Generation of Ptgr2 knockout mice
The generation of Ptgr2 knockout mice has been described previously (14). Briefly, the mouse Ptgr2 gene comprises 10 exons with exon 3 containing the catalytic domain. Deletion of exon 3 is predicted to not only delete the catalytic domain but also to create a frameshift mutation resulting in a stop codon in exon 4. The construct used for targeting the Ptgr2 gene was designed to insert a loxP sequence together with an "FRT-flanked" pgk-neo cassette in intron 2, and a loxP sequence in intron 4. The linearized targeting vector was electroporated into the 129/J embryonic stem cell line and selected by neomycin and ganciclovir. The selected clone was used for blastocyst injection. Immunoblots for Ptgr2 showed deletion of Ptgr2 in all tissues of knockout mice compared with wild-type controls (Supplementary Figure 1).
Animal model
All animal experiments were performed according to institutional ethical guidelines and were approved by the Institutional Animal Care and Use Committee of the Medical College (IACUC) of National Taiwan University. All mice were housed under standard conditions at 23°C and 12/12 hr light/dark (7AM.-7PM.) cycle in animal centers of National Taiwan University Medical College, which is accredited by the Association for Assessment and Accreditation of Laboratory Animal Care International (AAALAC). Mice were fed on either high-fat high-sucrose diet (HFHSD) (cat. no.D12331, Research Diets) or regular chow diet (cat no. 5001, Lab Diet). BPRPT245 was dissolved 3% dimethylacetamide and 10% cremophor in water and administered to orally HFHSD-fed obese C57BL6/J mice by oral gavage daily (100 mg/kg/day).
Glucose and insulin tolerance test
Glucose tolerance was evaluated by the oral (OGTT) and intraperitoneal glucose tolerance test (ipGTT) after a 6-hr fast. For the OGTT, glucose water (1 mg/g) was given by oral gavage and tail blood glucose was measured with a glucometer (ACCU-CHECK Performa, Roche) at 0, 15, 30, 45, 60, 90, and 120 min. For ipGTT, tail blood glucose was measured at 0, 15, 30, 45, 60, 90, and 120 min after intraperitoneal injection of glucose water (1 mg/g). For the insulin tolerance test (ITT), mice were fasted for 4 hr and then injected intraperitoneally with 1 U/kg of insulin (Humulin R, Eli Lilly). Tail blood glucose was measured at 0, 15, 30, 45, 60, 90, 120, and 180 min.
Measurement of insulin signaling
To evaluate insulin signaling in vivo, we fasted the mice overnight. Tissue was harvested 15 min after intraperitoneal insulin injection. The samples (perigonadal fat, inguinal fat, brown adipose tissue and liver) were extracted with RIPA buffer (50 mM Tris-HCl, pH 7.4, 2 mM ethylenediaminetetraacetic acid [EDTA]), 150 mM NaCl, 50 mM NaF, 1% Nonidet P-40, 1 mM phenylmethylsulfonyl fluoride (PMSF), 0.5% sodium deoxycholate, and 0.1% sodium dodecyl sulfate (SDS) containing phosphatase inhibitor cocktail (cat. no. 04693132001, Roche) and homogenized. The homogenates were then centrifuged at 13,000 rpm for 10 min at 4°C to remove debris. Samples were separated by SDS-polyacrylamide gel electrophoresis, transferred to polyvinylidene difluoride (PVDF) membrane, and probed with anti-phospho-Akt antibody (cat. no. 4058, Cell Signaling) and anti-Akt antibody (cat. no. 9272, Cell Signaling) and then with HRP conjugated anti-rabbit IgG antibody (1:10000; cat. no. GTX26721, GeneTex).
Positron emission tomography (PET) / CT (computer tomography) for assessment of glucose uptake
The measurement of glucose uptake in different tissues followed previous protocols (15,16). Briefly, mice were fasted for 4 hrs. A total of 0.5 MBq [18F]-FDG in a 0.1-ml volume was administered through the tail vein and 1.6 U/kg of insulin was injected intraperitoneally immediately following [18F]-FDG administration. Whole body scan was performed for a total of three cycles (10 min per cycle) using PET/CT scanner (eXplore Vista DR, GE). Images were analyzed using the Amide software (17). Region of interest (ROI) was determined using manual selection of sagittal, horizontal, and vertical slices. Standardized uptake values of each ROI (for each tissue) were calculated to estimate glucose uptake.
Formulation for 15-keto-PGE2 for injection
For animal experiments, 15-keto-PGE2 was dissolved in liposome (40 mg/kg/day in 20 μL/g/day liposome, intra-peritoneal injection twice daily) for the treatment and was compared to the vehicle-control groups (liposome vehicle 20 μL/g/day, intra-peritoneal injection twice daily) for 3 weeks. The Taiwan Liposome Company designed and provided the liposome formulation.
Preparation of cysteine-coupled 15-keto-PGE2 and 15-keto-PGF2a proteins
Five mg of ovomucoid (OVO, cat. no. T9253, Sigma-Aldrich) and bovine serum albumin (BSA, cat. no. A2153, Sigma-Aldrich) were dissolved in 1 mL of PBS containing 5% 2-mercaptoethanol and gently agitated at room temperature for 1 hr, respectively. The reduced proteins were buffer-exchanged with PBS using Amicon Ultra-15 Centrifugal Filter Units (cat. no. UFC901024, Millipore) to remove 2-ME. The proteins were then incubated with tenfold molar excess of 15-keto-PGE2 or 15-keto-PGF2α at room temperature for 1 hr with gentle agitation. The cysteine-coupled proteins, 15-keto-PGE2-cysteine-OVO, 15-keto-PGE2-cysteine-BSA, and 15-keto-PGF2α-cysteine-BSA, were further buffer-exchanged with PBS to remove free 15-keto-PGE2 or 15-keto-PGF2α respectively, and stored at 1 mg/mL.
Generation of monoclonal antibodies against cysteine-coupled 15-keto-PGE2
Ten Balb/c mice (National Laboratory Animal Center, Taiwan) were immunized with 15-keto-PGE2-cysteine-OVO (100 μg/mouse) emulsified with complete Freund’s adjuvant (cat. no. F5881, Sigma-Aldrich) via subcutaneous injection. After four weeks, mice were subcutaneously injected with 15-keto-PGE2-cysteine-OVO (100 μg/mouse) emulsified with incomplete Freund’s adjuvants (cat. no. F5506, Sigma-Aldrich) three times at a two-week intervals. Mice received 10 μg of 15-keto-PGE2-cysteine-OVO via tail vein three days prior to spleen harvest. Hybridomas were prepared by fusing spleen cells with the mouse myeloma cell line FO (cat. no. CRL-1646, ATCC) using PEG 1500 (cat. no. 10783641001, Roche). The fusion procedures followed the manufacturer’s manual. Hybridomas were selected in complete Dulbecco's modified Eagle’s medium (DMEM) containing hypoxanthine-aminopterin-thymidine (cat. no. 11067030, Thermo Fisher) and UltraCruz Hybridoma Cloning Supplement (cat. no. sc-224479, Santa Cruz) for 12-14 days. Cell culture supernatants were screened with ELISA using 15-keto-PGE2-cysteine-BSA as antigens. Positive hybridomas were then cloned by limiting dilution. Antibody isotypes were determined with a Rapid ELISA Mouse mAb Isotyping Kit (cat. no. 37503, Thermo Fisher). Monoclonal antibodies were purified from hybridoma culture supernatants using Protein A Sepharose CL-4B (cat. no. 17078001, GE). The purification procedures were performed according to the manufacturer’s manual. Four anti-cysteine-coupled 15-keto-PGE2 hybridoma clones were prepared. Clones 1A6 and 17E7 had superior specificity and reacted only to cysteine-coupled 15-keto-PGE2 (Supplementary Figure 2).
Cell culture
All cells were cultured at 37°C, 5% CO2 in a humidified incubator. HEK293T cells were maintained in DMEM (cat. no. SH30003.02, HyClone) supplemented with 10% fetal bovine serum (FBS) (cat. no. 04-001-1A, Biological Industries) and 1% antibiotic/antimycotic solution (cat. no. SV30079.01, HyClone). 3T3-L1 preadipocytes were maintained at ~70 % confluence in DMEM with 10% calf serum (cat. no. 16170078, Gibco) and 1% antibiotic/antimycotic solution. For adipocyte differentiation, confluent 3T3-L1 cells (defined as day 0) were exposed to induction medium containing 10% FBS, 1 μM dexamethasone (cat. no. D4902, Sigma-Aldrich), and 1 μg/ml insulin (Humulin R, Eli Lilly) in DMEM with or without 0.5 mM isobutyl-methylxanthine (cat. no. sc-201188A, Santa Cruz Biotechnology) as indicated. After two days, the medium was replaced with DMEM containing 10% FBS and 1 μg/ml insulin, and was replenished every two days until assay.
RNA extraction, cDNA synthesis and real-time quantitative PCR (RT-qPCR)
A total of 1×105 of 3T3-L1 preadipocytes were seeded in 6-well plates, and differentiated for six days, then the 3T3-L1 adipocytes were harvested in 1 ml of REzol C&T (cat. no. PT-KP200CT, Protech), and total RNA was extracted according to the manufacturer’s instructions with slight modifications. Briefly, 200 μl of chloroform was added to 1 ml of sample in REzol, and samples were mixed vigorously by shaking for 30 sec, followed by incubation at room temperature for 5 min. Then, samples were centrifuged at 12,000 g for 15 min at 4°C, and 400 μl of the upper aqueous phase were transferred to a new 1.5-ml tube. An equal volume of isopropanol was added. Samples were inverted several times for mixing and then centrifuged at 12,000g for 10 min at 4°C. The RNA precipitate, which formed a pellet at the bottom of 1.5-ml tube, was washed for three times with 75% ethanol and air-dried for 15 min. Pellets were dissolved the in RNase-free water. The concentration of RNA was measured using Nanodrop. cDNA was synthesized using a reverse transcription kit (cat. no. K1622, Thermo Scientific) using the oligo(dT)18 primers. RT-qPCR was performed in a 10-μl reaction with 50 ng cDNA and 0.4 M primer using SYBR green reagent (cat. no. 11203ES08, YEASEN). Mouse peptidylprolyl isomerase A (Ppia) mRNA was used as the internal control. RT-qPCR reactions were performed using ABI 7900HT FAST (Applied Biosystems) and Sequence Detection Systems (SDS v2.3, Applied Biosystems). All qPCR reactions were run in duplicates. The primers used are listed in Supplementary Table 1.
Plasmid construction and site-directed mutagenesis
Murine PPARγ plasmid (pCMV6-Pparg) was purchased from OriGene (cat. no. MC201042, OriGene, USA). A cysteine-313 to alanine (C313A) substitution of mPPARγ construct was generated with the mutagenesis kit (cat. no. 210518, Agilent, USA) following the standard manufacturer's protocol. Primers site-directed mutagenesis was designed using a web-based program (http://www.genomics.agilent.com/primerDesignProgram.jsp) (Supplementary Table 2).
Reporter assay
Gal4-PPARγ/UAS-LUC reporter assay was conducted as previously described (12) with minor modifications. Briefly, HEK293T cells were seeded at 1×105 cells/well in 24-well plates. After 24- hrs growth, a DNA solution containing UASG reporter construct, GAL4-PPAR expression vector and TK-Rluc (Renilla luciferase) reporter construct (internal control) was transfected using TurboFect™ transfection reagent (cat. no. R0532, Thermo Fisher). Cells were treated and harvested after another 24 and 48 hrs, respectively. The luciferase activity was measured by Luc-Pair™ Duo-Luciferase HS Assay Kit (cat. no. LF600, Promega) and normalized to the TK reporter signal. For the PPRE reporter assay, as described above, 24 hrs after seeding HEK293T cells, PPRE-LUC reporter vector, TK-Rluc reporter and wild-type or mPPARγ C313A were transfected into cells using TurboFect transfection reagent.
Insulin-stimulated glucose uptake assay
Differentiated 3T3-L1 cells were starved with serum-free DMEM containing 0.2% BSA (cat. no. A9647, Sigma-Aldrich). Cells were rinsed twice with KRH buffer (137 mM NaCl, 4.7 mM KCl, 1.85 mM CaCl2, 1.3 mM MgSO4, 50 mM HEPES and 0.1% BSA, pH 7.4), and treated with or without 1 μg/mg insulin in the presence of 20 μM cytochalasin B (cat. no.11328, Cayman Chemical), a GLUT inhibitor for measuring non-specific glucose uptake, for 30 min. Cells were then incubated with 0.5 μCi [3H]-2-deoxy-D-Glucose (cat. no. NET328A001MC, PerkinElmer) and 0.1 μM 2-deoxy-D-Glucose (2DG) (cat. no. 14325, Cayman Chemical) for 5 min. Uptake of 2DG was terminated by a rapid removal of medium and washing with ice-cold PBS three times. Cells were lysed with 0.1% SDS, and the radioactivity was measured using a liquid scintillation counter. Insulin-mediated glucose uptake was calculated by subtracting insulin-treated glucose uptake with basal glucose uptake (in absence of insulin).
Computer modeling analysis binding between PPARγ and 15-keto-PGE2
Docking analyses were conducted using the CovalentDock program (18). The human X-ray structure of PPARγ (PDB ID: 5Y2O) (19) was used to assess for covalent binding with the small molecule 15-keto-PGE2. To illustrate the binding mode of 15-keto-PGE2, the covalent docking approach was applied. To ensure mimicking the experiment results of mouse species, four amino acids (S302N, V307I, L435V, and Q454H) were mutated by computer modeling. To bring the docked ligand-protein complex to equilibrium, molecular dynamics simulations were performed using the BIOVIA 2017/Standard dynamics Cascade program (BIOVIA, Inc., San Diego, CA). The minimization convergent was used in two steps, namely, Steepest Descent with RMS gradient convergent to 1, and the final step of Adopted Basis NR with RMS Gradient convergent to 0.1. The default parameters were used for the simulation, except for the Production parameter, which was set as 20 ns. All the minimization process was performed in an aqueous environment following the Distance-Dependent Dielectrics methods. After the molecular dynamics simulation, the binding energy of these two molecules was calculated using the BIOVIA 2017/Calculate Binding Energies program (BIOVIA, Inc., San Diego, CA). Three-dimensional models were displayed by using the PyMOL program (20).
To evaluate the interaction between PTGR2 inhibitor BPRPT245, 15-keto-PGE2, NADPH, and PTGR2, the X-ray structure of human PTGR2 (PDB ID: 2ZB4) was used. Ligand energy was minimized using PyRx program before docking (21). The 2D protein-ligand interaction diagrams are presented by using the LigPlot+ program (22).
Immunoprecipitation and proteomic analysis
For immunoprecipitation, HEK293T cells were transfected with mouse PPARγ (mPPARγ) and mPPARγ C313A plasmids, which are Myc-DDK-tagged (OriGene, USA). After 24 hr, cells were treated with dimethyl sulfoxide (DMSO) or 30 μM 15-keto-PGE2 for an additional 24 hrs and harvested with cell lysis buffer (50 mM Tris HCl, pH7.4, 1 mM EDTA, 150 mM NaCl, and 1% Triton X-100). Proteins were immunoprecipitated using Anti-FLAG® M2 Magnetic Beads (cat. no. F1804, Sigma-Aldrich) according to the manufacturer’s protocol. Subsequently, the eluted proteins were separated by SDS-PAGE and subjected to immunoblot analysis. Following separation by SDS-PAGE, proteins were stained with Coomassie blue and SYPRO-Ruby stain. The protein bands of interest were cut out for in-gel tryptic digestion followed by C18 Zip-Tip clean-up (Millipore, USA). For shotgun proteomic identifications, nanoLC-nanoESI-MS/MS analysis was performed on a nanoAcquity system (Waters, USA) connected to the LTQ Orbitrap Velos hybrid MS (Thermo Electron, USA) equipped with a PicoView nanospray interface (New Objective, USA). Samples were loaded onto a C18 BEH column (75 μm × 25 cm length, 130 Å , 1.7 μm particle size) (Waters, USA) and separated by a segmented gradient form in 60 min from 5% to 40% acetonitrile (with 0.1% formic acid) at a constant flow rate of 300 nL/min with a column temperature of 35°C. The mass spectrometer was operated in the data-dependent mode. Full scan MS spectra were acquired in the Orbitrap at 60,000 resolution (at m/z 400) with automatic gain control target of 5´105. The 10 most abundant ions were isolated for high-energy collision dissociation, MS/MS fragmentation and detection in the Orbitrap. For MS/MS measurements, a resolution of 7500, an isolation window of 2 m/z and a target value of 50,000 ions, with maximum accumulation times of 100 ms were used. Fragmentation was performed at 35% normalized collision energy and an activation time of 0.1 ms. Ions with single and unrecognized charge state were excluded. Protein identification and modification were analyzed using Mascot Daemon (Matrix Science Inc.).
Generation of PPARγ-null 3T3-L1 stable cell lines using CRISPR/Cas9 System
Gene editing was performed in 3T3-L1 preadipocytes using the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas 9 system. Two single-guide RNAs (sgRNA) (Supplementary Table 3) targeting exon 6 of mouse Pparg were designed using a web-based design tool (http://crispr.mit.edu/). These oligonucleotides were cloned into the pSurrogate reporter and pAll-Cas9.Ppuro vector obtained from Academia Sinica, Taiwan. The pSurrogate reporter contains an EGFP and an out-of-frame mCherry located downstream of the sgRNA; the pAll-Cas9.Ppuro vector expresses Cas9 nuclease and sgRNA. Once a double-stand break at target site in pSurrogate reporter plasmid was created by Cas9, insertions and deletions (indels) could be introduced at the cleaved site. The indels cause frameshifts and resulted in expression of the mCherry gene. 3T3-L1 preadipocytes were co-transfected with pSurrogate reporter and pAll-Cas9.Ppuro vector at 80-90% confluency using PolyJet™ transfection reagent (cat. no. SL100688, SignaGen Laboratories, USA). Two days after transfection, EGFP/mCherry double-positive cells were enriched by fluorescent-activated cell sorting. Single cells were isolated and expanded. The genomic DNA was amplified by PCR and sequenced to confirm the knockout cell lines. The primer sequences are shown in the Supplementary Table 3.
Lentivirus production and transduction
The mPPARγ C313A cDNA sequence was amplified from pCMV6-Pparg C313A and
subcloned into the XhoI/NotI restriction sites of the pLVX-IRES-Neo vector. The lentiviral expression plasmids, pMD.G and pCMVΔR8.91 (RNAi Core, Academia Sinica, Taiwan) were co-transfected into HEK293T cells. The medium containing lentivirus was collected at 40 and 64 hr after transfection, and centrifuged at 1,200 rpm for 5 min and then filtered through a 0.45-μm filter. PPARγ-null 3T3-L1 preadipocytes were infected with the lentivirus in the presence of 10 μg/ml polybrene (cat. no. sc-134220, Santa Cruz). Then, 48 hrs after infection, the cells were selected with 400 μg/ml G418. The expression of PPARγ was verified by immunoblotting using anti-PPARγ antibody (cat. no. sc-7273, Santa Cruz). HSP70 was probed with anti-HSP70 antibody (cat. no. ab45133, Abcam) as a loading control.
15-keto-PGE2 extraction from tissues and cultured cells
Tissue was homogenized in liquid nitrogen using pestle and mortar. Approximately 500 mg of tissue homogenate was treated with 400 μL of the upper layer of acetonitrile/hexane mixture supplemented with 2 μL formic acid. The homogenate was then added with 600 μL acetonitrile containing 13,14 dihydro-15-keto-PGE2-d4 (cat no. 10007978, Cayman Chemical, Ann Arbor, MI) in 1:20,000 dilution, followed by addition of 600 μL ddH2O. The mixture was centrifuged at 12,000 rpm for 10 min at 0°C. The lower layer was then transferred to a new tube. A C-18 solid-phase extraction (SPE) cartridge (Cat no. 400020, Cayman Chemical) was first activated with 20 mL methanol and then with 20 mL ddH2O. The SPE cartridge was subsequently washed with 5 mL 15% methanol and then with 5 mL ddH2O. The sample was then eluted with 10 mL HPLC-degree methanol (Methanol Chromasolv LC-MS, Fluka) into a new 2-mL tube and was then air-dried by the SpeedVac system (Savant SPD1010, Thermo Scientific). The dried samples were stored in -80°C. The samples were reconstituted with 100 μL methanol before LC-MS/MS.
For 15-keto-PGE2 extraction from cultured cells (in 6-well culture plates), the medium was removed and cells were scraped by adding 500 μL methanol containing 13,14 dihydro-15-keto-PGE2-d4 (1:20000 dilution), followed by addition of 1 mL PBS buffer. The homogenate was centrifuged at 300g for 5 min to remove cellular debris. The supernatant was transferred to a new tube. The C-18 SPE cartridge was first activated with 2 mL methanol and then 2 mL ddH2O. The lower-layer sample was transferred to C-18 SPE cartridge. The SPE cartridge was subsequently washed with 2 mL 15% methanol and 5 mL ddH2O. The sample was then eluted with 5 mL HPLC-degree methanol into a new 2-mL tube and air-dried using a SpeedVac system. The dried samples were stored in -80°C. The dried samples were reconstituted with 60 μL methanol before analysis.
Human subjects for measurement of serum 15-keto-PGE2 levels
Fifty non-diabetic male participants were recruited from a community-based screening for diabetes mellitus in the Yunlin County in Taiwan. We recruited another 24 male patients with type 2 diabetes from the metabolic clinic of Yunlin branch of National Taiwan University Hospital and selected 24 age- and body mass index (BMI)-matched male non-diabetic patients from a community-based screening program. The study protocol was approved by the Institutional Review Board of the National Taiwan University Hospital. Written informed consent was obtained from every participating subject. All procedures performed in studies involving human participants were in accordance with the Declaration of Helsinki.
Measurement of 15-keto-PGE2 by liquid chromatography-tandem mass spectrometry analysis (LC-MS/MS)
LC-MS/MS quantification methods were performed as described previously (14). In the LC-MS/MS analysis, an ultra-high performance liquid chromatography (UHPLC) system (ACQUITY UPLC, Waters, Milford, MA) was coupled to a linear ion trap-orbitrap mass spectrometer (Orbitrap Elite, Thermo Scientific). Solvent A of 2% acetonitrile and 0.1% formic acid in deionized water and solvent B of 0.1% formic acid in acetonitrile were used as the mobile phase for UHPLC separation. The metabolites were separated with a reverse phase column (HSS T3, 1.8 μm, 2.1 mm×100 mm, Waters, Milford, MA) at the flow rate of 500 μL/min using gradients of 40% solvent B at 0–1 min, 40–50% solvent B at 1–3 min, 50–80% solvent B at 3–6 min, 80–99.5% solvent B at 6–8 min, 99.5–99.5% solvent B at 8–9 min, 99.5–40% solvent B at 9–9.01 min and 40–40% solvent B at 9.01–10 min. The total chromatography separation time for each analysis was 10 min and the column temperature was set to 40 °C. The mass spectrometer was operated in the ESI negative-ion mode with an electrospray voltage of 2.5 kV, capillary temperature 360 °C and source heater temperature 350 °C. Mass spectra were acquired by cycling a full MS scan with a range of mass-to-charge ratios (m/z) of 275-360 and two product ion scans for 13,14-dihydro-15-keto-PGE2 (m/z 351.21) and 15-keto-PGE2 (m/z 349.2). The fragmentation reactions of m/z 351.21 to 333.207 and m/z 349.2 to 331.191 were selected for the quantitation of 13,14-dihydro-15-keto-PGE2 and 15-keto-PGE2. All data were acquired and processed using Xcalibur software (Thermo Scientific)
High-throughput compound screening (HTS)
High-throughput compound screening was conducted at the core service in Genomics Research Center (GRC) of Academia Sinica, Taiwan. The GRC 120K ReSet comprises more than 125,000 compounds, which were selected as representatives by structural similarity clustered from the 2M GRC compound library. The ReSet was arrayed in 1,536-well plates as single compounds at 1 mM in 100% DMSO. The quality of all compounds was assured by the vendor (purity is greater than 90%) and was verified internally with 5% random sampling. Recombinant human PTGR2 protein was purified and the screening was conducted using the NADPH-Glo Detection kit (Promega) to measure the amount of NADPH. The CV of HTS ranged from 4.8 % to 6.1 % with a Z' value of 0.7. The threshold was set to 1.5, resulting ~300 hits for further confirmation and determination of the half-maximal inhibitory concentration (IC50). Eight-point two-fold dilution of the compounds were prepared for IC50 determination and used in dose-dependent studies. The compounds showing dose-dependent increases of unused NADPH indicated the activities of recombinant human PTGR2 enzymatic activity.
Measurement of bone mineral density
The bone mineral density of cortical bone of femur were quantified and imaged using a benchtop Micro-CT imager (SkyScan 1076 in vivo, Bruker-MicroCT, Belgium) at 35 μm voxel image resolution with voltage of 50 kV, current of 200 μA, and exposure time of 140 ms, using a 0.5 mm aluminum filter.
Cold-induced and diet-induced thermogenesis
For the cold tolerance test, 24-week-old mice with matched average body weight from the two groups were placed individually on HFHSD in a 4°C chamber. The rectal temperature of the mice was measured after 0, 1, 2, 3, 4, 5, 6, 12, and 18 hours. For the diet-induced thermogenesis test, 24-week-old mice were fasted overnight for 18 hours. Then, their rectal temperature was measured at 0, 30, 60, 90, 120, 150, 180, and 240 min after HFHSD refeeding.
Statistical analysis
All values were expressed as mean ± S.E.M. All reported sample sizes were biologically independent values but not technically repeatedly measured values. Comparisons between two independent groups were performed using Student t-tests. Comparisons between multiple groups were conducted using one-way analysis of variance with post hoc analyses. Two-sided P-values < 0.05 were considered as statistically significant. Statistical analyses were conducted using GraphPad Prism 8.0 and SAS 9.0.