Walnut oil increases cholesterol efflux through inhibition of stearoyl CoA desaturase 1 in THP-1 macrophage-derived foam cells

Jun Zhang, Jessica A Grieger, Penny M Kris-Etherton, Jerry T Thompson, Peter J Gillies, Jennifer A Fleming, John P Vanden Heuvel, Jun Zhang, Jessica A Grieger, Penny M Kris-Etherton, Jerry T Thompson, Peter J Gillies, Jennifer A Fleming, John P Vanden Heuvel

Abstract

Background: Walnuts significantly decrease total and low-density lipoprotein cholesterol in normo- and hypercholesterolemic individuals. No study to date has evaluated the effects of walnuts on cholesterol efflux, the initial step in reverse cholesterol transport, in macrophage-derived foam cells (MDFC). The present study was conducted to investigate the mechanisms by which walnut oil affects cholesterol efflux.

Methods: The extract of English walnuts (walnut oil) was dissolved in DMSO and applied to cultured THP-1 MDFC cells (0.5 mg/mL). THP-1 MDFC also were treated with human sera (10%, v:v) taken from subjects in a walnut feeding study. Cholesterol efflux was examined by liquid scintillation counting. Changes in gene expression were quantified by real time PCR.

Results: Walnut oil treatment significantly increased cholesterol efflux through decreasing the expression of the lipogenic enzyme stearoyl CoA desaturase 1 (SCD1) in MDFC. Alpha-linolenic acid (ALA), the major n-3 polyunsaturated fatty acids found in walnuts, recaptured SCD1 reduction in MDFC, a mechanism mediated through activation of nuclear receptor farnesoid-X-receptor (FXR). Postprandial serum treatment also increased cholesterol efflux in MDFC. When categorized by baseline C-reactive protein (CRP; cut point of 2 mg/L), subjects in the lower CRP sub-group benefited more from dietary intervention, including a more increase in cholesterol efflux, a greater reduction in SCD1, and a blunted postprandial lipemia.

Conclusion: In conclusion, walnut oil contains bioactive molecules that significantly improve cholesterol efflux in MDFC. However, the beneficial effects of walnut intake may be reduced by the presence of a pro-inflammatory state.

Trial registration: ClinicalTrials.gov: NCT00938340.

Figures

Figure 1
Figure 1
Walnut oil affects cholesterol transport and storage in THP-1 macrophage-derived foam cells (MDFC). (A) Walnut oil increases cholesterol efflux. DMSO treated cells have 2.3% cholesterol efflux (of total 3H-Ch). Values of DMSO treated cells are normalized and expressed as 100%. (B) Expression change of genes related with cholesterol transport and storage following walnut oil treatment. Full names of all abbreviation are as described in Table S1 footnote. (C) Walnut oil affects gene expression related with lipogenesis and fatty acid oxidation. (D) Walnut oil treatment decreases SCD1 protein. Band intensities were determined by OptiQuant Image analysis software (Packard Instrument Co., Meriden, CT). (E) Effect of walnut oil on ABCA1 and ABCG1 protein levels. (F) Effect of SCD1 on cholesterol efflux. SCD1 overespression is shown by western blot. A total of 60 μg proteins were loaded on each lane of control (empty plasmid) and overexpression groups. Bars not sharing common letters (a, b or c) differ with P < 0.05. (G) Fatty acids affect SCD1 expression. Fatty acids are conjugated to BSA with a molar ratio of 4:1 modified from method described by Calder et al. [50]. PA: palmitic acid; OA: oleic acid; LA: linoleic acid; ALA: alpha linolenic acid. (H) Fatty acids affect cholesterol efflux. Symbol * in all figure panels indicates a significant difference from respective control with P < 0.05. The data presented are means ± SEM of triplicate wells of two independent experiments.
Figure 2
Figure 2
Walnut oil inhibits SCD1 through a FXR pathway in THP-1 MDFC. (A) Activation of walnut oil on nuclear receptor ligand binding domain of Dual Luciferase Reporter Assay. PPAR: peroxisome proliferator-activated receptor; LXR: liver-X-receptor; RXR: retinoic-X-receptor; PXR: pregnane-X-receptor; FXR: farnesoid-X-receptor. (B) SCD1 protein (top) and mRNA (bottom) changes following nuclear receptor agonists treatment. SCD1 protein change was detected by Western blot. Treatments are as follows: Lane 1: DMSO control; Lane 2: ciprofibrate 100 μM (PPAR-α agonist); Lane 3: GW501506 500 nM (PPAR-β agonist); Lane 4: rosiglitazone 10 μM (PPAR-γ agonist); Lane 5: TO901317 5 μM (LXR agonist); Lane 6: 9-cis RA 100 nM (RXR agonist); Lane 7: rifamipicin 25 μM (PXR agonist); Lane 8: GW4064 10 μM (FXR agonist). (C) FXR activation increases cholesterol efflux. (D) FXR activation decreases SCD1 expression. THP-1 MDFCs were treated with 10 μM GW4064 (FXR agonist), walnut oil (0.5 mg/mL), 100 μM ALA in the presence or absence of 50 μM GGS (FXR antagonist). Bars not sharing common letters are significantly different. (E) Walnut oil and ALA activate FXR and increase its target gene SHP expression. (F) Overexpression of SHP inhibits SCD1 expression in MDFC. THP-1 derived macrophages were transfected with pCMX-hSHP and empty plasmid (control) for 24 h. Symbol * in all figure panels indicates a significant difference from respective control (P < 0.05). The data presented are means ± SEM of triplicate wells of two independent experiments.
Figure 3
Figure 3
Human serum affects SCD1 expression and cholesterol efflux in THP-1 MDFC. Human serum affects SCD1 expression in THP-1 MDFCs. Samples were taken from 15 subjects consuming different walnut components: whole walnut, walnut oil, defatted walnut meat, and walnut skin. Baseline serum as well as postprandial serum of 1, 2, 4, 6 h were applied as treatment (10%, v:v) for 24 h in serum free media. Real time PCR value of individual serum treated sample is divided by a PCR value of a pooled serum treated sample to minimize the variation between PCR runs and to exclude a non-specific serum effect on gene expression. Value of each postprandial serum treated sample is adjusted by baseline value, which served as a control within the same dietary period. Data were logarithm transformed for analysis. Sera of fifteen participants at baseline and 1, 2, 4, and 6 h postprandially were applied as treatment. N = 75 of each dietary group.
Figure 4
Figure 4
Categorized by baseline CRP level (2 mg/L as a cutting point), subjects have different serum lipid response following walnut oil intake. (A) Changes in CRP between low- and high-CRP subgroups. CRP values were expressed as mean (mg/L) ± SEM. Subjects were categorized as low (8 subjects) or high (7 subjects) CRP sub-groups based on the mean of baseline CRP values (P < 0.001 between sub-groups). (B) Changes in TG between low- and high-CRP subgroups (P < 0.01 between sub-groups). (C) Changes inTC between low- and high-CRP subgroups. (D) Changes in LDL-C between low- and high-CRP subgroups. (E) Changes in HDL-C between low- and high-CRP subgroups. Lipids and lipoproteins were expressed as mean (mg/dL) ± SEM. Open square with dashed line represents the high CRP sub-group. Solid circle with straight line represents the low CRP sub-group.
Figure 5
Figure 5
Human sera of low and high CRP sub-groups differentially affect cholesterol efflux. (A) Sera of subjects consuming walnut oil affect cholesterol efflux. Following the induction of foam cells, human sera (10%, v:v) of baseline and 4 h postprandially were applied as treatment to induce cholesterol efflux for 24 h. Following efflux period, control cells (incubated in serum-free culture medium only) had a cholesterol efflux of 5.4%. The value is normalized and expressed as 100%. (B) Categorized by baseline CRP (cut point of 2 mg/L), changes of cholesterol efflux following treatment by human sera of walnut oil group. Experiment conditions were as described in Figure 3B. Symbol * indicates a significant difference between control and treatment groups (P < 0.05). Results are representative of two independent experiments with a duplicate at each time point (baseline and 4 h postprandially) of each subject.
Figure 6
Figure 6
Human sera of low and high CRP sub-groups differentially affect gene expression. (A) Changes in SCD1 mRNA expression following treatment of human sera from subjects consuming walnut oil. * indicates significant difference from baseline and between CRP sub-groups (P < 0.01). (B) Quantification of SCD1 protein. Value of baseline sera treated cells of low CRP sub-group was normalized as 100. Bars not sharing the same letters differ (P < 0.05). (C) Changes in membrane transporters ABCA1 mRNA following human sera treatment. (D) Changes in ABCA1 proteins categorized by baseline CRP level. In the GLM model, age, gender, BMI, and glucose are served as covariates and ID number as dummy variable.

References

    1. Griel AE, Kris-Etherton PM. Tree nuts and the lipid profile: a review of clinical studies. Br J Nutr. 2006;96(Suppl 2):S68–78.
    1. Banel DK, Hu FB. Effects of walnut consumption on blood lipids and other cardiovascular risk factors: a meta-analysis and systematic review. Am J Clin Nutr. 2009;90:56–63. doi: 10.3945/ajcn.2009.27457.
    1. Sampath H, Ntambi JM. Polyunsaturated fatty acid regulation of genes of lipid metabolism. Annu Rev Nutr. 2005;25:317–340. doi: 10.1146/annurev.nutr.25.051804.101917.
    1. Zhang J, Kris-Etherton PM, Thompson JT, Hannon DB, Gillies PJ, Vanden Heuvel JP. Alpha-linolenic acid increases cholesterol efflux in macrophage-derived foam cells by decreasing stearoyl CoA desaturase 1 expression: evidence for a farnesoid-X-receptor mechanism of action. J Nutr Biochem. 2011. Epub ahead of print.
    1. Ntambi JM, Miyazaki M. Recent insights into stearoyl-CoA desaturase-1. Curr Opin Lipidol. 2003;14:255–261. doi: 10.1097/00041433-200306000-00005.
    1. Dobrzyn A, Ntambi JM. Stearoyl-CoA desaturase as a new drug target for obesity treatment. Obes Rev. 2005;6:169–174. doi: 10.1111/j.1467-789X.2005.00177.x.
    1. Popeijus HE, Saris WH, Mensink RP. Role of stearoyl-CoA desaturases in obesity and the metabolic syndrome. Int J Obes (Lond) 2008;32:1076–1082. doi: 10.1038/ijo.2008.55.
    1. Sun Y, Hao M, Luo Y, Liang CP, Silver DL, Cheng C, Maxfield FR, Tall AR. Stearoyl-CoA desaturase inhibits ATP-binding cassette transporter A1-mediated cholesterol efflux and modulates membrane domain structure. J Biol Chem. 2003;278:5813–5820. doi: 10.1074/jbc.M208687200.
    1. MacDonald ML, Singaraja RR, Bissada N, Ruddle P, Watts R, Karasinska JM, Gibson WT, Fievet C, Vance JE, Staels B, Hayden MR. Absence of stearoyl-CoA desaturase-1 ameliorates features of the metabolic syndrome in LDLR-deficient mice. J Lipid Res. 2008;49:217–229.
    1. Savransky V, Jun J, Li J, Nanayakkara A, Fonti S, Moser AB, Steele KE, Schweitzer MA, Patil SP, Bhanot S. et al.Dyslipidemia and atherosclerosis induced by chronic intermittent hypoxia are attenuated by deficiency of stearoyl coenzyme A desaturase. Circ Res. 2008;103:1173–1180. doi: 10.1161/CIRCRESAHA.108.178533.
    1. Brown JM, Chung S, Sawyer JK, Degirolamo C, Alger HM, Nguyen TM, Zhu X, Duong MN, Brown AL, Lord C. et al.Combined therapy of dietary fish oil and stearoyl-CoA desaturase 1 inhibition prevents the metabolic syndrome and atherosclerosis. Arterioscler Thromb Vasc Biol. 2010;30:24–30. doi: 10.1161/ATVBAHA.109.198036.
    1. Brown JM, Chung S, Sawyer JK, Degirolamo C, Alger HM, Nguyen T, Zhu X, Duong MN, Wibley AL, Shah R. et al.Inhibition of stearoyl-coenzyme A desaturase 1 dissociates insulin resistance and obesity from atherosclerosis. Circulation. 2008;118:1467–1475. doi: 10.1161/CIRCULATIONAHA.108.793182.
    1. MacDonald ML, van Eck M, Hildebrand RB, Wong BW, Bissada N, Ruddle P, Kontush A, Hussein H, Pouladi MA, Chapman MJ. et al.Despite antiatherogenic metabolic characteristics, SCD1-deficient mice have increased inflammation and atherosclerosis. Arterioscler Thromb Vasc Biol. 2009;29:341–347. doi: 10.1161/ATVBAHA.108.181099.
    1. Meyer MD, Terry LA. Development of a rapid method for the sequential extraction and subsequent quantification of fatty acids and sugars from avocado mesocarp tissue. J Agric Food Chem. 2008;56:7439–7445. doi: 10.1021/jf8011322.
    1. Bligh EG, Dyer WJ. A rapid method of total lipid extraction and purification. Can J Biochem Physiol. 1959;37:911–917. doi: 10.1139/o59-099.
    1. Metcalfe LD, Schmitz AA, Pelka JR. Rapid preparation of fatty acid esters from lipids for gas chromatographic analysis. Anal Chem. 1966;38:514–515. doi: 10.1021/ac60235a044.
    1. Zhang J, Cai S, Peterson BR, Kris-Etherton PM, Heuvel JP. Development of a cell-based, high-throughput screening assay for cholesterol efflux using a fluorescent mimic of cholesterol. Assay Drug Dev Technol. 2010;9:136–146.
    1. Zhang J, Kris-Etherton PM, Thompson JT, Vanden Heuvel JP. Effect of pistachio oil on gene expression of IFN-induced protein with tetratricopeptide repeats 2: a biomarker of inflammatory response. Mol Nutr Food Res. 2010;54(Suppl 1):S83–92.
    1. Heinemann FS, Ozols J. Degradation of stearoyl-coenzyme A desaturase: endoproteolytic cleavage by an integral membrane protease. Mol Biol Cell. 1998;9:3445–3453.
    1. Vanden Heuvel JP, Thompson JT, Frame SR, Gillies PJ. Differential activation of nuclear receptors by perfluorinated fatty acid analogs and natural fatty acids: a comparison of human, mouse, and rat peroxisome proliferator-activated receptor-alpha, -beta, and -gamma, liver X receptor-beta, and retinoid X receptor-alpha. Toxicol Sci. 2006;92:476–489. doi: 10.1093/toxsci/kfl014.
    1. Alberti KG, Zimmet P, Shaw J. The metabolic syndrome--a new worldwide definition. Lancet. 2005;366:1059–1062. doi: 10.1016/S0140-6736(05)67402-8.
    1. Grundy SM, Cleeman JI, Daniels SR, Donato KA, Eckel RH, Franklin BA, Gordon DJ, Krauss RM, Savage PJ, Smith SC Jr. et al.Diagnosis and management of the metabolic syndrome: an American Heart Association/National Heart, Lung, and Blood Institute Scientific Statement. Circulation. 2005;112:2735–2752. doi: 10.1161/CIRCULATIONAHA.105.169404.
    1. Rader DJ. Molecular regulation of HDL metabolism and function: implications for novel therapies. J Clin Invest. 2006;116:3090–3100. doi: 10.1172/JCI30163.
    1. Tall AR. Cholesterol efflux pathways and other potential mechanisms involved in the athero-protective effect of high density lipoproteins. J Intern Med. 2008;263:256–273. doi: 10.1111/j.1365-2796.2007.01898.x.
    1. Pal S, Davis PJ. N-3 polyunsaturated fatty acids enhance cholesterol efflux from human fibroblasts in culture. Biochem Biophys Res Commun. 1990;173:566–570. doi: 10.1016/S0006-291X(05)80072-5.
    1. Dusserre E, Pulcini T, Bourdillon MC, Ciavatti M, Berthezene F. Omega-3 fatty acids in smooth muscle cell phospholipids increase membrane cholesterol efflux. Lipids. 1995;30:35–41. doi: 10.1007/BF02537039.
    1. Marmillot P, Rao MN, Liu QH, Chirtel SJ, Lakshman MR. Effect of dietary omega-3 fatty acids and chronic ethanol consumption on reverse cholesterol transport in rats. Metabolism. 2000;49:508–512. doi: 10.1016/S0026-0495(00)80017-7.
    1. Mathur SN, Watt KR, Field FJ. Regulation of intestinal NPC1L1 expression by dietary fish oil and docosahexaenoic acid. J Lipid Res. 2007;48:395–404.
    1. Knapp HR, Hullin F, Salem N Jr. Asymmetric incorporation of dietary n-3 fatty acids into membrane aminophospholipids of human erythrocytes. J Lipid Res. 1994;35:1283–1291.
    1. Spector AA, Yorek MA. Membrane lipid composition and cellular function. J Lipid Res. 1985;26:1015–1035.
    1. Wassall SR, Brzustowicz MR, Shaikh SR, Cherezov V, Caffrey M, Stillwell W. Order from disorder, corralling cholesterol with chaotic lipids. The role of polyunsaturated lipids in membrane raft formation. Chem Phys Lipids. 2004;132:79–88.
    1. Sweet WD, Schroeder F. Polyunsaturated fatty acids alter sterol transbilayer domains in LM fibroblast plasma membrane. FEBS Lett. 1988;229:188–192. doi: 10.1016/0014-5793(88)80824-X.
    1. Kilsdonk EP, Dorsman AN, van Gent T, van Tol A. Effect of phospholipid fatty acid composition of endothelial cells on cholesterol efflux rates. J Lipid Res. 1992;33:1373–1382.
    1. Wang Y, Oram JF. Unsaturated fatty acids inhibit cholesterol efflux from macrophages by increasing degradation of ATP-binding cassette transporter A1. J Biol Chem. 2002;277:5692–5697. doi: 10.1074/jbc.M109977200.
    1. Uehara Y, Engel T, Li Z, Goepfert C, Rust S, Zhou X, Langer C, Schachtrup C, Wiekowski J, Lorkowski S. et al.Polyunsaturated fatty acids and acetoacetate downregulate the expression of the ATP-binding cassette transporter A1. Diabetes. 2002;51:2922–2928. doi: 10.2337/diabetes.51.10.2922.
    1. Murthy S, Born E, Mathur SN, Field FJ. Liver-X-receptor-mediated increase in ATP-binding cassette transporter A1 expression is attenuated by fatty acids in CaCo-2 cells: effect on cholesterol efflux to high-density lipoprotein. Biochem J. 2004;377:545–552.
    1. Wang Y, Oram JF. Unsaturated fatty acids phosphorylate and destabilize ABCA1 through a protein kinase C delta pathway. J Lipid Res. 2007;48:1062–1068. doi: 10.1194/jlr.M600437-JLR200.
    1. Wang YD, Chen WD, Moore DD, Huang W. FXR: a metabolic regulator and cell protector. Cell Res. 2008;18:1087–1095. doi: 10.1038/cr.2008.289.
    1. Guo GL, Santamarina-Fojo S, Akiyama TE, Amar MJ, Paigen BJ, Brewer B Jr, Gonzalez FJ. Effects of FXR in foam-cell formation and atherosclerosis development. Biochim Biophys Acta. 2006;1761:1401–1409.
    1. Mencarelli A, Renga B, Distrutti E, Fiorucci S. Antiatherosclerotic effect of farnesoid X receptor. Am J Physiol Heart Circ Physiol. 2009;296:H272–281.
    1. Zhang Y, Wang X, Vales C, Lee FY, Lee H, Lusis AJ, Edwards PA. FXR deficiency causes reduced atherosclerosis in Ldlr-/- mice. Arterioscler Thromb Vasc Biol. 2006;26:2316–2321. doi: 10.1161/01.ATV.0000235697.35431.05.
    1. Levine AS, Silvis SE. Absorption of whole peanuts, peanut oil, and peanut butter. N Engl J Med. 1980;303:917–918. doi: 10.1056/NEJM198010163031605.
    1. Blomhoff R, Carlsen MH, Andersen LF, Jacobs DR Jr. Health benefits of nuts: potential role of antioxidants. Br J Nutr. 2006;96(Suppl 2):S52–60.
    1. Chen CY, Milbury PE, Lapsley K, Blumberg JB. Flavonoids from almond skins are bioavailable and act synergistically with vitamins C and E to enhance hamster and human LDL resistance to oxidation. J Nutr. 2005;135:1366–1373.
    1. Wang X, Liao D, Bharadwaj U, Li M, Yao Q, Chen C. C-reactive protein inhibits cholesterol efflux from human macrophage-derived foam cells. Arterioscler Thromb Vasc Biol. 2008;28:519–526. doi: 10.1161/ATVBAHA.107.159467.
    1. Persson J, Nilsson J, Lindholm MW. Interleukin-1beta and tumour necrosis factor-alpha impede neutral lipid turnover in macrophage-derived foam cells. BMC Immunol. 2008;9:70. doi: 10.1186/1471-2172-9-70.
    1. Ma KL, Ruan XZ, Powis SH, Chen Y, Moorhead JF, Varghese Z. Inflammatory stress exacerbates lipid accumulation in hepatic cells and fatty livers of apolipoprotein E knockout mice. Hepatology. 2008;48:770–781. doi: 10.1002/hep.22423.
    1. Uryu S, Tokuhiro S, Oda T. beta-Amyloid-specific upregulation of stearoyl coenzyme A desaturase-1 in macrophages. Biochem Biophys Res Commun. 2003;303:302–305. doi: 10.1016/S0006-291X(03)00334-6.
    1. Petersson H, Lind L, Hulthe J, Elmgren A, Cederholm T, Riserus U. Relationships between serum fatty acid composition and multiple markers of inflammation and endothelial function in an elderly population. Atherosclerosis. 2009;203:298–303. doi: 10.1016/j.atherosclerosis.2008.06.020.
    1. Calder PC, Bond JA, Harvey DJ, Gordon S, Newsholme EA. Uptake and incorporation of saturated and unsaturated fatty acids into macrophage lipids and their effect upon macrophage adhesion and phagocytosis. Biochem J. 1990;269:807–814.
    1. Friedewald WT, Levy RI, Fredrickson DS. Estimation of the concentration of low-density lipoprotein cholesterol in plasma, without use of the preparative ultracentrifuge. Clin Chem. 1972;18:499–502.

Source: PubMed

3
Subscribe