Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Arterioscler Thromb Vasc Biol ; 39(10): 1967-1985, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31434492

RESUMO

OBJECTIVE: Fatty acid uptake and oxidation characterize the metabolism of alternatively activated macrophage polarization in vitro, but the in vivo biology is less clear. We assessed the roles of LpL (lipoprotein lipase)-mediated lipid uptake in macrophage polarization in vitro and in several important tissues in vivo. Approach and Results: We created mice with both global and myeloid-cell specific LpL deficiency. LpL deficiency in the presence of VLDL (very low-density lipoproteins) altered gene expression of bone marrow-derived macrophages and led to reduced lipid uptake but an increase in some anti- and some proinflammatory markers. However, LpL deficiency did not alter lipid accumulation or gene expression in circulating monocytes nor did it change the ratio of Ly6Chigh/Ly6Clow. In adipose tissue, less macrophage lipid accumulation was found with global but not myeloid-specific LpL deficiency. Neither deletion affected the expression of inflammatory genes. Global LpL deficiency also reduced the numbers of elicited peritoneal macrophages. Finally, we assessed gene expression in macrophages from atherosclerotic lesions during regression; LpL deficiency did not affect the polarity of plaque macrophages. CONCLUSIONS: The phenotypic changes observed in macrophages upon deletion of Lpl in vitro is not mimicked in tissue macrophages.


Assuntos
Aterosclerose/metabolismo , Hiperlipoproteinemia Tipo I/metabolismo , Lipase Lipoproteica/metabolismo , Ativação de Macrófagos/genética , Animais , Aterosclerose/patologia , Células Cultivadas , Modelos Animais de Doenças , Humanos , Hiperlipoproteinemia Tipo I/patologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patologia , Papel (figurativo) , Sensibilidade e Especificidade , Triglicerídeos/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 38(3): 509-519, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29371243

RESUMO

OBJECTIVE: Tissue macrophages induce and perpetuate proinflammatory responses, thereby promoting metabolic and cardiovascular disease. Lipoprotein lipase (LpL), the rate-limiting enzyme in blood triglyceride catabolism, is expressed by macrophages in atherosclerotic plaques. We questioned whether LpL, which is also expressed in the bone marrow (BM), affects circulating white blood cells and BM proliferation and modulates macrophage retention within the artery. APPROACH AND RESULTS: We characterized blood and tissue leukocytes and inflammatory molecules in transgenic LpL knockout mice rescued from lethal hypertriglyceridemia within 18 hours of life by muscle-specific LpL expression (MCKL0 mice). LpL-deficient mice had ≈40% reduction in blood white blood cell, neutrophils, and total and inflammatory monocytes (Ly6C/Ghi). LpL deficiency also significantly decreased expression of BM macrophage-associated markers (F4/80 and TNF-α [tumor necrosis factor α]), master transcription factors (PU.1 and C/EBPα), and colony-stimulating factors (CSFs) and their receptors, which are required for monocyte and monocyte precursor proliferation and differentiation. As a result, differentiation of macrophages from BM-derived monocyte progenitors and monocytes was decreased in MCKL0 mice. Furthermore, although LpL deficiency was associated with reduced BM uptake and accumulation of triglyceride-rich particles and macrophage CSF-macrophage CSF receptor binding, triglyceride lipolysis products (eg, linoleic acid) stimulated expression of macrophage CSF and macrophage CSF receptor in BM-derived macrophage precursor cells. Arterial macrophage numbers decreased after heparin-mediated LpL cell dissociation and by genetic knockout of arterial LpL. Reconstitution of LpL-expressing BM replenished aortic macrophage density. CONCLUSIONS: LpL regulates peripheral leukocyte levels and affects BM monocyte progenitor differentiation and aortic macrophage accumulation.


Assuntos
Aorta/enzimologia , Doenças da Aorta/enzimologia , Aterosclerose/enzimologia , Hiperlipoproteinemia Tipo I/enzimologia , Lipase Lipoproteica/deficiência , Macrófagos/enzimologia , Monócitos/enzimologia , Células Progenitoras Mieloides/enzimologia , Mielopoese , Animais , Aorta/patologia , Doenças da Aorta/sangue , Doenças da Aorta/genética , Doenças da Aorta/patologia , Aterosclerose/sangue , Aterosclerose/genética , Aterosclerose/patologia , Proliferação de Células , Citocinas/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Hiperlipoproteinemia Tipo I/sangue , Hiperlipoproteinemia Tipo I/genética , Hiperlipoproteinemia Tipo I/patologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Lipase Lipoproteica/genética , Macrófagos/patologia , Camundongos Knockout , Monócitos/patologia , Células Progenitoras Mieloides/patologia , Transdução de Sinais , Triglicerídeos/metabolismo
4.
Circ Res ; 118(2): 241-53, 2016 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-26574507

RESUMO

RATIONALE: Fatty acid oxidation is transcriptionally regulated by peroxisome proliferator-activated receptor (PPAR)α and under normal conditions accounts for 70% of cardiac ATP content. Reduced Ppara expression during sepsis and heart failure leads to reduced fatty acid oxidation and myocardial energy deficiency. Many of the transcriptional regulators of Ppara are unknown. OBJECTIVE: To determine the role of Krüppel-like factor 5 (KLF5) in transcriptional regulation of Ppara. METHODS AND RESULTS: We discovered that KLF5 activates Ppara gene expression via direct promoter binding. This is blocked in hearts of septic mice by c-Jun, which binds an overlapping site on the Ppara promoter and reduces transcription. We generated cardiac myocyte-specific Klf5 knockout mice that showed reduced expression of cardiac Ppara and its downstream fatty acid metabolism-related targets. These changes were associated with reduced cardiac fatty acid oxidation, ATP levels, increased triglyceride accumulation, and cardiac dysfunction. Diabetic mice showed parallel changes in cardiac Klf5 and Ppara expression levels. CONCLUSIONS: Cardiac myocyte KLF5 is a transcriptional regulator of Ppara and cardiac energetics.


Assuntos
Cardiomiopatia Dilatada/metabolismo , Diabetes Mellitus Experimental/metabolismo , Metabolismo Energético , Fatores de Transcrição Kruppel-Like/metabolismo , Miócitos Cardíacos/metabolismo , PPAR alfa/metabolismo , Sepse/metabolismo , Animais , Sítios de Ligação , Ligação Competitiva , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/fisiopatologia , Linhagem Celular , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/fisiopatologia , Ácidos Graxos/metabolismo , Genótipo , Fatores de Transcrição Kruppel-Like/deficiência , Fatores de Transcrição Kruppel-Like/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxirredução , PPAR alfa/genética , Fenótipo , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Proto-Oncogênicas c-jun/metabolismo , Sepse/genética , Sepse/fisiopatologia , Transdução de Sinais , Transportador 2 de Glucose-Sódio/genética , Transportador 2 de Glucose-Sódio/metabolismo , Inibidores do Transportador 2 de Sódio-Glicose , Fatores de Tempo , Transcrição Gênica , Ativação Transcricional , Transfecção , Triglicerídeos/metabolismo , Regulação para Cima
5.
PLoS One ; 10(6): e0128996, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26046657

RESUMO

We tested whether a high fat diet (HFD) containing the inflammatory dietary fatty acid palmitate or insulin deficient diabetes altered the remodeling of atherosclerotic plaques in LDL receptor knockout (Ldlr-/-) mice. Cholesterol reduction was achieved by using a helper-dependent adenovirus (HDAd) carrying the gene for the low-density lipoprotein receptor (Ldlr; HDAd-LDLR). After injection of the HDAd-LDLR, mice consuming either HFD, which led to insulin resistance but not hyperglycemia, or low fat diet (LFD), showed regression compared to baseline. However there was no difference between the two groups in terms of atherosclerotic lesion size, or CD68+ cell and lipid content. Because of the lack of effects of these two diets, we then tested whether viral-mediated cholesterol reduction would lead to defective regression in mice with greater hyperglycemia. In both normoglycemic and streptozotocin (STZ)-treated hyperglycemic mice, HDAd-LDLR significantly reduced plasma cholesterol levels, decreased atherosclerotic lesion size, reduced macrophage area and lipid content, and increased collagen content of plaque in the aortic sinus. However, reductions in anti-inflammatory and ER stress-related genes were less pronounced in STZ-diabetic mice compared to non-diabetic mice. In conclusion, HDAd-mediated Ldlr gene therapy is an effective and simple method to induce atherosclerosis regression in Ldlr-/- mice in different metabolic states.


Assuntos
Aterosclerose/terapia , Diabetes Mellitus Experimental/genética , Dieta Hiperlipídica , Terapia Genética/métodos , Hiperglicemia/terapia , Placa Aterosclerótica/terapia , Receptores de LDL/genética , Adenoviridae/genética , Animais , Aorta/metabolismo , Aorta/patologia , Aterosclerose/complicações , Aterosclerose/genética , Aterosclerose/patologia , Colesterol na Dieta/administração & dosagem , Colágeno/genética , Colágeno/metabolismo , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/patologia , Expressão Gênica , Vetores Genéticos , Hiperglicemia/complicações , Hiperglicemia/genética , Hiperglicemia/patologia , Resistência à Insulina , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Placa Aterosclerótica/complicações , Placa Aterosclerótica/genética , Placa Aterosclerótica/patologia , Receptores de LDL/deficiência , Estreptozocina
6.
J Biol Chem ; 289(43): 29881-91, 2014 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-25157099

RESUMO

Diacylglycerol acyltransferase 1 (DGAT1) catalyzes the final step in triglyceride synthesis, the conversion of diacylglycerol (DAG) to triglyceride. Dgat1(-/-) mice exhibit a number of beneficial metabolic effects including reduced obesity and improved insulin sensitivity and no known cardiac dysfunction. In contrast, failing human hearts have severely reduced DGAT1 expression associated with accumulation of DAGs and ceramides. To test whether DGAT1 loss alone affects heart function, we created cardiomyocyte-specific DGAT1 knock-out (hDgat1(-/-)) mice. hDgat1(-/-) mouse hearts had 95% increased DAG and 85% increased ceramides compared with floxed controls. 50% of these mice died by 9 months of age. The heart failure marker brain natriuretic peptide increased 5-fold in hDgat1(-/-) hearts, and fractional shortening (FS) was reduced. This was associated with increased expression of peroxisome proliferator-activated receptor α and cluster of differentiation 36. We crossed hDgat1(-/-) mice with previously described enterocyte-specific Dgat1 knock-out mice (hiDgat1(-/-)). This corrected the early mortality, improved FS, and reduced cardiac ceramide and DAG content. Treatment of hDgat1(-/-) mice with the glucagon-like peptide 1 receptor agonist exenatide also improved FS and reduced heart DAG and ceramide content. Increased fatty acid uptake into hDgat1(-/-) hearts was normalized by exenatide. Reduced activation of protein kinase Cα (PKCα), which is increased by DAG and ceramides, paralleled the reductions in these lipids. Our mouse studies show that loss of DGAT1 reproduces the lipid abnormalities seen in severe human heart failure.


Assuntos
Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/enzimologia , Lipídeos/sangue , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Envelhecimento/patologia , Animais , Glicemia/metabolismo , Colesterol/sangue , Diacilglicerol O-Aciltransferase/antagonistas & inibidores , Diacilglicerol O-Aciltransferase/metabolismo , Inibidores Enzimáticos/farmacologia , Exenatida , Ácidos Graxos/sangue , Deleção de Genes , Regulação da Expressão Gênica/efeitos dos fármacos , Insuficiência Cardíaca/genética , Humanos , Intestinos/efeitos dos fármacos , Intestinos/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Especificidade de Órgãos , Peptídeos/farmacologia , Fenótipo , Proteína Quinase C/metabolismo , Triglicerídeos/sangue , Peçonhas/farmacologia
7.
Am J Physiol Endocrinol Metab ; 305(11): E1339-47, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-24085031

RESUMO

Hearts utilize fatty acids as a primary source of energy. The sources of those lipids include free fatty acids and lipoprotein triglycerides. Deletion of the primary triglyceride-hydrolyzing enzyme lipoprotein lipase (LPL) leads to cardiac dysfunction. Whether heart LPL-knockout (hLPL0) mice are compromised due a deficiency in energetic substrates is unknown. To test whether alternative sources of energy will prevent cardiac dysfunction in hLPL0 mice, two different models were used to supply nonlipid energy. 1) hLPL0 mice were crossed with mice transgenically expressing GLUT1 in cardiomyocytes to increase glucose uptake into the heart; this cross-corrected cardiac dysfunction, reduced cardiac hypertrophy, and increased myocardial ATP. 2) Mice were randomly assigned to a sedentary or training group (swimming) at 3 mo of age, which leads to increased skeletal muscle production of lactate. hLPL0 mice had greater expression of the lactate transporter monocarboxylate transporter-1 (MCT-1) and increased cardiac lactate uptake. Compared with hearts from sedentary hLPL0 mice, hearts from trained hLPL0 mice had adaptive hypertrophy and improved cardiac function. We conclude that defective energy intake and not the reduced uptake of fat-soluble vitamins or cholesterol is responsible for cardiac dysfunction in hLPL0 mice. In addition, our studies suggest that adaptations in cardiac metabolism contribute to the beneficial effects of exercise on the myocardium of patients with heart failure.


Assuntos
Metabolismo Energético/genética , Coração/fisiologia , Lipase Lipoproteica/genética , Miocárdio/metabolismo , Triglicerídeos/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/prevenção & controle , Ecocardiografia , Transportador de Glucose Tipo 1/genética , Lipase Lipoproteica/metabolismo , Masculino , Camundongos , Camundongos Knockout , Miócitos Cardíacos/metabolismo , Cadeias Pesadas de Miosina/genética , Especificidade de Órgãos/genética
8.
Cell Metab ; 17(5): 695-708, 2013 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-23663738

RESUMO

Diabetes is a major risk factor for atherosclerosis. Although atherosclerosis is initiated by deposition of cholesterol-rich lipoproteins in the artery wall, the entry of inflammatory leukocytes into lesions fuels disease progression and impairs resolution. We show that diabetic mice have increased numbers of circulating neutrophils and Ly6-C(hi) monocytes, reflecting hyperglycemia-induced proliferation and expansion of bone marrow myeloid progenitors and release of monocytes into the circulation. Increased neutrophil production of S100A8/S100A9, and its subsequent interaction with the receptor for advanced glycation end products on common myeloid progenitor cells, leads to enhanced myelopoiesis. Treatment of hyperglycemia reduces monocytosis, entry of monocytes into atherosclerotic lesions, and promotes regression. In patients with type 1 diabetes, plasma S100A8/S100A9 levels correlate with leukocyte counts and coronary artery disease. Thus, hyperglycemia drives myelopoiesis and promotes atherogenesis in diabetes.


Assuntos
Aterosclerose/patologia , Hiperglicemia/patologia , Mielopoese/fisiologia , Animais , Aterosclerose/metabolismo , Medula Óssea/metabolismo , Medula Óssea/patologia , Doença das Coronárias/metabolismo , Doença das Coronárias/patologia , Citocinas/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Glucose/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Humanos , Hiperglicemia/metabolismo , Leucócitos/metabolismo , Leucócitos/patologia , Leucocitose/metabolismo , Leucocitose/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/metabolismo , Monócitos/patologia , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/patologia , NF-kappa B/metabolismo , Neutrófilos/metabolismo , Neutrófilos/patologia , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/metabolismo
9.
J Biol Chem ; 288(20): 14046-14058, 2013 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-23542081

RESUMO

Adipose fat storage is thought to require uptake of circulating triglyceride (TG)-derived fatty acids via lipoprotein lipase (LpL). To determine how LpL affects the biology of adipose tissue, we created adipose-specific LpL knock-out (ATLO) mice, and we compared them with whole body LpL knock-out mice rescued with muscle LpL expression (MCK/L0) and wild type (WT) mice. ATLO LpL mRNA and activity were reduced, respectively, 75 and 70% in gonadal adipose tissue (GAT), 90 and 80% in subcutaneous tissue, and 84 and 85% in brown adipose tissue (BAT). ATLO mice had increased plasma TG levels associated with reduced chylomicron TG uptake into BAT and lung. ATLO BAT, but not GAT, had altered TG composition. GAT from MCK/L0 was smaller and contained less polyunsaturated fatty acids in TG, although GAT from ATLO was normal unless LpL was overexpressed in muscle. High fat diet feeding led to less adipose in MCK/L0 mice but TG acyl composition in subcutaneous tissue and BAT reverted to that of WT. Therefore, adipocyte LpL in BAT modulates plasma lipoprotein clearance, and the greater metabolic activity of this depot makes its lipid composition more dependent on LpL-mediated uptake. Loss of adipose LpL reduces fat accumulation only if accompanied by greater LpL activity in muscle. These data support the role of LpL as the "gatekeeper" for tissue lipid distribution.


Assuntos
Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Tecido Adiposo/metabolismo , Lipase Lipoproteica/deficiência , Lipase Lipoproteica/genética , Adipócitos/citologia , Animais , Transplante de Medula Óssea , Quilomícrons/farmacocinética , Lipídeos/química , Lipólise , Macrófagos/citologia , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Triglicerídeos/sangue , Triglicerídeos/metabolismo
10.
Am J Physiol Endocrinol Metab ; 300(3): E489-99, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21139071

RESUMO

Normal hearts have increased contractility in response to catecholamines. Because several lipids activate PKCs, we hypothesized that excess cellular lipids would inhibit cardiomyocyte responsiveness to adrenergic stimuli. Cardiomyocytes treated with saturated free fatty acids, ceramide, and diacylglycerol had reduced cellular cAMP response to isoproterenol. This was associated with increased PKC activation and reduction of ß-adrenergic receptor (ß-AR) density. Pharmacological and genetic PKC inhibition prevented both palmitate-induced ß-AR insensitivity and the accompanying reduction in cell surface ß-ARs. Mice with excess lipid uptake due to either cardiac-specific overexpression of anchored lipoprotein lipase, PPARγ, or acyl-CoA synthetase-1 or high-fat diet showed reduced inotropic responsiveness to dobutamine. This was associated with activation of protein kinase C (PKC)α or PKCδ. Thus, several lipids that are increased in the setting of lipotoxicity can produce abnormalities in ß-AR responsiveness. This can be attributed to PKC activation and reduced ß-AR levels.


Assuntos
Lipídeos/fisiologia , Miócitos Cardíacos/metabolismo , Proteína Quinase C/fisiologia , Receptores Adrenérgicos beta/fisiologia , Animais , Western Blotting , Ceramidas/metabolismo , AMP Cíclico/metabolismo , Dieta , Gorduras na Dieta/farmacologia , Diglicerídeos/metabolismo , Ecocardiografia , Ativação Enzimática/fisiologia , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Miócitos Cardíacos/enzimologia , RNA/genética , RNA/isolamento & purificação , RNA Interferente Pequeno/genética
11.
J Biol Chem ; 285(49): 37976-86, 2010 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-20852327

RESUMO

Lipids circulate in the blood in association with plasma lipoproteins and enter the tissues either after hydrolysis or as non-hydrolyzable lipid esters. We studied cardiac lipids, lipoprotein lipid uptake, and gene expression in heart-specific lipoprotein lipase (LpL) knock-out (hLpL0), CD36 knock-out (Cd36(-/-)), and double knock-out (hLpL0/Cd36(-/-)-DKO) mice. Loss of either LpL or CD36 led to a significant reduction in heart total fatty acyl-CoA (control, 99.5 ± 3.8; hLpL0, 36.2 ± 3.5; Cd36(-/-), 57.7 ± 5.5 nmol/g, p < 0.05) and an additive effect was observed in the DKO (20.2 ± 1.4 nmol/g, p < 0.05). Myocardial VLDL-triglyceride (TG) uptake was reduced in the hLpL0 (31 ± 6%) and Cd36(-/-) (47 ± 4%) mice with an additive reduction in the DKO (64 ± 5%) compared with control. However, LpL but not CD36 deficiency decreased VLDL-cholesteryl ester uptake. Endogenously labeled mouse chylomicrons were produced by tamoxifen treatment of ß-actin-MerCreMer/LpL(flox/flox) mice. Induced loss of LpL increased TG levels >10-fold and reduced HDL by >50%. After injection of these labeled chylomicrons in the different mice, chylomicron TG uptake was reduced by ∼70% and retinyl ester by ∼50% in hLpL0 hearts. Loss of CD36 did not alter either chylomicron TG or retinyl ester uptake. LpL loss did not affect uptake of remnant lipoproteins from ApoE knock-out mice. Our data are consistent with two pathways for fatty acid uptake; a CD36 process for VLDL-derived fatty acid and a non-CD36 process for chylomicron-derived fatty acid uptake. In addition, our data show that lipolysis is involved in uptake of core lipids from TG-rich lipoproteins.


Assuntos
Antígenos CD36/metabolismo , VLDL-Colesterol/metabolismo , Quilomícrons/metabolismo , Ácidos Graxos/metabolismo , Metabolismo dos Lipídeos/fisiologia , Lipase Lipoproteica/metabolismo , Lipoproteínas VLDL/metabolismo , Miocárdio/metabolismo , Triglicerídeos/metabolismo , Animais , Antineoplásicos Hormonais/farmacocinética , Antígenos CD36/genética , VLDL-Colesterol/genética , Quilomícrons/genética , Ácidos Graxos/genética , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipase Lipoproteica/genética , Lipoproteínas VLDL/genética , Camundongos , Camundongos Knockout , Tamoxifeno/farmacologia , Triglicerídeos/genética
12.
Diabetes ; 57(6): 1674-82, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18346984

RESUMO

OBJECTIVE: Patients with diabetes often have dyslipidemia and increased postprandial lipidmia. Induction of diabetes in LDL receptor (Ldlr(-/-)) knockout mice also leads to marked dyslipidemia. The reasons for this are unclear. RESEARCH DESIGN AND METHODS: We placed Ldlr(-/-) and heterozygous LDL receptor knockout (Ldlr(+/-)) mice on a high-cholesterol (0.15%) diet, induced diabetes with streptozotocin (STZ), and assessed reasons for differences in plasma cholesterol. RESULTS: STZ-induced diabetic Ldlr(-/-) mice had plasma cholesterol levels more than double those of nondiabetic controls. Fast-performance liquid chromatography and ultracentrifugation showed an increase in both VLDL and LDL. Plasma VLDL became more cholesterol enriched, and both VLDL and LDL had a greater content of apolipoprotein (apo)E. In LDL the ratio of apoB48 to apoB100 was increased. ApoB production, assessed using [(35)S]methionine labeling in Triton WR1339-treated mice, was not increased in fasting STZ-induced diabetic mice. Similarly, postprandial lipoprotein production was not increased. Reduction of cholesterol in the diet to normalize the amount of cholesterol intake by the control and STZ-induced diabetic animals reduced plasma cholesterol levels in STZ-induced diabetic mice, but plasma cholesterol was still markedly elevated compared with nondiabetic controls. LDL from STZ-induced diabetic mice was cleared from the plasma and trapped more rapidly by livers of control mice. STZ treatment reduced liver expression of the proteoglycan sulfation enzyme, heparan sulfate N-deacetylase/N-sulfotrasferase-1, an effect that was reproduced in cultured hepatocytyes by a high glucose-containing medium. CONCLUSIONS: STZ-induced diabetic, cholesterol-fed mice developed hyperlipidemia due to a non-LDL receptor defect in clearance of circulating apoB-containing lipoproteins.


Assuntos
Colesterol/sangue , Diabetes Mellitus Experimental/sangue , Lipídeos/sangue , Lipoproteínas/sangue , Receptores de LDL/deficiência , Triglicerídeos/sangue , Animais , Apolipoproteínas B/sangue , Apolipoproteínas E/sangue , Glicemia/metabolismo , Colesterol na Dieta , Cruzamentos Genéticos , Diabetes Mellitus Experimental/fisiopatologia , Dislipidemias/genética , Fígado/fisiopatologia , Neoplasias Hepáticas , Neoplasias Hepáticas Experimentais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Tumorais Cultivadas
13.
Arterioscler Thromb Vasc Biol ; 28(3): 455-62, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18258818

RESUMO

OBJECTIVE: Although epidemiologic data suggest that hypertriglyceridemia and elevated plasma levels of fatty acids are toxic to arteries, in vitro correlates have been inconsistent. To investigate whether increased endothelial cell expression of lipoprotein lipase (LpL), the primary enzyme creating free fatty acids from circulating triglycerides (TG), affects vascular function, we created transgenic mice that express human LpL (hLpL) driven by the promoter and enhancer of the Tie2 receptor. METHODS AND RESULTS: Mice expressing this transgene, denoted EC-hLpL and L for low and H for high expression, had decreased plasma TG levels compared with wild-type mice (WT): 106+/-31 in WT, 37+/-17 (line H), and 63+/-31 mg/dL (line L) because of a reduction in VLDL TG; plasma cholesterol and HDL levels were unaltered. Crossing a high expressing EC-hLpL transgene onto the LpL knockout background allowed for survival of the pups; TG in these mice was approximately equal to that of heterozygous LpL knockout mice. Surprisingly, under control conditions the EC-hLpL transgene did not alter arterial function or endothelial cell gene expression; however, after tumor necrosis factor (TNF)-alpha treatment, arterial vascular cell adhesion molecule-1 (VCAM-1), E-selectin, and endogenous TNF-alpha mRNA levels were increased and arteries had impaired endothelium-dependent vasodilatation. This was associated with reduced eNOS dimers. CONCLUSIONS: Therefore, we hypothesize that excess vascular wall LpL augments vascular dysfunction in the setting of inflammation.


Assuntos
Células Endoteliais/fisiologia , Hipertrigliceridemia/enzimologia , Lipase Lipoproteica/biossíntese , Lipoproteínas VLDL/metabolismo , Vasculite/enzimologia , Vasodilatação/fisiologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Expressão Gênica , Genótipo , Humanos , Hipertrigliceridemia/complicações , Hipertrigliceridemia/patologia , Imuno-Histoquímica , Lipase Lipoproteica/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Distribuição Aleatória , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Triglicerídeos/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Vasculite/complicações
14.
J Pharmacol Exp Ther ; 313(2): 586-93, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15671204

RESUMO

hLpL(GPI) transgenic mice that overexpress human lipoprotein lipase (hLpL) with a glycosylphosphatidylinositol anchor on cardiomyocytes develop lipotoxic cardiomyopathy associated with increased cardiac uptake of plasma lipids. We hypothesized that peroxisome proliferator-activated receptor (PPAR)alpha, PPARgamma, or a PPARalpha/gamma agonist would alter cardiac function by modulating lipid uptake by the heart. hLpL(GPI) mice were administered rosiglitazone (10 mg/kg/day), fenofibrate (100 mg/kg/day), or DRF2655, an alkoxy propanoic acid analog (10 mg/kg/day), for 16 days. Rosiglitazone reduced plasma triglyceride (TG) from 107.63 +/- 6.98 to 77.61 +/- 3.98 mg/dl, whereas fenofibrate had no effect. DRF2655 reduced TG to 33.17 +/- 4.12 mg/dl. Rosiglitazone and DRF2655 decreased heart TG and total cholesterol; fenofibrate had no effect. Molecular markers for cardiac dysfunction, atrial natriuretic factor, brain natriuretic peptide, and tumor necrosis factor-alpha were decreased with rosiglitazone and increased with fenofibrate. Echocardiographic measurements showed reduced fractional shortening and increased left ventricular systolic dimension with fenofibrate. No changes in these parameters were observed with rosiglitazone or DRF2655 treatment. Muscle-specific carnitine palmitoyltransferase-1 and fatty acid transporter protein-1 gene expression were increased with fenofibrate and DRF2655 treatment; no change in expression of these genes was noted with rosiglitazone treatment. Rosiglitazone and DRF2655 reduced TG uptake by the heart, and fenofibrate treatment increased fatty acid uptake. Thus, in a lipotoxic cardiomyopathy mouse model, a PPARgamma agonist reduced cardiac lipid and markers of cardiomyopathy, whereas an agonist of PPARalpha did not improve cardiac lipids and worsened heart function. These changes were paralleled by alterations in heart lipid uptake. Overall, PPAR activators exhibit differential effects in this model of lipotoxic dilated cardiomyopathy.


Assuntos
Cardiomiopatias/metabolismo , Lipase Lipoproteica/biossíntese , Receptores Ativados por Proliferador de Peroxissomo/agonistas , Função Ventricular Esquerda/fisiologia , Animais , Cardiomiopatias/sangue , Cardiomiopatias/genética , Humanos , Lipase Lipoproteica/sangue , Lipase Lipoproteica/genética , Masculino , Camundongos , Camundongos Transgênicos , Oxazinas/farmacologia , Receptores Ativados por Proliferador de Peroxissomo/sangue , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Propionatos/farmacologia , Função Ventricular Esquerda/efeitos dos fármacos
15.
J Biol Chem ; 277(12): 10037-43, 2002 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-11790777

RESUMO

Although very low density lipoprotein (VLDL) receptor (VLDLr) knockout mice have been reported to have no lipoprotein abnormalities, they develop less adipose tissue than control mice when fed a high calorie diet. Mice that are deficient in adipose tissue expression of lipoprotein lipase (LpL) also have less fat, but only when crossed with ob/ob mice. We hypothesized that the VLDLr, a protein that will bind and transport LpL, is required for optimal LpL actions in vivo and that hypertriglyceridemia due to VLDLr deficiency is exacerbated by either LpL deficiency or VLDL overproduction. Fasted VLDLr knockout (VLDLr0) mice were more hypertriglyceridemic than controls (2-fold greater triglyceride levels). The hypertriglyceridemia due to VLDLr0 was even more evident when VLDLr0 mice were crossed with heterozygous LpL-deficient (LpL1) and human apolipoprotein B (apoB) transgenic mice. This was due to an increase in apoB48-containing VLDL. [(3)H]VLDL turnover studies showed that VLDL-triglyceride clearance in VLDLr0/LpL1 mice was impaired by 50% compared with LpL1 mice. VLDLr0/LpL1 mice had less LpL activity in postheparin plasma, heart, and skeletal muscle. Infection of mice with an adenovirus-expressing receptor-associated protein, an inhibitor of the VLDLr, reduced LpL activity in wild type but not VLDLr0 mice. Therefore, the VLDLr is required for normal LpL regulation in vivo, and the disruption of VLDLr results in hypertriglyceridemia associated with decreased LpL activity.


Assuntos
Lipase Lipoproteica/metabolismo , Receptores de LDL/genética , Receptores de LDL/fisiologia , Tecido Adiposo/metabolismo , Animais , Apolipoproteínas B/metabolismo , Arteriosclerose/sangue , Glicemia/metabolismo , Northern Blotting , Peso Corporal , Cromatografia Líquida de Alta Pressão , Genótipo , Glucose/metabolismo , Hipertrigliceridemia/genética , Immunoblotting , Cinética , Proteína Associada a Proteínas Relacionadas a Receptor de LDL/genética , Proteína Associada a Proteínas Relacionadas a Receptor de LDL/metabolismo , Lipase Lipoproteica/sangue , Lipoproteínas/sangue , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Músculos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Fatores de Tempo , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA