Cold exposure induces dynamic changes in circulating triacylglycerol species, which is dependent on intracellular lipolysis: A randomized cross-over trial

Maaike E Straat, Lucas Jurado-Fasoli, Zhixiong Ying, Kimberly J Nahon, Laura G M Janssen, Mariëtte R Boon, Gernot F Grabner, Sander Kooijman, Robert Zimmermann, Martin Giera, Patrick C N Rensen, Borja Martinez-Tellez, Maaike E Straat, Lucas Jurado-Fasoli, Zhixiong Ying, Kimberly J Nahon, Laura G M Janssen, Mariëtte R Boon, Gernot F Grabner, Sander Kooijman, Robert Zimmermann, Martin Giera, Patrick C N Rensen, Borja Martinez-Tellez

Abstract

Background: The application of cold exposure has emerged as an approach to enhance whole-body lipid catabolism. The global effect of cold exposure on the lipidome in humans has been reported with mixed results depending on intensity and duration of cold.

Methods: This secondary study was based on data from a previous randomized cross-over trial (ClinicalTrials.gov ID: NCT03012113). We performed sequential lipidomic profiling in serum during 120 min cold exposure of human volunteers. Next, the intracellular lipolysis was blocked in mice (eighteen 10-week-old male wild-type mice C57BL/6J) using a small-molecule inhibitor of adipose triglyceride lipase (ATGL; Atglistatin), and mice were exposed to cold for a similar duration. The quantitative lipidomic profiling was assessed in-depth using the Lipidyzer platform.

Findings: In humans, cold exposure gradually increased circulating free fatty acids reaching a maximum at 60 min, and transiently decreased total triacylglycerols (TAGs) only at 30 min. A broad range of TAG species was initially decreased, in particular unsaturated and polyunsaturated TAG species with ≤5 double bonds, while after 120 min a significant increase was observed for polyunsaturated TAG species with ≥6 double bonds in humans. The mechanistic study in mice revealed that the cold-induced increase in polyunsaturated TAGs was largely prevented by blocking adipose triglyceride lipase.

Interpretation: We interpret these findings as that cold exposure feeds thermogenic tissues with TAG-derived fatty acids for combustion, resulting in a decrease of circulating TAG species, followed by increased hepatic production of polyunsaturated TAG species induced by liberation of free fatty acids stemming from adipose tissue.

Funding: This work was supported by the Netherlands CardioVascular Research Initiative: 'the Dutch Heart Foundation, Dutch Federation of University Medical Centers, the Netherlands Organisation for Health Research and Development and the Royal Netherlands Academy of Sciences' [CVON2017-20 GENIUS-II] to Patrick C.N. Rensen. Borja Martinez-Tellez is supported by individual postdoctoral grant from the Fundación Alfonso Martin Escudero and by a Maria Zambrano fellowship by the Ministerio de Universidades y la Unión Europea - NextGenerationEU (RR_C_2021_04). Lucas Jurado-Fasoli was supported by an individual pre-doctoral grant from the Spanish Ministry of Education (FPU19/01609) and with an Albert Renold Travel Fellowship from the European Foundation for the Study of Diabetes (EFSD). Martin Giera was partially supported by NWO XOmics project #184.034.019.

Keywords: Cold; Desaturation; Fatty acid metabolism; Intracellular lipolysis; Long-chain polyunsaturated fatty acids.

Conflict of interest statement

Declaration of interests None.

Copyright © 2022 The Authors. Published by Elsevier B.V. All rights reserved.

Figures

Fig. 1
Fig. 1
Study design and lipidomic changes during cold exposure. (a) Overview of the crossover study design. During the first occasion, participants were exposed to a personalized cooling protocol. During the other occasion, participants were exposed to constant room temperature. Under both conditions, venous blood was drawn at five sequential timepoints (i.e., baseline, 30, 60, 90, and 120 min) to obtain serum for longitudinal lipidomic profiling. (b) Dynamic changes in total free fatty acid and triacylglycerol concentration during cold exposure, as measured with enzymatic assays. Data is presented as mean and standard error of the mean. P values are obtained from ANOVA repeated measures. ∗P < 0.05, ∗∗P < 0.01; compared to baseline (i.e., 0 min). (c) Baseline relative abundance of the number of lipid classes. (d) Dynamic changes in lipid classes during cold exposure. Data is presented as the log2 fold change (log2FC) relative to baseline (i.e., 0 min). P values are obtained from ANOVA repeated measures. (e) Volcano plot showing the change of individual lipid species after 120 min of cold exposure. The X-axis shows the log2FC between 120 min of cold exposure vs. baseline, the Y-axis shows the P value. P values are obtained from paired Student's t test. Values between brackets indicate the absolute number and the percentage of lipid species within the lipid class that were modulated by cold exposure. (f) Heatmap of free fatty acid (FFA) species that significantly dynamically changed during cold exposure. The color of the squares represents the log2FC relative to baseline. FFAs are sorted by the number of double bonds. ANOVA repeated measures were used to compare the different timepoints. CE, cholesteryl esters; CER, ceramides; DAG, diacylglycerols; DCER, dihydroceramides; FFA, free fatty acids; FC, fold change; HCER, hexosylceramides; LCER, lactosylceramides; LPC, lysophosphatidylcholine; LPE, lysophosphatidylethanolamine; PC, phosphatidylcholine; PE, phosphatidylethanolamine; SM, sphingomyelin; TAG, triacylglycerols; TN, thermoneutral.
Fig. 2
Fig. 2
Triacylglycerol (TAG) species response to cold exposure. (a) Heatmap of TAG species that significantly dynamically changed during cold exposure. The color of the squares represents the log2 fold change (log2FC) relative to baseline. TAGs are sorted by the number of double bonds. ANOVA repeated measures were used to compare the different timepoints. (b) Bubble plot showing the P value (in color) and log2FC (on y-axis) after 120 min of cold exposure per TAG species categorized by the number of double bonds. P values are obtained from paired Student's t test. (c) Bubble plot showing the P value (in color) and log2FC (on y-axis) after 120 min of cold exposure per TAG species categorized by the total length of the TAG. P values are obtained from paired Student's t test. FA, fatty acid; FC, fold change; TAG, triacylglycerol.
Fig. 3
Fig. 3
Triacylglycerol (TAG) species' response to mild cold exposure in mice either treated with vehicle or with Atglistatin. (a and b) Bubble plots showing the P value (in color) and log2 fold change (log2FC; on y-axis) between mice exposed to cold and mice kept at thermoneutrality (n = 6 each group; both treated with vehicle) per TAG species categorized by the number of double bonds (a) and total length (b). P values are obtained from paired Student's t test. (c and d) Bubble plots showing the P value (in color) and log2FC (on y-axis) between mice exposed to cold and treated with Atglistatin and mice kept at thermoneutrality (n = 6 each group; treated with vehicle) per TAG species categorized by the number of double bonds (c) and total length (d). P values are obtained from paired Student's t test. FC, fold change; TAG, triacylglycerol.
Fig. 4
Fig. 4
Graphic summary of proposed mechanism. We propose a mechanism by which cold exposure induces intracellular lipolysis in white adipose tissue, increasing circulating free fatty acids that can subsequently drive hepatic biosynthesis and release of triacylglycerol (TAG)-containing very-low density lipoprotein (VLDL) particles to feed thermogenic tissues. The cold-activated thermogenic tissues (i.e., brown adipose tissue and skeletal muscle) take up TAG-derived fatty acids from VLDL particles as fuel for oxidation to generate heat. FA, fatty acid; FFA, free fatty acids; PUFA, polyunsaturated fatty acid; TAG, triacylglycerol; VLDL, very-low density lipoprotein.

References

    1. Malik V.S., Willett W.C., Hu F.B. Global obesity: trends, risk factors and policy implications. Nat Rev Endocrinol. 2013;9(1):13–27.
    1. Finucane M.M., Stevens G.A., Cowan M.J., et al. National, regional, and global trends in body-mass index since 1980: systematic analysis of health examination surveys and epidemiological studies with 960 country-years and 9.1 million participants. Lancet. 2011;377(9765):557–567.
    1. Carobbio S., Rodriguez-Cuenca S., Vidal-Puig A. Origins of metabolic complications in obesity: ectopic fat accumulation. The importance of the qualitative aspect of lipotoxicity. Curr Opin Clin Nutr Metab Care. 2011;14(6):520–526.
    1. Cannon B., Nedergaard J.A.N. Brown adipose tissue: function and physiological significance. Physiol Rev. 2004;84(1):277–359.
    1. Dijk W., Heine M., Vergnes L., et al. ANGPTL4 mediates shuttling of lipid fuel to brown adipose tissue during sustained cold exposure. Elife. 2015;4:e08428.
    1. Bartelt A., Bruns O.T., Reimer R., et al. Brown adipose tissue activity controls triglyceride clearance. Nat Med. 2011;17(2):200–205.
    1. Khedoe P.P.S.J., Hoeke G., Kooijman S., et al. Brown adipose tissue takes up plasma triglycerides mostly after lipolysis. J Lipid Res. 2015;56(1):51–59.
    1. Berbée J.F.P., Boon M.R., Khedoe P., et al. Brown fat activation reduces hypercholesterolaemia and protects from atherosclerosis development. Nat Commun. 2015;6(1):1–11.
    1. Hoeke G., Kooijman S., Boon M.R., Rensen P.C.N., Berbée J.F.P. Role of brown fat in lipoprotein metabolism and atherosclerosis. Circ Res. 2016;118(1):173–182.
    1. Bartelt A., John C., Schaltenberg N., et al. Thermogenic adipocytes promote HDL turnover and reverse cholesterol transport. Nat Commun. 2017;8(1):1–10.
    1. Ouellet V., Labbé S.M., Blondin D.P., et al. Brown adipose tissue oxidative metabolism contributes to energy expenditure during acute cold exposure in humans. J Clin Invest. 2012;122(2):545–552.
    1. U Din M., Raiko J., Saari T., et al. Human brown adipose tissue [15O] O2 PET imaging in the presence and absence of cold stimulus. Eur J Nucl Med Mol Imaging. 2016;43(10):1878–1886.
    1. Saari T.J., Raiko J., U-Din M., et al. Basal and cold-induced fatty acid uptake of human brown adipose tissue is impaired in obesity. Sci Rep. 2020;10(1):1–11.
    1. Din M.U., Saari T., Raiko J., et al. Postprandial oxidative metabolism of human brown fat indicates thermogenesis. Cell Metab. 2018;28(2):207–216.
    1. Blondin D.P., Labbé S.M., Noll C., et al. Selective impairment of glucose but not fatty acid or oxidative metabolism in brown adipose tissue of subjects with type 2 diabetes. Diabetes. 2015;64(7):2388–2397.
    1. Hoeke G., Nahon K.J., Bakker L.E.H., et al. Short-term cooling increases serum triglycerides and small high-density lipoprotein levels in humans. J Clin Lipidol. 2017;11(4):920–928.
    1. Straat M.E., Martinez-Tellez B., Sardjoe Mishre A., et al. Cold-induced thermogenesis shows a diurnal variation that unfolds differently in males and females. J Clin Endocrinol Metab. 2022;107(6):1626–1635.
    1. Ghorasaini M., Mohammed Y., Adamski J., et al. Cross-laboratory standardization of preclinical lipidomics using differential mobility spectrometry and multiple reaction monitoring. Anal Chem. 2021;93(49):16369–16378.
    1. Mayer N., Schweiger M., Romauch M., et al. Development of small-molecule inhibitors targeting adipose triglyceride lipase. Nat Chem Biol. 2013;9(12):785–787.
    1. Nahon K.J., Janssen L.G.M., Sardjoe Mishre A.S.D., et al. The effect of mirabegron on energy expenditure and brown adipose tissue in healthy lean South Asian and Europid men. Diabetes Obes Metab. 2020;22(11):2032–2044.
    1. van Eenige R., Verhave P.S., Koemans P.J., Tiebosch I.A.C.W., Rensen P.C.N., Kooijman S. RandoMice, a novel, user-friendly randomization tool in animal research. PLoS One. 2020;15(8):e0237096.
    1. Liebisch G., Vizcaíno J.A., Köfeler H., et al. Shorthand notation for lipid structures derived from mass spectrometry. J Lipid Res. 2013;54(6):1523–1530.
    1. Choi S.S., Diehl A.M. Hepatic triglyceride synthesis and nonalcoholic fatty liver disease. Curr Opin Lipidol. 2008;19(3):295–300.
    1. Chow S.C., Shao J., Wang H., Lokhnygina Y. Chapman and Hall/CRC; 2017. Sample size calculations in clinical research.
    1. Madden C.J., Morrison S.F. Central nervous system circuits that control body temperature. Neurosci Lett. 2019;696:225–232.
    1. Bartness T.J., Song C.K. Thematic review series: adipocyte biology. Sympathetic and sensory innervation of white adipose tissue. J Lipid Res. 2007;48(8):1655–1672.
    1. Labbé S.M., Caron A., Bakan I., et al. In vivo measurement of energy substrate contribution to cold-induced brown adipose tissue thermogenesis. FASEB J. 2015;29(5):2046–2058.
    1. Schilperoort M., Hoeke G., Kooijman S., Rensen P.C.N. Relevance of lipid metabolism for brown fat visualization and quantification. Curr Opin Lipidol. 2016;27(3):242–248.
    1. la Fountaine M.F., Cirnigliaro C.M., Kirshblum S.C., McKenna C., Bauman W.A. Effect of functional sympathetic nervous system impairment of the liver and abdominal visceral adipose tissue on circulating triglyceride-rich lipoproteins. PLoS One. 2017;12(3):e0173934.
    1. Geerling J.J., Boon M.R., Kooijman S., et al. Sympathetic nervous system control of triglyceride metabolism: novel concepts derived from recent studies. J Lipid Res. 2014;55(2):180–189.
    1. Russo G.L. Dietary n− 6 and n− 3 polyunsaturated fatty acids: from biochemistry to clinical implications in cardiovascular prevention. Biochem Pharmacol. 2009;77(6):937–946.
    1. Sprecher H. Biochemistry of essential fatty acids. Prog Lipid Res. 1981;20:13–22.
    1. Sprecher H., Luthria D.L., Mohammed B.S., Baykousheva S.P. Reevaluation of the pathways for the biosynthesis of polyunsaturated fatty acids. J Lipid Res. 1995;36(12):2471–2477.
    1. Zhang J.Y., Kothapalli K.S.D., Brenna J.T. Desaturase and elongase limiting endogenous long chain polyunsaturated fatty acid biosynthesis. Curr Opin Clin Nutr Metab Care. 2016;19(2):103.
    1. di Gregorio E., Borroni B., Giorgio E., et al. ELOVL5 mutations cause spinocerebellar ataxia 38. Am J Hum Genet. 2014;95(2):209–217.
    1. Leonard A.E., Kelder B., Bobik E.G., et al. Identification and expression of mammalian long-chain PUFA elongation enzymes. Lipids. 2002;37(8):733–740.
    1. Lynes M.D., Shamsi F., Sustarsic E.G., et al. Cold-activated lipid dynamics in adipose tissue highlights a role for cardiolipin in thermogenic metabolism. Cell Rep. 2018;24(3):781–790.
    1. Chen L., Yan G., Ohwada T. Building on endogenous lipid mediators to design synthetic receptor ligands. Eur J Med Chem. 2022;231:114154.
    1. Marcher A.B., Loft A., Nielsen R., et al. RNA-seq and mass-spectrometry-based lipidomics reveal extensive changes of glycerolipid pathways in brown adipose tissue in response to cold. Cell Rep. 2015;13(9):2000–2013.
    1. Pernes G., Morgan P.K., Huynh K., et al. Characterization of the circulating and tissue-specific alterations to the lipidome in response to moderate and major cold stress in mice. Am J Physiol Regul Integr Comp Physiol. 2021;320(2):R95–R104.
    1. Xu Z., You W., Zhou Y., Chen W., Wang Y., Shan T. Cold-induced lipid dynamics and transcriptional programs in white adipose tissue. BMC Biol. 2019;17(1):1–21.
    1. Nielsen S., Karpe F. Determinants of VLDL-triglycerides production. Curr Opin Lipidol. 2012;23(4):321–326.
    1. Lewis G.F. Fatty acid regulation of very low density lipoprotein production. Curr Opin Lipidol. 1997;8(3):146–153.
    1. Kershaw E.E., Hamm J.K., Verhagen L.A.W., Peroni O., Katic M., Flier J.S. Adipose triglyceride lipase: function, regulation by insulin, and comparison with adiponutrin. Diabetes. 2006;55(1):148–157.
    1. Zimmermann R., Strauss J.G., Haemmerle G., et al. Fat mobilization in adipose tissue is promoted by adipose triglyceride lipase. Science (1979) 2004;306(5700):1383–1386.
    1. Demetrius L. Of mice and men: when it comes to studying ageing and the means to slow it down, mice are not just small humans. EMBO Rep. 2005;6(S1):S39–S44.
    1. van den Beukel J.C., Boon M.R., Steenbergen J., et al. Cold exposure partially corrects disturbances in lipid metabolism in a male mouse model of glucocorticoid excess. Endocrinology. 2015;156(11):4115–4128.
    1. Carpentier A.C., Blondin D.P., Virtanen K.A., Richard D., Haman F., Turcotte É.E. Brown adipose tissue energy metabolism in humans. Front Endocrinol (Lausanne) 2018;9:447.
    1. Mika A., Sledzinski T. Alterations of specific lipid groups in serum of obese humans: a review. Obes Rev. 2017;18(2):247–272.
    1. Diraison F., Dusserre E., Vidal H., Sothier M., Beylot M. Increased hepatic lipogenesis but decreased expression of lipogenic gene in adipose tissue in human obesity. Am J Physiol Endocrinol Metab. 2002;282(1):E46–E51.
    1. Auguet T., Guiu-Jurado E., Berlanga A., et al. Downregulation of lipogenesis and fatty acid oxidation in the subcutaneous adipose tissue of morbidly obese women. Obesity. 2014;22(9):2032–2038.
    1. Fan R., Koehler K., Chung S. Adaptive thermogenesis by dietary n-3 polyunsaturated fatty acids: emerging evidence and mechanisms. Biochim Biophys Acta Mol Cell Biol Lipids. 2019;1864(1):59–70.
    1. Kim M., Goto T., Yu R., et al. Fish oil intake induces UCP1 upregulation in brown and white adipose tissue via the sympathetic nervous system. Sci Rep. 2015;5(1):1–12.
    1. Kim J., Okla M., Erickson A., Carr T., Natarajan S.K., Chung S. Eicosapentaenoic acid potentiates brown thermogenesis through FFAR4-dependent up-regulation of miR-30b and miR-378. J Biol Chem. 2016;291(39):20551–20562.
    1. Watanabe Y., Tatsuno I. Prevention of cardiovascular events with omega-3 polyunsaturated fatty acids and the mechanism involved. J Atheroscler Thromb. 2020;27(3):183–198.
    1. Denisenko Y.K., Kytikova O.Y., Novgorodtseva T.P., Antonyuk M v, Gvozdenko T.A., Kantur T.A. Lipid-induced mechanisms of metabolic syndrome. J Obes. 2020;2020:5762395.
    1. Tousoulis D., Plastiras A., Siasos G., et al. Omega-3 PUFAs improved endothelial function and arterial stiffness with a parallel antiinflammatory effect in adults with metabolic syndrome. Atherosclerosis. 2014;232(1):10–16.
    1. Bhatt D.L., Steg P.G., Miller M., et al. Cardiovascular risk reduction with icosapent ethyl for hypertriglyceridemia. N Engl J Med. 2019;380(1):11–22.
    1. Ong K.T., Mashek M.T., Bu S.Y., Greenberg A.S., Mashek D.G. Adipose triglyceride lipase is a major hepatic lipase that regulates triacylglycerol turnover and fatty acid signaling and partitioning. Hepatology. 2011;53(1):116–126.

Source: PubMed

3
Suscribir