Abstract

Background

Accumulating evidence links brown adipose tissue (BAT) to increased cold-induced energy expenditure (CIEE) and regulation of lipid metabolism in humans. BAT has also been proposed as a novel source for biologically active lipid mediators including polyunsaturated fatty acids (PUFAs) and oxylipins. However, little is known about cold-mediated differences in energy expenditure and various lipid species between individuals with detectable BAT positive (BATpos) and those without BAT negative (BATneg).

Methods

Here we investigated a unique cohort of matched BATpos and BATneg individuals identified by 18F-fluorodeoxyglucose positron emission tomography combined with computed tomography ([18F]-FDG PET/CT). BAT function, CIEE, and circulating oxylipins, were analyzed before and after short-term cold exposure using [18F]-FDG PET/CT, indirect calorimetry, and high-resolution mass spectrometry, respectively.

Results

We found that active BAT is the major determinant of CIEE since only BATpos individuals experienced significantly increased energy expenditure in response to cold. A single bout of moderate cold exposure resulted in the dissipation of an additional 20 kcal excess energy in BATpos but not in BATneg individuals. The presence of BAT was associated with a unique systemic PUFA and oxylipin profile characterized by increased levels of anti-inflammatory omega-3 fatty acids as well as cytochrome P450 products but decreased concentrations of some proinflammatory hydroxyeicosatetraenoic acids when compared with BATneg individuals. Notably, cold exposure raised circulating levels of various lipids, including the recently identified BAT-derived circulating factors (BATokines) DiHOME and 12-HEPE, only in BATpos individuals.

Conclusions

In summary, our data emphasize that BAT in humans is a major contributor toward cold-mediated energy dissipation and a critical organ in the regulation of the systemic lipid pool.

Brown adipose tissue (BAT) is a thermogenic organ that dissipates chemical energy through heat production. Promotion of BAT function counteracts adiposity in numerous animal models and holds great promise as novel antiobesity concept in humans (1-5). Cold exposure is the most potent physiologic stimulus of BAT thermogenesis that results in increased energy expenditure in mice and humans. In humans, cold-activated BAT can be specifically detected by 18F-fluorodeoxyglucose positron emission tomography combined with computed tomography ([18F]-FDG PET/CT) (1, 2, 5-8). Cold-induced [18F]-FDG uptake in BAT has been shown to correlate positively with energy expenditure and negatively with adiposity in humans (7, 9-11). Indeed, BAT-dependent cold-induced thermogenesis significantly contributes to total energy expenditure, and chronic cold exposure studies demonstrated a decrease in body fat mass (9-13). However, the presence of BAT is very heterogeneous in the general population with large variations even in young healthy adults. For example, in a Japanese cohort of 260 healthy volunteers, only 125 (48%) participants had detectable BAT depots (BATpos) after cold exposure (14). As previously reported in a number of other studies, participants without detectable BAT (BATneg) were significantly older, had higher fat mass and higher expression of adiposity-related parameters such as hemoglobin A1c, triglycerides, and cholesterol (14-16). However, very little is known about the differences in energy expenditure and lipid metabolites between age- and weight-matched BATpos and BATneg individuals and therefore the individual contribution of BAT toward systemic fuel metabolism. Studying such interindividual differences is of particular importance given accumulating evidence that cold-activated BAT also improves insulin sensitivity and glucose uptake and significantly alters lipid homeostasis (12, 17, 18). BAT activation promotes lipid mobilization from peripheral stores and oxidative disposal, presumably to accommodate the increased fuel needs of BAT mitochondrial activity (19, 20). In keeping with this notion are observations of elevated triglyceride-rich very low-density lipoprotein concentrations following short-term cold exposure, possibly as a consequence of increased hepatic output (21). Recent evidence suggests that activated BAT also represents a source of specific lipid species that may contribute to metabolic homoeostasis (19, 22, 23). These and other observations suggest that BAT may be associated with the individual risk for type 2 diabetes (24). Polyunsaturated fatty acids (PUFAs) as well as their oxidation products, known as eicosanoids, are the endogenous ligands for peroxisome proliferator-associated receptors (PPARs), which act as transcriptional regulators of glucose and lipid metabolism (25). Specific lipid species such as 12,13-dihydroxyoctadecenoic acid (DiHOME) have been described to be secreted from brown adipocytes upon exercise (23) and cold exposure (26) with effects on fatty acid release and metabolism, while circulating levels of the lysophosphatidylcholine-acyl C16:0 correlate with BAT activity in humans (27). The identification of specific lipid patterns associated with functional BAT contributes toward a better understanding of the importance of BAT for lipid metabolism and may help identify potential BAT biomarkers. In this study we investigated the differences in cold-induced energy expenditure (CIEE) and circulating oxylipin profiles between BATpos and BATneg individuals. We found that a single bout of cold exposure significantly elevated energy expenditure for more than 2 hours after cessation of the cold challenge, whereas CIEE was absent in BATneg individuals. The presence of active BAT was associated with an anti-inflammatory oxylipin/eicosanoid profile, and cold exposure increased systemic oxylipin levels only in BATpos but not BATneg individuals. We identified distinct PUFA and oxylipin clusters that were concomitantly regulated by the presence of BAT and cold exposure and were associated with CIEE.

Materials and Methods

Participants and study approval

We included 16 age- and body mass index (BMI)-matched healthy volunteers, all from the same geographical region (28) that were previously identified as BATpos or BATneg (n = 8/group) in a cross-sectional BAT imaging study (NCT02381483). While BATneg participants had no significant [18F]-FDG uptake at either room temperature (RT) or after cold exposure, BATpos participants had strong [18F]-FDG uptake in the typical cervical and thoracic BAT regions following cold exposure (Fig. 1A, B). For the initial imaging study, interested volunteers were recruited through printed advertisement. 16 age- and BMI-matched participants were invited for a follow-up study to analyze total CIEE after a single bout of cold exposure. All participants were screened for their medical history and underwent physical examination. Participants that fulfilled the inclusion criteria (28) were enrolled in the study after providing written informed consent. This study is registered at ClinicalTrials.gov (NCT02381483) and has been approved by the Ethics Committee of the Medical University of Vienna (no. 1032/2013). The study was conducted at the Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna in accordance with the principles of the Declaration of Helsinki.

BATneg individuals lack [18F]-FDG uptake only in BAT but not in other metabolic tissues. (A) Representative [18F]-FDG PET image of a BATpos participant before (left) and after (right) cold exposure. (B) Representative [18F]-FDG PET image of a BATneg participant before (left) and after (right) cold exposure. (C) [18F]-FDG uptake in BAT before and after cold exposure in BATpos and BATneg participants. (D) Changes in mean norepinephrine concentrations in BATpos (black line) and BATneg (interrupted gray line) before and after cold exposure. ***P < 0.001, **P < 0.01, *P < 0.05. [18F]-FDG PET/CT, 18F-fluorodeoxyglucose positron emission tomography combined with computed tomography; BAT, brown adipose tissue; neg, negative; pos, positive; SUV, standardized uptake value.
Figure 1.

BATneg individuals lack [18F]-FDG uptake only in BAT but not in other metabolic tissues. (A) Representative [18F]-FDG PET image of a BATpos participant before (left) and after (right) cold exposure. (B) Representative [18F]-FDG PET image of a BATneg participant before (left) and after (right) cold exposure. (C) [18F]-FDG uptake in BAT before and after cold exposure in BATpos and BATneg participants. (D) Changes in mean norepinephrine concentrations in BATpos (black line) and BATneg (interrupted gray line) before and after cold exposure. ***P < 0.001, **P < 0.01, *P < 0.05. [18F]-FDG PET/CT, 18F-fluorodeoxyglucose positron emission tomography combined with computed tomography; BAT, brown adipose tissue; neg, negative; pos, positive; SUV, standardized uptake value.

Study design

Study participants were examined at 3 separate study visits in the morning after an overnight fast (>10 hours) (28). At study visit 1, all participants underwent the first [18F]-FDG PET/CT scan at RT (23°C) to detect any basal BAT activity. Participants were instructed to wear comfortable clothing and stay warm until arriving in the hospital. At study visit 2 (no longer than 14 days after study visit 1), study participants underwent the second [18F]-FDG PET/CT scan, this time after cold exposure, to detect cold-induced BAT activity. A personalized cooling protocol (28) was applied using a water-perfused cooling vest (CoolShirt Systems, Stockbridge, Georgia, USA) similar to as previously described (29). The water temperature was kept slightly above the shivering temperature, and muscle activity was monitored by electromyography (OT Bioelettronica, Torino, Italy). After 90 minutes of cold exposure, [18F]-FDG was administered, and cold exposure was continued for another 60 minutes until PET/CT acquisition started between 10 am and 11am. Participants that had [18F]-FDG-positive BAT depots after cold exposure were deemed BAT positive, whereas participants that had no significant [18F]-FDG uptake at baseline and after cold exposure, respectively, were considered BAT negative. All PET/CT scans were performed during the cool season (fall to spring) to avoid any seasonal influence on BAT activity.

At study visit 3, eight BAT-positive and 8 BAT-negative participants underwent repeated indirect calorimetry examinations before, during, and after cold exposure to determine the magnitude and duration of CIEE after a single bout of cold exposure. All examinations started between 8 am and 9 am, and all participants were in fasting condition (>10 hours). After a 30-minute resting phase, the basal resting energy expenditure was determined by indirect calorimetry. Next, participants were cooled for 90 minutes using the same temperature settings as during study visit 2. During the final 30 minutes of the cooling period, another indirect calorimetry was performed. Thereafter, during rewarming, indirect calorimetry was repeated every hour until energy expenditure had returned to the baseline (resting) levels. Blood was taken before and after 90 minutes of cold exposure.

PET/CT scanning protocol

Each participant received a dose of 2.5 MBq kg-1 body weight [18F]-FDG (at least 150 MBq and a maximum of 350 MBq). A combined PET/CT acquisition started 60 minutes after the administration of the radioactive isotope on a Siemens Biograph 64 True Point scanner (Siemens Healthcare Sector, Erlangen, Germany). First, a low dose CT scan (120 kV, 50 mAs) was performed for scatter and attenuation correction as well as for precise anatomical localization of the BAT depots, followed by PET acquisition in 3D mode of 3 minutes/bed position. Both PET and CT were acquired from the base of the skull to mid thigh.

PET/CT analysis

PET/CT scans were analyzed using Hermes Hybrid 3D Viewer (Hermes Medical Solutions, Stockholm, Sweden). The identification of the BAT depots was performed using a multithreshold model in accordance with the Brown Adipose Reporting Criteria in Imaging Studies (30). Briefly, the regions of interest were delineated in the axial fusion images. Only regions of interest located in regions with a CT radio density of –180 to –10 Hounsfield units and with a minimal standardized uptake value higher than 2 were considered for brown fat quantification.

Indirect calorimetry

The resting energy expenditure was measured by computerized open circuit indirect calorimetry (Quark RMR, Cosmed, Rome, Italy). Briefly, after a resting phase of 30 minutes, participants were studied for 30 minutes in a supine position in a quiet room with a constant temperature of about 23° to 24°C, or during cold exposure. The participants were instructed to stay motionless and awake during the entire study period. The results of the last 20 minutes of each experiment were averaged. CIEE was calculated as the percentage change in total energy expenditure between RT and cold exposure.

PUFA and oxylipin extraction

Blood was drawn in cooled ethylenediaminetetraacetic acid-coated tubes and was immediately centrifuged at 4000 rpm at 4°C. After centrifugation, the samples were frozen at –80°C until use in further experiments. Frozen plasma samples were thawed on ice. In order to precipitate proteins, 500 µL plasma were mixed with 2.0 mL ethanol (abs 99%; AustroAlco) including 10 to 100 nM of each internal standard (12S-, 15S- hydroxyeicosatetraenoic acids [HETE]-d8, 5-Oxo-ETE-d7,11.12- dihydroxyeicosatrienoic acid [DiHETrE]-d11, prostaglandin E (PGE)-d4, 20-HETE-d6; Cayman Europe, Tallinn, Estonia) and stored over night at –20°C. Samples were centrifuged (30 minutes, 5000 rpm, 9 deceleration, 7 acceleration, 4°C), and the supernatant transferred to a 15 mL Falcon tube. Ethanol was evaporated via vacuum centrifugation at 37°C until the original sample volume was restored. Samples were loaded on preconditioned 30 mg mL-1 StrataX solid phase extraction columns (Phenomenex, Torrance, CA, USA), using Pasteur pipettes, washed with 2 mL mass spectrometry grade water, and lipids were eluted with 500 µL ice cold methanol (MeOH abs.; VWR International, Vienna, Austria) containing 2% formic acid (FA; Sigma-Aldrich). MeOH was evaporated using a nitrogen stream at RT, and samples were reconstituted in 150 µL of reconstitution buffer (H2O/acetonitrile/MeOH + 0.2% FA - 65:31.5:3.5), containing another set of 10 to 100 nM of internal standards (5S-HETE-d8, 14.15-DiHETrE-d11, 8-iso-PGF2a-d4; Cayman Europe, Tallinn, Estonia).

Liquid chromatography with tandem mass spectrometry method

Analytes were separated using a Thermo Scientific Vanquish (ultra high-performance liquid chromatography) system and a Kinetex C18—column (2.6 μm C18 100 Å, LC Column 150 × 2.1 mm; Phenomenex). Applying a 20-minute gradient flow method, starting at 35% solvent B steadily increasing to 90% B (1-10 minutes), going up to 99% B in 0.5 minutes and held for 5 minutes. Solvent B was dropped to 35% in 0.5 min and the column equilibrated for 4 minutes. With an injection volume of 20 µL, flow rate was kept at 200 µL minute-1, and column oven temperature was set to 40°C. Solvent A: 0.2% FA in H2O, eluent B: ratio of acetonitrile to MeOH (90:10) containing 0.2% FA. Mass spectrometric analysis was performed with a Q Exactive HF Quadrupole-Orbitrap mass spectrometer (Thermo Fisher Scientific, Austria), equipped with a heated electrospray ionization source for negative ionization. Mass spectra were recorded operating from 250 to 700 m/z at a resolution of 60 000 @ 200 m/z on MS1 (full scan) level. The 2 most abundant precursor ions were selected for fragmentation (higher energy collisional dissociation 24 normalized collision energy), preferably molecules from an inclusion list that contained 31 m/z values specific for PUFAs and oxylipins (28). MS2 (fragment ion scan) was operated at a resolution of 15 000 @ 200 m/z. For negative ionization, a spray voltage of 2.2 kV and a capillary temperature of 253°C were applied, with the sheath gas set to 46 and the auxiliary gas to 10 arbitrary units. For the liquid chromatography–mass spectrometry (LC-MS) data interpretation, raw files generated by the Q-Exactive Orbitrap were analyzed manually using Thermo Xcalibur 4.1.31.9 (Qual browser), comparing reference spectra from the Lipid Maps depository library from July 2018 (3). For peak integration, the software TraceFinder (version 4.1, Thermo Scientific, Austria) was used.

Statistics

Statistical analyses were performed in R version 3.5.2 and CRAN or Bioconductor R-packages as needed (31). P values were corrected for multiple testing according to the false discovery rate (FDR) method (also known as the Benjamini–Hochberg procedure (32), and FDR values <10% were considered as statistically significant. Analytes abundance values were log2 transformed to get near normal distributions. Lipids with missing values in more than one-third of samples were removed (4/100), and the remaining missing values imputed with R-package mice version 3.4.0 using the predictive mean matching method (33). Differences of analytes were calculated in a regression model using Bioconductor package limma version 3.38.3 (34), and P values were corrected for multiple testing. Pair-matched information and the confounders sex and BMI (represented by the first principal component, PC1, explaining 82.6% of variance) were included into the limma model. Differences for the following contrasts were estimated: (a) all BATpos versus BATneg samples, (b) BATpos versus BATneg samples from RT controls, (c) cold exposure versus RT (paired), (d) cold exposure versus RT (paired) in BATpos and, (e) cold exposure versus RT (paired) in BATneg participants. Clinical parameters and laboratory values were also log2 transformed, and the following contrasts were estimated (corrected also for PC1 comprising sex and BMI information): (a) cold exposure versus RT (paired) and (b) BATpos versus BATneg at RT.

To integrate lipid levels with clinical parameters and laboratory values, lipid abundance values were first used for graphical Gaussian modeling (GGM) using R-package GGMselect (0.1–12.1) (35). GGM, also known as covariance selection or concentration graph modeling, is used to build association networks from abundance data. The principle behind GGM is to use partial correlations as a measure of independence of any 2 analytes, which allows distinguishing direct from indirect interactions. The tuning parameter K for the penalty function was varied between 1 and 4 with 0.5 increments, and the function selectFast [family = c(“C01,” “LA,” “EW”)] was used for optimizing the model employing the C01 algorithm, the Lasso-And (LA), and the adaptive lasso (EW) families (Vignette of R-package GGmselect). To select the optimal K value, the GGM-obtained subclusters were analyzed together as analytes-set for significant associations with above defined contrasts employing the procedure “gene set analysis” with raw P values and regulation-directions (logFC-values) from the corresponding single analytes-limma analyses using the runGSA function of R-package piano version 1.22.0 (36). The K value with the highest relative number of significant analytes-sets (= GGM subclusters) across all contrasts (based on the absolute number of subclusters) was used for further analyses, that is, K = 2, yielding 20 subclusters containing between 2 and 8 lipid species.

We correlated significantly regulated lipids or lipid clusters with clinical parameters and laboratory values from cold exposure and RT respectively, by modeling the different comparisons (contrasts) with limma (corrected for the PC1, representing sex and BMI information). We used all significant (FDR < 10%) associations to build the final association networks.

Finally, these associations together with the subcluster information were plotted as rich annotated networks for each contrast. Node colors (green-red) represent the log2 fold-change between groups according to corresponding contrast (large node meaning significant; small node meaning not significant). The edge color (yellow-blue) represents the correlation between oxylipins and clinical parameters or laboratory values (labeled with “c” if RT control correlations were described and not labeled if cold exposure correlations were described). Edges in red, green, and gray between oxylipins indicate the subcluster structure (ie, whether the oxylipins were connected by graphical Gaussian modeling). The line thickness indicates the statistical significance of the subclusters between different conditions (BATneg vs BATpos, RT vs cold exposure) considering all 5 contrasts (bold line: significant; thin line: not significant; red: positive correlation; green: negative correlation; and gray, direction undefined).

Prediction of lipid candidates using CFM-ID 3.0

This analysis strategy was used to identify DiHOME candidates in plasma samples. Peaks with an exact mass of 313.2384 and a fragmentation pattern similar to 9,10-DiHOME were subjected to CFM-ID 3.0 as the identification tool (37), searching for candidate molecules following the protocol based on MS2-spectra obtained with fragmentation energies of 10, 20, and 40 eV. This was performed for the following molecules: DiHOME_10.81, 313_10.40, 313_10.45, and 313_10.75. The CFM-ID 3.0 can search against either HMDB 4.0 (38), other lipid databases, or manually generated candidate lists. Using HMDB 4.0 suggested 9,10-DiHOME and 12,13-DiHOME as potential candidate molecules for the DiHOMEs listed above. However, HMDB 4.0 contains only 9,10-DiHOME and 12,13-DiHOME and no other DiHOMEs. We can exclude that DiHOME_10.81, 313_10.40, 313_10.45, 313_10.75 were 9,10-DiHOME or 12,13-DiHOME as we have assigned these molecules in our dataset to the retention times 8.56 and 8.40, respectively, based on the lipid standards. However, characteristic fragment ions strongly suggested that DiHOMEs_10.81, 313_10.40, 313_10.45, 313_10.75 were indeed DiHOMEs. Therefore, an extended candidate list was generated manually using SMILES code for all DiHOMEs available on LIPID MAPS (28). MS2 peak list files containing exact masses and intensity values were exported from Xcalibur 4.1.31.9 (Qual browser) for each DiHOME candidate peak, respectively. Compound identification criteria included ESI spectra in negative ion mode, Jaccard as the scoring function, and a mass tolerance of 20 ppm. To control for the reliability of this software, the experimental MS data obtained for 9,10-DiHOME were processed and resulted in the correct identification of 9,10-DiHOME (28).

Results

Identification of BATpos versus BATneg participants

In order to study the relevance of active BAT for CIEE and cold-regulated lipid species, we performed [18F]-FDG PET/CT scans in 30 healthy normal-weight individuals at RT and after 2.5 hours of moderate cold exposure. Using this approach, we identified 16 age- and BMI-matched participants (28) of whom 8 had detectable BAT [18F]-FDG uptake after cold exposure (BATpos) and 8 that did not have any significant BAT activity at RT or at cold-stimulated conditions (BATneg, Fig. 1A-C). Metabolic parameters such as circulating glucose, triglycerides, cholesterol, and BAT activity parameters were similar in both groups at RT and after cold exposure (28). The water temperatures of the cooling garment did not differ between BATpos and BATneg (28), whereas the cold-induced increase of circulating norepinephrine concentrations was even higher in BATneg than BATpos participants (Fig. 1D), emphasizing that insufficient cooling or lower sympathetic tone was not the reason for a lack of [18F]-FDG uptake in BATneg participants.. In contrast to BAT, [18F]-FDG uptake was similar between groups in other metabolically relevant tissues including liver, subcutaneous abdominal WAT, and skeletal muscle (pectoralis major and erector spinae) (28).

CIEE is increased in BATpos but not in BATneg individuals and reverses 150 minutes after discontinuation of cold exposure

To better understand the importance of the BAT status for CIEE, we quantified whole body energy expenditure in BATpos and BATneg participants before, during, and after cold exposure. At baseline, resting energy expenditure and the respiratory quotient were similar in both groups (Fig. 2A, B). However, CIEE increased significantly in BATpos reaching a maximum during the 90-minute cold exposure period. In contrast, energy expenditure was unaltered in BATneg in response to cold (Fig. 2C), suggesting that BAT status is the primary determinant of cold-mediated energy consumption. Given the lack of information about the duration of cold-induced BAT activity, we continued to examine energy expenditure after the discontinuation of cold exposure. After cold exposure, CIEE slowly declined to baseline levels within 150 minutes in BATpos individuals, whereas BATneg individuals did not experience any changes in energy expenditure during the entire test period (Fig. 2C). The incremental area under the curve of energy consumption during cooling and rewarming was approximately 20 kcal/day in BATpos and, therefore, significantly higher compared with BATneg (Fig. 2D).

Cold-induced energy expenditure (CIEE) differs significantly between BATpos and BATneg individuals. (A) Respiratory quotient (RQ) and resting energy expenditure (REE) (B) of BATpos (black bars) and BATneg participants (gray bars) at room temperature. (C) Change of CIEE over time during and after the cessation of the cold stimulus in BATpos (black line) and BATneg (gray line) participants. CIEE was calculated as the percentual increase in REE after cold exposure as compared with baseline. (D) Integrated energy consumption following a single bout of 90-minute cold exposure. ***P < 0.001, **P < 0.01, *P < 0.05. BAT, brown adipose tissue; iAUC, incremental area under the curve; min, minute(s); neg, negative; pos, positive; REE, RQ,
Figure 2.

Cold-induced energy expenditure (CIEE) differs significantly between BATpos and BATneg individuals. (A) Respiratory quotient (RQ) and resting energy expenditure (REE) (B) of BATpos (black bars) and BATneg participants (gray bars) at room temperature. (C) Change of CIEE over time during and after the cessation of the cold stimulus in BATpos (black line) and BATneg (gray line) participants. CIEE was calculated as the percentual increase in REE after cold exposure as compared with baseline. (D) Integrated energy consumption following a single bout of 90-minute cold exposure. ***P < 0.001, **P < 0.01, *P < 0.05. BAT, brown adipose tissue; iAUC, incremental area under the curve; min, minute(s); neg, negative; pos, positive; REE, RQ,

BATpos have a distinct systemic PUFA and oxylipin profile compared with BATneg individuals

Next we investigated whether (a) the presence of BAT defined the systemic lipid pool in humans and (b) cold exposure altered the PUFA and oxylipin concentrations distinctly in BATpos and BATneg. Therefore, blood plasma was collected from all 16 participants before and after a 90-minute cold exposure period for subsequent lipid extraction and analysis by LC-MS/MS. Molecules were identified by exact mass, MS2 fragmentation pattern, and chromatographic retention time. A panel of 31 commercially available PUFA and oxylipin standards was included in the analyses, supporting the exact assignment of the corresponding molecules in the plasma samples (28). However, an additional 67 molecules were identified as oxylipins without more detailed structural specifications (28). These molecules were annotated according to their mass and retention time, that is, 319_10.31 corresponds to a molecule with 319 Da and a retention time of 10.31 minutes.

The abundance of detected plasma PUFAs and oxylipins was very heterogeneous, covering a concentration range of more than 3 orders of magnitude (28). Various molecules including arachidonic acid, 14,15-DiHETrE, and 11-HEPE showed less interindividual variation than others such as 5-HETE, 11-HETE, and (15d-) PGJ2.

A principle component analysis indicated that the general variance of analytes between individuals was larger than the variance between groups (28). For further statistical analysis, we thus performed analytes-set analyses and a coassociation network analysis as described in Materials and Methods.

This analysis strategy identified 4 coassociation clusters that were distinctly regulated between BATpos and BATneg participants (Fig. 3A). The first cluster included 20-HETE and 2 DiHETrEs, all of them significantly upregulated in BATpos individuals and all representing known cytochrome P450 products (39). Therefore, we termed this cluster 1, P450 products. The second cluster was also increased in BATpos participants and included the ω3 fatty acids stearidonic acid, eicosapentaenoic acid (EPA), (40) and was thus designated as ω3 precursors. The third cluster was decreased in BATpos versus BATneg individuals and was named cluster 3, HETEs, because it contained only HETEs formed by lipoxygenases (5, 12, and 15-isoform) or via free oxygen radicals (9 and 11 isoform). A fourth cluster comprising mainly PUFAs was also distinctly regulated between BATpos and BATneg participants; however, this analytical strategy could not distinguish between down- or upregulation.

Systemic PUFAs and oxylipins differ between BATpos and BATneg and correlate with clinical BAT parameters. Association networks of (A) various lipids in BATpos versus BATneg participants at room temperature and of (B) various lipid species and clinical parameters in BATpos versus BATneg participants before cold exposure. Gray ellipses indicate significantly regulated lipid clusters. Node colors “green-red” represent log2 fold-change of individual lipids between groups (red, high in BATpos). Colored molecule names indicate significant differences between BATpos and BATneg. The color code yellow-blue represents the correlation between various lipids and clinical BAT parameters or laboratory values (red, positive correlation; green, negative correlation). BAT, brown adipose tissue; FA, fatty acid; HETE, hydroxyeicosatetraenoic acid; neg, negative; pos, positive; PUFA, polyunsaturated fatty acid.
Figure 3.

Systemic PUFAs and oxylipins differ between BATpos and BATneg and correlate with clinical BAT parameters. Association networks of (A) various lipids in BATpos versus BATneg participants at room temperature and of (B) various lipid species and clinical parameters in BATpos versus BATneg participants before cold exposure. Gray ellipses indicate significantly regulated lipid clusters. Node colors “green-red” represent log2 fold-change of individual lipids between groups (red, high in BATpos). Colored molecule names indicate significant differences between BATpos and BATneg. The color code yellow-blue represents the correlation between various lipids and clinical BAT parameters or laboratory values (red, positive correlation; green, negative correlation). BAT, brown adipose tissue; FA, fatty acid; HETE, hydroxyeicosatetraenoic acid; neg, negative; pos, positive; PUFA, polyunsaturated fatty acid.

Next, we included clinical BAT parameters such as CIEE, mean standardized uptake value, and BAT volume in the model and found that cluster 1, containing the cytochrome P450 products 20-HETE and 11,12- and 14,15-DiHETrE, was significantly correlated with CIEE in BATpos participants. The PPAR-γ ligand 15-deoxy-Δ12,14-prostaglandin J2 as well as 315_9.55 and 313_12.63 were positively associated with BAT volume and total lesion glycolysis, a marker of BAT activity (Fig. 3B).

To further address the effect of the BAT status on PUFA and oxylipin profiles, we performed a comparative analysis of such circulating lipids in BATpos and BATneg individuals at RT. By using a linear model analysis corrected for sex and BMI, with subsequent correction for multiple testing, and applying a threshold of an FDR < 10%, we identified 36 lipids that were significantly different between BATpos and BATneg participants at baseline, including 19 upregulated and 17 downregulated molecules (Table 1, Fig. 3A). The lipid molecule with the highest relative expression in BATpos compared with BATneg participants was DiHOME_10.81, which belongs to a class of BAT-derived lipokines (BATokines) previously associated with increased brown adipocyte and skeletal muscle fatty acid uptake (23, 26). The matching molecular mass and isotope pattern, as well as characteristic fragment ions at 295.2 and 251.2 Da, strongly suggest that the molecule that we identified as DiHOME_10.81 indeed belongs to the family of DiHOMEs. Previously, 12,13-DiHOME has been reported as an oxylipin highly expressed in BAT (26). However, DiHOME_10.81 is distinct from 12,13-DiHOME, which we also identified here using a lipid standard with the retention time of 8.4 minutes. Despite thorough analysis of the mass spectrometry data with the specialized software CMF-ID 3.0 and due to the lack of commercially available standards, the exact molecular characterization of DiHOME_10.81 and other candidate molecules of the DiHOME family was not yet possible (28). Other known molecules that were significantly increased in BATpos compared to BATneg included the anti-inflammatory EPA and stearidonic acid and the recently identified BATokine 12-HEPE, which improve glucose metabolism in vitro and in vivo (40). In contrast, a number of proinflammatory arachidonic acid metabolites such as 5-oxo-eicosatetraenoic acid and a number of HETEs were decreased in BATpos individuals (Table 1).

Table 1.

Differences in the abundance of various lipid species between BATpos and BATneg individuals. Log2 fold change (logFC) and false discovery rates (FDR) for comparisons between BATpos and BATneg at baseline (before cold exposure)

IDLogFCFDR
DiHOME_10.818.250.016
335_9.862.760.015
EPA2.010.007
317_8.691.810.046
Stearidonic acid_11.631.410.085
3.HOTrE_9.391.410.007
HpODE_10.041.410.007
319_11.071.380.010
12-HEPE1.370.007
265_9.081.240.029
HETE_9.570.990.012
313_10.750.940.029
6trans-LTB4_7.980.930.086
11,12-DiHETrE0.780.025
HpODE_7.710.780.077
HHTrE_9.010.770.098
14,15-DiHETrE0.770.015
335_9.770.730.085
20-HETE0.580.094
313_10.45–0.710.098
15S-HETrE_10.67–1.050.055
13,14-dihydro-15-keto-PGF2a_6.3–1.400.077
Linoleic acid–1.650.012
9/10-DiHOME–2.100.002
15-HETE–2.180.001
313_12.27–2.190.015
343_10.07–2.300.009
319_10.31–2.880.001
12-HETE–2.890.001
metPGE2_6.35–2.910.067
5-HETE–2.940.007
11-HETE–3.100.002
9-HETE–3.110.001
15S-HETrE_10.77–3.280.019
14-HDoHE and 10-HDoHE–3.750.010
5-Oxo-ETE–3.750.094
IDLogFCFDR
DiHOME_10.818.250.016
335_9.862.760.015
EPA2.010.007
317_8.691.810.046
Stearidonic acid_11.631.410.085
3.HOTrE_9.391.410.007
HpODE_10.041.410.007
319_11.071.380.010
12-HEPE1.370.007
265_9.081.240.029
HETE_9.570.990.012
313_10.750.940.029
6trans-LTB4_7.980.930.086
11,12-DiHETrE0.780.025
HpODE_7.710.780.077
HHTrE_9.010.770.098
14,15-DiHETrE0.770.015
335_9.770.730.085
20-HETE0.580.094
313_10.45–0.710.098
15S-HETrE_10.67–1.050.055
13,14-dihydro-15-keto-PGF2a_6.3–1.400.077
Linoleic acid–1.650.012
9/10-DiHOME–2.100.002
15-HETE–2.180.001
313_12.27–2.190.015
343_10.07–2.300.009
319_10.31–2.880.001
12-HETE–2.890.001
metPGE2_6.35–2.910.067
5-HETE–2.940.007
11-HETE–3.100.002
9-HETE–3.110.001
15S-HETrE_10.77–3.280.019
14-HDoHE and 10-HDoHE–3.750.010
5-Oxo-ETE–3.750.094

Abbreviations: 6 metPGE2, 6 met prostaglandin E2; 6-trans-LTB, 6-trans-leucotriene B; 13,14-dihydro-15-keto-PGF2a, Prostaglandin F; DiHETrE, dihydroxyeicosatrienoic acid; HDoHE, hydroxy-docosahexaenoic acid; HETre, hydroxy-eicosatrienoic acid; HHTrE, hydroxy-heptadecatrienoic acid; HOTre, hydroxy-octadecatrienoic acid; HpODE, hydroperoxy-octadecadienoic acid; ID, identification; neg, negative; pos, positive.

Table 1.

Differences in the abundance of various lipid species between BATpos and BATneg individuals. Log2 fold change (logFC) and false discovery rates (FDR) for comparisons between BATpos and BATneg at baseline (before cold exposure)

IDLogFCFDR
DiHOME_10.818.250.016
335_9.862.760.015
EPA2.010.007
317_8.691.810.046
Stearidonic acid_11.631.410.085
3.HOTrE_9.391.410.007
HpODE_10.041.410.007
319_11.071.380.010
12-HEPE1.370.007
265_9.081.240.029
HETE_9.570.990.012
313_10.750.940.029
6trans-LTB4_7.980.930.086
11,12-DiHETrE0.780.025
HpODE_7.710.780.077
HHTrE_9.010.770.098
14,15-DiHETrE0.770.015
335_9.770.730.085
20-HETE0.580.094
313_10.45–0.710.098
15S-HETrE_10.67–1.050.055
13,14-dihydro-15-keto-PGF2a_6.3–1.400.077
Linoleic acid–1.650.012
9/10-DiHOME–2.100.002
15-HETE–2.180.001
313_12.27–2.190.015
343_10.07–2.300.009
319_10.31–2.880.001
12-HETE–2.890.001
metPGE2_6.35–2.910.067
5-HETE–2.940.007
11-HETE–3.100.002
9-HETE–3.110.001
15S-HETrE_10.77–3.280.019
14-HDoHE and 10-HDoHE–3.750.010
5-Oxo-ETE–3.750.094
IDLogFCFDR
DiHOME_10.818.250.016
335_9.862.760.015
EPA2.010.007
317_8.691.810.046
Stearidonic acid_11.631.410.085
3.HOTrE_9.391.410.007
HpODE_10.041.410.007
319_11.071.380.010
12-HEPE1.370.007
265_9.081.240.029
HETE_9.570.990.012
313_10.750.940.029
6trans-LTB4_7.980.930.086
11,12-DiHETrE0.780.025
HpODE_7.710.780.077
HHTrE_9.010.770.098
14,15-DiHETrE0.770.015
335_9.770.730.085
20-HETE0.580.094
313_10.45–0.710.098
15S-HETrE_10.67–1.050.055
13,14-dihydro-15-keto-PGF2a_6.3–1.400.077
Linoleic acid–1.650.012
9/10-DiHOME–2.100.002
15-HETE–2.180.001
313_12.27–2.190.015
343_10.07–2.300.009
319_10.31–2.880.001
12-HETE–2.890.001
metPGE2_6.35–2.910.067
5-HETE–2.940.007
11-HETE–3.100.002
9-HETE–3.110.001
15S-HETrE_10.77–3.280.019
14-HDoHE and 10-HDoHE–3.750.010
5-Oxo-ETE–3.750.094

Abbreviations: 6 metPGE2, 6 met prostaglandin E2; 6-trans-LTB, 6-trans-leucotriene B; 13,14-dihydro-15-keto-PGF2a, Prostaglandin F; DiHETrE, dihydroxyeicosatrienoic acid; HDoHE, hydroxy-docosahexaenoic acid; HETre, hydroxy-eicosatrienoic acid; HHTrE, hydroxy-heptadecatrienoic acid; HOTre, hydroxy-octadecatrienoic acid; HpODE, hydroperoxy-octadecadienoic acid; ID, identification; neg, negative; pos, positive.

Cold exposure significantly increases PUFA and oxylipin concentrations only in BATpos but not in BATneg individuals

The direct comparative analysis of cold-mediated changes in lipid species revealed 11 molecules that were significantly altered by cold exposure. All of them were cold-induced but only in BATpos and not in BATneg individuals (Table 2). Particularly, PUFAs such as docosahexaenoic acid (DHA) and docosapentaenoic acid (DPA) as well as members of cluster 2 ω3 precursors were elevated in BATpos individuals in response to cold exposure (Fig. 4A,B, Table 2) Fig. 4 depicts the unprocessed signal intensity normalized area under the curve (nAUC) values from the LC-MS experiments, whereas Table 2 lists the results obtained after data processing, including correction for sex and BMI. This list also included the ω3 oxylipin 12-HEPE, which was not only higher in BATpos compared with BATneg individuals (Table 1) but significantly increased with cold exposure, supporting the recent notion of 12-HEPE as a cold-regulated BAT-derived lipokine (40). In addition, 12-HEPE was one of only 8 lipids that were elevated in BATpos at RT and increased even further in response to cold exposure (Fig. 5) as would be expected from a typical BATokine. Other lipids in this group were the ω3 PUFAs and cluster 3 members EPA and stearidonic acid (Fig. 5). Interestingly, some molecules including the PPAR-γ agonist PGJ2 and its metabolite 15d-PGJ2 showed oppositional regulation; they were nonsignificantly increased in BATpos but reduced in BATneg after cold exposure (28).

Table 2.

Significant cold-induced changes in circulating fatty acids and oxylipins occurred only in BATpos but not BATneg individuals. LogFC and FDR values for comparisons before versus after cold exposure in BATpos and BATneg, respectively

BATposBATneg
IDLogFCFDRLogFCFDR
12-HETE0.770.0230.410.212
Oleic acid0.720.0920.120.874
DPA0.720.0920.650.187
305_13.040.670.0960.230.692
Stearidonic acid_11.630.630.0960.380.336
DPA_12.970.540.0920.290.336
Linoleic acid0.500.0960.360.267
12-HEPE0.490.0580.180.425
DHA0.460.0920.460.187
AA0.450.0960.440.187
319_11.450.320.0920.260.212
BATposBATneg
IDLogFCFDRLogFCFDR
12-HETE0.770.0230.410.212
Oleic acid0.720.0920.120.874
DPA0.720.0920.650.187
305_13.040.670.0960.230.692
Stearidonic acid_11.630.630.0960.380.336
DPA_12.970.540.0920.290.336
Linoleic acid0.500.0960.360.267
12-HEPE0.490.0580.180.425
DHA0.460.0920.460.187
AA0.450.0960.440.187
319_11.450.320.0920.260.212

Abbreviations: BAT, brown adipose tissue; DHA, docosahexaenoic acid; DPA, docosapentaenoic acid; FDR, false discovery rate; HEPE, hydroxyeicosapentaenoic acid; HETE, hydroxyeicosatetraenoic acid; ID, identification; LogFC, log2 fold change; neg, negative; pos, positive.

Table 2.

Significant cold-induced changes in circulating fatty acids and oxylipins occurred only in BATpos but not BATneg individuals. LogFC and FDR values for comparisons before versus after cold exposure in BATpos and BATneg, respectively

BATposBATneg
IDLogFCFDRLogFCFDR
12-HETE0.770.0230.410.212
Oleic acid0.720.0920.120.874
DPA0.720.0920.650.187
305_13.040.670.0960.230.692
Stearidonic acid_11.630.630.0960.380.336
DPA_12.970.540.0920.290.336
Linoleic acid0.500.0960.360.267
12-HEPE0.490.0580.180.425
DHA0.460.0920.460.187
AA0.450.0960.440.187
319_11.450.320.0920.260.212
BATposBATneg
IDLogFCFDRLogFCFDR
12-HETE0.770.0230.410.212
Oleic acid0.720.0920.120.874
DPA0.720.0920.650.187
305_13.040.670.0960.230.692
Stearidonic acid_11.630.630.0960.380.336
DPA_12.970.540.0920.290.336
Linoleic acid0.500.0960.360.267
12-HEPE0.490.0580.180.425
DHA0.460.0920.460.187
AA0.450.0960.440.187
319_11.450.320.0920.260.212

Abbreviations: BAT, brown adipose tissue; DHA, docosahexaenoic acid; DPA, docosapentaenoic acid; FDR, false discovery rate; HEPE, hydroxyeicosapentaenoic acid; HETE, hydroxyeicosatetraenoic acid; ID, identification; LogFC, log2 fold change; neg, negative; pos, positive.

Cold exposure raises PUFA and oxylipin levels only in BATpos individuals. A) Cold-induced changes in the abundance (nAUC) of AA, 12-HETE, DPA, and oleic acid in BATpos and BATneg, respectively. Unprocessed abundance values (nAUC) determined from LC-MS experiments are depicted. Fold-change values may thus slightly differ from processed and corrected values reported in Tables 1 and 2; Significance levels were used as given in Tables 1 and 2 and are corrected for age, sex, pairedness, cold exposure, and BAT status (see Material and Methods). * FDR < 10%, ** FDR < 5%. B) Selected association networks showing distinct effects of cold exposure on lipid clusters in BATpos and BATneg individuals. Red molecules were cold induced; red edges indicate lipid clusters that were increased after cold exposure. AA, arachidonic acid; BAT, brown adipose tissue; DPA, docosapentaenoic acid; FDR, false discovery rate; HEPE, hydroxyeicosapentaenoic acid; HETE, hydroxyeicosatetraenoic acid; neg, negative; pos, positive; PUFA, polyunsaturated fatty acid.
Figure 4.

Cold exposure raises PUFA and oxylipin levels only in BATpos individuals. A) Cold-induced changes in the abundance (nAUC) of AA, 12-HETE, DPA, and oleic acid in BATpos and BATneg, respectively. Unprocessed abundance values (nAUC) determined from LC-MS experiments are depicted. Fold-change values may thus slightly differ from processed and corrected values reported in Tables 1 and 2; Significance levels were used as given in Tables 1 and 2 and are corrected for age, sex, pairedness, cold exposure, and BAT status (see Material and Methods). * FDR < 10%, ** FDR < 5%. B) Selected association networks showing distinct effects of cold exposure on lipid clusters in BATpos and BATneg individuals. Red molecules were cold induced; red edges indicate lipid clusters that were increased after cold exposure. AA, arachidonic acid; BAT, brown adipose tissue; DPA, docosapentaenoic acid; FDR, false discovery rate; HEPE, hydroxyeicosapentaenoic acid; HETE, hydroxyeicosatetraenoic acid; neg, negative; pos, positive; PUFA, polyunsaturated fatty acid.

Potential BAT-derived lipids are elevated in BATpos and after cold exposure. Venn diagram showing the number of overlapping lipids that were significantly increased (“up in BATpos”) or decreased (“down in BATpos”) in BATpos compared with BATneg individuals as well as lipids that were significantly upregulated (“up with cold”) or downregulated (“down with cold”) in response to cold exposure in both cohorts. All comparisons were corrected for age, sex, and BAT status, the latter only for cold exposure comparisons. Eight lipids were significantly higher in BATpos and increased by cold exposure, particularly suggesting BAT as a potential source. BAT, brown adipose tissue; DiHETrE, dihydroxyeicosatrienoic acid; HEPE, hydroxyeicosapentaenoic acid; HETE, hydroxyeicosatetraenoic acid; neg, negative; pos, positive.
Figure 5.

Potential BAT-derived lipids are elevated in BATpos and after cold exposure. Venn diagram showing the number of overlapping lipids that were significantly increased (“up in BATpos”) or decreased (“down in BATpos”) in BATpos compared with BATneg individuals as well as lipids that were significantly upregulated (“up with cold”) or downregulated (“down with cold”) in response to cold exposure in both cohorts. All comparisons were corrected for age, sex, and BAT status, the latter only for cold exposure comparisons. Eight lipids were significantly higher in BATpos and increased by cold exposure, particularly suggesting BAT as a potential source. BAT, brown adipose tissue; DiHETrE, dihydroxyeicosatrienoic acid; HEPE, hydroxyeicosapentaenoic acid; HETE, hydroxyeicosatetraenoic acid; neg, negative; pos, positive.

Discussion

Despite accumulating evidence that BAT activation is associated with increased energy expenditure, improved insulin sensitivity, and lipid clearance in humans (11, 12, 17-19), direct comparison of substrate metabolism and lipid species in matched individuals with and without active BAT depots has not been studied yet. Here we demonstrated that CIEE increased by 15% in BATpos, with no changes in BATneg participants, suggesting that BAT is the predominant contributor to CIEE (Fig. 2). This notion is not only supported by the absence of CIEE in BATneg individuals but also the lack of cold-induced FDG uptake in skeletal muscle in the entire study cohort (28). Numerous studies have previously shown an association between BAT activation and increased energy expenditure in humans (7, 9-11), however it had remained unclear how long this effect persisted once BAT was activated. We showed here that energy expenditure was elevated for approximately 150 minutes after discontinuation of the cold challenge and thus culminates in an additional energy loss of 20 kcal/day in BATpos, which is in line with a previous report studying oxidative metabolism in BAT using oxygen-15(15O)/PET imaging (41). Considering that 20 kcal/day is the amount of excess energy consumption conveyed by a single 90-minute bout of cold exposure, chronic cold adaption may have more pronounced effects on energy expenditure given that repeated cold exposure over 6 weeks increased BAT activity by 150%. In addition, chronic cold exposure as well as weight loss resulted in the emergence of newly recruited BAT in individuals with previously no detectable BAT (11, 42) emphasizing that therapeutic interventions targeting BAT activation may not be limited to individuals with the presence of BAT before treatment.

Recently, human BAT has been established as a metabolic tissue that takes up and burns lipids but also represents a source of lipid production by releasing so-called lipokines into the circulation (19, 22). Lipokines act locally or systemically and control a number of metabolic processes such as glucose homeostasis and inflammation.

Oxylipins, including eicosanoids are a complex class of lipid-derived signaling molecules that govern inflammatory, anti-inflammatory, and metabolic processes (26, 40, 43, 44). In addition, eicosanoids represent the most important physiological ligands for PPARs, which are critical in regulating carbohydrate and lipid metabolism (45). BAT has recently been identified as a source of oxylipin production in humans (26, 40). Using high-resolution mass spectrometry analysis (46) we found that circulating PUFA and oxylipin profiles were distinct in BATpos and BATneg individuals and that cold exposure significantly altered plasma lipid composition only in BATpos, strongly suggesting that BAT is a significant contributor to the systemic oxylipin pool in humans. Indeed, the BATokine 12,13-DiHOME has recently been demonstrated to be induced in mice and humans upon cold exposure. In addition, 12,13-DiHOME administration in mice stimulated BAT fuel uptake, triglyceride clearance, and conferred protection against cold stress (23, 26). We detected 12,13-DiHOME in all participants and found it increased in BATneg and BATpos individuals upon cold exposure, reaching statistical significance when combining both groups (logFC 1.36, FDR 6.57 %). Notably, the potential lipokine that was most strongly upregulated in BATpos compared with BATneg in our study cohort also belonged to the DiHOME family (designated here as DiHOME_10.81, Table 1 (28), suggesting that DiHOMEs are a class of lipids regulated by BAT with a potential for therapeutic targeting. In keeping with this notion, 12,13-DiHOME was inversely associated with obesity and related metabolic complications in a recent cross-sectional study with more 2200 participants (47). Like DiHOMEs, DiHETrEs are another group of cytochrome P450 oxidase products that were significantly higher in BATpos versus BATneg participants (cluster 1 in Fig. 3A). Notably this was the only lipid cluster that correlated positively with CIEE in BATpos (Fig. 3B), directly linking specific systemic oxylipin availability with BAT thermogenesis. Indeed, DiHETrEs are known agonists of the nuclear receptor PPAR-α (39), a master regulator of fatty acid oxidation and lipid catabolism. These observations also suggest that BAT may be associated with higher P450 enzymatic activity resulting in the formation of PPAR-α agonists and thus affecting systemic lipid metabolism.

The presence of BAT was also characterized by increased concentrations of ω3 fatty acids and their precursor molecules such as stearidonic acid, EPA, and 12-HEPE (cluster 2). In accordance with previous reports (48, 49), plasma levels of PUFAs such as DPA and DHA and their precursors were elevated in response to cold exposure only in BATpos participants (Table 2, Fig. 4). In mice, DPA supplementation had a comparable favorable effect on cardiometabolic parameters as does treatment with DHA (50). Supplementation with the ω3 fatty acids EPA and DHA reduces cardiovascular risk in a dose-dependent manner in large randomized trials (51). In addition, EPA administration has been linked to BAT thermogenesis as well as WAT browning in vitro and in mice (49, 52, 53). One possible mediator of this effect is the G-protein–coupled free fatty acid receptor GPR120, which has been shown to induce mitochondrial fission and thermogenic gene expression via fibroblast growth factor 21 (54-56). Stearidonic acid, which was more abundant in BATpos at RT as well as after cold exposure, has been shown to suppress lipopolysaccharide-induced inflammation in murine adipocyte stem cells (57). Most recently, 12-HEPE has been identified as a potential novel cold-induced BATokine that promotes glucose uptake in adipocytes and skeletal muscle through activation of an insulin-like intracellular signaling pathway (40). These findings not only suggest BAT as an important source for PUFA release as previously suggested (58) but could hint toward potential anti-inflammatory and positive metabolic effects mediated by BAT-derived ω3 fatty acids (59). In contrast, a number of proinflammatory HETEs including 5-, 11-, 12-, and 15-HETE were significantly reduced in BATpos compared with BATneg (cluster 3 in Fig. 3A, Table 1). All of them have been associated with obesity-related complications (60, 61). Particularly 12-HETE has been suggested to be involved in the pathogenesis of type 2 diabetes by triggering insulin resistance and β-cell dysfunction (60, 62-64). Both, 12-HETE and 12-HEPE are products of 12-lipoxygenase (12-LOX) and rise with cold exposure, which is in line with the recent finding of a cold-mediated increase in 12-LOX activity in BAT (40). However, the observation that 12-HETE levels were generally higher in BATneg versus BATpos individuals (Fig. 4B), suggests that other tissues than BAT may also contribute toward systemic 12-HETE production. Particularly, WAT may represent a potential source for oxylipins as previously reported (60). In addition, WAT depots are significantly larger than BAT depots, and WAT is also known to respond to cold stimuli given that adrenergic receptors are abundantly expressed in white adipocytes (65). However, 12-HEPE was 1 of only 8 molecules that were elevated in BATpos at baseline and increased even further after cold exposure (Fig. 5), suggesting that BAT is indeed a significant source of 12-HEPE production. This further demonstrates that 12-LOX activity has both detrimental as well as positive effects in adipocyte biology. Many of the identified lipids are part of the same biosynthesis pathways (eg, the LOX pathway) (28).

A limitation of this study is the relatively small sample size due to the age- and BMI-matched design and the expensive technology used here. Also, the radioactive exposure associated with [18F]-FDG PET/CT scans would hardly justify the screening of a larger number of individuals given that these were healthy volunteers. Despite the small sample size and the exploratory nature of the study, we found a robust regulation of several lipid species by cold exposure and BAT status that is in accordance with previous reports of cold-induced BAT-derived oxylipins (26, 40). However, it cannot be excluded that other tissues responsive to cold stimulation contribute to the observed phenotype. The fact that very distinct lipid clusters are regulated concomitantly by BAT status and cold exposure strongly suggests that BAT is indeed an important organ orchestrating the systemic PUFA/oxylipin pool in a coordinated manner and therefore warrants further investigation.

In summary, cold exposure and the presence of active BAT were associated with a distinct PUFA and oxylipin profile with potentially anti-inflammatory and metabolically favorable characteristics (eg, increased ω3 fatty acids and endogenous PPAR ligands). These data add to the growing body of evidence that BAT may affect metabolic processes beyond thermogenesis, including lipid metabolism and inflammation, and may communicate with other metabolic tissues through BATokines.

Abbreviations

    Abbreviations
     
  • [18F]-FDG PET/CT

    18F-fluorodeoxyglucose positron emission tomography combined with computed tomography

  •  
  • 12-LOX

    12-lipoxygenase

  •  
  • 15d-PGJ2

    15-deoxy-Δ12,14-prostaglandin J2

  •  
  • 5-oxo-ETE

    5-oxo-eicosatetraenoic acid

  •  
  • BAT

    brown adipose tissue

  •  
  • BATokines

    BAT-derived circulating factors

  •  
  • BMI

    body mass index

  •  
  • CIEE

    cold-induced energy expenditure

  •  
  • DHA

    docosahexaenoic acid

  •  
  • DiHETrE

    dihydroxyeicosatrienoic acid

  •  
  • DiHOME

    dihydroxyoctadecenoic acid

  •  
  • DPA

    docosapentaenoic acid

  •  
  • EPA

    eicosapentaenoic acid

  •  
  • FA

    formic acid

  •  
  • FDR

    false discovery rate

  •  
  • GGM

    graphical Gaussian modeling

  •  
  • HETE

    hydroxyeicosatetraenoic acid

  •  
  • HMDB

    human metabolome database

  •  
  • LC

    liquid chromatography

  •  
  • MBq

    megabecquerel

  •  
  • MeOH

    methanol

  •  
  • MS

    mass spectrometry

  •  
  • P450

    cytochrome P450

  •  
  • PC1

    first principal component

  •  
  • PPAR-α

    peroxisome proliferator-activated receptor alpha

  •  
  • PPAR-γ

    peroxisome proliferator-activated receptor gamma

  •  
  • PUFAs

    polyunsaturated fatty acids

  •  
  • REE

    resting energy expenditure

  •  
  • RQ

    respiratory quotient

  •  
  • WAT

    white adipose tissue

Acknowledgments

Financial Support: This work was supported by the Vienna Science and Technology Fund, LS12-059, Austrian Science Fund, P 27391 and the Austrian Diabetes Association Research Fund all to F.W.K.

Clinical Trial Information: ClinicalTrials.gov. registration no. NCT02381483 (registered March 6, 2015).

Author Contributions: All authors listed on this manuscript researched and/or analyzed data, contributed intellectually, and revised the manuscript.

Additional Information

The authors have nothing to disclose.

Disclosure Summary: The authors have nothing to disclose.

Data Availability: The datasets generated during and/or analyzed during the current study are not publicly available but are available from the corresponding author on reasonable request.

References

1.

Bartelt
 
A
,
Heeren
J
.
Adipose tissue browning and metabolic health
.
Nat Rev Endocrinol.
2014
;
10
(
1
):
24
-
36
.

2.

Harms
 
M
,
Seale
P
.
Brown and beige fat: development, function and therapeutic potential
.
Nat Med.
2013
;
19
(
10
):
1252
-
1263
.

3.

Kiefer
 
FW
.
The significance of beige and brown fat in humans
.
Endocr Connect.
2017
;
6
(
5
):
R70
-
R79
.

4.

Kim
 
SH
,
Plutzky
J
.
Brown fat and browning for the treatment of obesity and related metabolic disorders
.
Diabetes Metab J.
2016
;
40
(
1
):
12
-
21
.

5.

Ravussin
 
Y
,
Xiao
C
,
Gavrilova
O
,
Reitman
ML
.
Effect of intermittent cold exposure on brown fat activation, obesity, and energy homeostasis in mice
.
PLoS One.
2014
;
9
(
1
):
e85876
.

6.

Cypess
 
AM
,
Lehman
S
,
Williams
G
, et al.  
Identification and importance of brown adipose tissue in adult humans
.
N Engl J Med.
2009
;
360
(
15
):
1509
-
1517
.

7.

van Marken Lichtenbelt
 
WD
,
Vanhommerig
JW
,
Smulders
NM
, et al.  
Cold-activated brown adipose tissue in healthy men
.
N Engl J Med.
2009
;
360
(
15
):
1500
-
1508
.

8.

Virtanen
 
KA
,
Lidell
ME
,
Orava
J
, et al.  
Functional brown adipose tissue in healthy adults
.
N Engl J Med.
2009
;
360
(
15
):
1518
-
1525
.

9.

Saito
 
M
,
Okamatsu-Ogura
Y
,
Matsushita
M
, et al.  
High incidence of metabolically active brown adipose tissue in healthy adult humans: effects of cold exposure and adiposity
.
Diabetes.
2009
;
58
(
7
):
1526
-
1531
.

10.

Yoneshiro
 
T
,
Aita
S
,
Matsushita
M
, et al.  
Brown adipose tissue, whole-body energy expenditure, and thermogenesis in healthy adult men
.
Obesity (Silver Spring).
2011
;
19
(
1
):
13
-
16
.

11.

Yoneshiro
 
T
,
Aita
S
,
Matsushita
M
, et al.  
Recruited brown adipose tissue as an antiobesity agent in humans
.
J Clin Invest.
2013
;
123
(
8
):
3404
-
3408
.

12.

Hanssen
 
MJ
,
Hoeks
J
,
Brans
B
, et al.  
Short-term cold acclimation improves insulin sensitivity in patients with type 2 diabetes mellitus
.
Nat Med.
2015
;
21
(
8
):
863
-
865
.

13.

Lee
 
P
,
Smith
S
,
Linderman
J
, et al.  
Temperature-acclimated brown adipose tissue modulates insulin sensitivity in humans
.
Diabetes.
2014
;
63
(
11
):
3686
-
3698
.

14.

Matsushita
 
M
,
Yoneshiro
T
,
Aita
S
,
Kameya
T
,
Sugie
H
,
Saito
M
.
Impact of brown adipose tissue on body fatness and glucose metabolism in healthy humans
.
Int J Obes (Lond).
2014
;
38
(
6
):
812
-
817
.

15.

Bakker
 
LE
,
Boon
MR
,
van der Linden
RA
, et al.  
Brown adipose tissue volume in healthy lean South Asian adults compared with white Caucasians: a prospective, case-controlled observational study
.
Lancet Diabetes Endocrinol.
2014
;
2
(
3
):
210
-
217
.

16.

Yoneshiro
 
T
,
Aita
S
,
Matsushita
M
, et al.  
Age-related decrease in cold-activated brown adipose tissue and accumulation of body fat in healthy humans
.
Obesity (Silver Spring).
2011
;
19
(
9
):
1755
-
1760
.

17.

Chondronikola
 
M
,
Volpi
E
,
Børsheim
E
, et al.  
Brown adipose tissue improves whole-body glucose homeostasis and insulin sensitivity in humans
.
Diabetes.
2014
;
63
(
12
):
4089
-
4099
.

18.

Iwen
 
KA
,
Backhaus
J
,
Cassens
M
, et al.  
Cold-induced brown adipose tissue activity alters plasma fatty acids and improves glucose metabolism in men
.
J Clin Endocrinol Metab.
2017
;
102
(
11
):
4226
-
4234
.

19.

Chondronikola
 
M
,
Volpi
E
,
Børsheim
E
, et al.  
Brown adipose tissue activation is linked to distinct systemic effects on lipid metabolism in humans
.
Cell Metab.
2016
;
23
(
6
):
1200
-
1206
.

20.

Ouellet
 
V
,
Labbé
SM
,
Blondin
DP
, 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
.

21.

Hoeke
 
G
,
Nahon
KJ
,
Bakker
LEH
, et al.  
Short-term cooling increases serum triglycerides and small high-density lipoprotein levels in humans
.
J Clin Lipidol.
2017
;
11
(
4
):
920
-
928.e2
.

22.

Villarroya
 
F
,
Cereijo
R
,
Villarroya
J
,
Giralt
M
.
Brown adipose tissue as a secretory organ
.
Nat Rev Endocrinol.
2017
;
13
(
1
):
26
-
35
.

23.

Stanford
 
KI
,
Lynes
MD
,
Takahashi
H
, et al.  
12,13-diHOME: an exercise-induced lipokine that increases skeletal muscle fatty acid uptake
.
Cell Metab.
2018
;
27
(
6
):
1357
.

24.

Hankir
 
MK
,
Cowley
MA
,
Fenske
WK
.
A BAT-centric approach to the treatment of diabetes: turn on the brain
.
Cell Metab.
2016
;
24
(
1
):
31
-
40
.

25.

Tan
 
CK
,
Zhuang
Y
,
Wahli
W
.
Synthetic and natural peroxisome proliferator-activated receptor (PPAR) agonists as candidates for the therapy of the metabolic syndrome
.
Expert Opin Ther Targets.
2017
;
21
(
3
):
333
-
348
.

26.

Lynes
 
MD
,
Leiria
LO
,
Lundh
M
, et al.  
The cold-induced lipokine 12,13-diHOME promotes fatty acid transport into brown adipose tissue
.
Nat Med.
2017
;
23
(
5
):
631
-
637
.

27.

Boon
 
MR
,
Bakker
LEH
,
Prehn
C
, et al.  
LysoPC-acyl C16:0 is associated with brown adipose tissue activity in men
.
Metabolomics.
2017
;
13
(
5
):
48
.

28.

Kulterer
 
OC
,
Niederstaetter
L
,
Herz
CT
, et al.  
The presence of active brown adipose tissue determines cold-induced energy expenditure and oxylipin profiles in humans: Supplementary Materials and Methods
.
2020
. doi: 10.6084/m9.figshare.12015819. https://figshare.com/s/1d10df48f9b7beaba397

29.

Cypess
 
AM
,
Chen
YC
,
Sze
C
, et al.  
Cold but not sympathomimetics activates human brown adipose tissue in vivo
.
Proc Natl Acad Sci U S A.
2012
;
109
(
25
):
10001
-
10005
.

30.

Chen
 
KY
,
Cypess
AM
,
Laughlin
MR
, et al.  
Brown adipose reporting criteria in imaging STudies (BARCIST 1.0): recommendations for standardized FDG-PET/CT experiments in humans
.
Cell Metab.
2016
;
24
(
2
):
210
-
222
.

31.

R Development Core Team
.
R: A Language and Environment for Statistical Computing
.
Vienna, Austria
:
R Foundation for Statistical Computing
;
2010
.

32.

Benjamini
 
Y
,
Hochberg
Y
.
Controlling the false discovery rate - a practical and powerful approach to multiple testing
.
J R Stat Soc B
.
1995
;
57
(
1
):
289
-
300
.

33.

van Buuren
 
S
,
Groothuis-Oudshoorn
K.
 
mice: multivariate imputation by chained equations in R
.
J Stat Softw
.
2011
;
45
(
3
):
1
-
67
.

34.

Ritchie
 
ME
,
Phipson
B
,
Wu
D
, et al.  
limma powers differential expression analyses for RNA-sequencing and microarray studies
.
Nucleic Acids Res.
2015
;
43
(
7
):
e47
.

35.

Giraud
 
C
,
Huet
S
,
Verzelen
N
.
Graph selection with GGMselect
.
Stat Appl Genet Mol Biol.
2012
;
11
(
3
):
Article 3
.

36.

Väremo
 
L
,
Nielsen
J
,
Nookaew
I
.
Enriching the gene set analysis of genome-wide data by incorporating directionality of gene expression and combining statistical hypotheses and methods
.
Nucleic Acids Res.
2013
;
41
(
8
):
4378
-
4391
.

37.

Djoumbou-Feunang
 
Y
,
Pon
A
,
Karu
N
, et al.  
CFM-ID 3.0: significantly improved ESI-MS/MS prediction and compound identification
.
Metabolites
.
2019
;
9
(
4
):72.

38.

Wishart
 
DS
,
Feunang
YD
,
Marcu
A
, et al.  
HMDB 4.0: the human metabolome database for 2018
.
Nucleic Acids Res.
2018
;
46
(
D1
):
D608
-
D617
.

39.

Ng
 
VY
,
Huang
Y
,
Reddy
LM
,
Falck
JR
,
Lin
ET
,
Kroetz
DL
.
Cytochrome P450 eicosanoids are activators of peroxisome proliferator-activated receptor alpha
.
Drug Metab Dispos.
2007
;
35
(
7
):
1126
-
1134
.

40.

Leiria
 
LO
,
Wang
CH
,
Lynes
MD
, et al.  
12-Lipoxygenase regulates cold adaptation and glucose metabolism by producing the omega-3 lipid 12-HEPE from Brown fat
.
Cell Metab.
2019
;
30
(
4
):
768
-
783.e7
.

41.

Muzik
 
O
,
Mangner
TJ
,
Granneman
JG
.
Assessment of oxidative metabolism in brown fat using PET imaging
.
Front Endocrinol (Lausanne).
2012
;
3
:
15
.

42.

Vijgen
 
GH
,
Bouvy
ND
,
Teule
GJ
, et al.  
Increase in brown adipose tissue activity after weight loss in morbidly obese subjects
.
J Clin Endocrinol Metab.
2012
;
97
(
7
):
E1229
-
E1233
.

43.

Vegiopoulos
 
A
,
Müller-Decker
K
,
Strzoda
D
, et al.  
Cyclooxygenase-2 controls energy homeostasis in mice by de novo recruitment of brown adipocytes
.
Science.
2010
;
328
(
5982
):
1158
-
1161
.

44.

Dennis
 
EA
,
Norris
PC
.
Eicosanoid storm in infection and inflammation
.
Nat Rev Immunol.
2015
;
15
(
8
):
511
-
523
.

45.

Marion-Letellier
 
R
,
Savoye
G
,
Ghosh
S
.
Fatty acids, eicosanoids and PPAR gamma
.
Eur J Pharmacol.
2016
;
785
:
44
-
49
.

46.

Tahir
 
A
,
Bileck
A
,
Muqaku
B
, et al.  
Combined proteome and eicosanoid profiling approach for revealing implications of human fibroblasts in chronic inflammation
.
Anal Chem.
2017
;
89
(
3
):
1945
-
1954
.

47.

Vasan
 
SK
,
Noordam
R
,
Gowri
MS
,
Neville
MJ
,
Karpe
F
,
Christodoulides
C
.
The proposed systemic thermogenic metabolites succinate and 12,13-diHOME are inversely associated with adiposity and related metabolic traits: evidence from a large human cross-sectional study
.
Diabetologia.
2019
;
62
(
11
):
2079
-
2087
.

48.

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
.

49.

Kim
 
J
,
Okla
M
,
Erickson
A
,
Carr
T
,
Natarajan
SK
,
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
.

50.

Guo
 
XF
,
Sinclair
AJ
,
Kaur
G
,
Li
D
.
Differential effects of EPA, DPA and DHA on cardio-metabolic risk factors in high-fat diet fed mice
.
Prostaglandins Leukot Essent Fatty Acids.
2018
;
136
:
47
-
55
.

51.

Hu
 
Y
,
Hu
FB
,
Manson
JE
.
Marine omega-3 supplementation and cardiovascular disease: an updated meta-analysis of 13 randomized controlled trials involving 127 477 participants
.
J Am Heart Assoc.
2019
;
8
(
19
):
e013543
.

52.

Pahlavani
 
M
,
Wijayatunga
NN
,
Kalupahana
NS
, et al.  
Transcriptomic and microRNA analyses of gene networks regulated by eicosapentaenoic acid in brown adipose tissue of diet-induced obese mice
.
Biochim Biophys Acta Mol Cell Biol Lipids.
2018
;
1863
(
12
):
1523
-
1531
.

53.

Zhao
 
M
,
Chen
X
.
Eicosapentaenoic acid promotes thermogenic and fatty acid storage capacity in mouse subcutaneous adipocytes
.
Biochem Biophys Res Commun.
2014
;
450
(
4
):
1446
-
1451
.

54.

Miyamoto
 
J
,
Hasegawa
S
,
Kasubuchi
M
,
Ichimura
A
,
Nakajima
A
,
Kimura
I
.
Nutritional signaling via free fatty acid receptors
.
Int J Mol Sci.
2016
;
17
(
4
):
450
.

55.

Schilperoort
 
M
,
van Dam
AD
,
Hoeke
G
, et al.  
The GPR120 agonist TUG-891 promotes metabolic health by stimulating mitochondrial respiration in brown fat
.
EMBO Mol Med.
2018
;
10
(
3
):e8047.

56.

Quesada-López
 
T
,
Cereijo
R
,
Turatsinze
JV
, et al.  
The lipid sensor GPR120 promotes brown fat activation and FGF21 release from adipocytes
.
Nat Commun.
2016
;
7
:
13479
.

57.

Hsueh
 
HW
,
Zhou
Z
,
Whelan
J
, et al.  
Stearidonic and eicosapentaenoic acids inhibit interleukin-6 expression in ob/ob mouse adipose stem cells via Toll-like receptor-2-mediated pathways
.
J Nutr.
2011
;
141
(
7
):
1260
-
1266
.

58.

Qin
 
X
,
Park
HG
,
Zhang
JY
, et al.  
Brown but not white adipose cells synthesize omega-3 docosahexaenoic acid in culture
.
Prostaglandins Leukot Essent Fatty Acids.
2016
;
104
:
19
-
24
.

59.

Calder
 
PC
.
Omega-3 fatty acids and inflammatory processes
.
Nutrients.
2010
;
2
(
3
):
355
-
374
.

60.

Cole
 
BK
,
Lieb
DC
,
Dobrian
AD
,
Nadler
JL
.
12- and 15-lipoxygenases in adipose tissue inflammation
.
Prostaglandins Other Lipid Mediat.
2013
;
104-105
:
84
-
92
.

61.

Pickens
 
CA
,
Sordillo
LM
,
Zhang
C
,
Fenton
JI
.
Obesity is positively associated with arachidonic acid-derived 5- and 11-hydroxyeicosatetraenoic acid (HETE)
.
Metabolism.
2017
;
70
:
177
-
191
.

62.

Nunemaker
 
CS
,
Chen
M
,
Pei
H
, et al.  
12-Lipoxygenase-knockout mice are resistant to inflammatory effects of obesity induced by Western diet
.
Am J Physiol Endocrinol Metab.
2008
;
295
(
5
):
E1065
-
E1075
.

63.

Ma
 
K
,
Nunemaker
CS
,
Wu
R
,
Chakrabarti
SK
,
Taylor-Fishwick
DA
,
Nadler
JL
.
12-Lipoxygenase products reduce insulin secretion and {beta}-cell viability in human islets
.
J Clin Endocrinol Metab.
2010
;
95
(
2
):
887
-
893
.

64.

Tersey
 
SA
,
Maier
B
,
Nishiki
Y
,
Maganti
AV
,
Nadler
JL
,
Mirmira
RG
.
12-Lipoxygenase promotes obesity-induced oxidative stress in pancreatic islets
.
Mol Cell Biol.
2014
;
34
(
19
):
3735
-
3745
.

65.

Evans
 
BA
,
Merlin
J
,
Bengtsson
T
,
Hutchinson
DS
.
Adrenoceptors in white, brown, and brite adipocytes
.
Br J Pharmacol.
2019
;
176
(
14
):
2416
-
2432
.

Author notes

These authors contributed equally.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/journals/pages/open_access/funder_policies/chorus/standard_publication_model)