Abstract Objective Cyclin C (CCNC) is the most conserved subunit of the Mediator complex, which is an important transcription cofactor. Recently, we have found that CCNC facilitates brown adipogenesis in vitro by activating C/EBPα-dependent transcription. However, the role of CCNC in brown adipose tissue (BAT) in vivo remains unclear. Methods We generated conditional knock-out mice by crossing Ccnc^flox/flox mice with Myf5^Cre, Ucp1^Cre or Adipoq^Cre transgenic mice to investigate the role of CCNC in BAT development and function. We applied glucose and insulin tolerance test, cold exposure and indirect calorimetry to capture the physiological phenotypes and used immunostaining, immunoblotting, qRT-PCR, RNA-seq and cell culture to elucidate the underlying mechanisms. Results Here, we show that deletion of CCNC in Myf5^+ progenitor cells caused BAT paucity, despite the fact that there was significant neonatal lethality. Mechanistically different from in vitro, CCNC deficiency impaired the proliferation of embryonic brown fat progenitor cells without affecting brown adipogenesis or cell death. Interestingly, CCNC deficiency robustly reduced age-dependent lipid accumulation in differentiated brown adipocytes in all three mouse models. Mechanistically, CCNC in brown adipocytes is required for lipogenic gene expression through the activation of the C/EBPα/GLUT4/ChREBP axis. Consistent with the importance of de novo lipogenesis under carbohydrate-rich diets, high-fat diet (HFD) feeding abolished CCNC deficiency -caused defects of lipid accumulation in BAT. Although insulin sensitivity and response to acute cold exposure were not affected, CCNC deficiency in Ucp1^+ cells enhanced the browning of white adipose tissue (beiging) upon prolonged cold exposure. Conclusions Together, these data indicate an important role of CCNC-Mediator in the regulation of BAT development and lipid accumulation in brown adipocytes. Keywords: CCNC, brown fat, Lipid droplet, Progenitor, Proliferation, Lipogenesis Abbreviations: CCNC, cyclin C; BAT, brown adipose tissue; UCP1, uncoupling protein 1; C/EBP, CCAAT/enhancer-binding protein; ChREBP, carbohydrate response element-binding protein; SREBP, sterol regulatory element-binding protein Highlights * • CCNC plays different roles at different developmental stages in BAT. * • CCNC controls the prenatal BAT development by regulating the proliferation of brown progenitors. * • CCNC critically supports lipid accumulation in adult BAT through the C/EBPα/GLUT4/ChREBP axis. 1. Introduction The current epidemic of obesity and type 2 diabetes has increased the need for novel approaches to reduce adiposity. Obesity is caused by prolonged periods of positive energy balance in which energy intake exceeds energy expenditure. Brown adipose tissue (BAT) is specialized to dissipate energy through uncoupling protein 1 (UCP1) and thus may counteract obesity [[37]1]. Rodents also displays an inducible thermogenic adipose tissue termed beige adipocytes that arises within white adipose tissue (WAT) depots, that can use both UCP1 and possibly other futile cycles for heat production [[38]2,[39]3]. In humans, BAT is abundant at birth but is rapidly replaced by WAT through unknown mechanisms and is relatively scarce in adults as an unidentifiable tissue [[40]4]. However, studies with ^18fluoro-labelled 2-deoxy-glucose positron emission tomography (^18FDG-PET) scanning demonstrated that adult humans could have active thermogenic adipose tissue deposits and the amount of this thermogenic tissue is inversely correlated with adiposity and body mass index (BMI) [[41]5,[42]6]. Whether human thermogenic adipose tissues are more reflective of rodent brown or beige adipocytes has not been fully resolved. In any case, activation of BAT thermogenesis by cold exposure, or by β3-adrenergic receptor agonist, has been linked to increased energy expenditure, reduced adiposity and lower plasma lipids [[43][7], [44][8], [45][9]], indicating that BAT plays an important role in energy homeostasis both in animal models and humans. Therefore, a better understanding of the molecular control of BAT development and function may lead to new therapeutic avenues to combat obesity and metabolic disorders. The classical brown fat is localized in the interscapular region in rodents and arises from a Myf5-positive lineage [[46]10]. PR-domain-containing protein 16 (PRDM16), a BAT-enriched transcription cofactor, acts as a fate switch to control brown adipocyte versus myocyte between days 9–12 of gestation in mice [[47]10,[48]11]. Mechanistically, PRDM16 forms a transcriptional complex with the active form of CCAAT/enhancer-binding protein-beta (C/EBPβ) in the myogenic precursors to activate the brown adipogenic gene program by inducing peroxisome proliferator-activated receptor-gamma (PPARγ) expression [[49]12]. Meanwhile, it has been shown that euchromatic histone-lysine N-methyltransferase 1 (EHMT1) is a BAT-enriched lysine methyltransferase in complex with PRDM16 and regulates brown adipocyte fate by stabilizing PRDM16 proteins [[50]13]. Extracellularly, BMP7 signaling promotes brown adipocyte differentiation and thermogenesis through the induction of PRDM16 and PPARγ coactivator-1a (PGC-1a), which increase the expression of the brown fat marker UCP1 and adipogenic transcription factors PPARγ and C/EBPs as well as mitochondria biogenesis [[51]14]. However, the regulatory mechanisms for BAT development are not completely understood. Unlike white adipocytes, differentiated brown adipocytes are characterized by multilocular lipid droplets, in which triglycerides are increasingly accumulated with age. Although the physiological functions remain less clear, lipolysis and lipogenesis in BAT are dynamic and highly active in response to environmental cues. The regulation of lipolysis in BAT has been well documented [[52]15,[53]16], but the regulation of lipogenesis and triglyceride biosynthesis in BAT is less studied. Recently, one study has shown that AKT2, a cold-induced kinase, promotes de novo lipogenesis (DNL) in BAT through carbohydrate response element-binding protein (ChREBP) but not sterol regulatory element-binding protein-1 (SREBP-1) [[54]17]. Moreover, DNL in BAT may play a role in optimizing fuel storage and thermogenesis in mice, as inhibition of DNL in BAT causes compensatory increase of WAT browning under prolonged cold exposure [[55]17]. Importantly, brown fat DNL in human is positively correlated with Ucp1 expression, and negatively correlated with BMI [[56]17,[57]18]. Although there have been significant insights into the signaling pathways and transcription factors that regulate DNL in BAT, the regulation of lipid metabolism in BAT remains largely unclear. The Mediator complex is a conserved transcription cofactor that primarily regulates activator-dependent transcription [[58][19], [59][20], [60][21], [61][22]]. In mammalian cells, the Mediator complex is comprised of up to 30 subunits [[62][19], [63][20], [64][21], [65][22]]. It has been shown that the Mediator subunit MED1 regulates white adipogenesis in vitro through PPARγ [[66]23,[67]24]. MED14 [[68]25], MED19 [[69]26], MED23 [[70]27,[71]28] and MED31 [[72]29] are also involved in the regulation of white adipogenesis in vitro and in mouse models through PPARγ and/or other aspects of insulin signaling pathway. Moreover, MED1 regulates the brown adipocyte transcriptional program by interacting with PRDM16 [[73]30,[74]31]. More recently, using tissue specific MED1 knockout mouse models, two studies have demonstrated a critical role of MED1 in adipose tissue formation in vivo [[75]32,[76]33]. On the other hand, the Mediator subunits MED1 [[77]33], MED15 [[78]34] and MED17 [[79]35,[80]36] are directly or indirectly involved in the induction of lipogenesis. Previously, we have shown that the Mediator subunit CDK8 together with its activating partner CCNC negatively regulates DNL in hepatocytes and Drosophila fat bodies by promoting nuclear SREBP-1a/c phosphorylation and subsequent degradation [[81]37]. Although initially identified as a potential cell cycle regulator [[82]38,[83]39], CCNC functions to regulate gene transcription in the context of the Mediator complex. As a highly conserved Mediator subunit, CCNC has been shown to play multiple roles in various biological processes including cell proliferation [[84][38], [85][39], [86][40]], cell apoptosis [[87]41,[88]42], and cellular lipid metabolism [[89]37,[90]43]. Recently, we have identified CCNC as an essential regulator for brown adipogenesis in vitro [[91]43]. Mechanistically, CCNC-Mediator physically interacts with C/EBPα and activates the C/EBPα transactivation domain activity to facilitate brown preadipocyte differentiation [[92]43]. Interestingly, the CCNC level is decreased during brown adipogenesis and during aging in mice [[93]43], suggesting that CCNC may play a role in BAT development and function. Human individuals with mutations in the genomic locus of Cdk8, one major effector of CCNC, are prone to develop type 2 diabetes [[94]44]. Moreover, the GWAS Central data indicate that human Ccnc gene is significantly associated with BMI. However, the underlying mechanism remains unknown. Based on these findings, we hypothesize that CCNC plays an important role in BAT development and function in vivo. Here, we show that CCNC deletion in Myf5^+ cells caused a partial neonatal lethality in mice likely due to the developmental defects in rib cartilage. The survived CCNC-deficient mice displayed a paucity in BAT due to the impaired proliferation of brown adipocyte progenitor cells. In contrast to tissue culture, CCNC deficiency did not affect brown adipogenesis in vivo. Interestingly, CCNC-deficient BAT displayed a reduced ability of accumulating lipids over time when mice were fed with a carbohydrate-rich normal chow diet. This phenotype was recapitulated when CCNC was deleted in either Ucp1^+ or Adipoq^+ cells. RNA-seq analyses revealed that CCNC deficiency reduced the expression of some key enzymes for fatty acid and triglyceride biosynthesis that are critically regulated by the C/EBPα/GLUT4/ChREBP pathway. Consistent with the regulation of DNL, CCNC depletion -caused lipid accumulation defects in BAT was abolished when mice were fed with a high fat diet (HFD). Moreover, deletion of CCNC in Ucp1^+ cells also enhanced WAT beiging upon chronic cold exposure. Thus, our results indicate that CCNC plays a pivotal role in BAT prenatal development and postnatal lipid metabolism. 2. Materials and methods 2.1. Animals and diets All mouse experiments conformed to the protocols approved by the Animal Care and Use Committees of the Albert Einstein College of Medicine in accordance with the National Institutes of Health guidelines. Myf5^Cre (Stock #010529), Ucp1^Cre (Stock #024670) and Adipoq^Cre (Stock #028020) transgenic mice were obtained from the Jackson Laboratory. Ccnc-floxed mice was generated and backcrossed for ten generations into the C57BL/6J strain. Mice were housed in a pathogen-free animal facility at 22 °C with a 12 h light/dark cycle (7:00 am - 7:00 pm) with free access to water and the normal chow diet (#5053, LabDiet; 13 kcal% fat and 62 kcal% carbohydrate) or the high-fat diet (no. D12492, Research Diets; 60 kcal% fat and 20 kcal% carbohydrate), which started at the age of six weeks. 2.2. Body composition and indirect calorimetry Fat/lean mass was measured by quantitative nuclear magnetic resonance (NMR) noninvasive imaging (EchoMRI, TX). Metabolic measurement was performed using an Oxymax indirect calorimetry system (Columbus Instruments, Columbus, OH). Mice were individually housed in metabolic chambers at 22 °C with a 12 h light/dark cycle and free access to food and water. After acclimation in metabolic chambers for 48 h, energy expenditure, oxygen consumption (VO[2]), carbon dioxide production (VCO[2]), and spontaneous locomotor activity were measured as previously described [[95]45]. VO[2], VCO[2], and energy expenditure of each mouse were normalized by its lean mass. 2.3. Glucose and insulin tolerance test (GTT and ITT) For GTT, mice were fasted for 14 h and then received an intraperitoneal injection of glucose (1 g/kg body weight). Two weeks later, ITT was performed by an intraperitoneal injection of insulin (1.5 U/kg body weight) after 4 h fasting. Blood was collected at consecutive time points after the injection of glucose or insulin by tail bleeding, and blood glucose levels were measured using the AlphaTRAK 2 Blood Glucose Monitoring System. 2.4. Cold exposure To assess cold tolerance, 8-month-old control and Ccnc^Ucp1(KO) mice were fasted for 4 h in the morning, and then placed at 6 °C individually in pre-chilled cages with free access to pre-chilled water and food. Intrarectal temperatures were monitored and recorded with a TH-8 Thermalert Monitoring Thermometer (Physitemp Instruments, Clifton, NJ) at consecutive time points up to 14 days. 2.5. Measurement of triglycerides Triglycerides in BAT were measured using the Triglyceride Quantification Colorimetric Fluorometric Kit (Biovision, Milpitas, CA) according to the manufacturer's instructions. Briefly, total lipids were extracted from homogenized BAT (∼20 mg each), and triglycerides were converted to free fatty acids and glycerol by the addition of lipases. Glycerol was detected by an enzyme-coupled reaction and using a VersaMax spectrometry microplate reader (Molecular Devices, Sunnyvale, CA). The triglyceride levels were normalized by genomic DNA. 2.6. Alcian Blue-Alizarin Red staining Dead Ccnc^Myf5(KO) newborns and littermate controls (alive) were fixed in 4% paraformaldehyde for Alcian Blue-Alizarin Red staining as previously described with minor modifications [[96]46]. Briefly, skins and tissues were carefully removed, and fat was depleted by washing with 95% ethanol followed by overnight incubation in acetone. The skeletons were immersed in 0.03% alcian blue solution for 24 h to stain cartilage, washed with 70% ethanol, and incubated in 1% potassium hydroxide until clear. Bone was counterstained with 0.03% alizarin red solution until sharply delineated. The stained samples were cleaned in 1% potassium hydroxide/glycerol (1:1) for a week and kept in glycerol at the end. Images were captured by a Nikon Digital Camera and processed using the Photoshop CS6 software. 2.7. TUNEL assay, immunofluorescence, immunohistochemistry and histological analyses An 8-μm thick, 4% neutral buffered paraformaldehyde-fixed cryosections of tissues were embedded in an optimal cutting temperature compound. TUNEL assay was performed using the ApopTag Red in situ Apoptosis Detection Kit (EMD Millipore, Burlington, MA) according to the manufacturer's instructions. Briefly, sections were treated with the Equilibration Buffer and the Working Strength TdT Enzyme sequentially, and counterstained with an anti-digoxigenin conjugate antibody and mounted with the prolong antifade reagent with DAPI (Invitrogen) before imaging. For Ki67 immunofluorescent staining, slides were permeabilized with 0.3% Triton X-100 (Sigma–Aldrich), washed with 1xPBS, and blocked with 5% normal donkey serum (ab166643, Abcam) before adding anti-Ki67 primary antibody (ab16667, Abcam, 1:500) that was diluted in blocking solution. After wash, Alexa Flour 488-labelled secondary antibody (Invitrogen, 1:500) was added. The slides were then washed, counterstained, and mounted with the prolong antifade reagent with DAPI (Invitrogen) before imaging. For UCP1 immunohistochemical staining, 5 μm thick paraffin sections were deparaffinized, rehydrated, and epitopes unmasked by boiling with the Citra buffer (BioGenex, Fremont, CA). Tissues were then blocked with 5% normal goat serum solution before incubation with anti-UCP1 primary antibody (PA1-24894, Pierce, 1:200). Vectashield DAB Peroxidase Substrate Kit was used to detect the primary antibody. Sections were counterstained with hematoxylin, mounted, and imaged. For histological analyses, samples were embedded, cut, and performed Hematoxylin and Eosin (H&E) staining by the Albert Einstein College of Medicine Histology Core facility. 2.8. Western blotting Cultured cells were washed with cold 1xPBS, scraped, and homogenized by pipetting in a lysis buffer containing 50 mM Tris–HCl (pH 8.0), 420 mM NaCl, 0.1 mM EDTA, 0.5% Nonidet P-40, 0.05% SDS, 10% Glycerol, and a Protease and Phosphatase Inhibitor Cocktail (78442, Thermo Fisher Scientific). Tissues were homogenized in the lysis buffer using Ceria Stabilized Zirconium Oxide Beads. The cell or tissue homogenates were centrifuged for 30 min at 21,000×g at 4 °C, and supernatants were collected for protein assays. Twenty μg of total proteins for each sample were loaded and separated by 4–12% SDS-PAGE and transferred to a nitrocellulose membrane using the iBlot Blotting System (Thermo Fisher Scientific). The membrane was blocked with 5% non-fat dry milk in 1x Tris-buffered saline containing Tween-20 (TBST) and incubated with a primary antibody in the blocking buffer. The membranes were washed with 1xTBST and incubated with HRP-conjugated secondary antibody in blocking buffer. The membrane was then washed with 1xTBST, visualized through ECL (Pierce, Thermo Fisher Scientific), and exposure to X-ray films. The following primary antibodies and dilutions were used: Anti-CCNC (558903,1:500) from BD Pharmingen; Anti-Actin (A5060, 1:200) from sigma; Anti-β-tubulin (1235662A, 1:2000) from Invitrogen; Anti-ChREBP (NBP2-44307, 1:500) from NOVUS; Anti-C/EBPa (8178, 1:250), Anti-FAS (3180, 1:1000) and Anti-ACC1 (3676, 1:1000) from Cell Signaling Technology; Anti-GLUT4 (ab654, 1:500) from Abcam; Anti-SREBP1 (sc-13551, 1:200) and Anti-CPT1B (sc-393070, 1:500) from Santa Cruz Biotechnology. 2.9. RNA extraction and real-time RT-PCR Total RNA was prepared from tissue or cell samples using the Trizol Reagent (Invitrogen) according to the manufacturer's instructions. Genomic DNA contamination was removed by RNase-free DNase I (Thermo Scientific, USA). The quantity and quality of total RNA were determined by a spectrophotometer (Nanodrop 2000, Thermo Fisher). Total cDNA was synthesized from 1 μg of total RNA using iScript cDNA Synthesis Kit (BIO-RAD). Real-time PCR was performed using the PowerUp SYBR Green Master Mix (Thermo Fisher, USA). Specific primers for each gene are listed in [97]Table S1. Tbp or Rpl7 was used as the invariant control. 2.10. RNA sequencing (RNA-seq) After RNA extraction from BAT of 6-month-old Ccnc^Ucp1(KO) and control mice (three biological replicates each), RNA-seq libraries were prepared by the Einstein Genomics Core Facility. The library quality was analyzed by a Bioanalyzer (Agilent Technologies). Deep sequencing was performed using a HiSeq 2500 instrument (Illumina). Raw reads for each library were mapped using TopHat version 2.0.8 against the indexed mouse (mm9) genome, and transcripts were assembled using Cufflinks. Genes that were not expressed in all samples (FPKM<1) were filtered out. Differentially expressed genes (DEGs) were identified using a RNA-seq processing tool DESeq2, selected by adjusted_p value < 0.05 and fold change >1.4 or < 0.5, and visualized using R tools ggplot2 and pheatmap, respectively. Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analysis of DEGs was obtained from DAVID database (version 6.8). The heat map for the eight down-regulated genes in the insulin signaling pathway was based on the normalized reads, and they were listed according to their p values. 2.11. Tissue culture Immortalized wildtype and CCNC-knockout brown preadipocytes were generated as shown previously [[98]43]. These cells were maintained in DMEM with 25 mM glucose plus 10% FBS, 1% penicillin-streptomycin (P/S) and 1% L-glutamine. For differentiation, cells were cultured to confuent and then treated with the differentiation medium of DMEM containing dexamethasone (0.5 mM), insulin (20 nM), isobutylmethylxanthine (0.5 mM), indomethacin (0.125 mM), T3 (1 nM) and rosiglitozone (0.5 μM). After two days of culture, cells were maintained in DMEM containing 1 nM T3 and 20 nM insulin. Medium was replaced every other day. Full differentiation was achieved in about seven days. 2.12. Retrovirus transduction Mouse ChREBPβ was amplified by PCR from pCMV-Flag-ChREBPα, which was a gift from Dr. Donald K. Scott of Icahn School of Medicine at Mount Sinai, and subcloned into the EcoR1 site of the retroviral vector pMSCV-PIG using an In-Fusion Cloning kit (Clontech) to generate a ChREBPβ-expressing plasmid, termed pMSCV-PIG-ChREBPβ, which was verified by DNA sequencing. Retroviral packaging plasmids, pUMVC (#8449) and pCMV-VSV-G (#8454), were obtained from Addgene. To generate pseudotyped virus, pUMVC, pCMV-VSV-G and pMSCV-PIG vectors were co-transfected into sub-confluent HEK293T cells using Lipofectamine 3000 Transfection Reagent (Invitrogen) according to the manufacturer's instructions. The virus stocks were collected at 48 h and 72 h after transfection, filtered through a 0.45 μm filter, and frozen at −80 °C. Brown preadipocytes (20–30% confluent) were infected with retrovirus stocks containing 8 μg/ml polybrene for 12 h, washed and cultured in DMEM. 2.13. Luciferase reporter assays The constructs pcDNA3-Flag-C/EBPα (#66978) and 3xPPRE-TK-luc (#1015) were obtained from Addgene. The Slc2a4 promoter was amplified from mouse genomic DNA by PCR [[99]47], and subcloned into 3xPPRE-TK-luc by replacing the 3xPPRE sequence, which is located between Hind III and BamH I sites, using In-Fusion Cloning kit (Clontech). The resulting plasmid, Slc2a4-TK-luc, was verified by DNA sequencing. DNA transfection and luciferase reporter assays were conducted as described previously [[100]43]. Briefly, brown preadipocytes were seeded at a density of 1 × 10^5 per well in 24-well plates. Next day, cells were transfected with a mix of 250 ng of pcDNA3-Flag-C/EBPα (or control), 250 ng of Slc2a4-TK-luc and 50 ng of renilla luciferase reporter using the Lipofectamine 3000 Transfection Reagent. After incubation for two days, cells were lysed and analyzed using the Dual-Luciferase System (Promega) according to the manufacturer's instructions. The firefly luciferase activity was normalized by the corresponding renilla luciferase activity. 2.14. Statistical analyses All numerical results were expressed as Mean ± SD. Statistical difference was determined by unpaired Student's t-test at a significance level of p < 0.05. The statistical analyses and figure preparation were performed using GraphPad Prism 9 (GraphPad software). 3. Results 3.1. CCNC deficiency in the Myf5^+ lineage causes partial neonatal lethality and reduction of brown adipose tissues To understand the role of CCNC in BAT development, we deleted CCNC in Myf5^+ cells by crossing Ccnc^flox/flox mice with Myf5^Cre transgenic mice, as Myf5 is expressed specifically in the progenitor cells that give rise to brown adipocytes and skeletal muscle cells [[101]10]. As shown in [102]Figure 1A, genotyping analyses revealed that approximately 10% of mice were homozygous CCNC knockout (Ccnc^Myf5(KO): Ccnc^flox/flox; Myf5^Cre) at the time of weaning. While the distribution of CCNC heterozygous or wildtype mice was closer to the expected 25% Mendelian ratio, the percentage of Ccnc^Myf5(KO) mice was significantly lower than 25%, suggesting that many Ccnc^Myf5(KO) mice may have died before weaning. Although a previous study has shown that germline CCNC ablation causes embryonic lethality at E10.5 [[103]40], partial lethality of Ccnc^Myf5(KO) pups was not expected. After closely monitoring the breeding pairs, we discovered an unusual number of dead newborns, and genotyping analyses showed that the majority of those dead pups (25 out of 30) were Ccnc^Myf5(KO). However, genotyping analyses of embryos showed normal Mendelian ratios for all genotypes ([104]Figure 1A), suggesting that the death of those Ccnc^Myf5(KO) mice may have occurred shortly after birth. Figure 1. [105]Figure 1 [106]Open in a new tab CCNC deficiency in the Myf5^+ lineage causes partial neonatal lethality and prenatal brown fat paucity. (A) Summary of the genotypes of mice at E18.5 and P1 stages. (B) Representative pictures of Alcian Blue-Alizarin Red Skeletal Staining of the live control and dead Ccnc^Myf5(KO) mice at P1. The abnormal ribs are indicated by red arrow. (C) Macroscopic examination of BAT of the control and Ccnc^Myf5(KO) mice at P1. BAT is highlighted by dashed lines. (D) H&E staining of cervical transverse sections of the control and Ccnc^Myf5(KO) mice at E18.5. BAT is highlighted by dashed lines. Scale bar, 200 μm. (E) Representative images of BAT isolated from the live control and Ccnc^Myf5(KO) mice at P1. Scale bar, 1 mm. (F) Body weight of the control and Ccnc^Myf5(KO) mice at P1. (G) Percentage of BAT weight over body weight of the control and Ccnc^Myf5(KO) mice at P1. (H) qRT-PCR analysis of Ccnc, Cdk8 and Cdk19 expression in BAT of the control and Ccnc^Myf5(KO) mice at E18.5. ∗∗∗ p < 0.001 vs control. (For interpretation of the references to color in this figure legend, the