Mice.All animal studies were carried out following protocols approved by the Animal Care and Institutional Biosafety Committee of the University of Illinois at Chicago. Mice were maintained under standard conditions (standard diet and water) at 23 °C and ~60% humidity with 12 h light and 12 h dark cycles. We purchased C57/BL6J mice from Jackson Laboratory (#000664) as control wild type (WT) mice. KPC (LSL-KrasG12D/+: LSL-Trp53R172H/R172H: Pdx1-Cre) mice were generated by crossing LSL-KrasG12D/+:LSL-Trp53R172H/R172H mice with Pdx1-Cre homozygous mice34. We received KPC mice from Dr. Paul Grippo at University of Illinois at Chicago. Both sexes of mice were used for all experiments.
For tumor syngeneic allograft assay,mouse skin melanoma B16F10 cells (ATCC CRL-6475, 106 cells/100 µL PBS) were implanted subcutaneously in the dorsal flank of WT mice (C57BL/6J). Control mice were injected with PBS. The mice were monitored every other day to measure tumor growth with electronic caliper and body weight for 3 weeks and tumor size was presented with tumor volume which was calculated with equation of width2 x height x 0.523 as previously described78. For lineage tracing of muscle endothelial cells during distal tumor growth, we used inducible endothelial specific tdTomato expressing mice (tdTomato flfl:Cdh5-CreERT2, ECtdTomato)49. To induce the Cre recombinase in tdTomato fl/fl: Cdh5-CreERT2 mice, 8 weeks ECtdTomato mice were intraperitoneally administrated with tamoxifen (100 mg/kg, body weight, Sigma-Aldrich T5648) once a day for consecutive three days. One month later, the mice were used for tumor syngeneic allograft assay.
For rescue experiments of overexpression of EC specific PGC1α, we generated lentivirus expressing mouse PGC1α under endothelial specific Cdh5 (CD144) promoter. This lenti-Cdh5-mPGC1α-EGFP virus specifically express mPGC1α-EGFP in vascular ECs. Thus, the mice were injected intramuscularly with total 200 µL lentivirus (1x109 pfu/µL, 40 µL per injection site) at 5 different areas of tibialis anterior and gastrocnemius right after tumor implantation. Lentivirus expressing empty vector were used as a control. All mice were examined in 3 weeks after tumor implantation.
Mouse grip force and fatigue.The 4 limbs grip strength of mice was measured five times for each mouse using a Bio-GS3 Grip Strength Test Meter5 and the data are presented as averages after normalization against tibial length, which is not altered by muscle loss79,80. The fatigue of mice was evaluated using the values of grip force. The degree of fatigue was estimated by comparing first two pulls with the last two pulls (formula (4+5)/(1+2) which would theoretically be 1 in mice that show no fatigue)81 andpresentedwith fold change of decrease of original force.
Mouse skeletal muscle collection.All mice were measured body weight and tibial length before dissecting tissues with digital caliper. Skeletal muscles (gastrocnemius, tibialis anterior, or quadriceps) from both hindlimbs were dissected and immediately weighed using a microelectronic weighing scale (Accuris instruments, W3100A-120) and then directly embedded in tissue plus O.C.T. compound (Fisher) on dry ice for cryosection. For paraffin blocking, the tissues were fixed with 10% neutral buffered formalin overnight and changed with 70% ethanol following paraffin embedding. The muscle mass (weight, g) was normalized with tibial length (mm). For Western blotting or RT-qPCR, the muscles were immediately frozen in liquid nitrogen and stored at -80 °C until use.
Muscle clearing and 3D imaging. Gastrocnemius muscles were freshly harvested and fixed with 2% PFA for 10 min at RT. After washing in PBS, the muscles were longitudinally sectioned to 400 µm thickness using a vibratome (Leica, VT1200S) and stained with primary rat anti-mouse CD31 antibody (BioLegend, 102502, 1:1000 dilution) for 18 h at 4 °C. Then, the tissues were incubated with fluorescent secondary anti-rat IgG2 antibody at a 1:100 dilution for 18 h at 4 °C. The secondary antibody was prepared by conjugating anti-rat IgG2 antibody (BioLegend, 407502) with DyLight 633 (Thermofisher, 46414) at a 1:10 ratio for 18 h at 4 °C. For optical tissue cleaning, the muscle tissues were incubated in a gradient D-fructose solution (20% for 30 min, 80% for 30 min, then 100% for 1 h) at room temperature (RT). The 3D images were taken by using a confocal fluorescence microscope (Caliber ID, RS-G4) with a 40x oil objective (Olympus UPLXAPO 40XO, NA 1.4, 0.13 mm WD). The light source was 640 nm excitation (Toptica iChrome MLE-LFA 50 mW diode laser) with a 630/69 nm filter (Semrock) for DyLight 633. The images were reconstructed for visualization with Imaris Viewer 64-bit version 9.6.0. and the surface volume of vessels was analyzed with Imaris 64-bit version 7.2.2.
Mouse skeletal muscle endothelial cell isolation. Skeletal muscles (gastrocnemius, tibialis anterior, or quadriceps) from hindlimbs were dissected, minced, and digested with digestion buffer (2 mg/mL collagenase A, 1 mg/mL dispase II, 0.1 mg/mL DNase I in 1x PBS without Ca2+/Mg2+) at 37 °C for 30 min with gentle shaking. At the end of the digestion process, the tissue was titrated using 18 G needles in syringes up and down 5 times and the cell suspension was filtered through 100 µm disposable cell strainer into a fresh 50 mL tube and centrifuged at 300x g for 5 min at 4 °C. The cell pellet was resuspended with wash buffer (20% FBS, 2 mM EDTA in 1x PBS without Ca2+/Mg2+) and filtered through 70 µm disposable cell strainer into a fresh 50 mL tube and centrifuged at 300 xg for 5 min 4 °C. This step was repeated with 40 µm disposable cell strainer into a fresh 50 mL tube. The cells were then resuspended with antibody binding buffer (0.5% BSA, 2 mM EDTA, 1% FBS in 1x PBS without Ca2+/Mg2+). To evaluate relative number of muscle EC in total muscle cells, the same number of cells (106 cells/100 µL) were stained with specific antibodies of CD31 (EC marker, eBiosciences 14-0311082) and CD45 (immune cell marker, BioLegend 103108) for 30 min on ice. Samples were run through a Gallios flow cytometer (Beckman Coulter, Pasadena, CA) and analyzed by Kaluza software (Beckman Coulter). Because some immune cells also express CD31, leukocytes and dead cells were excluded by CD45+ and DAPI+ gating and the CD31+CD45-DAPI- cells were considered as pure ECs and represented by the percent (%) of total muscle cells. The muscle ECs (CD31+CD45-DAPI-) from single cell suspension of total muscle cells were obtained by FACS sorting using MoFlo Astrios (Beckman Coulter) or also isolated using mouse CD31 microbeads (130-097-418, Miltenyi Biotec) and MS column (130-042-201, Miltenyi Biotec) with magnetic separator for some experiments.
Immunofluorescence imaging using confocal microscopy. 1. Muscle tissue imaging for cryosection- The 7 µm cross-cryosections of muscle were dried for 5 min at RT and washed with 1x PBS for 5 min. The sections were blocked with blocking buffer (1x PBS, 2% BSA, 0.05% Tween 20, and 5% goat serum) for 1h at RT and then stained with EC specific CD31 antibody (BD biosciences, 550274, 1:25 dilution) or muscle base membrane laminin antibody (Sigma, L9393, 1:100 dilution) overnight at 4 °C in a humidified chamber. The secondary antibodies were used with goat anti-Rat IgM cross-adsorbed secondary antibody DyLight 488 (Thermo fisher, SA5-10010, 1:400 dilution) or goat anti-Rabbit IgG (H+L) cross-adsorbed secondary antibody Alexa fluor 633 (Thermo fisher, A21070, 1:500 dilution) for 1h at RT, respectively. The nuclei were stained with DAPI. 2. Muscle tissue imaging for paraffin section- The 4 µm cross-paraffin sections of muscle were deparaffinized and hydrated at RT as following; 100% Xylene1, 5 min →100% Xylene2, 5 min →100% EtOH, 3 min →95% EtOH, 3 min →70% EtOH, 3 min →50% EtOH, 3 min → distilled water, 5 min → TBST (0.1% Tween 20), 5 min. For antigen retrieval, the slides were boiled in Sodium Citrate retrieval buffer (pH 6.0, 0.05% Tween 20) for 30 min and cooled for 20 min at RT. After washing with 1x PBS, the slides were permeabilized with 0.2 % Triton X-100 for 10 min at RT and followed by blocking with blocking buffer (1X PBS, 2% BSA, 0.05% Tween 20, and 5% goat serum) for 1 h at RT and then stained with EC specific biotinylated isolectin B4 (Vector lab, B-1205-.5, 1:100 dilution) overnight at 4 °C and followed by staining with FITC-streptavidin (Invitrogen, 11-4317-87, 1:400 dilution) for 1 h at RT. The images were taken by fluorescence microscopy or confocal microscopy (LSM880, x20 objective).3. In vitro cell imaging- The primary endothelial cells were cultured on coverslips in 6 well plates. After treatment with lenti-shRNA virus for indicated times, the cells were fixed with 4% PFA for 10 min at RT and then blocked with blocking buffer (1X PBS, 2% BSA, and 0.05% Tween 20) without permeabilization for 1 h. EC barrier integrity was visualized by immunostaining with 1:250 dilution of Alexa Fluor647 mouse anti-human CD144 (BD561567) for 1 h at RT. After mounting with antifade reagent with DAPI (Vector lab, H-1800-10), the images were taken using confocal microscopy (Zeiss LSM880, Plan Apo 1.46NA, 63x objective).
Human abdominal muscle sections. We obtained human muscle sections from de-identified FFPE blocks from the Pathology archives through the UIC IRB-approved Pathology Biorepository. Data was not collected via interactions/interventions with individuals for research purposes, and there was no use of private, identifiable information about subjects. The paraffin sections (4 µm) were stained with H&E by UIC histology core laboratory. The images were taken using Olympus BX51/IX70 microscopy (x20 and x40 objectives). Human endothelial cells in muscles were stained with biotinylated Ulex Europaeus Agglutinin I Lectin (UEA I, B-1065-2, Vectorlab, 1:100 dilution) and FITC-streptavidin (11-4317-87, Invitrogen, 1:250 dilution). Nuclei was stained with DAPI. The images were taken using confocal microscopy (LSM880, x20 objective).
Muscle capillary permeability assay.WT and tumor bearing mice for 3 weekswere anesthetized with Ketamine/xylazine and retro-orbitally administrated with FITC-albumin (250 mg/kg in PBS)82. After 10 min, circulating FITC-albumin was washed away by PBS perfusion and skeletal muscles (gastrocnemius and Tibial anterior) were immediately dissected and directly embedded in tissue plus O.C.T. compound on dry ice for cryosection. The frozen muscles were sectioned with 50 µm thickness using cryostat and dried for 5 min at RT, followed by PBS wash and directly mounted with antifade reagent with DAPI (Vector lab, H-1800-10). The FITC-albumin fluorescence images were taken by Z-sectioning using confocal microscopy (LSM710, X20 objective) and reconstructed for visualization using Imaris 64 bit 7.2.2. The intensity of FITC-albumin fluorescence in muscle was measured using Image J (1.52d, Java 1.8.0_172[64 bit]).
Muscle hypoxia assay.WT and tumor bearing mice for 3 weekswere intraperitoneally injected with hypoxic probe pimonidazole (100 mg/kg, Hypoxyprobe, Hypoxyprobe plus kit) 1 h before harvesting muscle83. The paraffin muscle sections were used to measure the extent of hypoxia in gastrocnemius muscle by following the manufacturers’ guide. Nuclei were stained with DAPI and the images were taken using confocal microscopy (LSM880, X20 objective).
In situTerminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. To determine apoptotic endothelial cells in muscles from KPC mice, the paraffin sections were deparaffinized, hydrated, and antigen retrieval was performed as described above. The apoptotic cells in sections were determined using TUNEL assay kit for in situ apoptosis detection (Invitrogen, C10618). After finishing TUNEL staining, the sections were stained with EC marker isolectin B4 (IB4) for overnight and followed FITC-streptavidin staining for 1h at RT. Nuclei were stained with DAPI and the images were taken by confocal microscopy (LSM880, x20 objective).
FACS analysis for infiltrated immune cells in muscle.Briefly,mixed skeletal muscles (gastrocnemius, tibialis anterior, or quadriceps) from hindlimbs of control and tumor bearing mice were dissected, minced, and digested with digestion buffer (2 mg/mL collagenase A, 1 mg/mL dispase II, 0.1 mg/mL DNase I in 1x PBS) at 37 °C for 30 min with gentle shaking. Single cell suspensions were counted and the same number of cells (106 cells/100 µL) were stained with 1:100 dilution of anti-CD45 (157610, BioLegend) antibody and DAPI. Samples were run through a Gallios flow cytometer (Beckman Coulter, Pasadena, CA) and analyzed by Kaluza software (Beckman Coulter). The inflammatory response (CD45+DAPI-) cells were represented by the percent (%) of total muscle cells.
Blood plasma Activin level by ELISA. Mouse whole blood was slowly withdrawn by cardiac puncture and collected in anti-coagulant (0.5 M EDTA, 5 µL/100 µL blood) containing tubes. The blood was centrifuged in 2000x g at 4 °C for 15 min and plasma was collected in new tubes and stored at -80 °C until use. Activin-A levels in plasma were measured with 100 µL plasma for each sample using ELISA Kit (DAC00B).
shRNA, plasmids, or lentivirus production.Five lenti-shPGC1α RNA constructs were purchased from Sigma (TRCN0000364084, TRCN0000364085, TRCN0000364086, TRCN0000001166, TRCN0000001167) and knockdown efficiency was examined in ECs. The TRCN0000001166 clone showed the best effect to deplete PGC1α in ECs and we used this clone for experiments. pcDNA4-myc-PGC-1α (Plasmid #10974) and PGC-1α promoter luciferase delta CRE (Plasmid #8888) were purchased from Addgene. pRL/TK (renilla-luciferase) was provided by Dr. Chinnaswamy Tiruppathi at University of Illinois at Chicago. Mouse endothelial specific lenti-viral vector for PGC1α (pLV-EGFP-Cd144_mPpargc1α [NM_008904.2]) were designed and produced by Vector builder. Lenti-viruses were produced in HEK293T cells (CRL-11268, ATCC) by transfection with DNAs (2.5 µg pMD2.G, 5 µg of psPAX2, and 7.5 µg of DNA expression vector) with 30 µg polyethylenimine (PEI, Polysciences, 23966, USA), and concentrated with Lenti-X concentrator (CloneTech, 631232) as described previously84.
Cell culture.Humanlung microvascular endothelial cells (HLMVECs, CC-2527, Lonza) were obtained from Lonza and cultured with EGM2 (Lonza) including all supplements and 10% FBS (Hyclone) until passage 8. The cells were transduced with lenti-shPGC1α RNA virus with 1:2000 dilution polybrene (Millipore, TR-1003-G) for 24 h and then media was changed. After 72 h from virus infection, the cells were used for experiments. For Activin treatment, the cells were starved with 2% FBS media overnight and treated with Activin-A (R&D system, 338-AC-010, 25 ng/mL or 50 ng/mL) for indicated times.
Cell apoptosis. To evaluate cell apoptosis, the cells were stained with Annexin-V-FITC and PI (Bio-Rad, ANNEX20F) following FACS analysis using Gallios flow cytometer (Beckman Coulter, Pasadena, CA). The data was analyzed by Kaluza software (Beckman Coulter). The apoptotic cells (Annexin V-FITC+PI+) were presented with percent (%) of total cells.
Western Blotting.The cells were lysed with lysis buffer (50 mM HEPES pH7.5, 120 mM NaCl, 5 mM EDTA, 10 mM Na pyrophosphate, 50 mM NaF, 1mM Na3VO4, 1% Triton X-100). The concentration of protein was measured with Bradford protein assay solution (Bio-Red, 5000006) and the same amount of total protein was loaded in SDS-PAGE for Western blotting and probed with specific antibodies; PGC1α (NOVUS, NBP1-04676, 1:1000 dilution), Actin (Santa Cruz, sc-517582 HRP, 1:1000 dilution), or VE-cadherin (Cayman #160840, 1:1000 dilution). Quantitative analysis of Western blotting was performed using Image J (1.52d, Java 1.8.0_172[64 bit]). All raw gel blots are available in the Source data file.
Quantitative real-time PCR.Total RNA was isolated by using TriZol Reagent (Invitrogen, 15596026). The muscle tissues were lysed with TriZol and homogenized in safe-locked 1.5 mL tubes with metal beads (Next advance, SSB14B) using tissue homogenizer (Next advance, bullet Blender, BBX24B-CE). Reverse transcription was carried out using high-capacity cDNA reverse transcription kit (Applied Biosystems, 4368814) using 1-2 µg of total RNA. Quantitative PCR for human genes (PGC1α, Mcl1, Bcl2, Snail, Vimentin, VCAM1, Caspase1, IL6, or 18S) and mouse genes (PGC1α, MuRF1, Atrogin1, Twist, Glut1, ICAM1, TNFα, IRF7, IL-10, IL-1β, IL-6, or PPIA) was performed in duplicates with fast start universal SYBR Green master (ROX) PCR kit (Roche, 04913914001) using QuantStudio7 (Thermofisher). Expression of genes was normalized and expressed as fold-changes relative to 18S or to PPIA. A complete list of all primers is available in the Source Data files.
PGC1α promoter luciferase assay.ECs were transfected with 1 µg of a PGC1α promoter luciferase(Addgene # Plasmid #8888) and 35 ng of pRL/TK using PEI transfection reagent (Polyethylenimine). 48 h after transfection, the cells were stimulated with vehicle (0.1% BSA), TNFα (10 ng/mL), or Activin-A (25 ng/mL) for 16 h and then 100 µL of cell lysate from each sample was used to measure reporter gene expression. Firefly and Renilla luciferase activity were determined by the dual luciferase reagent assay system (Promega). The relative luciferase activity represents the mean value of the firefly/Renilla luciferase.
Chromatin immunoprecipitation assay.Confluent ECs in 100 mm dishes were fixed with 1% PFA for 10 min at RT and washed with 1x cold PBS. To quench unreacted PFA, the cells were incubated with 1x Glycine for 5 min at RT and washed with 1x cold PBS. The nuclear fractions were obtained by using EZ-Magna Chip A/G kit (Millipore, #17-10086) and then followed by DNA shearing using Covaris (bath temperature 7.5 °C, acoustic power 3 W, duty cycle 2%, time 60 sec, cycle number 200). The sonicated nuclear fractions were centrifuged at 10k xg at 4 ⁰C for 10 min and then chromatin immunoprecipitation (ChIP) assay was followed with manufacturers’ guidance for EZ-Magna Chip A/G kit (Millipore, #17-10086) with minor modification. Each 50 µL aliquot was used for immunoprecipitation with 1 µg antibodies for normal IgG (negative control), RNA polymerase (positive control) or PGC1α antibody (NOVUS, NBP1-04676) for 3 h. For input control, 5 µL supernatant was aliquot and stored at 4 ⁰C until protein/DNA elution. The complex of protein and DNA (both input and immunoprecipitated samples) was incubated in elution buffer with proteinase K and RNase A at 37 ⁰C for 30 min and further incubated at 62 ⁰C overnight and then denatured at 95 ⁰C for 10 min. The purified DNA (2 µL) was enriched by qPCR (initial denaturation 94 ⁰C 10 min, 1 cycle; denature 94 ⁰C 20 sec, anneal and extension 60 ⁰C 1 min, 50 cycles) with primers for putative binding motifs of PPARγ on VE-cadherin (Cdh5) promoter. The gene amplified values were normalized with input values and presented with fold enrichment of normal IgG (negative control).
Bulk RNA-seq and computational data analysis.ECs from skeletal muscles (gastrocnemius, tibialis anterior, or quadriceps) from hindlimbs were isolated and stained with specific antibodies of CD31 and CD45, and pure muscle ECs (CD31+CD45-DAPI-) were sorted using MoFlo Astrios (Beckman Coulter) as mentioned above. The sorted ECs were used for total RNA extraction by TRIzol Reagent (Invitrogen, 15596026) and the RNA was treated with DNase and its quality was evaluated with gel QC. Bulk RNA sequencing for 4-6 samples (each sample indicates a mouse) with 4 time points was performed with oligo-dT mRNA directional using Illumina Novaseq (HiSeq SR50) for coding genes at Genomic Facility in University of Chicago. Sequenced reads were aligned to the Mus musculus reference genome GRCm39 (mm39) with STAR v.2.7.6a85. Then mRNA expression counts were quantified from the aligned reads by using STAR –quantMode option. Genes were annotated using biomaRt R package86. We applied calcNormFactors function from edgeR R package87 to normalize the counts. Principal component analysis (PCA) was performed after normalization. Dynamic differentially expressed genes (DDEGs) were identified using TrendCatcher46 with adjusted dynamic p-value <0.05 threshold. For each DDEG, we calculated its accumulated log2 fold change (log2FC) over the time compared to its baseline expression. Then, we performed Gene Ontology (GO) enrichment analysis on both positively and negatively accumulated log2FC DDEGs respectively using clusterProfiler R package88, and picked the top 10% most enriched dynamic biological pathways. After removing redundant GO terms, for each biological pathway we calculated the averaged accumulated log2FC (GO_mean_logFC) from its corresponding DDEGs to infer the biological process accumulative change. Based on the pathway accumulative change ranking, we selected top 10 positively and negatively changed biological pathways respectively. To show how these GO enrichment change over time, we applied draw_TimeHeatmap_GO() function from TrendCatcher. To build the TimeHeatmap, TrendCatcher first check all the DDEG’s changing direction from its inferred trajectory within each time interval (either up or down). Then it calculated GO enrichment analysis for both up and down directed genes within that time interval. For each GO term within a corresponding time window, it also calculated a series of log2FC (Avg_log2FC) gene expression values over time, to quantify the trajectory dynamics of each biological pathway. Then we subset the TimeHeatmap object using draw_TimeHeatmap_selGO() function to show only the top 20 GO terms with the highest accumulative change.
Single cell cDNA library preparation.ECtdTomato mice with or without tumor for 3 weeks were used to dissect skeletal muscles (gastrocnemius, tibialis anterior, or quadriceps) from hindlimbs. The muscle tissues were minced and enzymatically digested with digestion buffer (2 mg/mL collagenase A, 1 mg/mL dispase II, 0.1 mg/mL DNase I in 1x PBS) at 37 °C for 30 min with gentle shaking as mentioned above. Control samples were pooled from 3 mice and melanoma samples were pooled from 4 mice. The single muscle cell suspension was sorted by tdTomato+DAPI- (ECs) and tdTomato-DAPI- (non-ECs) using MoFlo Astrios (Beckman Coulter). The sorted cells were loaded into a 10x Genomics microfluidics chip and encapsulated with barcoded oligo-dT–containing gel beads using the 10X Genomics Chromium controller according to the manufacturer’s instructions. Cell viability was more than 90% with a target of sequencing 6000 cells. Single-cell libraries were constructed using a Chromium Single Cell 3’ reagent kit v3.1 according to the manufacturer’s instructions. The quality of cDNA libraries was checked before sequencing and the cDNA libraries were multiplexed into one lane for sequencing on NovaSeq S1 (28 × 91bp paired-read).
scRNA-seq data processing and analysis. Raw counts tables were generated for each sample with Cell Ranger (v6.0.2) using default parameters and the 10x mm10-2020-A reference. A total of 24107 cells passed quality control (muscle ECs: control tdTomato positive (+) 4046 cells, melanoma tdTomato positive (+) 7123 cells, muscle non-ECs: control tdTomato negative (-) 5280 cells, melanoma tdTomato negative (-) 7658 cells). Data analysis was performed using the Scanpy (v1.9) python package89. Cells were filtered if they had mitochondrial read counts above 20% or if they expressed a genes-by-count ratio within the top 2% or bottom 2% of all cells. Data were normalized to 10,000 reads per cell using normalize_total() and the highly variable genes were found with highly_variable_genes(). The effects of total counts per cell and mitochondrial counts were regressed out with regress_out(). Each gene was scaled to unit variance and clipped at a value of 10 with the scale() function. Principle component analysis was performed using the pca() function on the variable genes. The neighborhood graph was computed using neighbors() with the top 20 principal components. Clustering was done with the Leiden algorithm then projected to two dimensions using umap(). Corresponding samples were integrated with the ingest() function. For TD tomato negative samples, cluster marker genes were found with rank_gene_groups() and cell types were annotated using well established markers in combination with markers from PangloaDB90. Differential expression testing between melanoma and control cells were performed with the DiffEXpy python package91 using the test.wald() function on the raw normalized counts. Multiple testing correction was performed using the Benjamini-Hochberg method. Differentially expressed genes and marker genes were tested for enrichment using the goatools92 python package and all musculus coding genes as the background. Statistically significant enrichments were defined by a corrected P value (Benjamini-Hochberg method) less than or equal to 0.05. The MEME Suite Simple Enrichment tool (https://www.biorxiv.org/content/10.1101/2021.08.23.457422v1) was used to search for JASPAR (https://doi.org/10.1093/nar/gkab1113) vertebrate transcription factor binding motif enrichment in the promoter regions 1000 bp upstream and 500 bp downstream of the transcription starting sites.
Analysis of PGC1α binding enrichment. Previously reported ChIP-seq data from mouse adipose cells53 were used to annotate genes within 5000 bp of significant MACS peaks which intersected with study genes (n = 1112). Using a background of 15960 genes present in the study cells, the intersection of differentially downregulated and upregulated genes in EC cluster 2 were tested for overrepresentation using a hypergeometric test and visualized using GSEApy.
Statistics and Reproducibility.Quantitative analysis of images and Western Blotting was performed using ImageJ (NIH, 1.52d, Java 1.8.0_172[64 bit]) software. Quantification is presented as the mean ± SEM from at least 3 independent biological replicate experiments or mice. The Student t-test with unpaired or paired tests were used for two group comparisons to determine statistical significance, with a p value threshold of less than 0.05. Significance levels are indicated in the figures as *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001. The t-test analyses were conducted using Prism 9.3.0, GraphPad Software (La Jolla, CA). We also used one-way ANOVA for statistical analysis using aov() function in R, followed by post-hoc multiple comparison analysis using Tukey test. Adjusted p-values were reported and less than 0.05 is considered as statistically significant. The significance levels of adjusted p values are indicated in the figures as *p < 0.05, **p < 0.01, and ***p < 0.001.