Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Free Radic Biol Med ; 160: 246-262, 2020 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-32791186

RESUMO

A growing body of evidences indicate the major role of extracellular vesicles (EVs) as players of cell communication in the pathogenesis of liver diseases. EVs are membrane-enclosed vesicles released by cells into the extracellular environment. Oxidative stress is also a key component of liver disease pathogenesis, but no role for hepatocyte-derived EVs has yet been described in the development of this process. Recently, some polycyclic aromatic hydrocarbons (PAHs), widespread environmental contaminants, were demonstrated to induce EV release from hepatocytes. They are also well-known to trigger oxidative stress leading to cell death. Therefore, the aim of this work was to investigate the involvement of EVs derived from PAHs-treated hepatocytes (PAH-EVs) in possible oxidative damages of healthy recipient hepatocytes, using both WIF-B9 and primary rat hepatocytes. We first showed that the release of EVs from PAHs -treated hepatocytes depended on oxidative stress. PAH-EVs were enriched in proteins related to oxidative stress such as NADPH oxidase and ferritin. They were also demonstrated to contain more iron. PAH-EVs could then induce oxidative stress in recipient hepatocytes, thereby leading to apoptosis. Mitochondria and lysosomes of recipient hepatocytes exhibited significant structural alterations. All those damages were dependent on internalization of EVs that reached lysosomes with their cargoes. Lysosomes thus appeared as critical organelles for EVs to induce apoptosis. In addition, pro-oxidant components of PAH-EVs, e.g. NADPH oxidase and iron, were revealed to be necessary for this cell death.


Assuntos
Vesículas Extracelulares , Hidrocarbonetos Policíclicos Aromáticos , Animais , Vesículas Extracelulares/metabolismo , Hepatócitos , Ferro/metabolismo , Estresse Oxidativo , Hidrocarbonetos Policíclicos Aromáticos/metabolismo , Hidrocarbonetos Policíclicos Aromáticos/toxicidade , Ratos
2.
Toxicol Sci ; 171(2): 443-462, 2019 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-31368503

RESUMO

Extracellular vesicles (EVs) are membrane-enclosed nanostructures released by cells into the extracellular environment. As major actors of physiological intercellular communication, they have been shown to be pathogenic mediators of several liver diseases. Extracellular vesicles also appear to be potential actors of drug-induced liver injury but nothing is known concerning environmental pollutants. We aimed to study the impact of polycyclic aromatic hydrocarbons (PAHs), major contaminants, on hepatocyte-derived EV production, with a special focus on hepatocyte death. Three PAHs were selected, based on their presence in food and their affinity for the aryl hydrocarbon receptor (AhR): benzo[a]pyrene (BP), dibenzo[a,h]anthracene (DBA), and pyrene (PYR). Treatment of primary rat and WIF-B9 hepatocytes by all 3 PAHs increased the release of EVs, mainly comprised of exosomes, in parallel with modifying exosome protein marker expression and inducing apoptosis. Moreover, PAH treatment of rodents for 3 months also led to increased EV levels in plasma. The EV release involved CYP metabolism and the activation of the transcription factor, the AhR, for BP and DBA and another transcription factor, the constitutive androstane receptor, for PYR. Furthermore, all PAHs increased cholesterol levels in EVs but only BP and DBA were able to reduce the cholesterol content of total cell membranes. All cholesterol changes very likely participated in the increase in EV release and cell death. Finally, we studied changes in cell membrane fluidity caused by BP and DBA due to cholesterol depletion. Our data showed increased cell membrane fluidity, which contributed to hepatocyte EV release and cell death.

3.
Free Radic Biol Med ; 129: 323-337, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30268890

RESUMO

We previously demonstrated that co-exposing pre-steatotic hepatocytes to benzo[a]pyrene (B[a]P), a carcinogenic environmental pollutant, and ethanol, favored cell death. Here, the intracellular mechanisms underlying this toxicity were studied. Steatotic WIF-B9 hepatocytes, obtained by a 48h-supplementation with fatty acids, were then exposed to B[a]P/ethanol (10 nM/5 mM, respectively) for 5 days. Nitric oxide (NO) was demonstrated to be a pivotal player in the cell death caused by the co-exposure in steatotic hepatocytes. Indeed, by scavenging NO, CPTIO treatment of co-exposed steatotic cells prevented not only the increase in DNA damage and cell death, but also the decrease in the activity of CYP1, major cytochrome P450s of B[a]P metabolism. This would then lead to an elevation of B[a]P levels, thus possibly suggesting a long-lasting stimulation of the transcription factor AhR. Besides, as NO can react with superoxide anion to produce peroxynitrite, a highly oxidative compound, the use of FeTPPS to inhibit its formation indicated its participation in DNA damage and cell death, further highlighting the important role of NO. Finally, a possible key role for AhR was pointed out by using its antagonist, CH-223191. Indeed it prevented the elevation of ADH activity, known to participate to the ethanol production of ROS, notably superoxide anion. The transcription factor, NFκB, known to be activated by ROS, was shown to be involved in the increase in iNOS expression. Altogether, these data strongly suggested cooperative mechanistic interactions between B[a]P via AhR and ethanol via ROS production, to favor cell death in the context of prior steatosis.


Assuntos
Benzo(a)pireno/toxicidade , Citocromo P-450 CYP1A1/genética , Etanol/toxicidade , Ácidos Graxos/farmacologia , Hepatócitos/efeitos dos fármacos , Óxido Nítrico/metabolismo , Álcool Desidrogenase/genética , Álcool Desidrogenase/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Compostos Azo/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Benzoatos/farmacologia , Linhagem Celular Tumoral , Quimera , Citocromo P-450 CYP1A1/antagonistas & inibidores , Citocromo P-450 CYP1A1/metabolismo , Dano ao DNA , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Hepatócitos/patologia , Imidazóis/farmacologia , Metaloporfirinas/farmacologia , NF-kappa B/genética , NF-kappa B/metabolismo , Necrose/induzido quimicamente , Necrose/genética , Necrose/metabolismo , Óxido Nítrico/agonistas , Pirazóis/farmacologia , Ratos , Receptores de Hidrocarboneto Arílico/genética , Receptores de Hidrocarboneto Arílico/metabolismo , Transdução de Sinais , Superóxidos/agonistas , Superóxidos/antagonistas & inibidores , Superóxidos/metabolismo
4.
Biomolecules ; 8(2)2018 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-29757947

RESUMO

The rise in prevalence of non-alcoholic fatty liver disease (NAFLD) constitutes an important public health concern worldwide. Including obesity, numerous risk factors of NAFLD such as benzo[a]pyrene (B[a]P) and ethanol have been identified as modifying the physicochemical properties of the plasma membrane in vitro thus causing membrane remodeling-changes in membrane fluidity and lipid-raft characteristics. In this study, the possible involvement of membrane remodeling in the in vivo progression of steatosis to a steatohepatitis-like state upon co-exposure to B[a]P and ethanol was tested in obese zebrafish larvae. Larvae bearing steatosis as the result of a high-fat diet were exposed to ethanol and/or B[a]P for seven days at low concentrations coherent with human exposure in order to elicit hepatotoxicity. In this condition, the toxicant co-exposure raised global membrane order with higher lipid-raft clustering in the plasma membrane of liver cells, as evaluated by staining with the fluoroprobe di-4-ANEPPDHQ. Involvement of this membrane's remodeling was finally explored by using the lipid-raft disruptor pravastatin that counteracted the effects of toxicant co-exposure both on membrane remodeling and toxicity. Overall, it can be concluded that B[a]P/ethanol co-exposure can induce in vivo hepatotoxicity via membrane remodeling which could be considered as a good target mechanism for developing combination therapy to deal with steatohepatitis.


Assuntos
Benzo(a)pireno/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Etanol/toxicidade , Fígado Gorduroso/metabolismo , Microdomínios da Membrana/efeitos dos fármacos , Animais , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Fígado Gorduroso/etiologia , Fígado Gorduroso/patologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Peixe-Zebra
5.
Sci Rep ; 8(1): 5963, 2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29654281

RESUMO

Hepatic steatosis (i.e. lipid accumulation) and steatohepatitis have been related to diverse etiologic factors, including alcohol, obesity, environmental pollutants. However, no study has so far analyzed how these different factors might interplay regarding the progression of liver diseases. The impact of the co-exposure to the environmental carcinogen benzo[a]pyrene (B[a]P) and the lifestyle-related hepatotoxicant ethanol, was thus tested on in vitro models of steatosis (human HepaRG cell line; hybrid human/rat WIF-B9 cell line), and on an in vivo model (obese zebrafish larvae). Steatosis was induced prior to chronic treatments (14, 5 or 7 days for HepaRG, WIF-B9 or zebrafish, respectively). Toxicity and inflammation were analyzed in all models; the impact of steatosis and ethanol towards B[a]P metabolism was studied in HepaRG cells. Cytotoxicity and expression of inflammation markers upon co-exposure were increased in all steatotic models, compared to non steatotic counterparts. A change of B[a]P metabolism with a decrease in detoxification was detected in HepaRG cells under these conditions. A prior steatosis therefore enhanced the toxicity of B[a]P/ethanol co-exposure in vitro and in vivo; such a co-exposure might favor the appearance of a steatohepatitis-like state, with the development of inflammation. These deleterious effects could be partly explained by B[a]P metabolism alterations.


Assuntos
Benzo(a)pireno/efeitos adversos , Etanol/efeitos adversos , Fígado Gorduroso/patologia , Fígado/patologia , Animais , Biomarcadores/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Progressão da Doença , Poluentes Ambientais/efeitos adversos , Fígado Gorduroso/metabolismo , Humanos , Inflamação/metabolismo , Inflamação/patologia , Larva/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/metabolismo , Ratos , Peixe-Zebra
6.
Sci Rep ; 7(1): 195, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28298645

RESUMO

Most tumors undergo metabolic reprogramming towards glycolysis, the so-called Warburg effect, to support growth and survival. Overexpression of IF1, the physiological inhibitor of the F0F1ATPase, has been related to this phenomenon and appears to be a relevant marker in cancer. Environmental contributions to cancer development are now widely accepted but little is known about the underlying intracellular mechanisms. Among the environmental pollutants humans are commonly exposed to, benzo[a]pyrene (B[a]P), the prototype molecule of polycyclic aromatic hydrocarbons (PAHs), is a well-known human carcinogen. Besides apoptotic signals, B[a]P can also induce survival signals in liver cells, both likely involved in cancer promotion. Our previous works showed that B[a]P elicited a Warburg-like effect, thus favoring cell survival. The present study aimed at further elucidating the molecular mechanisms involved in the B[a]P-induced metabolic reprogramming, by testing the possible involvement of IF1. We presently demonstrate, both in vitro and in vivo, that PAHs, especially B[a]P, strongly increase IF1 expression. Such an increase, which might rely on ß2-adrenergic receptor activation, notably participates to the B[a]P-induced glycolytic shift and cell survival in liver cells. By identifying IF1 as a target of PAHs, this study provides new insights about how environmental factors may contribute to related carcinogenesis.


Assuntos
Carcinógenos Ambientais/toxicidade , Carcinoma Hepatocelular/genética , Glicólise , Neoplasias Hepáticas/genética , Hidrocarbonetos Policíclicos Aromáticos/toxicidade , Proteínas/genética , Animais , Apoptose , Benzo(a)pireno/toxicidade , Carcinoma Hepatocelular/induzido quimicamente , Carcinoma Hepatocelular/metabolismo , Linhagem Celular , Sobrevivência Celular , Progressão da Doença , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Hepáticas/induzido quimicamente , Neoplasias Hepáticas/metabolismo , Neoplasias Experimentais , Proteínas/metabolismo , Ratos , Receptores Adrenérgicos beta 2/genética , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima , Proteína Inibidora de ATPase
7.
J Appl Toxicol ; 37(6): 732-746, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27896850

RESUMO

The easy-to-use in vivo model, zebrafish larva, is being increasingly used to screen chemical-induced hepatotoxicity, with a good predictivity for various mechanisms of liver injury. However, nothing is known about its applicability in exploring the mechanism called membrane remodeling, depicted as changes in membrane fluidity or lipid raft properties. The aim of this study was, therefore, to substantiate the zebrafish larva as a suitable in vivo model in this context. Ethanol was chosen as a prototype toxicant because it is largely described, both in hepatocyte cultures and in rodents, as capable of inducing a membrane remodeling leading to hepatocyte death and liver injury. The zebrafish larva model was demonstrated to be fully relevant as membrane remodeling was maintained even after a 1-week exposure without any adaptation as usually reported in rodents and hepatocyte cultures. It was also proven to exhibit a high sensitivity as it discriminated various levels of cytotoxicity depending on the extent of changes in membrane remodeling. In this context, its sensitivity appeared higher than that of WIF-B9 hepatic cells, which is suited for analyzing this kind of hepatotoxicity. Finally, the protection afforded by a membrane stabilizer, ursodeoxycholic acid (UDCA), or by a lipid raft disrupter, pravastatin, definitely validated zebrafish larva as a reliable model to quickly assess membrane remodeling involvement in chemical-induced hepatotoxicity. In conclusion, this model, compatible with a high throughput screening, might be adapted to seek hepatotoxicants via membrane remodeling, and also drugs targeting membrane features to propose new preventive or therapeutic strategies in chemical-induced liver diseases. Copyright © 2016 John Wiley & Sons, Ltd.


Assuntos
Larva/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fluidez de Membrana/efeitos dos fármacos , Microdomínios da Membrana/efeitos dos fármacos , Modelos Biológicos , Peixe-Zebra , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Etanol/toxicidade , Humanos , Células Híbridas , Larva/metabolismo , Fígado/metabolismo , Fígado/patologia , Microdomínios da Membrana/patologia , Microscopia de Fluorescência , Estresse Oxidativo/efeitos dos fármacos , Pravastatina/farmacologia , Ratos , Ácido Ursodesoxicólico/farmacologia
8.
Free Radic Biol Med ; 72: 11-22, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24681337

RESUMO

Several epidemiologic studies have shown an interactive effect of heavy smoking and heavy alcohol drinking on the development of hepatocellular carcinoma. It has also been recently described that chronic hepatocyte death can trigger excessive compensatory proliferation resulting later in the formation of tumors in mouse liver. As we previously demonstrated that both benzo[a]pyrene (B[a]P), an environmental agent found in cigarette smoke, and ethanol possess similar targets, especially oxidative stress, to trigger death of liver cells, we decided to study here the cellular and molecular mechanisms of the effects of B[a]P/ethanol coexposure on cell death. After an 18-h incubation with 100nM B[a]P, primary rat hepatocytes were supplemented with 50mM ethanol for 5 or 8h. B[a]P/ethanol coexposure led to a greater apoptotic cell death that could be linked to an increase in lipid peroxidation. Plasma membrane remodeling, as depicted by membrane fluidity elevation and physicochemical alterations in lipid rafts, appeared to play a key role, because both toxicants acted with specific complementary effects. Membrane remodeling was shown to induce an accumulation of lysosomes leading to an important increase in low-molecular-weight iron cellular content. Finally, ethanol metabolism, but not that of B[a]P, by providing reactive oxygen species, induced the ultimate toxic process. Indeed, in lysosomes, ethanol promoted the Fenton reaction, lipid peroxidation, and membrane permeabilization, thereby triggering cell death. To conclude, B[a]P exposure, by depleting hepatocyte membrane cholesterol content, would constitute a favorable ground for a later toxic insult such as ethanol intoxication. Membrane stabilization of both plasma membrane and lysosomes might be a potential target for further investigation considering cytoprotective strategies.


Assuntos
Benzo(a)pireno/toxicidade , Membrana Celular/efeitos dos fármacos , Etanol/toxicidade , Hepatócitos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Carcinógenos/toxicidade , Depressores do Sistema Nervoso Central/toxicidade , Hepatócitos/patologia , Peroxidação de Lipídeos/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Microscopia Eletrônica de Transmissão , Ratos , Ratos Sprague-Dawley
9.
Chem Biol Interact ; 207: 41-51, 2014 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-24246761

RESUMO

Plasma membrane is an early target of polycyclic aromatic hydrocarbons (PAH). We previously showed that the PAH prototype, benzo[a]pyrene (B[a]P), triggers apoptosis via DNA damage-induced p53 activation (genotoxic pathway) and via remodeling of the membrane cholesterol-rich microdomains called lipid rafts, leading to changes in pH homeostasis (non-genotoxic pathway). As omega-3 (n-3) fatty acids can affect membrane composition and function or hamper in vivo PAH genotoxicity, we hypothesized that addition of physiologically relevant levels of polyunsaturated n-3 fatty acids (PUFAs) might interfere with B[a]P-induced toxicity. The effects of two major PUFAs, docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), were tested on B[a]P cytotoxicity in the liver epithelial cell line F258. Both PUFAs reduced B[a]P-induced apoptosis. Surprisingly, pre-treatment with DHA increased the formation of reactive B[a]P metabolites, resulting in higher levels of B[a]P-DNA adducts. EPA had no apparent effect on B[a]P metabolism or related DNA damage. EPA and DHA prevented B[a]P-induced apoptotic alkalinization by affecting Na(+)/H(+) exchanger 1 activity. Thus, the inhibitory effects of omega-3 fatty acids on B[a]P-induced apoptosis involve a non-genotoxic pathway associated with plasma membrane remodeling. Our results suggest that dietary omega-3 fatty acids may have marked effects on the biological consequences of PAH exposure.


Assuntos
Apoptose/efeitos dos fármacos , Membrana Celular/metabolismo , Ácidos Graxos Ômega-3/farmacologia , Substâncias Protetoras/farmacologia , Transdução de Sinais/efeitos dos fármacos , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Benzo(a)pireno , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Colesterol/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Dano ao DNA , Ácidos Docosa-Hexaenoicos/farmacologia , Ácido Eicosapentaenoico/farmacologia , Concentração de Íons de Hidrogênio/efeitos dos fármacos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Lipídeos/química , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Modelos Biológicos , Transporte Proteico/efeitos dos fármacos , Ratos , Trocador 1 de Sódio-Hidrogênio , Proteína Supressora de Tumor p53/metabolismo
10.
Food Chem Toxicol ; 60: 286-96, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23907024

RESUMO

Previously, we demonstrated that eicosapentaenoic acid enhanced ethanol-induced oxidative stress and cell death in primary rat hepatocytes via an increase in membrane fluidity and lipid raft clustering. In this context, another n-3 polyunsaturated fatty acid, docosahexaenoic acid (DHA), was tested with a special emphasis on physical and chemical alteration of lipid rafts. Pretreatment of hepatocytes with DHA reduced significantly ethanol-induced oxidative stress and cell death. DHA protection could be related to an alteration of lipid rafts. Indeed, rafts exhibited a marked increase in membrane fluidity and packing defects leading to the exclusion of a raft protein marker, flotillin. Furthermore, DHA strongly inhibited disulfide bridge formation, even in control cells, thus suggesting a disruption of protein-protein interactions inside lipid rafts. This particular spatial organization of lipid rafts due to DHA subsequently prevented the ethanol-induced lipid raft clustering. Such a prevention was then responsible for the inhibition of phospholipase C-γ translocation into rafts, and consequently of both lysosome accumulation and elevation in cellular low-molecular-weight iron content, a prooxidant factor. In total, the present study suggests that DHA supplementation could represent a new preventive approach for patients with alcoholic liver disease based upon modulation of the membrane structures.


Assuntos
Ácidos Docosa-Hexaenoicos/farmacologia , Etanol/toxicidade , Hepatócitos/efeitos dos fármacos , Microdomínios da Membrana/efeitos dos fármacos , Animais , Morte Celular/efeitos dos fármacos , Células Cultivadas , Glutationa/metabolismo , Glutationa Peroxidase/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Proteínas de Membrana/metabolismo , Peso Molecular , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Fosfolipases Tipo C/antagonistas & inibidores , Fosfolipases Tipo C/metabolismo
11.
Free Radic Biol Med ; 51(11): 2018-30, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21945097

RESUMO

Dietary n-3 polyunsaturated fatty acids (n-3 PUFAs) have been reported to modulate lipid raft-dependent signaling, but not yet lipid raft-dependent oxidative stress. Previously, we have shown that ethanol-induced membrane remodeling, i.e., an increase in membrane fluidity and alterations in physical and biochemical properties of lipid rafts, participated in the development of oxidative stress. Thus, we decided to study n-3 PUFA effects in this context, by pretreating hepatocytes with eicosapentaenoic acid (EPA), a long-chain n-3 PUFA, before addition of ethanol. EPA was found to increase ethanol-induced oxidative stress through membrane remodeling. Addition of EPA resulted in a marked increase in lipid raft aggregation compared to ethanol alone. In addition, membrane fluidity of lipid rafts was markedly enhanced. Interestingly, EPA was found to preferentially incorporate into nonraft membrane regions, leading to raft cholesterol increase. Lipid raft aggregation by EPA enhanced phospholipase Cγ translocation into these microdomains. Finally, phospholipase Cγ was shown to participate in the potentiation of oxidative stress by promoting lysosome accumulation, a major source of low-molecular-weight iron. To conclude, the ability of EPA to modify lipid raft physical and chemical properties plays a key role in the enhancement, by this dietary n-3 PUFA, of ethanol-induced oxidative stress.


Assuntos
Gorduras Insaturadas na Dieta/farmacologia , Ácido Eicosapentaenoico/farmacologia , Etanol/farmacologia , Microdomínios da Membrana/química , Microdomínios da Membrana/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Animais , Morte Celular/efeitos dos fármacos , Células Cultivadas , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Ratos , Ratos Sprague-Dawley
12.
Hepatology ; 47(1): 59-70, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18038449

RESUMO

UNLABELLED: The role of the hepatocyte plasma membrane structure in the development of oxidative stress during alcoholic liver diseases is not yet fully understood. Previously, we have established the pivotal role of membrane fluidity in ethanol-induced oxidative stress, but no study has so far tested the involvement of lipid rafts. In this study, methyl-beta-cyclodextrin or cholesterol oxidase, which were found to disrupt lipid rafts in hepatocytes, inhibited both reactive oxygen species production and lipid peroxidation, and this suggested a role for these microstructures in oxidative stress. By immunostaining of lipid raft components, a raft clustering was detected in ethanol-treated hepatocytes. In addition, we found that rafts were modified by formation of malondialdehyde adducts and disulfide bridges. Interestingly, pretreatment of cells by 4-methyl-pyrazole (to inhibit ethanol metabolism) and various antioxidants prevented the ethanol-induced raft aggregation. In addition, treatment of hepatocytes by a stabilizing agent (ursodeoxycholic acid) or a fluidizing compound [2-(2-methoxyethoxy)ethyl 8-(cis-2-n-octylcyclopropyl)octanoate] led to inhibition or enhancement of raft clustering, respectively, which pointed to a relationship between membrane fluidity and lipid rafts during ethanol-induced oxidative stress. We finally investigated the involvement of phospholipase C in raft-induced oxidative stress upon ethanol exposure. Phospholipase C was shown to be translocated into rafts and to participate in oxidative stress by controlling hepatocyte iron content. CONCLUSION: Membrane structure, depicted as membrane fluidity and lipid rafts, plays a key role in ethanol-induced oxidative stress of the liver, and its modulation may be of therapeutic relevance.


Assuntos
Etanol/efeitos adversos , Hepatócitos/metabolismo , Microdomínios da Membrana/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Colesterol Oxidase/farmacologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/enzimologia , Fluidez de Membrana/efeitos dos fármacos , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/enzimologia , Fosfoinositídeo Fosfolipase C/metabolismo , Ratos , Ratos Sprague-Dawley , beta-Ciclodextrinas/farmacologia
13.
Cancer Res ; 67(16): 7865-74, 2007 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-17699793

RESUMO

We have previously shown that cisplatin triggers an early acid sphingomyelinase (aSMase)-dependent ceramide generation concomitantly with an increase in membrane fluidity and induces apoptosis in HT29 cells. The present study further explores the role and origin of membrane fluidification in cisplatin-induced apoptosis. The rapid increase in membrane fluidity following cisplatin treatment was inhibited by membrane-stabilizing agents such as cholesterol or monosialoganglioside-1. In HT29 cells, these compounds prevented the early aggregation of Fas death receptor and of membrane lipid rafts on cell surface and significantly inhibited cisplatin-induced apoptosis without altering drug intracellular uptake or cisplatin DNA adducts formation. Early after cisplatin treatment, Na+/H+ membrane exchanger-1 (NHE1) was inhibited leading to intracellular acidification, aSMase was activated, and ceramide was detected at the cell membrane. Treatment of HT29 cells with Staphylococcus aureus sphingomyelinase increased membrane fluidity. Moreover, pretreatment with cariporide, a specific inhibitor of NHE1, inhibited cisplatin-induced intracellular acidification, aSMase activation, ceramide membrane generation, membrane fluidification, and apoptosis. Finally, NHE1-expressing PS120 cells were more sensitive to cisplatin than NHE1-deficient PS120 cells. Altogether, these findings suggest that the apoptotic pathway triggered by cisplatin involves a very early NHE1-dependent intracellular acidification leading to aSMase activation and increase in membrane fluidity. These events are independent of cisplatin-induced DNA adducts formation. The membrane exchanger NHE1 may be another potential target of cisplatin, increasing cell sensitivity to this compound.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Proteínas de Transporte de Cátions/antagonistas & inibidores , Cisplatino/farmacologia , Neoplasias do Colo/tratamento farmacológico , Fluidez de Membrana/efeitos dos fármacos , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Proteínas de Transporte de Cátions/metabolismo , Colesterol/farmacologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Interações Medicamentosas , Guanidinas/farmacologia , Células HCT116 , Células HT29 , Humanos , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Trocador 1 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/metabolismo , Sulfonas/farmacologia
14.
Ann N Y Acad Sci ; 1090: 108-12, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17384252

RESUMO

Polycyclic aromatic hydrocarbons (PAHs) such as benzo[a]yrene (B[a]P) constitute a widely distributed class of environmental pollutants, responsible for highly toxic effects. Elucidating the intracellular mechanisms of this cytotoxicity thus remains a major challenge. Besides the activation of the p53 apoptotic pathway, we have previously found in F258 hepatic cells that the B[a]P (50 nM)-induced apoptosis was also dependent upon the transmembrane transporter NHE1, whose activation might result from membrane alterations in our model. We here demonstrate that: (1) B[a]P induces a membrane fluidization surprisingly linked to NHE1 activation; (2) membrane stabilization by exogenous cholesterol protects cells from B[a]P-induced apoptosis, via an effect on late acidification and iron uptake.


Assuntos
Apoptose/efeitos dos fármacos , Benzo(a)pireno/farmacologia , Fluidez de Membrana , Animais , Linhagem Celular , Ratos
15.
J Pharmacol Exp Ther ; 313(1): 104-11, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15634942

RESUMO

The relationship between bulk membrane fluidizing effect of ethanol and its toxicity due to oxidative stress is still unknown. To elucidate this issue, membrane fluidity of primary rat hepatocytes was studied by measuring order parameter after inhibition of ethanol-induced oxidative stress. We showed that pretreating cells with either 4-methyl-pyrazole (to inhibit ethanol metabolism), thiourea [a reactive oxygen species (ROS) scavenger], or vitamin E (a free radical chain-breaking antioxidant) prevented the ethanol-induced increase in membrane fluidity, thus suggesting that ethanol metabolism and ROS formation were involved in this elevation. The effects of membrane stabilizing agents (ursodeoxycholic acid or ganglioside GM1), shown to prevent fluidification, next pointed to a role for this increase in membrane fluidity in the development of ethanol-induced oxidative stress. Indeed, ROS production, lipid peroxidation, and cell death were all inhibited by these agents. In contrast, the fluidizing compounds Tween 20 or 2-(2-methoxyethoxy) ethyl 8-(cis-2-n-octylcyclopropyl) octanoate, which increased the membrane fluidizing effect of ethanol, enhanced the related oxidative stress. Using electron paramagnetic resonance to determine low molecular weight iron, we finally demonstrated that membrane fluidity influence proceeded through an increase in low molecular weight iron to enhance oxidative stress. In conclusion, the present findings clearly highlight the pivotal role of membrane fluidity in ethanol-induced oxidative stress and the potential therapeutic effect of membrane stabilizing compounds.


Assuntos
Depressores do Sistema Nervoso Central/toxicidade , Etanol/toxicidade , Hepatócitos/efeitos dos fármacos , Fluidez de Membrana/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Morte Celular/efeitos dos fármacos , Separação Celular , Proteínas de Fluorescência Verde/metabolismo , Ferro/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Fluidez de Membrana/efeitos dos fármacos , Peso Molecular , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio
16.
Free Radic Biol Med ; 34(10): 1283-94, 2003 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-12726916

RESUMO

Various drugs and chemicals can cause a glutathione (GSH) depletion in the liver. Moreover, nitric oxide (NO) can be generated in response to physiological and pathological situations such as inflammation. The aim of this study was to estimate oxidative stress when primary rat hepatocytes were exposed to GSH depletion after NO production. For this purpose, cells were preincubated with lipopolysaccharide (LPS) and gamma-interferon (IFN) for 18 h in order to induce NO production by NO synthase and then L-buthionine sulfoximine (BSO), an inhibitor of GSH synthesis, was added for 5 h. In hepatocyte cultures preincubated with LPS and IFN before BSO addition, an increase in lipid peroxidation was noted. In those cells, an elevation of iron-bound NO and a decrease in free NO led us to suggest the involvement of low-molecular-weight iron (LMW iron) in the enhancement of oxidative stress. Indeed, addition of deferiprone, a chelator of LMW iron, reduced iron-bound NO levels and the extent of oxidative stress. Moreover, an important elevation of LMW iron levels was also observed. As both, N-acetylcysteine, a GSH precursor, and N(G)-monomethyl-L-arginine, a NO synthase inhibitor, totally inhibited the elevation of LMW iron and oxidative stress, a cooperative role could be attributed to NO production and GSH depletion.


Assuntos
Butionina Sulfoximina/farmacologia , Glutationa/deficiência , Hepatócitos/efeitos dos fármacos , Ferro/metabolismo , Óxido Nítrico/biossíntese , Estresse Oxidativo , Acetilcisteína/farmacologia , Animais , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Deferiprona , Ditiocarb/farmacologia , Espectroscopia de Ressonância de Spin Eletrônica , Inibidores Enzimáticos/farmacologia , Hepatócitos/metabolismo , Interferon gama/farmacologia , Quelantes de Ferro/farmacologia , Peroxidação de Lipídeos/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Óxido Nítrico Sintase/metabolismo , Nitritos/metabolismo , Nitritos/farmacologia , Piridonas/farmacologia , Ratos , Ratos Sprague-Dawley , ômega-N-Metilarginina/farmacologia
17.
Can J Appl Physiol ; 28(1): 79-92, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12671197

RESUMO

We tested seven physical education students whether 30-s sprint anaerobic exercise (Wingate test) would result in oxidative stress (evaluated by lipid radical levels) sufficient to alter plasma non-enzymatic antioxidant status (plasma uric acid, ascorbic acid, alpha-tocopherol, beta-carotene). This study demonstrates that 1) Wingate test increases plasma uric and ascorbic acid concentrations (p <.05), and decreases plasma alpha-tocopherol and beta-carotene levels (p <.05); 2) lipid radical levels at rest and sprint performance are negatively correlated with resting plasma uric acid and alpha-tocopherol concentrations (p <.05). In conclusion, this study 1) demonstrates that a 30-s sprint anaerobic exercise is associated with acute changes in plasma non-enzymatic antioxidant status, 2) indicates that the subjects with largest leg peak power are those who exhibit the lowest plasma antioxidant status at rest (uric acid and alpha-tocopherol), 3) and suggests that antioxidant intake by maintaining plasma antioxidant concentration at rest in the normal range might protect athletes against oxidative stress induced by exercise.


Assuntos
Antioxidantes/metabolismo , Estresse Oxidativo , Aptidão Física , Adulto , Análise de Variância , Ácido Ascórbico/sangue , Ciclismo/fisiologia , Teste de Esforço , Humanos , Lactatos/sangue , Masculino , Ácido Úrico/sangue , alfa-Tocoferol/sangue , beta Caroteno/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA