当前位置: 首页 > 医学版 > 期刊论文 > 内科学 > 糖尿病学杂志 > 2005年 > 第5期 > 正文
编号:11256521
Linoleic Acid Increases Lectin-Like Oxidized LDL Receptor-1 (LOX-1) Expression in Human Aortic Endothelial Cells
     1 Department of Nutrition, University of Montreal, Centre Hospitalier de l’Universitee de Montreeal (CHUM) Research Centre, Notre-Dame Hospital, Montreal, Quebec, Canada

    2 Department of Medicine, University of Montreal, CHUM Research Centre, Notre-Dame Hospital, Montreal, Quebec, Canada

    ABSTRACT

    Results from in vitro studies suggest that selected fatty acids, and especially linoleic acid (LA), can elicit endothelial dysfunction (ED). Because LA is increased in all LDL subfractions in patients with type 2 diabetes, this alteration may contribute to ED associated with diabetes. Lectin-like oxidized LDL receptor-1 (LOX-1) is the major endothelial receptor for oxidized LDL (oxLDL), and uptake of oxLDL through LOX-1 induces ED. To evaluate whether LA may contribute to the upregulation of endothelial LOX-1 in diabetes, we studied the effect of LA on LOX-1 expression in cultured human aortic endothelial cells (HAECs). Treatment of HAECs with LA increased, in a time- and dose-dependent manner, endothelial LOX-1 protein expression. Pretreatment of HAECs with antioxidants and inhibitors of NADPH oxidase, protein kinase C (PKC), and nuclear factor-B (NF-B) inhibited the stimulatory effect of LA on LOX-1 protein expression. Furthermore, in LA-treated HAECs, increased expression of classic PKC isoforms was observed. LA also led to a significant increase in LOX-1 gene expression and enhanced the binding of nuclear proteins extracted from HAECs to the NF-B regulatory element of the LOX-1 gene promoter. Finally, LA enhanced, through LOX-1, oxLDL uptake by endothelial cells. Overall, these results demonstrate that LA enhances endothelial LOX-1 expression through oxidative stress-sensitive and PKC-dependent pathways. This effect seems to be exerted at the transcriptional level and to involve the activation of NF-B. Upregulation of LOX-1 by LA may contribute to ED associated with type 2 diabetes.

    Endothelial dysfunction (ED), a characteristic feature of early-state atherosclerosis, is well documented in patients with diabetes and in individuals with insulin resistance or at high risk for developing type 2 diabetes (1,2). Critical factors that may induce activation and injury to the endothelium in the diabetic state include oxidized LDL (oxLDL) (3) and selected unsaturated fatty acids, such as linoleic acid (LA) (4). One potential key determinant of oxLDL-induced ED is lectin-like oxidized LDL receptor-1 (LOX-1), a newly identified limiting factor for oxLDL uptake by endothelial cells (5). Data showing that LOX-1 expression is enhanced by proatherogenic factors relevant to human diabetes, including oxLDL (6), high glucose (7), advanced glycation end products (8), and C-reactive protein (9), and is upregulated in endothelium and aortas of diabetic animals (8) support a role of this receptor in the pathogenesis of diabetic vascular dysfunction. Substantial amounts of data have been accumulated to show that LA is a pro-oxidative and proinflammatory molecule (10eC12) that can induce endothelial cell activation and dysfunction. These data and the observation that patients with type 2 diabetes show increased concentrations of LA in all LDL subfractions (13) support the significance of this fatty acid in ED associated with diabetes. In the present study, we investigated the role of LA in the regulation of endothelial LOX-1, the molecular mechanisms involved in this effect, and the role of LOX-1 in LA-induced oxLDL uptake by endothelial cells.

    RESEARCH DESIGN AND METHODS

    Reagents.

    FCS was purchased from Wisent (St. Bruno, PQ, Canada). Hank’s balanced salt solution, penicillin-streptomycin, glycine, SDS, and Trizol reagent were obtained from Invitrogen Life Technologies (Burlington, ON, Canada). Human aortic endothelial cells (HAECs), endothelial growth culture medium (EGM), and an EGM bullet kit were obtained from Cedarlane Laboratories Limited (Hornby, ON, Canada). RPMI-1640 was obtained from Gibco (Grand Island, NY). Sodium salt LA, fatty acid-free BSA fraction V, isopropanol, vitamin C, and monoclonal antibody against -actin were purchased from Sigma (St. Louis, MO). Ficoll and horseradish peroxidase-conjugated anti-mouse IgG were obtained from Amersham Biosciences (Buckinghamshire, U.K.). Monoclonal antibodies against LOX-1, p50, p65, and protein kinase C (PKC)-, -I, -II, and - were bought from Santa Cruz Biotechnology (Santa Cruz, CA). IgG1 neutralizing antibody was purchased from R&D Systems (Minneapolis, MN). Calphostin C, GF109203X, BAY11-7085, N-acetyl-L-cysteine (NAC), curcumin, apocynin, thenoyltrifluoroacetone, Mn(III)tetrakis (4-benzoic acid), and diphenyleneiodonium chloride were obtained from Calbiochem (La Jolla, CA).

    Cells.

    Commercially available HAECs (passage 3) were grown to confluence in EGM under recommended conditions. The EGM was supplemented with 10% FCS containing 0.01 e/ml human epidermal growth factor, 0.1% gentamicin sulfate amphotericin-B, 1 e/ml hydrocortisone, and 12 e/ml bovine brain extract protein content. Cells were used at passages 3eC5. Evidence that these cells express von Willebrand factor, adhesion molecules, and cytokines and demonstrate plasminogen activator inhibitor-1 activity supports their relevance as a model for the study of native vascular endothelium. Confluent HAECs were incubated for different time periods in serum-free EBM supplemented with 1% penicillin-streptomycin in the presence of 0.2 mmol/l LA bound to 2% BSA. Serum-free EBM supplemented with 2% BSA was used as a control. Under these experimental conditions, the fatty acid-to-albumin molar ratio was 1, a value that is within physiological levels.

    Analysis of mRNA expression.

    Expression of the LOX-1 gene in HAECs was measured by the PCR technique. Total RNA for use in the PCR was extracted from cells by an improvement of the acid-phenol technique of Chomczynski and Sacchi. Briefly, cells were lysed with Trizol reagent and chloroform was added to the solution. After centrifugation, the RNA present in the aqueous phase was precipitated and resuspended in diethyl pyrocarbonate water. cDNA was synthesized from RNA by incubating total cellular RNA with 0.1 e oligodT (Pharmacia) for 5 min at 98°C and then by incubating the mixture with reverse transcription buffer for 1 h at 37°C. The cDNA obtained was amplified by using 0.8 eol/l of two synthetic primers specific for human LOX-1 (5'-TTACTCTCCATGGTGGTGGTGCC-3') (5'-AGCTTCTTCTGCTTGTTGCC-3') and human -actin (5'-TCGAATTCTGAGAAGAGCTATGAGCTGCCG-3') (5'-TCGCATCCGTGCCACCAGACAGCACTGTGTTG-3') used as the internal standard in the PCR mixture. A 193-base pair human LOX-1 cDNA fragment and a 201-base pair human -actin cDNA fragment were amplified enzymatically by 40 and 20 repeated cycles, respectively. An aliquot of each reaction mixture was then subjected to electrophoresis on 1% agarose gel containing ethidium bromide. The intensity of the bands was measured by an image analysis scanning system (Alpha Imager 2000, Packard Instrument Company). Titrating the cDNA samples ensured that the signal lies on the exponential part of the standard curve.

    Western blot.

    HAEC protein extracts (8 e) were applied to 10% SDS-PAGE and transferred to a nitrocellulose membrane using a Bio-Rad transfer blotting system at 100 V for 1 h. Nonspecific binding was blocked with 5% milk for 1 h at room temperature. After washing with PBS-Tween 0.1%, blots were incubated overnight at 4°C with anti-LOX-1 or anti--actin antibodies. After further washing, membranes were incubated for 1 h at room temperature with a horseradish peroxidase-conjugated donkey anti-mouse IgG (1/5,000). Antigen detection was performed with an enhanced chemiluminescence detection system (Amersham).

    For PKC isoforms analysis, HAECs were recovered and homogenized (Dounce: 15 strokes) in 500 e蘬 ice-cold buffer A (20 mmol/l Tris [pH 7.5], 0.5 mmol/l EDTA, 0.5 mmol/l EGTA, 25 e/ml aprotinin, and 25 e/ml leupeptin). The membrane and cytosolic fractions were separated by ultracentrifugation (100,000g for 30 min at 4°C). After recovery of high-speed supernatants containing cytosolic PKC, the corresponding membrane pellets were homogenized in 500 e蘬 buffer A containing 0.5% Triton X-100 and assayed for classic PKC isoforms by Western blot analysis.

    Uptake of 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate-labeled oxLDL.

    Native LDL (density 1.019eC1.063) was isolated from plasma obtained from healthy donors by sequential ultracentrifugation using potassium bromide for density adjustment. Native LDL was extensively dialyzed for 24 h at 4°C against 5 mmol/l Tris/50 nmol/l NaCl to remove EDTA. 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI)-labeled oxLDL (DiI-oxLDL) was obtained from Biomedical Technologies (Stoughton, MA). To examine cellular uptake of oxLDL, HAECs were seated in eight-well culture slides (Falcon) and incubated for 3 h in medium containing 5% of lipoprotein-deficient serum with DiI-oxLDL (80 e/ml) in the presence or absence of a 500-fold excess of unlabeled oxLDL. At the end of the incubation period, cells were washed, mounted on coverslips with mounting medium, and examined by fluorescence microscopy. To measure amounts of DiI-oxLDL accumulated in cells, HAECs were seated in 12-well plates receiving the same treatment as mentioned above. At the end of the incubation period, DiI was extracted by isopropanol, and the fluorescence was determined at 520/564 nm. Results were normalized to total cell protein concentrations (Bradford).

    DNA binding assay.

    The isolation of the nuclei was performed as follows. Briefly, 5 x 107 HAECs were washed and collected with 10 ml ice-cold PBS/phosphatase inhibitor buffer (125 mmol/l sodium fluoride, 250 mmol/l -glycerophosphate, 250 mmol/l para-nitrophenyl phosphate, and 25 mmol/l sodium orthovanadate). The cells were transferred to a prechilled 15-ml tube and centrifuged at 3,000 rpm for 5 min at 4°C. The pellet was resuspended in 1 ml ice-cold hypotonic buffer (20 mmol/l HEPES, pH 7.5, 5 mmol/l sodium fluoride, 10 eol/l sodium molybdate, and 0.1 mmol/l EDTA). After a 15-min incubation on ice, 50 e蘬 Nonidet-P40 (NP-40) (10%) was added, and the homogenate was mixed and centrifuged for 30 s at 4°C in a microcentrifuge. The nuclear pellet was resuspended in 50 e蘬 lysis buffer (1 mmol/l dithiothreitol, 2 mol/l potassium chloride, 25 mmol/l HEPES, 0.1 mmol/l EDTA, 1 e/ml aprotinin, and 2 mmol/l phenylmethylsulfonyl fluoride) and rocked on ice for 30 min. Nuclei were collected by centrifugation for 20 min at 13,000 rpm. Aliquots (50 e蘬) of the supernatants were frozen at eC80°C, and the protein concentration was determined. DNA retardation (mobility shift) electrophoresis assays were performed as follows. Briefly, 5-e nuclear extracts were incubated for 15 min in the presence of 5x binding buffer (125 mmol/l HEPES, pH 7.5, 50% glycerol, 250 mmol/l sodium chloride, 0.25% NP-40, and 5 mmol/l dithiothreitol). End-labeled double-stranded consensus sequences of the LOX-1 promoter nuclear factor (NF)-B-enhancing elements (20,000 cpm per sample) were then added to the samples for 30 min. Samples were analyzed on a 4% PAGE containing 0.01% NP-40. The specificity of the nuclear protein binding was assessed by incubating the nuclear proteins isolated from HAECs with a labeled DNA probe in the presence of a 1,000-fold molar excess of unlabeled DNA probe.

    DNA probes.

    Double-stranded oligonucleotides containing the NF-B (5-CGTCTGCCCTTTCCCCCTCT-3 and 5-GAGAAGAGGGGGAAAGG-3) consensus sequence of the human LOX-1 gene promoter (14) were synthesized with the aid of an automated DNA synthesizer. After annealing, the oligonucleotides were labeled with [-32P]ATP using the Boehringer Mannheim 5' end-labeling kit (Indianapolis, IN).

    Determination of cell viability.

    Cell viability after treatment with LA was assessed by trypan blue exclusion and was consistently found to be >80%.

    Determination of total protein concentration.

    Total protein content was estimated according to the Bradford method using a colorimetric assay (Bio-Rad, Mississauga, ON, Canada).

    Statistical analysis.

    All values were expressed as means ± SE. Data were analyzed by one-way ANOVA, followed by the Turkey test. A P value <0.05 was considered statistically significant.

    RESULTS

    Effect of LA on endothelial LOX-1 mRNA expression.

    Treatment of HAECs for 15eC24 h with 0.2 mmol/l LA significantly increased LOX-1 mRNA levels (LOX-1 mRNA levels [percent increase over control values] at 15 h: 161 ± 13, P < 0.001; 18 h: 178 ± 40, P < 0.01; 24 h: 187 ± 19, P < 0.001) (Fig. 1A). Under these experimental conditions, no modulation of the mRNA expression of -actin was observed (Fig. 1B). LOX-1 mRNA levels normalized to the levels of -actin mRNA are presented in Fig. 1C.

    Effect of LA on endothelial LOX-1 protein expression.

    To determine whether the increased LOX-1 mRNA expression induced by LA resulted in enhanced protein expression, HAECs were treated with 0.2 mmol/l LA for 6eC48 h and LOX-1 protein expression was determined by Western blot analysis. Exposure of HAECs for 15eC48 h to LA significantly increased LOX-1 protein levels (Fig. 2A-a). Maximal effect was observed at 24 h. LOX-1 protein levels normalized to the levels of -actin (Fig. 2A-b) are illustrated in Fig. 2A-c. In contrast, treatment of HAECs for 24 h with 0.2 mmol/l palmitic acid, oleic acid, or docosahexaenoic acid did not affect LOX-1 protein expression (data not shown). LA-induced LOX-1 protein expression was dose dependent (Fig. 2B-a). Induction of LOX-1 protein was already observed at 0.05 mmol/l LA and reached a maximum at 0.2 mmol/l. LOX-1 protein levels normalized to the levels of -actin (Fig. 2B-b) are illustrated in Fig. 2B-c.

    Role of oxidative stress in LA-induced LOX-1 protein expression.

    To evaluate whether oxidative stress is involved in the upregulation of LOX-1 by LA, HAECs were preincubated for 1 h with vitamin C (10 eol/l) or NAC (10 mmol/l) before exposure to LA. Pretreatment of HAECs with these antioxidants totally prevented LA-induced LOX-1 protein expression (Fig. 3A). LOX-1 protein levels normalized to the levels of -actin (Fig. 3B) are illustrated in Fig. 3C. (LOX-1 protein levels [percent increase over control values] are as follows: LA: 199 ± 13; LA + vitamin C: 98 ± 22, P < 0.01 vs. LA; LA + NAC: 77 ± 9, P < 0.001 vs. LA).

    Role of NADPH oxidase in LA-induced LOX-1 protein expression.

    To ascertain the enzymatic superoxide sources activated by LA, HAECs were preincubated for 1 h with the NADPH oxidase inhibitors apocynin (10 eol/l) and diphenyleneiodonium chloride (10 eol/l) or with the mitochondrial electron transport chain inhibitors thenoyltrifluoroacetone (10 eol/l) and Mn(III)tetrakis (4-benzoic acid) (100 eol/l) prior to treatment with 0.2 mmol/l LA. Pretreatment of HAECs with NADPH oxidase inhibitors prevented LA-induced LOX-1 protein expression, whereas mitochondrial electron transport chain inhibitors were ineffective (Fig. 4).

    Role of PKC and NF-B in LA-induced LOX-1 protein expression.

    To examine the signaling pathways mediating the effect of LA on LOX-1, HAECs were treated for 1 h with the pan-specific PKC inhibitor calphostin C (0.1 e/ml), the NF-B inhibitor BAY11-7085 (40 eol/l), or the activator protein-1 inhibitor curcumin (15 eol/l). As shown in Fig. 5, pretreatment of the cells with calphostin C and BAY11-7085, but not with curcumin, totally abolished the stimulatory effect of LA on LOX-1 protein expression.

    Effect of LA on PKC expression.

    To determine the expression of classic PKC isozymes in LA-treated cells, HAECs were treated for 15 h with 0.2 mmol/l LA, and expression of these isoforms was determined by Western blot analysis. As illustrated in Fig. 6, LA increased the expression of PKC- (1.9-fold), PKC-I (2.3-fold), PKC-II (3.5-fold), and PKC- (3.7-fold). Under these experimental conditions, no modulation of -actin, used as an internal control, was observed.

    Effect of LA on the binding of nuclear proteins to the regulatory NF-B sequence of the LOX-1 gene promoter.

    To determine whether the incubation of HAECs with LA might result in changes at the level of LOX-1 gene binding proteins, we measured the binding activity of nuclear proteins extracted from LA-treated HAECs to the NF-B consensus sequence of the LOX-1 gene promoter. Experiments showed that incubation of these cells for 6 h with LA resulted in a significant increase in the binding of nuclear proteins to the regulatory NF-B sequence of the LOX-1 gene promoter (Fig. 7). This binding complex was specifically competed in the presence of a 1,000-fold molar excess of the unlabeled NF-B oligonucleotide. This effect was also reduced by pretreatment of HAECs with the p65 antibody, while preincubation of the cells with the p50 antibody calphostin C, GF109203X (a classic PKC selective inhibitor), BAY11-7085, NAC, or vitamin C totally abolished the stimulatory effect of LA on the binding of nuclear proteins to the NF-B sequence of the LOX-1 gene promoter (Fig. 7).

    LA stimulates the uptake of oxLDL by endothelial cells through LOX-1.

    To investigate whether LA might facilitate the uptake of oxLDL by endothelial cells and whether this effect involves LOX-1, HAECs were treated with 0.2 mmol/l LA for 23 h; then incubation was pursued for 1 h in the presence of saturating amounts of antibodies to LOX-1 or IgG1 or in the presence of a 500-fold excess of unlabeled oxLDL. At the end of the incubation period, cells were incubated for 3 h with DiI-oxLDL (80 e/ml), and uptake of oxLDL was determined by measurement of extracted DiI-oxLDL and fluorescence microscopy. Incubation of HAECs with LA increased the uptake of oxLDL by these cells by more than twofold. This effect was totally blocked by pretreatment of the cells with anti-LOX-1 antibody or in the presence of excess unlabeled oxLDL. In contrast, no effect of the anti-IgG1 antibody on LA-induced oxLDL uptake by endothelial cells was observed (Fig. 8).

    DISCUSSION

    The hypothesis that release of fatty acids during lipoprotein lipase-mediated triglyceride hydrolysis may be atherogenic by causing endothelial injury was first proposed in the 1970s by Zilversmit (15). Today, it is generally accepted that selected fatty acids, and especially n-6 unsaturated fatty acids derived from the hydrolysis of lipoproteins, may cause ED. Of all the fatty acids, LA appears to be the one with the most profound and deleterious effects on endothelial barrier function (4,16). This could be related to the very low basal activity of elongase and desaturase enzymes in endothelial cells, thus preventing arachidonic acid formation from LA in these cells (17). Mechanisms of LA-mediated ED are numerous. They include disruption of endothelial cell integrity with alterations of functions of gap-junctions proteins (18), cell morphology (19), membrane-bound enzymes (20), peroxysome (21), and induc-tion of apoptotic cell death (22). Last but not least, accumulating evidences suggest that increase in intracellular oxidative stress and calcium levels (10,23) could be the main mechanisms by which LA exerts its harmful effects on endothelial cells.

    The significance of fatty acids, and especially LA, in the development of ED associated with human diabetes is supported by several observations. First, elevated levels of fatty acid-containing lipoproteins and circulating levels of free fatty acids are reported in diabetes (24,25). Second, upregulation of stearoyl-CoA desaturase by glucose may enhance the conversion of saturated to unsaturated fatty acids in the diabetic state (26). Third, lipoprotein lipase, the rate-limiting enzyme for triglyceride hydrolysis (15), is increased in patients with type 2 diabetes (27) and upregulated by fatty acids, including LA (28). Such high lipoprotein lipase activity in diabetes is likely to favor endothelial cell exposure to high levels of free fatty acids. Finally, increased concentrations of LA have been documented in all LDL subfractions in patients with type 2 diabetes (13).

    A role for LOX-1 as a major pathogenic determinant of ED associated with type 2 diabetes has recently been proposed. Consistent with this concept, it has been shown that endothelial LOX-1 is increased in diabetic animals and upregulated by metabolic and inflammatory factors dysregulated in diabetes, including glucose (7), advanced glycation end products (8), and C-reactive protein (9). The present study, which demonstrates that LA, at concentrations similar to those found in diabetic patients (29), upregulates endothelial LOX-1, further supports a role for this receptor in diabetes-related ED. Induction of LOX-1 by LA appears to be exerted at the transcriptional level, as reflected by the parallel increase in LOX-1 gene and protein expression.

    Activation of the transcription factor NF-B is critical for the expression of inflammatory genes involved in ED, and evidence exists that LA activates this transcription factor and induces NF-B-dependent gene expression in endothelial cells (23,30). Consistent with these data, we found that LA enhances the binding of nuclear proteins to the NF-B consensus sequence of the LOX-1 gene promoter (14) and that inhibition of NF-B totally prevented LA-induced LOX-1 expression. On the whole, these data strongly suggest that activation of the LOX-1 gene by LA involves NF-B.

    Oxidative stress is a critical signal transduction pathway involved in NF-B activation, and several evidences suggest that LA-mediated ED involves increased cellular oxidative stress. This assumption is mainly based on results from in vitro studies showing, in LA-treated endothelial cells, an increased production of reactive oxygen species and lipid hydroperoxides (23) with significant depletion in intracellular glutathione levels (11). Additional support for this hypothesis comes from the observations that LA induces, through oxidative stress, several endothelial cell inflammatory genes in vitro (10,31,32) and increases oxidative stress in vivo (33). Like many other endothelial antigens involved in endothelial cell activation/dysfunction, regulation of LOX-1 expression is redox-sensitive (34), and molecules that induce oxidative stress, such as homocysteine and angiotensin II, enhance LOX-1 expression (34,35). Our results showing that vitamin C and NAC inhibit the stimulatory effect of LA on LOX-1 protein expression clearly identify endothelial LOX-1 as a novel oxidative stress-sensitive target for LA. In agreement with previous studies (36,37), we found that NADPH oxidase inhibitors prevented LA-induced protein expression, thus indicating that NADPH oxidase is one of the enzymatic superoxide sources activated by LA. Considering the potential key role of LOX-1 in ED, these results further support the concept that oxidative stress may constitute an important mechanism responsible for LA-mediated ED.

    Superoxide per se is a strong stimulus for the activation of PKC (38). PKC activation has been implicated in ED associated with type 2 diabetes (1), and LA has been shown to activate PKC activity in various cell types (39eC41). Considering the causal link between oxidative stress and PKC activation and the well-documented role of this kinase in NF-B activation, we elaborated a tentative model in which sequential activation of the redox-sensitive PKC and NF-B signaling pathways could mediate the regulatory effect of LA on endothelial LOX-1 gene expression. Our data, which demonstrate that calphostin C totally abolished the stimulatory effect of LA on NF-B activation and LOX-1 protein expression, support this hypothesis. Furthermore, our finding that incubation of the cells with GF109203X, a selective inhibitor of the conventional PKCs, exerts identical effects and that LA activates PKC-, -I, -II, and - further suggests a role for these isotype-specific PKC isoforms in the induction of endothelial LOX-1 by LA. These results are in agreement with previous studies showing that fatty acids activate preferentially "soluble" , , and isoenzymes of PKC (42,43) and that LA activates PKC- and - in mouse mammary epithelial cells (44) and PKC- in rat pancreatic acinar cells (41).

    Endothelial cells do express different types of scavenger receptors (SRs), including CD36, SR-A, SR-B, and LOX-1. Although the role of fatty acids on vascular cell SR expression has been poorly investigated, studies have shown that polyunsaturated fatty acids and myristic acid increase the expression of hepatic SR-B1 expression (45,46) and that LA enhances CD36 expression in human monocytic cells (47). To the best of our knowledge, the present study demonstrates for the first time a role for fatty acids in vascular LOX-1 regulation. In agreement with the notion that LOX-1 mediates the uptake of oxLDL by endothelial cells (5), we found that upregulation of LOX-1 by LA is associated with significant increased uptake of oxLDL by HAECs. Taken collectively, these data suggest a new mechanism by which LA may promote ED—that of triggering the toxic effects of oxLDL on vascular endothelium through LOX-1.

    Multiple evidences have linked LA to atherosclerosis. First, it has been shown that adipose tissue levels of LA correlate with the degree of coronary artery disease (48) and with the appearance of new atherosclerotic lesions in coronary arteries (49). Second, evidence has been provided that concentrations of LA are increased in the phospholipidic fractions of human coronary arteries in cases of sudden cardiac death due to ischemic heart disease (50). Finally, a study in the Israeli population, which has one of the highest dietary polyunsaturated/saturated fat ratio in the world, concluded that diets rich in n-6 fatty acids may contribute to an increased incidence of atherosclerosis (51). Considering the potential key relationship between LA and atherogenesis, induction of endothelial LOX-1 expression by LA may represent a new mechanism by which this fatty acid may favor the development of atherosclerosis. Strategies aimed at reducing vascular LOX-1 expression could be beneficial in reducing atherosclerotic lesion development associated with human diabetes.

    ACKNOWLEDGMENTS

    This study was supported in part by a grant from the Association Diabeete Queebec. F.M. is supported by studentships from the CHUM Research Center and from the department of Nutrition, University of Montreal.

    DiI-oxLDL, 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate-labeled oxLDL; ED, endothelial dysfunction; EGM, endothelial growth culture medium; HAEC, human aortic endothelial cell; LA, linoleic acid; LOX-1, lectin-like oxidized LDL receptor-1; NAC, N-acetyl-L-cysteine; oxLDL, oxidized LDL; NF, nuclear factor; PKC, protein kinase C; SR, scavenger receptor

    REFERENCES

    Calles-Escandon J, Cipolla M: Diabetes and endothelial dysfunction: a clinical perspective. Endocr Rev22 :36 eC52,2001

    Johnstone MT, Creager SJ, Scales KM, Cusco JA, Lee BK, Creager MA: Impaired endothelium-dependent vasodilation in patients with insulin-dependent diabetes mellitus. Circulation88 :2510 eC2516,1993

    Kita T, Kume N, Minami M, Hayashida K, Murayama T, Sano H, Moriwaki H, Kataoka H, Nishi E, Horiuchi H, Arai H, Yokode M: Role of oxidized LDL in atherosclerosis. Ann N Y Acad Sci947 :199 eC205,2001

    Hennig B, Toborek M, McClain CJ: High-energy diets, fatty acids and endothelial cell function: implications for atherosclerosis. J Am Coll Nutr20 :97 eC105,2001

    Sawamura T, Kume N, Aoyama T, Moriwaki H, Hoshikawa H, Aiba Y, Tanaka T, Miwa S, Katsura Y, Kita T, Masaki T: An endothelial receptor for oxidized low-density lipoprotein. Nature386 :73 eC77,1997

    Aoyama T, Fujiwara H, Masaki T, Sawamura T: Induction of lectin-like oxidized LDL receptor by oxidized LDL and lysophosphatidylcholine in cultured endothelial cells. J Mol Cell Cardiol31 :2101 eC2114,1999

    Li L, Sawamura T, Renier G: Glucose enhances endothelial LOX-1 expression: role for LOX-1 in glucose-induced human monocyte adhesion to endothelium. Diabetes52 :1843 eC1850,2003

    Chen M, Nagase M, Fujita T, Narumiya S, Masaki T, Sawamura T: Diabetes enhances lectin-like oxidized LDL receptor-1 (LOX-1) expression in the vascular endothelium: possible role of LOX-1 ligand and AGE. Biochem Biophys Res Commun287 :962 eC968,2001

    Li L, Roumeliotis N, Sawamura T, Renier G: C-reactive protein enhances LOX-1 expression in human aortic endothelial cells: relevance of LOX-1 to C-reactive protein-induced endothelial dysfunction. Circ Res29 :877 eC883,2004

    Young VM, Toborek M, Yang F, McClain CJ, Hennig B: Effect of linoleic acid on endothelial cell inflammatory mediators. Metabolism47 :566 eC572,1998

    Hennig B, Meerarani P, Ramadass P, Watkins BA, Toborek M: Fatty acid-mediated activation of vascular endothelial cells. Metabolism49 :1006 eC1013,2000

    Toborek M, Lee YW, Garrido R, Kaiser S, Hennig B: Unsaturated fatty acids selectively induce an inflammatory environment in human endothelial cells. Am J Clin Nutr75 :119 eC125,2002

    Prescott J, Owens D, Collins P, Johnson A, Tomkin GH: The fatty acid distribution in low density lipoprotein in diabetes. Biochim Biophys Acta1439 :110 eC116,1999

    Nagase M, Abe J, Takahashi K, Ando J, Hirose S, Fujita T: Genomic organization and regulation of expression of the lectin-like oxidized low-density lipoprotein receptor (LOX-1) gene. J Biol Chem273 :33702 eC33707,1998

    Zilversmit DB: A proposal linking atherogenesis to the interaction of endothelial lipoprotein lipase with triglyceride-rich lipoproteins. Circ Res33 :633 eC638,1973

    Toborek M, Hennig B: The role of linoleic acid in endothelial cell gene expression: relationship to atherosclerosis. Subcell Biochem30 :415 eC436,1998

    Hennig B, Watkins BA: Linoleic acid and linolenic acid: effect on permeability properties of cultured endothelial cell monolayers. Am J Clin Nutr49 :301 eC305,1989

    Jiang WG, Bryce RP, Horrobin DF, Mansel RE: Regulation of tight junction permeability and occludin expression by polyunsaturated fatty acids. Biochem Biophys Res Commun244 :414 eC420,1998

    Denning GM, Figard PH, Kaduce TL, Spector AA: Role of triglycerides in endothelial cell arachidonic acid metabolism. J Lipid Res24 :993 eC1001,1983

    Ramasamy S, Boissonneault GA, Decker EA, Hennig B: Linoleic acid-induced endothelial cell injury: role of membrane-bound enzyme activities and lipid oxidation. J Biochem Toxicol6 :29 eC35,1991

    Hennig B, Boissonneault GA, Chow CK, Wang Y, Matulionis DH, Glauert HP: Effect of vitamin E on linoleic acid-mediated induction of peroxisomal enzymes in cultured porcine endothelial cells. J Nutr120 :331 eC337,1990

    Toborek M, Blanc EM, Kaiser S, Mattson MP, Hennig B: Linoleic acid potentiates TNF-mediated oxidative stress, disruption of calcium homeostasis, and apoptosis of cultured vascular endothelial cells. J Lipid Res38 :2155 eC2167,1997

    Hennig B, Toborek M, Joshi-Barve S, Barger SW, Barve S, Mattson MP, McClain CJ: Linoleic acid activates nuclear transcription factor-kappa B (NF-kappa B) and induces NF-kappa B-dependent transcription in cultured endothelial cells. Am J Clin Nutr63 :322 eC328,1996

    Erkelens DW: Diabetic dyslipidaemia. Eur Heart J19 (Suppl. H) :H27 eCH40,1998

    Kelley DE, Simoneau JA: Impaired free fatty acid utilization by skeletal muscle in non-insulin-dependent diabetes mellitus. J Clin Invest94 :2349 eC2356,1994

    Jones BH, Standridge MK, Claycombe KJ, Smith PJ, Moustaid-Moussa N: Glucose induces expression of stearoyl-CoA desaturase in 3T3eCL1 adipocytes. Biochem J335 :405 eC408,1998

    Sartippour MR, Renier G: Upregulation of macrophage lipoprotein lipase in patients with type 2 diabetes: role of peripheral factors. Diabetes49 :597 eC602,2000

    Michaud SE, Renier G: Direct regulatory effect of fatty acids on macrophage lipoprotein lipase: potential role of PPARs. Diabetes50 :660 eC666,2001

    Kwok CF, Shih KC, Hwu CM, Ho LT: Linoleic acid and oleic acid increase the endothelin-1 binding and action in cultured rat aortic smooth muscle cells. Metabolism49 :1386 eC1389,2000

    Toborek M, Barger SW, Mattson MP, Barve S, McClain CJ, Hennig B: Linoleic acid and TNF-alpha cross-amplify oxidative injury and dysfunction of endothelial cells. J Lipid Res37 :123 eC135,1996

    Dichtl W, Ares MP, Jonson AN, Jovinge S, Pachinger O, Giachelli CM, Hamsten A, Eriksson P, Nilsson J: Linoleic acid-stimulated vascular adhesion molecule-1 expression in endothelial cells depends on nuclear factor-kappaB activation. Metabolism51 :327 eC333,2002

    Woo LY, Joo PH, Hennig B, Toborek M: Linoleic acid induces MCP-1 gene expression in human microvascular endothelial cells through an oxidative mechanism. J Nutr Biochem12 :648 eC654,2001

    Turpeinen AM, Basu S, Mutanen M: A high linoleic acid diet increases oxidative stress in vivo and affects nitric oxide metabolism in humans. Prostaglandins Leukot Essent Fatty Acids59 :229 eC233,1998

    Nagase M, Ando K, Nagase T, Kaname S, Sawamura T, Fujita T: Redox-sensitive regulation of lox-1 gene expression in vascular endothelium. Biochem Biophys Res Commun281 :720 eC725,2001

    Li DY, Zhang YC, Philips MI, Sawamura T, Mehta JL: Upregulation of endothelial receptor for oxidized low-density lipoprotein (LOX-1) in cultured human coronary artery endothelial cells by angiotensin II type 1 receptor activation. Circ Res84 :1043 eC1049,1999

    Cui XL, Douglas JG: Arachidonic acid activates c-jun N-terminal kinase through NADPH oxidase in rabbit proximal tubular epithelial cells. Proc Natl Acad Sci U S A94 :3771 eC3776,1997

    Cury-Boaventura MF, Curi R: Regulation of reactive oxygen species (ROS) production by 18 carbon fatty acids in Jurkat and Raji cells. Clin Sci (Lond)108 :245 eC253,2005

    Nishikawa T, Edelstein D, Du XL, Yamagishi S, Matsumura T, Kaneda Y, Yorek MA, Beebe D, Oates PJ, Hammes HP, Giardino I, Brownlee M: Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature404 :787 eC790,2000

    Graber R, Sumida C, Nunez EA: Fatty acids and cell signal transduction. J Lipid Mediat Cell Signal9 :91 eC116,1994

    Lester DS: In vitro linoleic acid activation of protein kinase C. Biochim Biophys Acta1054 :297 eC303,1990

    Wooten MW, Wrenn RW: Linoleic acid is a potent activator of protein kinase C type III-alpha isoform in pancreatic acinar cells: its role in amylase secretion. Biochem Biophys Res Commun153 :67 eC73,1988

    el Touny S, Khan W, Hannun Y: Regulation of platelet protein kinase C by oleic acid: kinetic analysis of allosteric regulation and effects on autophosphorylation, phorbol ester binding, and susceptibility to inhibition. J Biol Chem265 :16437 eC16443,1990

    Khan WA, Blobe GC, Hannun YA: Activation of protein kinase C by oleic acid: determination and analysis of inhibition by detergent micelles and physiologic membranes: requirement for free oleate. J Biol Chem267 :3605 eC3612,1992

    Bandyopadhyay GK, Hwang S, Imagawa W, Nandi S: Role of polyunsaturated fatty acids as signal transducers: amplification of signals from growth factor receptors by fatty acids in mammary epithelial cells. Prostaglandins Leukot Essent Fatty Acids48 :71 eC78,1993

    Loison C, Mendy F, Serougne C, Lutton C: Dietary myristic acid modifies the HDL-cholesterol concentration and liver scavenger receptor BI expression in the hamster. Br J Nutr87 :199 eC210,2002

    Spady DK, Kearney DM, Hobbs HH: Polyunsaturated fatty acids up-regulate hepatic scavenger receptor B1 (SR-BI) expression and HDL cholesteryl ester uptake in the hamster. J Lipid Res40 :1384 eC1394,1999

    Pietsch A, Weber C, Goretzki M, Weber PC, Lorenz RL: N-3 but not N-6 fatty acids reduce the expression of the combined adhesion and scavenger receptor CD36 in human monocytic cells. Cell Biochem.Funct.13 :211 eC216,1995

    Hodgson JM, Wahlqvist ML, Boxall JA, Balazs ND: Can linoleic acid contribute to coronary artery disease Am J Clin Nutr58 :228 eC234,1993

    Blankenhorn DH, Johnson RL, Mack WJ, el Zein HA, Vailas LI: The influence of diet on the appearance of new lesions in human coronary arteries. JAMA263 :1646 eC1652,1990

    Luostarinen R, Boberg M, Saldeen T: Fatty acid composition in total phospholipids of human coronary arteries in sudden cardiac death. Atherosclerosis99 :187 eC193,1993

    Yam D, Eliraz A, Berry EM: Diet and disease: the Israeli paradox: possible dangers of a high omega-6 polyunsaturated fatty acid diet. Isr J Med Sci32 :1134 eC1143,1996(Fritz Maingrette, and Gen)