Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Pathol ; 258(3): 312-324, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36148647

RESUMEN

Despite the well-known hepatoprotective role of the epidermal growth factor receptor (EGFR) pathway upon acute damage, its specific actions during chronic liver disease, particularly cholestatic injury, remain ambiguous and unresolved. Here, we analyzed the consequences of inactivating EGFR signaling in the liver on the regenerative response following cholestatic injury. For that, transgenic mice overexpressing a dominant negative mutant human EGFR lacking tyrosine kinase activity (ΔEGFR) in albumin-positive cells were submitted to liver damage induced by 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC), an experimental model resembling human primary sclerosing cholangitis. Our results show an early activation of EGFR after 1-2 days of a DDC-supplemented diet, followed by a signaling switch-off. Furthermore, ΔEGFR mice showed less liver damage and a more efficient regeneration following DDC injury. Analysis of the mechanisms driving this effect revealed an enhanced activation of mitogenic/survival signals, AKT and ERK1/2-MAPKs, and changes in cell turnover consistent with a quicker resolution of damage in response to DDC. These changes were concomitant with profound differences in the profile of intrahepatic immune cells, consisting of a shift in the M1/M2 balance towards M2 polarity, and the Cd4/Cd8 ratio in favor of Cd4 lymphocytes, overall supporting an immune cell switch into a pro-restorative phenotype. Interestingly, ΔEGFR livers also displayed an amplified ductular reaction, with increased expression of EPCAM and an increased number of CK19-positive ductular structures in portal areas, demonstrating an overexpansion of ductular progenitor cells. In summary, our work supports the notion that hepatocyte-specific EGFR activity acts as a key player in the crosstalk between parenchymal and non-parenchymal hepatic cells, promoting the pro-inflammatory response activated during cholestatic injury and therefore contributing to the pathogenesis of cholestatic liver disease. © 2022 The Pathological Society of Great Britain and Ireland.


Asunto(s)
Hepatopatías , Regeneración Hepática , Albúminas/metabolismo , Albúminas/farmacología , Animales , Descarboxilasas de Aminoácido-L-Aromático/metabolismo , Descarboxilasas de Aminoácido-L-Aromático/farmacología , Molécula de Adhesión Celular Epitelial/metabolismo , Molécula de Adhesión Celular Epitelial/farmacología , Receptores ErbB/metabolismo , Hepatocitos/patología , Humanos , Hígado/patología , Hepatopatías/patología , Regeneración Hepática/fisiología , Ratones , Ratones Transgénicos , Proteínas Tirosina Quinasas , Proteínas Proto-Oncogénicas c-akt/metabolismo
2.
Cell Death Differ ; 29(12): 2362-2380, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35681014

RESUMEN

Activation of oval cells (OCs) has been related to hepatocyte injury during chronic liver diseases including non-alcoholic fatty liver disease (NAFLD). However, OCs plasticity can be affected under pathological environments. We previously found protection against hepatocyte cell death by inhibiting protein tyrosine phosphatase 1B (PTP1B). Herein, we investigated the molecular and cellular processes involved in the lipotoxic susceptibility in OCs expressing or not PTP1B. Palmitic acid (PA) induced apoptotic cell death in wild-type (Ptpn1+/+) OCs in parallel to oxidative stress and impaired autophagy. This lipotoxic effect was attenuated in OCs lacking Ptpn1 that showed upregulated antioxidant defences, increased unfolded protein response (UPR) signaling, higher endoplasmic reticulum (ER) content and elevated stearoyl CoA desaturase (Scd1) expression and activity. These effects in Ptpn1-/- OCs concurred with an active autophagy, higher mitochondrial efficiency and a molecular signature of starvation, favoring lipid droplet (LD) formation and dynamics. Autophagy blockade in Ptpn1-/- OCs reduced Scd1 expression, mitochondrial fitness, LD formation and restored lipoapoptosis, an effect also recapitulated by Scd1 silencing. PTP1B immunostaining was detected in OCs from mouse liver and, importantly, LDs were found in OCs from Ptpn1-/- mice with NAFLD. In conclusion, we demonstrated that Ptpn1 deficiency restrains lipoapoptosis in OCs through a metabolic rewiring towards a "starvation-like" fate, favoring autophagy, mitochondrial fitness and LD formation. Dynamic LD-lysosomal interations likely ensure lipid recycling and, overall, these adaptations protect against lipotoxicity. The identification of LDs in OCs from Ptpn1-/- mice with NAFLD opens therapeutic perspectives to ensure OC viability and plasticity under lipotoxic liver damage.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Animales , Ratones , Hepatocitos/metabolismo , Gotas Lipídicas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Ácido Palmítico/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Estearoil-CoA Desaturasa/genética , Estearoil-CoA Desaturasa/metabolismo , Eliminación de Gen
3.
Redox Biol ; 48: 102171, 2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34736121

RESUMEN

Therapeutic potential of metformin in obese/diabetic patients has been associated to its ability to combat insulin resistance. However, it remains largely unknown the signaling pathways involved and whether some cell types are particularly relevant for its beneficial effects. M1-activation of macrophages by bacterial lipopolysaccharide (LPS) promotes a paracrine activation of hypoxia-inducible factor-1α (HIF1α) in brown adipocytes which reduces insulin signaling and glucose uptake, as well as ß-adrenergic sensitivity. Addition of metformin to M1-polarized macrophages blunted these signs of brown adipocyte dysfunction. At the molecular level, metformin inhibits an inflammatory program executed by HIF1α in macrophages by inducing its degradation through the inhibition of mitochondrial complex I activity, thereby reducing oxygen consumption in a reactive oxygen species (ROS)-independent manner. In obese mice, metformin reduced inflammatory features in brown adipose tissue (BAT) such as macrophage infiltration, proinflammatory signaling and gene expression, and restored the response to cold exposure. In conclusion, the impact of metformin on macrophages by suppressing a HIF1α-dependent proinflammatory program is likely responsible for a secondary beneficial effect on insulin-mediated glucose uptake and ß-adrenergic responses in brown adipocytes.

4.
Redox Biol ; 26: 101263, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31299613

RESUMEN

Inflammation is typically associated with the development of fibrosis, cirrhosis and hepatocellular carcinoma. The key role of protein tyrosine phosphatase 1B (PTP1B) in inflammatory responses has focused this study in understanding its implication in liver fibrosis. Here we show that hepatic PTP1B mRNA expression increased after bile duct ligation (BDL), while BDL-induced liver fibrosis was markedly reduced in mice lacking Ptpn1 (PTP1B-/-) as assessed by decreased collagen deposition and α-smooth muscle actin (α-SMA) expression. PTP1B-/- mice also showed a significant increase in mRNA levels of key markers of monocytes recruitment (Cd68, Adgre1 and Ccl2) compared to their wild-type (PTP1B+/+) littermates at early stages of injury after BDL. Interestingly, the lack of PTP1B strongly increased the NADPH oxidase (NOX) subunits Nox1/Nox4 ratio and downregulated Cybb expression after BDL, revealing a pro-survival pattern of NADPH oxidase induction in response to liver injury. Chimeric mice generated by transplantation of PTP1B-/- bone marrow (BM) into irradiated PTP1B+/+ mice revealed similar hepatic expression profile of NOX subunits than PTP1B-/- mice while these animals did not show differences in infiltration of myeloid cells at 7 days post-BDL, suggesting that PTP1B deletion in other liver cells is necessary for boosting the early inflammatory response to the BDL. PTP1B-/- BM transplantation into PTP1B+/+ mice also led to a blockade of TGF-ß and α-SMA induction after BDL. In vitro experiments demonstrated that deficiency of PTP1B in hepatocytes protects against bile acid-induced apoptosis and abrogates hepatic stellate cells (HSC) activation, an effect ameliorated by NOX1 inhibition. In conclusion, our results have revealed that the lack of PTP1B switches NOX expression pattern in response to liver injury after BDL and reduces HSC activation and liver fibrosis.


Asunto(s)
Cirrosis Hepática/etiología , Cirrosis Hepática/metabolismo , NADPH Oxidasas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/deficiencia , Animales , Apoptosis/genética , Ácidos y Sales Biliares/metabolismo , Biomarcadores , Línea Celular , Medios de Cultivo Condicionados/metabolismo , Medios de Cultivo Condicionados/farmacología , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Células Estrelladas Hepáticas/metabolismo , Hepatocitos/metabolismo , Inmunohistoquímica , Macrófagos del Hígado/metabolismo , Cirrosis Hepática/patología , Masculino , Ratones , NADPH Oxidasas/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
5.
Sci Rep ; 8(1): 16461, 2018 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-30405191

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) is associated with post-operative liver failure (PLF) and impaired liver regeneration. We investigated the effects of a glucagon-like peptide-1 (GLP-1) receptor agonist on NAFLD, PLF and liver regeneration in mice fed chow diet or methionine/choline-deficient diet (MCD) or high fat diet (HFD). Fc-GLP-1 decreased transaminases, reduced intrahepatic triglycerides (TG) and improved MCD-induced liver dysfuction. Macrophage/Kupffer cell-related markers were also reduced although Fc-GLP-1 increased expression of genes related to natural killer (NK), cytotoxic T lymphocytes and hepatic stellate cell (HSC) activation. After partial hepatectomy (PH), survival rates increased in mice receiving Fc-GLP-1 on chow or MCD diet. However, the benefit of Fc-GLP-1 on NASH-like features was attenuated 2 weeks post-PH and liver mass restoration was not improved. At this time-period, markers of NK cells and cytotoxic T lymphocytes were further elevated in Fc-GLP-1 treated mice. Increased HSC related gene expression in livers was observed together with decreased retinyl ester content and increased retinal and retinoic acid, reflecting HSC activation. Similar effects were found in mice fed HFD receiving Fc-GLP-1. Our results shed light on the differential effects of a long-acting GLP-1R agonist in improving NAFLD and PLF, but not enhancing liver regeneration in mice.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón/agonistas , Hepatectomía , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Animales , Biomarcadores , Biopsia , Modelos Animales de Enfermedad , Expresión Génica , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Inmunohistoquímica , Mediadores de Inflamación , Hígado/inmunología , Hígado/metabolismo , Hígado/patología , Hígado/cirugía , Regeneración Hepática , Ratones , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/patología
6.
Mol Metab ; 7: 132-146, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29126873

RESUMEN

OBJECTIVES: Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in Western countries. Protein tyrosine phosphatase 1B (PTP1B), a negative modulator of insulin and cytokine signaling, is a therapeutic target for type 2 diabetes and obesity. We investigated the impact of PTP1B deficiency during NAFLD, particularly in non-alcoholic steatohepatitis (NASH). METHODS: NASH features were evaluated in livers from wild-type (PTP1BWT) and PTP1B-deficient (PTP1BKO) mice fed methionine/choline-deficient diet (MCD) for 8 weeks. A recovery model was established by replacing MCD to chow diet (CHD) for 2-7 days. Non-parenchymal liver cells (NPCs) were analyzed by flow cytometry. Oval cells markers were measured in human and mouse livers with NASH, and in oval cells from PTP1BWT and PTP1BKO mice. RESULTS: PTP1BWT mice fed MCD for 8 weeks exhibited NASH, NPCs infiltration, and elevated Fgf21, Il6 and Il1b mRNAs. These parameters decreased after switching to CHD. PTP1B deficiency accelerated MCD-induced NASH. Conversely, after switching to CHD, PTP1BKO mice rapidly reverted NASH compared to PTP1BWT mice in parallel to the normalization of serum triglycerides (TG) levels. Among NPCs, a drop in cytotoxic natural killer T (NKT) subpopulation was detected in PTP1BKO livers during recovery, and in these conditions M2 macrophage markers were up-regulated. Oval cells markers (EpCAM and cytokeratin 19) significantly increased during NASH only in PTP1B-deficient livers. HGF-mediated signaling and proliferative capacity were enhanced in PTP1BKO oval cells. In NASH patients, oval cells markers were also elevated. CONCLUSIONS: PTP1B elicits a dual role in NASH progression and reversion. Additionally, our results support a new role for PTP1B in oval cell proliferation during NAFLD.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Animales , Células Cultivadas , Colina/administración & dosificación , Dieta/efectos adversos , Molécula de Adhesión Celular Epitelial/sangre , Factores de Crecimiento de Fibroblastos/sangre , Humanos , Interleucina-1beta/sangre , Interleucina-6/sangre , Queratina-19/sangre , Hígado/metabolismo , Hígado/patología , Macrófagos/metabolismo , Masculino , Metionina/administración & dosificación , Metionina/deficiencia , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/etiología , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética
7.
Antioxid Redox Signal ; 27(16): 1332-1346, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28269997

RESUMEN

AIMS: Liver steatosis is associated with mitochondrial dysfunction and elevated reactive oxygen species (ROS) levels together with enhanced sensitivity to ischemia-reperfusion (IR) injury and limited response to preconditioning protocols. Here, we sought to determine whether the downregulation in the steatotic liver of peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α), a master regulator of mitochondrial metabolism and ROS that is known to play a role in liver metabolic control, could be responsible for the sensitivity of the steatotic liver to ischemic damage. RESULTS: PGC-1α was induced in normal liver after exposure to an IR protocol, which was concomitant with an increase in the levels of antioxidant proteins. By contrast, its induction was severely blunted in the steatotic liver, resulting in a modest induction of antioxidant proteins. Livers of PGC-1α-/- mice on a chow diet were normal, but they exhibited an enhanced sensitivity to IR injury and also a lack of response to ischemic preconditioning (IPC), a phenotype that recapitulated the features of the steatotic liver in terms of liver damage, although the inflammatory response differed between both models. Utilizing an in vitro model of IPC, we found that PGC-1α expression was downregulated in hepatic cells cultured at 1% O2; whereas it was induced after reoxygenation (3% O2), and it was responsible for the recovery of antioxidant gene expression after the ischemic period. Innovation & Conclusion: PGC-1α plays an important role in the protection against IR injury in the liver, which is likely associated with its capacity to induce antioxidant gene expression. Antioxid. Redox Signal. 27, 1332-1346.


Asunto(s)
Regulación hacia Abajo , Hígado Graso/patología , Hígado/irrigación sanguínea , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Animales , Antioxidantes/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Hígado Graso/genética , Hígado Graso/metabolismo , Técnicas de Inactivación de Genes , Hepatocitos , Precondicionamiento Isquémico , Ratones , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo
8.
Hepatology ; 65(3): 950-968, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27880981

RESUMEN

Because nonalcoholic steatohepatitis (NASH) is associated with impaired liver regeneration, we investigated the effects of G49, a dual glucagon-like peptide-1/glucagon receptor agonist, on NASH and hepatic regeneration. C57Bl/6 mice fed chow or a methionine and choline-deficient (MCD) diet for 1 week were divided into 4 groups: control (chow diet), MCD diet, chow diet plus G49, and M+G49 (MCD diet plus G49). Mice fed a high-fat diet (HFD) for 10 weeks were divided into groups: HFD and H+G49 (HFD plus G49). Following 2 (MCD groups) or 3 (HFD groups) weeks of treatment with G49, partial hepatectomy (PH) was performed, and all mice were maintained on the same treatment schedule for 2 additional weeks. Analysis of liver function, hepatic regeneration, and comprehensive genomic and metabolic profiling were conducted. NASH was ameliorated in the M+G49 group, manifested by reduced inflammation, steatosis, oxidative stress, and apoptosis and increased mitochondrial biogenesis. G49 treatment was also associated with replenishment of intrahepatic glucose due to enhanced gluconeogenesis and reduced glucose use through the pentose phosphate cycle and oxidative metabolism. Following PH, G49 treatment increased survival, restored the cytokine-mediated priming phase, and enhanced the proliferative capacity and hepatic regeneration ratio in mice on the MCD diet. NASH markers remained decreased in M+G49 mice after PH, and glucose use was shifted to the pentose phosphate cycle and oxidative metabolism. G49 administered immediately after PH was also effective at alleviating the pathological changes induced by the MCD diet. Benefits in terms of liver regeneration were also found in mice fed HFD and treated with G49. CONCLUSION: Dual-acting glucagon-like peptide-1/glucagon receptor agonists such as G49 represent a novel therapeutic approach for patients with NASH and particularly those requiring PH. (Hepatology 2017;65:950-968).


Asunto(s)
Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Regeneración Hepática/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Receptores de Glucagón/antagonistas & inhibidores , Animales , Biopsia con Aguja , Modelos Animales de Enfermedad , Péptido 1 Similar al Glucagón/farmacología , Humanos , Inmunohistoquímica , Peroxidación de Lípido , Regeneración Hepática/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/patología , Estrés Oxidativo , Distribución Aleatoria , Receptores de Glucagón/administración & dosificación , Resultado del Tratamiento
9.
Biochim Biophys Acta ; 1862(9): 1710-23, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27321932

RESUMEN

Cyclooxygenase-2 (COX-2) is involved in different liver diseases but little is known about the significance of COX-2 in the development and progression of non-alcoholic steatohepatitis (NASH). This study was designed to elucidate the role of COX-2 expression in hepatocytes in the pathogenesis of steatohepatitis and hepatic fibrosis. In the present work, hepatocyte-specific COX-2 transgenic mice (hCOX-2-Tg) and their wild-type (Wt) littermates were either fed methionine-and-choline deficient (MCD) diet to establish an experimental non-alcoholic steatohepatitis (NASH) model or injected with carbon tetrachloride (CCl4) to induce liver fibrosis. In our animal model, hCOX-2-Tg mice fed MCD diet showed lower grades of steatosis, ballooning and inflammation than Wt mice, in part by reduced recruitment and infiltration of hepatic macrophages, with a corresponding decrease in serum levels of pro-inflammatory cytokines. Furthermore, hCOX-2-Tg mice showed a significant attenuation of the MCD diet-induced increase in oxidative stress and hepatic apoptosis observed in Wt mice. Even more, hCOX-2-Tg mice treated with CCl4 had significantly lower stages of fibrosis and less hepatic content of collagen, hydroxyproline and pro-fibrogenic markers than Wt controls. Collectively, our data indicates that constitutive hepatocyte COX-2 expression ameliorates NASH and liver fibrosis development in mice by reducing inflammation, oxidative stress and apoptosis and by modulating activation of hepatic stellate cells, respectively, suggesting a possible protective role for COX-2 induction in NASH/NAFLD progression.


Asunto(s)
Ciclooxigenasa 2/genética , Hepatocitos/enzimología , Cirrosis Hepática/prevención & control , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Animales , Apoptosis , Células Cultivadas , Deficiencia de Colina/complicaciones , Ciclooxigenasa 2/metabolismo , Dinoprostona/farmacología , Modelos Animales de Enfermedad , Expresión Génica , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/enzimología , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Cirrosis Hepática/enzimología , Cirrosis Hepática/etiología , Masculino , Metionina/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Enfermedad del Hígado Graso no Alcohólico/enzimología , Enfermedad del Hígado Graso no Alcohólico/etiología , Estrés Oxidativo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
10.
J Nutr Biochem ; 23(12): 1676-84, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22464149

RESUMEN

Nonalcoholic steatosis is an important hepatic complication of obesity linked to mitochondrial dysfunction and insulin resistance. Furthermore, lipoic acid has been reported to have beneficial effects on mitochondrial function. In this study, we analyzed the potential protective effect of lipoic acid supplementation against the development of nonalcoholic steatosis associated with a long-term high-fat diet feeding and the potential mechanism of this effect. Wistar rats were fed on a standard diet (n=10), a high-fat diet (n=10) and a high-fat diet supplemented with lipoic acid (n=10). A group pair-fed to the latter group (n=6) was also included. Lipoic acid prevented hepatic triglyceride accumulation and liver damage in rats fed a high-fat diet (-68%±11.3% vs. obese group) through the modulation of genes involved in lipogenesis and mitochondrial ß-oxidation and by improving insulin sensitivity. Moreover, this molecule showed an inhibitory action on electron transport chain complexes activities (P<.01-P<.001) and adenosine triphosphate synthesis (P<.05), and reduced significantly energy efficiency. By contrast, lipoic acid induced an increase in mitochondrial copy number and in Ucp2 gene expression (P<.001 vs. obese). In summary, this investigation demonstrated the ability of lipoic acid to prevent nonalcoholic steatosis induced by a high-fat intake. Finally, the novelty and importance of this study are the finding of how lipoic acid modulates some of the mitochondrial processes involved in energy homeostasis. The reduction in mitochondrial energy efficiency could also explain, at least in part, the beneficial effects of lipoic acid not only in fatty liver but also in preventing excessive body weight gain.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Hígado Graso/prevención & control , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Ácido Tióctico/farmacología , Adenosina Trifosfato/metabolismo , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Ciclo del Ácido Cítrico/efectos de los fármacos , Enzimas/metabolismo , Hígado Graso/etiología , Regulación de la Expresión Génica/efectos de los fármacos , Insulina/sangre , Canales Iónicos/genética , Metabolismo de los Lípidos/efectos de los fármacos , Lipogénesis/efectos de los fármacos , Lipogénesis/genética , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Proteínas Mitocondriales/genética , Tamaño de los Órganos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Sustancias Protectoras/farmacología , Ratas , Ratas Wistar , Transaminasas/sangre , Triglicéridos/metabolismo , Proteína Desacopladora 2
11.
Obesity (Silver Spring) ; 20(10): 1974-83, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22327056

RESUMEN

Nonalcoholic steatosis is an important hepatic complication of obesity linked to mitochondrial dysfunction and oxidative stress. Lipoic acid (LA) has been reported to have beneficial effects on mitochondrial function and to attenuate oxidative stress. The sirtuin (SIRT) family has been demonstrated to play an important role in the regulation of mitochondrial function and in the activation of antioxidant defenses. In this study, we analyzed the potential protective effect of LA supplementation, via the modulation of mitochondrial defenses through the SIRT pathway, against oxidative stress associated with high-fat feeding. Wistar rats were fed a standard diet (control group (C), n = 10), a high-fat diet (obese group (OB), n = 10) and a high-fat diet supplemented with LA (OLIP, n = 10). A group pair-fed to the latter group (pair-fed OLIP group (PFO), n = 6) was also included. LA prevented hepatic triglyceride (TG) accumulation (-68.2%) and liver oxidative damage (P < 0.01) through the inhibition of hydroperoxide (H(2)O(2)) production (P < 0.001) and the stimulation of mitochondrial antioxidant defenses. LA treatment upregulated manganese superoxide dismutase (SOD2) (60.6%) and glutathione peroxidase (GPx) (100.2%) activities, and increased the reduced glutathione (GSH): oxidized glutathione (GSSG) ratio and UCP2 mRNA levels (P < 0.001-P < 0.01). Moreover, this molecule reduced oxidative damage in mitochondrial DNA (mtDNA) and increased mitochondrial copy number (P < 0.001- P < 0.01). LA treatment decreased the acetylation levels of Forkhead transcription factor 3a (Foxo3a) and PGC1ß (P < 0.001- P < 0.01) through the stimulation of SIRT3 and SIRT1 (P < 0.001). In summary, our results demonstrate that the beneficial effects of LA supplementation on hepatic steatosis could be mediated by its ability to restore the oxidative balance by increasing antioxidant defenses through the deacetylation of Foxo3a and PGC1ß by SIRT1 and SIRT3.


Asunto(s)
Antioxidantes/farmacología , Hígado Graso/tratamiento farmacológico , Hígado/patología , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Sirtuina 1/efectos de los fármacos , Sirtuina 3/efectos de los fármacos , Ácido Tióctico/farmacología , Animales , Antioxidantes/metabolismo , Western Blotting , Peso Corporal , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Hígado Graso/metabolismo , Hígado Graso/patología , Peroxidación de Lípido , Masculino , Enfermedad del Hígado Graso no Alcohólico , Tamaño de los Órganos , Oxidación-Reducción , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Ácido Tióctico/metabolismo
12.
Biomarkers ; 16(8): 670-8, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21999619

RESUMEN

The need for minimally invasive biomarkers to predict the progression of non-alcoholic fatty liver disease to non-alcoholic steatohepatitis is a priority. Oxidative stress and mitochondrial dysfunction contribute in this physiopathological process. The aim of this study was to analyze the potential role of erythrocytes as surrogate biomarkers of hepatic mitochondrial oxidative status in an animal model under different dietary oxidative conditions. Interestingly, we found that erythrocyte antioxidant status correlated with triglyceride content (p < 0.05-p < 0.001), thiobarbituric acid reactive species levels (p < 0.001) and with liver mitochondrial antioxidant levels (p < 0.001). These data suggest that erythrocyte antioxidant defenses could be used as sensitive and minimally invasive biomarkers of mitochondrial status in diverse oxidative conditions.


Asunto(s)
Biomarcadores/metabolismo , Eritrocitos/metabolismo , Mitocondrias Hepáticas/metabolismo , Estrés Oxidativo , Animales , Secuencia de Bases , Cartilla de ADN , Masculino , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa
13.
Redox Rep ; 15(5): 207-16, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21062536

RESUMEN

Modulating mitochondrial antioxidant status is a nutritional issue of great interest in the treatment or prevention of several oxidative stress related diseases such as obesity. Thus, the aim of the present study was to analyze the effects of three antioxidants on hepatic mitochondrial function and antioxidant status. Isolated rat liver mitochondria were incubated with vitamin C, resveratrol and lipoic acid. The activity of antioxidant enzymes (manganese superoxide dismutase and glutathione peroxidase), ROS generation and respiratory parameters (RCR, P/O ratio and respiratory states) were measured. Vitamin C influenced mitochondrial function by decreasing of ROS generation (P < 0.0001), by stimulating the activity of manganese superoxide dismutase (197.60 ± 35.99%; P < 0.001) as well as glutathione peroxidase (15.70 ± 5.76%; P < 0.05) and by altering the activity of the electron transport chain, mainly by decreasing the P/O ratio (P < 0.05). Resveratrol induced a significant increase in manganese superoxide dismutase activity (160 ± 11.78%; P < 0.0001) and a decrease in ROS generation (P < 0.05 to P < 0.0001). By contrast, lipoic acid inhibited glutathione peroxidase activity (16.48 ± 3.27%; P < 0.05) and induced the uncoupling of the electron transport chain (P < 0.01). Moreover, this antioxidant induced a strong decrease in the P/O ratio (P < 0.05 to P < 0.0001). In conclusion, our results suggest that the three tested antioxidants produced direct effects on mitochondrial function, although the magnitude and intensity of these actions were significantly different, which may have implications when administrated as antioxidants.


Asunto(s)
Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Mitocondrias Hepáticas/efectos de los fármacos , Estilbenos/farmacología , Ácido Tióctico/farmacología , Animales , Glutatión Peroxidasa/metabolismo , Peróxido de Hidrógeno/metabolismo , Masculino , Mitocondrias Hepáticas/metabolismo , Oxidantes/metabolismo , Estrés Oxidativo/efectos de los fármacos , Consumo de Oxígeno , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno/metabolismo , Resveratrol , Superóxido Dismutasa/metabolismo , Superóxidos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA