paramagnetic resonance measurements uncover the molecular basis of the so-called bridging glutamate (E205) residue in intersubunit electron transfer. The structures of the substrate-bound CntA help to define the substrate pocket. Importantly, a tyrosine residue (Y203) is essential for ligand recognition through a π-cation interaction with the quaternary ammonium group. This interaction between an aromatic residue and quaternary amine substrates allows us to delineate a subgroup of Rieske oxygenases (group V) from the prototype ring-hydroxylating Rieske oxygenases involved in bioremediation of aromatic pollutants in the environment. Furthermore, we report the discovery of the first known CntA inhibitors and solve the structure of CntA in complex with the inhibitor, demonstrating the pivotal role of Y203 through a π-π stacking interaction with the inhibitor. Our study provides the structural and molecular basis for future discovery of drugs targeting this TMA-producing enzyme in human gut.


Assuntos
Carnitina/metabolismo , Oxigenases de Função Mista/metabolismo , Catálise , Espectroscopia de Ressonância de Spin Eletrônica , Transporte de Elétrons , Oxigenases de Função Mista/antagonistas & inibidores , Oxigenases de Função Mista/química , Conformação Proteica , Especificidade por Substrato
16.
Bioorg Med Chem ; 28(20): 115675, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-33069066

RESUMO

Human aspartate/asparagine-ß-hydroxylase (AspH) is a 2-oxoglutarate (2OG) dependent oxygenase that catalyses the hydroxylation of Asp/Asn-residues of epidermal growth factor-like domains (EGFDs). AspH is reported to be upregulated on the cell surface of invasive cancer cells in a manner distinguishing healthy from cancer cells. We report studies on the effect of small-molecule active pharmaceutical ingredients (APIs) of human cancer therapeutics on the catalytic activity of AspH using a high-throughput mass spectrometry (MS)-based inhibition assay. Human B-cell lymphoma-2 (Bcl-2)-protein inhibitors, including the (R)-enantiomer of the natural product gossypol, were observed to efficiently inhibit AspH, as does the antitumor antibiotic bleomycin A2. The results may help in the design of AspH inhibitors with the potential of increased selectivity compared to the previously identified Fe(II)-chelating or 2OG-competitive inhibitors. With regard to the clinical use of bleomycin A2 and of the Bcl-2 inhibitor venetoclax, the results suggest that possible side-effects mediated through the inhibition of AspH and other 2OG oxygenases should be considered.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Bleomicina/farmacologia , Inibidores Enzimáticos/farmacologia , Gossipol/farmacologia , Oxigenases de Função Mista/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Antibióticos Antineoplásicos/química , Bleomicina/química , Relação Dose-Resposta a Droga , Composição de Medicamentos , Inibidores Enzimáticos/química , Gossipol/química , Humanos , Oxigenases de Função Mista/isolamento & purificação , Oxigenases de Função Mista/metabolismo , Simulação de Acoplamento Molecular , Estrutura Molecular , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade
17.
Oncol Rep ; 44(6): 2364-2372, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33125119

RESUMO

Aspartate/asparagine ß­hydroxylase (AspH) is a type II transmembrane protein that catalyzes the post­translational hydroxylation of definite aspartyl and asparaginyl residues in epidermal growth factor­like domains of substrates. In the last few decades, accumulating evidence has indicated that AspH expression is upregulated in numerous types of human malignant cancer and is associated with poor survival and prognosis. The AspH protein aggregates on the surface of tumor cells, which contributes to inducing tumor cell migration, infiltration and metastasis. However, small­molecule inhibitors targeting hydroxylase activity can markedly block these processes, both in vitro and in vivo. Immunization of tumor­bearing mice with a phage vaccine fused with the AspH protein can substantially delay tumor growth and progression. Additionally, AspH antigen­specific CD4+ and CD8+ T cells were identified in the spleen of tumor­bearing mice. Therefore, these agents may be used as novel strategies for cancer treatment. The present review summarizes the current progress on the underlying mechanisms of AspH expression in cancer development.


Assuntos
Antígenos de Neoplasias/genética , Proteínas de Ligação ao Cálcio/genética , Regulação Neoplásica da Expressão Gênica/imunologia , Proteínas de Membrana/genética , Oxigenases de Função Mista/genética , Proteínas Musculares/genética , Neoplasias/genética , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Proteínas de Ligação ao Cálcio/imunologia , Proteínas de Ligação ao Cálcio/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Movimento Celular/imunologia , Furanos/farmacologia , Furanos/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Linfócitos do Interstício Tumoral/imunologia , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Oxigenases de Função Mista/antagonistas & inibidores , Oxigenases de Função Mista/imunologia , Oxigenases de Função Mista/metabolismo , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/imunologia , Proteínas Musculares/metabolismo , Invasividade Neoplásica/genética , Invasividade Neoplásica/imunologia , Invasividade Neoplásica/patologia , Neoplasias/imunologia , Neoplasias/mortalidade , Neoplasias/patologia , Prognóstico , Ácidos Sulfônicos/farmacologia , Ácidos Sulfônicos/uso terapêutico , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Chemphyschem ; 21(19): 2150-2154, 2020 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-32797657

RESUMO

The enzyme system mimicking Implication (IMPLY) and Inhibition (INHIB) Boolean logic gates has been designed. The same enzyme system was used to operate as the IMPLY or INHIB gate simply by reformulating the input signals. The optical analysis of the logic operation confirmed the output generation as expected for the studied logic gates. The conceptual approach to the IMPLY and INHIB logic gates allows their construction with many other enzymes operating in a similar way.


Assuntos
Inibidores Enzimáticos/farmacologia , Glucose Oxidase/antagonistas & inibidores , Hexoquinase/antagonistas & inibidores , Peroxidase do Rábano Silvestre/antagonistas & inibidores , Oxigenases de Função Mista/antagonistas & inibidores , Aspergillus niger/enzimologia , Biocatálise , Glucose Oxidase/metabolismo , Hexoquinase/metabolismo , Peroxidase do Rábano Silvestre/metabolismo , Oxigenases de Função Mista/metabolismo , Pediococcus/enzimologia , Saccharomyces cerevisiae/enzimologia
19.
J Exp Clin Cancer Res ; 39(1): 163, 2020 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-32811566

RESUMO

As metastasis is a major cause of death in cancer patients, new anti-metastatic strategies are needed to improve cancer therapy outcomes. Numerous pathways have been shown to contribute to migration and invasion of malignant tumors. Aspartate ß-hydroxylase (ASPH) is a key player in the malignant transformation of solid tumors by enhancing cell proliferation, migration, and invasion. ASPH also promotes tumor growth by stimulation of angiogenesis and immunosuppression. These effects are mainly achieved via the activation of Notch and SRC signaling pathways. ASPH expression is upregulated by growth factors and hypoxia in different human tumors and its inactivation may have broad clinical impact. Therefore, small molecule inhibitors of ASPH enzymatic activity have been developed and their anti-metastatic effect confirmed in preclinical mouse models. ASPH can also be targeted by monoclonal antibodies and has also been used as a tumor-associated antigen to induce both cluster of differentiation (CD) 8+ and CD4+ T cells in mice. The PAN-301-1 vaccine against ASPH has already been tested in a phase 1 clinical trial in patients with prostate cancer. In summary, ASPH is a promising target for anti-tumor and anti-metastatic therapy based on inactivation of catalytic activity and/or immunotherapy.


Assuntos
Antineoplásicos/uso terapêutico , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Neoplasias Pulmonares/tratamento farmacológico , Proteínas de Membrana/antagonistas & inibidores , Oxigenases de Função Mista/antagonistas & inibidores , Terapia de Alvo Molecular , Proteínas Musculares/antagonistas & inibidores , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/patologia , Prognóstico
20.
Int J Mol Sci ; 21(13)2020 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-32635392

RESUMO

Despite the existence of considerable research on somatic embryogenesis (SE), the molecular mechanism that regulates the biosynthesis of auxins during the SE induction process remains unknown. Indole-3-acetic acid (IAA) is an auxin that is synthesized in plants through five pathways. The biosynthetic pathway most frequently used in this synthesis is the conversion of tryptophan to indol-3-pyruvic acid (IPA) by tryptophan aminotransferase of Arabidopsis (TAA) followed by the conversion of IPA to IAA by enzymes encoded by YUCCA (YUC) genes of the flavin monooxygenase family; however, it is unclear whether YUC-mediated IAA biosynthesis is involved in SE induction. In this study, we report that the increase of IAA observed during SE pre-treatment (plants in MS medium supplemented with 1-naphthaleneacetic acid (NAA) 0.54 µM and kinetin (Kin) 2.32 µM for 14 days) was due to its de novo biosynthesis. By qRT-PCR, we demonstrated that YUC gene expression was consistent with the free IAA signal found in the explants during the induction of SE. In addition, the use of yucasin to inhibit the activity of YUC enzymes reduced the signal of free IAA in the leaf explants and dramatically decreased the induction of SE. The exogenous addition of IAA restored the SE process in explants treated with yucasin. Our findings suggest that the biosynthesis and localization of IAA play an essential role during the induction process of SE in Coffea canephora.


Assuntos
Coffea/embriologia , Coffea/metabolismo , Ácidos Indolacéticos/metabolismo , Reguladores de Crescimento de Plantas/biossíntese , Vias Biossintéticas/efeitos dos fármacos , Vias Biossintéticas/genética , Coffea/genética , Inibidores Enzimáticos/farmacologia , Perfilação da Expressão Gênica , Genes de Plantas , Oxigenases de Função Mista/antagonistas & inibidores , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Família Multigênica , Reguladores de Crescimento de Plantas/genética , Proteínas de Plantas/antagonistas & inibidores , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Técnicas de Embriogênese Somática de Plantas , Triazóis/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...