Identification of a botanical inhibitor of intestinal diacylglyceride acyltransferase 1 activity via in vitro screening and a parallel, randomized, blinded, placebo-controlled clinical trial

Rodney A Velliquette, Kerry Grann, Stephen R Missler, Jennifer Patterson, Chun Hu, Kevin W Gellenbeck, Jeffrey D Scholten, R Keith Randolph, Rodney A Velliquette, Kerry Grann, Stephen R Missler, Jennifer Patterson, Chun Hu, Kevin W Gellenbeck, Jeffrey D Scholten, R Keith Randolph

Abstract

Background: Diacylglyceride acyltransferase 1 (DGAT1) is the enzyme that adds the final fatty acid on to a diacylglyceride during triglyceride (TG) synthesis. DGAT1 plays a key role in the repackaging of dietary TG into circulating TG rich chylomicrons. A growing amount of research has indicated that an exaggerated postprandial circulating TG level is a risk indicator for cardiovascular and metabolic disorders. The aim of this research was to identify a botanical extract that inhibits intestinal DGAT1 activity and attenuates postprandial hypertriglyceridemia in overweight and obese humans.

Methods: Twenty individual phytochemicals and an internal proprietary botanical extract library were screened with a primary cell-free DGAT1 enzyme assay that contained dioleoyl glycerol and palmitoleoyl Coenzyme A as substrates plus human intestinal microsomes as the DGAT1 enzyme source. Botanical extracts with IC50 values < 100 μg/mL were evaluated in a cellular DGAT1 assay. The cellular DGAT1 assay comprised the analysis of (14)C labeled TG synthesis in cells incubated with (14)C-glycerol and 0.3 mM oleic acid. Lead botanical extracts were then evaluated in a parallel, double-blind, placebo-controlled clinical trial. Ninety healthy, overweight and obese participants were randomized to receive 2 g daily of placebo or individual botanical extracts (the investigational product) for seven days. Serum TG levels were measured before and after consuming a high fat meal (HFM) challenge (0.354 L drink/shake; 77 g fat, 25 g carbohydrate and 9 g protein) as a marker of intestinal DGAT1 enzyme activity.

Results: Phenolic acids (i.e., gallic acid) and polyphenols (i.e., cyanidin) abundantly found in nature appeared to inhibit DGAT1 enzyme activity in vitro. Four polyphenolic rich botanical extracts were identified from in vitro evaluation in both cell-free and cellular model systems: apple peel extract (APE), grape extract (GE), red raspberry leaf extract (RLE) and apricot/nectarine extract (ANE) (IC50 = 1.4, 5.6, and 10.4 and 3.4 μg/mL, respectively). In the seven day clinical trial, compared to placebo, only GE significantly reduced the baseline subtracted change in serum TG AUC following consumption of the HFM (AUC = 281 ± 37 vs. 181 ± 30 mg/dL*h, respectively; P = 0.021). Chromatographic characterization of the GE revealed a large number of closely eluting components containing proanthocyanidins, catechins, anthocyanins and their secondary metabolites that corresponded with the observed DGAT1 enzyme inhibition in the cell-free model.

Conclusion: These data suggest that a dietary GE has the potential to attenuate postprandial hypertriglyceridemia in part by the inhibition of intestinal DGAT1 enzyme activity without intolerable side effects.

Trial registration: This trial was registered with ClinicalTrials.gov NCT02333461.

Figures

Fig. 1
Fig. 1
Role of DGAT1 in the assimilation of dietary TG into circulating TG rich chylomicrons (CM). Dietary TG are first broken down into monoacylglycerides (MG) and fatty acids (FA) by a host of pancreatic lipases. MG and FA are then absorbed into the small intestinal enterocytes and repackaged into diacylglyercides (DG) by monoacylglyceride acyltransferase transferase. DGAT1 then acylates the DG into TG (yellow circles), which are incorporated into CM and secreted into the lymphatic circulation then enter the blood circulation via the thoracic duct
Fig. 2
Fig. 2
Clinical trial participant flow
Fig. 3
Fig. 3
Percent DGAT1 enzyme inhibition and IC50 values for single phytochemicals in the cell-free assay. Twenty individual phytochemicals were screened through the cell-free DGAT1 enzyme assay. Six of the twenty phytochemicals were dose dependent inhibitors of DGAT1 enzyme activity (IC50 ranged from 0.667 to 8.60 μM, compared to A-922500 = 40nM) and all were phenolic acids or polyphenols. Results are the mean of duplicates
Fig. 4
Fig. 4
Percent DGAT1 enzyme inhibition and IC50 values for the four lead botanical extracts. a APE, GE, RLE and ANE exhibited dose responsive inhibition of DGAT1 enzyme activity in the cell-free assay. The IC50 values ranged from 1.41 to 10.4 μg/mL and 17.1 ng/mL for A-922500. Results are the mean of triplicates. b The cellular DGAT1 assay was comprised of adding [14C]-glycerol to label newly synthesized TG and 0.3 mM oleic acid/BSA to stimulate DGAT1 activity. All botanical extracts and A-922500 inhibited oleic acid induced DGAT1 enzyme activity as measured by 14C label TG levels. GE was statistically more potent than APE, RLE and ANE, defined as greatest inhibition at the lowest dose (100 μg/mL) (Two-way ANOVA with Bonferroni's multiple comparisons test; P < 0.001). Result are the mean ± SEM (n = 3). Different letters, within each dose, indicate statistically significant
Fig. 5
Fig. 5
Baseline (fasting levels) subtracted change in serum TG levels following a HFM challenge after seven days of placebo or investigational product. a Baseline subtracted serum TG response over a 6 h period. GE significantly reduced the serum TG levels at 2 h (p = 0.014) and 4 h (p = (0.029) after the HFM compared to placebo. b AUC for the baseline subtracted serum TG was also significantly reduced by seven days of dietary GE (p = 0.021). Fasting serum TG = 95.8 ± 8.8; 94.7 ± 6.8; 90.3 ± 6.2; 102.1 ± 13.8; 103.8 ± 9.8 mg/dL, for placebo, APE, GE, RLE and ANE, respectively. Results are mean ± SEM. Comparisons between placebo and investigational product test groups were made using unpaired t-test (*P < 0.05)
Fig. 6
Fig. 6
Cell-free DGAT1 inhibition from chromatographic fractionation of GE with overlay of the corresponding LC-MS and LC-UV chromatograms. a The LC-MS chromatogram was generated using base peak index utility to differentiate major compounds. b The LC-UV chromatogram represents the sum of all wavelengths to better show the complex mixture and abundance of phytochemicals in the GE extract. DGAT1 enzyme inhibition (right side x-axis) correlates with the LC-UV pattern and abundance (left side x-axis), showing that multiple phytochemicals in the GE extract are responsible for the observed inhibition. Identities of numbered peaks are given in Table 5

References

    1. Swinburn BA, Sacks G, Hall KD, McPherson K, Finegood DT, Moodie ML, Gortmaker SL. The global obesity pandemic: shaped by global drivers and local environments. Lancet. 2011;378:804–14. doi: 10.1016/S0140-6736(11)60813-1.
    1. Chun OK, Chung SJ, Song WO. Estimated dietary flavonoid intake and major food sources of U.S. adults. J Nutr. 2007;137:1244–52.
    1. Miller M, Stone NJ, Ballantyne C, Bittner V, Criqui MH, Ginsberg HN, Goldberg AC, Howard WJ, Jacobson MS, Kris-Etherton PM, et al. Triglycerides and cardiovascular disease: a scientific statement from the American Heart Association. Circulation. 2011;123:2292–333. doi: 10.1161/CIR.0b013e3182160726.
    1. Boren J, Matikainen N, Adiels M, Taskinen MR. Postprandial hypertriglyceridemia as a coronary risk factor. Clin Chim Acta. 2014;431:131–42. doi: 10.1016/j.cca.2014.01.015.
    1. Katsanos CS. Clinical considerations and mechanistic determinants of postprandial lipemia in older adults. Adv Nutr. 2014;5:226–34. doi: 10.3945/an.113.004903.
    1. Chan DC, Pang J, Romic G, Watts GF. Postprandial hypertriglyceridemia and cardiovascular disease: current and future therapies. Curr Atheroscler Rep. 2013;15:309. doi: 10.1007/s11883-013-0309-9.
    1. Zilversmit DB. Atherogenic nature of triglycerides, postprandial lipidemia, and triglyceride-rich remnant lipoproteins. Clin Chem. 1995;41:153–8.
    1. Zilversmit DB. Atherogenesis: a postprandial phenomenon. Circulation. 1979;60:473–85. doi: 10.1161/01.CIR.60.3.473.
    1. Bansal S, Buring JE, Rifai N, Mora S, Sacks FM, Ridker PM. Fasting compared with nonfasting triglycerides and risk of cardiovascular events in women. JAMA. 2007;298:309–16. doi: 10.1001/jama.298.3.309.
    1. Cohn JS. Are we ready for a prospective study to investigate the role of chylomicrons in cardiovascular disease? Atheroscler Suppl. 2008;9:15–8. doi: 10.1016/j.atherosclerosissup.2008.05.003.
    1. Redgrave TG. Chylomicrons in disease-future challenges Invited keynote address. Atheroscler Suppl. 2008;9:3–6. doi: 10.1016/j.atherosclerosissup.2008.05.002.
    1. DeVita RJ, Pinto S. Current status of the research and development of diacylglycerol O-acyltransferase 1 (DGAT1) inhibitors. J Med Chem. 2013;56:9820–5. doi: 10.1021/jm4007033.
    1. Nordestgaard BG, Benn M, Schnohr P, Tybjaerg-Hansen A. Nonfasting triglycerides and risk of myocardial infarction, ischemic heart disease, and death in men and women. JAMA. 2007;298:299–308. doi: 10.1001/jama.298.3.299.
    1. Sasase T, Morinaga H, Yamamoto H, Ogawa N, Matsui K, Miyajima K, Kawai T, Mera Y, Masuyama T, Shinohara M, et al. Increased fat absorption and impaired fat clearance cause postprandial hypertriglyceridemia in Spontaneously Diabetic Torii rat. Diabetes Res Clin Pract. 2007;78:8–15. doi: 10.1016/j.diabres.2007.02.020.
    1. Ceriello A, Quagliaro L, Piconi L, Assaloni R, Da Ros R, Maier A, Esposito K, Giugliano D. Effect of postprandial hypertriglyceridemia and hyperglycemia on circulating adhesion molecules and oxidative stress generation and the possible role of simvastatin treatment. Diabetes. 2004;53:701–10. doi: 10.2337/diabetes.53.3.701.
    1. Bae JH, Schwemmer M, Lee IK, Lee HJ, Park KR, Kim KY, Bassenge E. Postprandial hypertriglyceridemia-induced endothelial dysfunction in healthy subjects is independent of lipid oxidation. Int J Cardiol. 2003;87:259–67. doi: 10.1016/S0167-5273(02)00347-9.
    1. Bae JH, Bassenge E, Kim KB, Kim YN, Kim KS, Lee HJ, Moon KC, Lee MS, Park KY, Schwemmer M. Postprandial hypertriglyceridemia impairs endothelial function by enhanced oxidant stress. Atherosclerosis. 2001;155:517–23. doi: 10.1016/S0021-9150(00)00601-8.
    1. Teno S, Uto Y, Nagashima H, Endoh Y, Iwamoto Y, Omori Y, Takizawa T. Association of postprandial hypertriglyceridemia and carotid intima-media thickness in patients with type 2 diabetes. Diabetes Care. 2000;23:1401–6. doi: 10.2337/diacare.23.9.1401.
    1. Mitsuguchi Y, Ito T, Ohwada K. Pathologic findings in rabbit models of hereditary hypertriglyceridemia and hereditary postprandial hypertriglyceridemia. Comp Med. 2008;58:465–80.
    1. Botham KM, Wheeler-Jones CP. Postprandial lipoproteins and the molecular regulation of vascular homeostasis. Prog Lipid Res. 2013;52:446–64. doi: 10.1016/j.plipres.2013.06.001.
    1. Schwander F, Kopf-Bolanz KA, Buri C, Portmann R, Egger L, Chollet M, McTernan PG, Piya MK, Gijs MA, Vionnet N, et al. A Dose–response Strategy Reveals Differences between Normal-Weight and Obese Men in Their Metabolic and Inflammatory Responses to a High-Fat Meal. J Nutr. 2014;144:1517–23. doi: 10.3945/jn.114.193565.
    1. Yen CL, Stone SJ, Koliwad S, Harris C, Farese RV., Jr Thematic review series: glycerolipids. DGAT enzymes and triacylglycerol biosynthesis. J Lipid Res. 2008;49:2283–301. doi: 10.1194/jlr.R800018-JLR200.
    1. Smith SJ, Cases S, Jensen DR, Chen HC, Sande E, Tow B, Sanan DA, Raber J, Eckel RH, Farese RV., Jr Obesity resistance and multiple mechanisms of triglyceride synthesis in mice lacking Dgat. Nat Genet. 2000;25:87–90. doi: 10.1038/75651.
    1. Ables GP, Yang KJ, Vogel S, Hernandez-Ono A, Yu S, Yuen JJ, Birtles S, Buckett LK, Turnbull AV, Goldberg IJ, et al. Intestinal DGAT1 deficiency reduces postprandial triglyceride and retinyl ester excursions by inhibiting chylomicron secretion and delaying gastric emptying. J Lipid Res. 2012;53:2364–79. doi: 10.1194/jlr.M029041.
    1. Lee B, Fast AM, Zhu J, Cheng JX, Buhman KK. Intestine-specific expression of acyl CoA:diacylglycerol acyltransferase 1 reverses resistance to diet-induced hepatic steatosis and obesity in Dgat1−/− mice. J Lipid Res. 2010;51:1770–80. doi: 10.1194/jlr.M002311.
    1. Birch AM, Buckett LK, Turnbull AV. DGAT1 inhibitors as anti-obesity and anti-diabetic agents. Curr Opin Drug Discov Devel. 2010;13:489–96.
    1. Yeh VS, Beno DW, Brodjian S, Brune ME, Cullen SC, Dayton BD, Dhaon MK, Falls HD, Gao J, Grihalde N, et al. Identification and preliminary characterization of a potent, safe, and orally efficacious inhibitor of acyl-CoA:diacylglycerol acyltransferase 1. J Med Chem. 2012;55:1751–7. doi: 10.1021/jm201524g.
    1. Cheng D, Iqbal J, Devenny J, Chu CH, Chen L, Dong J, Seethala R, Keim WJ, Azzara AV, Lawrence RM, et al. Acylation of acylglycerols by acyl coenzyme A:diacylglycerol acyltransferase 1 (DGAT1). Functional importance of DGAT1 in the intestinal fat absorption. J Biol Chem. 2008;283:29802–11. doi: 10.1074/jbc.M800494200.
    1. Schober G, Arnold M, Birtles S, Buckett LK, Pacheco-Lopez G, Turnbull AV, Langhans W, Mansouri A. Diacylglycerol acyltransferase-1 inhibition enhances intestinal fatty acid oxidation and reduces energy intake in rats. J Lipid Res. 2013;54:1369–84. doi: 10.1194/jlr.M035154.
    1. Langhans W, Leitner C, Arnold M. Dietary fat sensing via fatty acid oxidation in enterocytes: possible role in the control of eating. Am J Physiol Regul Integr Comp Physiol. 2011;300:R554–65. doi: 10.1152/ajpregu.00610.2010.
    1. Lin HV, Chen D, Shen Z, Zhu L, Ouyang X, Vongs A, Kan Y, Levorse JM, Kowalik EJ, Jr, Szeto DM, et al. Diacylglycerol acyltransferase-1 (DGAT1) inhibition perturbs postprandial gut hormone release. PLoS One. 2013;8:e54480. doi: 10.1371/journal.pone.0054480.
    1. Kolovou GD, Mikhailidis DP, Kovar J, Lairon D, Nordestgaard BG, Ooi TC, Perez-Martinez P, Bilianou H, Anagnostopoulou K, Panotopoulos G. Assessment and clinical relevance of non-fasting and postprandial triglycerides: an expert panel statement. Curr Vasc Pharmacol. 2011;9:258–70. doi: 10.2174/157016111795495549.
    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.
    1. Qi J, Lang W, Giardino E, Caldwell GW, Smith C, Minor LK, Darrow AL, Willemsens G, Dewaepenaert K, Roevens P, et al. High-content assays for evaluating cellular and hepatic diacylglycerol acyltransferase activity. J Lipid Res. 2010;51:3559–67. doi: 10.1194/jlr.D008029.
    1. Maciejewski BS, LaPerle JL, Chen D, Ghosh A, Zavadoski WJ, McDonald TS, Manion TB, Mather D, Patterson TA, Hanna M, et al. Pharmacological inhibition to examine the role of DGAT1 in dietary lipid absorption in rodents and humans. Am J Physiol Gastrointest Liver Physiol. 2013;304:G958–69. doi: 10.1152/ajpgi.00384.2012.
    1. Denison H, Nilsson C, Lofgren L, Himmelmann A, Martensson G, Knutsson M, Al-Shurbaji A, Tornqvist H, Eriksson JW. Diacylglycerol acyltransferase 1 inhibition with AZD7687 alters lipid handling and hormone secretion in the gut with intolerable side effects: a randomized clinical trial. Diabetes Obes Metab. 2014;16:334–43. doi: 10.1111/dom.12221.
    1. Denison H, Nilsson C, Kujacic M, Lofgren L, Karlsson C, Knutsson M, Eriksson JW. Proof of mechanism for the DGAT1 inhibitor AZD7687: results from a first-time-in-human single-dose study. Diabetes Obes Metab. 2013;15:136–43. doi: 10.1111/dom.12002.
    1. Cao J, Zhou Y, Peng H, Huang X, Stahler S, Suri V, Qadri A, Gareski T, Jones J, Hahm S, et al. Targeting Acyl-CoA:diacylglycerol acyltransferase 1 (DGAT1) with small molecule inhibitors for the treatment of metabolic diseases. J Biol Chem. 2011;286:41838–51. doi: 10.1074/jbc.M111.245456.
    1. Hiramine Y, Tanabe T. Characterization of acyl-coenzyme A:diacylglycerol acyltransferase (DGAT) enzyme of human small intestine. J Physiol Biochem. 2011;67:259–64. doi: 10.1007/s13105-010-0071-1.
    1. Serrano-Wu MH, Coppola GM, Gong Y, Neubert AD, Chatelain R, Clairmont KB, Commerford R, Cosker T, Daniels T, Hou Y, et al. Intestinally Targeted Diacylglycerol Acyltransferase 1 (DGAT1) Inhibitors Robustly Suppress Postprandial Triglycerides. ACS Med Chem Lett. 2012;3:411–5. doi: 10.1021/ml3000512.
    1. King AJ, Segreti JA, Larson KJ, Souers AJ, Kym PR, Reilly RM, Collins CA, Voorbach MJ, Zhao G, Mittelstadt SW, Cox BF. In vivo efficacy of acyl CoA: diacylglycerol acyltransferase (DGAT) 1 inhibition in rodent models of postprandial hyperlipidemia. Eur J Pharmacol. 2010;637:155–61. doi: 10.1016/j.ejphar.2010.03.056.
    1. Yang J, Xiao YY. Grape phytochemicals and associated health benefits. Crit Rev Food Sci Nutr. 2013;53:1202–25. doi: 10.1080/10408398.2012.692408.
    1. Dohadwala MM, Vita JA. Grapes and cardiovascular disease. J Nutr. 2009;139:1788S–93. doi: 10.3945/jn.109.107474.
    1. Joshi SS, Kuszynski CA, Bagchi D. The cellular and molecular basis of health benefits of grape seed proanthocyanidin extract. Curr Pharm Biotechnol. 2001;2:187–200. doi: 10.2174/1389201013378725.
    1. Lefevre M, Wiles JE, Zhang X, Howard LR, Gupta S, Smith AA, Ju ZY, DeLany JP. Gene expression microarray analysis of the effects of grape anthocyanins in mice: a test of a hypothesis-generating paradigm. Metabolism. 2008;57:S52–7. doi: 10.1016/j.metabol.2008.03.005.
    1. Georgiev V, Ananga A, Tsolova V. Recent advances and uses of grape flavonoids as nutraceuticals. Nutrients. 2014;6:391–415. doi: 10.3390/nu6010391.
    1. Xia EQ, Deng GF, Guo YJ, Li HB. Biological activities of polyphenols from grapes. Int J Mol Sci. 2010;11:622–46. doi: 10.3390/ijms11020622.
    1. Yadav M, Jain S, Bhardwaj A, Nagpal R, Puniya M, Tomar R, Singh V, Parkash O, Prasad GB, Marotta F, Yadav H. Biological and medicinal properties of grapes and their bioactive constituents: an update. J Med Food. 2009;12:473–84. doi: 10.1089/jmf.2008.0096.
    1. Chuang CC, McIntosh MK. Potential mechanisms by which polyphenol-rich grapes prevent obesity-mediated inflammation and metabolic diseases. Annu Rev Nutr. 2011;31:155–76. doi: 10.1146/annurev-nutr-072610-145149.
    1. Ali K, Maltese F, Choi Y, Verpoorte R. Metabolic constituents of grapevine and grape-derived products. Phytochemistry Reviews. 2010;9:357–78. doi: 10.1007/s11101-009-9158-0.
    1. Klein A, Wrulich OA, Jenny M, Gruber P, Becker K, Fuchs D, Gostner JM, Uberall F. Pathway-focused bioassays and transcriptome analysis contribute to a better activity monitoring of complex herbal remedies. BMC Genomics. 2013;14:133. doi: 10.1186/1471-2164-14-133.
    1. Stevenson DE, Hurst RD. Polyphenolic phytochemicals--just antioxidants or much more? Cell Mol Life Sci. 2007;64:2900–16. doi: 10.1007/s00018-007-7237-1.
    1. Milella RA, Antonacci D, Crupi P, Incampo F, Carrieri C, Semeraro N, Colucci M. Skin extracts from 2 Italian table grapes (Italia and Palieri) inhibit tissue factor expression by human blood mononuclear cells. J Food Sci. 2012;77:H154–9. doi: 10.1111/j.1750-3841.2012.02818.x.
    1. Forester SC, Waterhouse AL. Identification of Cabernet Sauvignon anthocyanin gut microflora metabolites. J Agric Food Chem. 2008;56:9299–304. doi: 10.1021/jf801309n.
    1. Cardona F, Andres-Lacueva C, Tulipani S, Tinahones FJ, Queipo-Ortuno MI. Benefits of polyphenols on gut microbiota and implications in human health. J Nutr Biochem. 2013;24:1415–22. doi: 10.1016/j.jnutbio.2013.05.001.
    1. Hidalgo M, Oruna-Concha MJ, Kolida S, Walton GE, Kallithraka S, Spencer JP, de Pascual-Teresa S. Metabolism of anthocyanins by human gut microflora and their influence on gut bacterial growth. J Agric Food Chem. 2012;60:3882–90. doi: 10.1021/jf3002153.
    1. Cases S, Stone SJ, Zhou P, Yen E, Tow B, Lardizabal KD, Voelker T, Farese RV., Jr Cloning of DGAT2, a second mammalian diacylglycerol acyltransferase, and related family members. J Biol Chem. 2001;276:38870–6. doi: 10.1074/jbc.M106219200.
    1. Haas JT, Winter HS, Lim E, Kirby A, Blumenstiel B, DeFelice M, Gabriel S, Jalas C, Branski D, Grueter CA, et al. DGAT1 mutation is linked to a congenital diarrheal disorder. J Clin Invest. 2012;122:4680–4. doi: 10.1172/JCI64873.
    1. Little TJ, Horowitz M, Feinle-Bisset C. Modulation by high-fat diets of gastrointestinal function and hormones associated with the regulation of energy intake: implications for the pathophysiology of obesity. Am J Clin Nutr. 2007;86:531–41.

Source: PubMed

3
Sottoscrivi