Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 39(9): 1776-1786, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31340670

RESUMEN

OBJECTIVE: Air pollution is associated with increased cardiovascular morbidity and mortality, as well as dyslipidemia and metabolic syndrome. Our goal was to dissect the mechanisms involved. Approach and Results: We assessed the effects of exposure to air pollution on lipid metabolism in mice through assessment of plasma lipids and lipoproteins, oxidized fatty acids 9-HODE (9-hydroxyoctadecadienoic) and 13-HODE (13-hydroxyoctadecadienoic), lipid, and carbohydrate metabolism. Findings were corroborated, and mechanisms were further assessed in HepG2 hepatocytes in culture. ApoE knockout mice exposed to inhaled diesel exhaust (DE, 6 h/d, 5 days/wk for 16 weeks) exhibited elevated plasma cholesterol and triglyceride levels, increased hepatic triglyceride content, and higher hepatic levels of 9-HODE and 13-HODE, as compared to control mice exposed to filtered air. A direct effect of DE exposure on hepatocytes was demonstrated by treatment of HepG2 cells with a methanol extract of DE particles followed by loading with oleic acid. As observed in vivo, this led to increased triglyceride content and significant downregulation of ACAD9 mRNA expression. Treatment of HepG2 cells with DE particles and oleic acid did not alter de novo lipogenesis but inhibited total, mitochondrial, and ATP-linked oxygen consumption rate, indicative of mitochondrial dysfunction. Treatment of isolated mitochondria, prepared from mouse liver, with DE particles and oleic acid also inhibited mitochondrial complex activity and ß-oxidation. CONCLUSIONS: DE exposure leads to dyslipidemia and liver steatosis in ApoE knockout mice, likely due to mitochondrial dysfunction and decreased lipid catabolism.


Asunto(s)
Hígado Graso/inducido químicamente , Hiperlipidemias/inducido químicamente , Mitocondrias/metabolismo , Emisiones de Vehículos/toxicidad , Animales , Células Hep G2 , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Triglicéridos/metabolismo
2.
Arterioscler Thromb Vasc Biol ; 39(6): 1045-1054, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31070450

RESUMEN

Objective- FMO (flavin-containing monooxygenase) 3 converts bacterial-derived trimethylamine to trimethylamine N-oxide (TMAO), an independent risk factor for cardiovascular disease. We generated FMO3 knockout (FMO3KO) mouse to study its effects on plasma TMAO, lipids, glucose/insulin metabolism, thrombosis, and atherosclerosis. Approach and Results- Previous studies with an antisense oligonucleotide (ASO) knockdown strategy targeting FMO3 in LDLRKO (low-density lipoprotein receptor knockout) mice resulted in major reductions in TMAO levels and atherosclerosis, but also showed effects on plasma lipids, insulin, and glucose. Although FMO3KO mice generated via CRISPR/Cas9 technology bred onto the LDLRKO background did exhibit similar effects on TMAO levels, the effects on lipid metabolism were not as pronounced as with the ASO knockdown model. These differences could result from either off-target effects of the ASO or from a developmental adaptation to the FMO3 deficiency. To distinguish these possibilities, we treated wild-type and FMO3KO mice with control or FMO3 ASOs. FMO3-ASO treatment led to the same extent of lipid-lowering effects in the FMO3KO mice as the wild-type mice, indicating off-target effects. The levels of TMAO in LDLRKO mice fed an atherogenic diet are very low in both wild-type and FMO3KO mice, and no significant effect was observed on atherosclerosis. When FMO3KO and wild-type mice were maintained on a 0.5% choline diet, FMO3KO showed a marked reduction in both TMAO and in vivo thrombosis potential. Conclusions- FMO3KO markedly reduces systemic TMAO levels and thrombosis potential. However, the previously observed large effects of an FMO3 ASO on plasma lipid levels appear to be due partly to off-target effects.


Asunto(s)
Aterosclerosis/metabolismo , Colina/metabolismo , Metilaminas/metabolismo , Oxigenasas/genética , Trombosis/metabolismo , Animales , Aterosclerosis/genética , Colina/farmacología , Modelos Animales de Enfermedad , Metabolismo de los Lípidos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxigenasas/metabolismo , Reacción en Cadena de la Polimerasa/métodos , Distribución Aleatoria , Valores de Referencia , Trombosis/fisiopatología
3.
Biochem Biophys Res Commun ; 505(1): 87-92, 2018 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-30241945

RESUMEN

The mammalian paraoxonases (PONs 1, 2 and 3) are a family of esterases that are highly conserved within and between species. They exhibit antioxidant and anti-inflammatory activities. However, their physiological function(s) and native substrates are uncertain. Previous structure-activity relationship studies demonstrate that PONs have a high specificity for lipophilic lactones, suggesting that such compounds may be representative of native substrates. This report describes the ability of PONs to hydrolyze two bioactive δ-lactones derived from arachidonic acid, 5,6-dihydroxy-eicosatrienoic acid lactone (5,6-DHTL) and cyclo-epoxycyclopentenone (cyclo-EC). Both lactones were very efficiently hydrolyzed by purified PON3. PON1 efficiently hydrolyzed 5,6-DHTL, but with a specific activity about 15-fold lower than PON3. 5,6-DHTL was a poor substrate for PON2. Cyclo-EC was a poor substrate for PON1 and not hydrolyzed by PON2. Studies with the PON inhibitor EDTA and a serine esterase inhibitor indicated that the PONs are the main contributors to hydrolysis of the lactones in human and mouse liver homogenates. Studies with homogenates from PON3 knockout mouse livers indicated that >80% of the 5,6-DHTL and cyclo-EC lactonase activities were attributed to PON3. The findings provide further insight into the structural requirements for PONs substrates and support the hypothesis that PONs, particularly PON1 and PON3, evolved to hydrolyze and regulate a class of lactone lipid mediators derived from polyunsaturated fatty acids.


Asunto(s)
Arildialquilfosfatasa/metabolismo , Eicosanoides/metabolismo , Lactonas/metabolismo , Animales , Ácido Araquidónico/química , Ácido Araquidónico/metabolismo , Arildialquilfosfatasa/genética , Eicosanoides/química , Células HEK293 , Humanos , Hidrólisis , Lactonas/química , Hígado/metabolismo , Ratones Noqueados , Estructura Molecular , Especificidad por Sustrato
4.
FASEB J ; 29(4): 1185-97, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25477283

RESUMEN

We report the engineering and characterization of paraoxonase-3 knockout mice (Pon3KO). The mice were generally healthy but exhibited quantitative alterations in bile acid metabolism and a 37% increased body weight compared to the wild-type mice on a high fat diet. PON3 was enriched in the mitochondria-associated membrane fraction of hepatocytes. PON3 deficiency resulted in impaired mitochondrial respiration, increased mitochondrial superoxide levels, and increased hepatic expression of inflammatory genes. PON3 deficiency did not influence atherosclerosis development on an apolipoprotein E null hyperlipidemic background, but it did lead to a significant 60% increase in atherosclerotic lesion size in Pon3KO mice on the C57BL/6J background when fed a cholate-cholesterol diet. On the diet, the Pon3KO had significantly increased plasma intermediate-density lipoprotein/LDL cholesterol and bile acid levels. They also exhibited significantly elevated levels of hepatotoxicity markers in circulation, a 58% increase in gallstone weight, a 40% increase in hepatic cholesterol level, and increased mortality. Furthermore, Pon3KO mice exhibited decreased hepatic bile acid synthesis and decreased bile acid levels in the small intestine compared with wild-type mice. Our study suggests a role for PON3 in the metabolism of lipid and bile acid as well as protection against atherosclerosis, gallstone disease, and obesity.


Asunto(s)
Arildialquilfosfatasa/deficiencia , Aterosclerosis/enzimología , Cálculos Biliares/enzimología , Obesidad/enzimología , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Arildialquilfosfatasa/genética , Arildialquilfosfatasa/metabolismo , Aterosclerosis/etiología , Aterosclerosis/genética , Ácidos y Sales Biliares/metabolismo , Quimiocina CCL2/metabolismo , Colesterol en la Dieta/administración & dosificación , Ácido Cólico/administración & dosificación , Dieta/efectos adversos , Modelos Animales de Enfermedad , Femenino , Cálculos Biliares/etiología , Cálculos Biliares/genética , Expresión Génica , Predisposición Genética a la Enfermedad , Mediadores de Inflamación/metabolismo , Interleucina-6/metabolismo , Intestino Delgado/metabolismo , Riñón/metabolismo , Metabolismo de los Lípidos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Hepáticas/metabolismo , Obesidad/etiología , Obesidad/genética
5.
J Lipid Res ; 56(1): 22-37, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25378658

RESUMEN

We performed silencing and overexpression studies of flavin containing monooxygenase (FMO) 3 in hyperlipidemic mouse models to examine its effects on trimethylamine N-oxide (TMAO) levels and atherosclerosis. Knockdown of hepatic FMO3 in LDL receptor knockout mice using an antisense oligonucleotide resulted in decreased circulating TMAO levels and atherosclerosis. Surprisingly, we also observed significant decreases in hepatic lipids and in levels of plasma lipids, ketone bodies, glucose, and insulin. FMO3 overexpression in transgenic mice, on the other hand, increased hepatic and plasma lipids. Global gene expression analyses suggested that these effects of FMO3 on lipogenesis and gluconeogenesis may be mediated through the PPARα and Kruppel-like factor 15 pathways. In vivo and in vitro results were consistent with the concept that the effects were mediated directly by FMO3 rather than trimethylamine/TMAO; in particular, overexpression of FMO3 in the human hepatoma cell line, Hep3B, resulted in significantly increased glucose secretion and lipogenesis. Our results indicate a major role for FMO3 in modulating glucose and lipid homeostasis in vivo, and they suggest that pharmacologic inhibition of FMO3 to reduce TMAO levels would be confounded by metabolic interactions.


Asunto(s)
Aterosclerosis/enzimología , Glucosa/metabolismo , Metabolismo de los Lípidos , Oxigenasas/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Dieta Occidental , Heces/química , Femenino , Regulación Enzimológica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Glucosa/biosíntesis , Homeostasis , Humanos , Insulina/sangre , Mucosa Intestinal/metabolismo , Factores de Transcripción de Tipo Kruppel , Lipogénesis , Lipoproteínas/sangre , Hígado/metabolismo , Metilaminas/metabolismo , Ratones , Oxigenasas/deficiencia , Oxigenasas/genética , PPAR alfa/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/genética , Factores de Transcripción/metabolismo
6.
FASEB J ; 27(9): 3805-17, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23752203

RESUMEN

We examined the effects of a natural secondary bile acid, hyodeoxycholic acid (HDCA), on lipid metabolism and atherosclerosis in LDL receptor-null (LDLRKO) mice. Female LDLRKO mice were maintained on a Western diet for 8 wk and then divided into 2 groups that received chow, or chow + 1.25% HDCA, diets for 15 wk. We observed that mice fed the HDCA diet were leaner and exhibited a 37% (P<0.05) decrease in fasting plasma glucose level. HDCA supplementation significantly decreased atherosclerotic lesion size at the aortic root region, the entire aorta, and the innominate artery by 44% (P<0.0001), 48% (P<0.01), and 94% (P<0.01), respectively, as compared with the chow group. Plasma VLDL/IDL/LDL cholesterol levels were significantly decreased, by 61% (P<0.05), in the HDCA group as compared with the chow diet group. HDCA supplementation decreased intestinal cholesterol absorption by 76% (P<0.0001) as compared with the chow group. Furthermore, HDL isolated from the HDCA group exhibited significantly increased ability to mediate cholesterol efflux ex vivo as compared with HDL of the chow diet group. In addition, HDCA significantly increased the expression of genes involved in cholesterol efflux, such as Abca1, Abcg1, and Apoe, in a macrophage cell line. Thus, HDCA is a candidate for antiatherosclerotic drug therapy.


Asunto(s)
Aterosclerosis/metabolismo , Aterosclerosis/prevención & control , Ácido Desoxicólico/uso terapéutico , Lipoproteínas HDL/sangre , Receptores de LDL/deficiencia , Animales , HDL-Colesterol/sangre , LDL-Colesterol/sangre , VLDL-Colesterol/sangre , Femenino , Absorción Intestinal/efectos de los fármacos , Lipoproteínas LDL/sangre , Ratones , Ratones Noqueados , Receptores de LDL/genética
7.
J Proteome Res ; 12(4): 1946-55, 2013 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-23448543

RESUMEN

Oxidative stress is a determinant of liver steatosis and the progression to more severe forms of disease. The present study investigated the effect of paraoxonase-1 (PON1) deficiency on histological alterations and hepatic metabolism in mice fed a high-fat high-cholesterol diet. We performed nontargeted metabolomics on liver tissues from 8 male PON1-deficient mice and 8 wild-type animals fed a high-fat, high-cholesterol diet for 22 weeks. We also measured 8-oxo-20-deoxyguanosine, reduced and oxidized glutathione, malondialdehyde, 8-isoprostanes and protein carbonyl concentrations. Results indicated lipid droplets in 14.5% of the hepatocytes of wild-type mice and in 83.3% of the PON1-deficient animals (P < 0.001). The metabolomic assay included 322 biochemical compounds, 169 of which were significantly decreased and 16 increased in PON1-deficient mice. There were significant increases in lipid peroxide concentrations and oxidative stress markers. We also found decreased glycolysis and the Krebs cycle. The urea cycle was decreased, and the pyrimidine cycle had a significant increase in orotate. The pathways of triglyceride and phospholipid synthesis were significantly increased. We conclude that PON1 deficiency is associated with oxidative stress and metabolic alterations leading to steatosis in the livers of mice receiving a high-fat high-cholesterol diet.


Asunto(s)
Arildialquilfosfatasa/deficiencia , Colesterol/efectos adversos , Dieta Alta en Grasa/efectos adversos , Hígado Graso/etiología , Metabolismo de los Lípidos/efectos de los fármacos , Aminoácidos/metabolismo , Animales , Arildialquilfosfatasa/genética , Biomarcadores/metabolismo , Hígado Graso/metabolismo , Hígado Graso/patología , Glucosa/metabolismo , Glutatión/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Metabolómica/métodos , Ratones , Ratones Endogámicos C57BL , Ácido Orótico/metabolismo , Estrés Oxidativo
8.
Biochem Biophys Res Commun ; 431(1): 19-24, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23291186

RESUMEN

Hyperlipidemia blunts anabolic effects of intermittent parathyroid hormone (PTH) on cortical bone, and the responsiveness to PTH are restored in part by oral administration of the antioxidant ApoA-I mimetic peptide, D-4F. To evaluate the mechanism of this rescue, hyperlipidemic mice overexpressing the high-density lipoprotein-associated antioxidant enzyme, paraoxonase 1 (Ldlr(-/-)PON1(tg)) were generated, and daily PTH injections were administered to Ldlr(-/-)PON1(tg) and to littermate Ldlr(-/-) mice. Expression of bone regulatory genes was determined by realtime RT-qPCR, and cortical bone parameters of the femoral bones by micro-computed tomographic analyses. PTH-treated Ldlr(-/-)PON1(tg) mice had significantly greater expression of PTH receptor (PTH1R), activating transcription factor-4 (ATF4), and osteoprotegerin (OPG) in femoral cortical bone, as well as significantly greater cortical bone mineral content, thickness, and area in femoral diaphyses compared with untreated Ldlr(-/-)PON1(tg) mice. In contrast, in control mice (Ldlr(-/-)) without PON1 overexpression, PTH treatment did not induce these markers. Calvarial bone of PTH-treated Ldlr(-/-)PON1(tg) mice also had significantly greater expression of osteoblastic differentiation marker genes as well as BMP-2-target and Wnt-target genes. Untreated Ldlr(-/-)PON1(tg) mice had significantly greater expression of PTHR1 than untreated Ldlr(-/-) mice, whereas sclerostin expression was reduced. In femoral cortical bones, expression levels of transcription factors, FoxO1 and ATF4, were also elevated in the untreated, control Ldlr(-/-)PON1(tg) mice, suggesting enhancement of cellular protection against oxidants. These findings suggest that PON1 restores responsiveness to PTH through effects on oxidant stress, PTH receptor expression, and/or Wnt signaling.


Asunto(s)
Anabolizantes/administración & dosificación , Arildialquilfosfatasa/fisiología , Huesos/efectos de los fármacos , Hiperlipidemias/enzimología , Hormona Paratiroidea/administración & dosificación , Animales , Huesos/enzimología , Expresión Génica/efectos de los fármacos , Humanos , Hiperlipidemias/sangre , Lípidos/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteocitos/efectos de los fármacos , Osteocitos/metabolismo , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Receptores de LDL/genética
9.
Mediators Inflamm ; 2013: 156053, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23766557

RESUMEN

We studied the influence of PON1 on metabolic alterations induced by oxidized LDL when incubated with endothelial cells. HUVEC cells were incubated with native LDL, oxidized LDL, oxidized LDL plus HDL from wild type mice, and oxidized LDL plus HDL from PON1-deficient mice. Results showed alterations in carbohydrate and phospholipid metabolism and increased apoptosis in cells incubated with oxidized LDL. These changes were partially prevented by wild type mouse HDL, but the effects were less effective with HDL from PON1-deficient mice. Our results suggest that PON1 may play a significant role in endothelial cell survival by protecting cells from alterations in the respiratory chain induced by oxidized LDL. These results extend current knowledge on the protective role of HDL and PON1 against oxidation and apoptosis in endothelial cells.


Asunto(s)
Arildialquilfosfatasa/metabolismo , Células Endoteliales/metabolismo , Lipoproteínas LDL/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Arildialquilfosfatasa/genética , Caspasa 9/metabolismo , Ciclo del Ácido Cítrico/fisiología , Citometría de Flujo , Glucólisis/fisiología , Humanos , Lipoproteínas HDL/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Fosfolípidos/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 31(11): 2624-33, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21836061

RESUMEN

OBJECTIVE: Chronic infection has long been postulated as a stimulus for atherogenesis. Pseudomonas aeruginosa infection has been associated with increased atherosclerosis in rats, and these bacteria produce a quorum-sensing molecule 3-oxo-dodecynoyl-homoserine lactone (3OC12-HSL) that is critical for colonization and virulence. Paraoxonase 2 (PON2) hydrolyzes 3OC12-HSL and also protects against the effects of oxidized phospholipids thought to contribute to atherosclerosis. We now report the response of human aortic endothelial cells (HAECs) to 3OC12-HSL and oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine (Ox-PAPC) in relation to PON2 expression. METHODS AND RESULTS: Using expression profiling and network modeling, we identified the unfolded protein response (UPR), cell cycle genes, and the mitogen-activated protein kinase signaling pathway to be heavily involved in the HAEC response to 3OC12-HSL. The network also showed striking similarities to a network created based on HAEC response to Ox-PAPC, a major component of minimally modified low-density lipoprotein. HAECs in which PON2 was silenced by small interfering RNA showed increased proinflammatory response and UPR when treated with 3OC12-HSL or Ox-PAPC. CONCLUSION: 3OC12-HSL and Ox-PAPC influence similar inflammatory and UPR pathways. Quorum sensing molecules, such as 3OC12-HSL, contribute to the proatherogenic effects of chronic infection. The antiatherogenic effects of PON2 include destruction of quorum sensing molecules.


Asunto(s)
4-Butirolactona/análogos & derivados , Arildialquilfosfatasa/metabolismo , Endotelio Vascular/metabolismo , Homoserina/análogos & derivados , Fosfolípidos/farmacología , Percepción de Quorum , Estrés Fisiológico/efectos de los fármacos , 4-Butirolactona/farmacología , Aorta/citología , Aorta/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Arildialquilfosfatasa/antagonistas & inhibidores , Arildialquilfosfatasa/efectos de los fármacos , Células Cultivadas , Endotelio Vascular/citología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Homoserina/farmacología , Humanos , Lipoproteínas LDL/metabolismo , Oxidación-Reducción , ARN Interferente Pequeño/farmacología , Estrés Fisiológico/fisiología
11.
JACC Basic Transl Sci ; 6(2): 103-115, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33665512

RESUMEN

This study demonstrates, for the first time, that renal tubular excretion of trimethylamine N-oxide (TMAO) is inhibited by concomitant loop diuretic administration. The observed marked accumulation in the renal parenchyma, and to lesser extent, plasma, implies differential distributions of TMAO across various tissues and/or systems as a consequence of efflux channel control. A better understanding of TMAO renal clearance and its potential interactions with current and future therapies in patients with heart failure are warranted.

12.
Sci Rep ; 11(1): 16409, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34385484

RESUMEN

We recently showed that NOTUM, a liver-secreted Wnt inhibitor, can acutely promote browning of white adipose. We now report studies of chronic overexpression of NOTUM in liver indicating that it protects against diet-induced obesity and improves glucose homeostasis in mice. Adeno-associated virus (AAV) vectors were used to overexpress GFP or mouse Notum in the livers of male C57BL/6J mice and the mice were fed an obesifying diet. After 14 weeks of high fat, high sucrose diet feeding, the AAV-Notum mice exhibited decreased obesity and improved glucose tolerance compared to the AAV-GFP mice. Gene expression and immunoblotting analysis of the inguinal fat and brown fat revealed increased expression of beige/brown adipocyte markers in the AAV-Notum group, suggesting enhanced thermogenic capacity by NOTUM. A ß3 adrenergic receptor agonist-stimulated lipolysis test suggested increased lipolysis capacity by NOTUM. The levels of collagen and C-C motif chemokine ligand 2 (CCL2) in the epididymal white adipose tissue of the AAV-Notum mice were significantly reduced, suggesting decreased fibrosis and inflammation, respectively. RNA sequencing analysis of inguinal white adipose of 4-week chow diet-fed mice revealed a highly significant enrichment of extracellular matrix (ECM) functional cluster among the down-regulated genes in the AAV-Notum group, suggesting a potential mechanism contributing to improved glucose homeostasis. Our in vitro studies demonstrated that recombinant human NOTUM protein blocked the inhibitory effects of WNT3A on brown adipocyte differentiation. Furthermore, NOTUM attenuated WNT3A's effects on upregulation of TGF-ß signaling and its downstream targets. Overall, our data suggest that NOTUM modulates adipose tissue function by promoting thermogenic capacity and inhibiting fibrosis through inhibition of Wnt signaling.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Esterasas/metabolismo , Obesidad/metabolismo , Termogénesis/fisiología , Adipocitos Beige/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Metabolismo Energético/fisiología , Intolerancia a la Glucosa/metabolismo , Lipólisis/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL
13.
Sci Rep ; 11(1): 518, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33436815

RESUMEN

Patients with chronic kidney disease (CKD) have elevated circulating levels of trimethylamine N-oxide (TMAO), a metabolite derived from gut microbes and associated with cardiovascular diseases. High circulating levels of TMAO and its dietary precursor, choline, predict increased risk for development of CKD in apparently healthy subjects, and studies in mice fed TMAO or choline suggest that TMAO can contribute to kidney impairment and renal fibrosis. Here we examined the interactions between TMAO, kidney disease, and cardiovascular disease in mouse models. We observed that while female hyperlipidemic apoE KO mice fed a 0.2% adenine diet for 14 weeks developed CKD with elevated plasma levels of TMAO, provision of a non-lethal inhibitor of gut microbial trimethylamine (TMA) production, iodomethylcholine (IMC), significantly reduced multiple markers of renal injury (plasma creatinine, cystatin C, FGF23, and TMAO), reduced histopathologic evidence of fibrosis, and markedly attenuated development of microalbuminuria. In addition, while the adenine-induced CKD model significantly increased heart weight, a surrogate marker for myocardial hypertrophy, this was largely prevented by IMC supplementation. Surprisingly, adenine feeding did not increase atherosclerosis and significantly decreased the expression of inflammatory genes in the aorta compared to the control groups, effects unrelated to TMAO levels. Our data demonstrate that inhibition of TMAO production attenuated CKD development and cardiac hypertrophy in mice, suggesting that TMAO reduction may be a novel strategy in treating CKD and its cardiovascular disease complications.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Metilaminas/efectos adversos , Metilaminas/metabolismo , Insuficiencia Renal Crónica/etiología , Adenina/administración & dosificación , Adenina/efectos adversos , Albuminuria/etiología , Animales , Cardiomegalia/etiología , Cardiomegalia/prevención & control , Colina/administración & dosificación , Colina/efectos adversos , Colina/análogos & derivados , Colina/farmacología , Modelos Animales de Enfermedad , Femenino , Factor-23 de Crecimiento de Fibroblastos , Fibrosis , Riñón/patología , Metilaminas/administración & dosificación , Ratones , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/prevención & control
14.
J Biol Chem ; 284(51): 35564-71, 2009 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-19840942

RESUMEN

Paraoxonases (PONs) are a family of lactonases with promiscuous enzyme activity that has been implicated in multiple diseases. PON2 is intracellularly located, is the most ubiquitously expressed PON, and has the highest lactonase activity of the PON family members. Whereas some single-nucleotide polymorphisms (SNPs) in PON1 have resulted in altered enzymatic activity in serum, to date the functional consequences of SNPs on PON2 function remain unknown. We hypothesized that a common PON2 SNP would result in impaired lactonase activity. Substitution of cysteine for serine at codon 311 in recombinant PON2 resulted in normal protein production and localization but altered glycosylation and decreased lactonase activity. Moreover, we screened 200 human lung samples for the PON2 Cys(311) variant and found that in vivo this mutation impaired lactonase activity. These data suggest that impaired lactonase activity may play a role in innate immunity, atherosclerosis, and other diseases associated with the PON2 311 SNP.


Asunto(s)
Arildialquilfosfatasa/metabolismo , Aterosclerosis/enzimología , Inmunidad Innata , Polimorfismo de Nucleótido Simple , Sustitución de Aminoácidos , Animales , Arildialquilfosfatasa/genética , Aterosclerosis/genética , Células CHO , Cricetinae , Cricetulus , Glicosilación , Humanos , Pulmón/enzimología
15.
FASEB J ; 23(6): 1721-7, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19204075

RESUMEN

Genetic disorders of homocysteine (Hcy) or folate metabolism or high-methionine diets elevate plasma Hcy and its atherogenic metabolite Hcy-thiolactone. In humans, severe hyperhomocysteinemia due to genetic alterations in cystathionine beta-synthase (Cbs) or methylenetetrahydrofolate reductase (Mthfr) results in neurological abnormalities and premature death from vascular complications. In mouse models, dietary or genetic hyperhomocysteinemia results in liver or brain pathological changes and accelerates atherosclerosis. Hcy-thiolactone has the ability to form isopeptide bonds with protein lysine residues, which generates modified proteins (N-Hcy-protein) with autoimmunogenic and prothrombotic properties. Our aim was to determine how N-Hcy-protein levels are affected by genetic or nutritional disorders in Hcy or folate metabolism in mice. We found that plasma N-Hcy-protein was elevated 10-fold in mice fed a high-methionine diet compared with the animals fed a normal commercial diet. We also found that inactivation of Cbs, Mthfr, or the proton-coupled folate transporter (Pcft) gene resulted in a 10- to 30-fold increase in plasma or serum N-Hcy-protein levels. Liver N-Hcy-protein was elevated 3.4-fold in severely and 11-fold in extremely hyperhomocysteinemic Cbs-deficient mice, 3.6-fold in severely hyperhomocysteinemic Pcft mice, but was not elevated in mildly hyperhomocysteinemic Mthfr-deficient animals, suggesting that mice have a capacity to prevent accumulation of N-Hcy-protein in their organs. These findings provide evidence that N-Hcy-protein is an important metabolite associated with Hcy pathophysiology in the mouse.


Asunto(s)
Ácido Fólico/metabolismo , Homocisteína/metabolismo , Hiperhomocisteinemia , Animales , Cistationina betasintasa/genética , Cistationina betasintasa/metabolismo , Dieta , Homocisteína/análogos & derivados , Humanos , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/metabolismo , Hiperhomocisteinemia/patología , Hígado/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Metionina/administración & dosificación , Metionina/metabolismo , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Metilenotetrahidrofolato Reductasa (NADPH2)/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
16.
Adv Exp Med Biol ; 660: 73-87, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20221872

RESUMEN

PON3 is a member of the paraoxonase gene family that includes PON1 and PON2. For example, PON3 and PON1 share approximately 60% identity at the amino acid level. Recent studies have demonstrated that PON3 is present in human and rabbit HDL but not in mouse HDL. Mouse PON3 appears to be cell-associated and is expressed in a wide range of tissues such as liver, adipose, macrophage, and the artery wall. In vitro studies have shown that PON3 can prevent LDL oxidation and destroy bacterial quorum-sensing molecules. Previous studies also showed that human PON3 transgenic mice were protected from obesity and atherosclerosis in both the C57BL/6 J wild-type and LDLR knockout genetic background. Administration of adenovirus expressing the human PON3 gene into apoE -/- mice also decreased atherosclerotic lesion formation. In order to further understand the functions of PON3 in physiology and disease, we performed in situ hybridization analysis to examine Pon3 gene expression patterns in newborn and adult mice, in various tissues, including atherosclerotic lesions of apoE -/- mice. Our results show relatively high levels of Pon3 mRNA labeling in the adrenal gland, submaxillary gland, lung, liver, adipose, pancreas, large intestine, and other tissues of newborn mice. In the adult mouse, Pon3 mRNA levels were much lower in the corresponding tissues as mentioned above for the newborn mouse. Sections of the aortic root from the hearts of both wild-type and apoE -/- mice displayed moderate levels of Pon3 mRNA labeling. Pon3 mRNA was also detected in the atherosclerotic lesion areas at the aortic root of apoE -/- hearts. Our data revealed that mouse Pon3 is expressed in a wide range of tissues, and that its expression is temporally controlled.


Asunto(s)
Arildialquilfosfatasa/biosíntesis , Arildialquilfosfatasa/genética , Animales , Regulación de la Expresión Génica , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Genéticos , Miocardio/metabolismo , ARN Complementario/metabolismo , ARN Mensajero/metabolismo , Factores de Tiempo , Distribución Tisular
17.
Curr Opin Lipidol ; 20(4): 288-92, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19474728

RESUMEN

PURPOSE OF REVIEW: The paraoxonase (PON) gene family includes three members, PON1, PON2, and PON3. In-vitro and mouse studies have demonstrated that all three PONs are atheroprotective. Some, but not all, human epidemiologic studies have observed associations between PON gene polymorphisms and risk of cardiovascular disease (CVD). In this review, we summarize studies published within the last year, elucidating involvement of PON1 and PON2 in oxidative stress, CVD, and innate immune responses. RECENT FINDINGS: In a prospective study, the PON1 192QQ genotype and low PON1 activity were associated with increased systemic oxidative stress and increased risk for CVD. PON1 expression protected against Pseudomonas aeruginosa lethality in Drosophila, suggesting that PON1 can interfere with quorum sensing in vivo. PON2 attenuated macrophage triglyceride accumulation via inhibition of diacylglycerol acyltransferase 1. Overexpression of PON2 protected against endoplasmic reticulum stress-induced apoptosis when the stress was induced by interference with protein modification but not when endoplasmic reticulum stress was induced by Ca2+ deregulation. SUMMARY: Both mouse and human studies have demonstrated the antioxidative and atheroprotective effects of PON1. The mechanisms by which PON2 exerts its atheroprotective effects are emerging. Large-scale epidemiologic studies are needed to further examine the relationship between PON2 genetic polymorphisms and risk for CVD. Elucidation of the physiologic substrates of the PON proteins is of particular importance to further advance this field.


Asunto(s)
Arildialquilfosfatasa/inmunología , Arildialquilfosfatasa/metabolismo , Enfermedades Cardiovasculares/enzimología , Enfermedades Cardiovasculares/inmunología , Inmunidad Innata/fisiología , Animales , Antioxidantes , Arildialquilfosfatasa/genética , Regulación Enzimológica de la Expresión Génica , Humanos
18.
Sci Rep ; 10(1): 16848, 2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-33033318

RESUMEN

Paraoxonase 1(PON1) is an HDL-associated protein, which metabolizes inflammatory, oxidized lipids associated with atherosclerotic plaque development. Because oxidized lipid mediators have also been implicated in the pathogenesis of rheumatoid arthritis (RA), we evaluated the role of PON1 in murine inflammatory arthritis. K/BxN serum transfer (STIA) or collagen antibody transfer (CAIA) was used for arthritis induction in B6 mice homozygous for the PON1 human transgene [PON1Tg], PON1 knock-out mice [PON1KO], and wild type littermate control mice [WT]. Experiments were also performed in K/BxN mice with chronic arthritis, and in RA patients and healthy controls. Arthritis activity in K/BxN mice was associated with a marked dyslipidemia, lower PON1 activity and higher bioactive lipid mediators (BLM), as well as a dysregulated hepatic lipid gene expression profile. Higher serum PON1 activity correlated with lower BLM and lower arthritis activity in both K/BxN mice and RA patients. Overexpression of the human PON1 transgene was associated with reduced inflammatory arthritis, which correlated strongly with higher circulating PON1 activity, upregulation of the hepatic glutathione pathway, and reduction of circulating BLM. These results implicate PON1 as a potential novel therapeutic target for joint disease in RA with potential for vascular benefit, which warrants further investigation.


Asunto(s)
Artritis Reumatoide/genética , Arildialquilfosfatasa/genética , Arildialquilfosfatasa/metabolismo , Expresión Génica , Animales , Artritis Reumatoide/etiología , Artritis Reumatoide/terapia , Enfermedad Crónica , Dislipidemias/genética , Glutatión/metabolismo , Humanos , Inflamación , Hígado/metabolismo , Ratones Transgénicos , Terapia Molecular Dirigida , Transducción de Señal , Transgenes
19.
Circ Res ; 100(12): 1703-11, 2007 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-17495224

RESUMEN

Heme oxygenase (HO-1) is the rate-limiting enzyme in the catabolism of heme, which leads to the generation of biliverdin, iron, and carbon monoxide. It has been shown to have important antioxidant and antiinflammatory properties that result in a vascular antiatherogenic effect. To determine whether HO-1 expression in macrophages constitutes a significant component of the protective role in atherosclerosis, we evaluated the effect of decreased or absent HO-1 expression in peritoneal macrophages on oxidative stress and inflammation in vitro, and the effect of complete deficiency of HO-1 expression in macrophages in atherosclerotic lesion formation in vivo. We found that compared with HO-1(+/+) controls, peritoneal macrophages from HO-1(-/-) and HO-1(+/-) mice exhibited (1) increased reactive oxygen species (ROS) generation, (2) increased proinflammatory cytokines such as monocyte chemotactic protein 1 (MCP-1) and interleukin 6 (IL-6), and (3) increased foam cell formation when treated with oxLDL, attributable in part to increased expression of scavenger receptor A (SR-A). Bone marrow transplantation experiments performed in lethally irradiated LDL-R null female mice, reconstituted with bone marrow from HO-1(-/-) versus HO-1(+/+) mice, revealed that HO-1(-/-) reconstituted animals exhibited atherosclerotic lesions with a greater macrophage content as evaluated by immunohistochemistry and planimetric assessment. We conclude that HO-1 expression in macrophages constitutes an important component of the antiatherogenic effect by increasing antioxidant protection and decreasing the inflammatory component of atherosclerotic lesions.


Asunto(s)
Aterosclerosis/metabolismo , Aterosclerosis/prevención & control , Hemo-Oxigenasa 1/metabolismo , Macrófagos/enzimología , Animales , Antioxidantes/metabolismo , Aterosclerosis/patología , Trasplante de Médula Ósea/patología , Quimiocina CCL2/metabolismo , Células Espumosas/patología , Regulación de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Interleucina-6/metabolismo , Macrófagos/patología , Ratones , Estrés Oxidativo/fisiología , Especies Reactivas de Oxígeno/metabolismo , Receptores de LDL/genética , Receptores de LDL/fisiología , Receptores Depuradores de Clase A/genética , Receptores Depuradores de Clase A/fisiología
20.
Circ Res ; 100(8): 1200-7, 2007 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-17379834

RESUMEN

Paraoxonase 3 (PON3) is a member of the PON family, which includes PON1, PON2, and PON3. Recently, PON3 was shown to prevent the oxidation of low-density lipoprotein in vitro. To test the role of PON3 in atherosclerosis and related traits, 2 independent lines of human PON3 transgenic (Tg) mice on the C57BL/6J (B6) background were constructed. Human PON3 mRNA was detected in various tissues, including liver, lung, kidney, brain, adipose, and aorta, of both lines of Tg mice. The human PON3 mRNA levels in the livers of PON3 Tg mice were 4- to 7-fold higher as compared with the endogenous mouse Pon3 mRNA levels. Human PON3 protein and activity were detected in the livers of Tg mice as well. No significant differences in plasma total, high-density lipoprotein, and very-low-density lipoprotein/low-density lipoprotein cholesterol and triglyceride and glucose levels were observed between the PON3 Tg and non-Tg mice. Interestingly, atherosclerotic lesion areas were significantly smaller in both lines of male PON3 Tg mice as compared with the male non-Tg littermates on B6 background fed an atherogenic diet. When bred onto the low-density lipoprotein receptor knockout mouse background, the male PON3 Tg mice also exhibited decreased atherosclerotic lesion areas and decreased expression of monocyte chemoattractant protein-1 in the aorta as compared with the male non-Tg littermates. In addition, decreased adiposity and lower circulating leptin levels were observed in both lines of male PON3 Tg mice as compared with the male non-Tg mice. In an F2 cross, adipose Pon3 mRNA levels inversely correlated with adiposity and related traits. Our study demonstrates that elevated PON3 expression significantly decreases atherosclerotic lesion formation and adiposity in male mice. PON3 may play an important role in protection against obesity and atherosclerosis.


Asunto(s)
Arteriosclerosis/enzimología , Arteriosclerosis/genética , Esterasas/genética , Obesidad/enzimología , Obesidad/genética , Animales , Arildialquilfosfatasa , Esterasas/biosíntesis , Esterasas/fisiología , Femenino , Regulación Enzimológica de la Expresión Génica/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA