Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Arterioscler Thromb Vasc Biol ; 32(12): 2839-46, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23023374

ABSTRACT

OBJECTIVE: To determine the impact of hematopoietic deletion of nuclear factor- (erythroid-derived 2) like 2 factor (Nrf2) on the development of atherosclerosis and liver injury in an obese, hypercholesterolemic mouse model. METHODS AND RESULTS: Two-month-old male low-density lipoprotein receptor-deficient mice were lethally irradiated and transplanted with either wild type or Nrf2-deficient (Nrf2(-/-)) bone marrow cells. At 3 months of age, mice were placed on an obesogenic high-fat diet (HFD), high-cholesterol diet for 7 months. Despite no differences in body weight, body fat percentage, liver fat, plasma glucose, lipids, or insulin, the HFD-fed Nrf2(-/-) bone marrow recipients had increased proinflammatory vascular gene expression, a significant increase in atherosclerosis area (18% versus 28%; P=0.018) and lesion complexity, and a marked increase in liver fibrosis. The acceleration of vascular and liver injury may arise from enhanced macrophage migration, inflammation, and oxidative stress resulting from myeloid Nrf2 deficiency. CONCLUSIONS: Myeloid-derived Nrf2 activity attenuates atherosclerosis development and liver inflammation and fibrosis associated with obesity. Prevention of oxidative stress in macrophage and other myeloid lineage cells may be an important therapeutic target to reduce inflammation-driven complications of obesity.


Subject(s)
Atherosclerosis/epidemiology , Gene Deletion , Hypercholesterolemia/complications , Liver Cirrhosis/epidemiology , Myeloid Cells/metabolism , NF-E2-Related Factor 2/deficiency , Obesity/complications , Animals , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Bone Marrow Transplantation , Cell Movement/physiology , Comorbidity , Disease Models, Animal , Hypercholesterolemia/epidemiology , Liver Cirrhosis/metabolism , Liver Cirrhosis/physiopathology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Obesity/epidemiology , Oxidative Stress/physiology , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, LDL/metabolism , Risk Factors
2.
J Environ Manage ; 99: 27-35, 2012 May 30.
Article in English | MEDLINE | ID: mdl-22306087

ABSTRACT

In this paper we describe elements of a field research project that presented farmers with economic incentives to control nitrate runoff. The approach used is novel in that payments are based on ambient water quality and water quantity produced by a watershed rather than proxies for water quality conservation. Also, payments are made based on water quality relative to a control watershed, and therefore, account for stochastic fluctuations in background nitrate levels. Finally, the program pays farmers as a group to elicit team behavior. We present our approach to modeling that allowed us to estimate prices for water and resulting payment levels. We then compare these preliminary estimates to the actual values recorded over 33 months of fieldwork. We find that our actual payments were 29% less than our preliminary estimates, due in part to the failure of our ecological model to estimate discharge accurately. Despite this shortfall, the program attracted the participation of 53% of the farmers in the watershed, and resulted in substantial nitrate abatement activity. Given this favorable response, we propose that research efforts focus on implementing field trials of group-level performance-based payments. Ideally these programs would be low risk and control for naturally occurring contamination.


Subject(s)
Models, Economic , Motivation , Water Pollution/prevention & control , Water Quality , Agriculture
3.
Hepatology ; 52(6): 2001-11, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20938947

ABSTRACT

UNLABELLED: Nonalcoholic fatty liver disease (NAFLD) is a common complication of obesity that can progress to nonalcoholic steatohepatitis (NASH), a serious liver pathology that can advance to cirrhosis. The mechanisms responsible for NAFLD progression to NASH remain unclear. Lack of a suitable animal model that faithfully recapitulates the pathophysiology of human NASH is a major obstacle in delineating mechanisms responsible for progression of NAFLD to NASH and, thus, development of better treatment strategies. We identified and characterized a novel mouse model, middle-aged male low-density lipoprotein receptor (LDLR)(-/-) mice fed a high-fat diet (HFD), which developed NASH associated with four of five metabolic syndrome (MS) components. In these mice, as observed in humans, liver steatosis and oxidative stress promoted NASH development. Aging exacerbated the HFD-induced NASH such that liver steatosis, inflammation, fibrosis, oxidative stress, and liver injury markers were greatly enhanced in middle-aged versus young LDLR(-/-) mice. Although expression of genes mediating fatty acid oxidation and antioxidant responses were up-regulated in young LDLR(-/-) mice fed HFD, they were drastically reduced in MS mice. However, similar to recent human trials, NASH was partially attenuated by an insulin-sensitizing peroxisome proliferator-activated receptor-gamma (PPARγ) ligand, rosiglitazone. In addition to expected improvements in MS, newly identified mechanisms of PPARγ ligand effects included stimulation of antioxidant gene expression and mitochondrial ß-oxidation, and suppression of inflammation and fibrosis. LDLR-deficiency promoted NASH, because middle-aged C57BL/6 mice fed HFD did not develop severe inflammation and fibrosis, despite increased steatosis. CONCLUSION: MS mice represent an ideal model to investigate NASH in the context of MS, as commonly occurs in human disease, and NASH development can be substantially attenuated by PPARγ activation, which enhances ß-oxidation.


Subject(s)
Fatty Liver/prevention & control , Receptors, LDL/deficiency , Thiazolidinediones/therapeutic use , Aging/physiology , Animals , Antioxidants/metabolism , Dietary Fats/adverse effects , Fatty Liver/genetics , Gene Expression , Hepatitis/etiology , Liver/drug effects , Liver/metabolism , Male , Metabolic Syndrome , Mice , Mice, Knockout , Mitochondria, Liver/physiology , Oxidative Stress , PPAR gamma/metabolism , Rosiglitazone
4.
Circ Res ; 104(6): e42-54, 2009 Mar 27.
Article in English | MEDLINE | ID: mdl-19265038

ABSTRACT

Excess food intake leads to obesity and diabetes, both of which are well-known independent risk factors for atherosclerosis, and both of which are growing epidemics in an aging population. We hypothesized that aging enhances the metabolic and vascular effects of high fat diet (HFD) and therefore examined the effect of age on atherosclerosis and insulin resistance in lipoprotein receptor knockout (LDLR(-/-)) mice. We found that 12-month-old (middle-aged) LDLR(-/-) mice developed substantially worse metabolic syndrome, diabetes, and atherosclerosis than 3-month-old (young) LDLR(-/-) mice when both were fed HFD for 3 months, despite similar elevations in total cholesterol levels. Microarray analyses were performed to analyze the mechanism responsible for the marked acceleration of atherosclerosis in middle-aged mice. Chow-fed middle-aged mice had greater aortic expression of multiple antioxidant genes than chow-fed young mice, including glutathione peroxidase-1 and -4, catalase, superoxide dismutase-2, and uncoupling protein-2. Aortic expression of these enzymes markedly increased in young mice fed HFD but decreased or only modestly increased in middle-aged mice fed HFD, despite the fact that systemic oxidative stress and vascular reactive oxygen species generation, measured by plasma F2alpha isoprostane concentration (systemic) and dihydroethidium conversion and p47phox expression (vascular), were greater in middle-aged mice fed HFD. Thus, the mechanism for the accelerated vascular injury in older LDLR(-/-) mice was likely the profound inability to mount an antioxidant response. This effect was related to a decrease in vascular expression of 2 key transcriptional pathways regulating the antioxidant response, DJ-1 and forkhead box, subgroup O family (FOXOs). Treatment of middle-aged mice fed HFD with the antioxidant apocynin attenuated atherosclerosis, whereas treatment with the insulin sensitizer rosiglitazone attenuated both metabolic syndrome and atherosclerosis. Both treatments decreased oxidative stress. A novel effect of rosiglitazone was to increase expression of Nrf2 (nuclear factor [erythroid-derived 2]-like 2), a downstream target of DJ-1 contributing to enhanced expression of vascular antioxidant enzymes. This investigation underscores the role of oxidative stress when multiple atherosclerotic risk factors, particularly aging, converge on the vessel wall and emphasizes the need to develop effective strategies to inhibit oxidative stress to protect aging vasculature.


Subject(s)
Aging/metabolism , Antioxidants/metabolism , Atherosclerosis/metabolism , Diet, Atherogenic , Gene Expression Regulation , Acetophenones/pharmacology , Aging/genetics , Aging/pathology , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Eating , Enzyme Inhibitors/pharmacology , Gene Expression Profiling , Gene Knockout Techniques , Humans , Insulin Resistance/genetics , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Mice , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Oligonucleotide Array Sequence Analysis , Oxidative Stress/drug effects , Oxidative Stress/genetics , Receptors, LDL/genetics
5.
Proc Natl Acad Sci U S A ; 105(11): 4277-82, 2008 Mar 18.
Article in English | MEDLINE | ID: mdl-18337495

ABSTRACT

Activation of the nuclear hormone receptor peroxisome proliferator-activated receptor delta (PPARdelta) has been shown to improve insulin resistance, adiposity, and plasma HDL levels. However, its antiatherogenic role remains controversial. Here we report atheroprotective effects of PPARdelta activation in a model of angiotensin II (AngII)-accelerated atherosclerosis, characterized by increased vascular inflammation related to repression of an antiinflammatory corepressor, B cell lymphoma-6 (Bcl-6), and the regulators of G protein-coupled signaling (RGS) proteins RGS4 and RGS5. In this model, administration of the PPARdelta agonist GW0742 (1 or 10 mg/kg) substantially attenuated AngII-accelerated atherosclerosis without altering blood pressure and increased vascular expression of Bcl-6, RGS4, and RGS5, which was associated with suppression of inflammatory and atherogenic gene expression in the artery. In vitro studies demonstrated similar changes in AngII-treated macrophages: PPARdelta activation increased both total and free Bcl-6 levels and inhibited AngII activation of MAP kinases, p38, and ERK1/2. These studies uncover crucial proinflammatory mechanisms of AngII and highlight actions of PPARdelta activation to inhibit AngII signaling, which is atheroprotective.


Subject(s)
Angiotensin II/pharmacology , Atherosclerosis/metabolism , PPAR delta/metabolism , Adipokines/blood , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Cell Movement/drug effects , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Hypercholesterolemia/blood , Hypercholesterolemia/pathology , Hypertriglyceridemia/metabolism , Hypertriglyceridemia/pathology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Ligands , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , PPAR delta/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-6/genetics , Proto-Oncogene Proteins c-bcl-6/metabolism , Receptors, LDL/deficiency , Receptors, LDL/genetics , Receptors, LDL/metabolism , Signal Transduction/drug effects , Thiazoles/blood , Thiazoles/pharmacology , Transcription, Genetic/genetics
6.
Kidney Int ; 77(7): 588-600, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20130530

ABSTRACT

The profibrotic adhesion molecule, osteopontin (OPN), is upregulated in kidneys of humans and mice with diabetes. The thiazolidinedione (TZD) insulin sensitizers decrease albuminuria in diabetic nephropathy (DN) and reduce OPN expression in vascular and cardiac tissue. To examine whether OPN is a critical mediator of DN we treated db/db mice with insulin, rosiglitazone, or pioglitazone to achieve similar fasting plasma glucose levels. The urine albumin-to-creatinine ratio and glomerular OPN expression were increased in diabetic mice, but both were reduced by the TZDs more than by insulin. We administered streptozotocin to OPN-null and OPN-wild-type mice, and OPN-null mice were bred into both type 1 (Ins2(akita/+)) and 2 (db/db) diabetic mice. In each case, OPN deletion decreased albuminuria, mesangial area, and glomerular collagen IV, fibronectin and transforming growth factor (TGF)-beta in the diabetic mice compared with their respective controls. In cultured mouse mesangial cells, TZDs but not insulin decreased angiotensin II-induced OPN expression, while recombinant OPN upregulated TGF-beta, ERK/MAPK, and JNK/MAPK signaling. These studies show that OPN expression in DN mouse models enhances glomerular damage, likely through the expression of TGF-beta, while its deletion protects against disease progression, suggesting that OPN might serve as a therapeutic target.


Subject(s)
Albuminuria/etiology , Diabetic Nephropathies/metabolism , Kidney Glomerulus/metabolism , Osteopontin/metabolism , Thiazolidinediones/therapeutic use , Albuminuria/drug therapy , Angiotensin II/metabolism , Animals , Cells, Cultured , Collagen Type IV/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/complications , Diabetic Nephropathies/drug therapy , Fibronectins/metabolism , Hypoglycemic Agents/therapeutic use , Insulin/therapeutic use , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR gamma/metabolism , Transforming Growth Factor beta/metabolism
7.
Diabetes ; 55(8): 2265-71, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16873689

ABSTRACT

Previous characterization of mouse chromosome 2 identified genomic intervals that influence obesity, insulin resistance, and dyslipidemia. For this, resistant CAST/Ei (CAST) alleles were introgressed onto a susceptible C57BL/6J background to generate congenic strains with CAST alleles encompassing 67-162 Mb (multigenic obesity 6 [MOB6]) and 84-180 Mb (MOB5) from mouse chromosome 2. To examine the effects of each congenic locus on atherosclerosis and glucose disposal, we bred each strain onto a sensitizing LDL receptor-null (LDLR(-/-)) C57BL/6J background to predispose them to hypercholesterolemia and insulin resistance. LDLR(-/-) congenics and controls were characterized for measures of atherogenesis, insulin sensitivity, and obesity. We identified a genomic interval unique to the MOB6 congenic (72-84 Mb) that dramatically decreased atherosclerosis by approximately threefold and decreased insulin resistance. This region also reduced adiposity twofold. Conversely, the congenic region unique to MOB5 (162-180 Mb) increased insulin resistance but had little effect on atherosclerosis and adiposity. The MOB congenic intervals are concordant to human and rat quantitative trait loci influencing diabetes and atherosclerosis traits. Thus, our results define a strategy for studying the poorly understood interactions between diabetes and atherosclerosis and for identifying genes underlying the cardiovascular complications of insulin resistance.


Subject(s)
Cardiovascular Diseases/genetics , Insulin Resistance/genetics , Obesity/genetics , Receptors, LDL/deficiency , Alleles , Animals , Atherosclerosis/genetics , Blood Glucose/analysis , Breeding , Cholesterol/blood , Chromosome Mapping , Chromosomes , Chromosomes, Mammalian , Genetic Predisposition to Disease , Hypercholesterolemia/genetics , Hyperglycemia/genetics , Hyperlipidemias/genetics , Male , Mice , Mice, Inbred C57BL , Receptors, LDL/physiology
8.
J Clin Invest ; 112(9): 1318-31, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14597759

ABSTRACT

Osteopontin (OPN) is expressed in atherosclerotic lesions, particularly in diabetic patients. To determine the role of OPN in atherogenesis, ApoE-/-OPN+/+, ApoE-/-OPN+/-, and ApoE-/-OPN-/- mice were infused with Ang II, inducing vascular OPN expression and accelerating atherosclerosis. Compared with ApoE-/-OPN+/+ mice, ApoE-/-OPN+/- and ApoE-/-OPN-/- mice developed less Ang II-accelerated atherosclerosis. ApoE-/- mice transplanted with bone marrow derived from ApoE-/-OPN-/- mice had less Ang II-induced atherosclerosis compared with animals receiving ApoE-/-OPN+/+ cells. Aortae from Ang II-infused ApoE-/-OPN-/- mice expressed less CD68, C-C-chemokine receptor 2, and VCAM-1. In response to intraperitoneal thioglycollate, recruitment of leukocytes in OPN-/- mice was impaired, and OPN-/- leukocytes exhibited decreased basal and MCP-1-directed migration. Furthermore, macrophage viability in atherosclerotic lesions from Ang II-infused ApoE-/-OPN-/- mice was decreased. Finally, Ang II-induced abdominal aortic aneurysm formation in ApoE-/-OPN-/- mice was reduced and associated with decreased MMP-2 and MMP-9 activity. These data suggest an important role for leukocyte-derived OPN in mediating Ang II-accelerated atherosclerosis and aneurysm formation.


Subject(s)
Angiotensin II/pharmacology , Aortic Aneurysm, Abdominal/etiology , Arteriosclerosis/etiology , Sialoglycoproteins/physiology , Animals , Aortic Aneurysm, Abdominal/therapy , Apolipoproteins E/physiology , Arteriosclerosis/therapy , Cell Movement , Cell Survival , Chemokine CCL2/physiology , Cytokines/biosynthesis , Gene Expression Regulation/drug effects , Leukocytes/physiology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Osteopontin , RNA, Messenger/analysis , Receptors, CCR2 , Receptors, Chemokine/physiology , Sialoglycoproteins/genetics
9.
Circ Res ; 97(9): e88-96, 2005 Oct 28.
Article in English | MEDLINE | ID: mdl-16224068

ABSTRACT

Angiotensin II (Ang II) is a powerful accelerator of atherosclerosis. Herein, we describe a novel transcription mechanism through which Ang II inhibits macrophage expression of the ATP-binding cassette transporter A1 (ABCA1), a key regulator of reverse cholesterol transport. We demonstrate that chronic Ang II infusion substantially promotes macrophage infiltration, foam cell formation, and atherosclerosis in low-density lipoprotein receptor-deficient mice and significantly reduces ABCA1 expression in peripheral macrophages. Administration of the Ang II type 1 receptor blocker valsartan inhibited Ang II-induced ABCA1 mRNA repression, macrophage cholesterol accumulation, and atherosclerosis. Ang II treatment reduced ABCA1 promoter activity of in vitro cultured mouse peritoneal macrophages, inducing fos-related antigen 2 (Fra2) protein binding to an ABCA1 promoter E-box motif, a site known to negatively regulate macrophage ABCA1 transcription. Valsartan pretreatment blocked Fra2 binding to the ABCA1 promoter, and Fra2 small interfering RNA pretreatment attenuated Ang II-mediated ABCA1 transcriptional inhibition, confirming the role of Fra2 in this process. This new evidence suggests that Ang II, a well-known proinflammatory and pro-oxidative factor, alters macrophage cholesterol homeostasis by repressing ABCA1 to promote foam cell formation and atherosclerosis.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Angiotensin II/pharmacology , Atherosclerosis/chemically induced , Macrophages/metabolism , Repressor Proteins/pharmacology , ATP Binding Cassette Transporter 1 , Animals , Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/genetics , Cholesterol/metabolism , Fos-Related Antigen-2/genetics , Fos-Related Antigen-2/physiology , Mice , Promoter Regions, Genetic , RNA, Messenger/analysis , RNA, Small Interfering/pharmacology , Receptors, LDL/physiology , Transcription, Genetic/drug effects
10.
Circ Res ; 95(12): e110-23, 2004 Dec 10.
Article in English | MEDLINE | ID: mdl-15539633

ABSTRACT

The liver X receptors alpha and beta (LXRalpha and LXRbeta) are important regulators of cholesterol homeostasis in liver and macrophages. Synthetic LXR ligands prevent the development of atherosclerosis in murine models; however, the potential functional relevance of LXRs in vascular smooth muscle cells (VSMCs) has not been investigated. In the present study, we demonstrate that LXRs are expressed and functional in primary human coronary artery VSMCs (CASMCs). LXR ligands inhibited mitogen-induced VSMC proliferation and G1-->S phase progression of the cell cycle. Inhibition of G1 exit by LXR ligands was accompanied by a dose-dependent inhibition of retinoblastoma protein (Rb) phosphorylation, which functions as the key switch for G1-->S cell cycle progression. LXR ligands suppressed mitogen-induced degradation of the cyclin-dependent kinase inhibitor p27Kip1, attenuated cyclin D1 and cyclin A expression, and inhibited the expression of S phase-regulatory minichromosome maintenance protein 6. Stabilization of p27kip1 by LXR ligands was mediated by supressing the transcriptional activation of the S phase kinase-associated protein 2 (Skp2), an F-box protein that targets p27Kip1 for degradation. Inhibition of Rb phosphorylation and G1-->S cell cycle progression by LXR ligands was reversed in VSMCs overexpressing Skp2, indicating that Skp2 as an upstream regulator of p27Kip1 degradation plays a central role in LXR ligand-mediated inhibition of VSMC proliferation. Furthermore, adenovirus-mediated overexpression of the S phase transcription factor E2F, which is released after Rb phosphorylation, reversed the inhibitory effect of LXR ligands on VSMC proliferation and S phase gene expression, suggesting that the primary mechanisms by which LXR ligands inhibit VSMC proliferation occur upstream of Rb phosphorylation. Finally, neointima formation in a model of rat carotid artery balloon injury was significantly attenuated after treatment with the LXR ligand T1317 compared with vehicle-treated animals. These data demonstrate that LXR ligands inhibit VSMC proliferation and neointima formation after balloon injury and suggest that LXR ligands may constitute a novel therapy for proliferative vascular diseases. The full text of this article is available online at http://circres.ahajournals.org.


Subject(s)
Carotid Artery Injuries/pathology , DNA-Binding Proteins/physiology , Muscle, Smooth, Vascular/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Tunica Intima/pathology , Tunica Media/pathology , Animals , Anticholesteremic Agents/pharmacology , Benzoates/pharmacology , Benzylamines/pharmacology , Cell Cycle/drug effects , Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Division/drug effects , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Coronary Vessels/cytology , Cyclin-Dependent Kinase Inhibitor p27 , DNA-Binding Proteins/agonists , G1 Phase/drug effects , Gene Expression Regulation/drug effects , Humans , Hydrocarbons, Fluorinated , Hyperplasia , Insulin/pharmacology , Ligands , Liver X Receptors , Minichromosome Maintenance Complex Component 6 , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Orphan Nuclear Receptors , Phosphorylation/drug effects , Platelet-Derived Growth Factor/pharmacology , Protein Processing, Post-Translational/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/agonists , Recombinant Fusion Proteins/physiology , Retinoblastoma Protein/metabolism , S-Phase Kinase-Associated Proteins/biosynthesis , S-Phase Kinase-Associated Proteins/genetics , S-Phase Kinase-Associated Proteins/physiology , Sulfonamides , Transfection , Tumor Suppressor Proteins/metabolism , Tunica Intima/drug effects , Tunica Media/drug effects
11.
J Am Coll Cardiol ; 43(9): 1698-705, 2004 May 05.
Article in English | MEDLINE | ID: mdl-15120833

ABSTRACT

OBJECTIVES: Osteopontin (OPN) is upregulated in left ventricular hypertrophy and is stimulated by angiotensin II (AngII). Our objective was to determine whether mice deficient in OPN would be protected from AngII-induced cardiac fibrosis. BACKGROUND: Interstitial fibrosis can lead to myocardial dysfunction and ultimately heart failure. Osteopontin activates integrins that regulate cell adhesion, migration, and growth, thus implicating OPN in the process of cardiac fibrosis. METHODS: Osteopontin null (OPN(-/-)) mice (n = 18) and wild-type controls (n = 20) were infused with AngII (2.5 or 3.0 microg/kg/min) for four days or three weeks via osmotic mini-pumps. Hearts were assessed morphometrically and histologically, including quantitative assessment of fibrosis via optical microscopic imaging analysis. Cardiac fibroblasts derived from these mice were evaluated for adhesion and proliferation. Cardiac transcript expression for cytokines, extracellular matrix (ECM), integrin, and atrial natriuretic peptide were assessed. RESULTS: Osteopontin(-/-) mice exhibited less cardiac fibrosis (0.7%) than wild-type mice (8.0%) (p < 0.01) and lowered heart/body weight ratios (0.10% vs. 0.23%) (p < 0.01) after three weeks of AngII infusion. Expression of transforming growth factor-beta, fibronectin, and collagen was not different between OPN(-/-) and wild-type mice, despite the decrease in ECM accumulation in the OPN(-/-) mice. Adhesion to ECM substrates decreased by 30% to 50% in cardiac fibroblasts of OPN(-/-) mice but was restored in OPN(-/-) cells by the addition of recombinant osteopontin. CONCLUSIONS: Osteopontin mediates cardiac fibrosis, probably through the modulation of cellular adhesion and proliferation. Because OPN is increased in cardiac hypertrophy and its lack attenuates fibrosis, understanding of OPN function is essential to extend our knowledge about molecular determinants of cardiac hypertrophy and failure.


Subject(s)
Angiotensin II/adverse effects , Myocardium/metabolism , Myocardium/pathology , Sialoglycoproteins/deficiency , Vasoconstrictor Agents/adverse effects , Angiotensin II/administration & dosage , Animals , Atrial Natriuretic Factor/drug effects , Atrial Natriuretic Factor/metabolism , Blood Pressure/drug effects , Blotting, Northern , Cardiomegaly/etiology , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cell Adhesion/drug effects , Cell Division/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Extracellular Matrix/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Mice , Mice, Knockout , Models, Cardiovascular , Muscle Proteins , Nuclear Proteins/drug effects , Nuclear Proteins/metabolism , Osteopontin , Repressor Proteins/drug effects , Repressor Proteins/metabolism , Up-Regulation/drug effects , Vasoconstrictor Agents/administration & dosage
12.
Drug News Perspect ; 16(4): 197-204, 2003 May.
Article in English | MEDLINE | ID: mdl-12942148

ABSTRACT

Peroxisome proliferator-activated receptor-gamma (PPARgamma) ligands have been used for several years as modulators of insulin sensitivity and glucose metabolism. Recent data from numerous studies have shown that PPARgamma ligands have numerous beneficial effects in the vasculature. They have been shown to regulate proliferation and migration of vascular smooth muscle cells as well as improving endothelial cell function. They improve blood pressure by actions at the resistance arteries and kidneys. Clinical trials have indicated that PPARgamma ligands can minimize the development of atherosclerosis as well as regulating other vascular inflammation. PPARgamma has been detected in all the cells found in the vessel wall as well as those cells associated with vascular pathophysiologies. In the monocyte/macrophage, PPARgamma ligands downregulate production of inflammatory cytokines and migration. Also, PPARgamma ligands regulate the expression of SR-A and CD36 receptors that take up lipids in the macrophage. PPARgamma has also been demonstrated to act through the liver X receptor alpha to increase the activity of reverse cholesterol transport in these cells. In dendritic cells and T-cells, PPARgamma ligands have been shown to inhibit activation and the initiation of inflammation. Inflammatory cytokines are downregulated in animal models administered PPARgamma ligands, leading to decreased atherosclerosis. While PPARgamma ligands have been useful in the treatment of type 2 diabetes, the important vasculoprotection elicited by these compounds could prove to be of greater benefit in the future.


Subject(s)
Endothelium, Vascular/pathology , Ligands , Muscle, Smooth, Vascular/pathology , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Vascular Diseases/prevention & control , Animals , Arteriosclerosis/drug therapy , Arteriosclerosis/etiology , Arteriosclerosis/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Humans , Inflammation/metabolism , Inflammation/prevention & control , Insulin Resistance , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Vascular Diseases/etiology , Vascular Diseases/metabolism
13.
Plant Dis ; 84(10): 1140-1146, 2000 Oct.
Article in English | MEDLINE | ID: mdl-30831908

ABSTRACT

Conventional and alternative integrated pest management (IPM) programs for managing arthropods and diseases affecting processing apple production were compared over 4 years. The effects of the two programs on populations of pest and beneficial insects, and on disease incidence and severity, were evaluated in the field and laboratory and at a commercial fruit processing plant by federal inspectors. The economic implications of the alternative management program were assessed by examining its relative costs and benefits compared with conventional management. In the alternative program, arthropods were managed with oil and Bacillus thuringiensis (Bt), and early-season diseases were managed with fungicides. Calcium chloride was used to suppress rot diseases. The alternative plots showed lower levels of indirect pests (aphids, leaf-hoppers, and mites) and increased numbers of arthropod natural enemies. On harvested fruit, there was significantly more injury in the alternative plots from codling moth or oriental fruit moth, plum curculio, and apple maggot. Levels of foliar diseases were similar between treatments; however, fruit from the alternative program exhibited significantly more sooty blotch, flyspeck, and rots. At the commercial processor, levels of cull fruit were higher for the alternative program in 2 of the 3 years with harvestable yield. Loads of fruit that were not acceptable for processing were due mostly to internal worms and worm damage. Incidence of decay was within the quality limits set by the processor, except for one conventional sample, in which decay exceeded 5%. Of the 36 loads of fruit that were examined from each program, 4 and 13 loads were rejected from the conventional and alternative programs, respectively. Net present value comparisons were dependent upon how rejected loads were valued. At cider prices for rejected loads, net returns were similar between programs. When rejected loads were valued at a complete loss, conventional net present value was over 140% higher than the alternative program. Among cultivars, Golden Delicious was most profitable and York was least profitable.

14.
Diabetes ; 57(9): 2470-9, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18511847

ABSTRACT

OBJECTIVE: Cardiac fibrosis is an important component of diabetic cardiomyopathy. Peroxisome proliferator-activated receptor gamma (PPARgamma) ligands repress proinflammatory gene expression, including that of osteopontin, a known contributor to the development of myocardial fibrosis. We thus investigated the hypothesis that PPARgamma ligands could attenuate cardiac fibrosis. RESEARCH DESIGN AND METHODS: Wild-type cardiomyocyte- and macrophage-specific PPARgamma(-/-) mice were infused with angiotensin II (AngII) to promote cardiac fibrosis and treated with the PPARgamma ligand pioglitazone to determine the roles of cardiomyocyte and macrophage PPARgamma in cardiac fibrosis. RESULTS: Cardiomyocyte-specific PPARgamma(-/-) mice (cPPARgamma(-/-)) developed spontaneous cardiac hypertrophy with increased ventricular osteopontin expression and macrophage content, which were exacerbated by AngII infusion. Pioglitazone attenuated AngII-induced fibrosis, macrophage accumulation, and osteopontin expression in both wild-type and cPPARgamma(-/-) mice but induced hypertrophy in a PPARgamma-dependent manner. We pursued two mechanisms to explain the antifibrotic cardiomyocyte-PPARgamma-independent effects of pioglitazone: increased adiponectin expression and attenuation of proinflammatory macrophage activity. Adenovirus-expressed adiponectin had no effect on cardiac fibrosis and the PPARgamma ligand pioglitazone did not attenuate AngII-induced cardiac fibrosis, osteopontin expression, or macrophage accumulation in monocyte-specific PPARgamma(-/-) mice. CONCLUSIONS: We arrived at the following conclusions: 1) both cardiomyocyte-specific PPARgamma deficiency and activation promote cardiac hypertrophy, 2) both cardiomyocyte and monocyte PPARgamma regulate cardiac macrophage infiltration, 3) inflammation is a key mediator of AngII-induced cardiac fibrosis, 4) macrophage PPARgamma activation prevents myocardial macrophage accumulation, and 5) PPARgamma ligands attenuate AngII-induced cardiac fibrosis by inhibiting myocardial macrophage infiltration. These observations have important implications for potential interventions to prevent cardiac fibrosis.


Subject(s)
Cardiomegaly/pathology , Macrophages/metabolism , Myocardium/pathology , Myocytes, Cardiac/pathology , PPAR gamma/genetics , PPAR gamma/metabolism , Adiponectin/blood , Adiponectin/pharmacology , Angiotensin II/pharmacology , Animals , Cardiomegaly/drug therapy , Cardiomegaly/immunology , Chemotaxis/immunology , Female , Fibrosis , Hypoglycemic Agents/pharmacology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Myocytes, Cardiac/drug effects , Pioglitazone , Thiazolidinediones/pharmacology , Vasoconstrictor Agents/pharmacology
15.
J Environ Manage ; 67(4): 373-83, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12710925

ABSTRACT

Grassroots watershed associations have formed as an avenue to facilitate public involvement in protecting watersheds. Growth in the number of watershed associations has created a need to evaluate whether their existence enhances environmental protection by local communities. In this research, environmental protection was measured by the number of watershed protection activities conducted and amount of funding directed towards protecting surface waters by non-profit, volunteer organizations. Regression models were utilized to examine the influence of population demographic and watershed characteristics variables to explain the formation of watershed associations along with watershed protection activities and funding. Regression results showed that the presence of watershed associations had positive impacts on both the number of activities and financial resources acquired. The results confirm that watershed associations enhance the ability of communities to obtain funding and to implement watershed protection actions. Thus, watershed associations serve as an effective institution for promoting environmental protection.


Subject(s)
Conservation of Natural Resources , Environmental Pollution/prevention & control , Public Sector , Water Supply , Community-Institutional Relations , Demography , Humans , Program Evaluation , Volunteers , West Virginia
16.
Proc Natl Acad Sci U S A ; 99(11): 7604-9, 2002 May 28.
Article in English | MEDLINE | ID: mdl-12032330

ABSTRACT

The nuclear receptors LXRalpha and LXRbeta have been implicated in the control of cholesterol and fatty acid metabolism in multiple cell types. Activation of these receptors stimulates cholesterol efflux in macrophages, promotes bile acid synthesis in liver, and inhibits intestinal cholesterol absorption, actions that would collectively be expected to reduce atherosclerotic risk. However, synthetic LXR ligands have also been shown to induce lipogenesis and hypertriglyceridemia in mice, raising questions as to the net effects of these compounds on the development of cardiovascular disease. We demonstrate here that the nonsteroidal LXR agonist GW3965 has potent antiatherogenic activity in two different murine models. In LDLR(-/-) mice, GW3965 reduced lesion area by 53% in males and 34% in females. A similar reduction of 47% was observed in male apoE(-/-) mice. Long-term (12-week) treatment with LXR agonist had differential effects on plasma lipid profiles in LDLR(-/-) and apoE(-/-) mice. GW3965 induced expression of ATP-binding cassettes A1 and G1 in modified low-density lipoprotein-loaded macrophages in vitro as well as in the aortas of hyperlipidemic mice, suggesting that direct actions of LXR ligands on vascular gene expression are likely to contribute to their antiatherogenic effects. These observations provide direct evidence for an atheroprotective effect of LXR agonists and support their further evaluation as potential modulators of human cardiovascular disease.


Subject(s)
Arteriosclerosis/prevention & control , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Aorta/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Arteriosclerosis/blood , Arteriosclerosis/physiopathology , Body Weight , Cholesterol/blood , DNA-Binding Proteins , Female , Ligands , Liver X Receptors , Macrophages, Peritoneal/physiology , Mice , Mice, Knockout , Orphan Nuclear Receptors , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Triglycerides/blood
SELECTION OF CITATIONS
SEARCH DETAIL