Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 181
Filtrar
1.
J Biochem Mol Toxicol ; 35(8): e22821, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34036678

RESUMO

The preservation of the redox homeostasis is critical for cell survival and functionality. Redox imbalance is an essential inducer of several pathological states. CD4+ /helper T cells are highly dependent on the redox state of their surrounding milieu. The potential of the aryl hydrocarbon receptor (AhR) engagement in controlling CD4+ T-cell fate during redox alteration is still challenging. C57BL/6 mice were treated with AhR agonist 6-formylindolo[3,2-b]carbazole (FICZ), AhR antagonist CH223191, an inhibitor of glutathione biosynthesis buthionine sulfoximine (BSO), and the antioxidant N-acetylcysteine (NAC) alone or in combination. Six days later, splenocytes were evaluated for the expression of the redox-related genes and the possible changes in T-cell subsets. FICZ like BSO significantly elevated the expression of HMOX1, GCLC, and GCLM genes but it failed to increase the expression of the Nrf2 gene. Moreover, FICZ + BSO increased while FICZ + CH223191 or NAC decreased the expression of these genes. FICZ also significantly increased Th1 cell numbers but decreased Tregs in a dose-dependent manner. Furthermore, a high dose of FICZ + CH223191 + NAC significantly enhanced Th1, Th17, and Treg cells but its low dose in such a situation increased Th2 and Th17 while decreased Treg cells. AhR engagement during redox alteration can determine the fate of CD4 + T cells, so, AhR agonists or antagonists might be useful in assessing immune responses. However, these results need further verifications in vitro and in animal models of various diseases.


Assuntos
Receptores de Hidrocarboneto Arílico , Linfócitos T Auxiliares-Indutores/metabolismo , Acetilcisteína/farmacologia , Animais , Compostos Azo/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glutamato-Cisteína Ligase/biossíntese , Heme Oxigenase-1/biossíntese , Proteínas de Membrana/biossíntese , Camundongos , Fator 2 Relacionado a NF-E2/biossíntese , Oxirredução/efeitos dos fármacos , Pirazóis/farmacologia , Receptores de Hidrocarboneto Arílico/agonistas , Receptores de Hidrocarboneto Arílico/metabolismo
2.
Molecules ; 25(14)2020 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-32650607

RESUMO

Activation of nuclear factor erythroid-2-related factor 2 (NRF2) has been proven to be an effective means to prevent the development of cancer, and natural curcumin stands out as a potent NRF2 activator and cancer chemopreventive agent. In this study, we have synthesized a series of 4-anilinoquinolinylchalcone derivatives, and used a NRF2 promoter-driven firefly luciferase reporter stable cell line, the HaCaT/ARE cells, to screen a panel of these compounds. Among them, (E)-3-{4-[(4-acetylphenyl)amino]quinolin-2-yl}-1-(4-fluorophenyl)prop-2-en-1-one (13b) significantly increased NRF2 activity in the HaCaT cell with a half maximal effective concentration (EC50) value of 1.95 µM. Treatment of compound 13b upregulated HaCaT cell NRF2 expression at the protein level. Moreover, the mRNA level of NRF2 target genes, heme oxygenase-1 (HO-1), glutamate-cysteine ligase catalytic subunit (GCLC), and glucose-6-phosphate dehydrogenase (G6PD) were significantly increased in HaCaT cells upon the compound 13b treatment. The molecular docking results exhibited that the small molecule 13b is well accommodated by the bound region of Kelch-like ECH-associated protein 1 (Keap1)-Kelch and NRF2 through stable hydrogen bonds and hydrophobic interaction, which contributed to the enhancement of affinity and stability between the ligand and receptor. Compound 13b has been identified as the lead compound for further structural optimization.


Assuntos
Chalconas , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína 1 Associada a ECH Semelhante a Kelch , Queratinócitos , Simulação de Acoplamento Molecular , Fator 2 Relacionado a NF-E2/biossíntese , Linhagem Celular Transformada , Chalconas/síntese química , Chalconas/química , Chalconas/farmacologia , Glucosefosfato Desidrogenase , Glutamato-Cisteína Ligase/biossíntese , Heme Oxigenase-1/biossíntese , Humanos , Proteína 1 Associada a ECH Semelhante a Kelch/química , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Queratinócitos/química , Queratinócitos/metabolismo , Fator 2 Relacionado a NF-E2/genética
3.
Int J Neurosci ; 130(1): 1-8, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31518514

RESUMO

Aim: Sevoflurane could induce apoptosis of rat hippocampal neurons, while theaflavins (TFs) have antioxidant and anti-inflammatory properties. This study aims to explore whether TFs could alleviate sevoflurane-induced neuronal cell injury.Materials and methods: Cells were treated by concentration gradient of sevoflurane and TFs. Cell viability, level of reactive oxygen species (ROS) and apoptosis rate were determined by cell counting kit-8 (CCK-8) and flow cytometry, respectively. Quantitative PCR (qPCR) and western blot were performed to determine mRNA and protein expressions.Results: TFs promoted viability of cells under the treatment of sevoflurane, while it suppressed apoptosis and down-regulated ROS level in a concentration-dependent manner. TFs could also down-regulate expression levels of caspase-3 and caspase-9 and cytosol and intranuclear nuclear factor E2-related factor 2 (Nrf2) in rat hippocampal nerve cells, while it up-regulated those of heme oxygenase 1 (HO-1), NADPH quinine oxidoreductase 1 (NQO1), glutamate cysteine ligase (GCL) and peroxiredoxin 1 (Prx1).Conclusions: Our study suggests that TFs exert protective effects on sevoflurane-induced neurocytotoxicity and therefore could be used as a potential drug for treatment of neuronal injury.


Assuntos
Biflavonoides/farmacologia , Catequina/farmacologia , Hipocampo/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/fisiologia , Neurônios/metabolismo , Síndromes Neurotóxicas/prevenção & controle , Sevoflurano/toxicidade , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Caspase 3/biossíntese , Caspase 9/biossíntese , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Glutamato-Cisteína Ligase/biossíntese , Heme Oxigenase-1/biossíntese , Hipocampo/metabolismo , NAD(P)H Desidrogenase (Quinona)/biossíntese , Fator 2 Relacionado a NF-E2/biossíntese , Neurônios/fisiologia , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/fisiopatologia , Peroxirredoxinas/biossíntese , Cultura Primária de Células , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia , Regulação para Cima/efeitos dos fármacos
4.
Artigo em Inglês | MEDLINE | ID: mdl-28760906

RESUMO

The combination of trimethoprim and sulfamethoxazole (TMP-SMX) is the most effective regimen for therapy of Pneumocystis pneumonia (PCP). As many patients with PCP are allergic or do not respond to it, efforts have been devoted to develop alternative therapies for PCP. We have found that the combination of vitamin D3 (VitD3) (300 IU/kg/day) and primaquine (PMQ) (5 mg/kg/day) was as effective as TMP-SMX for therapy of PCP. In this study, we investigated the mechanisms by which vitamin D enhances the efficacy of PMQ. C57BL/6 mice were immunosuppressed by CD4+ cell depletion, infected with Pneumocystismurina for 8 weeks, and then treated for 9 days with the combination of VitD3 and PMQ (VitD3-PMQ) or with TMP-SMX or PMQ to serve as controls. The results showed that vitamin D supplementation increased the number of CD11c+ cells, suppressed the production of proinflammatory cytokines (tumor necrosis factor alpha [TNF-α], gamma interferon [IFN-γ], and interleukin-6 [IL-6]) and inducible nitric oxide synthase (iNOS), and enhanced the expression of genes related to antioxidation (glutathione reductase and glutamate-cysteine ligase modifier subunit), antimicrobial peptides (cathelicidin), and autophagy (ATG5 and beclin-1). These results suggest that the main action of vitamin D is enhancing the ability of the host to defend against Pneumocystis infection.


Assuntos
Antibacterianos/uso terapêutico , Pneumocystis/efeitos dos fármacos , Pneumonia por Pneumocystis/tratamento farmacológico , Primaquina/uso terapêutico , Vitamina D/uso terapêutico , Animais , Peptídeos Catiônicos Antimicrobianos/biossíntese , Proteína 5 Relacionada à Autofagia/biossíntese , Proteína Beclina-1/biossíntese , Sinergismo Farmacológico , Feminino , Glutamato-Cisteína Ligase/biossíntese , Glutationa Redutase/biossíntese , Humanos , Interferon gama/biossíntese , Interleucina-6/biossíntese , Macrófagos Alveolares/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II/biossíntese , Pneumonia por Pneumocystis/microbiologia , Combinação Trimetoprima e Sulfametoxazol/uso terapêutico , Fator de Necrose Tumoral alfa/biossíntese , Catelicidinas
5.
Toxicology ; 374: 1-9, 2016 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-27865893

RESUMO

In addition to increased morbidity and mortality caused by respiratory and cardiovascular diseases, air pollution may also contribute to central nervous system (CNS) diseases. Traffic-related air pollution is a major contributor to global air pollution, and diesel exhaust (DE) is its most important component. DE contains more than 40 toxic air pollutants and is a major constituent of ambient particulate matter (PM), particularly of ultrafine-PM. Limited information suggests that exposure to DE may cause oxidative stress and neuroinflammation in the CNS. We hypothesized that males may be more susceptible than females to DE neurotoxicity, because of a lower level of expression of paraoxonase 2 (PON2), an intracellular anti-oxidant and anti-inflammatory enzyme. Acute exposure of C57BL/6 mice to DE (250-300µg/m3 for 6h) caused significant increases in lipid peroxidation and of pro-inflammatory cytokines (IL-1α, IL-1ß, IL-3, IL-6, TNF-α) in various brain regions (particularly olfactory bulb and hippocampus). In a number of cases the observed effects were more pronounced in male than in female mice. DE exposure also caused microglia activation, as measured by increased Iba1 (ionized calcium-binding adapter molecule 1) expression, and of TSPO (translocator protein) binding. Mice heterozygotes for the modifier subunit of glutamate cysteine ligase (the limiting enzyme in glutathione biosynthesis; Gclm+/- mice) appeared to be significantly more susceptible to DE-induced neuroinflammation than wild type mice. These findings indicate that acute exposure to DE causes neuroinflammation and oxidative stress in brain, and suggest that sex and genetic background may play important roles in modulating susceptibility to DE neurotoxicity.


Assuntos
Poluentes Atmosféricos/toxicidade , Química Encefálica/efeitos dos fármacos , Encéfalo/patologia , Inflamação/induzido quimicamente , Estresse Oxidativo/efeitos dos fármacos , Emissões de Veículos/toxicidade , Animais , Arildialquilfosfatase/biossíntese , Arildialquilfosfatase/genética , Citocinas/biossíntese , Feminino , Variação Genética , Glutamato-Cisteína Ligase/biossíntese , Glutamato-Cisteína Ligase/genética , Inflamação/genética , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Síndromes Neurotóxicas/patologia , Tamanho da Partícula , Material Particulado/toxicidade , Caracteres Sexuais
6.
Blood ; 127(21): 2587-97, 2016 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-26907633

RESUMO

Multiple myeloma (MM) cell lines and primary tumor cells are addicted to the MYC oncoprotein for survival. Little is known, however, about how MYC expression is upregulated in MM cells. The mucin 1 C-terminal subunit (MUC1-C) is an oncogenic transmembrane protein that is aberrantly expressed in MM cell lines and primary tumor samples. The present studies demonstrate that targeting MUC1-C with silencing by clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein 9 editing or with the GO-203 inhibitor is associated with downregulation of MYC messenger RNA and protein. The results show that MUC1-C occupies the MYC promoter and thereby activates the MYC gene by a ß-catenin/transcription factor 4 (TCF4)-mediated mechanism. In this way, MUC1-C (1) increases ß-catenin occupancy on the MYC promoter, (2) forms a complex with ß-catenin and TCF4, and, in turn, (3) drives MYC transcription. Analysis of MM cells using quantitative real-time reverse transcription polymerase chain reaction arrays further demonstrated that silencing MUC1-C is associated with downregulation of MYC target genes, including CCND2, hTERT, and GCLC Analysis of microarray data sets further demonstrated that MUC1 levels positively correlate with MYC expression in MM progression and in primary cells from over 800 MM patients. These findings collectively provide convincing evidence that MUC1-C drives MYC expression in MM.


Assuntos
Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Mucina-1/biossíntese , Mieloma Múltiplo/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Elementos de Resposta , Transcrição Gênica , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Ciclina D2/biossíntese , Ciclina D2/genética , Glutamato-Cisteína Ligase/biossíntese , Glutamato-Cisteína Ligase/genética , Humanos , Mucina-1/genética , Mieloma Múltiplo/genética , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas c-myc/genética , Telomerase/biossíntese , Telomerase/genética , beta Catenina/genética , beta Catenina/metabolismo
7.
Tumour Biol ; 37(4): 4813-29, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26520442

RESUMO

Chemoresistance to platinum-based antineoplastic agents is a consistent feature among ovarian carcinomas; however, whereas high-grade serous carcinoma (OSC) acquires resistance during chemotherapy, ovarian clear cell carcinoma (OCCC) is intrinsically resistant. The main objective of this study was to explore, in vitro and in vivo, if hepatocyte nuclear factor 1ß (HNF1ß) and glutaminolysis contribute for the resistance of OCCC to carboplatin through the intrinsically increased GSH bioavailability. To disclose the role of HNF1ß, experiments were also performed in an OSC cell line, which does not express HNF1ß. Metabolic profiles, GSH quantification, HNF1ß, and γ-glutamylcysteine ligase catalytic subunit (GCLC) and modifier subunit (GCLM) expression, cell cycle, and death were assessed in ES2 cell line (OCCC) and OVCAR3 cell line (OSC); HNF1ß knockdown was performed in ES2 and murine model of subcutaneous and peritoneal OCCC tumors was established to test buthionine sulphoxamine (BSO), as a sensitizer to carboplatin. Glutaminolysis is activated in ES2 and OVCAR3, though ES2 exclusively synthesizes amino acids and GSH. ES2 cells are more resistant to carboplatin than OVCAR3 and the abrogation of GSH production by BSO sensitizes ES2 to carboplatin. HNF1ß regulates the expression of GCLC, but not GCLM, and consequently GSH production in ES2. In vivo, BSO prior to carboplatin reduces dramatically subcutaneous tumor size and GSH levels, as well as peritoneal dissemination. Our study discloses HNF1ß as the mediator of intrinsic OCCC chemoresistance and sheds a light to re-explore a cancer adjuvant therapeutic approach using BSO to overcome the lack of efficient therapy in OCCC.


Assuntos
Adenocarcinoma de Células Claras/tratamento farmacológico , Glutamato-Cisteína Ligase/biossíntese , Glutamato-Cisteína Ligase/sangue , Fator 1-beta Nuclear de Hepatócito/biossíntese , Neoplasias Ovarianas/tratamento farmacológico , Adenocarcinoma de Células Claras/genética , Adenocarcinoma de Células Claras/patologia , Animais , Carboplatina/administração & dosagem , Linhagem Celular Tumoral , Cisplatino/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glutamato-Cisteína Ligase/genética , Glutationa/biossíntese , Fator 1-beta Nuclear de Hepatócito/genética , Humanos , Camundongos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Oxid Med Cell Longev ; 2016: 9151290, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26640618

RESUMO

Toll-like receptor 4 (TLR4) is an important sensor of Gram-negative bacteria and can trigger activation of the innate immune system. Increased activation of TLR4 can lead to the induction of oxidative stress. Herein, the pathway whereby TLR4 affects antioxidant activity was studied. In TLR4-overexpressing sheep, TLR4 expression was found to be related to the integration copy number when monocytes were challenged with lipopolysaccharide (LPS). Consequently, production of malondialdehyde (MDA) was increased, which could increase the activation of prooxidative stress enzymes. Meanwhile, activation of an antioxidative enzyme, glutathione peroxidase (GSH-Px), was increased. Real-time PCR showed that expression of activating protein-1 (AP-1) and the antioxidative-related genes was increased. By contrast, the expression levels of superoxide dismutase 1 (SOD1) and catalase (CAT) were reduced. In transgenic sheep, glutathione (GSH) levels were dramatically reduced. Furthermore, transgenic sheep were intradermally injected with LPS in each ear. The amounts of inflammatory infiltrates were correlated with the number of TLR4 copies that were integrated in the genome. Additionally, the translation of γ-glutamylcysteine synthetase (γ-GCS) was increased. Our findings indicated that overexpression of TLR4 in sheep could ameliorate oxidative injury through GSH secretion that was induced by LPS stimulation. Furthermore, TLR4 promoted γ-GCS translation through the AP-1 pathway, which was essential for GSH synthesis.


Assuntos
Glutationa/metabolismo , Lipopolissacarídeos/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo , Animais , Animais Geneticamente Modificados , Regulação da Expressão Gênica , Glutamato-Cisteína Ligase/biossíntese , Glutamato-Cisteína Ligase/genética , Glutationa/genética , Glutationa Peroxidase/biossíntese , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Ovinos , Receptor 4 Toll-Like/genética , Fator de Transcrição AP-1/biossíntese , Fator de Transcrição AP-1/genética
9.
J Pineal Res ; 60(2): 217-27, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26681403

RESUMO

Smoking is one of the most harmful lifestyles in the world. Very few studies have investigated the effects of melatonin in smoke-induced vascular injury. This study was designed to investigate whether melatonin could protect rats and humans from smoke-induced vascular injury. 32 male rats and a double-blind randomized controlled trial (RCT) containing 63 participants formed the subjects of this study. In rats, 10 mg/kg of melatonin was intraperitoneally injected. Blood samples and abdominal artery were harvested two weeks later. Melatonin decreased the expression of platelet endothelial cell adhesion molecule-1 (CD31), intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1) and endothelin-1 (ET-1) compared with the smoke exposed group (P < 0.05), whereas endothelial nitric oxide synthase (eNOS), nuclear erythroid 2-related factor 2 (Nrf2), NAD(P)H quinone oxidoreductase 1 (NQO-1), catalytic glutamate cysteine ligase (GCLC) and heme oxygenase-1 (HO-1) recovered markedly (P < 0.05). In humans, 3 mg/day of melatonin was taken orally by the participants. Blood samples were drawn at baseline and after two weeks of treatment. Compared with the oral placebo group, melatonin decreased the concentration of fibrinogen (Fbg) (P = 0.04) and free fatty acids (FFA) (P = 0.04) in smokers, along with the decreased expression of ICAM-1, VCAM-1 and ET-1 (P = 0.004, P = 0.001, P < 0.0001, respectively). In contrast, Nrf2 and HO-1 expression were markedly increased (P = 0.0001, P = 0.0049, respectively) after smokers took melatonin orally. In summary, our present data suggest that melatonin could ameliorate smoke-induced vascular injury.


Assuntos
Melatonina , Fumar , Doenças Vasculares , Adulto , Animais , Feminino , Humanos , Masculino , Ratos , Administração Oral , Moléculas de Adesão Celular/biossíntese , Endotelina-1/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Glutamato-Cisteína Ligase/biossíntese , Heme Oxigenase-1/biossíntese , Melatonina/administração & dosagem , NAD(P)H Desidrogenase (Quinona)/biossíntese , Fator 2 Relacionado a NF-E2/biossíntese , Óxido Nítrico Sintase Tipo III/biossíntese , Ratos Sprague-Dawley , Fumar/efeitos adversos , Fumar/tratamento farmacológico , Fumar/metabolismo , Doenças Vasculares/tratamento farmacológico , Doenças Vasculares/etiologia , Doenças Vasculares/metabolismo
10.
Eur J Pharmacol ; 771: 99-106, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26687633

RESUMO

Glutathione is a key regulator of oxidative balance in all mammals, especially in the central nervous system. The first step of glutathione synthesis is catalyzed by glutamate-cysteine ligase (GCL), which is composed of catalytic and modifier subunits (GCLC and GCLM, respectively). In non-neural cells and tissues, insulin and insulin-like growth factor 1 (IGF-1) have been found to stimulate transcription of GCLC gene. Here we found that treatment of human neuroblastoma SH-SY5Y cells with insulin or IGF-1 increased mRNA level of GCLM, but not of GCLC, in a concentration- and time-dependent manner. In contrast, insulin did not increase GCL expression in rat C6 glioma cells. We also confirmed that IGF-1 increased protein level of GCLM and cellular glutathione content in SH-SY5Y cells. In addition, IGF-1 increased nuclear factor erythroid 2-related factor 2 (Nrf2) protein in the nuclear fraction of SH-SY5Y cells. siRNA-mediated knockdown of Nrf2 protein expression abrogated IGF-1-induced up-regulation of GCLM mRNA expression. Finally, IGF-1-induced increase in nuclear Nrf2 protein and GCLM mRNA expression was abolished by LY294002, a phosphoinositide 3-kinase inhibitor. These results indicate that insulin and IGF-1 have the ability to enhance glutathione biosynthesis in neuronal cells via specific up-regulation of GCLM expression.


Assuntos
Glutamato-Cisteína Ligase/biossíntese , Glutationa/biossíntese , Fator de Crescimento Insulin-Like I/farmacologia , Proteínas Quinases Dependentes de 3-Fosfoinositídeo/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Cromonas/farmacologia , Expressão Gênica/efeitos dos fármacos , Glutamato-Cisteína Ligase/efeitos dos fármacos , Glutamato-Cisteína Ligase/genética , Humanos , Morfolinas/farmacologia , Fator 2 Relacionado a NF-E2/biossíntese , Fator 2 Relacionado a NF-E2/genética , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , RNA Interferente Pequeno/farmacologia , Ratos , Regulação para Cima/efeitos dos fármacos
11.
PLoS One ; 9(9): e106711, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25184212

RESUMO

Exposure to nanoparticles (NPs) may cause vascular effects including endothelial dysfunction and foam cell formation, with oxidative stress and inflammation as supposed central mechanisms. We investigated oxidative stress, endothelial dysfunction and lipid accumulation caused by nano-sized carbon black (CB) exposure in cultured human umbilical vein endothelial cells (HUVECs), THP-1 (monocytes) and THP-1 derived macrophages (THP-1a). The proliferation of HUVECs or co-cultures of HUVECs and THP-1 cells were unaffected by CB exposure, whereas there was increased cytotoxicity, assessed by the LDH and WST-1 assays, especially in THP-1 and THP-1a cells. The CB exposure decreased the glutathione (GSH) content in THP-1 and THP-1a cells, whereas GSH was increased in HUVECs. The reactive oxygen species (ROS) production was increased in all cell types after CB exposure. A reduction of the intracellular GSH concentration by buthionine sulfoximine (BSO) pre-treatment further increased the CB-induced ROS production in THP-1 cells and HUVECs. The expression of adhesion molecules ICAM-1 and VCAM-1, but not adhesion of THP-1 to HUVECs or culture dishes, was elevated by CB exposure, whereas these effects were unaffected by BSO pre-treatment. qRT-PCR showed increased VCAM1 expression, but no change in GCLM and HMOX1 expression in CB-exposed HUVECs. Pre-exposure to CB induced lipid accumulation in THP-1a cells, which was not affected by the presence of the antioxidant N-acetylcysteine. In addition, the concentrations of CB to induce lipid accumulation were lower than the concentrations to promote intracellular ROS production in THP-1a cells. In conclusion, exposure to nano-sized CB induced endothelial dysfunction and foam cell formation, which was not dependent on intracellular ROS production.


Assuntos
Células Endoteliais da Veia Umbilical Humana/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Macrófagos/metabolismo , Nanopartículas , Espécies Reativas de Oxigênio/metabolismo , Fuligem/farmacologia , Linhagem Celular Tumoral , Regulação da Expressão Gênica/efeitos dos fármacos , Glutamato-Cisteína Ligase/biossíntese , Heme Oxigenase-1/biossíntese , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Macrófagos/citologia , Estresse Oxidativo/efeitos dos fármacos , Molécula 1 de Adesão de Célula Vascular/biossíntese
12.
Free Radic Biol Med ; 75: 129-39, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25016074

RESUMO

Diabetes increases oxidative stress in the retina and decreases the levels of the intracellular antioxidant glutathione (GSH). The transcriptional factor Nrf2 regulates the expression of Gclc, the enzyme important in the biosynthesis of GSH, and in diabetes the binding of Nrf2 at the antioxidant response element region 4 (ARE4) is decreased. Our aim was to investigate the role of epigenetic modifications in the decreased Nrf2 binding at Gclc-ARE4 in the development of diabetic retinopathy and in the metabolic memory associated with its continued progression. The effect of hyperglycemia on H3K4 methylation in Nrf2 binding at Gclc-ARE4 was investigated by chromatin immunoprecipitation in the rat retina and was confirmed in retinal endothelial cells in which histone demethylase (LSD1) was manipulated. The role of histone methylation at Gclc-ARE4 in the metabolic memory was examined in rats maintained under poor control for 3 months followed by good control (GC) for 3 months. Although H3K4me2 at Gclc-ARE4 was increased in diabetes, H3K4me3 and H3K4me1 were decreased. LSD1 siRNA abrogated the glucose-induced decrease in H3K4me1 at Gclc-ARE4 and ameliorated decreases in Nrf2 binding at Gclc-ARE4 and Gclc transcripts. Reestablishment of GC failed to provide any benefits to histone methylation, and Nrf2 binding activity remained compromised. Thus, in diabetic retinopathy, histone methylation at Gclc-ARE4 plays an important role in regulating the Nrf2-Gclc-GSH cascade. Targeting histone methylation could help inhibit/slow down this blinding disease.


Assuntos
Retinopatia Diabética/patologia , Glutamato-Cisteína Ligase/metabolismo , Histona Desmetilases/genética , Histonas/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Idoso , Animais , Elementos de Resposta Antioxidante , Bovinos , Linhagem Celular , Metilação de DNA , Diabetes Mellitus Experimental/patologia , Epigênese Genética , Glucose/metabolismo , Glutamato-Cisteína Ligase/biossíntese , Glutationa/biossíntese , Glutationa/metabolismo , Humanos , Hiperglicemia/metabolismo , Masculino , Pessoa de Meia-Idade , Estresse Oxidativo , Ligação Proteica/genética , Interferência de RNA , RNA Interferente Pequeno , Ratos , Ratos Wistar , Retina/citologia , Retina/patologia
13.
Oncogene ; 33(5): 556-66, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-23353822

RESUMO

Retinoblastoma (Rb) protein is a tumor suppressor that is dysregulated in a majority of human cancers. Rb functions to inhibit cell cycle progression in part by directly disabling the E2F family of cell cycle-promoting transcription factors. Because the de novo synthesis of multiple glutamine-derived anabolic precursors is required for cell cycle progression, we hypothesized that Rb also may directly regulate proteins involved in glutamine metabolism. We examined glutamine metabolism in mouse embryonic fibroblasts (MEFs) isolated from mice that have triple knock-outs (TKO) of all three Rb family members (Rb-1, Rbl1 and Rbl2) and found that loss of global Rb function caused a marked increase in (13)C-glutamine uptake and incorporation into glutamate and tricarboxylic acid cycle (TCA) intermediates in part via upregulated expression of the glutamine transporter ASCT2 and the activity of glutaminase 1 (GLS1). The Rb-controlled transcription factor E2F-3 altered glutamine uptake by direct regulation of ASCT2 mRNA and protein expression, and E2F-3 was observed to associate with the ASCT2 promoter. We next examined the functional consequences of the observed increase in glutamine uptake and utilization and found that glutamine exposure potently increased oxygen consumption, whereas glutamine deprivation selectively decreased ATP concentration in the Rb TKO MEFs but not the wild-type (WT) MEFs. In addition, TKO MEFs exhibited elevated production of glutathione from exogenous glutamine and had increased expression of gamma-glutamylcysteine ligase relative to WT MEFs. Importantly, this metabolic shift towards glutamine utilization was required for the proliferation of Rb TKO MEFs but not for the proliferation of the WT MEFs. Last, addition of the TCA cycle intermediate α-ketoglutarate to the Rb TKO MEFs reversed the inhibitory effects of glutamine deprivation on ATP, GSH levels and viability. Taken together, these studies demonstrate that the Rb/E2F cascade directly regulates a major energetic and anabolic pathway that is required for neoplastic growth.


Assuntos
Fator de Transcrição E2F3/metabolismo , Glutamina/metabolismo , Proteína do Retinoblastoma/metabolismo , Proteína p107 Retinoblastoma-Like/metabolismo , Proteína p130 Retinoblastoma-Like/metabolismo , Trifosfato de Adenosina/biossíntese , Sistema ASC de Transporte de Aminoácidos/biossíntese , Sistema ASC de Transporte de Aminoácidos/genética , Sistema ASC de Transporte de Aminoácidos/metabolismo , Animais , Transporte Biológico/genética , Ciclo Celular , Proliferação de Células , Células Cultivadas , Fator de Transcrição E2F3/biossíntese , Fibroblastos , Glutamato-Cisteína Ligase/biossíntese , Glutaminase/biossíntese , Glutaminase/genética , Glutaminase/metabolismo , Glutationa/biossíntese , Ácidos Cetoglutáricos/metabolismo , Camundongos , Camundongos Knockout , Antígenos de Histocompatibilidade Menor , Oxigênio/metabolismo , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno , Espécies Reativas de Oxigênio/metabolismo , Proteína do Retinoblastoma/genética , Proteína p107 Retinoblastoma-Like/genética , Proteína p130 Retinoblastoma-Like/genética
14.
Free Radic Biol Med ; 68: 148-58, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24296246

RESUMO

Endotoxemia correlates with the degree of liver failure and may participate in worsening of liver diseases. Lipopolysaccharide (LPS; synonymous with endotoxin) treatment in mice lowered the hepatic glutathione (GSH) level, which in turn is a variable that determines susceptibility to LPS-induced injury. We previously showed that LPS treatment in mice lowered hepatic expression of the rate-limiting enzyme in GSH synthesis, glutamate-cysteine ligase (GCL). The aim of our current work was to determine the molecular mechanism(s) responsible for these changes. Studies were done using RAW cells (murine macrophages), in vivo LPS-treated mice, and mouse hepatocytes. We found that LPS treatment lowered GCL catalytic and modifier (Gclc and Gclm) subunit expression at the transcriptional level, which was unrelated to alterations in nitric oxide production or induction of NF-κB/p65 subunit. The key mechanism was a decrease in sumoylation of nuclear factor-erythroid 2-related factor 2 (Nrf2) and MafG, which is required for their heterodimerization and subsequent binding and trans-activation of the antioxidant-response element (ARE) present in the promoter region of these genes that is essential for their expression. LPS treatment lowered markedly the expression of ubiquitin-conjugating enzyme 9 (Ubc9), which is required for sumoylation. Similar findings also occurred in liver after in vivo LPS treatment and in LPS-treated mouse hepatocytes. Overexpression of Ubc9 protected against LPS-mediated inhibition of Gclc and Gclm expression in RAW cells and hepatocytes. In conclusion, LPS-mediated lowering of GCL expression in hepatocytes and macrophages is due to lowering of sumoylation of Nrf2 and MafG, leading to reduced heterodimerization, binding, and trans-activation of ARE.


Assuntos
Glutamato-Cisteína Ligase/biossíntese , Glutationa/biossíntese , Falência Hepática/metabolismo , NF-kappa B/metabolismo , Animais , Regulação Enzimológica da Expressão Gênica , Glutamato-Cisteína Ligase/genética , Glutationa/antagonistas & inibidores , Glutationa/genética , Lipopolissacarídeos/toxicidade , Falência Hepática/induzido quimicamente , Falência Hepática/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Fator de Transcrição MafG/metabolismo , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , Óxido Nítrico/metabolismo , Regiões Promotoras Genéticas , Proteínas Repressoras/metabolismo , Sumoilação
15.
Free Radic Biol Med ; 65: 1506-1515, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23277146

RESUMO

Willow bark extract (WBE) is listed in the European Pharmacopoeia and has been traditionally used for treating fever, pain, and inflammation. Recent studies have demonstrated its clinical usefulness. This study investigated the antioxidative effects of WBE in human umbilical vein endothelial cells (HUVECs) and Caenorhabditis elegans. WBE prevented oxidative-stress-induced cytotoxicity of HUVECs and death of C. elegans. WBE dose-dependently increased mRNA and protein expression levels of the nuclear factor erythroid 2-related factor 2 (Nrf2) target genes heme oxygenase-1, γ-glutamylcysteine ligase modifier and catalytic subunits, and p62 and intracellular glutathione (GSH) in HUVECs. In the nematode C. elegans, WBE increased the expression of the gcs-1::green fluorescent protein reporter, a well-characterized target of the Nrf2 ortholog SKN-1, in a manner that was SKN-1-dependent. WBE increased intranuclear expression and DNA binding of Nrf2 and the activity of an antioxidant response element (ARE) reporter plasmid in HUVECs. WBE-induced expression of Nrf2-regulated genes and increased GSH levels in HUVECs were reduced by Nrf2 and p38 small interfering (si) RNAs and by the p38-specific inhibitor SB203580. Nrf2 siRNA reduced the cytoprotective effect of WBE against oxidative stress in HUVECs. Salicin, a major anti-inflammatory ingredient of WBE, failed to activate ARE-luciferase activity, whereas a salicin-free WBE fraction showed intensive activity. WBE induced antioxidant enzymes and prevented oxidative stress through activation of Nrf2 independent of salicin, providing a new potential explanation for the clinical usefulness of WBE.


Assuntos
Caenorhabditis elegans/enzimologia , Células Endoteliais da Veia Umbilical Humana/enzimologia , Fator 2 Relacionado a NF-E2/metabolismo , Extratos Vegetais/farmacologia , Salix/química , Animais , Elementos de Resposta Antioxidante/genética , Antioxidantes , Álcoois Benzílicos/farmacologia , Proteínas de Caenorhabditis elegans/biossíntese , Proteínas de Caenorhabditis elegans/genética , Hidrolases de Éster Carboxílico/metabolismo , Células Cultivadas , Inibidores de Ciclo-Oxigenase/farmacologia , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Células Endoteliais/enzimologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Glucosídeos/farmacologia , Glutamato-Cisteína Ligase/biossíntese , Glutamato-Cisteína Ligase/genética , Glutationa/biossíntese , Glutationa/genética , Heme Oxigenase-1/biossíntese , Heme Oxigenase-1/genética , Imidazóis/farmacologia , Fator 2 Relacionado a NF-E2/genética , Estresse Oxidativo/efeitos dos fármacos , Casca de Planta/química , Proteínas Proto-Oncogênicas c-myc/biossíntese , Proteínas Proto-Oncogênicas c-myc/genética , Piridinas/farmacologia , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Interferente Pequeno , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/genética
16.
Free Radic Res ; 47(2): 89-94, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23136969

RESUMO

Glutathione (GSH) is an important cellular antioxidant and has a critical role in maintaining the balance of cellular redox. In this study, we investigated the GSH biosynthesis genes involved in the elevation of endogenous GSH levels using an irradiation system with an irradiation dose rate of 1.78 mGy/h, which was about 40,000 times less than the dose rates used in other studies. The results showed that GSH levels were significantly increased in the low-dose (0.02 and 0.2 Gy) irradiated group compared to those in the non-irradiated group, but enzymatic antioxidants such as superoxide dismutase and catalase were not induced at any doses tested. The elevation in GSH was accompanied by elevated expression of glutamate-cysteine ligase modifier subunit, but no changes were observed in the expression of glutamate-cysteine ligase catalytic subunit and thioredoxin in de novo GSH synthesis. In the case of genes involved in the GSH regeneration cycle, the expression of glutathione reductase was not changed after irradiation, whereas glutathione peroxidase was only increased in the 0.2 Gy irradiated group. Collectively, our results suggest that the de novo pathway, rather than the regeneration cycle, may be mainly switched on in response to stimulation with long-term low-dose radiation in the spleen.


Assuntos
Glutamato-Cisteína Ligase/biossíntese , Glutationa/biossíntese , Glutationa/efeitos da radiação , Baço/efeitos da radiação , Animais , Catalase/efeitos da radiação , Raios gama , Glutamato-Cisteína Ligase/genética , Glutamato-Cisteína Ligase/efeitos da radiação , Glutationa/metabolismo , Glutationa Peroxidase/efeitos da radiação , Glutationa Redutase/metabolismo , Glutationa Redutase/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/biossíntese , Baço/metabolismo , Superóxido Dismutase/efeitos da radiação , Tiorredoxinas
17.
Toxicol Appl Pharmacol ; 266(3): 439-42, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23201461

RESUMO

Mustard gas, used in chemical warfare since 1917, is a mutagenic and carcinogenic agent that produces severe dermal lesions for which there are no effective therapeutics; it is currently seen as a potential terrorist threat to civilian populations. Sulforaphane, found in cruciferous vegetables, is known to induce enzymes that detoxify compounds such as the sulfur mustards that react through electrophilic intermediates. Here, we observe that a single topical treatment with sulforaphane induces mouse epidermal levels of the regulatory subunit of glutamate-cysteine ligase, the rate-limiting enzyme in glutathione biosynthesis, and also increases epidermal levels of reduced glutathione. Furthermore, a glutathione S-transferase, GSTA4, is also induced in mouse skin by sulforaphane. In an in vivo model in which mice are given a single mutagenic application of the sulfur mustard analog 2-(chloroethyl) ethyl sulfide (CEES), we now show that therapeutic treatment with sulforaphane abolishes the CEES-induced increase in mutation frequency in the skin, measured four days after exposure. Sulforaphane, a natural product currently in clinical trials, shows promise as an effective therapeutic against mustard gas.


Assuntos
Substâncias para a Guerra Química/toxicidade , Glutamato-Cisteína Ligase/biossíntese , Gás de Mostarda/análogos & derivados , Gás de Mostarda/toxicidade , Pele/efeitos dos fármacos , Tiocianatos/farmacologia , Animais , Indução Enzimática/efeitos dos fármacos , Feminino , Glutationa/biossíntese , Glutationa Transferase/biossíntese , Immunoblotting , Isotiocianatos , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Pele/enzimologia , Pele/metabolismo , Sulfóxidos
18.
J Biol Chem ; 287(43): 36341-55, 2012 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-22942279

RESUMO

GSH is synthesized sequentially by glutamate-cysteine ligase (GCL) and GSH synthase and defends against oxidative stress, which promotes hepatic stellate cell (HSC) activation. Changes in GSH synthesis during HSC activation are poorly characterized. Here, we examined the expression of GSH synthetic enzymes in rat HSC activation and reversion to quiescence. Expression of the GCL catalytic subunit (GCLC) fell during HSC activation and increased when activated HSCs revert back to quiescence. Blocking the increase in GCLC expression kept HSCs in an activated state. Activated HSCs have higher nuclear levels and binding activity of MafG to the antioxidant response element (ARE) of GCLC but lower Nrf2/MafG heterodimer binding to the ARE. Quiescent HSCs have a lower nuclear MafG level but higher Nrf2/MafG heterodimer binding to ARE. This occurred because of enhanced sumoylation of Nrf2 and MafG by SUMO-1, which promoted Nrf2 binding to ARE and heterodimerization with MafG. In vivo, knockdown of GCLC exacerbated bile duct ligation-induced liver injury and fibrosis. Ursodeoxycholic acid and S-adenosylmethionine are anti-fibrotic in bile duct ligation, but this effect was nearly lost if GCLC induction was blocked. In conclusion, sumoylation of Nrf2 and MafG enhances heterodimerization and increases GCLC expression, which keeps HSCs in a quiescent state. Antifibrotic agents require activation of GCLC to fully exert their protective effect.


Assuntos
Regulação Enzimológica da Expressão Gênica , Glutamato-Cisteína Ligase/biossíntese , Células Estreladas do Fígado/enzimologia , Cirrose Hepática/enzimologia , Animais , Sequência de Bases , Células Cultivadas , Ativação Enzimática , Técnicas de Silenciamento de Genes , Glutamato-Cisteína Ligase/genética , Células Estreladas do Fígado/patologia , Cirrose Hepática/genética , Cirrose Hepática/patologia , Fator de Transcrição MafG/genética , Fator de Transcrição MafG/metabolismo , Dados de Sequência Molecular , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Elementos de Resposta/genética , S-Adenosilmetionina/genética , S-Adenosilmetionina/metabolismo , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Sumoilação/genética , Ácido Ursodesoxicólico/genética , Ácido Ursodesoxicólico/metabolismo
19.
Free Radic Res ; 46(10): 1220-9, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22762311

RESUMO

Diquat is an herbicide that generates superoxide anions through redox cycling. Nuclear factor erythroid-derived 2- like 2 (Nrf2) is a transcription factor that up-regulates cytoprotective genes in response to oxidative stress. To investigate the protective effect of Nrf2 against diquat-induced toxicity, wild-type, Nrf2-null and Kelch-like ECH-associated protein 1-knockdown (Keap1-KD) mice with enhanced Nrf2 activity were treated with diquat dibromide (125 mg/kg, i.p.). Blood and tissues were collected at 1, 2, 4 and 6 hours after treatment. Administration of diquat resulted in lipid peroxidation and lethality in wild-type mice, which were more in Nrf2-null mice and less in Keap1-KD mice. Diquat produced liver injury in Nrf2-null mice, as evidenced by increased serum ALT activity and extensive hepatic necrosis, but not in wild-type and Keap1-KD mice. Diquat produced more severe lung injury in Nrf2-null than in wild-type mice, as evidenced by increased lung weight and alveolar collapse. In contrast, Keap1-KD mice had attenuated lung edema and no histopathological alterations. To further investigate the mechanism of the protective effects of Nrf2, lung and liver glutathione (GSH) concentrations were quantified. Diquat decreased GSH in lung and liver in wild-type mice, and the decrease was more in Nrf2-null mice, and less in Keap1-KD mice. After diquat treatment, the mRNA of the GSH synthesis enzyme Gclc was increased in Keap1-KD, but not in Nrf2-null mice. Collectively, Nrf2 plays an important role in preventing diquat-induced liver and lung injury, and this protective effect results from Nrf2-regulated elevation of cellular GSH and expression of GSH synthetic genes.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Diquat/toxicidade , Lesão Pulmonar/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Alanina Transaminase/sangue , Animais , Antioxidantes/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Glutamato-Cisteína Ligase/biossíntese , Glutationa/metabolismo , Dissulfeto de Glutationa/metabolismo , Herbicidas/toxicidade , Endogamia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator 2 Relacionado a NF-E2/deficiência , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Análise de Sobrevida , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
20.
J Neuroendocrinol ; 24(10): 1346-55, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22672343

RESUMO

The Zucker diabetic fatty (ZDF) rat is an obesity and type 2 diabetes model. Progression to diabetes is well characterised in ZDF rats, but only in the fasted state. We evaluated the mechanisms underlying postprandial insulin resistance in young ZDF rats. We tested the hypothesis that the overall postprandial action of insulin is affected in ZDF rats as a result of impairment of the hepatic parasympathetic-nitric oxide (PSN-NO) axis and/or glutathione (GSH), resulting in decreased indirect (PSN-NO axis) and direct actions of insulin. Nine-week-old male ZDF rats and lean Zucker rats (LZR, controls) were used. The action of insulin was assessed in the fed state before and after parasympathetic antagonism atropine. Basal hepatic NO and GSH were measured, as well as NO synthase (NOS) and γ-glutamyl-cysteine synthethase (GCS) activity and expression. ZDF rats presented postprandial hyperglycaemia (ZDF, 201.4 ± 12.9 mg/dl; LZR, 107.7 ± 4.3 mg/dl), but not insulinopaenia (ZDF, 5.9 ± 0.8 ng/ml; LZR, 1.5 ± 0.3 ng/ml). Total postprandial insulin resistance was observed (ZDF, 78.6 ± 7.5 mg glucose/kg; LZR, 289.2 ± 24.7 mg glucose/kg), with a decrease in both the direct action of insulin (ZDF, 54.8 ± 7.0 mg glucose/kg; LZR, 173.3 ± 20.5 mg glucose/kg) and the PSN-NO axis (ZDF, 24.5 ± 3.9 mg glucose/kg; LZR, 115.9 ± 19.4 mg glucose/kg). Hepatic NO (ZDF, 117.2 ± 11.4 µmol/g tissue; LZR, 164.6 ± 4.9 µmol/g tissue) and GSH (ZDF, 4.9 ± 0.3 µmol/g; LZR, 5.9 ± 0.2 µmol/g) were also compromised as a result of decreased NOS and GCS activity, respectively. These results suggest a compromise of the mechanism responsible for potentiating insulin action after a meal in ZDF rats. We show that defective PSN-NO axis and GSH synthesis, together with an impaired direct action of insulin, appears to contribute to postprandial insulin resistance in this model.


Assuntos
Diabetes Mellitus/metabolismo , Resistência à Insulina/fisiologia , Óxido Nítrico/deficiência , Sistema Nervoso Parassimpático/fisiologia , Período Pós-Prandial/fisiologia , Animais , Glicemia/metabolismo , Glutamato-Cisteína Ligase/biossíntese , Glutationa/metabolismo , Insulina/sangue , Fígado/enzimologia , Fígado/metabolismo , Masculino , Camundongos , Óxido Nítrico Sintase/biossíntese , Ratos Zucker
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...