Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
PLoS One ; 12(8): e0181052, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28797077

RESUMO

Myeloid cell and hepatocyte IKKß may mediate the genesis of obesity and insulin resistance in mice fed high fat diet. However, their gender-specific roles in the pathogenesis of non-alcoholic steatohepatitis (NASH) are not known. Here we demonstrate myeloid IKKß deficiency prevents Western diet-induced obesity and visceral adiposity in females but not in males, and attenuates hyperglycemia, global IR, and NASH in both genders. In contrast, all metabolic sequela including NASH are aggravated by hepatocyte IKKß deficiency (IkbkbΔhep) in male but not female mice. Gene profiling identifies sulfotransferase family 1E (Sult1e1), which encodes a sulfotransferase E1 responsible for inactivation of estrogen, as a gene upregulated in NASH in both genders and most conspicuously in male IkbkbΔhep mice having worst NASH and lowest plasma estradiol levels. LXRα is enriched to LXRE on Sult1e1 promoter in male WT and IkbkbΔhep mice with NASH, and a Sult1e1 promoter activity is increased by LXRα and its ligand and augmented by expression of a S32A mutant of IκBα. These results demonstrate striking gender differences in regulation by IKKß of high cholesterol saturated fat diet-induced metabolic changes including NASH and suggest hepatocyte IKKß is protective in male due at least in part to its ability to repress LXR-induced Sult1e1. Our findings also raise a caution for systemic IKK inhibition for the treatment of NASH as it may exacerbate the disease in male patients.


Assuntos
Quinase I-kappa B/genética , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/genética , Sulfotransferases/genética , Adiposidade , Animais , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Hepatócitos/patologia , Hiperglicemia/etiologia , Hiperglicemia/genética , Hiperglicemia/patologia , Quinase I-kappa B/análise , Resistência à Insulina/genética , Gordura Intra-Abdominal/metabolismo , Gordura Intra-Abdominal/patologia , Masculino , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/patologia , Obesidade/etiologia , Obesidade/genética , Obesidade/patologia , Fatores Sexuais , Sulfotransferases/análise , Transcriptoma
2.
J Immunol ; 187(10): 5408-18, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22013115

RESUMO

We recently demonstrated that ω-3-polyunsaturated fatty acids ameliorate obesity-induced adipose tissue inflammation and insulin resistance. In this study, we report novel mechanisms underlying ω-3-polyunsaturated fatty acid actions on adipose tissue, adipocytes, and stromal vascular cells (SVC). Inflamed adipose tissue from high-fat diet-induced obese mice showed increased F4/80 and CD11b double-positive macrophage staining and elevated IL-6 and MCP-1 levels. Docosahexaenoic acid (DHA; 4 µg/g) did not change the total number of macrophages but significantly reduced the percentage of high CD11b/high F4/80-expressing cells in parallel with the emergence of low-expressing CD11b/F4/80 macrophages in the adipose tissue. This effect was associated with downregulation of proinflammatory adipokines in parallel with increased expression of IL-10, CD206, arginase 1, resistin-like molecule α, and chitinase-3 like protein, indicating a phenotypic switch in macrophage polarization toward an M2-like phenotype. This shift was confined to the SVC fraction, in which secretion of Th1 cytokines (IL-6, MCP-1, and TNF-α) was blocked by DHA. Notably, resolvin D1, an anti-inflammatory and proresolving mediator biosynthesized from DHA, markedly attenuated IFN-γ/LPS-induced Th1 cytokines while upregulating arginase 1 expression in a concentration-dependent manner. Resolvin D1 also stimulated nonphlogistic phagocytosis in adipose SVC macrophages by increasing both the number of macrophages containing ingested particles and the number of phagocytosed particles and by reducing macrophage reactive oxygen species production. No changes in adipocyte area and the phosphorylation of hormone-sensitive lipase, a rate-limiting enzyme regulating adipocyte lipolysis, were observed. These findings illustrate novel mechanisms through which resolvin D1 and its precursor DHA confer anti-inflammatory and proresolving actions in inflamed adipose tissue.


Assuntos
Tecido Adiposo/imunologia , Tecido Adiposo/patologia , Polaridade Celular/imunologia , Ácidos Docosa-Hexaenoicos/fisiologia , Mediadores da Inflamação/fisiologia , Macrófagos/imunologia , Macrófagos/patologia , Animais , Modelos Animais de Doenças , Imunofenotipagem , Ativação de Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/imunologia , Obesidade/metabolismo , Obesidade/patologia , Distribuição Aleatória , Transdução de Sinais/imunologia
3.
Curr Opin Clin Nutr Metab Care ; 14(4): 347-53, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21587068

RESUMO

PURPOSE OF REVIEW: Obesity is a major risk factor for metabolic syndrome-related comorbidities such as insulin resistance, type-II diabetes, and nonalcoholic fatty liver disease (NAFLD). A wealth of evidence indicates that the associated pathologies of the metabolic syndrome are aggravated by the presence of a chronic state of 'low-grade' inflammation in the adipose tissue. This article discusses recent data implicating lipoxygenases and especially 5-lipoxygenase and its derived products, the leukotrienes, in mounting adipose tissue inflammation and related pathologies in obesity. RECENT FINDINGS: Overexpression of selected members of the 5-lipoxygenase pathway and increased leukotriene production are common findings in excessive visceral fat depots. In these conditions, 5-lipoxygenase products exert potent proinflammatory actions including induction of nuclear factor-κB and secretion of proinflammatory and insulin resistant adipokines (i.e., monocyte chemotactic protein-1, tumor necrosis factor-α, macrophage inflammatory protein-1γ, and interleukin-6) by adipose tissue. The 5-lipoxygenase pathway also plays a major role in mounting inflammation in hepatic tissue and has emerged as a pathogenic factor in obesity-induced NAFLD. Similar role in NAFLD has been proposed for the 12/15-lipoxygenase pathway. SUMMARY: Modulation of lipoxygenases represents a novel target in the prevention of adipose tissue and hepatic dysfunction related to the metabolic syndrome.


Assuntos
Araquidonato 5-Lipoxigenase/metabolismo , Fígado Gorduroso/fisiopatologia , Resistência à Insulina , Leucotrienos/metabolismo , Obesidade/fisiopatologia , Adipocinas/metabolismo , Tecido Adiposo/patologia , Animais , Araquidonato 12-Lipoxigenase/metabolismo , Araquidonato 15-Lipoxigenase/metabolismo , Araquidonato 5-Lipoxigenase/genética , Quimiocina CCL2/metabolismo , Diabetes Mellitus Tipo 2/etiologia , Fígado Gorduroso/etiologia , Humanos , Inflamação/patologia , Insulina/metabolismo , Interleucina-6/metabolismo , Leucotrienos/genética , Proteínas Inflamatórias de Macrófagos/metabolismo , Síndrome Metabólica/etiologia , NF-kappa B/metabolismo , Hepatopatia Gordurosa não Alcoólica , Obesidade/complicações , Fatores de Risco , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
4.
FASEB J ; 25(8): 2538-50, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21507897

RESUMO

Peroxisome proliferator-activated receptor (PPAR) γ is a nuclear receptor central to glucose and lipid homeostasis. PPARγ role in nonalcoholic fatty liver disease is controversial because PPARγ overexpression is a general property of steatotic livers, but its activation by thiazolidinediones reduces hepatic steatosis. Here, we investigated hepatic PPARγ function by using Cre-loxP technology to generate hepatocyte (PPARγ(Δhep))- and macrophage (PPARγ(Δmac))-specific PPARγ-knockout mice. Targeted deletion of PPARγ in hepatocytes, and to a lesser extent in macrophages, protected mice against high-fat diet-induced hepatic steatosis. Down-regulated expression of genes involved in lipogenesis (SCD1, SREBP-1c, and ACC), lipid transport (CD36/FAT, L-FABP, and MTP), and ß-oxidation (PPARα and ACO) was observed in PPARγ(Δhep) mice. Moreover, PPARγ(Δhep) mice showed improved glucose tolerance and reduced PEPCK expression without changes in Pcx, Fbp1, and G6Pc expression and CREB and JNK phosphorylation. In precision-cut liver slices (PCLSs) and hepatocytes, rosiglitazone either alone or in combination with oleic acid increased triglyceride accumulation, an effect that was blocked by the PPARγ antagonist biphenol A diglycidyl ether (BADGE). PCLSs and hepatocytes from PPARγ(Δhep) mice showed blunted responses to rosiglitazone and oleic acid, whereas the response to these compounds remained intact in PCLSs from PPARγ(Δmac) mice. Collectively, these findings establish PPARγ expression in hepatocytes as a prosteatotic factor in fatty liver disease.


Assuntos
Fígado Gorduroso/etiologia , Fígado Gorduroso/fisiopatologia , Obesidade/complicações , Obesidade/fisiopatologia , PPAR gama/fisiologia , Animais , Sequência de Bases , Primers do DNA/genética , Gorduras na Dieta/administração & dosagem , Fígado Gorduroso/genética , Fígado Gorduroso/patologia , Expressão Gênica , Marcação de Genes , Glucose/metabolismo , Hepatócitos/fisiologia , Células de Kupffer/fisiologia , Camundongos , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica , Obesidade/genética , Obesidade/patologia , Especificidade de Órgãos , PPAR gama/deficiência , PPAR gama/genética
5.
Hepatology ; 52(6): 1980-91, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20967760

RESUMO

UNLABELLED: We have shown that Alox15, the gene encoding for 12/15-lipoxygenase (12/15-LO), is markedly up-regulated in livers from apolipoprotein E-deficient (ApoE(-/-)) mice, which spontaneously develop nonalcoholic fatty liver disease secondary to hyperlipidemia. In the current study, we used ApoE(-/-) mice with a targeted disruption of the Alox15 gene to assess the role of 12/15-LO in the development and progression of hepatic steatosis and inflammation. Compared with ApoE(-/-) mice, which exhibited extensive hepatic lipid accumulation and exacerbated inflammatory injury, ApoE/12/15-LO double-knockout (ApoE(-/-)/12/15-LO(-/-)) mice showed reduced serum alanine aminotransferase levels; decreased hepatic steatosis, inflammation, and macrophage infiltration; and decreased fatty acid synthase, tumor necrosis factor α (TNFα), monocyte chemoattractant protein-1 (MCP-1), interleukin (IL)-18, and IL-6 expression. Remarkably, disruption of Alox15 attenuated glucose intolerance and high-fat diet-induced insulin resistance, up-regulated insulin receptor substrate-2, and exerted opposite effects on hepatic c-Jun amino-terminal kinase and adenosine monophosphate-activated protein kinase phosphorylation, known negative and positive regulators of insulin signaling, respectively. In adipose tissue, the absence of Alox15 induced significant reductions in the expression of the proinflammatory and insulin-resistant adipokines MCP-1, TNFα, and resistin while increasing the expression of glucose transporter-4. Interestingly, compared with ApoE(-/-) mice, which exhibited increased hepatic caspase-3 staining, ApoE(-/-)/12/15-LO(-/-) mice showed attenuated hepatocellular injury. Consistent with this finding, hepatocytes isolated from ApoE(-/-) mice were more vulnerable to TNFα-induced programmed cell death, an effect that was not observed in hepatocytes carrying a targeted disruption of the Alox15 gene. CONCLUSION: Collectively, our data suggest a potentially relevant mechanism linking 12/15-LO to the promotion of hepatic steatosis, insulin resistance, and inflammation in experimental liver disease of metabolic origin.


Assuntos
Apolipoproteínas E/deficiência , Araquidonato 12-Lipoxigenase/genética , Araquidonato 15-Lipoxigenase/genética , Fígado Gorduroso/prevenção & controle , Alanina Transaminase/sangue , Animais , Antígenos de Diferenciação/imunologia , Apoptose , Araquidonato 12-Lipoxigenase/fisiologia , Araquidonato 15-Lipoxigenase/fisiologia , Fígado Gorduroso/genética , Teste de Tolerância a Glucose , Resistência à Insulina , Fígado/patologia , Masculino , Camundongos , Camundongos Knockout , Regulação para Cima
6.
J Immunol ; 184(7): 3978-87, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20207999

RESUMO

The presence of the so-called low-grade inflammatory state is recognized as a critical event in adipose tissue dysfunction, leading to altered secretion of adipokines and free fatty acids (FFAs), insulin resistance, and development of hepatic complications associated with obesity. This study was designed to investigate the potential contribution of the proinflammatory 5-lipoxygenase (5-LO) pathway to adipose tissue inflammation and lipid dysfunction in experimental obesity. Constitutive expression of key components of the 5-LO pathway, as well as leukotriene (LT) receptors, was detected in adipose tissue as well as in adipocyte and stromal vascular fractions. Adipose tissue from obese mice, compared with that from lean mice, exhibited increased 5-LO activating protein (FLAP) expression and LTB(4) levels. Incubation of adipose tissue with 5-LO products resulted in NF-kappaB activation and augmented secretion of proinflammatory adipokines such as MCP-1, IL-6, and TNF-alpha. In addition, LTB(4), but not LTD(4), reduced FFA uptake in primary adipocytes, whereas 5-LO inhibition suppressed isoproterenol-induced adipose tissue lipolysis. In mice with dietary obesity, elevated FLAP expression in adipose tissue was paralleled with macrophage infiltration, increased circulating FFA levels, and hepatic steatosis, phenomena that were reversed by FLAP inhibition with Bay-X-1005. Interestingly, FLAP inhibition induced AMP-activated protein kinase phosphorylation in parallel with decreases in hormone-sensitive lipase activity and the expression and secretion of TNF-alpha and IL-6. Similar effects were observed in differentiated 3T3-L1 adipocytes incubated with either Bay-X-1005 or the selective LTB(4) receptor antagonist U-75302. Taken together, these findings indicate that the 5-LO pathway signals the adipose tissue low-grade inflammatory state and steatogenic potential in experimental obesity.


Assuntos
Tecido Adiposo/metabolismo , Proteínas de Transporte/metabolismo , Inflamação/metabolismo , Proteínas de Membrana/metabolismo , Obesidade/metabolismo , Proteínas Ativadoras de 5-Lipoxigenase , Tecido Adiposo/patologia , Animais , Cromatografia Líquida de Alta Pressão , Citocinas/metabolismo , Modelos Animais de Doenças , Eicosanoides/análise , Eicosanoides/metabolismo , Ensaio de Imunoadsorção Enzimática , Ácidos Graxos/metabolismo , Fígado Gorduroso , Expressão Gênica , Perfilação da Expressão Gênica , Imuno-Histoquímica , Inflamação/patologia , Inflamação/fisiopatologia , Metabolismo dos Lipídeos , Lipídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Obesidade/patologia , Obesidade/fisiopatologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Hepatology ; 51(3): 817-27, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20112424

RESUMO

UNLABELLED: The actual risk factors that drive hepatic inflammation during the transition from steatosis to steatohepatitis are unknown. We recently demonstrated that hyperlipidemia-prone apolipoprotein E-deficient (ApoE(-/-)) mice exhibit hepatic steatosis and increased susceptibility to hepatic inflammation and advanced fibrosis. Because the proinflammatory 5-lipoxygenase (5-LO) pathway was found to be up-regulated in these mice and given that 5-LO deficiency confers cardiovascular protection to ApoE(-/-) mice, we determined the extent to which the absence of 5-LO would alter liver injury in these mice. Compared with ApoE(-/-) mice, which showed expected hepatic steatosis and inflammation, ApoE/5-LO double-deficient (ApoE(-/-)/5-LO(-/-)) mice exhibited reduced hepatic inflammation, macrophage infiltration, tumor necrosis factor alpha (TNF-alpha), monocyte chemoattractant protein-1 (MCP-1) and interleukin (IL)-18 expression, caspase-3 and nuclear factor-kappaB (NF-kappaB) activities, and serum alanine aminotransferase levels in the absence of changes in hepatic steatosis. The lack of 5-LO produced a remarkable insulin-sensitizing effect in the adipose tissue because peroxisome proliferator-activated receptor gamma, insulin receptor substrate-1, and adiponectin were up-regulated, whereas c-Jun amino-terminal kinase phosphorylation and MCP-1 and IL-6 expression were down-regulated. On the other hand, hepatocytes isolated from ApoE(-/-)/5-LO(-/-) mice were more resistant to TNF-alpha-induced apoptosis. The 5-LO products leukotriene (LT) B(4), LTD(4), and 5-HETE consistently triggered TNF-alpha-induced apoptosis and compromised hepatocyte survival by suppressing NF-kappaB activity in the presence of actinomycin D. Moreover, ApoE(-/-)/5-LO(-/-) mice were protected against sustained high-fat diet (HFD)-induced liver injury and hepatic inflammation, macrophage infiltration and insulin resistance were significantly milder than those of ApoE(-/-) mice. Finally, pharmacological inhibition of 5-LO significantly reduced hepatic inflammatory infiltrate in the HFD and ob/ob models of fatty liver disease. CONCLUSION: These combined data indicate that hyperlipidemic mice lacking 5-LO are protected against hepatic inflammatory injury, suggesting that 5-LO is involved in mounting hepatic inflammation in metabolic disease.


Assuntos
Araquidonato 5-Lipoxigenase/deficiência , Araquidonato 5-Lipoxigenase/fisiologia , Hepatócitos/patologia , Hiperlipidemias/enzimologia , Fator de Necrose Tumoral alfa/fisiologia , Animais , Apolipoproteínas E/genética , Hepatite , Hiperlipidemias/genética , Hiperlipidemias/metabolismo , Camundongos , Camundongos Knockout
8.
FASEB J ; 23(6): 1946-57, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19211925

RESUMO

Omega-3-polyunsaturated fatty acids (omega-3-PUFAs) have well-documented protective effects that are attributed not only to eicosanoid inhibition but also to the formation of novel biologically active lipid mediators (i.e., resolvins and protectins). In this study, we examined their effects on ob/ob mice, an obesity model of insulin resistance and fatty liver disease. Dietary intake of omega-3-PUFAs had insulin-sensitizing actions in adipose tissue and liver and improved insulin tolerance in obese mice. Genes involved in insulin sensitivity (PPARgamma), glucose transport (GLUT-2/GLUT-4), and insulin receptor signaling (IRS-1/IRS-2) were up-regulated by omega-3-PUFAs. Moreover, omega-3-PUFAs increased adiponectin, an anti-inflammatory and insulin-sensitizing adipokine, and induced AMPK phosphorylation, a fuel-sensing enzyme and a gatekeeper of the energy balance. Concomitantly, hepatic steatosis was alleviated by omega-3-PUFAs. A lipidomic analysis with liquid chromatography/mass spectrometry/mass spectrometry revealed that omega-3-PUFAs inhibited the formation of omega-6-PUFA-derived eicosanoids, while triggering the formation of omega-3-PUFA-derived resolvins and protectins. Moreover, representative members of these lipid mediators, namely resolvin E1 and protectin D1, mimicked the insulin-sensitizing and antisteatotic effects of omega-3-PUFAs and induced adiponectin expression to a similar extent that of rosiglitazone, a member of the thiazolidinedione family of antidiabetic drugs. Taken together, these findings uncover beneficial actions of omega-3-PUFAs and their bioactive lipid autacoids in preventing obesity-induced insulin resistance and hepatic steatosis.


Assuntos
Ácidos Docosa-Hexaenoicos/metabolismo , Ácido Eicosapentaenoico/análogos & derivados , Ácidos Graxos Ômega-3 , Fígado Gorduroso/dietoterapia , Fígado Gorduroso/metabolismo , Resistência à Insulina , Obesidade , Proteínas Quinases Ativadas por AMP/metabolismo , Adiponectina/genética , Adiponectina/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Animais , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Dieta , Gorduras na Dieta/metabolismo , Ácido Eicosapentaenoico/metabolismo , Ácidos Graxos Ômega-3/química , Ácidos Graxos Ômega-3/metabolismo , Ácidos Graxos Ômega-3/uso terapêutico , Fígado Gorduroso/patologia , Transportador de Glucose Tipo 4/genética , Transportador de Glucose Tipo 4/metabolismo , Hipoglicemiantes/metabolismo , Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/metabolismo , Masculino , Camundongos , Camundongos Obesos , Músculo Esquelético/metabolismo , Obesidade/metabolismo , Obesidade/patologia , PPAR gama/genética , PPAR gama/metabolismo , Resistina/genética , Resistina/metabolismo , Rosiglitazona , Tiazolidinedionas/metabolismo
9.
Am J Physiol Gastrointest Liver Physiol ; 296(3): G553-62, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19136384

RESUMO

The contribution of metabolic factors to the severity of liver disease is not completely understood. In this study, apolipoprotein E-deficient (ApoE-/-) mice were evaluated to define potential effects of hypercholesterolemia on the severity of carbon tetrachloride (CCl4)-induced liver injury. Under baseline conditions, hypercholesterolemic ApoE-/- mice showed increased hepatic oxidative stress (SOD activity/4-hydroxy-2-nonenal immunostaining) and higher hepatic TGF-beta1, MCP-1, and TIMP-1 expression than wild-type control mice. After CCl4 challenge, ApoE-/- mice exhibited exacerbated steatosis (Oil Red O staining), necroinflammation (hematoxylin-eosin staining), macrophage infiltration (F4/80 immunohistochemistry), and fibrosis (Sirius red staining and alpha-smooth muscle actin immunohistochemistry) and more severe liver injury [alanine aminotransferase (ALT) and aspartate aminotransferase] than wild-type controls. Direct correlations were identified between serum cholesterol and hepatic steatosis, fibrosis, and ALT levels. These changes did not reflect the usual progression of the disease in ApoE-/- mice, since exacerbated liver injury was not present in untreated age-paired ApoE-/- mice. Moreover, hepatic cytochrome P-450 expression was unchanged in ApoE-/- mice. To explore potential mechanisms, cell types relevant to liver pathophysiology were exposed to selected cholesterol-oxidized products. Incubation of hepatocytes with a mixture of oxysterols representative of those detected by GC-MS in livers from ApoE-/- mice resulted in a concentration-dependent increase in total lipoperoxides and SOD activity. In hepatic stellate cells, oxysterols increased IL-8 secretion through a NF-kappaB-independent mechanism and upregulated TIMP-1 expression. In macrophages, oxysterols increased TGF-beta1 secretion and MCP-1 expression in a concentration-dependent manner. Oxysterols did not compromise cell viability. Taken together, these findings demonstrate that hypercholesterolemic mice are sensitized to liver injury and that cholesterol-derived products (i.e., oxysterols) are able to induce proinflammatory and profibrogenic mechanisms in liver cells.


Assuntos
Apolipoproteínas E/genética , Hipercolesterolemia/genética , Hipercolesterolemia/metabolismo , Hepatopatias/genética , Hepatopatias/metabolismo , Estresse Oxidativo/fisiologia , Animais , Tetracloreto de Carbono/toxicidade , Doença Hepática Induzida por Substâncias e Drogas , Quimiocina CCL2/metabolismo , Colesterol/metabolismo , Predisposição Genética para Doença , Células Estreladas do Fígado/metabolismo , Hidroxicolesteróis/metabolismo , Interleucina-8/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , NF-kappa B/metabolismo , Superóxido Dismutase/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
10.
Gastroenterol. hepatol. (Ed. impr.) ; 31(10): 682-692, dic. 2008. ilus, tab
Artigo em Es | IBECS | ID: ibc-71557

RESUMO

La presencia de lesión en el parénquima celular es común a un gran número de enfermedades crónicas del hígado, como por ejemplo las hepatitis virales, la hepatitis alcohólica, las colestasis crónicas o la esteatohepatitis. Aunque la patogenia puede variar según el agente etiológico, hay una serie de mecanismos comunes a todas ellas. Entre estos mecanismos destacan la activación de las células de Kupffer y el reclutamiento de células inflamatorias, la formación de radicales libres del oxígeno y la aparición de estrés oxidativo, la producción de citocinas, principalmente del factor de necrosis tumoral alfa y el factor de crecimiento transformante beta, y la liberación de mediadores de inflamación derivados de la oxidación del ácido araquidónico a través de la ciclooxigenasa 2 y la 5-lipooxigenasa


The presence of a lesion in the cellular parenchyma is common to a large number of chronic liver diseases, such as viral hepatitides, alcoholic hepatitis, chronic cholestasis and steatohepatitis. Although the pathogenesis may vary according to the etiological agent, a series of mechanisms is common to all. Notable among these mechanisms are Kupffer cell activation and inflammatory cell recruitment, free oxygen radical formation and the development of oxidative stress, cytokine production, mainly TNF and TGF , andinflammatory mediator release due to arachidonic acid oxidation through the COX-2 and 5-LO pathways (AU)


Assuntos
Humanos , Hepatopatias/patologia , Hepatopatias/imunologia , Células de Kupffer/patologia , Mediadores da Inflamação , Inflamação/patologia , Estresse Oxidativo , Citocinas/biossíntese , Doença Crônica , Espécies Reativas de Oxigênio , Radicais Livres
11.
J Lipid Res ; 49(12): 2513-23, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18645210

RESUMO

As 5-lipoxygenase (5-LO) is an emerging target in obesity and insulin resistance, we have investigated whether this arachidonate pathway is also implicated in the progression of obesity-related fatty liver disease. Our results show that 5-LO activity and 5-LO-derived product levels are significantly elevated in the liver of obese ob/ob mice with respect to wild-type controls. Treatment of ob/ob mice with a selective 5-LO inhibitor exerted a remarkable protection from hepatic steatosis as revealed by decreased oil red-O staining and reduced hepatic triglyceride (TG) concentrations. In addition, 5-LO inhibition in ob/ob mice downregulated genes involved in hepatic fatty acid uptake (i.e., L-FABP and FAT/CD36) and normalized peroxisome proliferator-activated receptor alpha (PPARalpha) and acyl-CoA oxidase expression, whereas the expression of lipogenic genes [i.e., fatty acid synthase (FASN) and SREBP-1c] remained unaltered. Furthermore, 5-LO inhibition restored hepatic microsomal TG transfer protein (MTP) activity in parallel with a stimulation of hepatic VLDL-TG and apoB secretion in ob/ob mice. Consistent with these findings, 5-LO products directly inhibited MTP activity and triggered cytosolic TG accumulation in CC-1 cells, a murine hepatocyte cell line. Taken together, these findings identify a novel steatogenic role for 5-LO in the liver through mechanisms involving the regulation of hepatic MTP activity and VLDL-TG and apoB secretion.


Assuntos
Apolipoproteínas B/metabolismo , Araquidonato 5-Lipoxigenase/metabolismo , Proteínas de Transporte/metabolismo , Lipoproteínas VLDL/metabolismo , Fígado/enzimologia , Obesidade/enzimologia , Triglicerídeos/metabolismo , Animais , Células Cultivadas , Fígado/metabolismo , Masculino , Camundongos , Camundongos Obesos , Obesidade/metabolismo , Ratos
12.
Gastroenterol Hepatol ; 31(10): 682-92, 2008 Dec.
Artigo em Espanhol | MEDLINE | ID: mdl-19174085

RESUMO

The presence of a lesion in the cellular parenchyma is common to a large number of chronic liver diseases, such as viral hepatitides, alcoholic hepatitis, chronic cholestasis and steatohepatitis. Although the pathogenesis may vary according to the etiological agent, a series of mechanisms is common to all. Notable among these mechanisms are Kupffer cell activation and inflammatory cell recruitment, free oxygen radical formation and the development of oxidative stress, cytokine production, mainly TNFa and TGFb, and inflammatory mediator release due to arachidonic acid oxidation through the COX-2 and 5-LO pathways.


Assuntos
Hepatopatias/etiologia , Animais , Citocinas/fisiologia , Humanos , Inflamação/complicações , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Células de Kupffer/fisiologia , Lipoxigenase/fisiologia , Hepatopatias/enzimologia , Hepatopatias/imunologia , Prostaglandina-Endoperóxido Sintases/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...