Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Front Chem ; 10: 896386, 2022.
Article in English | MEDLINE | ID: mdl-35720993

ABSTRACT

Several neurodegenerative diseases are driven by misfolded proteins that assemble into soluble aggregates. These "toxic oligomers" have been associated with a plethora of cellular dysfunction and dysregulation, however the structural features underlying their toxicity are poorly understood. A major impediment to answering this question relates to the heterogeneous nature of the oligomers, both in terms of structural disorder and oligomer size. This not only complicates elucidating the molecular etiology of these disorders, but also the druggability of these targets as well. We have synthesized a class of bifunctional stilbenes to modulate both the conformational toxicity within amyloid beta oligomers (AßO) and the oxidative stress elicited by AßO. Using a neuronal culture model, we demonstrate this bifunctional approach has the potential to counter the molecular pathogenesis of Alzheimer's disease in a powerful, synergistic manner. Examination of AßO structure by various biophysical tools shows that each stilbene candidate uniquely alters AßO conformation and toxicity, providing insight towards the future development of structural correctors for AßO. Correlations of AßO structural modulation and bioactivity displayed by each provides insights for future testing in vivo. The multi-target activity of these hybrid molecules represents a highly advantageous feature for disease modification in Alzheimer's, which displays a complex, multifactorial etiology. Importantly, these novel small molecules intervene with intraneuronal AßO, a necessary feature to counter the cycle of dysregulation, oxidative stress and inflammation triggered during the earliest stages of disease progression.

2.
Free Radic Biol Med ; 143: 25-46, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31356870

ABSTRACT

Elevation of blood triglycerides, primarily triglyceride-rich lipoproteins (TGRL), is an independent risk factor for cardiovascular disease and vascular dementia (VaD). Accumulating evidence indicates that both atherosclerosis and VaD are linked to vascular inflammation. However, the role of TGRL in vascular inflammation, which increases risk for VaD, remains largely unknown and its underlying mechanisms are still unclear. We strived to determine the effects of postprandial TGRL exposure on brain microvascular endothelial cells, the potential risk factor of vascular inflammation, resulting in VaD. We showed in Aung et al., J Lipid Res., 2016 that postprandial TGRL lipolysis products (TL) activate mitochondrial reactive oxygen species (ROS) and increase the expression of the stress-responsive protein, activating transcription factor 3 (ATF3), which injures human brain microvascular endothelial cells (HBMECs) in vitro. In this study, we deployed high-throughput sequencing (HTS)-based RNA sequencing methods and mito stress and glycolytic rate assays with an Agilent Seahorse XF analyzer and profiled the differential expression of transcripts, constructed signaling pathways, and measured mitochondrial respiration, ATP production, proton leak, and glycolysis of HBMECs treated with TL. Conclusions: TL potentiate ROS by mitochondria which activate mitochondrial oxidative stress, decrease ATP production, increase mitochondrial proton leak and glycolysis rate, and mitochondria DNA damage. Additionally, CPT1A1 siRNA knockdown suppresses oxidative stress and prevents mitochondrial dysfunction and vascular inflammation in TL treated HBMECs. TL activates ATF3-MAPKinase, TNF, and NRF2 signaling pathways. Furthermore, the NRF2 signaling pathway which is upstream of the ATF3-MAPKinase signaling pathway, is also regulated by the mitochondrial oxidative stress. We are the first to report differential inflammatory characteristics of transcript variants 4 (ATF3-T4) and 5 (ATF3-T5) of the stress responsive gene ATF3 in HBMECs induced by postprandial TL. Specifically, our data indicates that ATF3-T4 predominantly regulates the TL-induced brain microvascular inflammation and TNF signaling. Both siRNAs of ATF3-T4 and ATF3-T5 suppress cells apoptosis and lipotoxic brain microvascular endothelial cells. These novel signaling pathways triggered by oxidative stress-responsive transcript variants, ATF3-T4 and ATF3-T5, in the brain microvascular inflammation induced by TGRL lipolysis products may contribute to pathophysiological processes of vascular dementia.


Subject(s)
Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Brain/pathology , Microvessels/injuries , Mitochondria/metabolism , Oxidative Stress , Apoptosis , Brain Injuries/metabolism , DNA Damage , Endothelial Cells/cytology , Endothelial Cells/metabolism , Genetic Variation , Glycolysis , Humans , Inflammation , Lipolysis , Microvessels/metabolism , Oxygen Consumption , Postprandial Period , Protons , RNA, Small Interfering/metabolism , RNA-Seq , Reactive Oxygen Species/metabolism , Signal Transduction , Superoxides/metabolism
3.
Food Funct ; 9(12): 6245-6256, 2018 Dec 13.
Article in English | MEDLINE | ID: mdl-30402637

ABSTRACT

Our lab previously demonstrated that triglyceride-rich lipoprotein (TGRL) lipolysis products induce lipid droplet formation and pro-inflammatory gene expression in monocytes. We hypothesized that the inhibition of perilipin 2 expression in THP-1 monocytes would reduce lipid droplet formation and suppress pro-inflammatory gene expression induced by TGRL lipolysis products. In the current study, we use microarray analysis to identify gene expression altered by TGRL lipolysis products in THP-1 monocytes. We confirmed the expression of selected genes by quantitative reverse transcription PCR and characterized lipid droplet formation in these cells after exposure to TGRL lipolysis products. Using siRNA inhibition of perilipin 2 expression, we examined the role of perilipin 2 in the response of THP-1 monocytes to TGRL lipolysis products. We found that perilipin 2 siRNA increased the intracellular triglyceride content, increased the size of lipid droplets, and reduced pro-atherogenic and pro-inflammatory gene expression. We saw a reduction of serum/glucocorticoid kinase 1, v-maf musculoaponeurotic fibrosarcoma oncogene homolog F (avian), chemokine (C-C motif) ligand 3, and interleukin 8 gene expression induced by TGRL lipolysis products. This study supports previous findings that reduction of perilipin 2 expression is protective against atherogenesis, while finding an unexpected increase in lipid droplet size with reduced perilipin 2 expression.


Subject(s)
Chemokine CCL3/genetics , Interleukin-8/genetics , Lipid Droplets/metabolism , Monocytes/drug effects , Perilipin-2/genetics , Adult , Chemokine CCL3/metabolism , Down-Regulation , Female , Humans , Interleukin-8/metabolism , Lipid Droplets/chemistry , Lipolysis , Lipoproteins/metabolism , Male , Monocytes/metabolism , Perilipin-2/metabolism , Triglycerides/metabolism , Young Adult
4.
Am J Physiol Cell Physiol ; 312(4): C500-C516, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28077357

ABSTRACT

Elevation of blood triglycerides, primarily as triglyceride-rich lipoproteins (TGRL), has been linked to cerebrovascular inflammation, vascular dementia, and Alzheimer's disease (AD). Brain microvascular endothelial cells and astrocytes, two cell components of the neurovascular unit, participate in controlling blood-brain barrier (BBB) permeability and regulating neurovascular unit homeostasis. Our studies showed that infusion of high physiological concentrations of TGRL lipolysis products (TGRL + lipoprotein lipase) activate and injure brain endothelial cells and transiently increase the BBB transfer coefficient (Ki = permeability × surface area/volume) in vivo. However, little is known about how blood lipids affect astrocyte lipid accumulation and inflammation. To address this, we first demonstrated TGRL lipolysis products increased lipid droplet formation in cultured normal human astrocytes. We then evaluated the transcriptional pathways activated in astrocytes by TGRL lipolysis products and found upregulated stress and inflammatory-related genes including activating transcription factor 3 (ATF3), macrophage inflammatory protein-3α (MIP-3α), growth differentiation factor-15 (GDF15), and prostaglandin-endoperoxide synthase 2 (COX2). TGRL lipolysis products also activated the JNK/cJUN/ATF3 pathway, induced endoplasmic reticulum stress protein C/EBP homologous protein (CHOP), and the NF-κB pathway, while increasing secretion of MIP-3α, GDF15, and IL-8. Thus our results demonstrate TGRL lipolysis products increase the BBB transfer coefficient (Ki), induce astrocyte lipid droplet formation, activate cell stress pathways, and induce secretion of inflammatory cytokines. Our observations are consistent with evidence for lipid-induced neurovascular injury and inflammation, and we, therefore, speculate that lipid-induced astrocyte injury could play a role in cognitive decline.


Subject(s)
Astrocytes/metabolism , Blood-Brain Barrier/physiology , Lipid Droplets/metabolism , Lipolysis/physiology , Lipoproteins/metabolism , Oxidative Stress/physiology , Triglycerides/metabolism , Animals , Cells, Cultured , Male , Mice , Mice, Inbred C57BL
5.
PLoS One ; 10(12): e0145523, 2015.
Article in English | MEDLINE | ID: mdl-26709509

ABSTRACT

Studies have suggested a link between the transforming growth factor beta 1 (TGF-ß1) signaling cascade and the stress-inducible activating transcription factor 3 (ATF3). We have demonstrated that triglyceride-rich lipoproteins (TGRL) lipolysis products activate MAP kinase stress associated JNK/c-Jun pathways resulting in up-regulation of ATF3, pro-inflammatory genes and induction of apoptosis in human aortic endothelial cells. Here we demonstrate increased release of active TGF-ß at 15 min, phosphorylation of Smad2 and translocation of co-Smad4 from cytosol to nucleus after a 1.5 h treatment with lipolysis products. Activation and translocation of Smad2 and 4 was blocked by addition of SB431542 (10 µM), a specific inhibitor of TGF-ß-activin receptor ALKs 4, 5, 7. Both ALK receptor inhibition and anti TGF-ß1 antibody prevented lipolysis product induced up-regulation of ATF3 mRNA and protein. ALK inhibition prevented lipolysis product-induced nuclear accumulation of ATF3. ALKs 4, 5, 7 inhibition also prevented phosphorylation of c-Jun and TGRL lipolysis product-induced p53 and caspase-3 protein expression. These findings demonstrate that TGRL lipolysis products cause release of active TGF-ß and lipolysis product-induced apoptosis is dependent on TGF-ß signaling. Furthermore, signaling through the stress associated JNK/c-Jun pathway is dependent on TGF-ß signaling suggesting that TGF-ß signaling is necessary for nuclear accumulation of the ATF3/cJun transcription complex and induction of pro-inflammatory responses.


Subject(s)
Activating Transcription Factor 3/biosynthesis , Endothelial Cells/metabolism , Lipoproteins/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Triglycerides/metabolism , Active Transport, Cell Nucleus , Animals , Aorta/metabolism , Apoptosis , Caspase 3/metabolism , Cells, Cultured , Humans , Lipolysis , Lipoproteins/genetics , Mice , Phosphorylation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Smad2 Protein/metabolism , Smad4 Protein/metabolism , Stress, Physiological , Transforming Growth Factor beta1/metabolism , Triglycerides/genetics , Tumor Suppressor Protein p53/metabolism
6.
Phys Chem Chem Phys ; 16(39): 21595-21601, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25189785

ABSTRACT

Activating transcription factor 3 (ATF3) is a member of the mammalian activation transcription factor/cAMP, physiologically important in the regulation of pro- and anti-inflammatory target genes. We compared the induction of ATF3 protein as measured by Western blot analysis with single-molecule localization microscopy dSTORM to quantify the dynamics of accumulation of intranuclear ATF3 of triglyceride-rich (TGRL) lipolysis product-treated HAEC (Human Aortic Endothelial Cells). The ATF3 expression rate within the first three hours after treatment with TGRL lipolysis products is about 3500 h(-1). After three hours we detected 33,090 ± 3491 single-molecule localizations of ATF3. This was accompanied by significant structural changes in the F-actin network of the cells at ∼3-fold increased localization precision compared to widefield microscopy after treatment. Additionally, we discovered a cluster size of approximately 384 nanometers of ATF3 molecules. We show for the first time the time course of ATF3 accumulation in the nucleus undergoing lipotoxic injury. Furthermore, we demonstrate ATF3 accumulation associated with increased concentrations of TGRL lipolysis products occurs in large aggregates.


Subject(s)
Activating Transcription Factor 3/biosynthesis , Activating Transcription Factor 3/metabolism , Aorta/cytology , Aorta/metabolism , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Lipolysis
7.
BMC Syst Biol ; 8: 80, 2014 Jul 04.
Article in English | MEDLINE | ID: mdl-24993133

ABSTRACT

BACKGROUND: Neurovascular inflammation is associated with a number of neurological diseases including vascular dementia and Alzheimer's disease, which are increasingly important causes of morbidity and mortality around the world. Lipotoxicity is a metabolic disorder that results from accumulation of lipids, particularly fatty acids, in non-adipose tissue leading to cellular dysfunction, lipid droplet formation, and cell death. RESULTS: Our studies indicate for the first time that the neurovascular circulation also can manifest lipotoxicity, which could have major effects on cognitive function. The penetration of integrative systems biology approaches is limited in this area of research, which reduces our capacity to gain an objective insight into the signal transduction and regulation dynamics at a systems level. To address this question, we treated human microvascular endothelial cells with triglyceride-rich lipoprotein (TGRL) lipolysis products and then we used genome-wide transcriptional profiling to obtain transcript abundances over four conditions. We then identified regulatory genes and their targets that have been differentially expressed through analysis of the datasets with various statistical methods. We created a functional gene network by exploiting co-expression observations through a guilt-by-association assumption. Concomitantly, we used various network inference algorithms to identify putative regulatory interactions and we integrated all predictions to construct a consensus gene regulatory network that is TGRL lipolysis product specific. CONCLUSION: System biology analysis has led to the validation of putative lipid-related targets and the discovery of several genes that may be implicated in lipotoxic-related brain microvascular endothelial cell responses. Here, we report that activating transcription factors 3 (ATF3) is a principal regulator of TGRL lipolysis products-induced gene expression in human brain microvascular endothelial cell.


Subject(s)
Brain/blood supply , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Lipid Metabolism/drug effects , Metabolic Diseases/metabolism , Microvessels/pathology , Systems Biology , Activating Transcription Factor 3/metabolism , Apoptosis/drug effects , Gene Expression Regulation/drug effects , Humans , Lipolysis/drug effects , Lipoproteins/chemistry , Lipoproteins/pharmacology , Metabolic Diseases/pathology , Signal Transduction/drug effects , Triglycerides/chemistry
8.
Arterioscler Thromb Vasc Biol ; 33(9): 2088-96, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23868936

ABSTRACT

OBJECTIVE: Elevation of triglyceride-rich lipoproteins (TGRLs) contributes to the risk of atherosclerotic cardiovascular disease. Our work has shown that TGRL lipolysis products in high physiological to pathophysiological concentrations cause endothelial cell injury; however, the mechanisms remain to be delineated. APPROACH AND RESULTS: We analyzed the transcriptional signaling networks in arterial endothelial cells exposed to TGRL lipolysis products. When human aortic endothelial cells in culture were exposed to TGRL lipolysis products, activating transcription factor 3 (ATF3) was identified as a principal response gene. Induction of ATF3 mRNA and protein was confirmed by quantitative reverse-transcription polymerase chain reaction and Western blot respectively. Immunofluorescence analysis showed that ATF3 accumulated in the nuclei of cells treated with lipolysis products. Nuclear expression of phosphorylated c-Jun N-terminal kinase (JNK), previously shown to be an initiator of the ATF3 signaling cascade, also was demonstrated. Small interfering RNA (siRNA)-mediated inhibition of ATF3 blocked lipolysis products-induced transcription of E-selectin and interleukin-8, but not interleukin-6 or nuclear factor-κB. c-Jun, a downstream protein in the JNK pathway, was phosphorylated, whereas expression of nuclear factor-κB-dependent JunB was downregulated. Additionally, JNK siRNA suppressed ATF3 and p-c-Jun protein expression, suggesting that JNK is upstream of the ATF3 signaling pathway. In vivo studies demonstrated that infusion of TGRL lipolysis products into wild-type mice induced nuclear ATF3 accumulation in carotid artery endothelium. ATF3(-/-) mice were resistant to vascular apoptosis precipitated by treatment with TGRL lipolysis products. Also peripheral blood monocytes isolated from postprandial humans had increased ATF3 expression as compared with fasting monocytes. CONCLUSIONS: This study demonstrates that TGRL lipolysis products activate ATF3-JNK transcription factor networks and induce endothelial cells inflammatory response.


Subject(s)
Activating Transcription Factor 3/metabolism , Apoptosis , Endothelial Cells/metabolism , Inflammation/metabolism , Lipoproteins/metabolism , Triglycerides/metabolism , Activating Transcription Factor 3/deficiency , Activating Transcription Factor 3/genetics , Animals , Blotting, Western , Cells, Cultured , E-Selectin/metabolism , Endothelial Cells/immunology , Endothelial Cells/pathology , Enzyme Activation , Fluorescent Antibody Technique , Gene Expression Profiling/methods , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Inflammation Mediators/metabolism , Interleukin-8/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Leukocytes, Mononuclear/metabolism , Lipolysis , Lipoprotein Lipase/metabolism , Lipoproteins/blood , Lipoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Phosphorylation , RNA Interference , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Triglycerides/blood
9.
Physiol Genomics ; 43(15): 917-29, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21652769

ABSTRACT

Epidemiologic studies associate exposure to ambient particulate matter (APM) with increased cardiovascular mortality. Since both pulmonary inflammation and systemic circulation of ultrafine particles are hypothesized as initiating cardiovascular effects, we examined responses of potential target cells in vitro. Human aortic endothelial cells (HAEC) were exposed to 10 µg/ml fine and ultrafine APM collected in an urban setting in summer 2006 or winter 2007 in the San Joaquin Valley, California. RNA isolated after 3 h was analyzed with high-density oligonucleotide arrays. Summer APM treatment affected genes involved in xenobiotic and oxidoreductase activity, transcription factors, and inflammatory responses in HAEC, while winter APM had a robust xenobiotic but lesser inflammatory response. Real-time polymerase chain reaction analysis confirmed that particulate matter (PM)-treated HAEC increased mRNA levels of xenobiotic response enzymes CYP1A1, ALDH1A3, and TIPARP and cellular stress response transcription factor ATF3. Inflammatory response genes included E-selectin, PTGS2, CXCL-2 (MIP-2α), and CCL-2 (MCP-1). Multiplex protein assays showed secretion of IL-6 and MCP-1 by HAEC. Since induction of CYP1A1 is mediated through the ligand-activated aryl hydrocarbon receptor (AhR), we demonstrated APM induced AhR nuclear translocation by immunofluorescence and Western blotting and activation of the AhR response element using a luciferase reporter construct. Inhibitor studies suggest differential influences of polycyclic aromatic hydrocarbon signaling, ROS-mediated responses and endotoxin alter stress and proinflammatory endothelial cell responses. Our findings demonstrate gene responses correlated with current concepts that systemic inflammation drives cardiovascular effects of particulate air pollution. We also demonstrate a unique pattern of gene responses related to xenobiotic metabolism in PM-exposed HAEC.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/metabolism , Particulate Matter/toxicity , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CXCL2/genetics , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 Enzyme System/genetics , E-Selectin/genetics , Humans , Interleukin-6/metabolism , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Receptors, Aryl Hydrocarbon/genetics
10.
Nat Rev Nephrol ; 6(6): 361-70, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20440276

ABSTRACT

Diabetic nephropathy is an increasingly important cause of morbidity and mortality worldwide. A large body of evidence suggests that dyslipidemia has an important role in the progression of kidney disease in patients with diabetes. Lipids may induce renal injury by stimulating TGF-beta, thereby inducing the production of reactive oxygen species and causing damage to the glomeruli and glomerular glycocalyx. Findings from basic and clinical studies strongly suggest that excess amounts of a variety of lipoproteins and lipids worsens diabetes-associated microvascular and macrovascular disease, increases glomerular injury, increases tubulointerstitial fibrosis, and accelerates the progression of diabetic nephropathy. The increasing prevalence of obesity, type 2 diabetes mellitus, and diabetic nephropathy means that interventions that can interrupt the pathophysiological cascade of events induced by lipoproteins and lipids could enable major life and cost savings. This Review discusses the structural, cellular, and microscopic findings associated with diabetic nephropathy and the influence of lipoproteins, specifically triglyceride-rich lipoproteins (TGRLs), on the development and perpetuation of diabetic nephropathy. Some of the accepted and hypothesized mechanisms of renal injury relating to TGRLs are also described.


Subject(s)
Diabetic Nephropathies/metabolism , Diabetic Nephropathies/physiopathology , Dyslipidemias/metabolism , Dyslipidemias/physiopathology , Lipoproteins/metabolism , Triglycerides/metabolism , Animals , Diabetic Nephropathies/drug therapy , Dyslipidemias/drug therapy , Humans , Hypolipidemic Agents/therapeutic use , Lipoproteins/drug effects
11.
Genes Nutr ; 4(1): 23-39, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19104882

ABSTRACT

Male C57BL/6 mice were fed diets supplemented with either beta-carotene (BC) or lycopene (LY) that were formulated for human consumption. Four weeks of dietary supplementations results in plasma and lung carotenoid (CAR) concentrations that approximated the levels detected in humans. Bioactivity of the CARs was determined by assaying their effects on the activity of the lung transcriptome (~8,500 mRNAs). Both CARs activated the cytochrome P450 1A1 gene but only BC induced the retinol dehydrogenase gene. The contrasting effects of the two CARs on the lung transcriptome were further uncovered in mice exposed to cigarette smoke (CS) for 3 days; only LY activated ~50 genes detected in the lungs of CS-exposed mice. These genes encoded inflammatory-immune proteins. Our data suggest that mice offer a viable in vivo model for studying bioactivities of dietary CARs and their modulatory effects on lung genomic expression in both health and after exposure to CS toxicants.

12.
J Am Coll Nutr ; 27(2): 267-73, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18689558

ABSTRACT

OBJECTIVE: While tomato product supplementation, containing antioxidant carotenoids, including lycopene, decreases oxidative stress, the role of purified lycopene as an antioxidant remains unclear. Thus, we tested the effects of different doses of purified lycopene supplementation on biomarkers of oxidative stress in healthy volunteers. METHODS: This was a double-blind, randomized, placebo-controlled trial, examining the effects of 8-week supplementation of purified lycopene, on plasma lycopene levels, biomarkers of lipid peroxidation {LDL oxidizability, malondialdehyde & hydroxynonenals (MDA & HNE), urinary F(2)-isoprostanes}, and markers of DNA damage in urine and lymphocytes. Healthy adults (n = 77, age > or = 40 years), consumed a lycopene-restricted diet for 2 weeks, and were then randomized to receive 0, 6.5, 15, or 30 mg lycopene/day for 8 weeks, while on the lycopene-restricted diet. Blood and urine samples were collected at the beginning and end of Week 2 of lycopene-restricted diet, and at end of Week 10 of the study. RESULTS: Independent of the dose, plasma lycopene levels significantly increased in all lycopene supplemented groups versus placebo (p < 0.05). ANOVA revealed a significant decrease in DNA damage by the comet assay (p = 0.007), and a significant decrease in urinary 8-hydroxy deoxoguanosine (8-OHdG) at 8 weeks versus baseline (p = 0.0002), with 30 mg lycopene/day. No significant inter- or intra-group differences were noted for glucose, lipid profile, or other biomarkers of lipid peroxidation at any dose/time point. CONCLUSIONS: Thus, purified lycopene was bioavailable and was shown to decrease DNA oxidative damage and urinary 8-OHdG at the high dose.


Subject(s)
Antioxidants/administration & dosage , Carotenoids/administration & dosage , Carotenoids/blood , 8-Hydroxy-2'-Deoxyguanosine , Adult , Aldehydes/blood , Biomarkers/metabolism , Blood Glucose/metabolism , Comet Assay , DNA/drug effects , DNA/metabolism , DNA Damage , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/urine , Dietary Supplements , Dose-Response Relationship, Drug , Double-Blind Method , F2-Isoprostanes/urine , Humans , Lipid Peroxidation/drug effects , Lipoproteins, LDL/blood , Lycopene , Malondialdehyde/blood , Oxidative Stress/drug effects , Oxidative Stress/physiology
13.
J Lipid Res ; 49(3): 521-30, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18033753

ABSTRACT

Angiotensin II (angII) is known to promote atherosclerosis; however, the mechanisms involved are not fully understood. To determine whether angII stimulates proteoglycan production and LDL retention, LDL receptor-deficient mice were infused with angII (1,000 ng/kg/min) or saline via osmotic minipumps. To control for the hypertensive effect of angII, a parallel group received norepinephrine (NE; 5.6 mg/kg/day). Arterial lipid accumulation was evaluated by measuring the retention rate of LDL in isolated carotid arteries perfused ex vivo. Mice infused with angII had increased vascular content of biglycan and perlecan and retained twice as much LDL as saline- or NE-infused mice, although no group developed atherosclerosis at this time. To determine whether this increase in biglycan and perlecan content predisposed to atherosclerosis development, mice were infused with angII, saline, or NE for 4 weeks, then pumps were removed and mice received an atherogenic Western diet for another 6 weeks. Mice that had received angII infusions had 3-fold increased atherosclerosis compared with mice that had received saline or NE, and apolipoprotein B colocalized with both proteoglycans. Thus, one mechanism by which angII promotes atherosclerosis is increased proteoglycan synthesis and increased arterial LDL retention, which precedes and contributes to atherosclerosis development.


Subject(s)
Angiotensin II/pharmacology , Atherosclerosis/etiology , Blood Vessels/chemistry , Proteoglycans/biosynthesis , Angiotensin II/administration & dosage , Animals , Apolipoproteins B/metabolism , Carotid Arteries , Lipoproteins, LDL/analysis , Mice , Mice, Knockout , Norepinephrine/administration & dosage , Proteoglycans/drug effects , Receptors, LDL/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...