Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
2.
Free Radic Biol Med ; 178: 42-53, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34848368

RESUMEN

Insulin resistance (IR) promotes atherosclerosis and increases the risk of diabetes and cardiovascular diseases. Our previous studies have demonstrated that high uric acid (HUA) increased oxidative stress, leading to IR in cardiomyocytes and pancreatic ß cells. However, whether HUA can induce IR in monocytes/macrophages, which play critical roles in all stages of atherosclerosis, is unclear. Recent findings revealed that thioredoxin-interacting protein (TXNIP) negatively regulates insulin signaling; however, the roles and mechanisms of TXNIP in HUA-induced IR remain unclear. Therefore, in this study, we investigated the function of TXNIP in macrophages treated with UA. Transcriptomic profiling revealed TXNIP as one of the most upregulated genes, and subsequent RT-PCR and Western blot analyses confirmed that TXNIP was upregulated by HUA. HUA treatment significantly increased mitochondrial reactive oxygen species (MtROS) levels and decreased insulin-stimulated glucose uptake. Silencing TXNIP by RNA interference significantly diminished HUA-induced oxidative stress and IR. Mechanistically, silencing TXNIP reversed the inhibition of the phosphorylation of insulin receptor substrate 2 (IRS2)/protein kinase B (AKT) pathway induced by HUA. Additional study revealed that HUA induced the activation of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) signaling pathway, but silencing TXNIP abolished it. Moreover, Nrf2 inhibitor (ML385) ameliorated HUA-induced IR independent of IRS2/AKT signaling. Probenecid, a well-known UA-lowering drug, significantly suppressed the activation of TXNIP and Nrf2/HO-1 signaling. Furthermore, RNA-seq revealed that activation of the TXNIP-related redox pathway may be a key regulator in patients with asymptomatic hyperuricemia. These data suggest that silencing TXNIP could ameliorate HUA-induced IR via the IRS2/AKT and Nrf2/HO-1 pathways in macrophages. Additionally, TXNIP might be a promising therapeutic target for preventing and treating oxidative stress and IR induced by HUA.


Asunto(s)
Resistencia a la Insulina , Ácido Úrico , Proteínas Portadoras/genética , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Humanos , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Macrófagos/metabolismo , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo
3.
Am J Transl Res ; 13(2): 659-671, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33594316

RESUMEN

Berberine (BBR) confers potential cardioprotective effects. However, the relevant mechanisms underlying its regulation of cardiomyocyte survival following hypoxia/reoxygenation (H/R) treatment remain unknown. The present study investigated whether BBR could protect H/R by suppressing apoptosis and explored how TGF-ß/Smad4 signaling pathway influenced H/R in vitro. Two cardiomyocyte cell lines-AC16 and H9c2- were treated with H/R and BBR. The survival and apoptosis of these two cell lines were assessed using the MTT and BrdU assays and western blotting (WB) and flow cytometry. Mitochondrial reactive oxygen species (ROS) and caspase (Cas)-3, Cas-8, and Cas-9 activation were evaluated using enzyme-linked immunosorbent assay as well as WB. Compared to the control group, H/R resulted in notable cell apoptosis, whereas BBR treatment evidently counteracted the process. BBR also markedly suppressed H/R-triggered excessive mitochondrial ROS generation and inhibited Smad4 expression. Overexpressing Smad4 in BBR-treated H/R-exposed cardiomyocytes reversed the effect of BBR treatment on apoptosis. Therefore, BBR protects H/R-treated cardiomyocytes from apoptosis by inhibiting the TGF-ß/Smad4 signaling pathway.

4.
Mol Cell Endocrinol ; 443: 138-145, 2017 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-28042024

RESUMEN

Hyperuricemia occurs together with abnormal glucose metabolism and insulin resistance. Skeletal muscle is an important organ of glucose uptake, disposal, and storage. Metformin activates adenosine monophosphate-activated protein kinase (AMPK) to regulate insulin signaling and promote the translocation of glucose transporter type 4 (GLUT4), thereby stimulating glucose uptake to maintain energy balance. Our previous study showed that high uric acid (HUA) induced insulin resistance in skeletal muscle tissue. However, the mechanism of metformin ameliorating UA-induced insulin resistance in muscle cells is unknown and we aimed to determine it. In this study, differentiated C2C12 cells were exposed to UA (15 mg/dl), then reactive oxygen species (ROS) was detected with DCFH-DA and glucose uptake with 2-NBDG. The levels of phospho-insulin receptor substrate 1 (IRS1; Ser307), phospho-AKT (Ser473) and membrane GLUT4 were examined by western blot analysis. The impact of metformin on UA-induced insulin resistance was monitored by adding Compound C, an AMPK inhibitor, and LY294002, a PI3K/AKT inhibitor. Our data indicate that UA can increase ROS production, inhibit IRS1-AKT signaling and insulin-stimulated glucose uptake, and induce insulin resistance in C2C12 cells. Metformin can reverse this process by increasing intracellular glucose uptake and ameliorating UA-induced insulin resistance.


Asunto(s)
Resistencia a la Insulina , Metformina/farmacología , Células Musculares/metabolismo , Músculo Esquelético/citología , Ácido Úrico/toxicidad , 4-Cloro-7-nitrobenzofurazano/análogos & derivados , 4-Cloro-7-nitrobenzofurazano/metabolismo , Acetilcisteína/farmacología , Adenilato Quinasa/metabolismo , Animales , Antioxidantes/farmacología , Línea Celular , Desoxiglucosa/análogos & derivados , Desoxiglucosa/metabolismo , Insulina/farmacología , Proteínas Sustrato del Receptor de Insulina/metabolismo , Ratones , Modelos Biológicos , Células Musculares/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
5.
Cell Physiol Biochem ; 40(3-4): 538-548, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27889764

RESUMEN

BACKGROUND/AIMS: Hyperuricemia is part of the metabolic-syndrome cluster of abdominal obesity, impaired glucose tolerance, insulin resistance, dyslipidemia, and hypertension. Monocytes/macrophages are critical in the development of metabolic syndrome, including gout, obesity and atherosclerosis. However, how high uric acid (HUA) exposure affects monocyte/macrophage function remains unclear. In this study, we investigated the molecular mechanism of HUA exposure in monocytes/macrophages and its impact on oxidized low-density lipoprotein (oxLDL)-induced foam-cell formation in a human monocytic cell line, THP-1. METHODS: We primed THP-1 cells with phorbol-12-myristate-13-acetate (PMA) for differentiation, then exposed cells to HUA and detected the production of reactive oxygen species (ROS) and analyzed the level of phospho-AMPKα. THP-1 cells were pre-incubated with Compound C, an AMPK inhibitor, or N-acetyl-L-cysteine (NAC), a ROS scavenger, or HUA before PMA, to assess CD68 expression and phospho-AMPKα level. PMA-primed THP-1 cells were pre-treated with oxLDL before Compound C and HUA treatment. Western blot analysis was used to examine the levels of phospho-AMPKα, CD68, ABCG1, ABCA1, cyclooxygenase-2 (COX-2) and NF-κB (p65). Flow cytometry was used to assess ROS production and CD68 expression in live cells. Oil-red O staining was used to observe oxLDL uptake in cells. RESULTS: HUA treatment increased ROS production in PMA-primed THP-1 cells; NAC blocked HUA-induced oxidative stress. HUA treatment time-dependently increased phospho-AMPKα level in PMA-primed THP-1 cells. The HUA-induced oxidative stress increased phospho-AMPKα levels, which was blocked by NAC. HUA treatment impaired CD68 expression during cell differentiation by activating the AMPK pathway, which was reversed by Compound C treatment. Finally, HUA treatment inhibited oxLDL uptake in the formation of foam cells in THP-1 cells, which was blocked by Compound C treatment. HUA treatment significantly increased the expression of ABCG1 and reversed the oxLDL-reduced ABCG1 expression but did not affect the expression of ABCA1, NF-κB (p65) or COX-2. CONCLUSIONS: HUA exposure activated the ROS-AMPK pathway, impaired CD68 expression, and inhibited oxLDL-induced foam-cell formation in a human monocytic cell line, THP-1.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Células Espumosas/citología , Lipoproteínas LDL/farmacología , Monocitos/citología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Ácido Úrico/farmacología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1/metabolismo , Acetilcisteína/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Ciclooxigenasa 2/metabolismo , Células Espumosas/efectos de los fármacos , Células Espumosas/metabolismo , Humanos , Modelos Biológicos , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología , Factores de Tiempo , Factor de Transcripción ReIA/metabolismo
6.
Cancer Sci ; 107(12): 1806-1817, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27636742

RESUMEN

Metformin is an oral biguanide commonly used for treating type II diabetes and has recently been reported to possess antiproliferative properties that can be exploited for the prevention and treatment of a variety of cancers. The mechanisms underlying this effect have not been fully elucidated. Our study shows a marked loss of AMP-activated protein kinase (AMPK) phosphorylation and nuclear human Forkhead box O1 (FOXO1) protein in estrogen-dependent endometrial cancer (EC) tumors compared to normal control endometrium. Metformin treatment suppressed EC cell growth in a time-dependent manner in vitro; this effect was cancelled by cotreatment with an AMPK inhibitor, compound C. Metformin decreased FOXO1 phosphorylation and increased FOXO1 nuclear localization in Ishikawa and HEC-1B cells, with non-significant increase in FOXO1 mRNA expression. Moreover, compound C blocked the metformin-induced changes of FOXO1 and its phosphorylation protein, suggesting that metformin upregulated FOXO1 activity by AMPK activation. Similar results were obtained after treatment with insulin. In addition, transfection with siRNA for FOXO1 cancelled metformin-inhibited cell growth, indicating that FOXO1 mediated metformin to inhibit EC cell proliferation. A xenograft mouse model further revealed that metformin suppressed HEC-1B tumor growth, accompanied by downregulated ki-67 and upregulated AMPK phosphorylation and nuclear FOXO1 protein. Taken together, these data provide a novel mechanism of antineoplastic effect for metformin through the regulation of FOXO1, and suggest that the AMPK-FOXO1 pathway may be a therapeutic target to the development of new antineoplastic drugs.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Estrógenos/metabolismo , Proteína Forkhead Box O1/metabolismo , Metformina/farmacología , Transducción de Señal/efectos de los fármacos , Adulto , Anciano , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/genética , Activación Enzimática , Femenino , Humanos , Ratones , Persona de Mediana Edad , Fosforilación , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA