Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Lipid Res ; 62: 100093, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34153284

RESUMEN

The liver is the central organ regulating cholesterol synthesis, storage, transport, and elimination. Mouse carboxylesterase 1d (Ces1d) and its human ortholog CES1 have been described to possess lipase activity and play roles in hepatic triacylglycerol metabolism and VLDL assembly. It has been proposed that Ces1d/CES1 might also catalyze cholesteryl ester (CE) hydrolysis in the liver and thus be responsible for the hydrolysis of HDL-derived CE; this could contribute to the final step in the reverse cholesterol transport (RCT) pathway, wherein cholesterol is secreted from the liver into bile and feces, either directly or after conversion to water-soluble bile salts. However, the proposed function of Ces1d/CES1 as a CE hydrolase is controversial. In this study, we interrogated the role hepatic Ces1d plays in cholesterol homeostasis using liver-specific Ces1d-deficient mice. We rationalized that if Ces1d is a major hepatic CE hydrolase, its absence would (1) reduce in vivo RCT flux and (2) provoke liver CE accumulation after a high-cholesterol diet challenge. We found that liver-specific Ces1d-deficient mice did not show any difference in the flux of in vivo HDL-to-feces RCT nor did it cause additional liver CE accumulation after high-fat, high-cholesterol Western-type diet feeding. These findings challenge the importance of Ces1d as a major hepatic CE hydrolase.


Asunto(s)
Ésteres del Colesterol/metabolismo , Hígado/metabolismo , Animales , Carboxilesterasa/deficiencia , Carboxilesterasa/metabolismo , Células Cultivadas , Hidrólisis , Ratones , Ratones Noqueados , Ratones Transgénicos
2.
Chem Res Toxicol ; 34(6): 1556-1571, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-33900070

RESUMEN

Chlorpyrifos (CPF) is an organophosphate (OP) pesticide that causes acute toxicity by inhibiting acetylcholinesterase (AChE) in the nervous system. However, endocannabinoid (eCB) metabolizing enzymes in brain of neonatal rats are more sensitive than AChE to inhibition by CPF, leading to increased levels of eCBs. Because eCBs are immunomodulatory molecules, we investigated the association between eCB metabolism, lipid mediators, and immune function in adult and neonatal mice exposed to CPF. We focused on lung effects because epidemiologic studies have linked pesticide exposures to respiratory diseases. CPF was hypothesized to disrupt lung eCB metabolism and alter lung immune responses to lipopolysaccharide (LPS), and these effects would be more pronounced in neonatal mice due to an immature immune system. We first assessed the biochemical effects of CPF in adult mice (≥8 weeks old) and neonatal mice after administering CPF (2.5 mg/kg, oral) or vehicle for 7 days. Tissues were harvested 4 h after the last CPF treatment and lung microsomes from both age groups demonstrated CPF-dependent inhibition of carboxylesterases (Ces), a family of xenobiotic and lipid metabolizing enzymes, whereas AChE activity was inhibited in adult lungs only. Activity-based protein profiling (ABPP)-mass spectrometry of lung microsomes identified 31 and 32 individual serine hydrolases in neonatal lung and adult lung, respectively. Of these, Ces1c/Ces1d/Ces1b isoforms were partially inactivated by CPF in neonatal lung, whereas Ces1c/Ces1b and Ces1c/BChE were partially inactivated in adult female and male lungs, respectively, suggesting age- and sex-related differences in their sensitivity to CPF. Monoacylglycerol lipase (MAGL) and fatty acid amide hydrolase (FAAH) activities in lung were unaffected by CPF. When LPS (1.25 mg/kg, i.p.) was administered following the 7-day CPF dosing period, little to no differences in lung immune responses (cytokines and immunophenotyping) were noted between the CPF and vehicle groups. However, a CPF-dependent increase in the amounts of dendritic cells and certain lipid mediators in female lung following LPS challenge was observed. Experiments in neonatal and adult Ces1d-/- mice yielded similar results as wild type mice (WT) following CPF treatment, except that CPF augmented LPS-induced Tnfa mRNA in adult Ces1d-/- mouse lungs. This effect was associated with decreased expression of Ces1c mRNA in Ces1d-/- mice versus WT mice in the setting of LPS exposure. We conclude that CPF exposure inactivates several Ces isoforms in mouse lung and, during an inflammatory response, increases certain lipid mediators in a female-dependent manner. However, it did not cause widespread altered lung immune effects in response to an LPS challenge.


Asunto(s)
Cloropirifos/farmacología , Inhibidores Enzimáticos/farmacología , Hidrolasas/antagonistas & inhibidores , Metabolismo de los Lípidos/efectos de los fármacos , Pulmón/efectos de los fármacos , Serina/antagonistas & inhibidores , Animales , Cloropirifos/química , Inhibidores Enzimáticos/química , Hidrolasas/inmunología , Pulmón/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estructura Molecular , Serina/inmunología
3.
J Lipid Res ; 60(4): 880-891, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30737251

RESUMEN

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.


Asunto(s)
Carboxilesterasa/metabolismo , Dieta Alta en Grasa/efectos adversos , Hígado/metabolismo , Sacarosa/antagonistas & inhibidores , Triglicéridos/metabolismo , Animales , Carboxilesterasa/deficiencia , Hígado/química , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sacarosa/administración & dosificación , Sacarosa/efectos adversos
4.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(7): 688-699, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29631096

RESUMEN

Obesity often leads non-alcoholic fatty liver disease, insulin resistance and hyperlipidemia. Expression of carboxylesterase CES1 is positively correlated with increased lipid storage and plasma lipid concentration. Here we investigated structural and metabolic consequences of a single nucleotide polymorphism in CES1 gene that results in p.Gly143Glu amino acid substitution. We generated a humanized mouse model expressing CES1WT (control), CES1G143E and catalytically dead CES1S221A (negative control) in the liver in the absence of endogenous expression of the mouse orthologous gene. We show that the CES1G143E variant exhibits only 20% of the wild-type lipolytic activity. High-fat diet fed mice expressing CES1G143E had reduced liver and plasma triacylglycerol levels. The mechanism by which decreased CES1 activity exerts this hypolipidemic phenotype was determined to include decreased very-low density lipoprotein secretion, decreased expression of hepatic lipogenic genes and increased fatty acid oxidation as determined by increased plasma ketone bodies and hepatic mitochondrial electron transport chain protein abundance. We conclude that attenuation of human CES1 activity provides a beneficial effect on hepatic lipid metabolism. These studies also suggest that CES1 is a potential therapeutic target for non-alcoholic fatty liver disease management.


Asunto(s)
Hidrolasas de Éster Carboxílico/genética , Predisposición Genética a la Enfermedad , Hígado/patología , Enfermedad del Hígado Graso no Alcohólico/genética , Obesidad/metabolismo , Animales , Hidrolasas de Éster Carboxílico/metabolismo , Quimera/genética , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Femenino , Humanos , Metabolismo de los Lípidos/genética , Lípidos/sangre , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedad del Hígado Graso no Alcohólico/sangre , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Obesidad/sangre , Obesidad/etiología , Polimorfismo de Nucleótido Simple
5.
J Biol Chem ; 291(4): 1974-1990, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26627833

RESUMEN

Cytopathic effects are currently believed to contribute to hepatitis C virus (HCV)-induced liver injury and are readily observed in Huh7.5 cells infected with the JFH-1 HCV strain, manifesting as apoptosis highly correlated with growth arrest. Reactive oxygen species, which are induced by HCV infection, have recently emerged as activators of AMP-activated protein kinase. The net effect is ATP conservation via on/off switching of metabolic pathways that produce/consume ATP. Depending on the scenario, this can have either pro-survival or pro-apoptotic effects. We demonstrate reactive oxygen species-mediated activation of AMP-activated kinase in Huh7.5 cells during HCV (JFH-1)-induced growth arrest. Metabolic labeling experiments provided direct evidence that lipid synthesis is attenuated, and ß-oxidation is enhanced in these cells. A striking increase in nuclear peroxisome proliferator-activated receptor α, which plays a dominant role in the expression of ß-oxidation genes after ligand-induced activation, was also observed, and we provide evidence that peroxisome proliferator-activated receptor α is constitutively activated in these cells. The combination of attenuated lipid synthesis and enhanced ß-oxidation is not conducive to lipid accumulation, yet cellular lipids still accumulated during this stage of infection. Notably, the serum in the culture media was the only available source for polyunsaturated fatty acids, which were elevated (2-fold) in the infected cells, implicating altered lipid import/export pathways in these cells. This study also provided the first in vivo evidence for enhanced ß-oxidation during HCV infection because HCV-infected SCID/Alb-uPA mice accumulated higher plasma ketones while fasting than did control mice. Overall, this study highlights the reprogramming of hepatocellular lipid metabolism and bioenergetics during HCV infection, which are predicted to impact both the HCV life cycle and pathogenesis.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Ácidos Grasos/metabolismo , Hepacivirus/fisiología , Hepatitis C/metabolismo , Lípidos/biosíntesis , Neoplasias Hepáticas/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/virología , Hepatitis C/virología , Humanos , Hígado/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/virología , Ratones , Ratones SCID , Mitocondrias/genética , Oxidación-Reducción , PPAR alfa/genética , PPAR alfa/metabolismo
6.
Biochim Biophys Acta ; 1861(5): 482-90, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26976727

RESUMEN

Ces1g/Es-x deficiency in mice results in weight gain, insulin resistance, fatty liver and hyperlipidemia through upregulation of de novo lipogenesis and oversecretion of triacylglycerol (TG)-rich lipoproteins. Here, we show that restoration of Ces1g/Es-x expression only in the liver significantly reduced hepatic TG concentration accompanied by decreased size of lipid droplets, reduced secretion of very low-density lipoproteins and improved insulin-mediated signal transduction in the liver. Collectively, these results demonstrate that hepatic Ces1g/Es-x plays a critical role in limiting hepatic steatosis, very low-density lipoprotein assembly and in augmenting insulin sensitivity.


Asunto(s)
Hidrolasas de Éster Carboxílico/metabolismo , Hígado Graso/prevención & control , Terapia Genética , Hiperlipidemias/prevención & control , Resistencia a la Insulina , Insulina/sangre , Metabolismo de los Lípidos , Hígado/enzimología , Animales , Glucemia/metabolismo , Hidrolasas de Éster Carboxílico/deficiencia , Hidrolasas de Éster Carboxílico/genética , Modelos Animales de Enfermedad , Hígado Graso/sangre , Hígado Graso/enzimología , Hígado Graso/genética , Femenino , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Predisposición Genética a la Enfermedad , Hiperlipidemias/sangre , Hiperlipidemias/enzimología , Hiperlipidemias/genética , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos/genética , Lipoproteínas VLDL/sangre , Ratones Noqueados , Fenotipo , Transducción de Señal , Factores de Tiempo , Triglicéridos/sangre
7.
Br J Cancer ; 117(1): 148-155, 2017 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-28588319

RESUMEN

BACKGROUND: Prognostic significance of adiposity, at the time of cancer diagnosis, on survival is not clear. Body mass index (kg m-2) does not provide an appropriate assessment of body composition; therefore, the concept of the 'obesity paradox' needs to be investigated based on the prognostic significance of fat and muscle. Independent prognostic significance of adipose tissue in predicting mortality, importance of visceral and subcutaneous adiposity in the presence and absence of sarcopenia on survival, was investigated. METHODS: Adiposity markers including total adipose index (TATI), visceral adipose tissue index (VATI) and subcutaneous adipose tissue index (SATI) were estimated for 1473 gastrointestinal and respiratory cancer patients and 273 metastatic renal cell carcinoma patients using computed tomography. Univariate and multivariate analysis to determine mortality hazard ratios (HR) were conducted using cox proportional hazard models. RESULTS: Low SATI (SATI <50.0 cm2 m-2 in males and <42.0 cm2 m-2 in females) independently associated with increased mortality (HR: 1.26; 95% CI: 1.11-1.43; P<0.001) and shorter survival (13.1 months; 95% CI, 11.4-14.7) compared to patients with high SATI (19.3 months; 95% CI, 17.6-21.0; P<0.001). In the presence of sarcopenia, the longest survival was observed in patients with high subcutaneous adiposity. CONCLUSIONS: Subcutaneous adipose tissues appear to associate with reduction in mortality risk demonstrating the prognostic importance of fat distribution. The effect of sarcopenia on survival was more pronounced in patients with low subcutaneous adiposity.


Asunto(s)
Grasa Intraabdominal/diagnóstico por imagen , Neoplasias/mortalidad , Obesidad Abdominal/epidemiología , Sarcopenia/epidemiología , Grasa Subcutánea/diagnóstico por imagen , Adiposidad , Anciano , Alberta/epidemiología , Carcinoma de Células Renales/mortalidad , Femenino , Neoplasias Gastrointestinales/mortalidad , Humanos , Neoplasias Renales/mortalidad , Masculino , Persona de Mediana Edad , Análisis Multivariante , Obesidad Abdominal/diagnóstico por imagen , Pronóstico , Modelos de Riesgos Proporcionales , Factores Protectores , Neoplasias del Sistema Respiratorio/mortalidad , Factores de Riesgo , Sarcopenia/diagnóstico por imagen , Tomografía Computarizada por Rayos X
8.
Lipids Health Dis ; 16(1): 247, 2017 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-29258509

RESUMEN

BACKGROUND: Accelerated loss of adipose tissue in cancer is associated with shorter survival, and reduced quality of life. Evidence is emerging suggesting tumour association with alterations in adipose tissue, but much less is known about drug-related mechanisms contributing to adipose atrophy. Identification of mechanisms by which tumour and cancer treatments, such as chemotherapy, affect adipose tissue are required to develop appropriate therapeutic interventions to prevent fat depletion in cancer. This pre-clinical study aimed to assess alterations in adipose tissue during the clinical course of cancer. METHODS: Fischer 344 rats bearing the Ward colorectal tumour were euthanized before chemotherapy, after 1- cycle, or 2-cycles of a combination chemotherapy consisting of Irinotecan (CPT-11) combined with 5-fluorouracil (5-FU), which recapitulates first line treatment for human colorectal cancer. Periuterine adipose tissue was isolated. Healthy rats served as a reference group. Histological analysis (hematoxylin and eosin), Real-time PCR (TaqMan) and proteomic analysis (LC-MS/MS) were performed. RESULTS: Larger adipocytes (3993.7 ± 52.6 µm2) in tumour-bearing animals compared to the reference group (3227.7 ± 36.7 µm2; p < 0.001) was associated with reduced expression of proteins involved in mitochondrial fatty acid oxidation. The presence of a tumour has a significant effect on phospholipid but not triglyceride fatty acid composition. There were greater proportions of saturated fatty acids concurrent with lower monounsaturated fatty acids within the PL fraction of adipocytes in tumour-bearing animals. Chemotherapy treatment decreased the size of adipocytes (2243.9 ± 30.4 µm2; p < 0.001) and led to depletion of n-3 polyunsaturated fatty acids in adipose tissue triglyceride. Evaluation of the proteome profile revealed decreased expression of proteins involved in ATP generation, ß-oxidation, and lipogenesis. Overall, adipose tissue may not be able to efficiently oxidize fatty acids to provide energy to maintain energy demanding pathways like lipogenesis inside the tissue. CONCLUSIONS: In conclusion, metabolic adaptations to mitochondrial impairment may contribute to diminished lipid storage capacity of adipose tissue following chemotherapy delivery.


Asunto(s)
Adipocitos/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias del Colon/metabolismo , Lipogénesis/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Adipocitos/metabolismo , Adipocitos/patología , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Camptotecina/análogos & derivados , Camptotecina/farmacología , Tamaño de la Célula , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Ácidos Grasos/agonistas , Ácidos Grasos/metabolismo , Ácidos Grasos Monoinsaturados/antagonistas & inhibidores , Ácidos Grasos Monoinsaturados/metabolismo , Ácidos Grasos Omega-3/antagonistas & inhibidores , Ácidos Grasos Omega-3/metabolismo , Femenino , Fluorouracilo/farmacología , Humanos , Irinotecán , Lipogénesis/genética , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Fosfolípidos/metabolismo , Proteoma/antagonistas & inhibidores , Proteoma/genética , Proteoma/metabolismo , Ratas , Ratas Endogámicas F344 , Triglicéridos/metabolismo
9.
Am J Physiol Gastrointest Liver Physiol ; 310(7): G526-38, 2016 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-26797396

RESUMEN

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.


Asunto(s)
Antiinfecciosos/farmacología , Hepatitis/prevención & control , Cirrosis Hepática Experimental/prevención & control , Hígado/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/prevención & control , PPAR gamma/agonistas , Fosfatidiletanolamina N-Metiltransferasa/deficiencia , Tiazolidinedionas/farmacología , Actinas/genética , Actinas/metabolismo , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/enzimología , Adipocitos Blancos/patología , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/enzimología , Tejido Adiposo Blanco/patología , Adiposidad/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Dieta Alta en Grasa , Predisposición Genética a la Enfermedad , Hepatitis/enzimología , Hepatitis/genética , Hepatitis/patología , Resistencia a la Insulina , Hígado/enzimología , Hígado/patología , Cirrosis Hepática Experimental/enzimología , Cirrosis Hepática Experimental/genética , Cirrosis Hepática Experimental/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/enzimología , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/patología , Obesidad/enzimología , Obesidad/genética , Obesidad/prevención & control , Estrés Oxidativo/efectos de los fármacos , PPAR gamma/metabolismo , Fenotipo , Fosfatidiletanolamina N-Metiltransferasa/genética , Pioglitazona , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 35(5): 1080-91, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25792450

RESUMEN

OBJECTIVE: Very low-density lipoprotein assembly and secretion are regulated by the availability of triacylglycerol. Although compelling evidence indicates that the majority of triacylglycerol in very low-density lipoprotein is derived from re-esterification of lipolytic products released by endoplasmic reticulum-associated lipases, little is known about roles of acyl-CoA:diacylglycerol acyltransferases (DGATs) in this process. We aimed to investigate the contribution of DGAT1 and DGAT2 in lipid metabolism and lipoprotein secretion in primary mouse and human hepatocytes. APPROACH AND RESULTS: We used highly selective small-molecule inhibitors of DGAT1 and DGAT2, and we tracked storage and secretion of lipids synthesized de novo from [(3)H]acetic acid and from exogenously supplied [(3)H]oleic acid. Inactivation of individual DGAT activity did not affect incorporation of either radiolabeled precursor into intracellular triacylglycerol, whereas combined inactivation of both DGATs severely attenuated triacylglycerol synthesis. However, inhibition of DGAT2 augmented fatty acid oxidation, whereas inhibition of DGAT1 increased triacylglycerol secretion, suggesting preferential channeling of separate DGAT-derived triacylglycerol pools to distinct metabolic pathways. Inactivation of DGAT2 impaired cytosolic lipid droplet expansion, whereas DGAT1 inactivation promoted large lipid droplet formation. Moreover, inactivation of DGAT2 attenuated expression of lipogenic genes. Finally, triacylglycerol secretion was significantly reduced on DGAT2 inhibition without altering extracellular apolipoprotein B levels. CONCLUSIONS: Our data suggest that DGAT1 and DGAT2 can compensate for each other to synthesize triacylglycerol, but triacylglycerol synthesized by DGAT1 is preferentially channeled to oxidation, whereas DGAT2 synthesizes triacylglycerol destined for very low-density lipoprotein assembly.


Asunto(s)
Diacilglicerol O-Acetiltransferasa/metabolismo , Hepatocitos/enzimología , Triglicéridos/biosíntesis , Acilcoenzima A/metabolismo , Animales , Células Cultivadas , Diacilglicerol O-Acetiltransferasa/genética , Hepatocitos/metabolismo , Humanos , Metabolismo de los Lípidos/fisiología , Lipogénesis/fisiología , Ratones , Rol , Sensibilidad y Especificidad
11.
Biochim Biophys Acta ; 1831(6): 1113-23, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23376223

RESUMEN

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.


Asunto(s)
Retículo Endoplásmico/enzimología , Hepatocitos/enzimología , Lipasa/metabolismo , Lipoproteínas VLDL/metabolismo , Hígado/enzimología , Triglicéridos/metabolismo , Animales , Apolipoproteínas B/metabolismo , Células Cultivadas , Ésteres del Colesterol/metabolismo , Ácidos Grasos/metabolismo , Hepatocitos/citología , Metabolismo de los Lípidos , Hígado/citología , Ratones , Oxidación-Reducción , PPAR alfa/metabolismo
12.
Breast Cancer Res Treat ; 143(2): 301-12, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24337703

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Resistencia a Antineoplásicos/genética , Ácidos Grasos Monoinsaturados/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Indoles/farmacología , Antineoplásicos/farmacología , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Receptor alfa de Estrógeno/biosíntesis , Femenino , Fluvastatina , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Hidroximetilglutaril-CoA-Reductasas NADP-Dependientes/biosíntesis , Células MCF-7 , Ácido Mevalónico/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , ARN Mensajero/biosíntesis , Receptor ErbB-2
13.
Circ Res ; 111(8): 982-90, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22872154

RESUMEN

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.


Asunto(s)
Enfermedad de la Arteria Coronaria/metabolismo , Dislipidemias/metabolismo , Lipasa/genética , Receptores de LDL/genética , Animales , Aorta/metabolismo , Aorta/patología , Apolipoproteínas B/sangre , Glucemia/metabolismo , HDL-Colesterol/sangre , LDL-Colesterol/sangre , VLDL-Colesterol/sangre , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/patología , Modelos Animales de Enfermedad , Dislipidemias/genética , Ácidos Grasos/metabolismo , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado Graso/patología , Femenino , Insulina/sangre , Resistencia a la Insulina/fisiología , Lipasa/deficiencia , Lipasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Receptores de LDL/metabolismo
14.
Biochim Biophys Acta ; 1821(5): 762-9, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21963564

RESUMEN

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.


Asunto(s)
Hepatocitos/enzimología , Lipasa , Hígado/enzimología , Triglicéridos , Animales , Autofagia , Diabetes Mellitus Tipo 2/enzimología , Esterasas/metabolismo , Hígado Graso/enzimología , Hígado Graso/etiología , Humanos , Lipasa/clasificación , Lipasa/metabolismo , Cirrosis Hepática/enzimología , Enfermedad del Hígado Graso no Alcohólico , Triglicéridos/biosíntesis , Triglicéridos/metabolismo
15.
J Hepatol ; 59(2): 336-43, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23542347

RESUMEN

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.


Asunto(s)
Hidrolasas de Éster Carboxílico/metabolismo , Hepacivirus/fisiología , Lipoproteínas VLDL/metabolismo , Triglicéridos/metabolismo , Apolipoproteínas B/metabolismo , Hidrolasas de Éster Carboxílico/antagonistas & inhibidores , Hidrolasas de Éster Carboxílico/genética , Línea Celular , Técnicas de Silenciamiento del Gen , Hepacivirus/crecimiento & desarrollo , Hepacivirus/patogenicidad , Interacciones Huésped-Patógeno , Humanos , Lipólisis , Modelos Biológicos , Virulencia , Replicación Viral
16.
Hepatology ; 56(6): 2188-98, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22806626

RESUMEN

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.


Asunto(s)
Hidrolasas de Éster Carboxílico/deficiencia , Colesterol/metabolismo , Hígado Graso/enzimología , Hiperlipidemias/enzimología , Obesidad/enzimología , Análisis de Varianza , Animales , Glucemia/metabolismo , Hidrolasas de Éster Carboxílico/genética , Hidrolasas de Éster Carboxílico/metabolismo , Ácidos Docosahexaenoicos/metabolismo , Metabolismo Energético , Ayuno , Ácidos Grasos Insaturados/administración & dosificación , Ácidos Grasos Insaturados/metabolismo , Hígado Graso/genética , Femenino , Aceites de Pescado/administración & dosificación , Expresión Génica , Hepatocitos/metabolismo , Hiperlipidemias/genética , Insulina/sangre , Lipoproteínas VLDL/metabolismo , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico , Obesidad/genética , Fenotipo , Fosfolípidos/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Triglicéridos/metabolismo
17.
Hepatology ; 56(6): 2154-62, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22707181

RESUMEN

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.


Asunto(s)
VLDL-Colesterol/sangre , Lipasa/deficiencia , Lipoproteínas VLDL/sangre , Hígado/enzimología , Hígado/metabolismo , Triglicéridos/sangre , Animales , Colesterol en la Dieta/administración & dosificación , VLDL-Colesterol/metabolismo , Grasas de la Dieta/administración & dosificación , Ácidos Grasos/metabolismo , Hígado Graso/metabolismo , Femenino , Glucosa/biosíntesis , Prueba de Tolerancia a la Glucosa , Insulina/metabolismo , Resistencia a la Insulina , Cuerpos Cetónicos/sangre , Lipasa/genética , Lipoproteínas VLDL/metabolismo , Masculino , Ratones , Ratones Noqueados , Transducción de Señal , Triglicéridos/metabolismo
18.
Arterioscler Thromb Vasc Biol ; 32(5): 1087-93, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22517367

RESUMEN

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.


Asunto(s)
Metabolismo de los Lípidos , Lípidos/biosíntesis , Lipoproteínas/biosíntesis , Hígado/metabolismo , Animales , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Retículo Endoplásmico/metabolismo , Humanos , Hígado/citología
19.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1868(10): 159376, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37516308

RESUMEN

Two distinct diacylglycerol acyltransferases (DGAT1 and DGAT2) catalyze the final committed step of triacylglycerol (TG) synthesis in hepatocytes. After its synthesis in the endoplasmic reticulum (ER) TG is either stored in cytosolic lipid droplets (LDs) or is assembled into very low-density lipoproteins in the ER lumen. TG stored in cytosolic LDs is hydrolyzed by adipose triglyceride lipase (ATGL) and the released fatty acids are converted to energy by oxidation in mitochondria. We hypothesized that targeting/association of ATGL to LDs would differ depending on whether the TG stores were generated through DGAT1 or DGAT2 activities. Individual inhibition of DGAT1 or DGAT2 in Huh7 hepatocytes incubated with oleic acid did not yield differences in TG accretion while combined inhibition of both DGATs completely prevented TG synthesis suggesting that either DGAT can efficiently esterify exogenously supplied fatty acid. DGAT2-made TG was stored in larger LDs, whereas TG formed by DGAT1 accumulated in smaller LDs. Inactivation of DGAT1 or DGAT2 did not alter expression (mRNA or protein) of ATGL, the ATGL activator ABHD5/CGI-58, or LD coat proteins PLIN2 or PLIN5, but inactivation of both DGATs increased PLIN2 abundance despite a dramatic reduction in the number of LDs. ATGL was found to preferentially target to LDs generated by DGAT1 and fatty acids released from TG in these LDs were also preferentially used for fatty acid oxidation. Combined inhibition of DGAT2 and ATGL resulted in larger LDs, suggesting that the smaller size of DGAT1-generated LDs is the result of increased lipolysis of TG in these LDs.


Asunto(s)
Hepatocitos , Lipólisis , Diacilglicerol O-Acetiltransferasa/genética , Diacilglicerol O-Acetiltransferasa/metabolismo , Ácidos Grasos/metabolismo , Hepatocitos/metabolismo , Triglicéridos/metabolismo
20.
Cell Mol Gastroenterol Hepatol ; 15(2): 281-306, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36243320

RESUMEN

BACKGROUND & AIMS: Binge drinking in patients with metabolic syndrome accelerates the development of alcohol-associated liver disease. However, the underlying mechanisms remain elusive. We investigated if oxidative and nonoxidative alcohol metabolism pathways, diet-induced obesity, and adipose tissues influenced the development of acute liver injury in a single ethanol binge model. METHODS: A single ethanol binge was administered to chow-fed or high-fat diet (HFD)-fed wild-type and genetically modified mice. RESULTS: Oral administration of a single dose of ethanol induced acute liver injury and hepatic endoplasmic reticulum (ER) stress in chow- or HFD-fed mice. Disruption of the Adh1 gene increased blood ethanol concentration and exacerbated acute ethanol-induced ER stress and liver injury in both chow-fed and HFD-fed mice, while disruption of the Aldh2 gene did not affect such hepatic injury despite high blood acetaldehyde levels. Mechanistic studies showed that alcohol, not acetaldehyde, promoted hepatic ER stress, fatty acid synthesis, and increased adipocyte death and lipolysis, contributing to acute liver injury. Increased serum fatty acid ethyl esters (FAEEs), which are formed by an enzyme-mediated esterification of ethanol with fatty acids, were detected in mice after ethanol gavage, with higher levels in Adh1 knockout mice than in wild-type mice. Deletion of the Ces1d gene in mice markedly reduced the acute ethanol-induced increase of blood FAEE levels with a slight but significant reduction of serum aminotransferase levels. CONCLUSIONS: Ethanol and its nonoxidative metabolites, FAEEs, not acetaldehyde, promoted acute alcohol-induced liver injury by inducing ER stress, adipocyte death, and lipolysis.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Estrés del Retículo Endoplásmico , Etanol , Lipólisis , Animales , Ratones , Acetaldehído/metabolismo , Adipocitos/metabolismo , Ésteres/metabolismo , Etanol/toxicidad , Ácidos Grasos/metabolismo , Hígado/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA