Mice
All animal protocols were approved by the Institutional Animal Care and Use Committee at Seoul National University (SNU-211209-4-1, SNU-230130-5, SNU-210403-2) and conducted in strict compliance with the guidelines for humane care and use of laboratory animals specified by the Ministry of Food and Drug Safety. Mice were housed at 22 ± 1°C and maintained on a 12-h light/12-h dark cycle with free access to food and water. Only male mice were used for experiments. C57BL/6N-Tm4sf19em1cyagen (TM4SF19 KO; Cyagen, KOCMP-20925-Tm4sf19) mice were established by the CRISPR/Cas9 system with the use of single guide RNA (5′-CTGACTCGTCTAGTGG-3′). The primers used for genotyping to confirm TM4SF19 KO were as follow: forward: 5’-TTAGAGGAAGTCCTTGAGACCCC-3’ and Reverse: 5’- ATCTGTATGACCGTTCACTTTGGA-3’.
Inducible macrophage-specific TM4SF19 KO (Tm4sf19flox/flox/Csf1r-CreER: TM4SF19 MKO) mice were generated by crossing Tm4sf19flox/flox mice (C57BL/6JSmoc-Tm4sf19em1(flox)Smoc, Shanghai model organism, stock#: NM-CKO-200286) with Csf1r-CreER (FVB-Tg (Csf1r-cre/Esr1*)1Jwp/J, Jackson laboratory, stock# 019098) mice. Tm4sf19flox/flox mice without Csf1r-CreER were used for wild-type (WT) control. For macrophage-specific TM4SF19 KO induction, tamoxifen (75mg/kg, Cayman, 13258) dissolved in sunflower oil was treated to Tm4sf19flox/flox/Csf1r-CreER mice and WT controls at 6 weeks of age by oral gavage on each of 5 consecutive days. Ten days after the last dose of tamoxifen treatment, mice were used for each experiment. To prolong the duration of Cre recombinase activity in mice, tamoxifen treatment was performed at the age of 6, 10, 14 weeks and mice were sacrificed after 12 weeks of HFD feeding. The genotyping primers to detect the flox or WT were as follows: forward: 5′- AGGGCAAAGAAGGAAGTGGCTAAT -3′, reverse: 5′-CAGGAAGGGGGCAGACAAGGAGT-3′. The genotyping primers to detect CreER were as follows: forward: 5′-CTTCCAAAGCATGGTCCAGT-3′, reverse: 5′-TGAACCAGCTCCCTATCTGC-3′.
For the diet-induced obesity model, mice at 6 weeks of age were fed a 60% fat diet (HFD; Research Diets, D12492) for 12 weeks. A standardized rodent pellet diet (NCD; Purina, 38057) was used for a control chow diet. Energy expenditure, locomotor activities, and food intake were estimated by indirect calorimetry system (PhenoMaster; TSE Systems). Body composition was measured by nuclear magnetic resonance scanning EchoMRI-700 (Echo Medical Systems).
For the glucose tolerance test, mice were fasted for 12 hours before measuring blood glucose levels. Mice were injected with d-glucose (2 g/kg body weights, Sigma, G7021) by intraperitoneal injection. For the insulin tolerance test, mice were treated with insulin by intraperitoneal injection (0.75 units/kg, Sigma, 91077C). Blood glucose levels were measured from tail vein blood samples at an indicated time using GlucoDoctor Top meter (Allmedicus, AGM-4100) and appropriate glucose indicator strips. For insulin tolerance test, mice were fasted for 6 hours and insulin was injected by intraperitoneal injection. To examine insulin signaling, insulin (0.75 units/kg) was injected into anesthetized mice by 2,2,2-tribromoethyl alcohol (avertin; Sigma, T48402) with the inferior vena cava and adipose tissues were harvested 10 min after the injection. To keep mice temperature at 37℃, we used the heating pad system (Toyotech, DP30-05A). For serum insulin, total cholesterol, and triglyceride levels, mouse insulin ELISA kit (Fujifilm, AKRIN-011T) and chemical blood analyzer (Fujifilm, DRI-CHEM 3500s) were used.
