Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Molecules ; 25(19)2020 Sep 23.
Article in English | MEDLINE | ID: mdl-32977558

ABSTRACT

Overfeeding of a hypercaloric diet leads to obesity, diabetes, chronic inflammation, and fatty liver disease. Although limiting fat or carbohydrate intake is the cornerstone for obesity management, whether lowering fat or reducing carbohydrate intake is more effective for health management remains controversial. This study used murine models to determine how dietary fat and carbohydrates may influence metabolic disease manifestation. Age-matched C57BL/6J mice were fed 2 hypercaloric diets with similar caloric content, one with very high fat and low carbohydrate content (VHF) and the other with moderately high fat levels with high sucrose content (HFHS) for 12 weeks. Both groups gained more weight and displayed hypercholesterolemia, hyperglycemia, hyperinsulinemia, and liver steatosis compared to mice fed a normal low-fat (LF) diet. Interestingly, the VHF-fed mice showed a more robust adipose tissue inflammation compared to HFHS-fed mice, whereas HFHS-fed mice showed liver fibrosis and inflammation that was not observed in VHF-fed mice. Taken together, these results indicate macronutrient-specific tissue inflammation with excess dietary fat provoking adipose tissue inflammation, whereas moderately high dietary fat with extra sucrose is necessary and sufficient for hepatosteatosis advancement to steatohepatitis. Hence, liver and adipose tissues respond to dietary fat and sucrose in opposite manners, yet both macronutrients are contributing factors to metabolic diseases.


Subject(s)
Adipose Tissue/drug effects , Dietary Carbohydrates/pharmacology , Dietary Fats/adverse effects , Eating/drug effects , Liver/drug effects , Sucrose/adverse effects , Adipose Tissue/pathology , Animals , Inflammation/chemically induced , Inflammation/pathology , Insulin Resistance , Lipoproteins/blood , Liver/pathology , Male , Mice , Mice, Inbred C57BL
2.
Arterioscler Thromb Vasc Biol ; 37(6): 1046-1049, 2017 06.
Article in English | MEDLINE | ID: mdl-28473440

ABSTRACT

OBJECTIVE: Mice with adipocyte-specific inactivation of low-density lipoprotein receptor-related protein-1 (LRP1) are resistant to diet-induced obesity and hyperglycemia because of compensatory thermogenic response by muscle. However, the physiological function of LRP1 in mature adipocytes and its role in cardiovascular disease modulation are unknown. This study compared perivascular adipose tissues (PVAT) from wild-type (adLrp1+/+) and adipocyte-specific LRP1 knockout (adLrp1-/-) mice in modulation of atherosclerosis progression. APPROACH AND RESULTS: Analysis of adipose tissues from adLrp1+/+ and adLrp1-/- mice after Western diet feeding for 16 weeks revealed that, in comparison to adLrp1+/+ mice, the adipocytes in adLrp1-/- mice were smaller, but their adipose tissues were more inflamed with increased monocyte-macrophage infiltration and inflammatory gene expression. The transplantation of PVAT from chow-fed adLrp1+/+ and adLrp1-/- mice into the area surrounding the carotid arteries of Ldlr-/- mice before feeding the Western diet revealed a contributory role of PVAT toward hypercholesterolemia-induced atherosclerosis. Importantly, recipients of adLrp1-/- PVAT displayed a 3-fold increase in atherosclerosis compared with adLrp1+/+ PVAT recipients. The increased atherosclerosis invoked by LRP1-deficient PVAT was associated with elevated monocyte-macrophage infiltration and inflammatory cytokine expression in the transplanted fat. CONCLUSIONS: PVAT provide outside-in signals through the adventitia to modulate atherosclerotic lesion progression in response to hypercholesterolemia. Moreover, adipocytes with LRP1 deficiency are dysfunctional and more inflamed. This latter observation adds the adipose tissue to the list of anatomic sites where LRP1 expression is important to protect against diet-induced atherosclerosis.


Subject(s)
Adipocytes/metabolism , Atherosclerosis/metabolism , Diet, Western , Inflammation/metabolism , Receptors, LDL/deficiency , Tumor Suppressor Proteins/deficiency , Adipocytes/pathology , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Disease Models, Animal , Disease Progression , Genetic Predisposition to Disease , Hypercholesterolemia/genetics , Hypercholesterolemia/metabolism , Inflammation/genetics , Inflammation/pathology , Low Density Lipoprotein Receptor-Related Protein-1 , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, LDL/genetics , Signal Transduction , Tumor Suppressor Proteins/genetics
3.
J Biol Chem ; 291(32): 16610-9, 2016 08 05.
Article in English | MEDLINE | ID: mdl-27317662

ABSTRACT

LRP1 (LDL receptor-related protein-1) is a ubiquitous receptor with both cell signaling and ligand endocytosis properties. In the liver, LRP1 serves as a chylomicron remnant receptor and also participates in the transport of extracellular cathepsin D to the lysosome for prosaposin activation. The current study showed that in comparison with wild type mice, hepatocyte-specific LRP1 knock-out (hLrp1(-/-)) mice were more susceptible to fasting-induced lipid accumulation in the liver. Primary hepatocytes isolated from hLrp1(-/-) mice also accumulated more intracellular lipids and experienced higher levels of endoplasmic reticulum (ER) stress after palmitate treatment compared with similarly treated hLrp1(+/+) hepatocytes. Palmitate-treated hLrp1(-/-) hepatocytes displayed similar LC3-II levels, but the levels of p62 were elevated in comparison with palmitate-treated hLrp1(+/+) hepatocytes, suggesting that the elevated lipid accumulation in LRP1-defective hepatocytes was not due to defects in autophagosome formation but was due to impairment of lipophagic lipid hydrolysis in the lysosome. Additional studies showed increased palmitate-induced oxidative stress, mitochondrial and lysosomal permeability, and cell death in hLrp1(-/-) hepatocytes. Importantly, the elevated cell death and ER stress observed in hLrp1(-/-) hepatocytes were abrogated by E64D treatment, whereas inhibiting ER stress diminished cell death but not lysosomal permeabilization. Taken together, these results documented that LRP1 deficiency in hepatocytes promotes lipid accumulation and lipotoxicity through lysosomal-mitochondrial permeabilization and ER stress that ultimately result in cell death. Hence, LRP1 dysfunction may be a major risk factor in fatty liver disease progression.


Subject(s)
Endoplasmic Reticulum Stress , Fatty Liver , Hepatocytes , Oxidative Stress , Palmitic Acid/toxicity , Receptors, LDL/deficiency , Tumor Suppressor Proteins/deficiency , Animals , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Fatty Liver/chemically induced , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/pathology , Hepatocytes/metabolism , Hepatocytes/pathology , Low Density Lipoprotein Receptor-Related Protein-1 , Lysosomes/genetics , Lysosomes/metabolism , Lysosomes/pathology , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/pathology , Oxidative Stress/drug effects , Oxidative Stress/genetics , Permeability
4.
J Biol Chem ; 289(45): 31638-46, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25248747

ABSTRACT

Previous studies have shown that the myeloid-specific deficiency of the transcription factor Krüppel-like factor 2 (KLF2) accelerates atherosclerosis in hypercholesterolemic Ldlr(-/-) mice due to the enhanced adhesion of myeloid cells to activated endothelial cells in the vessel wall. This study revealed elevated basal inflammation with elevated plasma levels of Ccl2, Ccl4, Ccl5, and Ccl11 in the myeloid-specific KLF2 knock-out (myeKlf2(-/-)) mice. Peritoneal macrophages isolated from myeKlf2(-/-) mice showed increased mRNA levels of several inflammatory mediators, including Ccl2, Ccl5, Ccl7, Cox-2, Cxcl1, and IL-6. In contrast, the levels of two microRNAs, miR-124a and miR-150, were lower in Klf2(-/-) macrophages compared with Klf2(+/+) macrophages. Additional studies showed a direct inverse relationship between miR-124a levels with Ccl2 expression, with anti-miR-124a increasing Ccl2 mRNA levels in Klf2(+/+) macrophages, whereas the restoration of miR-124a levels in Klf2(-/-) macrophages significantly reduced Ccl2 mRNA expression. Likewise, the inverse relationship was observed between miR-150 levels and Cxcl1 expression in Klf2(+/+) and Klf2(-/-) mice. Moreover, miR150 likely regulates the miR124a expression and thus augments expression of inflammatory mediators in myeKlf2(-/-) macrophages. This study documented that the transcription factor KLF2 modulates inflammatory chemokine production via regulation of microRNA expression levels in immune cells.


Subject(s)
Gene Expression Regulation , Kruppel-Like Transcription Factors/genetics , Macrophages, Peritoneal/metabolism , MicroRNAs/blood , Animals , Atherosclerosis/genetics , Base Sequence , Binding Sites , Chemokines/metabolism , Female , Inflammation , Kruppel-Like Transcription Factors/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Myeloid Cells/cytology , Myeloid Cells/metabolism
5.
Arterioscler Thromb Vasc Biol ; 34(8): 1723-30, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24947528

ABSTRACT

OBJECTIVE: Perivascular adipose tissue (PVAT) expands during obesity, is highly inflamed, and correlates with coronary plaque burden and increased cardiovascular risk. We tested the hypothesis that PVAT contributes to the vascular response to wire injury and investigated the underlying mechanisms. APPROACH AND RESULTS: We transplanted thoracic aortic PVAT from donor mice fed a high-fat diet to the carotid arteries of recipient high-fat diet-fed low-density lipoprotein receptor knockout mice. Two weeks after transplantation, wire injury was performed, and animals were euthanized 2 weeks later. Immunohistochemistry was performed to quantify adventitial macrophage infiltration and neovascularization and neointimal lesion composition and size. Transplanted PVAT accelerated neointimal hyperplasia, adventitial macrophage infiltration, and adventitial angiogenesis. The majority of neointimal cells in PVAT-transplanted animals expressed α-smooth muscle actin, consistent with smooth muscle phenotype. Deletion of monocyte chemoattractant protein-1 in PVAT substantially attenuated the effects of fat transplantation on neointimal hyperplasia and adventitial angiogenesis, but not adventitial macrophage infiltration. Conditioned medium from perivascular adipocytes induced potent monocyte chemotaxis in vitro and angiogenic responses in cultured endothelial cells. CONCLUSIONS: These findings indicate that PVAT contributes to the vascular response to wire injury, in part through monocyte chemoattractant protein-1-dependent mechanisms.


Subject(s)
Adipose Tissue/transplantation , Carotid Artery Injuries/metabolism , Chemokine CCL2/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima , Actins/metabolism , Adipocytes/metabolism , Adipocytes/transplantation , Adipose Tissue/metabolism , Animals , Biomarkers/metabolism , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Injuries/etiology , Carotid Artery Injuries/genetics , Carotid Artery Injuries/pathology , Cells, Cultured , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Chemotaxis , Coculture Techniques , Culture Media, Conditioned/metabolism , Diet, High-Fat , Disease Models, Animal , Endothelial Cells/metabolism , Humans , Hyperplasia , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Neovascularization, Pathologic , Phenotype , Receptors, LDL/deficiency , Receptors, LDL/genetics , Signal Transduction , Time Factors , Transendothelial and Transepithelial Migration
6.
Biochim Biophys Acta ; 1842(9): 1395-405, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24840660

ABSTRACT

Genome-wide association studies have linked LRP8 polymorphisms to premature coronary artery disease and myocardial infarction in humans. However, the mechanisms by which dysfunctions of apolipoprotein E receptor-2 (apoER2), the protein encoded by LRP8 gene, influence atherosclerosis have not been elucidated completely. The current study focused on the role of apoER2 in macrophages, a cell type that plays an important role in atherosclerosis. Results showed that apoER2-deficient mouse macrophages accumulated more lipids and were more susceptible to oxidized LDL (oxLDL)-induced death compared to control cells. Consistent with these findings, apoER2 deficient macrophages also displayed defective serum-induced Akt activation and higher levels of the pro-apoptotic protein phosphorylated p53. Furthermore, the expression and activation of peroxisome proliferator-activated receptor γ (PPARγ) were increased in apoER2-deficient macrophages. Deficiency of apoER2 in hypercholesterolemic LDL receptor-null mice (Lrp8(-/-)Ldlr(-/-) mice) also resulted in accelerated atherosclerosis with more complex lesions and extensive lesion necrosis compared to Lrp8(+/+)Ldlr(-/-) mice. The atherosclerotic plaques of Lrp8(-/-)Ldlr(-/-) mice displayed significantly higher levels of p53-positive macrophages, indicating that the apoER2-deficient macrophages contribute to the accelerated atherosclerotic lesion necrosis observed in these animals. Taken together, this study indicates that apoER2 in macrophages limits PPARγ expression and protects against oxLDL-induced cell death. Thus, abnormal apoER2 functions in macrophages may at least in part contribute to the premature coronary artery disease and myocardial infarction in humans with LRP8 polymorphisms. Moreover, the elevated PPARγ expression in apoER2-deficient macrophages suggests that LRP8 polymorphism may be a genetic modifier of cardiovascular risk with PPARγ therapy.


Subject(s)
Hypercholesterolemia/pathology , LDL-Receptor Related Proteins/physiology , Macrophages/pathology , PPAR gamma/metabolism , Plaque, Atherosclerotic/pathology , Receptors, LDL/physiology , Animals , Apoptosis , Blotting, Western , Cells, Cultured , Female , Flow Cytometry , Humans , Hypercholesterolemia/complications , Hypercholesterolemia/metabolism , Lipids/analysis , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , Plaque, Atherosclerotic/etiology , Plaque, Atherosclerotic/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Risk Factors
7.
Adipocyte ; 3(4): 333-8, 2014.
Article in English | MEDLINE | ID: mdl-26317058

ABSTRACT

Adipose tissue serves as both a storage site for excess calories and as an endocrine organ, secreting hormones such as adiponectin that promote metabolic homeostasis. In obesity, adipose tissue expands primarily by hypertrophy (enlargement of existing adipocytes) rather than hyperplasia (generation of new adipocytes via adipogenic differentiation of preadipocytes). Progressive adipocyte hypertrophy leads to inflammation, insulin resistance, dyslipidemia, and ectopic lipid deposition, the hallmark characteristics of metabolic disease. We demonstrate that during chronic high fat feeding in mice, adipogenic differentiation is impaired due to the actions of histone deacetylase 9 (HDAC9), a member of the class II family of HDACs. Mechanistically, upregulated HDAC9 expression blocks the adipogenic differentiation program during chronic high fat feeding, leading to accumulation of improperly differentiated adipocytes with diminished expression of adiponectin. These adipocytes are inefficient at storing lipid, resulting in ectopic lipid deposition in the liver. HDAC9 gene deletion prevents the detrimental effects of chronic high fat feeding on adipogenic differentiation, increases adiponectin expression, and enhances energy expenditure by promoting beige adipogenesis, thus leading to reduced body mass and improved metabolic homeostasis. HDAC9 is therefore emerging as a critical regulator of adipose tissue health and a novel therapeutic target for obesity-related disease.

8.
Diabetes ; 63(1): 176-87, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24101673

ABSTRACT

During chronic caloric excess, adipose tissue expands primarily by enlargement of individual adipocytes, which become stressed with lipid overloading, thereby contributing to obesity-related disease. Although adipose tissue contains numerous preadipocytes, differentiation into functionally competent adipocytes is insufficient to accommodate the chronic caloric excess and prevent adipocyte overloading. We report for the first time that a chronic high-fat diet (HFD) impairs adipogenic differentiation, leading to accumulation of inefficiently differentiated adipocytes with blunted expression of adipogenic differentiation-specific genes. Preadipocytes from these mice likewise exhibit impaired adipogenic differentiation, and this phenotype persists during in vitro cell culture. HFD-induced impaired adipogenic differentiation is associated with elevated expression of histone deacetylase 9 (HDAC9), an endogenous negative regulator of adipogenic differentiation. Genetic ablation of HDAC9 improves adipogenic differentiation and systemic metabolic state during an HFD, resulting in diminished weight gain, improved glucose tolerance and insulin sensitivity, and reduced hepatosteatosis. Moreover, compared with wild-type mice, HDAC9 knockout mice exhibit upregulated expression of beige adipocyte marker genes, particularly during an HFD, in association with increased energy expenditure and adaptive thermogenesis. These results suggest that targeting HDAC9 may be an effective strategy for combating obesity-related metabolic disease.


Subject(s)
Adipose Tissue/metabolism , Histone Deacetylases/metabolism , Insulin Resistance/genetics , Metabolic Diseases/metabolism , Obesity/metabolism , Repressor Proteins/metabolism , Adipocytes/metabolism , Adipogenesis/physiology , Adiponectin/blood , Animals , Diet, High-Fat , Glucose Tolerance Test , Histone Deacetylases/genetics , Insulin/blood , Leptin/blood , Metabolic Diseases/genetics , Metabolic Diseases/prevention & control , Mice , Mice, Knockout , Obesity/genetics , Obesity/prevention & control , Repressor Proteins/genetics , Resistin/blood , Thermogenesis/physiology
9.
Biochim Biophys Acta ; 1841(6): 888-95, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24315825

ABSTRACT

Mice deficient in group 1b phospholipase A2 have decreased plasma lysophosphatidylcholine and increased hepatic oxidation that is inhibited by intraperitoneal lysophosphatidylcholine injection. This study sought to identify a mechanism for lysophosphatidylcholine-mediated inhibition of hepatic oxidative function. Results showed that in vitro incubation of isolated mitochondria with 40-200µM lysophosphatidylcholine caused cyclosporine A-resistant swelling in a concentration-dependent manner. However, when mitochondria were challenged with 220µM CaCl2, cyclosporine A protected against permeability transition induced by 40µM, but not 80µM lysophosphatidylcholine. Incubation with 40-120µM lysophosphatidylcholine also increased mitochondrial permeability to 75µM CaCl2 in a concentration-dependent manner. Interestingly, despite incubation with 80µM lysophosphatidylcholine, the mitochondrial membrane potential was steady in the presence of succinate, and oxidation rates and respiratory control indices were similar to controls in the presence of succinate, glutamate/malate, and palmitoyl-carnitine. However, mitochondrial oxidation rates were inhibited by 30-50% at 100µM lysophosphatidylcholine. Finally, while 40µM lysophosphatidylcholine has no effect on fatty acid oxidation and mitochondria remained impermeable in intact hepatocytes, 100µM lysophosphatidylcholine inhibited fatty acid stimulated oxidation and caused intracellular mitochondrial permeability. Taken together, these present data demonstrated that LPC concentration dependently modulates mitochondrial microenvironment, with low micromolar concentrations of lysophosphatidylcholine sufficient to change hepatic oxidation rate whereas higher concentrations are required to disrupt mitochondrial integrity.


Subject(s)
Lysophosphatidylcholines/administration & dosage , Mitochondria, Liver/metabolism , Oxidation-Reduction/drug effects , Permeability/drug effects , Animals , Calcium Chloride/administration & dosage , Cyclosporine/administration & dosage , Dose-Response Relationship, Drug , Group IV Phospholipases A2/deficiency , Group IV Phospholipases A2/genetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Lysophosphatidylcholines/metabolism , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/genetics , Mice , Mitochondria, Liver/drug effects
10.
PLoS One ; 8(11): e82026, 2013.
Article in English | MEDLINE | ID: mdl-24312398

ABSTRACT

Valvular disease is common in patients with Marfan syndrome and can lead to cardiomyopathy. However, some patients develop cardiomyopathy in the absence of hemodynamically significant valve dysfunction, suggesting alternative mechanisms of disease progression. Disruption of LDL receptor-related protein-1 (Lrp1) in smooth muscle cells has been shown to cause vascular pathologies similar to Marfan syndrome, with activation of smooth muscle cells, vascular dysfunction and aortic aneurysms. This study used echocardiography and blood pressure monitoring in mouse models to determine whether inactivation of Lrp1 in vascular smooth muscle leads to cardiomyopathy, and if so, whether the mechanism is a consequence of valvular disease. Hemodynamic changes during treatment with captopril were also assessed. Dilation of aortic roots was observed in young Lrp1-knockout mice and progressed as they aged, whereas no significant aortic dilation was detected in wild type littermates. Diastolic blood pressure was lower and pulse pressure higher in Lrp1-knockout mice, which was normalized by treatment with captopril. Aortic dilation was followed by development of aortic insufficiency and subsequent dilated cardiomyopathy due to valvular disease. Thus, smooth muscle cell Lrp1 deficiency results in aortic dilation and insufficiency that causes secondary cardiomyopathy that can be improved by captopril. These findings provide novel insights into mechanisms of cardiomyopathy associated with vascular activation and offer a new model of valvular cardiomyopathy.


Subject(s)
Aortic Valve Insufficiency/genetics , Blood Vessels/physiopathology , Cardiomyopathies/physiopathology , Gene Deletion , Low Density Lipoprotein Receptor-Related Protein-1/physiology , Muscle, Smooth/metabolism , Aging/physiology , Animals , Aortic Valve Insufficiency/diagnostic imaging , Blood Pressure , Cardiomyopathies/diagnostic imaging , Low Density Lipoprotein Receptor-Related Protein-1/genetics , MAP Kinase Signaling System , Male , Mice , Mice, Knockout , Phosphorylation , Smad2 Protein/metabolism , Ultrasonography
11.
J Lipid Res ; 54(5): 1430-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23434611

ABSTRACT

Ginsenoside Rb1 (Rb1), a natural compound extracted from ginseng, exerts anti-obesity activity and improves insulin sensitivity in high-fat diet (HFD)-induced obese rats. The objective of the current study was to evaluate the protective effect of Rb1 on fatty liver in HFD-induced obese rats and to elucidate underlying mechanisms. After chronic intraperitoneal administration, Rb1 (10 mg/kg) significantly ameliorated hepatic fat accumulation in HFD-induced obese rats, as demonstrated by reduced liver weight, hepatic triglyceride content, and histological evaluation of liver sections by hematoxylin and eosin and Oil Red O staining. Using primary cultured rat hepatic cells, we found that the rate of fatty acid oxidation and the activity of carnitine palmitoyltransferase 1 (CPT1), a key enzyme in fatty acid ß-oxidation, were significantly elevated in Rb1-treated hepatocytes compared with those of vehicle-treated cells. HPLC analysis revealed that Rb1 increased the cellular AMP/ATP ratio, which is associated with elevated activation of hepatic AMP-activated protein kinase (AMPK) and phosphorylated acetyl-CoA carboxylase. Consistent with the activation of AMPK, Rb1 stimulated the expression of genes encoding fatty acid oxidative enzymes and proteins, and suppressed the expression of genes encoding enzymes or proteins that function in lipogenesis, assessed by quantitative PCR. We conclude that Rb1 has a potent ability to reduce hepatic fat accumulation and might be useful as a therapeutic agent for fatty liver disorder.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Ginsenosides/administration & dosage , Obesity/drug therapy , Obesity/metabolism , Animals , Diet, High-Fat , Fat Body/drug effects , Fat Body/metabolism , Fatty Liver/drug therapy , Lipid Metabolism/drug effects , Lipogenesis/drug effects , Liver/drug effects , Liver/metabolism , Male , Obesity/pathology , Rats
12.
Diabetes ; 62(2): 382-91, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22961083

ABSTRACT

Genetic studies have revealed the association between the ε2 allele of the apolipoprotein E (apoE) gene and greater risk of metabolic diseases. This study compared C57BL/6 mice in which the endogenous mouse gene has been replaced by the human APOE2 or APOE3 gene (APOE2 and APOE3 mice) to identify the mechanism underlying the relationship between ε2 and obesity and diabetes. In comparison with APOE3 mice, the APOE2 mice had elevated fasting plasma lipid and insulin levels and displayed prolonged postprandial hyperlipidemia accompanied by increased granulocyte number and inflammation 2 h after being fed a lipid-rich meal. In comparison with APOE3 mice, the APOE2 mice also showed increased adiposity when maintained on a Western-type, high-fat, high-cholesterol diet. Adipose tissue dysfunction with increased macrophage infiltration, abundant crown-like structures, and inflammation were also observed in adipose tissues of APOE2 mice. The severe adipocyte dysfunction and tissue inflammation corresponded with the robust hyperinsulinemia observed in APOE2 mice after being fed the Western-type diet. Taken together, these data showed that impaired plasma clearance of apoE2-containing, triglyceride-rich lipoproteins promotes lipid redistribution to neutrophils and adipocytes to accentuate inflammation and adiposity, thereby accelerating the development of hyperinsulinemia that will ultimately lead to advanced metabolic diseases.


Subject(s)
Apolipoprotein E2/metabolism , Hyperinsulinism/metabolism , Inflammation/metabolism , Obesity/metabolism , Postprandial Period/physiology , Adipocytes/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adiposity/genetics , Adiposity/physiology , Animals , Apolipoprotein E2/genetics , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Cholesterol, Dietary/adverse effects , Cholesterol, Dietary/metabolism , Diet, High-Fat/adverse effects , Fasting/metabolism , Granulocytes/metabolism , Humans , Hyperinsulinism/etiology , Hyperlipidemias/blood , Hyperlipidemias/etiology , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Inflammation/etiology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity/etiology
13.
J Biol Chem ; 287(33): 27876-84, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22730380

ABSTRACT

Apolipoprotein (apo) E4 is a major genetic risk factor for a wide spectrum of inflammatory metabolic diseases, including atherosclerosis, diabetes, and Alzheimer disease. This study compared diet-induced adipose tissue inflammation as well as functional properties of macrophages isolated from human APOE3 and APOE4 mice to identify the mechanism responsible for the association between apoE4 and inflammatory metabolic diseases. The initial study confirmed previous reports that APOE4 gene replacement mice were less sensitive than APOE3 mice to diet-induced body weight gain but exhibited hyperinsulinemia, and their adipose tissues were similarly inflamed as those in APOE3 mice. Peritoneal macrophages isolated from APOE4 mice were defective in efferocytosis compared with APOE3 macrophages. Increased cell death was also observed in APOE4 macrophages when stimulated with LPS or oxidized LDL. Western blot analysis of cell lysates revealed that APOE4 macrophages displayed elevated JNK phosphorylation indicative of cell stress even under basal culturing conditions. Significantly higher cell stress due mainly to potentiation of endoplasmic reticulum (ER) stress signaling was also observed in APOE4 macrophages after LPS and oxidized LDL activation. The defect in efferocytosis and elevated apoptosis sensitivity of APOE4 macrophages was ameliorated by treatment with the ER chaperone tauroursodeoxycholic acid. Taken together, these results showed that apoE4 expression causes macrophage dysfunction and promotes apoptosis via ER stress induction. The reduction of ER stress in macrophages may be a viable option to reduce inflammation and inflammation-related metabolic disorders associated with the apoE4 polymorphism.


Subject(s)
Apolipoprotein E4/metabolism , Endoplasmic Reticulum Stress , Macrophages, Peritoneal/metabolism , Signal Transduction , Adipose Tissue/metabolism , Adipose Tissue/pathology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Apolipoprotein E4/genetics , Apoptosis/drug effects , Apoptosis/genetics , Cells, Cultured , Cholagogues and Choleretics/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hyperinsulinism/genetics , Hyperinsulinism/metabolism , Hyperinsulinism/pathology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/pharmacology , Lipoproteins, LDL/genetics , Lipoproteins, LDL/metabolism , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Macrophages, Peritoneal/pathology , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Metabolic Diseases/pathology , Mice , Mice, Transgenic , Phosphorylation , Polymorphism, Genetic , Taurochenodeoxycholic Acid/pharmacology
14.
Circ Res ; 110(10): 1294-302, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22474254

ABSTRACT

RATIONALE: Hemizygous deficiency of the transcription factor Krüppel-like factor 2 (KLF2) has been shown previously to augment atherosclerosis in hypercholesterolemic mice. However, the cell type responsible for the increased atherosclerosis due to KLF2 deficiency has not been identified. This study examined the consequence of myeloid cell-specific KLF2 inactivation in atherosclerosis. METHODS AND RESULTS: Cell-specific knockout mice were generated by Cre/loxP recombination. Macrophages isolated from myeloid-specific Klf2 knockout (myeKlf2(-/-)) mice were similar to myeKlf2(+/+) macrophages in response to activation, polarization, and lipid accumulation. However, in comparison to myeKlf2(+/+) macrophages, myeKlf2(-/-) macrophages adhered more robustly to endothelial cells. Neutrophils from myeKlf2(-/-) mice also adhered more robustly to endothelial cells, and fewer myeKlf2(-/-) neutrophils survived in culture over a 24-hour period in comparison with myeKlf2(+/+) neutrophils. When myeKlf2(-/-) mice were mated to Ldlr(-/-) mice and then fed a high fat and high cholesterol diet, significant increase in atherosclerosis was observed in the myeKlf2(-/-)Ldlr(-/-) mice compared with myeKlf2(+/+)Ldlr(-/-) littermates. The increased atherosclerosis in myeKlf2(-/-)Ldlr(-/-) mice was associated with elevated presence of neutrophils and macrophages, with corresponding increase of myeloperoxidase as well as chlorinated and nitrosylated tyrosine epitopes in their lesion areas compared with myeKlf2(+/+)Ldlr(-/-) mice. CONCLUSIONS: This study documents a role for myeloid KLF2 expression in modulating atherosclerosis. The increased neutrophil accumulation and atherosclerosis progression with myeloid-specific KLF2 deficiency also underscores the importance of neutrophils in promoting vascular oxidative stress and atherosclerosis. Collectively, these results suggest that elevating KLF2 expression may be a novel strategy for prevention and treatment of atherosclerosis.


Subject(s)
Atherosclerosis/immunology , Cell Adhesion/immunology , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Macrophages/immunology , Neutrophils/immunology , Animals , Atherosclerosis/pathology , Cell Death/immunology , Endothelial Cells/cytology , Endothelial Cells/immunology , Female , Hypercholesterolemia/immunology , Hypercholesterolemia/pathology , Lymphocyte Count , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/cytology , Vasculitis/immunology , Vasculitis/pathology
15.
J Biol Chem ; 286(15): 13079-87, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21343303

ABSTRACT

The low density lipoprotein receptor-related protein-1 (LRP1) is known to serve as a chylomicron remnant receptor in the liver responsible for the binding and plasma clearance of apolipoprotein E-containing lipoproteins. Previous in vitro studies have provided evidence to suggest that LRP1 expression may also influence high density lipoprotein (HDL) metabolism. The current study showed that liver-specific LRP1 knock-out (hLrp1(-/-)) mice displayed lower fasting plasma HDL cholesterol levels when compared with hLrp1(+/+) mice. Lecithin:cholesterol acyl transferase and hepatic lipase activities in plasma of hLrp1(-/-) mice were comparable with those observed in hLrp1(+/+) mice, indicating that hepatic LRP1 inactivation does not influence plasma HDL remodeling. Plasma clearance of HDL particles and HDL-associated cholesteryl esters was also similar between hLrp1(+/+) and hLrp1(-/-) mice. In contrast, HDL secretion from primary hepatocytes isolated from hLrp1(-/-) mice was significantly reduced when compared with that observed with hLrp1(+/+) hepatocytes. Biotinylation of cell surface proteins revealed decreased surface localization of the ATP-binding cassette, subfamily A, member 1 (ABCA1) protein, but total cellular ABCA1 level was not changed in hLrp1(-/-) hepatocytes. Finally, hLrp1(-/-) hepatocytes displayed reduced binding capacity for extracellular cathepsin D, resulting in lower intracellular cathepsin D content and impairment of prosaposin activation, a process that is required for membrane translocation of ABCA1 to facilitate cholesterol efflux and HDL secretion. Taken together, these results documented that hepatic LRP1 participates in cellular activation of lysosomal enzymes and through this mechanism, indirectly modulates the production and plasma levels of HDL.


Subject(s)
Cell Membrane/metabolism , Hepatocytes/metabolism , Lipoproteins, HDL/blood , Liver/metabolism , Lysosomes/metabolism , Receptors, LDL/metabolism , Tumor Suppressor Proteins/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Cathepsin D/genetics , Cathepsin D/metabolism , Cell Membrane/genetics , Fasting/blood , Lipase/genetics , Lipase/metabolism , Lipoproteins, HDL/genetics , Low Density Lipoprotein Receptor-Related Protein-1 , Lysosomes/genetics , Mice , Mice, Knockout , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Protein Transport/physiology , Receptors, LDL/genetics , Saposins/genetics , Saposins/metabolism , Tumor Suppressor Proteins/genetics
16.
Arterioscler Thromb Vasc Biol ; 30(6): 1159-65, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20299691

ABSTRACT

OBJECTIVE: Adiponectin is an adipocyte-derived, secreted protein that is implicated in protection against a cluster of related metabolic disorders. Mice lacking adiponectin display impaired hepatic insulin sensitivity and respond only partially to peroxisome proliferator-activated receptor gamma agonists. Adiponectin has been associated with antiinflammatory and antiatherogenic properties; however, the direct involvement of adiponectin on the atherogenic process has not been studied. METHODS AND RESULTS: We crossed adiponectin knockout mice (Adn(-/-)) or mice with chronically elevated adiponectin levels (Adn(Tg)) into the low-density lipoprotein receptor-null (Ldlr(-/-)) and the apoliprotein E-null (Apoe(-/-)) mouse models. Adiponectin levels did not correlate with a suppression of the atherogenic process. Plaque volume in the aortic root, cholesterol accumulation in the aorta, and plaque morphology under various dietary conditions were not affected by circulating adiponectin levels. In light of the strong associations reported for adiponectin with cardiovascular disease in humans, the lack of a phenotype in gain- and loss-of-function studies in mice suggests a lack of causation for adiponectin in inhibiting the buildup of atherosclerotic lesions. CONCLUSIONS: These data indicate that the actions of adiponectin on the cardiovascular system are complex and multifaceted, with a minimal direct impact on atherosclerotic plaque formation in preclinical rodent models.


Subject(s)
Aortic Diseases/metabolism , Atherosclerosis/metabolism , Acetates/pharmacology , Adiponectin/blood , Adiponectin/deficiency , Adiponectin/genetics , Adiponectin/metabolism , Animals , Aortic Diseases/blood , Aortic Diseases/drug therapy , Aortic Diseases/genetics , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Atherosclerosis/pathology , Cholesterol/metabolism , Disease Models, Animal , Female , Genotype , Indoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , PPAR gamma/agonists , PPAR gamma/metabolism , Phenotype , Receptors, LDL/deficiency , Receptors, LDL/genetics , Time Factors
17.
Arterioscler Thromb Vasc Biol ; 29(11): 1772-8, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19729608

ABSTRACT

OBJECTIVE: Defective smooth muscle expression of LDL receptor-related protein-1 (Lrp1) increases atherosclerosis in hypercholesterolemic mice. This study explored the importance of smooth muscle Lrp1 expression under normolipidemic conditions. METHODS AND RESULTS: Smooth muscle cells isolated from control (smLrp1(+/+)) and smooth muscle-specific Lrp1 knockout (smLrp1(-/-)) mice were characterized based on morphology, smooth muscle marker protein expression levels, and growth rates in vitro. Vascular functions were assessed by aortic constrictive response to agonist stimulation in situ and neointimal hyperplasia to carotid arterial injury in vivo. The smLrp1(-/-) smooth muscle cells displayed reduced alpha-actin and calponin expression and an accelerated growth rate attribtuable to sustained phosphorylation of platelet-derived growth factor receptor (PRGFR) and protein kinase B/Akt. Vasoconstrictive response to agonist stimulation was impaired in aortic rings isolated from smLrp1(-/-) mice. Injury-induced neointimal hyperplasia was significantly increased in smLrp1(-/-) mice. The increase in neointima was associated with corresponding elevated activation of PDGFR signaling pathway. CONCLUSIONS: Smooth muscle expression of Lrp1 is important in maintaining normal vascular functions under normolipidemic conditions. The absence of Lrp1 expression results in greater smooth muscle cell proliferation, deficient contractile protein expression, impairment of vascular contractility, and promotion of denudation-induced neointimal hyperplasia.


Subject(s)
Carotid Artery Injuries/pathology , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Muscle, Smooth, Vascular/metabolism , Platelet-Derived Growth Factor/pharmacology , Receptors, LDL/metabolism , Tunica Intima/metabolism , Analysis of Variance , Animals , Blotting, Western , Carotid Artery Injuries/metabolism , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Immunohistochemistry , Low Density Lipoprotein Receptor-Related Protein-1/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Probability , Random Allocation , Reference Values , Tunica Intima/cytology , Vasoconstriction/physiology
18.
Diabetes ; 57(1): 5-12, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17914034

ABSTRACT

OBJECTIVE: Apolipoprotein E (ApoE) regulates plasma lipid levels via modulation of lipolysis and serving as ligand for receptor-mediated clearance of triglyceride (TG)-rich lipoproteins. This study tested the impact of modulating lipid delivery to tissues on insulin responsiveness and diet-induced obesity. RESEARCH DESIGN AND METHODS: ApoE(+/+) and apoE(-/-) mice were placed on high-fat-high-sucrose diabetogenic diet or control diet for 24 weeks. Plasma TG clearance, glucose tolerance, and tissue uptake of dietary fat and glucose were assessed. RESULTS: Plasma TG clearance and lipid uptake by adipose tissue were impaired, whereas glucose tolerance was improved in control diet-fed apoE(-/-) mice compared with apoE(+/+) mice after an oral lipid load. Fat mass was reduced in apoE(-/-) mice compared with apoE(+/+) mice under both dietary conditions. The apoE(-/-) mice exhibited lower body weight and insulin levels than apoE(+/+) mice when fed the diabetogenic diet. Glucose tolerance and uptake by muscle and brown adipose tissue (BAT) was also improved in mice lacking apoE when fed the diabetogenic diet. Indirect calorimetry studies detected no difference in energy expenditure and respiratory quotient between apoE(+/+) and apoE(-/-) mice on control diet. Energy expenditure and uncoupling protein-1 expression in BAT were slightly but not significantly increased in apoE(-/-) mice on diabetogenic diet. CONCLUSIONS: These results demonstrated that decreased lipid delivery to insulin-sensitive tissues improves insulin sensitivity and ameliorates diet-induced obesity.


Subject(s)
Apolipoproteins E/deficiency , Lipids/blood , Animals , Blood Glucose/metabolism , Deoxyglucose/metabolism , Diet, Diabetic , Energy Metabolism , Glucose Tolerance Test , Lipoproteins/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Triglycerides/blood , Tritium
19.
J Clin Invest ; 117(11): 3271-82, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17948131

ABSTRACT

Diet-induced obesity and its serious consequences such as diabetes, cardiovascular disease, and cancer are rapidly becoming a major global health threat. Therefore, understanding the cellular and molecular mechanisms by which dietary fat causes obesity and diabetes is of paramount importance in order to identify preventive and therapeutic strategies. Increased dietary fat intake results in high plasma levels of triglyceride-rich lipoproteins (TGRL). Tissue uptake of TGRL has been shown to promote glucose intolerance. We generated mice with an adipocyte-specific inactivation of the multifunctional receptor LDL receptor-related protein-1 (LRP1) to determine its role in mediating the effects of TGRL on diet-induced obesity and diabetes. Knockout mice displayed delayed postprandial lipid clearance, reduced body weight, smaller fat stores, lipid-depleted brown adipocytes, improved glucose tolerance, and elevated energy expenditure due to enhanced muscle thermogenesis. We further demonstrated that inactivation of adipocyte LRP1 resulted in resistance to dietary fat-induced obesity and glucose intolerance. These findings identify LRP1 as a critical regulator of adipocyte energy homeostasis, where functional disruption leads to reduced lipid transport, increased insulin sensitivity, and muscular energy expenditure.


Subject(s)
Adipocytes/metabolism , Glucose/metabolism , Homeostasis , Lipid Metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Tumor Suppressor Proteins/metabolism , Adipocytes/cytology , Adipose Tissue/anatomy & histology , Adipose Tissue/metabolism , Animals , Behavior, Animal/physiology , Biological Transport/physiology , Diet , Dietary Fats , Energy Metabolism , Glucose Intolerance/metabolism , Lipoproteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Postprandial Period , Tumor Suppressor Proteins/genetics
20.
Neurobiol Aging ; 26(3): 317-23, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15639309

ABSTRACT

Over the years, the vascular protective role of apolipoprotein (apo) E has been attributed to the ability of apoE to induce cholesterol efflux from macrophage foam cells and its transport of extrahepatic cholesterol to the liver for excretion out of the body. Recently, apoE has been shown to protect against vascular disease by additional mechanisms that are independent of its cholesterol transport functions. This review summarizes data demonstrating apoE binding to specific cell surface receptors and proteoglycans in smooth muscle cells triggers distinct signalling pathways that result in inhibition of cell migration, proliferation, and excessive extracellular matrix deposition. apoE binding to the low density lipoprotein receptor-related protein is responsible for inhibition of cell migration, due to the induction of cyclic AMP accumulation and protein kinase A activation. apoE inhibition of cell proliferation is mediated by its binding to proteoglycans and the resulting activation of inducible nitric oxide synthase. apoE also inhibits excessive extracellular matrix protein synthesis. The receptor responsible for this latter apoE function remains to be identified.


Subject(s)
Apolipoproteins E/metabolism , Cell Movement/physiology , Cell Proliferation , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Signal Transduction/physiology , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Down-Regulation , Gene Expression Regulation/physiology , Heparan Sulfate Proteoglycans/metabolism , Mice , Muscle, Smooth, Vascular/cytology , Nitric Oxide/metabolism , Protein Isoforms/metabolism , Protein Kinase C/metabolism , Receptors, Cell Surface/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...