RESUMO
The complexity of hepatocellular carcinoma (HCC) signaling and the failure of pharmacological therapeutics reveal the significance of establishing new anti-cancer strategies. Interferon alpha (IFN-α) has been used as adjuvant therapy for reducing HCC recurrence and improving survival. Delta-tocotrienol (δ-tocotrienol), a natural unsaturated isoform of vitamin E, is a promising candidate for cancer treatment. In this study, we evaluated whether the combination of δ-tocotrienol with IFN-α displays significant advantages in the treatment of HCC cells. Results showed that the combination significantly decreased cell viability, migration and invasion of HCC cells compared with single therapies. Combining δ-tocotrienol and IFN-α enhanced the decrease in proliferating cell nuclear antigen (PCNA) and matrix metalloproteinase (MMP) 7 and MMP-9. The combination also produced an enhancement of apoptosis together with increased Bax/Bcl-xL ratio and reactive oxygen species (ROS) generation. δ-tocotrienol induced Notch1 activation and changes in Erk and p38 MAPK signaling status. Blocking experiments confirmed that ROS and Erk are involved, at least in part, in the anti-cancer effects of the combined treatment. In conclusion, the combination of δ-tocotrienol with IFN-α therapy showed promising results for HCC cell treatment, which makes the combination of cytokine-based immunotherapy with natural products a potential strategy against liver cancer.
Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Apoptose , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Humanos , Interferon-alfa/farmacologia , Interferon-alfa/uso terapêutico , Neoplasias Hepáticas/patologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Vitamina E/análogos & derivados , Vitamina E/farmacologia , Vitamina E/uso terapêuticoRESUMO
IFN-α administration to patients has been long discouraged and pushed back by new and apparently better drugs; however the adverse secondary effect, the high costs and the lack of specific action, make these new drugs hard to be used and put IFN-α again in the eye of the researchers. IFN-α-2b was demonstrated to induce apoptosis and modulation of lipid metabolism and the mechanisms are still unknown. Here, we sought to find the link between these features using a model of early stage cancer development. Using in vitro and in vivo approaches, we evaluated apoptosis and lipid metabolism. IFN-α-2b induced changes in hepatic cholesterol mass due to decreased synthesis and increased secretion. Interestingly, the drop in cellular cholesterol levels was necessary for IFN-α-2b to induce apoptosis. Results presented in this paper show the complexity of the action of IFN-α-2b on the early stages of liver cancer development. We show for the first time an interrelationship between cholesterol, apoptosis and IFN-α-2b. This makes clear the need for a reevaluation of IFN-α-2b action in order to develop softer, safer and more bearable derivatives. In this regard, knowing the molecular mechanisms by which IFN-α exerts its cellular actions is of crucial importance, and it is the main condition for therapy success for classical and new malignancies.
Assuntos
Apoptose/efeitos dos fármacos , Colesterol/metabolismo , Hepatócitos/efeitos dos fármacos , Interferon alfa-2/farmacologia , Animais , Linhagem Celular Tumoral , Hepatócitos/metabolismo , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Neoplasias Hepáticas/metabolismo , Masculino , Ratos , Ratos WistarRESUMO
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease. Triacylglycerol accumulation in the liver is a hallmark of NAFLD. Metabolic studies have confirmed that increased hepatic de novo lipogenesis (DNL) in humans contributes to fat accumulation in the liver and to NAFLD progression. Mice deficient in carboxylesterase (Ces)1d expression are protected from high-fat diet-induced hepatic steatosis. To investigate whether loss of Ces1d can also mitigate steatosis induced by over-activated DNL, WT and Ces1d-deficient mice were fed a lipogenic high-sucrose diet (HSD). We found that Ces1d-deficient mice were protected from HSD-induced hepatic lipid accumulation. Mechanistically, Ces1d deficiency leads to activation of AMP-activated protein kinase and inhibitory phosphorylation of acetyl-CoA carboxylase. Together with our previous demonstration that Ces1d deficiency attenuated high-fat diet-induced steatosis, this study suggests that inhibition of CES1 (the human ortholog of Ces1d) might represent a novel pharmacological target for prevention and treatment of NAFLD.
Assuntos
Carboxilesterase/metabolismo , Dieta Hiperlipídica/efeitos adversos , Fígado/metabolismo , Sacarose/antagonistas & inibidores , Triglicerídeos/metabolismo , Animais , Carboxilesterase/deficiência , Fígado/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Sacarose/administração & dosagem , Sacarose/efeitos adversosRESUMO
IFN-α is used for inflammatory purposes, and obesity and NAFLD are strongly correlated with inflammatory processes. We wondered whether IFN-α-2b can attenuate obesity development and its associated NAFLD in mice fed high fat diet (HFD) for 10â¯weeks. IFN-α-2b had a robust effect on body weight loss associated with NAFLD amelioration by decreasing hepatic inflammation. IFN-α-2b-treated mice showed increased plasma cholesterol levels together with decreased hepatic cholesterol, both on chow and HF diets. Interestingly, mice on IFN-α-2b treatment secreted smaller VLDL particles highly enriched in cholesterol. Mechanistically, we found that IFN-α-2b antiobesity effects were related to increased fatty acid oxidation; and its effects on cholesterol metabolism were due to both a decrease in the master cholesterogenic transcription factor SREBP-2 and in the rate limiting enzyme in cholesterol synthesis, HMGCR. To our knowledge, this is the first report showing the effects of IFN-α-2b on the prevention of the development of HFD-induced body weight gain and dyslipidemia through a mechanism that involves fatty acid oxidation and cholesterol decrease. These studies comprise necessary steps for understanding the amelioration of obesity and NAFLD. Results shed some light into the mechanism of action of natural cytokines, and their effects on ameliorating obesity and its related diseases.
Assuntos
Interferon alfa-2/uso terapêutico , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Obesidade/prevenção & controle , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Dislipidemias/tratamento farmacológico , Ácidos Graxos/metabolismo , Lipídeos/sangue , Lipoproteínas/sangue , Lipoproteínas VLDL/sangue , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/patologia , Hipernutrição/complicações , Reação em Cadeia da Polimerase em Tempo RealRESUMO
PURPOSE: Glycerol usage is increasing in food industry for human and animal nutrition. This study analyzed the impact of glycerol metabolism when orally supplemented during the early stage of rat liver carcinogenesis. METHODS: Wistar rats were subjected to a 2-phase model of hepatocarcinogenesis (initiated-promoted, IP group). IP animals also received glycerol by gavage (200 mg/kg body weight, IPGly group). RESULTS: Glycerol treatment reduced the volume of preneoplastic lesions by decreasing the proliferative status of liver foci, increasing the expression of p53 and p21 proteins and reducing the expression of cyclin D1 and cyclin-dependent kinase 1. Besides, apoptosis was enhanced in IPGly animals, given by an increment of Bax/Bcl-2 ratio, Bad and PUMA mitochondrial expression, a concomitant increase in cytochrome c release and caspase-3 activation. Furthermore, hepatic levels of glycerol phosphate and markers of oxidative stress were increased in IPGly rats. Oxidative stress intermediates act as intracellular messengers, inducing p53 activation and changes in JNK and Erk signaling pathways, with JNK activation and Erk inhibition. CONCLUSION: The present work provides novel data concerning the preventive actions of glycerol during the development of liver cancer and represents an economically feasible intervention to treat high-risk individuals.
Assuntos
Anticarcinógenos/uso terapêutico , Apoptose , Suplementos Nutricionais , Glicerol/uso terapêutico , Neoplasias Hepáticas Experimentais/prevenção & controle , Estresse Oxidativo , Lesões Pré-Cancerosas/prevenção & controle , Animais , Anticarcinógenos/sangue , Anticarcinógenos/metabolismo , Biomarcadores/sangue , Carcinogênese , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Glicerol/sangue , Glicerol/metabolismo , Peroxidação de Lipídeos , Fígado/enzimologia , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas Experimentais/sangue , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/patologia , Sistema de Sinalização das MAP Quinases , Masculino , Mitocôndrias Hepáticas/enzimologia , Mitocôndrias Hepáticas/metabolismo , Mitocôndrias Hepáticas/patologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Fosforilação , Lesões Pré-Cancerosas/sangue , Lesões Pré-Cancerosas/metabolismo , Lesões Pré-Cancerosas/patologia , Ratos Wistar , Carga TumoralRESUMO
It is accepted that cancer development is associated with metabolic changes. Previously, we established a model of hepatic preneoplasia in which adult rats were subjected to a 2-phase model of hepatocarcinogenesis (initiated-promoted, IP) for 6weeks until they develop altered hepatic foci (AHF). Here, we found that a whole metabolic shift occurs in order to favor cancer development. IP animals presented with increased plasma lipids due to increased VLDL secretion as well as increased liver lipid accretion due to stimulated transacetylase activity rather than lipogenesis, compared to control rats. We found that carboxylesterase 3/triacylglycerol hydrolase (Ces3/Tgh) presented with a perilobular distribution surrounding lipid droplets in normal livers. However, it is downregulated both at the protein and mRNA level in liver homogenates and is almost undetectable inside the AHF with no changes in the surrounding tissue. Ces3/Tgh expression is regulated by ω-3 fatty acids, thus, supplementation of diet with fish oil, allowed the restoration of Ces3/Tgh expression inside the foci and, more interestingly, led to the decrease in number and volume of the AHF. These studies show a preventive role of Ces3/Tgh in liver cancer development.
RESUMO
Phosphatidylethanolamine N-methyltransferase (PEMT) is an important enzyme in hepatic phosphatidylcholine (PC) biosynthesis. Pemt(-/-) mice are protected against high-fat diet (HFD)-induced obesity and insulin resistance; however, these mice develop nonalcoholic fatty liver disease (NAFLD). We hypothesized that peroxisomal proliferator-activated receptor-γ (PPARγ) activation by pioglitazone might stimulate adipocyte proliferation, thereby directing lipids from the liver toward white adipose tissue. Pioglitazone might also act directly on PPARγ in the liver to improve NAFLD. Pemt(+/+) and Pemt(-/-) mice were fed a HFD with or without pioglitazone (20 mg·kg(-1)·day(-1)) for 10 wk. Pemt(-/-) mice were protected from HFD-induced obesity but developed NAFLD. Treatment with pioglitazone caused an increase in body weight gain in Pemt(-/-) mice that was mainly due to increased adiposity. Moreover, pioglitazone improved NAFLD in Pemt(-/-) mice, as indicated by a 35% reduction in liver weight and a 57% decrease in plasma alanine transaminase levels. Livers from HFD-fed Pemt(-/-) mice were steatotic, inflamed, and fibrotic. Hepatic steatosis was still evident in pioglitazone-treated Pemt(-/-) mice; however, treatment with pioglitazone reduced hepatic fibrosis, as evidenced by reduced Sirius red staining and lowered mRNA levels of collagen type Iα1 (Col1a1), tissue inhibitor of metalloproteinases 1 (Timp1), α-smooth muscle actin (Acta2), and transforming growth factor-ß (Tgf-ß). Similarly, oxidative stress and inflammation were reduced in livers from Pemt(-/-) mice upon treatment with pioglitazone. Together, these data show that activation of PPARγ in HFD-fed Pemt(-/-) mice improved liver function, while these mice were still protected against diet-induced obesity and insulin resistance.
Assuntos
Anti-Infecciosos/farmacologia , Hepatite/prevenção & controle , Cirrose Hepática Experimental/prevenção & controle , Fígado/efeitos dos fármacos , Hepatopatia Gordurosa não Alcoólica/prevenção & controle , PPAR gama/agonistas , Fosfatidiletanolamina N-Metiltransferase/deficiência , Tiazolidinedionas/farmacologia , Actinas/genética , Actinas/metabolismo , Adipócitos Brancos/efeitos dos fármacos , Adipócitos Brancos/enzimologia , Adipócitos Brancos/patologia , Tecido Adiposo Branco/efeitos dos fármacos , Tecido Adiposo Branco/enzimologia , Tecido Adiposo Branco/patologia , Adiposidade/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Dieta Hiperlipídica , Predisposição Genética para Doença , Hepatite/enzimologia , Hepatite/genética , Hepatite/patologia , Resistência à Insulina , Fígado/enzimologia , Fígado/patologia , Cirrose Hepática Experimental/enzimologia , Cirrose Hepática Experimental/genética , Cirrose Hepática Experimental/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/enzimologia , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/patologia , Obesidade/enzimologia , Obesidade/genética , Obesidade/prevenção & controle , Estresse Oxidativo/efeitos dos fármacos , PPAR gama/metabolismo , Fenótipo , Fosfatidiletanolamina N-Metiltransferase/genética , Pioglitazona , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Inibidor Tecidual de Metaloproteinase-1/genética , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismoRESUMO
Hepatic triacylglycerol levels are governed through synthesis, degradation and export of this lipid. Here we demonstrate that enforced expression of hepatic lipase in the endoplasmic reticulum in McArdle RH7777 hepatocytes resulted in a significant decrease in the incorporation of fatty acids into cellular triacylglycerol and cholesteryl ester accompanied by attenuation of secretion of apolipoprotein B-containing lipoproteins. Hepatic lipase-mediated depletion of intracellular lipid storage increased the expression of peroxisome proliferator-activated receptor α and its target genes and augmented oxidation of fatty acids. These data show that 1) hepatic lipase is active in the endoplasmic reticulum and 2) intracellular hepatic lipase modulates cellular lipid metabolism and lipoprotein secretion.
Assuntos
Retículo Endoplasmático/enzimologia , Hepatócitos/enzimologia , Lipase/metabolismo , Lipoproteínas VLDL/metabolismo , Fígado/enzimologia , Triglicerídeos/metabolismo , Animais , Apolipoproteínas B/metabolismo , Células Cultivadas , Ésteres do Colesterol/metabolismo , Ácidos Graxos/metabolismo , Hepatócitos/citologia , Metabolismo dos Lipídeos , Fígado/citologia , Camundongos , Oxirredução , PPAR alfa/metabolismoRESUMO
Statins, routinely used to treat hypercholesterolemia, selectively induce apoptosis in some tumor cells by inhibiting the mevalonate pathway. Recent clinical studies suggest that a subset of breast tumors is particularly susceptible to lipophilic statins, such as fluvastatin. To quickly advance statins as effective anticancer agents for breast cancer treatment, it is critical to identify the molecular features defining this sensitive subset. We have therefore characterized fluvastatin sensitivity by MTT assay in a panel of 19 breast cell lines that reflect the molecular diversity of breast cancer, and have evaluated the association of sensitivity with several clinicopathological and molecular features. A wide range of fluvastatin sensitivity was observed across breast tumor cell lines, with fluvastatin triggering cell death in a subset of sensitive cell lines. Fluvastatin sensitivity was associated with an estrogen receptor alpha (ERα)-negative, basal-like tumor subtype, features that can be scored with routine and/or strong preclinical diagnostics. To ascertain additional candidate sensitivity-associated molecular features, we mined publicly available gene expression datasets, identifying genes encoding regulators of mevalonate production, non-sterol lipid homeostasis, and global cellular metabolism, including the oncogene MYC. Further exploration of this data allowed us to generate a 10-gene mRNA abundance signature predictive of fluvastatin sensitivity, which showed preliminary validation in an independent set of breast tumor cell lines. Here, we have therefore identified several candidate predictors of sensitivity to fluvastatin treatment in breast cancer, which warrant further preclinical and clinical evaluation.
Assuntos
Neoplasias da Mama/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/genética , Ácidos Graxos Monoinsaturados/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Indóis/farmacologia , Antineoplásicos/farmacologia , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Receptor alfa de Estrogênio/biossíntese , Feminino , Fluvastatina , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Hidroximetilglutaril-CoA-Redutases NADP-Dependentes/biossíntese , Células MCF-7 , Ácido Mevalônico/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/biossíntese , Receptor ErbB-2RESUMO
BACKGROUND: FoxO3a, a member of the FOXO family of transcription factors, is expressed in adult liver and modulates the expression of genes involved in apoptosis. FoxO3a is post-translationally regulated, negatively by PI3K/Akt and MAPK/Erk and positively by oxidative stress/JNK pathways. In previous works, we have demonstrated that interferon-α2b (IFN-α2b) induces apoptosis of hepatic preneoplastic foci through the production of reactive oxygen species (ROS). AIMS: To investigate the post-translational signal events triggered by the oxidative stress induced by IFN-α2b and the modulation of FoxO3a transcriptional activity during these events in rat preneoplastic liver. METHODS: Adult male Wistar rats were subjected to a two-phase model of hepatocarcinogenesis. A group of animals received IFN-α2b and another group received IFN-α2b and ascorbic acid (ASC), by intraperitoneal injection. Lipid peroxidation, immunohistochemistry, immunoblotting, co-immunoprecipitation and sqRT-PCR assays were performed to explore the role of ROS, JNK, Akt, Erk, FoxO3a, ß-catenin and PUMA in the IFN-α2b-mediated apoptotic mechanism. RESULTS: In vivo IFN-α2b treatment induced endogenous production of ROS which activated JNK. IFN-α2b blocked the activation of Akt and Erk, avoiding FoxO3a activity repression. Activated JNK was responsible for the nuclear translocation and transcriptional activity of FoxO3a which positively modulated the expression of PUMA, a proapoptotic player. In addition, nuclear FoxO3a competed for the nuclear ß-catenin associated to TCF, inhibiting the canonical Wnt signalling pathway. CONCLUSIONS: The data presented here propose a model in which in vivo IFN-α2b treatment induces nuclear translocation and transcriptional activity of FoxO3a, triggering the mitochondrial apoptotic pathway in hepatic preneoplastic foci.
Assuntos
Apoptose/genética , Carcinogênese/efeitos dos fármacos , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica/fisiologia , Fígado/metabolismo , Estresse Oxidativo/fisiologia , Transdução de Sinais/fisiologia , Animais , Proteína Forkhead Box O3 , Regulação da Expressão Gênica/genética , Immunoblotting , Imuno-Histoquímica , Imunoprecipitação , Interferon alfa-2 , Interferon-alfa/farmacologia , Peroxidação de Lipídeos , Masculino , Modelos Biológicos , Processamento de Proteína Pós-Traducional/genética , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genéticaRESUMO
RATIONALE: Carboxylesterase 3/triacylglycerol hydrolase (TGH) has been shown to participate in hepatic very low-density lipoprotein (VLDL) assembly. Deficiency of TGH in mice lowers plasma lipids and atherogenic lipoproteins without inducing hepatic steatosis. OBJECTIVE: To investigate the contribution of TGH to atherosclerotic lesion development in mice that lack low-density lipoprotein receptor (LDLR). METHODS AND RESULTS: Mice deficient in LDL receptor (Ldlr(-/-)) and mice lacking both TGH and LDLR (Tgh(-/-)/Ldlr(-/-)) were fed with a Western-type diet for 12 weeks. Analysis of Tgh(-/-)/Ldlr(-/-) plasma showed an atheroprotective lipoprotein profile with decreased cholesterol in the VLDL and the LDL fractions, concomitant with elevated high-density lipoprotein cholesterol. Significantly reduced plasma apolipoprotein B levels were also observed in Tgh(-/-)/Ldlr(-/-) mice. Consequently, Tgh(-/-)/Ldlr(-/-) mice presented with a significant reduction (54%, P<0.01) of the high-fat, high-cholesterol dieteninduced atherosclerotic plaques when compared with Tgh(+/+)/Ldlr(-/-) mice in the cross-sectional aortic root analysis. TGH deficiency did not further increase liver steatosis despite lowering plasma lipids, mainly due to reduced hepatic lipogenesis. The ameliorated dyslipidemia in Tgh(-/-)/Ldlr(-/-) mice was accompanied with significantly improved insulin sensitivity. CONCLUSIONS: Inhibition of TGH activity ameliorates atherosclerosis development and improves insulin sensitivity in Ldlr(-/-) mice.
Assuntos
Doença da Artéria Coronariana/metabolismo , Dislipidemias/metabolismo , Lipase/genética , Receptores de LDL/genética , Animais , Aorta/metabolismo , Aorta/patologia , Apolipoproteínas B/sangue , Glicemia/metabolismo , HDL-Colesterol/sangue , LDL-Colesterol/sangue , VLDL-Colesterol/sangue , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/patologia , Modelos Animais de Doenças , Dislipidemias/genética , Ácidos Graxos/metabolismo , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Feminino , Insulina/sangue , Resistência à Insulina/fisiologia , Lipase/deficiência , Lipase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Receptores de LDL/metabolismoRESUMO
The hallmark of obesity and one of the key contributing factors to insulin resistance, type 2 diabetes and cardiovascular disease is excess triacylglycerol (TG) storage. In hepatocytes, excessive accumulation of TG is the common denominator of a wide range of clinicopathological entities known as nonalcoholic fatty liver disease, which can eventually progress to cirrhosis and associated complications including hepatic failure, hepatocellular carcinoma and death. A tight regulation between TG synthesis, hydrolysis, secretion and fatty acid oxidation is required to prevent lipid accumulation as well as lipid depletion from hepatocytes. Therefore, understanding the pathways that regulate hepatic TG metabolism is crucial for development of therapies to ameliorate pathophysiological conditions associated with excessive hepatic TG accumulation, including dyslipidemias, viral infection and atherosclerosis. This review highlights the physiological roles of liver lipases that degrade TG in cytosolic lipid droplets, endoplasmic reticulum, late endosomes/lysosomes and along the secretory route. This article is part of a Special Issue entitled Triglyceride Metabolism and Disease.
Assuntos
Hepatócitos/enzimologia , Lipase , Fígado/enzimologia , Triglicerídeos , Animais , Autofagia , Diabetes Mellitus Tipo 2/enzimologia , Esterases/metabolismo , Fígado Gorduroso/enzimologia , Fígado Gorduroso/etiologia , Humanos , Lipase/classificação , Lipase/metabolismo , Cirrose Hepática/enzimologia , Hepatopatia Gordurosa não Alcoólica , Triglicerídeos/biossíntese , Triglicerídeos/metabolismoRESUMO
BACKGROUND & AIMS: Very low density lipoproteins (VLDLs) are triacylglycerol (TG)-rich lipoproteins produced by the human liver. VLDLs derive the majority of their TG cargo from the lipolysis of TG stored in hepatocellular lipid droplets (LDs). Important roles for LDs and the VLDL secretory pathway in the cell culture production of infectious hepatitis C virus (HCV) have been established. We hypothesized that TG lipolysis and VLDL production are impaired during HCV infection so that these cellular processes can be diverted towards HCV production. METHODS: We used an HCV permissive cell culture system (JFH-1/HuH7.5 cells) to examine the relationship between TG lipolysis, VLDL assembly, and the HCV lifecycle using standard biochemical approaches. RESULTS: Lipolysis of cellular TG and VLDL production were impaired in HCV infected cells during the early peak of viral infection. This was partially explained by an apparent deficiency of a putative TG lipase, arylacetamide deacetylase (AADAC). The re-introduction of AADAC to infected cells restored cellular TG lipolysis, indicating a role for HCV-mediated downregulation of AADAC in this process. Defective lipolysis of cellular TG stores and VLDL production were also observed in HuH7.5 cells stably expressing a short hairpin RNA targeting AADAC expression, proving AADAC deficiency contributes to these defective pathways. Finally, impaired production of HCV was observed with AADAC knockdown cells, demonstrating a role for AADAC in the HCV lifecycle. CONCLUSIONS: This insight into the biology of HCV infection and possibly pathogenesis identifies AADAC as a novel and translationally relevant therapeutic target.
Assuntos
Hidrolases de Éster Carboxílico/metabolismo , Hepacivirus/fisiologia , Lipoproteínas VLDL/metabolismo , Triglicerídeos/metabolismo , Apolipoproteínas B/metabolismo , Hidrolases de Éster Carboxílico/antagonistas & inibidores , Hidrolases de Éster Carboxílico/genética , Linhagem Celular , Técnicas de Silenciamento de Genes , Hepacivirus/crescimento & desenvolvimento , Hepacivirus/patogenicidade , Interações Hospedeiro-Patógeno , Humanos , Lipólise , Modelos Biológicos , Virulência , Replicação ViralRESUMO
UNLABELLED: Increased lipogenesis, together with hyperlipidemia and increased fat deposition, contribute to obesity and associated metabolic disorders including nonalcoholic fatty liver disease. Here we show that carboxylesterase 1/esterase-x (Ces1/Es-x) plays a regulatory role in hepatic fat metabolism in the mouse. We demonstrate that Ces1/Es-x knockout mice present with increased hepatic lipogenesis and with oversecretion of apolipoprotein B (apoB)-containing lipoproteins (hepatic very-low density lipoproteins), which leads to hyperlipidemia and increased fat deposition in peripheral tissues. Consequently, Ces1/Es-x knockout mice develop obesity, fatty liver, hyperinsulinemia, and insulin insensitivity on chow diet without change in food intake and present with decreased energy expenditure. Ces1/Es-x deficiency prevents the release of polyunsaturated fatty acids from triacylglycerol stores, leading to an up-regulation of sterol regulatory element binding protein 1c-mediated lipogenesis, which can be reversed with dietary ω-3 fatty acids. CONCLUSION: These studies support a role for Ces1/Es-x in the partitioning of regulatory fatty acids and concomitant control of hepatic lipid biosynthesis, secretion, and deposition.
Assuntos
Hidrolases de Éster Carboxílico/deficiência , Colesterol/metabolismo , Fígado Gorduroso/enzimologia , Hiperlipidemias/enzimologia , Obesidade/enzimologia , Análise de Variância , Animais , Glicemia/metabolismo , Hidrolases de Éster Carboxílico/genética , Hidrolases de Éster Carboxílico/metabolismo , Ácidos Docosa-Hexaenoicos/metabolismo , Metabolismo Energético , Jejum , Ácidos Graxos Insaturados/administração & dosagem , Ácidos Graxos Insaturados/metabolismo , Fígado Gorduroso/genética , Feminino , Óleos de Peixe/administração & dosagem , Expressão Gênica , Hepatócitos/metabolismo , Hiperlipidemias/genética , Insulina/sangue , Lipoproteínas VLDL/metabolismo , Camundongos , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica , Obesidade/genética , Fenótipo , Fosfolipídeos/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Triglicerídeos/metabolismoRESUMO
UNLABELLED: Carboxylesterase 3/triacylglycerol hydrolase (Ces3/TGH) participates in hepatic very low-density lipoprotein (VLDL) assembly and in adipose tissue basal lipolysis. Global ablation of Ces3/Tgh expression decreases serum triacylglycerol (TG) and nonesterified fatty acid levels and improves insulin sensitivity. To understand the tissue-specific role of Ces3/TGH in lipid and glucose homeostasis, we generated mice with a liver-specific deletion of Ces3/Tgh expression (L-TGH knockout [KO]). Elimination of hepatic Ces3/Tgh expression dramatically decreased plasma VLDL TG and VLDL cholesterol concentrations but only moderately increased liver TG levels in mice fed a standard chow diet. Significantly reduced plasma TG and cholesterol without hepatic steatosis were also observed in L-TGH KO mice challenged with a high-fat, high-cholesterol diet. L-TGH KO mice presented with increased plasma ketone bodies and hepatic fatty acid oxidation. Intrahepatic TG in L-TGH KO mice was stored in significantly smaller lipid droplets. Augmented hepatic TG levels in chow-fed L-TGH KO mice did not affect glucose tolerance or glucose production from hepatocytes, but impaired insulin tolerance was observed in female mice. CONCLUSION: Our data suggest that ablation of hepatic Ces3/Tgh expression decreases plasma lipid levels without causing severe hepatic steatosis.
Assuntos
VLDL-Colesterol/sangue , Lipase/deficiência , Lipoproteínas VLDL/sangue , Fígado/enzimologia , Fígado/metabolismo , Triglicerídeos/sangue , Animais , Colesterol na Dieta/administração & dosagem , VLDL-Colesterol/metabolismo , Gorduras na Dieta/administração & dosagem , Ácidos Graxos/metabolismo , Fígado Gorduroso/metabolismo , Feminino , Glucose/biossíntese , Teste de Tolerância a Glucose , Insulina/metabolismo , Resistência à Insulina , Corpos Cetônicos/sangue , Lipase/genética , Lipoproteínas VLDL/metabolismo , Masculino , Camundongos , Camundongos Knockout , Transdução de Sinais , Triglicerídeos/metabolismoRESUMO
Overproduction of apolipoprotein B (apoB)-containing lipoproteins by the liver and the intestine is 1 of the hallmarks of insulin resistance and type 2 diabetes and a well-established risk factor of cardiovascular disease. The assembly of apoB lipoproteins is regulated by the availability of lipids that form the neutral lipid core (triacylglycerol and cholesteryl ester) and the limiting lipoprotein monolayer (phospholipids and cholesterol). Although tremendous advances have been made over the past decade toward understanding neutral lipid and phospholipid biosynthesis and neutral lipid storage in cytosolic lipid droplets (LDs), little is known about the mechanisms that govern the transfer of lipids to the lumen of the endoplasmic reticulum for apoB lipidation. ApoB-synthesizing organs can deposit synthesized neutral lipids into at least 3 different types of LDs, each decorated with a subset of specific proteins: perilipin-decorated cytosolic LDs, and 2 types of LDs formed in the lumen of the endoplasmic reticulum, the secretion-destined LDs containing apoB, and resident lumenal LDs coated with microsomal triglyceride transfer protein and exchangeable apolipoproteins. This brief review will address the current knowledge of lumenal lipid metabolism in the context of apoB assembly and lipid storage.
Assuntos
Metabolismo dos Lipídeos , Lipídeos/biossíntese , Lipoproteínas/biossíntese , Fígado/metabolismo , Animais , Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/metabolismo , Retículo Endoplasmático/metabolismo , Humanos , Fígado/citologiaRESUMO
A new tioconazole (TCZ) mucoadhesive film, based on a biodegradable chitosan/ hydroxypropyl methylcellulose (CH/HPMC) blend, was developed for treatment of vaginal candidiasis. The formulation was optimized through an I-optimal design (minimizing the integral of the prediction variance across the factor space), where the impact of the proportion of the ingredients and processing variables on the quality of the final product was evaluated. Both, the thickness of the film and the swelling index, which affect patients' comfort and compliance, were considered. Mechanical testing, such as load at break, elongation at break, and mucoadhesive strength were also included as dependent variables. The optimal mucoadhesive film formulation, which should be obtained at a drying temperature of 30 °C, was found to include the combination of CH and HPMC (forming polymers) at 0.25:0.75 ratio, a mixture of polyethylene glycol 400 and propylene glycol as plasticizers (0.07:0.93, 5% w/w), and TCZ loaded at 15 % w/w. The optimal preparation was subjected to exhaustive characterization studies, which revealed that the drug was entrapped in the polymeric matrix in an amorphous state and that the film exhibited a smooth and uniform surface, demonstrating excellent component compatibility. In vitro tests showed that the formulation has an excellent time to kill value (3 min) and lacks cytotoxicity, suggesting that it should be highly effective and safe.
Assuntos
Imidazóis , Projetos de Pesquisa , Feminino , Humanos , Derivados da Hipromelose , PolímerosRESUMO
Many cancer patients receive their classical therapies together with vitamin supplements. However, the effectiveness of these strategies is on debate. Here we aimed to evaluate how vitamin E supplementation affects the anticancer effects of interferon (IFN-α) using an early-model of liver cancer development (initiation-promotion, IP). Male Wistar rats subjected to this model were divided as follows: untreated (IP), IP treated with recombinant IFN-α-2b (6.5 × 105 U/kg), IP treated with vitamin E (50 mg/kg), and IP treated with combination of vitamin E and IFN-α-2b. After treatments rats were fasted and euthanized and plasma and livers were collected. Combined administration of vitamin E and IFN-α-2b induced body weight drop, increased liver apoptosis, and low levels of hepatic lipids. Interestingly, vitamin E and IFN-α-2b combination also induced an increase in altered hepatic foci number, but not in size. It seems that vitamin E acts on its antioxidant capability in order to block the oxidative stress induced by IFN-α-2b, blocking in turn its beneficial effects on preneoplastic livers, leading to harmful final effects. In conclusion, this study shows that vitamin E supplementation in IFN-α-2b-treated rats exerts unwanted effects; and highlights that in spite of being natural, nutritional supplements may not always exert beneficial outcomes when used as complementary therapy for the treatment of cancer.
Assuntos
Anticarcinógenos/farmacologia , Interferon alfa-2/farmacologia , Neoplasias Hepáticas/prevenção & controle , Vitamina E/farmacologia , Vitaminas/farmacologia , Animais , Carcinogênese/efeitos dos fármacos , Carcinogênese/patologia , Interações Medicamentosas , Fígado/efeitos dos fármacos , Fígado/patologia , Neoplasias Hepáticas/patologia , Masculino , Ratos WistarRESUMO
Contacts between organelles create microdomains that play major roles in regulating key intracellular activities and signaling pathways, but whether they also regulate systemic functions remains unknown. Here, we report the ultrastructural organization and dynamics of the inter-organellar contact established by sheets of curved rough endoplasmic reticulum closely wrapped around the mitochondria (wrappER). To elucidate the in vivo function of this contact, mouse liver fractions enriched in wrappER-associated mitochondria are analyzed by transcriptomics, proteomics, and lipidomics. The biochemical signature of the wrappER points to a role in the biogenesis of very-low-density lipoproteins (VLDL). Altering wrappER-mitochondria contacts curtails VLDL secretion and increases hepatic fatty acids, lipid droplets, and neutral lipid content. Conversely, acute liver-specific ablation of Mttp, the most upstream regulator of VLDL biogenesis, recapitulates this hepatic dyslipidemia phenotype and promotes remodeling of the wrappER-mitochondria contact. The discovery that liver wrappER-mitochondria contacts participate in VLDL biology suggests an involvement of inter-organelle contacts in systemic lipid homeostasis.
Assuntos
Retículo Endoplasmático/metabolismo , Homeostase , Lipídeos/química , Fígado/metabolismo , Mitocôndrias/metabolismo , Animais , Retículo Endoplasmático/ultraestrutura , Enterócitos/metabolismo , Inativação Gênica , Hepatócitos/metabolismo , Imageamento Tridimensional , Intestino Delgado/citologia , Lipoproteínas VLDL/biossíntese , Masculino , Metabolômica , Camundongos Endogâmicos C57BL , Mitocôndrias/ultraestrutura , Membranas Mitocondriais/metabolismo , Fosfolipídeos/biossíntese , Proteínas/metabolismoRESUMO
Wnt/beta-catenin/T cell factor (TCF) pathway is activated in several types of human cancers, promoting cell growth and proliferation. Forkhead box containing protein class O (FOXO) transcription factors compete with TCF for beta-catenin binding, particularly under cellular oxidative stress conditions. Contrary to beta-catenin/TCF, beta-catenin/FOXO promotes the transcription of genes involved in cell cycle arrest and apoptosis. We have previously demonstrated that in vivo interferon-alpha2b (IFN-alpha2b) administration induces apoptosis in preneoplastic livers, a mechanism mediated by reactive oxygen species (ROS) and transforming growth factor-beta(1) (TGF-beta(1)). This study was aimed to assess the status of the Wnt/beta-catenin/TCF pathway in a very early stage of rat hepatocarcinogenesis and to further evaluate the effects of in vivo IFN-alpha2b treatment on it. We demonstrated that the Wnt/beta-catenin/TCF pathway is activated in preneoplastic rat livers. More important, in vivo IFN-alpha2b treatment inhibits Wnt/beta-catenin/TCF pathway and promotes programed cell death possibly providing a link with FOXO pathway.