Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Nutr Metab Cardiovasc Dis ; 28(6): 582-591, 2018 06.
Article in English | MEDLINE | ID: mdl-29691148

ABSTRACT

BACKGROUND AND AIMS: Angiotensin receptor blockers (ARB) and angiotensin converting enzyme inhibitors (ACEI) reduce cardiovascular events in the general population. Maintenance hemodialysis (MHD) patients are at high cardiovascular risk but few studies have directly addressed the comparative efficacy of these drugs. MHD disrupts the normally atheroprotective actions of high density lipoprotein (HDL), therefore, we compared ACEI or ARB treatment on HDL functions in MHD. METHODS AND RESULTS: HDL was isolated at the starting point (pre) and 3-6 months later (post) in 30 MHD randomly assigned to placebo, ramipril or valsartan. Outcomes included cholesterol efflux, inflammatory cytokine response, effects on Toll-like receptors (TLR), superoxide production, methylarginine and serum amyloid A (SAA) levels. HDL from ARB- or ACEI-treated subjects was more effective in maintaining efflux than HDL of placebo. HDL from ARB- or ACEI-treated subjects but not placebo lessened cellular superoxide production. In contrast, neither ARB nor ACEI improved HDL anti-inflammatory effect. Indeed, HDL of ACEI-treated subjects potentiated the cytokine responses in association with activation of TLR but did not alter the HDL content of methylarginines or SAA. CONCLUSION: Both ACEI and ARB stabilized HDL cholesterol acceptor function and sustained cellular anti-oxidative effects but not anti-inflammatory effects, and ACEI-treatment instead amplified the HDL inflammatory response. The findings reveal possible utility of antagonizing angiotensin actions in MDH and suggest a possible mechanism for superiority of ARB vs ACEI in the setting of advanced kidney disease.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Cholesterol, HDL/blood , Kidney Failure, Chronic/therapy , Ramipril/therapeutic use , Renal Dialysis , Valsartan/therapeutic use , Adult , Angiotensin II Type 1 Receptor Blockers/adverse effects , Angiotensin-Converting Enzyme Inhibitors/adverse effects , Biomarkers/blood , Double-Blind Method , Female , Humans , Inflammation Mediators/blood , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/diagnosis , Male , Middle Aged , Oxidative Stress/drug effects , Ramipril/adverse effects , Renal Dialysis/adverse effects , Tennessee , Time Factors , Treatment Outcome , Valsartan/adverse effects
2.
Lupus ; 22(1): 26-33, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23060481

ABSTRACT

Free fatty acids (FFAs) are implicated in the pathogenesis of insulin resistance and atherosclerosis. Inflammatory cytokines promote lipolysis and increase FFAs, a cause of endothelial dysfunction and increased atherosclerosis risk. We hypothesized that increased inflammation is associated with increased FFAs, resulting in insulin resistance and atherosclerosis in patients with systemic lupus erythematosus (SLE). We measured clinical variables, serum FFAs, homeostasis model assessment for insulin resistance (HOMA), inflammatory cytokines, markers of endothelial activation, cholesterol concentrations and coronary artery calcium in 156 patients with SLE and 90 controls. We compared FFAs in patients with SLE and controls using Wilcoxon rank sum tests and further tested for the independent association between FFAs and disease status with adjustment for age, race and sex using multivariable regression models. We assessed the relationship between FFAs and continuous variables of interest using Spearman correlation and multivariable regression analysis. Levels of FFAs were higher in patients with SLE than controls (0.55 mmol/l (0.37-0.71) vs 0.44 mmol/l (0.32-0.60), P = 0.02). Levels of FFAs remained significantly higher among patients with SLE after adjustment for age, race and sex (P = 0.03) but not after further adjustment for body mass index (P = 0.13). FFA levels did not differ according to the usage of current immunosuppressive medications in univariate and adjusted analysis (all P > 0.05). Among patients with SLE, concentrations of FFAs were higher among those with metabolic syndrome compared to those without (0.66 mmol/l (0.46-0.81) vs 0.52 mmol/l (0.35-0.66), P < 0.001). FFAs were positively correlated with insulin resistance (HOMA) (rho = 0.23, P = 0.004, P adjusted = 0.006) and triglyceride levels (rho = 0.22, P = 0.01, P adjusted = 0.004). FFAs were not associated with inflammatory cytokines (IL-6, TNF-α) (all P > 0.05) but were positively associated with levels of E-selectin (rho = 0.33, P = < 0.001, P adjusted = 0.001) and ICAM-1 (rho = 0.35, P < 0.001, P adjusted = 0.001). FFAs were correlated with coronary artery calcium score (rho = 0.20, P = 0.01) but this was attenuated after adjustment for age, race and sex (P = 0.33). From our study we concluded that FFAs are elevated in patients with SLE, particularly those with metabolic syndrome. FFAs in patients with SLE are not associated with markers of generalized inflammation but are associated with insulin resistance and markers of endothelial activation.


Subject(s)
Fatty Acids, Nonesterified/blood , Inflammation/blood , Insulin Resistance , Lupus Erythematosus, Systemic/blood , Metabolic Syndrome/blood , Adult , Biomarkers/blood , Calcium/metabolism , Case-Control Studies , Cholesterol/blood , Coronary Angiography/methods , Coronary Artery Disease/blood , Coronary Artery Disease/diagnosis , Coronary Artery Disease/epidemiology , Coronary Vessels/diagnostic imaging , Coronary Vessels/metabolism , Cross-Sectional Studies , Cytokines/blood , Endothelial Cells/immunology , Endothelial Cells/metabolism , Female , Humans , Immunosuppressive Agents/therapeutic use , Inflammation/diagnosis , Inflammation/epidemiology , Inflammation/immunology , Inflammation Mediators/blood , Logistic Models , Lupus Erythematosus, Systemic/diagnosis , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/epidemiology , Lupus Erythematosus, Systemic/immunology , Male , Metabolic Syndrome/diagnosis , Metabolic Syndrome/epidemiology , Metabolic Syndrome/immunology , Middle Aged , Multivariate Analysis , Odds Ratio , Prevalence , Prognosis , Risk Factors , Tennessee/epidemiology , Tomography, X-Ray Computed , Triglycerides/blood , Up-Regulation
3.
Gene Ther ; 19(11): 1041-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22130447

ABSTRACT

In gene therapy, tissue-specific promoters are useful tools to direct transgene expression and improve efficiency and safety. Macrophage-specific promoters (MSPs) have previously been published using different delivery systems. In this study, we evaluated five different MSPs fused with green fluorescent protein (GFP) to delineate the one with highest specificity using lentiviral delivery. We compared three variants of the CD68 promoter (full length, the 343-bp proximal part and the 150-bp proximal part) and two variants (in forward and reverse orientation) of a previously characterized synthetic promoter derived from elements of transcription factor genes. We transduced a number of cell lines and primary cells in vitro. In addition, hematopoietic stem cells were transduced with MSPs and transferred into lethally irradiated recipient mice. Fluorescence activated cell sorting analysis was performed to determine the GFP expression in different cell populations both in vitro and in vivo. We showed that MSPs can efficiently be used for lentiviral gene delivery and that the 150-bp proximal part of the CD68 promoter provides primarily macrophage-specific expression of GFP. We propose that this is the best currently available MSP to use for directing transgene expression to macrophage populations in vivo using lentiviral vectors.


Subject(s)
Genetic Vectors/genetics , Lentivirus/genetics , Macrophages/metabolism , Promoter Regions, Genetic , Animals , Cell Line , Gene Dosage , Gene Expression , Gene Order , Genetic Therapy , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Mice , Organ Specificity/genetics , Transduction, Genetic , Transgenes
4.
Nutr Metab Cardiovasc Dis ; 22(5): 409-16, 2012 May.
Article in English | MEDLINE | ID: mdl-21277179

ABSTRACT

BACKGROUND AND AIMS: Diet may play an important role in the development of hyperuricemia and gout. However, the association between dietary factors and hyperuricemia remains unclear, and few studies have investigated direct links between food intake and hyperuricemia. The aim of this study was to investigate associations between high purine-content foods and protein intake with the prevalence of hyperuricemia by using data from a cross-sectional study of 3978 men aged 40-74 yrs living in Shanghai, China. METHODS AND RESULTS: Hyperuricemia was defined as blood uric acid level >7.0 mg/dl. One quarter of this population had hyperuricemia. Dietary information was collected by using a food frequency questionnaire. We collected information on anthropometric measurements and lifestyle factors and other potential confounding factors and disease history via interviews. Total protein consumption was not associated with hyperuricemia. We found a positive association between protein from animal sources and prevalence of hyperuricemia and an inverse association between protein from plant sources and hyperuricemia. However, these associations failed to reach significance in mutually adjusted analysis. Seafood intake was associated with higher prevalence of hyperuricemia. The ORs for quintiles of seafood intake (including fish and shellfish) were 1.00, 1.49, 1.35, 1.34, and 1.56 (p for trend: 0.01). An inverse association approaching significance between soy food consumption and hyperuricemia was observed (ORs: 1.00, 0.90, 0.70, 0.89, and 0.77 for quintiles of intake; p for trend: 0.07). No associations between consumption of purine-rich vegetables or meat and prevalence of hyperuricemia were observed. CONCLUSIONS: Our data suggest a direct association between seafood consumption and hyperuricemia and an inverse association between consumption of soy food and hyperuricemia among middle-aged, Chinese men.


Subject(s)
Dietary Proteins/adverse effects , Hyperuricemia/etiology , Purines/adverse effects , Urban Health , Adult , Aged , China/epidemiology , Cohort Studies , Cross-Sectional Studies , Diet/adverse effects , Diet/ethnology , Dietary Proteins/administration & dosage , Humans , Hyperuricemia/blood , Hyperuricemia/epidemiology , Hyperuricemia/ethnology , Male , Meat/adverse effects , Meat/analysis , Middle Aged , Prevalence , Purines/administration & dosage , Purines/analysis , Seafood/adverse effects , Seafood/analysis , Soy Foods/adverse effects , Soy Foods/analysis , Surveys and Questionnaires , Urban Health/ethnology , Uric Acid/blood
5.
Nutr Metab Cardiovasc Dis ; 22(3): 223-30, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21111583

ABSTRACT

BACKGROUND AND AIM: Increased levels of C-reactive protein (CRP), common in aging populations, are associated with higher risk for chronic diseases, including diabetes and coronary heart disease. The aim of this study was to investigate associations between lifestyle factors and high CRP among middle-aged men living in Shanghai, China. METHODS AND RESULTS: In this cross-sectional study, 3978 urban Chinese men aged 40-74 years who were free of type-2 diabetes at baseline provided fasting blood samples, anthropometric measurements and information on lifestyle factors and disease history. Dietary patterns were assessed by factor analysis. Participants were categorised into two groups according to CRP level: normal (≤ 3 mg/L) and high (> 3 mg/L). Associations between CRP categories and lifestyle factors were investigated by using logistic regression. Obesity, weight gain, cigarette smoking and alcohol intake were positively associated with high CRP levels, while physical activity and a dietary pattern with high consumption of fruit were inversely related to high CRP levels. A positive trend of marginal significance between quintiles of a dietary pattern with high consumption of meat and high CRP levels was also observed. No association between tea intake and CRP level was observed. CONCLUSIONS: Components of an adverse lifestyle were associated with high CRP levels. Obesity, smoking and alcohol intake were associated with high CRP, a biomarker of low-grade inflammation in middle-aged men, while a dietary pattern rich in fruit and high physical activity were inversely associated with the prevalence of high CRP.


Subject(s)
Asian People , C-Reactive Protein/analysis , Inflammation Mediators/blood , Inflammation/ethnology , Life Style/ethnology , Urban Population , Adult , Age Factors , Aged , Alcohol Drinking/ethnology , Alcohol Drinking/immunology , China/epidemiology , Cross-Sectional Studies , Exercise , Factor Analysis, Statistical , Feeding Behavior , Humans , Inflammation/blood , Inflammation/immunology , Logistic Models , Male , Middle Aged , Obesity/ethnology , Obesity/immunology , Odds Ratio , Risk Assessment , Risk Factors , Sex Factors , Smoking/ethnology , Smoking/immunology , Weight Gain/ethnology
6.
Circulation ; 104(23): 2820-5, 2001 Dec 04.
Article in English | MEDLINE | ID: mdl-11733401

ABSTRACT

BACKGROUND: We recently used a bone marrow-based gene therapy approach to show that small amounts of retrovirus-derived human apolipoprotein E3 (apoE3) produced by macrophages are protective against early atherosclerosis in apoE-deficient mice. METHODS AND RESULTS: In the present study, we evaluated whether the effect produced by macrophage-derived apoE3 is related to its ability to bind cellular membranes. To this end, we used apoE2 and apoEcys142, dysfunctional human variants with reduced binding to the LDL receptor or to heparan sulfate proteoglycans, respectively. ApoE-deficient mice, 5 weeks of age, received transplants of apoE(-/-) bone marrow cells transduced with either parental retrovirus or apoE3, apoE2, or apoEcys142 retroviral vectors. Human apoE was detected by ELISA in the serum of apoE3, apoE2, and apoEcys142 mice as early as 4 weeks after bone marrow transplantation, and at 8 weeks, plasma apoE levels were 55.5+/-20.3, 50.5+/-8.7, and 15.3+/-7.3 microgram/dL, respectively. In all groups, cholesterol levels increased with age but were not affected by apoE expression. As previously demonstrated, the lesion area in male apoE3 mice (3808+/-2224 micrometer(2)/section) was 40% smaller than that in control mice (6503+/-3475 micrometer(2)/section). In apoE2 mice, however, the lesion area was similar to that of controls (5991+/-2771 micrometer(2)/section), and apoEcys142 mice showed an unexpected and significant increase in lesion size (10 320+/-6128 micrometer(2)/section). Thus, transplantation with marrow transfected with receptor binding-defective apoE variants did not replicate the antiatherogenic effect of apoE3. CONCLUSIONS: These data provide in vivo evidence suggesting that macrophage-derived apoE delays development of atherosclerosis through a receptor-dependent pathway.


Subject(s)
Apolipoproteins E/physiology , Arteriosclerosis/pathology , Animals , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/blood , Apolipoproteins E/genetics , Arteriosclerosis/genetics , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Cloning, Molecular , Female , Gene Transfer Techniques , Hematopoietic Stem Cell Transplantation/methods , Humans , Male , Mice , Mice, Knockout , Protein Isoforms/blood , Protein Isoforms/genetics , Protein Isoforms/physiology , Retroviridae/genetics , Time Factors
7.
Arterioscler Thromb Vasc Biol ; 21(11): 1790-5, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11701467

ABSTRACT

The concentration of apolipoprotein (apo) AI in the artery wall is thought to enhance cellular cholesterol efflux and protect against atherosclerosis. It has been shown that although macrophages do not make apoAI, they respond to it by increased cholesterol efflux. We hypothesized that macrophage production of apoAI would increase cholesterol efflux and reduce atherogenesis. In this study, we produced mice expressing human apoAI under the control of the macrophage-specific scavenger receptor-A promoter (mphi-AI). Human apoAI was detectable in the serum HDL fraction of mphi-AI transgenic mice at concentrations too low to affect serum cholesterol or HDL levels. Immunoblotting showed the presence of human apoAI in transgenic macrophage culture supernatants, mostly as lipoprotein-free protein, with a small component associated with HDL-like particles. Atherosclerosis studies using apoAI((-/-)) mice transplanted with mphi-AI bone marrow showed that in the absence of macrophage-derived apoE, local expression of apoAI reduced diet-induced lesions in the proximal aorta. Additionally, mphi-AI macrophages showed a 40% increase in cholesterol efflux compared with control macrophages. These data support the hypothesis that apoAI production by macrophages in the artery wall is protective against atherosclerosis. This protection is likely mediated by increased cholesterol efflux and decreased foam cell formation in vivo.


Subject(s)
Apolipoprotein A-I/biosynthesis , Apolipoprotein A-I/genetics , Arteriosclerosis/metabolism , Cholesterol/metabolism , Macrophages/metabolism , Animals , Arteriosclerosis/etiology , Biological Transport , Cells, Cultured , Diet, Atherogenic , Foam Cells/physiology , Humans , Lipoproteins, HDL/chemistry , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
8.
Curr Opin Lipidol ; 12(5): 489-95, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11561167

ABSTRACT

The scope of this review is to discuss the new advances in our understanding of the role of scavenger receptor class A in the initiation and modulation of the atherosclerotic process. Through the approaches of gene manipulation in the mouse model, a substantial body of literature has accumulated that depicts scavenger receptor class A as a central player in atherogenesis. In studies of scavenger receptor class A overexpression in macrophages through bone marrow transplantation using transgenic donor material, recipient mice with hyperlipidemia caused either by apolipoprotein E or LDL receptor deficiency did not show convincing changes in the degree of atherosclerosis development compared with controls. Conversely, the deletion of the scavenger receptor class A gene in the mouse has shown, in a consistent and significant fashion, that this receptor serves a pro-atherogenic function under hyperlipidemic conditions, as both apolipoprotein E and LDL receptor-deficient mice had reduced atherosclerosis in the absence of scavenger receptor class A. In addition, we have recently shown that C57BL/6 mice are protected from diet-induced atherosclerosis when they lack scavenger receptor class A, and that the macrophage is the cell type responsible for the effect of scavenger receptor class A deficiency in reducing lesion formation in C57BL/6 and LDL receptor null mice. Together, these results demonstrate that macrophage scavenger receptor class A contributes significantly to atherosclerotic lesion formation, and suggest that the uptake of oxidized or modified lipoproteins by vessel wall macrophages is a central process in atherogenesis.


Subject(s)
Arteriosclerosis/physiopathology , CD36 Antigens/metabolism , Foam Cells/metabolism , Macrophages/metabolism , Receptors, Immunologic/metabolism , Animals , Disease Models, Animal , Gene Deletion , Humans , Hyperlipidemias/physiopathology , Mice , Mice, Inbred C57BL , Receptors, LDL/metabolism , Receptors, Scavenger , Scavenger Receptors, Class A
9.
J Biol Chem ; 276(39): 36742-8, 2001 Sep 28.
Article in English | MEDLINE | ID: mdl-11477092

ABSTRACT

We have previously reported that the lack of apolipoprotein (apo) E expression by macrophages promotes foam cell formation in vivo. Because transgenic mice overexpressing human apoA-I from the liver (h-apoA-I TgN) are protected from the atherogenesis induced by apoE deficiency, we hypothesized that the presence of apoA-I in the vessel wall could reduce the negative effect of apoE deficiency on lesion growth. To address this issue, we used both retroviral transduction and transgenic approaches to produce in vivo systems where apoA-I is expressed from macrophages. In the retroviral transduction study, apoA-I-deficient (apoA-I(-/-)) mice reconstituted with apoE-deficient (apoE(-/-)) bone marrow cells that were infected with a retroviral vector expressing human apoA-I (MFG-HAI) had 95% lower atherosclerotic lesion area than that of recipients of apoE(-/-) bone marrow cells infected with the parental virus (MFG). To determine whether the protective effect of locally produced apoA-I was due to the lack of systemic apoA-I, we conducted a different experiment using h-apoA-I TgN mice as recipients of apoE(-/-) bone marrow with or without human apoA-I (driven by a macrophage-specific transgene defined as mphi-AI). Aortic lesion area in apoE(-/-)/mphi-AI --> h-apoA-I TgN mice was decreased by 85% compared with apoE(-/-) --> h-apoA-I TgN mice. These data demonstrate that expression of apoA-I from macrophages protects against atherogenesis without affecting plasma apoA-I and high density lipoprotein cholesterol levels.


Subject(s)
Apolipoprotein A-I/biosynthesis , Arteriosclerosis/metabolism , Macrophages/metabolism , Retroviridae/genetics , Animals , Apolipoprotein A-I/chemistry , Apolipoproteins/metabolism , Blotting, Western , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Cells, Cultured , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Humans , Immunoblotting , Immunohistochemistry , Lipid Metabolism , Lipoproteins, HDL/metabolism , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Retroviridae/metabolism , Time Factors , Transduction, Genetic , Transgenes
10.
Am J Cardiol ; 88(2A): 12E-15E, 2001 Jul 19.
Article in English | MEDLINE | ID: mdl-11473738

ABSTRACT

Although the concept that inflammation plays a role in the biology of atherosclerosis is now well accepted, the basic feature of the arterial lesion remains the accumulation of clusters of foam cells. These clusters are the consequence of the enhanced recruitment of monocytes in the vessel wall induced by the hyperlipidemia and of the disproportionate accumulation of lipids in the cytoplasm of macrophages deriving from monocytes. Ultimately, every molecular force and pathway with modulating activity over the developing lesion will have to act on a convergence point with factors regulating cholesterol balance in the macrophage. Consistent with this view is the recent report that cytokines, such as tumor necrosis factor-alpha, can influence the expression of the scavenger receptor, whereas interferon-gamma can inhibit adenosine triphosphate-binding cassette transporter-1, the main effector of cholesterol efflux in the peripheral cell. Conversely, recent data have shown that primary alterations in macrophage cholesterol balance, such as those produced by the total absence of acylcoenzyme A:cholesterol acyltransferase-1, may determine local changes compatible with the activation of inflammatory pathways. In this brief review, we discuss some of the convergence points between inflammation and cholesterol balance, and we highlight the additional therapeutic targets suggested by these new developments in vascular biology.


Subject(s)
Arteriosclerosis , Calcinosis/complications , Cholesterol/metabolism , Cytokines/biosynthesis , Hyperlipidemias/complications , Inflammation/complications , Animals , Arteriosclerosis/etiology , Arteriosclerosis/metabolism , Calcinosis/metabolism , Humans , Inflammation/metabolism
11.
Kidney Int ; 59(5): 1899-910, 2001 May.
Article in English | MEDLINE | ID: mdl-11318962

ABSTRACT

BACKGROUND: Peroxisome proliferator-activated receptor-gamma (PPAR gamma) is a member of the nuclear receptor superfamily of ligand-dependent transcriptional factors with beneficial effects in diabetes mediated by improved insulin sensitivity and lipid metabolism, but potential adverse effects in atherosclerosis by promoting in vitro foam cell formation. We explored whether a PPAR gamma agonist, troglitazone (TGL), affects sclerosis by mechanisms unrelated to insulin and lipid effects in a model of nondiabetic glomerulosclerosis. METHODS: Adult male Sprague Dawley rats underwent 5/6 nephrectomy and were treated for 12 weeks as follows: control (CONT), no further treatment; triple antihypertensive therapy (TRX); and TGL or TGL + TRX. Functional, morphological, and molecular analyses were performed. RESULTS: Systolic blood pressure (SBP) was increased in CONT and TGL groups (161 +/- 1 and 160 +/- 3 mm Hg), but not in TGL + TRX and TRX (120 +/- 3 vs. 126 +/- 1 mm Hg, P < 0.0001 vs. non-TRX). Serum triglyceride and cholesterol levels in all groups remained normal except for slightly higher serum cholesterol levels in TRX group. TGL groups had reduced proteinuria, serum creatinine, and glomerulosclerosis versus CONT, in contrast to no significant effect with TRX alone (sclerosis index, 0 to 4+ scale: CONT 1.99 +/- 0.42, TGL 0.85 +/- 0.12, TGL + TRX 0.56 +/- 0.14, TRX 1.30 +/- 0.21; TGL, P < 0.05; TGL + TRX, P = 0.01 vs. CONT). Glomerular cell proliferation, assessed by proliferating cell nuclear antigen (PCNA), was decreased after treatment with TGL or TGL + TRX, in parallel with decreases in glomerular p21 mRNA and p27 protein compared with CONT and TRX (PCNA + cells/glomerulus: CONT 2.04 +/- 0.64, TGL 0.84 +/- 0.21, TGL + TRX 0.30 +/- 0.07, TRX 1.38 +/- 0.37; TGL, P < 0.05, TGL + TRX, P < 0.01 vs. CONT). Glomerular plasminogen activator inhibitor-1 (PAI-1) immunostaining was decreased in TGL or TGL + TRX groups (0 to 4+ scale, CONT 2.42 +/- 0.32, TGL 1.40 +/- 0.24, TGL + TRX 1.24 +/- 0.17, TRX 2.53 +/- 0.24; TGL or TGL + TRX vs. CONT, P < 0.05), with a parallel decrease in PAI-1 mRNA by in situ hybridization. Glomerular and tubular transforming growth factor-beta (TGF-beta) mRNA expression was decreased with TGL treatment. Glomerular macrophages, present in CONT and TRX rats, did not express PPAR gamma, in contrast to PPAR gamma + macrophages in control carotid artery plaque. PPAR gamma was expressed in resident cells. CONCLUSIONS: Our results demonstrate in vivo that the PPAR gamma ligand TGL ameliorates the progression of glomerulosclerosis in a nondiabetic model. Macrophages show phenotypic diversity in glomerular versus vascular sclerosis, with macrophage PPAR gamma expression in only the latter. PPAR gamma beneficial effects are independent of insulin/glucose effects and are associated with regulation of glomerular cell proliferation, hypertrophy, and decreased PAI-1 and TGF-beta expression.


Subject(s)
Chromans/pharmacology , Glomerulosclerosis, Focal Segmental/prevention & control , Hypoglycemic Agents/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Thiazoles/pharmacology , Thiazolidinediones , Transcription Factors/agonists , Animals , Antihypertensive Agents/pharmacology , Base Sequence , DNA Primers/genetics , Disease Models, Animal , Glomerulosclerosis, Focal Segmental/etiology , Glomerulosclerosis, Focal Segmental/pathology , Glomerulosclerosis, Focal Segmental/physiopathology , Insulin Resistance , Kidney/pathology , Lipids/blood , Macrophages/pathology , Male , Nephrectomy , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Transforming Growth Factor beta/genetics , Troglitazone
12.
J Biol Chem ; 276(25): 22965-70, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11304532

ABSTRACT

We have investigated the recycling of apoE in livers of apoE(-)/- mice transplanted with wild type bone marrow (apoE(+/+) --> apoE(-)/-), a model in which circulating apoE is derived exclusively from macrophages. Nascent Golgi lipoproteins were recovered from livers of apoE(+/+) --> apoE(-)/- mice 8 weeks after transplantation. ApoE was identified with nascent d < 1.006 and with d 1.006-1.210 g/ml lipoproteins at a level approximately 6% that of nascent lipoproteins from C57BL/6 mice. Hepatocytes from apoE(+/+) --> apoE(-)/- mice were isolated and cultured in media free of exogenous apoE. ApoE was found in the media primarily on the d < 1.006 g/ml fraction, indicating a resecretion of internalized apoprotein. Secretion of apoE from C57BL/6 hepatocytes was consistent with constitutive production, whereas the majority of apoE secreted from apoE(+/+) --> apoE(-)/- hepatocytes was recovered in the last 24 h of culture. This suggests that release may be triggered by accumulation of an acceptor, such as very low density lipoproteins, in the media. In agreement with the in vivo data, total recovery of apoE from apoE(+/+) --> apoE(-)/- hepatocytes was approximately 6% that of the apoE recovered from C57BL/6 hepatocytes. Since plasma apoE levels in the transplanted mice are approximately 10% of control levels, the findings indicate that up to 60% of the internalized apoE may be reutilized under physiologic conditions. These studies provide definitive evidence for the sparing of apoE and its routing through the secretory pathway and demonstrate that internalized apoE can be resecreted in a quantitatively significant fashion.


Subject(s)
Apolipoproteins E/metabolism , Endocytosis , Hepatocytes/metabolism , Animals , Apolipoproteins E/genetics , Cells, Cultured , Hepatocytes/cytology , Mice , Mice, Inbred C57BL
13.
Front Biosci ; 6: D515-25, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11229870

ABSTRACT

The mouse is the most utilized model to study lipids and atherosclerosis. Before the advent of the techniques of genetic manipulation it was well known that different inbred strains had varying degrees of susceptibility to diet-induced atherosclerosis. The C57BL/6 mouse was adopted as the standard even though the arterial lesions induced by even extreme diets were limited in size, complexity, and distribution. This changed with the production of several gene knockout and transgenic mice, which in many cases produced remarkable effects on plasma lipoproteins and arterial lesions even without dietary manipulations. The most typical example remains the apoE deficient model, in which a massive hyperlipidemia is accompanied with the development of severe atherosclerotic plaques at the aortic root and throughout the aortic tree. With the creation of the LDL receptor knockout, and the different knockout and transgenic mice with changes in apoB, apoE, and the HDL system, a solid body of new information has emerged on the mechanisms regulating plasma lipoprotein levels and controlling the initial stages of atherogenesis. This paper presents an overview of the most utilized mouse models and a summary of the results obtained with the technique of bone marrow transplantation, an approach to study macrophages in atherosclerosis.


Subject(s)
Arteriosclerosis/metabolism , Disease Models, Animal , Hyperlipidemias/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Arteriosclerosis/genetics , Hyperlipidemias/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, LDL/deficiency , Receptors, LDL/genetics
14.
J Clin Invest ; 107(2): 163-71, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11160132

ABSTRACT

During atherogenesis, circulating macrophages migrate into the subendothelial space, internalize cholesterol-rich lipoproteins, and become foam cells by progressively accumulating cholesterol esters. The inhibition of macrophage acyl coenzyme A:cholesterol acyltransferase (ACAT), which catalyzes the formation of cholesterol esters, has been proposed as a strategy to reduce foam cell formation and to treat atherosclerosis. We show here, however, that hypercholesterolemic LDL receptor-deficient (LDLR(-/-)) mice reconstituted with ACAT1-deficient macrophages unexpectedly develop larger atherosclerotic lesions than control LDLR(-/-) mice. The ACAT1-deficient lesions have reduced macrophage immunostaining and more free cholesterol than control lesions. Our findings suggest that selective inhibition of ACAT1 in lesion macrophages in the setting of hyperlipidemia can lead to the accumulation of free cholesterol in the artery wall, and that this promotes, rather than inhibits, lesion development.


Subject(s)
Arteriosclerosis/genetics , Macrophages/enzymology , Receptors, LDL/deficiency , Sterol O-Acyltransferase/deficiency , Animals , Aorta/metabolism , Aorta/pathology , Arteriosclerosis/metabolism , Arteriosclerosis/pathology , Bone Marrow Transplantation , Cell Transplantation , Cholesterol/metabolism , Coloring Agents , Female , Immunohistochemistry , Liver/cytology , Liver/embryology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sterol O-Acyltransferase/antagonists & inhibitors , Up-Regulation
15.
Curr Atheroscler Rep ; 2(1): 29-35, 2000 Jan.
Article in English | MEDLINE | ID: mdl-11122722

ABSTRACT

It is now widely accepted that low-density lipoprotein (LDL) is not the only atherogenic component of the lipid profile and that abnormalities in the metabolism and plasma levels of triglycerides and high-density lipoprotein (HDL) may lead to accelerated growth of atherosclerotic lesions. Fibrates are the drugs of first choice in the management of hypertriglyceridemia, and are also able to substantially raise HDL. The recently published Veterans Administration-High-density Lipoprotein Intervention Trial (VA-HIT) trial showed that fibrate treatment in patients with coronary heart disease (CHD), low HDL, modestly elevated triglycerides, and normal LDL reduces the risk of a recurrent coronary event by 25%. A reasonable approach to the dyslipidemic patient with high CHD risk is to tailor the intervention to the specific lipoprotein abnormality. Under these assumptions fibrate therapy should become widespread, considering that the most common lipid alteration in CHD and patients with diabetes is low HDL and high triglycerides.


Subject(s)
Cardiovascular Diseases/prevention & control , Clofibrate/pharmacology , Hyperlipidemias/therapy , Hypolipidemic Agents/pharmacology , Angiography , Cardiovascular Diseases/diagnostic imaging , Cholesterol, HDL/metabolism , Cholesterol, LDL/metabolism , Clinical Trials as Topic , Drug Interactions , Humans , Hyperlipidemias/complications , Hyperlipidemias/prevention & control , Preventive Medicine , Risk Factors , Ultrasonography
16.
Arterioscler Thromb Vasc Biol ; 20(12): 2593-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11116058

ABSTRACT

The absence of the scavenger receptor A (SR-A)-I/II has produced variable effects on atherosclerosis in different murine models. Therefore, we examined whether SR-AI/II deficiency affected atherogenesis in C57BL/6 mice, an inbred strain known to be susceptible to diet-induced atherosclerotic lesion formation, and whether the deletion of macrophage SR-AI/II expression would modulate lesion growth in C57BL/6 mice and LDL receptor (LDLR)(-/-) mice. SR-AI/II-deficient (SR-AI/II(-/-)) female and male mice on the C57BL/6 background were challenged with a butterfat diet for 30 weeks. No differences were detected in plasma lipids between SR-AI/II(-/-) and SR-AI/II(+/+) mice, whereas both female and male SR-AI/II(-/-) mice had a tremendous reduction (81% to 86%) in lesion area of the proximal aorta compared with SR-AI/II(+/+) mice. Next, to analyze the effect of macrophage-specific SR-AI/II deficiency in atherogenesis, female C57BL/6 mice were lethally irradiated, transplanted with SR-AI/II(-/-) or SR-AI/II(+/+) fetal liver cells, and challenged with the butterfat diet for 16 weeks. In a separate experiment, male LDLR(-/-) mice were reconstituted with SR-AI/II(-/-) or SR-AI/II(+/+) fetal liver cells and challenged with a Western diet for 10 weeks. No significant differences in plasma lipids and lipoprotein profiles were noted between the control and experimental groups in either experiment. SR-AI/II(-/-)-->C57BL/6 mice, however, had a 60% reduction in lesion area of the proximal aorta compared with SR-AI/II(+/+)-->C57BL/6 mice. A similar level of reduction (60%) in lesion area was noted in the proximal aorta and the entire aorta en face of SR-AI/II(-/-)-->LDLR(-/-) mice compared with SR-AI/II(+/+)-->LDLR(-/-) mice. These results demonstrate in vivo that SR-AI/II expression has no impact on plasma lipid levels and that macrophage SR-AI/II contributes significantly to atherosclerotic lesion formation.


Subject(s)
Arteriosclerosis/etiology , Macrophages/metabolism , Membrane Proteins , Receptors, Immunologic/biosynthesis , Receptors, Lipoprotein , Animals , Aorta/pathology , Aorta/ultrastructure , Arteriosclerosis/genetics , Arteriosclerosis/pathology , Body Weight , CD36 Antigens/genetics , Cell Transplantation , Cholesterol/blood , Diet, Atherogenic , Dietary Fats/administration & dosage , Disease Models, Animal , Female , Fetus/chemistry , Lipoproteins/blood , Liver/cytology , Male , Mice , Mice, Inbred C57BL , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Receptors, LDL/deficiency , Receptors, Scavenger , Scavenger Receptors, Class A , Scavenger Receptors, Class B , Time Factors , Triglycerides/blood
17.
J Biol Chem ; 275(34): 26293-9, 2000 Aug 25.
Article in English | MEDLINE | ID: mdl-10858435

ABSTRACT

The role of macrophage lipoprotein lipase (LPL) expression in atherosclerotic lesion formation was examined in low density lipoprotein receptor (LDLR(-/-)) mice using dietary conditions designed to induce either fatty streak lesions or complex atherosclerotic lesions. First, LDLR(-/-) mice chimeric for macrophage LPL expression were created by transplantation of lethally irradiated female LDLR(-/-) mice with LPL(-/-) (n = 12) or LPL(+/+) (n = 14) fetal liver cells as a source of hematopoietic cells. To induce fatty streak lesions, these mice were fed a Western diet for 8 weeks, resulting in severe hypercholesterolemia. There were no differences in plasma post-heparin LPL activity, serum lipid levels, or lipoprotein distribution between these two groups. The mean lesion area in the proximal aorta in LPL(-/-) --> LDLR(-/-) mice was significantly reduced by 33% compared with LPL(+/+) --> LDLR(-/-) mice, and a similar reduction (38%) in lesion area was found by en face analysis of the aortae. To induce complex atherosclerotic lesions, female LDLR(-/-) mice were lethally irradiated, transplanted with LPL(-/-) (n = 14), LPL(+/-) (n = 13), or LPL(+/+) (n = 14) fetal liver cells, and fed the Western diet for 19 weeks. Serum cholesterol and triglyceride levels did not differ between the three groups. After 19 weeks of diet, the lesions in the proximal aorta were complex with relatively few macrophages expressing LPL protein and mRNA in LPL(+/+) --> LDLR(-/-) mice. Analysis of cross-sections of the proximal aorta demonstrated no differences in the extent of lesion area between the groups, whereas en face analysis of the aortae revealed a dose-dependent effect of macrophage LPL on mean aortic lesion area in LPL(-/-) --> LDLR(-/-), LPL(-/+) --> LDLR(-/-), and LPL(+/+) --> LDLR(-/-) mice (1.8 +/- 0. 2%, 3.5 +/- 0.5% and 5.9 +/- 0.8%, respectively). Taken together, these data indicate that macrophage LPL expression in the artery wall promotes atherogenesis during foam cell lesion formation, but this impact may be limited to macrophage-rich lesions.


Subject(s)
Arteriosclerosis/pathology , Foam Cells/metabolism , Lipoprotein Lipase/metabolism , Macrophages/enzymology , Receptors, LDL/physiology , Animals , Aorta/pathology , Arteriosclerosis/metabolism , Chimera , Diet , Female , Mice , Mice, Inbred C57BL
18.
J Clin Invest ; 105(6): 711-9, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10727439

ABSTRACT

Inhibitors of acyl CoA:cholesterol acyltransferase (ACAT) have attracted considerable interest as a potential treatment for atherosclerosis. Currently available inhibitors probably act nonselectively against the two known ACATs. One of these enzymes, ACAT1, is highly expressed in macrophages in atherosclerotic lesions, where it contributes to foam-cell formation. In this study, we examined the effects of selective ACAT1 deficiency in two mouse models of atherosclerosis. In the setting of severe hypercholesterolemia caused by deficiency in apoE or the LDL receptor (LDLR), total ACAT1 deficiency led to marked alterations in cholesterol homeostasis and extensive deposition of unesterified cholesterol in the skin and brain. Bone marrow transplantation experiments demonstrated that ACAT1 deficiency in macrophages was sufficient to cause dermal xanthomas in hyperlipidemic LDLR-deficient mice. ACAT1 deficiency did not prevent the development of atherosclerotic lesions in either apoE-deficient or LDLR-deficient mice, despite causing relatively lower serum cholesterol levels. However, the lesions in ACAT1-deficient mice were atypical in composition, with reduced amounts of neutral lipids and a paucity of macrophages in advanced lesions. Although the latter findings may be associated with increased lesion stability, the marked alterations in cholesterol homeostasis indicate that selectively inhibiting ACAT1 in the setting of severe hyperlipidemia may have detrimental consequences.


Subject(s)
Arteriosclerosis/etiology , Cholesterol/metabolism , Foam Cells/pathology , Hypercholesterolemia/genetics , Isoenzymes/physiology , Macrophages/enzymology , Sterol O-Acyltransferase/physiology , Xanthomatosis/etiology , Animals , Aorta/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apolipoproteins E/physiology , Arteriosclerosis/enzymology , Arteriosclerosis/genetics , Arteriosclerosis/pathology , Bone Marrow Transplantation , Crosses, Genetic , Diet, Atherogenic , Foam Cells/enzymology , Hypercholesterolemia/complications , Hypercholesterolemia/enzymology , Hypercholesterolemia/pathology , Isoenzymes/deficiency , Isoenzymes/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, LDL/physiology , Skin/enzymology , Skin/pathology , Sterol O-Acyltransferase/deficiency , Sterol O-Acyltransferase/genetics , Xanthomatosis/enzymology , Xanthomatosis/genetics , Xanthomatosis/pathology
19.
Circulation ; 101(2): 207-13, 2000 Jan 18.
Article in English | MEDLINE | ID: mdl-10637210

ABSTRACT

Statins (HMG-CoA reductase inhibitors) are used widely for the treatment of hypercholesterolemia. They inhibit HMG-CoA reductase competitively, reduce LDL levels more than other cholesterol-lowering drugs, and lower triglyceride levels in hypertriglyceridemic patients. Statins are well tolerated and have an excellent safety record. Clinical trials in patients with and without coronary heart disease and with and without high cholesterol have demonstrated consistently that statins reduce the relative risk of major coronary events by approximately 30% and produce a greater absolute benefit in patients with higher baseline risk. Proposed mechanisms include favorable effects on plasma lipoproteins, endothelial function, plaque architecture and stability, thrombosis, and inflammation. Mechanisms independent of LDL lowering may play an important role in the clinical benefits conferred by these drugs and may ultimately broaden their indication from lipid-lowering to antiatherogenic agents.


Subject(s)
Cardiology/trends , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Hypercholesterolemia/drug therapy , Clinical Trials as Topic , Drug Therapy, Combination , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hypercholesterolemia/prevention & control , Hypolipidemic Agents/therapeutic use , Practice Guidelines as Topic
20.
Trends Cardiovasc Med ; 10(1): 23-30, 2000 Jan.
Article in English | MEDLINE | ID: mdl-11150725

ABSTRACT

Apolipoprotein E (apoE) is unique among the different apoproteins because of its many functions in the assembly, processing, and removal of plasma lipoproteins. Some of these functions require the presence of apoE inside the cell or in close association with its outer membrane. For example, the presence of apoE increases triglyceride content in newly formed very low-density lipoprotein (VLDL), whereas the fast removal of chylomicrons requires the presence of a cellular pool of apoE ready to surface and bind to membrane proteoglycans and possibly to lipoprotein receptors such as the LDL receptor-related protein (LRP). Our recent discovery that VLDL-apoE internalized by hepatocytes is partially protected from lysosomal degradation and recycles through the Golgi apparatus suggests that the biologic cycle of apoE may not be concluded when the lipoprotein is taken up by the cell, and that recycling apoE may have a physiologic role in lipoprotein assembly, remnant removal, and cholesterol efflux.


Subject(s)
Apolipoproteins E/physiology , Cell Physiological Phenomena , Animals , Apolipoproteins E/metabolism , Cholesterol/metabolism , Chylomicrons/metabolism , Humans , Lipoproteins/metabolism , Liver/cytology , Liver/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...