Bone marrow transplantation
Bone marrow transplantation (BMT) was performed with whole bone marrow cells from WT C57BL/6N and TM4SF19 KO mice. Bone marrow cells were prepared by flushing the tibia and femur with DMEM 51. Tail vein injection (2x106 cells) was performed on lethally irradiated 6-week-old recipient WT mice under anesthesia. After a 4 weeks recovery period, mice were initiated to be fed an HFD for 12 weeks.
Human
Human subcutaneous fat tissues were collected from Korea University Guro Hospital (IRB no.:2022GR0095), and tissues were stored at -80℃. The characteristic of human fat tissues from 24 individuals is available in Supplementary Table 2.
Histology
BAT, IWAT, and GWAT were harvested and fixed in 10% formalin (Sigma, HT50128) for 24 hours at 4°C and then embedded in paraffin. 5 µm paraffin sections were prepared from the paraffin-embedded tissue blocks. For hematoxylin and eosin (H&E) staining, deparaffinized sections were stained with ClearView Staining Hematoxylin (BBC Biochemical, MA010081) and Eosin Y Alcoholic solution (BBC Biochemical, 3610). The images were obtained with Nikon Elements (NIS BR Analysis ver 5.10.00). For F4/80 staining in liver and GWAT, 5 µm paraffin sections were deparaffinized with 2 x 10 min in 100% xylene (Daejung, 1330-20-7), 10 min in 50:50 xylene:ethanol, 5 min in 100% ethanol (Daejung, 4022–4410), 5 min in 95% ethanol, 5 min in 70% ethanol. After deparaffinization step, the slides were rinsed with distilled water, boiled for 10 min in citrate buffer (pH 6.0) for antigen retrieval, and cooled to room temperature for 10 min. Blocking was performed with PBS mixed with 3% BSA for 1hours, and F4/80 was stained with antibody. DAPI (1000x) was stained for 5min after antibody staining. Images were acquired with Zeiss confocal microscope (LSM800) and analyzed with Zen software (version 3.0).
Western blot and quantitative PCR
To perform western blot analysis, BAT, IWAT, and GWAT depots from WT and TM4SF19 KO mice were homogenized in PRO-PREP Protein Extraction Solution (iNtRON Biotechnology, 17081) containing SIGMAFAST Protease Inhibitor Cocktail (Sigma, S8820) and PhoSTOP phosphatase inhibitors (Roche, 4906845001) using tacoPrep Bead Beater homogenization system. The protein concentration of protein samples was quantified by BCA assay and measured by spectrophotometry (MultiSkan GO, 51119000, Thermo Fisher Scientific) at 562 nm using SkanIt software ver 5.0. 10µg protein samples were denatured in 5x sample buffer (Elpis Biotech, EBA 1052) in 95 ℃ for 5 mins and separated on 8% or 12% of SDS-PAGE gel and transferred to PVDF membrane (Bio-Rad). The membrane was blocked with 5% non-fat dry milk or BSA in TBST (Tris-buffered saline with 0.1% Tween 20) and incubated with primary antibody at 4°C overnight followed by horseradish peroxidase (HRP)-conjugated secondary antibody incubation at room temperature for 1 hour. Anti-rabbit TM4SF19 antibody (YT6290, WB 1:1000) were purchased from Immunoway. Anti-rabbit α/β-Tubulin (2148 ,WB 1:2000), anti-rabbit NF-ĸB p105/50 (13586 ,WB 1:1000), anti-rabbit ATP6V1B2 (14617, WB 1:1000), anti-rabbit LAMP1 (3243, WB 1:1000), anti-rabbit F4/80 (30325, IHC 1:200), anti-rabbit p-IRS1 (S612) (3203, WB 1:1000), anti-rabbit IRS1 (3407, WB 1:1000), anti-rabbit COXIV (4850, WB 1:1000), anti-rabbit Myc-Tag (2278, IP 1:200), and normal rabbit IgG (2729, IP 1:100) were purchased in Cell Signaling. Anti-rabbit ATP6V0B (NBP2-83943 WB 1:1000) was purchased in Novus Biological. Anti-rabbit ATP6V1A (GTX110815, WB 1:1000) was purchased in GeneTEX. Anti-mouse p-AKT (S473) (Sc-514032, WB (1:1000)) and anti-mouse AKT (Sc-81434, WB 1:1000) were purchased from Santa Cruz. Anti-mouse total oxphos cocktail (Ab110413,WB 1:1000) was purchased from Abcam.
For quantitative PCR (qPCR) analysis, total RNA from fat tissues from mice and humans were isolated by TRIzol reagent (Thermo Fisher Scientific, 15596018) according to the manufacturer’s protocols. High-Capacity cDNA Reverse Transcription kit (Applied Biosystems, 4368814) was used to synthesize cDNA and qPCR reaction was performed with iQ SYBR Green Supermix (Bio-Rad, 170–8884) in Bio-Rad CFX Connect Real Time PCR Detection system. β-Actin and Peptidylprolyl isomerase A (PPIA) were used as a housekeeping gene and primers used for qPCR are listed in Supplementary table 3. 2-ΔCt method was used to calculate the relative expression levels of each gene.
Flow cytometric analysis
GWAT from WT and TM4SF19 KO mice after feeding NCD or HFD were used for flow cytometric analysis. Adipose tissues were isolated and digested with filtered 2 mg/mL collagenase type I (Gibco, 17100-017) in KRBB buffer containing 3% BSA at 37°C. Fully dissociated preparations were then passed through a 100 µm cell strainer and centrifuged at 500 × g for 5 min. Pellets were then washed with DMEM containing 20% FBS two times. Stroma vascular fraction (SVF) stained with fluorescence labeled appropriate primary antibodies at room temperature. BV421 Rat Anti-Mouse F4/80 (565411, 1:50) were purchased from BD Biosciences. Human/Mouse TREM2 Allophycocyanin Mab (FAB17291A, 1:50) were purchased from R&D systems. APC/Cyanine7 anti-mouse CD45 (103116, 1:100), FITC anti-mouse/human CD11b (101206, 1:100), Brilliant Violet 421™ anti-human CD206 (141717, 1:100), PE/Cyanine7 anti-mouse CD11c (117317, 1:100) were purchased from Biolegend.
Unstained and single stained controls were used for compensation. LSRFortessa X-20 Flow Cytometer (BD Biosciences) was used to analyze the samples and the data was acquired by BD FACSDiva 8.0 software which then was further analyzed by Flowjo software (version 10.5.3; TreeStar). The gating strategies are provided in Supplementary Fig. 2.
Adipocyte fractionation and magnetically activated cell sorting (MACS)
To collect floating adipocytes, fully dissociated GWAT as described above were centrifuged at 300 x g for 5min. Red blood cells were lysed with RBC lysis buffer (15.5 mM NH4Cl, 1.2 mM NaHCO3 and 50 mM EDTA, pH 7.4) and pellets (SVF) were then washed with PBS with 3% BSA (500 x g for 5min). SVF were incubated with anti-F4/80-FITC antibody (Biolegend) for 2 hours at 4℃ followed by anti-FITC-microbeads (Miltenyi Biotec, 130-048-701, 1:10) incubation for 1 hour at 4°C. After incubation, F4/80 + cells were isolated by column (Miltenyi Biotec, 130-122-727) according to the manufacturer’s instruction (Miltenyi Biotec, MACS technique).
Cell culture
Dulbecco’s Modified Eagle Medium (DMEM; Welgene, LM001-07) supplemented with 10% fetal bovine serum (FBS; Gibco, 16000044) and 1% penicillin/streptomycin (PS; Welgene, LS202-02) were used for growth medium. RAW264.7 macrophages (ATCC, TIB-71) were maintained in growth medium until experiments were initiated. To check the localization of TM4SF19, we used lysotracker following manufacturer’s protocols. For colocalization analysis, the pearson correlation coefficient was measured by JACOP in ImageJ software (version 1.52a).
C3H10T1/2 cells (ATCC, CCL-226) were used to conduct in vitro adipocyte experiments. For adipogenic lineage commitment, C3H10T1/2 cells were exposed to 20 ng/mL bone morphogenetic protein 4 (BMP4) (R&D systems, 314-BP) in the growth medium with for 3 days. For adipogenic differentiation, the cells were incubated with the differentiation medium [growth medium containing 0.125 mmol/L indomethacin (Cayman Chemical, 70270), 2.5 mmol/L isobutyl methylxanthine (Cayman Chemical, I5879), 1 µmol/L dexamethasone (Cayman Chemical, D4902), 10 µg/mL insulin, and 1 nmol/L triiodothyronine (Cayman Chemical, 16028)] for 3 days. Then the cells were maintained with the growth medium containing 10 µg/mL insulin and 1 nmol/L triiodothyronine until the experiments performed.
Bone marrow-derived macrophages (BMDMs) were extracted from femur and tibia of 6 weeks WT and TM4SF19 KO mice 51. Briefly, the bone marrows from the femur and tibia were washed and resuspended by injecting 2mL of DMEM and centrifuge bone marrow suspension at 500 x g for 5 minutes at 4 ℃. Bone marrow was resuspended in 1mL of RBC lysis buffer and centrifuge at 500 x g for 5 minutes at 4℃. For BMDM differentiation, bone marrow cells were incubated with bone marrow culture medium [growth medium with 1ng/mL M-CSF1 (Peprotech Ltd, 315-02)] for 3days at 37℃, 5% CO2. On the third day, media was changed with fresh bone marrow culture medium for an additional 3 day of differentiation. LysoSensor Yellow/Blue dextran (Invitrogen, L22460) staining was performed to measure lysosomal pH following the manufacturer’s protocols. 1mg/mL of LysoSensor Yellow/Blue dextran was incubated with 24 hours, and 4% PFA was incubated 10 min for fixation. Images were acquired with Zeiss (LSM800) and analyzed with Zen software (version 3.0).
Subcellular fractionation
To collect the membrane and cytosolic fractions, we performed subcellular fractionation using the method described previously 54. A total of 2x10^8 cells were lysed and homogenized using fractionation buffer. The homogenates were then centrifuged at 500 x g for 10 minutes to clear the nuclei and intact cells. The resulting supernatant was collected and subjected to further centrifugation at 100,000 x g for 4 hours to isolate the cytosolic (supernatant) and membrane (pellet) fractions. The resulting pellet was lysed with RIPA buffer (Thermo Fisher Scientific, 89901). To confirm the purity of the isolated membrane and cytosol fractions, two marker proteins, LAMP1 and b-actin, were used for membrane and cytosolic fraction markers, respectively.
Lysosome isolation
Lysosome isolation was performed using a previously described method 55. A total of 2x108 cells were rinsed and collected with a fractionation buffer. The samples were then centrifuged at 1000 x g for 10 minutes, and collect the resulting supernatant. The supernatant was centrifuged at 20000 x g for 20 minutes and the resulting pellets were mixed with 19% diluted Optiprep density gradient medium (Biovision, M1248). The mixture was then loaded onto a low osmotic discontinuous density gradient with 27%, 22.5%, 19%, 16%, 12%, and 8% 55 and centrifuged at 150,000 x g for 4 hours. Subcellular fractions were eluted with RIPA lysis buffers (Thermo Fisher Scientific, 89901). To verify the purity of the lysosome fractions, the lysosome marker LAMP1 was tested in individual fractions by immunoblot analysis. It was found that fraction 2 (22.5%) was enriched with the lysosome. High-purity lysosome fractions (fraction 2) were used to measure V-ATPase activity by ATPase activity assay (Abcam, ab234055) following the manufacturer’s protocols. Briefly, 50µg of fresh lysosomal proteins were incubated with 5mM sodium azide (mitochondrial ATPase inhibitor) in the presence or absence of concanamycin A (ConA: specific inhibitor of V-ATPase) for 30 minutes at 37℃. The V-ATPase activity was normalized with experimental values in the absence of ConA and was then subtracted with control values in the presence of ConA.
Gene overexpression
Lentiviral transfer plasmid Tm4sf19 ORF was generated by inserting Tm4sf19 (NM_001160402.1) ORF clone to the transfer vector pLenti-EF1a-C-mGFP-P2A-Puro lentiviral vector (Origene, PS100121). Myc-tagged transfer plasmid was generated through GFP-fused TM4SF19 by replacing mGFP with myc-DDK. Lentivirus was produced by transfection of HEK293T cells with transfer plasmids (5 µg), psPAX2 (3.75 µg, Addgene, 12260), and pMD2.G (1.25 µg, Addgene, 12259) in 4:3:1 ratio using jetPRIME transfection reagent (Polyplus, 114 − 15). The medium containing viral particles was collected after 48 h of transfection, centrifuged, and filtered through a 0.45-nm syringe filter (Sartorius, STR.16555K). RAW264.7 macrophages were transduced with the viral supernatants containing 8 µg/mL polybrene (Santa Cruz, sc-134220) for 72 hours and were replaced with growth medium containing 2 µg/mL puromycin (Sigma, P8833) for selection for 48 hours.
Immunoprecipitation of Myc-tagged proteins
For immunoprecipitation analysis, RAW264.7 macrophages overexpressing Myc-fused TM4SF19 were harvested by lysis buffer (50 mM Tris-HCl (pH 7.6), 150 mM NaCl, 2 mM EDTA, and protease inhibitor). After 10 minutes of incubation on ice, lysates were centrifuged at 13,000 rpm, at 4℃ for 15 minutes and the pellet was discarded. 50 µL of Pierce™ Protein A/G Magnetic Beads (Invitrogen, 88802) were incubated with 5 µL of Myc and IgG antibodies for 2 hours rotating at 4℃. Beads were washed once in lysis buffer before incubation with 300 µg of lysate for 4 hours, rotating at 4℃. Beads were then washed five times in a buffer containing Tris-buffered saline (TBS) containing 0.05% Tween-20 Detergent. Bound proteins were eluted by 0.1M glycine (pH 2.0).
Phagocytosis assay
For phagocytosis analysis, fully differentiated C3H10T1/2 adipocytes were treated 5µg/ml brefeldin A (BFA; Biolegend, 420601) for 24 hours and were stained with 4,4-difluoro-5-(2-thienyl)-4-bora-3a, 4a-diaza-s-indacene3-dodecanoic acid (BODIPY 558/568 C12) (Invitrogen, D3835), and macrophages were labeled with Vybrant DiO Cell-Labeling Solution (Invitrogen, V22886) overnight. BODIPY-labeled C3H10T1/2 cells were detached, added to the DiO-labeled macrophages, and then co-cultured for 16 hours. Live cell imaging was performed with Operetta CLS High-Content Analysis System (Perkin Elmer) and analyzed by Harmony High-Content Imaging and Analysis Software (Perkin Elmer). For detection of high-resolution images of phagocytosis of dying adipocytes by the macrophages, fully differentiated adipocytes were treated 5µg/ml BFA(Biolegend) for 24 hours and then co-cultured with BMDM stained by Alexa488-cholera toxin B (CtB, Invitrogen, C22841) for 90 min. After co-culturing, fixation was performed with 4% PFA for 15 min and then LipidTOX (Invitrogen, H34476) was stained for 1 hour according to the manufacturer’s protocols. Images were acquired with Zeiss (LSM800) or Leica (TCSSP8) confocal microscope and analyzed with Zen software (version 3.0) or Leica software (LASX).
Promoter analysis
CiiiDER, an integrated computational toolkit for transcription factor binding analysis, was performed to analyze the promoter reigon (2.5kb) of mouse Tm4sf19 and human TM4SF19 gene (ENSMUSG00000079625, ENSG00000145107). Only match scores of 0.85 or above were accepted to retrieve the accurate transcription factor binding sites. For the chromatin immunoprecipitation (ChIP) analysis, MAGnify Chromatin Immunoprecipitation System kit (Invitrogen, 49-2024) was used. RAW264.7 macrophages were treated with LPS for 4 hours prior to crosslink. The macrophages were crosslinked with 1% formaldehyde for 10 minutes at room temperature and the reaction was stopped by adding 1.25 M glycine. Immunoprecipitation was performed using the antibody against NF-κB1 p105/p50 (Cell Signaling, 13586) or a normal IgG (Cell Signaling, 2729) as a negative control. To perform ChIP-qPCR analysis and gel-electrophoresis, precipitated DNA fragments were analyzed by specific primers for the putative NF-κB binding site bearing promoter region of TM4SF19 (Forward: 5′- AGGGGTGCTCTCTCAGAAG-3′ and Reverse: 5′-GCTCAACGGTCTTAGT-3′).
Single-nucleus RNA sequencing
Tissues were incubated in TST lysis buffer (146 mM NaCl, 10mM Tris-Cl pH7.5, 1mM CaCl2, 21 mM MgCl2, 0.03% Tween-20, 0.01% BSA) for 10 minutes at 4℃ and minced using Noyes spring scissors. Samples were filtered through a 70µm strainer and centrifuged at 500 x g for 5 min at 4℃. Pellets were washed once with TST lysis buffer and twice with 1% BSA/PBS. Pelleted nuclei were resuspended in 1% BSA/PBS with 0.2U/µl RNase inhibitor and filtered through a 30µm strainer. Nuclei were stained with acridine orange/propidium iodide solution and counted using an automated counter (LUNA-FX7, Logos Biosystems, L70001).
For sample multiplexing, 1–2 x 106 nuclei per a sample were stained with 100µl of CellPlex (10X Genomics, PN-1000261). Samples were washed three times with 2ml of cold 1% BSA/ PBS and centrifuged at 500 x g for 5 min at 4℃. Nuclei were resuspended in 1% BSA/PBS and equal numbers of cells from each biological replicate for a condition were mixed. Nuclei were counted and added to the reverse transcription mixture with the aim of capturing 20,000 nuclei of the combined sample.
Libraries for single-nucleus RNA sequencing were generated using the Chromium Next GEM Single Cell 3’Kit (10X Genomics, PN-1000269), 3’Feature Barcode Kit (10X Genomics, PN-1000262), Dual Index Kit TT Set A (10X Genomics, PN-1000215), and Dual Index Kit NN Set A (10X Genomics, PN-1000243). To generate gel bead-in-emulsions (GEMs), samples were loaded onto the Chromium Next GEM Chip G (10X Genomics, PN-2000127) and the subsequent microfluidic process was performed on a Chromium controller (10X Genomics). Reverse transcription was performed by incubating GEMs in a thermal cycler (C1000 Touch Thermal Cycler with Deep Well, Bio-Rad). After incubation, GEMs were disrupted and pooled cDNA was purified using silane magnetic beads. Amplification of cDNA was performed using a thermal cycler. During cDNA size selection using SPRIselect beads, the supernatant and bead pellet portion were handled separately to obtain cDNA from poly-adenylated mRNA and DNA from cell multiplexing oligo feature barcode. To generate cell multiplexing libraries, 75µl of supernatant was retained and further purified. The cell multiplexing barcode DNA and indexing primers (10X Genomics, PN-3000482) were added to the PCR reaction mixture and PCR amplification was performed. For cDNA library generation, bead pellets were purified and cDNA was eluted. Subsequent library construction procedures, including enzymatic fragmentation, end repair, A-tailing, adaptor ligation, and sample index PCR, were performed according to the manufacturer’s protocol.
Size and concentration of cDNA and libraries were analyzed using a Bioanalyzer (Agilent). Libraries were pooled and sequenced on a HiSeq X Ten sequencer (Illumina) using a 100bp paired-end protocol to generate a minimum of 20,000 read pairs per cell for 3’gene expression library and 5,000 read pairs per cell for the cell multiplexing library.
Single-nucleus RNA sequencing data pre-processing
Raw reads from multiplexed single-nucleus RNA sequencing data were aligned to the mouse genome (mm10) and demultiplexed by sample-specific barcodes using cellranger (v6.1.1). The raw count matrix for each sample was generated using cellranger multi with min-assignment-confidence = 0.7, expect-cells = 20000 and include-introns = true options. R (v4.1.0) was used for analysis. Low-quality cells with detected UMIs lower than 1,000 or percentage of genes mapped to mitochondrial genes higher than 5% were filtered out for further analysis. The threshold was determined by visually checking the distribution of per-cell UMI count using addPerCellQC function of scater (v1.22.0) R package 56. All count matrices were combined and cells were clustered using quickCluster function of scran (v1.22.1) R package 57. Cell-specific size factors were computed using computeSumFactors function of the same package. The normalized count matrix was generated with logNormCounts function using the calculated size factors with pseudo_count = 1. To select highly variable genes (HVGs), variance of normalized expression for each gene was modeled using modelGeneVar and getTopHVGs function of scran R package with fdr.threshold = 0.05. The dataset was scaled and centered using ScaleData function of Seurat (v4.0.5) 58. The top 25 principal components (PCs) are computed on HVGs and used for clustering and UMAP dimension reduction. To identify molecular characteristics of each cluster, marker genes of each cluster were obtained using FindAllMarkers function of Seurat. Cell types were manually annotated in resolution of clusters based on identified molecular characteristics. Putative doublets were confirmed using scDblFinder (v1.8.0) R package 59. These cells were manually double-checked whether they expressed two or more cell type markers from different lineages and removed. Cells expressing erythroid cell markers such as Hba-a1, Hba-a2 and Alas were removed. There were some clusters presumed to be derived from epididymis, anatomically close to the GWAT. These cells were annotated as epididymal cells (Abcb5, Ces5a, Adam7), spermatozoas (Dnah12, Spef2, Hydin) and efferent duct cells (Adcy8, Dnah5, Slc9a3) and removed for further analysis. The remained cells after quality control were re-clustered using the same functions described above with the top 15 PCs. The batch effect caused by different samples was corrected by RunHarmony function of harmony (v0.1.0) R package 60.
Single-nucleus RNA sequencing data analysis
Trajectory analysis
Pseudotime analysis was conducted using Palantir (v1.0.0) python package for monocyte/macrophage and APC/Adipocyte cell types 61. To construct pseudotime trajectory on tSNE plot, the variable features were selected using getTopHVGs function and renormalized count matrix of intended populations was extracted. For monocyte and macrophage lineage trajectory, the start_cell was chosen randomly in the monocyte cluster. The first 150 PCs were used to make a diffusion map using run_diffusion_maps with n_components = 10 and determine_multiscale_space function with the default options. Pseudotime and differentiation trajectory were obtained using run_palantir function with num_waypoints = 2000. The coordinates of tSNE on diffusion components were computed using run_tsne function with perplexity = 700. The constructed trajectory was visualized on tSNE plot using plot_palantir_results function. After that, to find the more reliable starting cell, the cell which has the lowest pseudotime was selected as a start_cell and the above process was repeated. For adipose lineage trajectory, the start_cell was designated as one of the cells expressing Dpp4 and Cd34 (Figure S2F). Trajectory was constructed following the same methods as mentioned above except for these options: the number of used PC = 150, num_waypoints = 5000 for run_palantir function and perplexity = 900 for run_tsne function. The start_cell was selected again as described above.
Monocle3 (v1.3.1) was used for confirmation of monocyte to macrophage trajectory 62,63. We used same HVGs and normalized count matrix of monocyte and macrophage populations as used in Palantir. We preprocessed the data using preprocess_cds function with 20 PCs and reduced dimension using UMAP. Clustering was performed using cluster_cells function with resolution of 0.0001. The trajectory was constructed using learn_graph with default parameters.
Cell type composition
To calculate the cell type composition, the number of cells in each cell type for each sample or condition was divided by the total cell number of each sample. The total count of the cell in each sample was scaled as 100%.
DE (Differential Expression) and GO (Gene Ontology) analysis
For publicly available transcriptomic database analysis, differentially expressed genes (DEGs) between different body fat conditions (NCD vs HFD and lean vs obese) were demonstrated by iDEP 9.664 and GEO2R provided by NCBI. The significant DEGs of top 100 upregulated genes in each dataset were determined by adjusted P-value < 0.05 and fold change > 2. DAVID (https://david.ncifcrf.gov) was used for functional enrichment analysis 65. DEGs were determined between Adipocyte.1 and Adipocyte.3 using limma (v3.50.0) R package 66. Briefly, lmFit function was used to fit the model on the normalized count for each gene and batch effects were considered using duplicateCorrelation function by adding sample information to the block option. Statistical test was conducted using eBayes function with trend = T and robust = T options. The significant DEGs (adjusted P-value < 0.05) between Adipocyte.1 and Adipocyte.3 were used as input. The DEGs up-regulated in Adipocyte.1 and up-regulated in Adipocyte.3 were separately used for each cluster-enriched pathway analysis.
For gene set enrichment analysis (GSEA), DEGs between the WT HFD and TM4SF19 KO HFD conditions were obtained using FindMarkers function of Seurat R package and Molecular Signatures Database (MSigDB) was downloaded using msigdbr (v7.4.1) R package. DEGs were sorted by average log2 fold change and applied for GSEA using fgsea (v1.21.2) R package.
Calculating Signature score
Trem2 + macrophage and Lyve1 + macrophage signature scores were calculated using AddModuleScore function of Seurat. Up-regulated DEGs were used as signature genes. DEGs were determined between Lyve1 + macrophage and Trem2 + macrophage in wild-type mice (WT NCD, WT HFD), filtered with adjusted P-value < 0.05. Mac1 and Mac3 signature scores were computed using same function. The DEGs between Mac3 (Trem2-expressing macrophage) and Mac1 (Lyve1-expressing macrophage) was obtained from the previous study (Jatin et al., 2019) and filtered with p-value < 0.05 and log2fc > 1. The significance was determineded by MASC (v0.0.0.9000) R package 67 and batch was considered as random effect.
Co-expression network analysis
The co-expression gene modules were obtained using hdWGCNA (v0.2.04) R package, which was applied after sub-clustering the monocyte and macrophage population of the WT HFD condition. 1837 metacells were constructed using the MetacellsByGroups function with k = 25 parameter and genes expressed in at least 5% of the cells used for downstream analysis. Co-expression gene network was constructed using the ConstructNetwork function with soft power threshold of 3, chosen using the TestSoftPowers function. DEGs between WT HFD and TM4SF19 KO HFD conditions of monocyte and macrophage population were obtained using FindMarkers function of Seurat R package, and down- or up-regulated genes were determined with adjusted P-value < 0.05 and |log2fc| > 0.2.
Statistics
GraphPad Prism 7 software (GraphPad Software, USA) was used for statistical analysis and correlation analysis. Data are presented as mean ± standard errors of the mean (SEM) or mean ± standard deviation (SD) as indicated in the Figure Legends. National Institutes of Health ImageJ software (version 1.52a) was used to quantify the intensity of immunoblot and immunostaining.