Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
1.
IUBMB Life ; 73(1): 213-222, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33249758

RESUMEN

Diabetes mellitus (DM)-induced glucolipotoxicity is a factor strongly contributing to alveolar bone deficiency. Parathyroid hormone (PTH) has been identified as a main systemic mediator to balance physiological calcium in bone. This study aimed to uncover PTH's potential role in ameliorating the osteogenic capacity of human bone marrow mesenchymal stem cells (HBMSCs) against glucolipotoxicity. Optimal PTH concentrations and high glucose and palmitic acid (GP) were administered to cells, followed by alkaline phosphatase (ALP) staining and ALP activity assay. Quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR) and Immunoblot were carried out for assessing mRNA and protein amounts, respectively. Cell counting kit-8 (CCK-8) and flow cytometry were performed for quantitating cell proliferation. Osteogenesis and oxidative stress were determined, and the involvement of mitogen-activated protein kinase (MAPK) signaling was further verified. About 1-50 mmol/ml GP significantly inhibited the osteogenic differentiation of HBMSCs. 10-9 mol/L PTH was found to be the optimal concentration for HBMSC induction. PTH had no effects on HBMSC proliferation, with or without GP treatment. PTH reversed inadequate osteogenesis and excessive oxidative stress in GP-treated HBMSCs. Mechanistically, PTH activated p38 MAPK signaling, while inhibiting p38 MAPK-suppressed PTH's beneficial impacts on HBMSCs. Collectively, PTH promotes osteogenic differentiation in HBMSCs against glucolipotoxicity via p38 MAPK signaling.


Asunto(s)
Glucosa/efectos adversos , Células Madre Mesenquimatosas/citología , Osteogénesis , Ácido Palmítico/efectos adversos , Hormona Paratiroidea/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Adulto , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Inhibidores Enzimáticos/efectos adversos , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Transducción de Señal , Edulcorantes/efectos adversos , Adulto Joven , Proteínas Quinasas p38 Activadas por Mitógenos/genética
2.
Molecules ; 26(11)2021 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-34204938

RESUMEN

The aim of the study was to evaluate the influence of vitamin K2 (VK2) supplementation on the sphingolipid metabolism pathway in palmitate-induced insulin resistant hepatocytes. The study was carried out on human hepatocellular carcinoma cells (HepG2) incubated with VK2 and/or palmitic acid (PA). The concentrations of sphingolipids were measured by high-performance liquid chromatography. The expression of enzymes from the sphingolipid pathway was assessed by Western blotting. The same technique was used in order to determine changes in the expression of the proteins from the insulin signaling pathway in the cells. Simultaneous incubation of HepG2 cells with palmitate and VK2 elevated accumulation of sphinganine and ceramide with increased expression of enzymes from the ceramide de novo synthesis pathway. HepG2 treatment with palmitate and VK2 significantly decreased the insulin-stimulated expression ratio of insulin signaling proteins. Moreover, we observed that the presence of PA w VK2 increased fatty acid transport protein 2 expression. Our study showed that VK2 activated the ceramide de novo synthesis pathway, which was confirmed by the increase in enzymes expression. VK2 also intensified fatty acid uptake, ensuring substrates for sphingolipid synthesis through the de novo pathway. Furthermore, increased concentration of sphingolipids, mainly sphinganine, inhibited insulin pathway proteins phosphorylation, increasing insulin resistance development.


Asunto(s)
Vías Biosintéticas/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Ceramidas/análisis , Resistencia a la Insulina , Neoplasias Hepáticas/metabolismo , Ácido Palmítico/efectos adversos , Vitamina K 2/farmacología , Cromatografía Líquida de Alta Presión , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Humanos , Insulina/metabolismo , Modelos Biológicos , Fosforilación , Esfingosina/análogos & derivados , Esfingosina/análisis , Regulación hacia Arriba
3.
Mol Med ; 25(1): 17, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-31060494

RESUMEN

BACKGROUND: Obesity is one of the leading causes of insulin resistance. Accumulating reports have highlighted that serum amyloid A-1 (SAA1) is a potential candidate that is capable of attenuating insulin resistance. Hence, we conducted the current study with aims of investigating our proposed hypothesis that silencing SAA1 could inhibit the progression of obesity-induced insulin resistance through the NF-κB pathway. METHODS: Gene expression microarray analysis was initially performed to screen differentially expressed genes (DEGs) associated with obesity. Palmitate (PA)-induced insulin resistance Huh7 cell models and high-fat diet (HFD)-induced mouse models were established to elucidate the effect of SAA1/Saa1 on insulin resistance. The NF-κB pathway-related expression was subsequently determined through the application of reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis. RESULTS: Saa1 was identified as an obesity-related gene based on the microarray data of GSE39549. Saa1 was determined to be highly expressed in HFD-induced insulin resistance mouse models. PA-induced Huh7 cells, treated with silenced SAA1 or NF-κB pathway inhibition using BAY 11-7082, displayed a marked decrease in both Saa1 and SOCS3 as well as an elevation in 2DG, IRS1 and the extent of IRS1 phosphorylation. HFD mice treated with silenced Saa1 or inhibited NF-κB pathway exhibited improved fasting blood glucose (FBG) levels as well as fasting plasma insulin (FPI) levels, glucose tolerance and systemic insulin sensitivity. Saa1/SAA1 was determined to show a stimulatory effect on the transport of the NF-κBp65 protein from the cytoplasm to the nucleus both in vivo and in vitro, suggesting that Saa1/SAA1 could activate the NF-κB pathway. CONCLUSION: Taken together, our key findings highlight a novel mechanism by which silencing of SAA1 hinders PA or HFD-induced insulin resistance through inhibition of the NF-κB pathway.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Resistencia a la Insulina/fisiología , FN-kappa B/metabolismo , Ácido Palmítico/efectos adversos , Proteína Amiloide A Sérica/metabolismo , Animales , Glucemia/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Ayuno/sangre , Prueba de Tolerancia a la Glucosa , Humanos , Insulina/sangre , Resistencia a la Insulina/genética , Ratones , Ratones Endogámicos C57BL , Análisis por Matrices de Proteínas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Amiloide A Sérica/genética , Transducción de Señal/efectos de los fármacos
4.
Biochem Biophys Res Commun ; 513(1): 201-206, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-30952426

RESUMEN

Elevated saturated free fatty acid levels during over-nutrition lead to hypothalamic inflammation, which perturbs energy homeostasis. Whether brain-derived metabolites are coupled to the development of obesity pathogenesis during the early over-nutrition period has not been thoroughly investigated. In this study, we found increased linoleic acid, an unsaturated fatty acid, in both the whole brain and hypothalamus of mice fed a high-fat diet for 4 weeks. Furthermore, we observed that linoleic acid effectively reversed the inflammatory responses induced by palmitic acid treatment in microglial cells. Collectively, this study suggests the reversible function of linoleic acid on brain inflammation in association with microglial activation during short-term exposure to a high-fat diet.


Asunto(s)
Antiinflamatorios/uso terapéutico , Dieta Alta en Grasa/efectos adversos , Encefalitis/tratamiento farmacológico , Ácido Linoleico/uso terapéutico , Microglía/efectos de los fármacos , Animales , Antiinflamatorios/inmunología , Encefalitis/etiología , Encefalitis/inmunología , Ácido Linoleico/inmunología , Ratones Endogámicos C57BL , Microglía/inmunología , Ácido Palmítico/efectos adversos , Ácido Palmítico/inmunología
5.
Biol Pharm Bull ; 42(5): 671-679, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31061309

RESUMEN

Nicotiana glauca is a cosmopolitan shrub, used in medicine to treat swellings, wounds, sores and cancer. However, its users lack of knowledge of the adverse effects. We seek to evaluate the effects of lipid extracts from N. glauca on myoblasts, identifying the compounds which cause undesirable effects. Myoblasts are important in muscle homeostasis, thus a high death rate of them cause myopathies. We performed an ethanolic extraction from leaves of N. glauca and the extract was successively partitioned with hexane, chloroform and ethyl acetate. The effects of extracts in C2C12 cells were analysed by terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL), Mitotracker and 4',6-diamidino-2-phenylindole (DAPI) staining, Western blotting, real-time PCR and immunofluorescence assays. Caspase activity was studied. The fraction with the highest apoptotic effects was analysed by chromatography, NMR and GC-MS spectrometry were used to identify the apoptotic agent, after which its biological activity was evaluated. The extracts from N. glauca induced apoptosis in C2C12 cells involving caspase-3/7. We found that the extracts trigger a defence response in muscle through Akt and heat shock protein 27 (HSP27). We identified an apoptotic agent as palmitic acid. These data suggest that the use of N. glauca in hormone replacement therapy, or in other therapies affects skeletal muscle homeostasis, worsening the negative effects of the menopause. Thus, the relevance of this work lies in the fact that it is the first time that a report about the molecular mechanism responsible for the side effects of medicinal use of N. glauca, has been shown. Moreover the compound responsible for these effects has been identified.


Asunto(s)
Mioblastos Esqueléticos/efectos de los fármacos , Nicotiana , Ácido Palmítico/efectos adversos , Fitoterapia/efectos adversos , Extractos Vegetales/efectos adversos , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Proteínas de Choque Térmico HSP27/metabolismo , Ratones , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética
6.
Can J Physiol Pharmacol ; 97(12): 1132-1140, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31374178

RESUMEN

Long-chain saturated fatty acids, especially palmitic acid (PA), contribute to cardiomyocyte lipotoxicity. This study tests the effects of PA on adult rat cardiomyocyte contractile function and proteins associated with calcium regulating cardiomyocyte contraction and relaxation. Adult rat cardiomyocytes were pretreated with resveratrol (Resv) and then treated with PA. For the reversal study, cardiomyocytes were incubated with PA prior to treatment with Resv. Cardiomyocyte contractility, ratio of rod- to round-shaped cardiomyocytes, and Hoechst staining were used to measure functional and morphological changes in cardiomyocytes. Protein expression of sarco-endoplasmic reticulum ATPase 2a (SERCA2a), native phospholamban (PLB) and phosphorylated PLB (pPLB ser16 and pPLB thr17), and troponin I (TnI) and phosphorylated TnI (pTnI) were measured. SERCA2a activity was also measured. Our results show that PA (200 µM) decreased the rate of cardiomyocyte relaxation, reduced the number of rod-shaped cardiomyocytes, and increased the number of cells with condensed nuclei; pre-treating cardiomyocytes with Resv significantly prevented these changes. Post-treatment with Resv did not reverse morphological changes induced by PA. Protein expression levels of SERCA2a, PLB, pPLBs, TnI, and pTnI were unchanged by PA or Resv. SERCA2a activity assay showed that Vmax and Iono ratio were increased with PA and pre-treatment with Resv prevented this increase. In conclusion, our results show that Resv protect cardiomyocytes from contractile dysfunction induced by PA.


Asunto(s)
Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Ácido Palmítico/efectos adversos , Resveratrol/farmacología , Animales , Apoptosis/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Ratas , Ratas Sprague-Dawley , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Troponina I/metabolismo
7.
Cell Mol Biol Lett ; 24: 5, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30820195

RESUMEN

OBJECTIVE: Tetrahydroxy stilbene glucoside (TSG) has been reported to exert a cytoprotective effect against various toxicants. However, the function and mechanism of TSG in palmitic acid (PA)-induced inflammation and apoptosis in cardiomyocytes are still unknown. The present study was designed to investigate the post-transcriptional mechanism in TSG-treated cardiomyocytes' inflammation and apoptosis induced by PA. METHODS: The mRNA and protein levels were assayed by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting, respectively. The targeted genes were predicted by a bioinformatics algorithm and confirmed by a dual luciferase reporter assay. Cell proliferation was analyzed by CCK-8 assay. Annexin V-fluorescein isothiocyanate/polyimide (annexin V-FITC/PI) staining was used to evaluate apoptosis using flow cytometry. RESULTS: TSG restricted the detrimental effects, including the activated inflammatory response and apoptosis, of PA in cardiomyocytes, as well as the up-regulation of miR-129-3p and down-regulation of p-Smad3 expression. In addition, bioinformatics and experimental analysis suggested that Smad3 was a direct target of miR-129-3p, which could inhibit or enhance the expression of p-Smad by transfection with miR-129-3p mimics or inhibitors, respectively. Furthermore, our results demonstrated that overexpression of Smad3 reversed the inhibition of inflammation and apoptosis by overexpression of miR-129-3p in PA-stimulated cardiomyocytes. CONCLUSION: TSG targeted to miR-129-3p/Smad3 signaling inhibited PA-induced inflammation and apoptosis in cardiomyocytes.


Asunto(s)
Apoptosis/efectos de los fármacos , Glucósidos/farmacología , Inflamación/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Transducción de Señal , Estilbenos/farmacología , Animales , Línea Celular , Inflamación/inducido químicamente , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ácido Palmítico/efectos adversos , Sustancias Protectoras/farmacología , Ratas , Proteína smad3/metabolismo
8.
BMC Complement Altern Med ; 19(1): 255, 2019 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-31519174

RESUMEN

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease and is characterized by excessive hepatic lipid accumulation. Many studies have suggested that lipid overload is the key initial factor that contributes to hepatic steatosis. Our previous study indicated that diosgenin (DSG) has a beneficial effect on energy metabolism, but the underlying mechanism remains unclear. METHODS: Human normal hepatocytes (LO2 cells) were incubated with palmitic acid to establish the cell model of nonalcoholic fatty liver. The effects of DSG on lipid metabolism, glucose uptake and mitochondrial function were evaluated. Furthermore, the mechanism of DSG on oxidative stress, lipid consumption and lipid synthesis in LO2 cells was investigated. RESULTS: The results indicated that palmitic acid induced obvious lipid accumulation in LO2 cells and that DSG treatment significantly reduced the intracellular lipid content. DSG treatment upregulated expression of lipolysis proteins, including phospho-AMP activated protein kinase (p-AMPK), phospho-acetyl-coA carboxylase (p-ACC) and carnitine acyl transferase 1A (CPT-1A), and inhibited expression of lipid synthesis-related proteins, including sterol regulatory element-binding protein 1c (SREBP-1c) and fatty acid synthase (FAS). Additionally, DSG-treated cells displayed a marked improvement in mitochondrial function, with less production of reactive oxygen species and a higher mitochondrial membrane potential compared with the model group. CONCLUSION: This study suggests that DSG can reduce intracellular lipid accumulation in LO2 cells and that the underlying mechanism may be related to the improving oxidative stress, increasing fatty acid ß-oxidation and decreasing lipid synthesis. The above changes might be mediated by the activation of the AMPK/ACC/CPT-1A pathway and inhibition of the SREBP-1c/FAS pathway.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Diosgenina/farmacología , Ácido Graso Sintasas/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Ácido Palmítico/efectos adversos , Proteínas Quinasas Activadas por AMP/genética , Acetil-CoA Carboxilasa/genética , Carnitina O-Acetiltransferasa/genética , Carnitina O-Acetiltransferasa/metabolismo , Línea Celular , Ácido Graso Sintasas/genética , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/genética , Transducción de Señal/efectos de los fármacos , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
9.
J Cell Biochem ; 119(12): 9752-9763, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30129212

RESUMEN

The formation of atherosclerotic changes leads to dysfunction in numerous cell types, especially endothelial cells. In the current experiment, we aimed to show the therapeutic effect of Docosahexaenoic acid on palmitic-induced atherosclerotic changes in the human endothelial lineage. Human Umbilical Vein Endothelial cells were incubated with 1 mM palmitic acid for 48 hours and then exposed to 40 µM docosahexaenoic acid for next 24 hours. Cellular atherosclerosis and lipid removal were confirmed by the application of Oil red O solution. The cell survival rate was studied by using MTT assay and flow cytometry analysis of Annexin V. We also measured the protein level of tumor necrosis factor-α and granulocyte-macrophage colony-stimulating factor by immunofluorescence imaging. The transcription level of genes participating in the atherosclerosis signaling pathway was monitored in atherosclerotic endothelial cells before and after treatment with docosahexaenoic acid. The viability of the cells was reduced after 48 hours incubation with palmitic acid. It is noteworthy that the number of viable endothelial cells was increased after exposure to docosahexaenoic acid. Compared with the cells that received palmitic acid, Oil red O staining showed a decrease in the cellular content of fatty acid after incubation with docosahexaenoic acid (P < 0.05). PCR array indicated that the modulation of key genes played a role in atherosclerosis and reached near-control levels. These data support the notion that incubation of atherosclerotic human endothelial cells with docosahexaenoic acid could return the detrimental effects of palmitic acid by modulation of the atherosclerosis signaling pathway.


Asunto(s)
Aterosclerosis/genética , Ácidos Docosahexaenoicos/farmacología , Ácido Palmítico/efectos adversos , Apoptosis/efectos de los fármacos , Aterosclerosis/metabolismo , Aterosclerosis/patología , Supervivencia Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ácido Palmítico/farmacocinética , Reacción en Cadena de la Polimerasa , Transducción de Señal/genética , Factor de Necrosis Tumoral alfa/metabolismo
10.
J Cell Biochem ; 119(4): 3363-3372, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29130531

RESUMEN

Metabolic syndrome is a risk factor for the development of cardiovascular diseases. Myocardial cell damage leads to an imbalance of energy metabolism, and many studies have indicated that short-term hypoxia during myocardial cell injury has a protective effect. In our previous animal studies, we found that short-term hypoxia in the heart has a protective effect, but long-term hypoxia increases myocardial cell injury. Palmitic acid (PA)-treated H9c2 cardiomyoblasts and neonatal rat ventricle cardiomyocytes were used to simulate hyperlipidemia model, which suppress cluster of differentiation 36 (CD36) and activate glucose transporter type 4 (GLUT4). We exposed the cells to short- and long-term hypoxia and investigated the protective effects of hypoxic preconditioning on PA-induced lipotoxicity in H9c2 cardiomyoblasts and neonatal rat cardiomyocytes. Preconditioning with short-term hypoxia enhanced both CD36 and GLUT4 metabolism pathway protein levels. Expression levels of phospho-PI3K, phospho-Akt, phospho-AMPK, SIRT1, PGC1α, PPARα, CD36, and CPT1ß induced by PA was reversed by short-term hypoxia in a time-dependent manner. PA-induced increased GLUT4 membrane protein level was reduced in the cells exposed to short-term hypoxia and si-PKCζ. Short-term hypoxia, resveratrol and si-PKCζ rescue H9c2 cells from apoptosis induced by PA and switch the metabolic pathway from GLUT4 dependent to CD36 dependent. We demonstrate short-term hypoxic preconditioning as a more efficient way as resveratrol in maintaining the energy metabolism of hearts during hyperlipidemia and can be used as a therapeutic strategy.


Asunto(s)
Antígenos CD36/metabolismo , Proteínas de Unión al ADN/metabolismo , Ácidos Grasos/metabolismo , Hiperlipidemias/metabolismo , Miocitos Cardíacos/citología , Ácido Palmítico/efectos adversos , Factores de Transcripción/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Hipoxia de la Célula , Línea Celular , Glucosa/metabolismo , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Redes y Vías Metabólicas , Modelos Biológicos , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ácido Palmítico/farmacología , Ratas
11.
J Pineal Res ; 65(2): e12493, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29607540

RESUMEN

Melatonin plays an important role in regulating circadian rhythms. It also acts as a potent antioxidant and regulates glucose and lipid metabolism, although the exact action mechanism is not clear. The α2-HS-glycoprotein gene (AHSG) and its protein, fetuin-A (FETUA), are one of the hepatokines and are known to be associated with insulin resistance and type 2 diabetes. The aim of this study was to determine whether melatonin improves hepatic insulin resistance and hepatic steatosis in a FETUA-dependent manner. In HepG2 cells treated with 300 µmol/L of palmitic acid, phosphorylated AKT expression decreased, and FETUA expression increased, but this effect was inhibited by treatment with 10 µmol/L of melatonin. However, melatonin did not improve insulin resistance in FETUA-overexpressing cells, indicating that improvement in insulin resistance by melatonin was dependent on downregulation of FETUA. Moreover, melatonin decreased palmitic acid-induced ER stress markers, CHOP, Bip, ATF-6, XBP-1, ATF-4, and PERK. In addition, in the high-fat diet (HFD) mice, oral treatment with 100 mg/kg/day melatonin for 10 weeks reduced body weight gain to one-third of that of the HFD group and hepatic steatosis. Insulin sensitivity and glucose intolerance improved with the upregulation of muscle p-AKT protein expression. FETUA expression and ER stress markers in the liver and serum of HFD mice were decreased by melatonin treatment. In conclusion, melatonin can improve hepatic insulin resistance and hepatic steatosis through reduction in ER stress and the resultant AHSG expression.


Asunto(s)
Grasas de la Dieta/efectos adversos , Hígado Graso/tratamiento farmacológico , Hígado Graso/metabolismo , Resistencia a la Insulina , Melatonina/farmacología , alfa-2-Glicoproteína-HS/metabolismo , Animales , Grasas de la Dieta/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Hígado Graso/inducido químicamente , Hígado Graso/patología , Células Hep G2 , Humanos , Ratones , Ácido Palmítico/efectos adversos , Ácido Palmítico/farmacología
12.
J Biol Chem ; 291(9): 4462-72, 2016 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26733200

RESUMEN

Autophagy is an evolutionarily conserved degradative process that allows cells to maintain homoeostasis in numerous physiological situations. This process also functions as an essential protective response to endoplasmic reticulum (ER) stress, which promotes the removal and degradation of unfolded proteins. However, little is known regarding the mechanism by which autophagy is initiated and regulated in response to ER stress. In this study, different types of autophagy were identified in human gastric cancer MKN45 cells in response to the stress induced by nutrient starvation or lipotoxicity in which the regulation of these pathways is mammalian target of rapamycin (mTOR)-dependent or -independent, respectively. Interestingly, we found that p8, a stress-inducible transcription factor, was enhanced in MKN45 cells treated with palmitic acid to induce lipotoxicity. Furthermore, an increase in autophagy was observed in MKN45 cells stably overexpressing p8 using a lentivirus system, and autophagy induced by palmitic acid was blocked by p8 RNAi compared with the control. Western blotting analyses showed that autophagy was regulated by p8 or mTOR in response to the protein kinase-like endoplasmic reticulum kinase/activating transcription factor 6-mediated ER stress of lipotoxicity or the parkin-mediated mitochondrial stress of nutrient starvation, respectively. Furthermore, our results indicated that autophagy induced by palmitic acid is mTOR-independent, but this autophagy pathway was regulated by p8 via p53- and PKCα-mediated signaling in MKN45 cells. Our findings provide insights into the role of p8 in regulating autophagy induced by the lipotoxic effects of excess fat accumulation in cells.


Asunto(s)
Autofagia , Proteínas de Unión al ADN/metabolismo , Estrés del Retículo Endoplásmico , Ácidos Grasos no Esterificados/efectos adversos , Transducción de Señal , Factores de Transcripción/metabolismo , Regulación hacia Arriba , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Estrés del Retículo Endoplásmico/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Microscopía Fluorescente , Ácido Palmítico/efectos adversos , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Regulación hacia Arriba/efectos de los fármacos
13.
Arterioscler Thromb Vasc Biol ; 36(9): 2011-8, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27417581

RESUMEN

OBJECTIVE: We assessed whether the association between dietary saturated fatty acids (SFA) and incident coronary heart disease (CHD) depends on the food source, the carbon chain length of SFA, and the substituting macronutrient. APPROACH AND RESULTS: From the Rotterdam Study, 4722 men and women (≥55 years) were included. Baseline (1990-1993) SFA intake was assessed using a validated food frequency questionnaire. CHD (nonfatal myocardial infarction and fatal CHD) was ascertained by medical records. Using multivariable Cox regression analysis, we calculated CHD risks for higher intakes of total SFA, SFA from specific food sources, SFA differing in carbon chain length, and substituting other macronutrients instead of SFA. During a median follow-up of 16.3 years, 659 CHD events occurred. Total SFA intake was not associated with CHD risk (hazard ratio [HR] per 5 en%, 1.13; 95% confidence interval, 0.94-1.22), and neither was SFA from specific food sources. A higher CHD risk was observed for palmitic acid (16:0) intake (HRSD, 1.26; 95% confidence interval, 1.05-1.15) but not for SFA with other chain lengths. Except for a higher CHD risk for substitution of SFA with animal protein (HR5en%, 1.24; 95% confidence interval, 1.01-1.51), substitution with other macronutrients was not associated with CHD. CONCLUSIONS: In this Dutch population, we observed that a higher intake of palmitic acid, which accounts for ≈50% of the total SFA intake, was associated with a higher CHD risk, as was substitution of total SFA with animal protein. Nevertheless, we found no association between total SFA intake and CHD risk, which did not differ by food source.


Asunto(s)
Enfermedad Coronaria/epidemiología , Dieta , Conducta Alimentaria , Ácido Palmítico/efectos adversos , Factores de Edad , Anciano , Enfermedad Coronaria/diagnóstico , Dieta/efectos adversos , Proteínas en la Dieta/efectos adversos , Femenino , Estudios de Seguimiento , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Análisis Multivariante , Países Bajos , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Medición de Riesgo , Factores de Riesgo
14.
Mol Ther ; 24(11): 1926-1938, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27502608

RESUMEN

Growth differentiation factor 11 (GDF11) reduces cardiac hypertrophy, improves cerebral vasculature and enhances neurogenesis in ageing mice. Higher growth differentiation factor 11/8 (GDF11/8) is associated with lower risk of cardiovascular events in humans. Here, we showed that adeno-associated viruses-GDF11 and recombinant GDF11 protein improve endothelial dysfunction, decrease endothelial apoptosis, and reduce inflammation, consequently decrease atherosclerotic plaques area in apolipoprotein E-/- mice. Moreover, adeno-associated viruses-GDF11 and recombinant GDF11 stabilize atherosclerotic plaques by selectively decreasing in macrophages and T lymphocytes, while increasing in collagen and vascular smooth muscle cells within plaques. In addition, GDF11 inhibit palmitic acid-induced endothelial apoptosis and ameliorate palmitic acid-induced inflammatory response in RAW264.7 macrophages in vitro. Mechanistically, GDF11 activates the TGF-ß/Smad2/3, AMPK/endothelial nitricoxide synthase (eNOS) while suppresses JNK and NF-κB pathways. In humans, circulating GDF11/8 is positively associated with flow-mediated endothelium-dependent dilation in overweight subjects. We concluded that adeno-associated viruses-GDF11 and recombinant GDF11 protect against endothelial injury and reduce atherosclerosis in apolipoprotein E-/- mice, thus may be providing a novel approach to the treatment of atherosclerosis.


Asunto(s)
Apolipoproteínas E/genética , Aterosclerosis/terapia , Proteínas Morfogenéticas Óseas/genética , Células Endoteliales/efectos de los fármacos , Terapia Genética/métodos , Factores de Diferenciación de Crecimiento/genética , Proteínas Recombinantes/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Aterosclerosis/genética , Aterosclerosis/metabolismo , Células Cultivadas , Dependovirus/genética , Modelos Animales de Enfermedad , Células Endoteliales/citología , Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/farmacología , Humanos , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Ácido Palmítico/efectos adversos , Células RAW 264.7 , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Linfocitos T/metabolismo
15.
J Biol Chem ; 290(38): 23282-90, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26240153

RESUMEN

Aberrant deposition of fat including free fatty acids in the liver often causes damage to hepatocytes, namely lipotoxicity, which is a key pathogenic event in the development and progression of fatty liver diseases. This study demonstrates a pivotal role of sphingosine kinase 1 (SphK1) in protecting hepatocytes from lipotoxicity. Exposure of primary murine hepatocytes to palmitate resulted in dose-dependent cell death, which was enhanced significantly in Sphk1-deficient cells. In keeping with this, expression of dominant-negative mutant SphK1 also markedly promoted palmitate-induced cell death. In contrast, overexpression of wild-type SphK1 profoundly protected hepatocytes from lipotoxicity. Mechanistically, the protective effect of SphK1 is attributable to suppression of ER stress-mediated pro-apoptotic pathways, as reflected in the inhibition of IRE1α activation, XBP1 splicing, JNK phosphorylation, and CHOP induction. Of note, SphK1 inhibited the IRE1α pathway by reducing IRE1α expression at the transcriptional level. Moreover, S1P mimicked the effect of SphK1, suppressing IRE1α expression in a receptor-dependent manner. Furthermore, enforced overexpression of IRE1α significantly blocked the protective effect of SphK1 against lipotoxicity. Therefore, this study provides new insights into the role of SphK1 in hepatocyte survival and uncovers a novel mechanism for protection against ER stress-mediated cell death.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Endorribonucleasas/biosíntesis , Inhibidores Enzimáticos/efectos adversos , Hepatocitos/metabolismo , Ácido Palmítico/efectos adversos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Proteínas Serina-Treonina Quinasas/biosíntesis , Animales , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/genética , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/genética , Endorribonucleasas/genética , Inhibidores Enzimáticos/farmacología , Hepatocitos/patología , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Ácido Palmítico/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Proteínas Serina-Treonina Quinasas/genética , Empalme del ARN/efectos de los fármacos , Empalme del ARN/genética , Factores de Transcripción del Factor Regulador X , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética , Proteína 1 de Unión a la X-Box
16.
Biochem Biophys Res Commun ; 476(4): 665-669, 2016 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-27246737

RESUMEN

It is known that endoplasmic reticulum stress (ERS) contributes to insulin resistance (IR) and non-alcoholic fatty liver disease (NAFLD) in mammals. However, we recently demonstrated that overfeeding with a traditional diet (mainly consisting of cooked maize) does not induce ERS in goose. As cellular studies show that high glucose and palmitate can trigger ERS in mammalian cells, we hypothesized that supplementing sugar to the traditional diet could induce ERS, thus promoting insulin resistance and fatty liver. To test the hypothesis, we first treated goose primary hepatocytes with high glucose (25 mM and 50 mM) and palmitate (0.5 mM) supplemented with or without 0.25 mM oleate. Data indicated that, as in mammalian cells, high glucose and palmitate indeed induced ERS in goose primary hepatocytes, and palmitate-induced ERS was suppressed by supplemental 0.25 mM oleate. We then tested the hypothesis with an in vivo study, in which Landes geese overfed with traditional or novel diets (i.e., the traditional diet supplemented with sugar) were compared with control geese (normally fed with cooked maize) for ERS, IR and fatty liver. The differences in glucose tolerance, insulin tolerance and postprandial blood glucose between the geese overfed with traditional and novel diets suggested that supplementing dietary sugar promoted IR. This promotion was accompanied with an increasing trend of liver weight and abdominal fat weight relative to body weight. Surprisingly, compared to overfeeding with the traditional diet, overfeeding with the novel diet did not induce ERS, even further suppressed ERS in goose fatty liver. Together, our findings suggest that supplementing dietary sugar promotes ERS-independent IR and fatty liver in goose. It is intriguing to discover the factor(s) protecting goose liver from ERS as well as the non-ERS mechanism underlying IR.


Asunto(s)
Carbohidratos de la Dieta/administración & dosificación , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/fisiología , Hígado Graso/etiología , Resistencia a la Insulina/fisiología , Animales , Células Cultivadas , Carbohidratos de la Dieta/efectos adversos , Chaperón BiP del Retículo Endoplásmico , Hígado Graso/metabolismo , Hígado Graso/patología , Gansos , Expresión Génica/efectos de los fármacos , Glucosa/administración & dosificación , Glucosa/efectos adversos , Prueba de Tolerancia a la Glucosa , Proteínas de Choque Térmico/genética , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Ácido Oléico/administración & dosificación , Tamaño de los Órganos/efectos de los fármacos , Ácido Palmítico/administración & dosificación , Ácido Palmítico/efectos adversos
17.
Klin Lab Diagn ; 61(2): 68-77, 2016 Feb.
Artículo en Ruso | MEDLINE | ID: mdl-27455558

RESUMEN

In phylogenesis, becoming of biologicalfunctions and biological reactions proceeds with the purpose ofpermanent increasing of "kinetic perfection ". The main role belongs to factors ofphysical, chemical and biological kinetics, their evaluation using systemic approach technique under permanent effect of natural selection. The late-in-phylogenesis insulin, proceeded with, in development of biological function of locomotion, specialization of insulin-dependent cells: skeletal myocytes, syncytium of cardiomyocytes, subcutaneous adipocytes, periportal hepatocytes, Kupffer's macrophages and ß-cells of islets of pancreas. The insulin initiated formation of new, late in phylogenesis, large pool of fatty cells-subcutaneous adipocytes that increased kinetic parameters of biological function of locomotion. In realization of biological function of locomotion only adipocytes absorb exogenous mono unsaturated and saturated fatty acids in the form of triglycerides in composition of oleic and palmitic lipoproteins of very low density using apoE/B-100 endocytosis. The rest of insulin-dependent cells absorb fatty acids in the form of unesterified fatty acids from associates with albumin and under effect of CD36 of translocase offatty acids. The insulin in all insulin-depended cells inhibits biological reaction of lipolysis enhancing contributing into development of lipoidosis. The insulin expresses transfer offatty acids in the form of unsaturated fatty acids from adipocytes into matrix of mitochondria. The insulin supplies insulin-dependent cells with substrates for acquiring energy subject to that in pool of unsaturated fatty acids in adipocytes prevails hydrophobic palmitic unsaturated fatiy acid that slowly passes into matrix through external membrane ofmitochondria; oxidases of mitochondria so slowly implement its ß-oxidation that content of exogenous palmitic unsaturatedfatty acid can't be higher than phylogenetic, physiological level - 15% of all amount offatty acids transferring to insulin-dependent cells. The insulin can't both to decrease content of exogenous palmitic fatty acid and inhibit lipolysis in visceral fatty cells of omentum.


Asunto(s)
Linfocitos B/efectos de los fármacos , Grasas de la Dieta/efectos adversos , Hepatocitos/efectos de los fármacos , Macrófagos del Hígado/efectos de los fármacos , Lipogénesis/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Ácido Palmítico/efectos adversos , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Linfocitos B/citología , Linfocitos B/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Insulina/metabolismo , Resistencia a la Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Macrófagos del Hígado/citología , Macrófagos del Hígado/metabolismo , Lipólisis/fisiología , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Miocardio/citología , Miocardio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Triglicéridos/metabolismo
18.
Biochem Biophys Res Commun ; 458(1): 1-7, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25592967

RESUMEN

Fenofibrate (FF) is widely used to lower blood lipids in clinical practice, but whether its protective effect on endothelium-dependent vasodilatation (EDV) in thoracic aorta is related with endoplasmic reticulum (ER) stress remains unknown. In this study, female Sprauge Dawley rats were divided into standard chow diets (SCD), high-fat diets (HFD) and HFD plus FF treatment group (HFD + FF) randomly. The rats of latter two groups were given HFD feeding for 5 months, then HFD + FF rats were treated with FF (30 mg/kg, once daily) via gavage for another 2 months. The pathological and tensional changes, protein expression of eNOS, and ER stress related genes in thoracic aorta were measured. Then impacts of palmitic acid (PA) and FF on EDV of thoracic aorta from normal female SD rats were observed. Ultimately the expression of ER stress related genes were assessed in primary mouse aortic endothelial cells (MAEC) treated by fenofibric acid (FA) and PA. We found that FF treatment improved serum lipid levels and pathological changes in thoracic aorta, accompanied with decreased ER stress and increased phosphorylation of eNOS. FF pretreatment also improved EDV impaired by different concentrations of PA treatment. The dose- and time-dependent inhibition of cell proliferation by PA were inverted by FA pretreatment. Phosphorylation of eNOS and expression of ER stress related genes were all inverted by FA pretreatment in PA-treated MAEC. Our findings show that fenofibrate recovers damaged EDV by chronic HFD feeding and acute stimulation of PA, this effect is related with decreased ER stress and increased phosphorylation of eNOS.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Fenofibrato/farmacología , Ácido Palmítico/efectos adversos , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aorta/patología , Células Cultivadas , Estrés del Retículo Endoplásmico/genética , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Femenino , Fenofibrato/análogos & derivados , Regulación de la Expresión Génica/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Obesidad/sangre , Obesidad/tratamiento farmacológico , Ratas Sprague-Dawley , Vasodilatación/efectos de los fármacos
19.
Molecules ; 20(9): 17339-61, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26393565

RESUMEN

A growing body of evidence highlights the close association between nutrition and human health. Fat is an essential macronutrient, and vegetable oils, such as palm oil, are widely used in the food industry and highly represented in the human diet. Palmitic acid, a saturated fatty acid, is the principal constituent of refined palm oil. In the last few decades, controversial studies have reported potential unhealthy effects of palm oil due to the high palmitic acid content. In this review we provide a concise and comprehensive update on the functional role of palm oil and palmitic acid in the development of obesity, type 2 diabetes mellitus, cardiovascular diseases and cancer. The atherogenic potential of palmitic acid and its stereospecific position in triacylglycerols are also discussed.


Asunto(s)
Grasas de la Dieta/efectos adversos , Ácido Palmítico/efectos adversos , Aceites de Plantas/química , Animales , Enfermedades Cardiovasculares/etiología , Diabetes Mellitus Tipo 2/etiología , Humanos , Ratones , Neoplasias/etiología , Obesidad/etiología , Aceite de Palma , Aceites de Plantas/efectos adversos
20.
Molecules ; 20(4): 5456-67, 2015 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-25826786

RESUMEN

It is well known that Phellinus linteus, which produces hispidin and its derivatives, possesses antioxidant activities. In this study, we investigated whether hispidin has protective effects on palmitate-induced oxidative stress in C2C12 skeletal muscle cells. Our results showed that palmitate treatment in C2C12 myotubes increased ROS generation and cell death as compared with the control. However, pretreatment of hispidin for 8 h improved the survival of C2C12 myotubes against palmitate-induced oxidative stress via inhibition of intracellular ROS production. Hispidin also inhibited palmitate-induced apoptotic nuclear condensation in C2C12 myotubes. In addition, we found that hispidin can suppress cleavage of caspase-3, expression of Bax, and NF-κB translocation. Therefore, these results suggest that hispidin is capable of protecting C2C12 myotubes against palmitate-induced oxidative stress.


Asunto(s)
Citoprotección , Fibras Musculares Esqueléticas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Pironas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Animales , Caspasa 3/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Fibras Musculares Esqueléticas/citología , FN-kappa B/metabolismo , Ácido Palmítico/efectos adversos , Ácido Palmítico/farmacología , Proteína X Asociada a bcl-2/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda