Efficacy of dietary odd-chain saturated fatty acid pentadecanoic acid parallels broad associated health benefits in humans: could it be essential?

Stephanie Venn-Watson, Richard Lumpkin, Edward A Dennis, Stephanie Venn-Watson, Richard Lumpkin, Edward A Dennis

Abstract

Dietary odd-chain saturated fatty acids (OCFAs) are present in trace levels in dairy fat and some fish and plants. Higher circulating concentrations of OCFAs, pentadecanoic acid (C15:0) and heptadecanoic acid (C17:0), are associated with lower risks of cardiometabolic diseases, and higher dietary intake of OCFAs is associated with lower mortality. Population-wide circulating OCFA levels, however, have been declining over recent years. Here, we show C15:0 as an active dietary fatty acid that attenuates inflammation, anemia, dyslipidemia, and fibrosis in vivo, potentially by binding to key metabolic regulators and repairing mitochondrial function. This is the first demonstration of C15:0's direct role in attenuating multiple comorbidities using relevant physiological mechanisms at established circulating concentrations. Pairing our findings with evidence that (1) C15:0 is not readily made endogenously, (2) lower C15:0 dietary intake and blood concentrations are associated with higher mortality and a poorer physiological state, and (3) C15:0 has demonstrated activities and efficacy that parallel associated health benefits in humans, we propose C15:0 as a potential essential fatty acid. Further studies are needed to evaluate the potential impact of decades of reduced intake of OCFA-containing foods as contributors to C15:0 deficiencies and susceptibilities to chronic disease.

Conflict of interest statement

S.V. is a co-founder of and employed by Epitracker, Inc and Seraphina Therapeutics, Inc, which financed studies in this paper and hold exclusive licensing rights from the U.S. Navy to commercialize odd-chain saturated fatty acids as human and animal health products. R. Lumpkin’s work related to this study was also financed by Epitracker. E.A. Dennis is a member of the Scientific Advisory Board of Epitracker and holds equity.

Figures

Figure 1
Figure 1
Reduced mitochondrial reactive oxygen species production in stressed human cell systems supplemented with C15:0 at 10, 20 and 50 µM compared to non-supplemented controls. Endpoint of upper whisker, maximum; upper edge of box, 75th percentile; line inside box, median; diamond, mean; lower edge of box, 25th percentile; endpoint of lower whisker, minimum; circle, outlier.
Figure 2
Figure 2
Annotated dose-dependent anti-inflammatory and antifibrotic activities of C15:0 (0.74, 2.2, 6.7 and 20 µM) in primary human cell systems mimicking inflammation and fibrosis. MCP-1 = monocyte chemoattractant protein 1, sIgG = secreted immunoglobulin G, PAI-1 = plasminogen activation inhibitor 1.
Figure 3
Figure 3
Annotated dose-dependent anti-inflammatory and antifibrotic activities of saturated fatty acids (C13:0, C14:0, C15:0, and C16:0 in 20 µM) in primary human cell systems mimicking inflammation and fibrosis. MCP-1 = monocyte chemoattractant protein 1; IgG = immunoglobulin G; IL-17A, IL-17F, IL-6 = interleukin 17 A, 17 F and 6; sTNFα = secreted tumor necrosis factor alpha; VCAM-1=vascular adhesion molecule 1, CXCL10, CXLX11, CXCL9 = chemokine ligand 10, 11 and 9; PAI-1 = plasminogen activation inhibitor 1). BT System = Primary human cell system in Eurofins/DiscoverX BioMAP Diversity PLUS panel modeling T cell dependent B cell proliferation; HDF3CGF System = Primary human cell system in Eurofins/DiscoverX BioMAP Diversity PLUS panel modeling matrix/tissue remodeling in the context of Th1-type inflammation, including fibrosis and chronic inflammation.
Figure 4
Figure 4
Plasma deuterated C15:0 (a), C17:0 (b), and C13:0 (c) concentrations achieved over 24 h in male Sprague Dawley rats (n = 6 total, 3 per time point between 15 min and 12 h) dosed orally once with deuterated C15:0 (35 mg/kg body weight). Upper edge of box, maximum; line inside box, median; diamond, mean; lower edge of box, minimum.
Figure 5
Figure 5
Comparisons of circulating concentrations of the pro-inflammatory chemokine, monocyte chemoattractant 1 (MCP-1) (a), pro-inflammatory cytokine, interleukin 6 (IL-6) (b), glucose (c), and total cholesterol (d) between high fat diet-induced obese in vivo model supplemented with low dose daily oral C15:0 (pentadecanoic acid, 5 mg/kg) over 12 weeks and non-supplemented controls. Wilcoxon two-sided p values.
Figure 6
Figure 6
Comparisons of red blood cell indices, including hemoglobin (a), red blood cell counts (b), red blood cell distribution width (c), and reticulocytes (d) between high fat-diet fed in vivo models for nonalcoholic steatohepatitis (NASH) supplemented with higher doses of daily oral C15:0 (pentadecanoic acid, 35 mg/kg) over 11 weeks and non-supplemented controls. Wilcoxon two-sided p values.
Figure 7
Figure 7
Comparisons of circulating globulins (a), platelets (b), cholesterol (c), and triglycerides (d) between high fat-diet fed in vivo models for nonalcoholic steatohepatitis (NASH) supplemented with higher doses of daily oral C15:0 (pentadecanoic acid, 35 mg/kg) over 11 weeks and non-supplemented controls. Wilcoxon two-sided p values.

References

    1. Dietary goals for the United States prepared by the staff of the Select Committee on Nutrition and Human Needs, United States Senate. United States. Washington. U.S. Govt. Print. Off (1977).
    1. U.S. Department of Agriculture, Human Nutrition Information Service. Food intakes: Individuals in 48 states, year 1977-78. Nationwide food consumption survey 1977-78 Report No. I-1. NFCS 1977-78 Report No. I-1 (1983).
    1. U.S. Department of Agriculture, Agricultural Research Service. Data tables: Results from USDA’s 1994-1996 continuing survey of food intakes by individuals and 1996-96 diet and health knowledge survey. NTIS accession number: PB98-500457 (1999).
    1. Bluher M. Obesity: global epidemiology and pathogenesis. Nature Rev Endocrinol. 2019;15:288–298. doi: 10.1038/s41574-019-0176-8.
    1. Zheng Y, Ley SH, Hu FB. Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nature Rev Endocrinol. 2018;14:88–98. doi: 10.1038/nrendo.2017.151.
    1. Tapper EB, Loomba R. NAFLD, metabolic syndrome and the fight that will define clinical practice for a generation of hepatologists. Hepatol. 2018;67:1657–1659. doi: 10.1002/hep.29722.
    1. White, M. J., Armstrong, S. C., Kay, M. C., Perrin, E. M. & Skinner, A. Associations between milk fat content and obesity, 1999–2016. Pediatric Obesity e12612 (2020).
    1. Mitri J, et al. Dairy intake and type 2 diabetes risk factors: a narrative review. Diab Metab Synd Clin Res Rev. 2019;13:2879–2887.
    1. Bhupathi V, Mazariegos M, Rodriquez JBC, Doeker A. Dairy intake and risk of cardiovascular disease. Curr Cardiol Rep. 2020;22:11. doi: 10.1007/s11886-020-1263-0.
    1. Astrup A, Geiker NRW, Magkos F. Effects of full-fat and fermented dairy products on cardiometabolic disease: food is more than the sum of its parts. Adv Nutr. 2019;10:924S–930S. doi: 10.1093/advances/nmz069.
    1. Mansson HL. Fatty acids in bovine milk fat. Food Nutr Res. 2008;52:1.
    1. Rocha DM, Caldas AP, Oliveira LL, Bressan J, Hermsdorff HH. Saturated fatty acids trigger TLR4-mediated inflammatory response. Atheroscler. 2016;244:211–215. doi: 10.1016/j.atherosclerosis.2015.11.015.
    1. Zong G, et al. Intake of individual saturated fatty acids and risk of coronary heart disease in US men and women: two prospective longitudinal cohorts. BMJ. 2016;355:i5796. doi: 10.1136/bmj.i5796.
    1. Forouhi NG, et al. Differences in the prospective association between individual plasma phospholipid saturated fatty acids and incident type 2 diabetes: the EPIC-InterAct case-cohort study. Lancet Diab Endocrinol. 2014;14:70146–9.
    1. Kurotani K, et al. Even- and odd-chain saturated fatty acids in serum phospholipids are differentially associated with adipokines. PLOS ONE. 2017;12:e0178192. doi: 10.1371/journal.pone.0178192.
    1. Aglago EK, et al. Association between serum phospholipid fatty acid levels and adiposity in Mexican women. J Lipid Res. 2017;58:1462–1470. doi: 10.1194/jlr.P073643.
    1. Huang L, et al. Circulating saturated fatty acids and incident type 2 diabetes: A systematic review and meta-analysis. Nutrients. 2019;11:998. doi: 10.3390/nu11050998.
    1. Jimenez-Cepeda A, et al. Dietary intake of fatty acids and its relationship with FEV1/FVC in patients with chronic obstructive pulmonary disease. Clin Nutr. 2019;29:92–96.
    1. Khaw KT, Friesen MD, Riboli E, Luben R, Wareham N. Plasma phospholipid fatty acid concentration and incident coronary heart disease in men and women: the EPIC-Norfolk prospective study. PLOS Med. 2012;9:e1001255. doi: 10.1371/journal.pmed.1001255.
    1. Krachler B, et al. Fatty acid profile of the erythrocyte membrane preceding development of type 2 diabetes. Nutr Metab Cardiovasc Dis. 2008;18:503–510. doi: 10.1016/j.numecd.2007.04.005.
    1. Liu S, et al. Intake of dietary saturated fatty acids and risk of type 2 diabetes in the European Prospective Investigation into Cancer and Nutrition-Netherlands cohort: associations by types, sources of fatty acids and substitution by macronutrients. Eur J Nutrition. 2019;58:1125–1136. doi: 10.1007/s00394-018-1630-4.
    1. Maruyama C, et al. Differences in serum phospholipid fatty acid compositions and estimated desaturate activities between Japanese men with and without metabolic syndrome. J Atheroscler Thromb. 2008;15:306–313. doi: 10.5551/jat.E564.
    1. Matejcic M, et al. Circulating plasma phospholipid fatty acids and risk of pancreatic cancer in a large European cohort. Int J Cancer. 2018;143:2437–2448. doi: 10.1002/ijc.31797.
    1. Unger AL, Torres-Gonzales M, Kraft J. Dairy fat consumption and the risk of metabolic syndrome: an examination of the saturated fatty acids in dairy. Nutrients. 2019;11:220. doi: 10.3390/nu11092200.
    1. Villamor E, Villar LA, Lozano A, Herrera VM, Herran OF. Serum fatty acids and progression from dengue fever to dengue hemorrhagic fever / dengue shock syndrome. Br J Nutr. 2018;120:787–796. doi: 10.1017/S0007114518002039.
    1. Yoo W, et al. Fatty acids in non-alcoholic steatohepatitis: focus on pentadecanoic acid. PLOS ONE. 2017;12:e0189965. doi: 10.1371/journal.pone.0189965.
    1. Zhu Y, et al. A prospective and longitudinal study of plasma phospholipid saturated fatty acid profile in relation to cardiometabolic markers and the risk of gestational diabetes. Am J Clin Nutr. 2018;107:1017–1026. doi: 10.1093/ajcn/nqy051.
    1. Zhuang P, Cheng L, Wang J, Zhang Y. Saturated fatty acid intake is associated with total mortality in a nationwide cohort study. J Nutrition. 2019;149:68–77. doi: 10.1093/jn/nxy237.
    1. Mozaffarian D. Saturated fatty acids and type 2 diabetes: more evidence to re-invent dietary guidelines. Lancet Diab Endocrinol. 2014;2:770–772. doi: 10.1016/S2213-8587(14)70166-4.
    1. Astrup A, et al. WHO draft guidelines on dietary saturated and trans fatty acids: time for a new approach? BMJ. 2019;366:14137.
    1. Zheng JS, et al. Changes in plasma phospholipid fatty acid profiles over 13 years and correlates of change: European Prospective Investigation into Cancer and Nutrition-Norfolk Study. Am J Clin Nutr. 2019;109:1527–1534. doi: 10.1093/ajcn/nqz030.
    1. Brevik A, Veierod MB, Drevon CA, Andersen LF. Evaluation of the odd fatty acids 15:0 and 17:0 in serum and adipose tissue as markers of intake of milk and dairy fat. Eur J Clin Nutr. 2005;59:1417–1422. doi: 10.1038/sj.ejcn.1602256.
    1. Venn-Watson S, et al. Blood-based indicators of insulin resistance and metabolic syndrome in bottlenose dolphins (Tursiops truncatus) Frontiers Endo. 2013;4:136.
    1. Venn-Watson S, Benham C, Carlin K, St. Leger J. Hemochromatosis and fatty change: building evidence for insulin resistance in bottlenose dolphins (Tursiops truncatus) J Zoo Wildlf Med. 2012;43:S35–S47. doi: 10.1638/2011-0146.1.
    1. Venn-Watson S, et al. Increased dietary intake of saturated fatty acid heptadecanoic acid (C17:0) associated with decreasing ferritin and alleviated metabolic syndrome in dolphins. PLOS ONE. 2015;10:e0132117. doi: 10.1371/journal.pone.0132117.
    1. Wang DH, et al. Saturated branched chain, normal odd-carbon-numbered, and n-3 (Omega-3) polyunsaturated fatty acids in freshwater fish in the Northeastern United States. J Agric Food Chem. 2016;64:7512–7519. doi: 10.1021/acs.jafc.6b03491.
    1. Meluchova B, et al. Seasonal variations in fatty acid composition of pasture forage plants and CLA content in ewe milk fat. Small Rum Res. 2008;78:56–65. doi: 10.1016/j.smallrumres.2008.05.001.
    1. Bas P, Archimede H, Rouzeau A, Sauvant D. Fatty acid composition of mixed-rumen bacteria: effect of concentration and type of forage. J Dairy Sci. 2003;86:2940–2948. doi: 10.3168/jds.S0022-0302(03)73891-0.
    1. Harwood, J. L. Membrane lipids in algae. In: Paul-Andre, S. & Norio, M. (eds) Lipids in photosynthesis: Structure, function and genetics. Adv Photosyn Respir6:53–63 (1998).
    1. Forman BM, Chen J, Evans RM. Hypolipidemic drugs, polyunsaturated fatty acids, and eicosanoids are ligands for peroxisome proliferator-activated receptors α and δ. Proc Natl Acac Sci. 1997;94:4312–4317. doi: 10.1073/pnas.94.9.4312.
    1. Gross B, Pawlak M, Lefebvre P, Staels B. PPARs in obesity-induced T2DM, dyslipidemia and NAFLD. Nature Rev Endocrinol. 2017;13:36–49. doi: 10.1038/nrendo.2016.135.
    1. Kilgore KS, Billin AN. PPAR β/δ ligands as modulators of the inflammatory response. Curr Opin Invest Drugs. 2008;9:463–469.
    1. Pawlak M, Lefebvre P, Staels B. Molecular mechanism of PPARα action and its impact on lipid metabolism, inflammation and fibrosis in non-alcoholic fatty liver disease. J Hepatol. 2015;62:720–733. doi: 10.1016/j.jhep.2014.10.039.
    1. Haas RH. Mitochondrial dysfunction in aging and diseases of aging. Biology. 2019;8:48. doi: 10.3390/biology8020048.
    1. Wojtczak L, Schonfeld P. Effect of fatty acids on energy coupling processes in mitochondria. BBA. 1993;1183:31–57.
    1. Otogawa K, et al. Erythrophagocytosis by liver macrophages (Kupffer cells) promotes oxidative stress, inflammation, and fibrosis in a rabbit model of steatohepatitis: implications for the pathogenesis of human nonalcoholic steatohepatitis. Am J Pathol. 2007;170:967–980. doi: 10.2353/ajpath.2007.060441.
    1. Holman R. Essential fatty acids. Nutr Rev. 1958;16:33–35. doi: 10.1111/j.1753-4887.1958.tb00660.x.
    1. Donath MY, Shoelson SE. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol. 2011;11:98–107. doi: 10.1038/nri2925.
    1. Esser N, Paquot N, Scheen AJ. Anti-inflammatory agents to treat or prevent type 2 diabetes, metabolic syndrome and cardiovascular disease. Expert Opin Investig Drugs. 2015;24:283–307. doi: 10.1517/13543784.2015.974804.
    1. Fulop T, Larbi A, Witkowski JM. Human inflammaging. Gertontol. 2019;65:495–504. doi: 10.1159/000497375.
    1. Wang X, et al. Lipopolysaccharaide and palmitic acid synergistically induced MCP-1 production in MAPK-mediated TLR4 signaling pathway in RAW264.7 cells. Lipids Health Dis. 2019;18:71. doi: 10.1186/s12944-019-1017-4.
    1. Takaishi H, Taniguchi T, Takahashi A, Ishikawa Y, Yokoyama M. High glucose accelerates MCP-1 production via p38 MAPK in vascular endothelial cells. Biochem Biophys Res Commun. 2003;305:122–8. doi: 10.1016/S0006-291X(03)00712-5.
    1. Monti P, et al. The CC chemokine MCP-1/CCL2 in pancreatic Cancer progression: regulation of expression and potential mechanisms of antimalignant activity. Cancer Res. 2003;63:7451–61.
    1. Yousefzadeh MJ, et al. Circulating levels of monocyte chemoattractant protein-1 as a potential measure of biological age in mice and frailty in humans. Aging Cell. 2018;17:e12706. doi: 10.1111/acel.12706.
    1. Egin, A. The definition and prevalence of obesity and metabolic syndrome. In: Engin A., Engin A. (eds) Obesity and Lipotoxicity. Adv Exp Med and Biol, 960, 1–17. Springer, Cham.
    1. Zheng JS, et al. Association between plasma phospholipid saturated fatty acids and metabolic markers of lipid, hepatic, inflammation and glycaemic pathways in eight European countries: a cross-sectional analysis in the EPIC-InterAct study. BMC Medicine. 2017;15:203. doi: 10.1186/s12916-017-0968-4.
    1. Jenkins B, et al. Odd chain fatty acids: new insights of the relationship between the gut microbiota, dietary intake, biosynthesis and glucose intolerance. Sci Rep. 2017;7:44845. doi: 10.1038/srep44845.
    1. Wang Z, et al. The elongation of very long-chain fatty acid 6 gene product catalyzes elongation of n-13:0 and n-15:0 odd-chain SFA in human cells. Br J Nutr. 2019;121:241–248. doi: 10.1017/S0007114518003185.
    1. Younossi Z, et al. Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nature Rev Gastroenterol Hepatol. 2018;15:11–20. doi: 10.1038/nrgastro.2017.109.
    1. Ogawa T, Fujii H, Yoshizato K, Kawada N. A human-type nonalcoholic steatohepatitis model with advanced fibrosis in rabbits. Am J Pathol. 2010;177:153–165. doi: 10.2353/ajpath.2010.090895.
    1. Angulo P, et al. Liver fibrosis, but no other histologic features, is associated with long-term outcomes of patients with nonalcoholic fatty liver disease. Gastroenterol. 2015;149:389–397. doi: 10.1053/j.gastro.2015.04.043.
    1. Weiss G, Goodnough LT. Anemia of chronic disease. NEJM. 2005;352:1011–1023. doi: 10.1056/NEJMra041809.
    1. Sarnak MJ, et al. Anemia as a risk factor for cardiovascular disease in the atherosclerosis risk in communities (ARIC) study. J Am Coll Cardiol. 2002;40:27–33. doi: 10.1016/S0735-1097(02)01938-1.
    1. Kratz M, et al. Dairy fat intake is associated with glucose tolerance, hepatic and systemic insulin sensitivity, and liver fat but not β-cell function in humans. Am. J. Clin. Nutr. 2014;99:1385–1396. doi: 10.3945/ajcn.113.075457.
    1. Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153:1194–1217. doi: 10.1016/j.cell.2013.05.039.
    1. Pfeuffer M, Jaudszus A. Pentadecanoic and heptadecanoic acids: multifaceted odd-chain fatty acids. Adv Nutr. 2016;7:730–743. doi: 10.3945/an.115.011387.
    1. Kaur N, Chugh V, Gupta AK. Essential fatty acids as functional components of foods – a review. J Food Sci Technol. 2014;51:2289–2303. doi: 10.1007/s13197-012-0677-0.
    1. Katare A, Thakkar S, Dhepale S, Khunt D, Misra M. Fatty acids as essential adjuvants to treat various ailments and their role in drug delivery: a review. Nutrition. 2019;65:138–157. doi: 10.1016/j.nut.2019.03.008.
    1. Burr GO, Burr MM. On the nature and role of the fatty acids essential in nutrition. J Biol Chem. 1930;86:587–621.
    1. Carlson SJ, et al. A diet with docosahexaenoic and arachidonic acids as the sole source of polyunsaturated fatty acids is sufficient to support visual, cognitive, motor, and social development in mice. Front Neurosci. 2019;13:72. doi: 10.3389/fnins.2019.00072.
    1. Jenkins B, et al. The dietary total-fat content affects the in vivo circulating C15:0 and C17:0 fatty acid levels independently. Nutrients. 2018;10:1646. doi: 10.3390/nu10111646.
    1. Weitkunat K, et al. Odd-chain fatty acids as a biomarker for dietary fiber intake: a novel pathway for endogenous production from propionate. Am J Clin Nutr. 2017;105:1544–1551.
    1. Jenkins B, de Schryver E, Van Veldhoven PP, Koulman AA. Peroxisomal 2-hydroxyacyl-CoA lyase is involved in endogenous biosynthesis of heptadecanoic acid. Molecules. 2017;13:E1718. doi: 10.3390/molecules22101718.
    1. Mascarehenas MR, et al. Malabsorption blood test: assessing fat absorption in patients with cystic fibrosis and pancreatic insufficiency. J Clin Pharmacol. 2015;55:854–865. doi: 10.1002/jcph.484.
    1. Stallings VA, et al. Diagnosing malabsorption with systemic lipid profiling: pharmacokinetics of pentadecanoic acid and triheptadecanoic acid following oral administration in healthy subjects and subjects with cystic fibrosis. Int J Clin Pharmacol Ther. 2013;51:263–273. doi: 10.5414/CP201793.
    1. Wolk A, Vessby B, Ljung H, Barrefors P. Evaluation of a biological marker of dairy fat intake. Am J Clin Nutr. 1998;68:291–295. doi: 10.1093/ajcn/68.2.291.
    1. Chaikoff IL, Bloom B, Stevens BP, Reinhardt WO, Dauben WG. Pentadecanoic acid-5-C14: its absorption and lymphatic transport. J Biol Chem. 1951;190:431–435.
    1. Ikwobe OJ, et al. 418 – Odd-chain fatty acids predict insulin sensitivity in people with T2DM and protect HepG2 cells from palmitate-induced insulin resistance via PPARα. Free Radical Biol Med. 2016;100:S174. doi: 10.1016/j.freeradbiomed.2016.10.459.
    1. Lagerstedt SA, et al. Quantitative determination of plasma C8-C26 total fatty acids for the biochemical diagnosis of nutritional and metabolic disorders. Mol. Gen. Metabol. 2001;73:38–45. doi: 10.1006/mgme.2001.3170.

Source: PubMed

3
Abonneren