Abstract Regulatory T cells (T[regs]) in non-lymphoid organs provide critical brakes on inflammation and regulate tissue homeostasis. While so-called “tissue-T[regs]” are phenotypically and functionally diverse, serving to optimize their performance and survival, upregulation of pathways related to circadian rhythms is a feature they share. Yet, the diurnal regulation of T[regs] and its consequences are controversial and poorly understood. Here, we profiled diurnal variations in visceral-adipose tissue (VAT) and splenic T[regs] in the presence and absence of core-clock genes. VAT, but not splenic, T[regs] upregulated their cell-intrinsic circadian program, and exhibited diurnal variations in their activation and metabolic state. BMAL1 deficiency specifically in T[regs] led to constitutive activation and poor oxidative metabolism in VAT, but not splenic, T[regs]. Disruption of core-clock components resulted in loss of fitness: BMAL1-deficient VAT T[regs] were preferentially lost during competitive transfers and in heterozygous BMAL1-deficient females. After 16 weeks of high-fat-diet feeding, VAT inflammation was increased in mice harboring BMAL1-deficient T[regs], and the remaining cells lost the transcriptomic signature of bona fide VAT T[regs]. Unexpectedly, VAT T[regs] suppressed adipocyte lipolysis, and BMAL1 deficiency specifically in T[regs] abrogated the characteristic diurnal variation in adipose-tissue lipolysis, resulting in enhanced suppression of lipolysis throughout the day. These findings argue for the importance of the cell-intrinsic clock program in optimizing VAT T[reg] function and fitness. ONE-SENTENCE SUMMARY: Visceraladipose tissue Tregs turn up cell-intrinsic circadian rhythms to promote cellular fitness and regulate diurnal lipolysis INTRODUCTION Regulatory T cells (T[regs]), particularly those expressing the transcription factor (TF) Foxp3, play an essential role in restraining overexuberant immune responses. Although early studies focused on T[regs] circulating through lymphoid organs, it is by now well established that T[regs] are located in non-lymphoid tissues as well. These so-called “tissue T[regs]” have unique transcriptomes, T cell receptor (TCR) repertoires and growth/survival-factor dependencies that enable them to thrive in particular tissue environments, where they help maintain homeostasis ([36]1). Even though tissue T[regs] exhibit a diversity of phenotypes and functions, they also share transcriptomic modules that set them apart from their lymphoid-organ counterparts ([37]1, [38]2). Intriguingly, pathways related to circadian rhythms are amongst the upregulated pathways shared most prominently, raising the question of whether tissue T[regs] are subject to distinct diurnal regulation. Mammalian circadian rhythms evolved to anticipate and adapt to diurnal variations in the environment. A set of “core-clock” genes encodes the molecular drivers of this process, the expression, activity and degradation of which form an autoregulatory feedback loop lasting approximately 24 hours ([39]3). Heterodimeric complexes of BMAL1:CLOCK drive a core component of circadian transcription, including genes encoding its inhibitors, PER and CRY, and genes specifying its transcriptional regulators, the REV-ERBs and ROR [reviewed in ([40]3)]. These molecular oscillators are stratified into a hierarchy of central and peripheral pacemakers according to whether they are localized in cells of the suprachiasmic nucleus of the brain or in cells of peripheral tissues. Peripheral clocks interpret synchronization signals from the central pacemaker, which is primarily synchronized by light, and from other environmental cues, like feeding and temperature, to generate cell-intrinsic rhythmicity ([41]4–[42]6). Our understanding of circadian regulation of T cells and of the functions of their core-clock genes is just beginning to emerge. Rhythmic T cell migration between the circulation and lymphoid organs is the best-described circadian behavior: in mice, peak numbers of lymphocytes are found in circulation around zeitgeber time (ZT) 5 (ZT0 signalling onset of the light period), and in lymphoid organs around ZT13 ([43]7–[44]10). Additionally, naïve murine T cells isolated during the day are more likely to differentiate into T helper 17 (Th17) cells than those taken at night ([45]11). However, core-clock genes were reported to be dispensable for T cell differentiation and for their responses to bacterial and viral infection ([46]12), and expression of core-clock genes in splenic T[regs] seemed to lack rhythmicity ([47]13). Thus, the contexts wherein T[regs] are subject to circadian regulation, as well as the consequences of such regulation, remain in question. We decided to investigate the importance and function of circadian rhythms in the paradigmatic tissue-T[reg] population found in epididymal visceral-adipose tissue (VAT) of lean mice ([48]14). Thymic T[regs] seed VAT in the first weeks of life, where they proliferate indolently until they dominate the CD4^+ T cell pool around 20–30 weeks of age ([49]15). Their accumulation and survival depend on a unique transcriptome largely driven by the critical transcriptional regulator of adipocyte differentiation, PPARγ, as well as on their TCR and cytokines such as IL-33 ([50]15–[51]18). Importantly, VAT T[regs] regulate local and systemic metabolic tenor by controlling both immunologic and non-immunologic cells ([52]14, [53]16, [54]17). Combining population-level and single-cell transcriptomics, adoptive transfer experiments, genetic ablation, obesity-inducing challenges, lipolysis assays, and cytofluorimetry-based single-cell metabolic assays, we uncovered a role for the cell-intrinsic clock in promoting the fitness of VAT, but not splenic, T[regs], regulating, for example, adipose-tissue lipolysis. The heightened circadian regulation in T[regs]operating in non-lymphoid-tissue environments re-emphasizes their adaptability and striking divergence from their lymphoid-organ counterparts. RESULTS Elevated expression of core-clock genes in VAT T[regs] Since pathways related to circadian rhythms appeared to be upregulated in tissue T[regs] ([55]1, [56]2), we employed published microarray data to compare expression of the core-clock genes in VAT ([57]16), muscle ([58]19) and colonic lamina propria ([59]20) T[regs] with that of their lymphoid-organ counterparts. Core-clock genes, across all major arms of the cell-intrinsic clock machinery ([60]Fig. 1A), were expressed at higher levels in T[regs] from non-lymphoid than lymphoid tissues ([61]Fig. 1B and [62]fig. S1A). Focusing on VAT, we found much less evident up-regulation of the core-clock genes in conventional CD4^+ T cells (T[convs]) than in T[regs] ([63]Fig. 1C and [64]fig. S1B). Data from a T[reg] adoptive-transfer system confirmed that the VAT microenvironment up-regulated core-clock gene expression: transfer of T[regs] from a VAT-T[reg] TCR-transgenic (tg) mouse line ([65]18) into non-tg recipients resulted in accumulation of donor T[regs] with elevated expression of core-clock genes in VAT in comparison with spleen ([66]Fig. 1D and [67]fig. S1C). Thus, expression of core-clock genes was elevated in tissue Tregs, VAT T[regs] in particular, in comparison with their splenic counterparts, an increase induced by the tissular microenvironment. Fig. 1: Upregulation of core-clock gene expression in VAT T[regs]. Fig. 1: [68]Open in a new tab (A) Schematic of the cell-intrinsic circadian rhythms executed by core-clock gene products. (B) Core-clock gene expression by tissue T[regs] from published microarray data: VAT ([69]16), skeletal muscle 4 days after cardiotoxin injury ([70]19), and colonic lamina propria ([71]20) T[regs] in comparison with lymphoid-organ T[regs] from the same mice. (C) Core-clock gene expressions in VAT-T[reg] or -T[conv] cells, from published microarray data on retired male breeders ([72]28) in comparison with lymphoid-organ T[regs] or T[convs] from the same mice. (B, C) p: * ≤ 0.05, ** ≤ 0.01, *** ≤ 0.001 according to Student’s t-test comparing expression in tissue versus paired lymphoid-organ T[regs]. (D) Core-clock gene expression at zeitgeber 0 (onset of light phase) in donor T[regs] 9 weeks after transfer in recipients’ VAT and spleen. T[regs] from Foxp3-Cre.YFP^+CD45.1^+TCR-tg^+ donors were transferred into Foxp3-Cre.YFP^+CD45.1/2^+ recipients. FDR: * ≤ 0.05, ** ≤ 0.01, *** ≤ 0.001 according to the quasi-likelihood F-test by EdgeR. n ≥ 3 per condition. VAT, visceral adipose tissue; Spl, spleen; AU, arbitrary units; TCR, T cell receptor; tg, transgenic. Rhythmic expression of VAT-T[reg] core-clock genes We next assessed diurnal expression profiles of the core-clock genes by population-level RNA sequencing of cytofluorimetrically sorted VAT T[regs]. Plotting gene expression versus time, where zeitgeber (ZT) 0 denotes onset of the light phase and ZT12 the dark phase, revealed that expression of most of the core-clock genes (such as Bmal1, Per1-3, and Nr1d1,2) exhibited significant rhythmicity in VAT T[regs] ([73]Fig. 2A). The times of peak core-clock gene expression in VAT T[regs] were similar to those previously reported for liver cells ([74]21, [75]22), reinforcing the notion that peripheral tissues are usually synchronized. Expression of a few core-clock genes, specifically Cry1-2 and Rorα, lacked rhythmicity in VAT T[regs], which is not surprising given the report that not all core-clock genes are rhythmically expressed in peripheral tissues ([76]23). In stark contrast, rhythmicity of core-clock genes in splenic T[regs] appeared muted and was not statistically significant, with the exception of Nr1d2 ([77]Fig. 2A). Fig. 2: Rhythmic expression of core-clock genes and other loci in VAT T[regs]. Fig. 2: [78]Open in a new tab Retired B6 male breeders (>35 weeks old) were sacrificed every 4 hours during a 12:12 light:dark cycle. ZT0 signals onset of the light period (see bars below relevant panels). T[regs] were sorted as CD4^+CD25^+. (A) Expression of core-clock genes over time. (B) Heatmap of rhythmically expressed genes in VAT T[regs]. (C) Distribution of the time of peak expression for rhythmically expressed genes in VAT T[regs]. (D) Fold-change of maximum/minimum gene expression over time in rhythmically expressed genes. (E) Ingenuity Pathway Analysis of rhythmic genes. (F, G) Expression profile of selected rhythmicaly expressed genes in the TCR signaling pathway (F) and cholesterol biosynthesis pathways (G) from panel E. Circadian rhythmicity determined by JTK_Cycle. p: * ≤ 0.05, ** ≤ 0.01, *** ≤ 0.001. n = 3 for each time point from ZT0 to ZT16, n = 2 for ZT20. ZT, Zeitgeiber time. TCA, tricarboxylic acid; VAT, visceral adipose tissue; Spl, spleen; AU, arbitrary units; TCR, T cell receptor. Flow-cytometric gating strategies for all quantified or isolated cell-types can be found in [79]fig. S7. Looking beyond the core-clock genes to loci they might regulate, we identified over 600 genes with rhythmic expression in VAT T[regs] after filtering out genes with low expression or a period of rhythmicity less than 16 hours ([80]Fig. 2B, [81]Table S1). Peak expression of most of these genes was near either ZT0 or ZT12 ([82]Fig. 2C), with about a 1.5-to 2-fold change in expression between their highest and lowest expression ([83]Fig. 2D). Pathway analysis on the rhythmically expressed genes in VAT T[regs] revealed an enrichment in pathways related to protein ubiquitination, sirtuin signalling, cholesterol biosynthesis, T cell activation and mitochondrial metabolism ([84]Fig. 2E). Transcripts encoding molecules involved in TCR signaling and cholesterol biosynthesis peaked mostly around ZT0/24 ([85]Fig. 2F, [86]G). Thus, clock genes were rhythmically expressed in VAT Tregs but generally not in splenic Tregs. Core-clock-gene products enhance the fitnessof VAT T[regs] To assess the functional importance of the T[reg]-intrinsic clock, we abrogated expression of BMAL1 (which drives the forward loop of the clock and thereby promotes rhythmicity) specifically in T[regs] by crossing Foxp3-Cre.YFP mice with a Bmal1^flox/flox strain ([87]24). The mutant offspring will hereafter be referred to as T[reg]^Bmal1Δ mice (Foxp3-Cre.YFP^+Bmal1^flox/flox) as opposed to the T[reg]^Bmal1WT controls (Foxp3-Cre.YFP^+Bmal1^wt/wt). We confirmed that the mutant T[regs] lacked the floxed exon 8 ([88]fig. S2A) by inspecting pile-ups generated from population-level RNA sequencing (RNA-seq). Furthermore, we verified the specificity of deletion under the Foxp3-Cre driver by quantitative RT-qPCR, and observed that Bmal1 expression was substantially decreased in T[regs] but remained unaffected in CD4^+ T[convs] or CD8^+ T cells from the spleen ([89]fig. S2B). As expected, loss of BMAL1 led to significant alterations in the expression of other core-clock genes at ZT0 in sorted VAT T[regs]. Specifically, there was elevated expression of some genes, such as Cry1 and Npas2, and reduced expression of others, such as Nr1d1 and Nr1d2 ([90]Fig. 3A). Fig. 3: Loss of competitive fitness in VAT T[regs] in the absence of BMAL1 or REV-ERBα. Fig. 3: [91]Open in a new tab (A, B): VAT T[regs] were isolated at ZT0 from 18-to-30-week-old male T[reg]^Bmal1WT (WT) or Treg^Bmal1Δ (Δ) mice. (A) Expression of core-clock genes at 18 weeks of age determined by RNA-seq analysis. (B) Fraction of VAT T[regs] across age groups at ZT0 (left) and at ZT0 vs ZT12 (right). Left: pooled data from 4 independent experiments; right: representative data of 3 independent experiments. n ≥ 2 per group per experiment. (C-G) Competitive transfer of a 1:1 mix of T[regs] from 6-to-8-week-old male mice of the indicated genotype into 8-to-10-week-old recipients, as schematized in (C). (D, E) Fractions of Bmal1 WT and Bmal1 Δ donor cells expressed as a percentage of total T[regs] (D) or of total donor T[regs] (E) in the recipients’ VAT. (F) similar to panel E but from the recipients’ spleen 12 weeks after transfer. (G) Fraction of donor cells in cycle (Ki67^+). For 9 and 12 weeks after transfer, representative data of at least 3 experiments, n ≥ 3 per experiment. (H) T[regs] in gonadal fat of Foxp3-Cre^+/−.YFP^+/−Bmal1^flox/flox female heterozygotes. Fraction of Cre^− versus Cre^+ T[regs]; left: representative flow plot; middle: frequency; right: cell number. (I-K) Competitive transfer of a 1:1 mix of T[regs] from the indicated genotypes into Foxp3-Cre.YFP^+CD45.1/2^+ recipients and assessed 12 weeks after transfer. Data pooled from 2 independent experiments. (I,J) Fractions of Nr1d1 WT and Nr1d1 Δ donors expressed as a percentage of total T[regs] (I) or of total donor T[regs](J) in the recipient’s VAT. (K) Same as panel I, but from the recipients’ spleen. (p: * ≤0.05, ** ≤0.01, *** ≤ 0.001 according to Student’s t-test for the transfer experiments; quasi-likelihood F-test by EdgeR for RNA-seq data. wks, weeks; VAT, visceral adipose tissue; Spl, spleen; AU, arbitrary units; TCR, T cell receptor; tg, transgenic. Flow-cytometric gating strategies for all quantified or isolated cell-types can be found in [92]fig. S7. Since the VAT-T[reg] compartment in adult mice receives little contribution from circulating T[regs] ([93]15), we expected diurnal variation in the circulation of lymphoid-organ T cells ([94]8) to have little impact on the VAT-T[reg] pool size. Accordingly, the number and fraction of VAT T[regs] remained similar at ZT0 and ZT12, and between T[reg]^Bmal1WT and T[reg]^Bmal1Δ mice at ZT0 ([95]Fig. 3B and [96]fig. S2C). The frequency of T[regs] in the spleen also lacked diurnal variation and appeared normal in the absence of BMAL1 ([97]fig. S2D). We suspected that the T[reg]-intrinsic clock might promote fitness without impacting accumulation at homeostasis. Thus, we assessed the fitness of BMAL1-deficient T[regs] in a competitive adoptive-transfer system. When polyclonal lymphoid-organ T[regs] are transferred under this protocol, they do not accrue in VAT; however, Tregs from the VAT-T[reg] TCR-tg mouse line mentioned above do accumulate after transfer, reflecting efficient recognition of their TCR’s ligand ([98]18). We generated Foxp3-Cre.YFP^+.CD45.1^+.TCR-tg^+Bmal1 WT and Foxp3-Cre.YFP^+.CD45.2^+.TCR-tg^+Bmal1 Δ donor strains. Bmal1 WT and Bmal1 Δ T[regs] were transferred at a 1:1 ratio into Foxp3-Cre.YFP^+.CD45.1/2^+ recipients, and donor-T[reg] accumulation was quantified in the VAT and spleen at various timepoints thereafter ([99]Fig. 3C). Donor T[regs] were first detected in VAT 6 weeks after transfer, when the Bmal1 WT and Bmal1 Δ cells were present at an equal ratio. By 7.5 to 9 weeks, the Bmal1 Δ population had undergone a preferential expansion, representing over 70% of the total donor-T[reg] pool. However, this transient dominance was followed by a crash of the Bmal1 Δ T[reg] population, such that mutant cells represented less than 10% of donor T[regs] 12 weeks after transfer ([100]Fig. 3D–[101]E). Similar dynamics were observed for donor-T[reg] numbers ([102]fig. S2E). In contrast, Bmal1 WT and Bmal1 Δ T[regs] persisted equally well in the spleen at 12 weeks after transfer ([103]Fig. 3F), suggesting a tissue-specific role for the T[reg]-intrinsic clock in promoting fitness. The reduced number of Bmal1 Δ cells was not due to a lack of proliferation as the fractions of Ki67^+ donor cells were similar in the presence and absence of Bmal1 ([104]Fig. 3G); nor did it appear to reflect migration to other tissues ([105]fig. S2F). We also verified that the reduction of Bmal1 Δ T[regs] was not due to the CD45 allele they expressed, as competitive transfer of CD45.1^+ and CD45.2^+ WT cells led to similar persistence in the recipients’ VAT ([106]fig. S2G). Therefore, loss of Bmal1 Δ donor cells most likely issued from a difference in survival. Finally, loss of fitness was not restricted to adoptive transfer settings nor to TCR-tg T[regs]. Because the Foxp3 gene maps to the X chromosome, random X-inactivation in female mice randomly silences one of the Foxp3 alleles in each cell. Thus, in Foxp3-Cre.YFP^+/−Bmal1^flox/flox heterozygotes, half of the T[regs] express BMAL1 (Cre^-) while half do not (Cre^+). The number and fraction of Cre^+ T[regs] in the gonadal fat of female heterozygotes were substantially lower than those of co-resident Cre^− T[regs] ([107]Fig. 3H). Only 20% of total T[regs] were Cre^+ and their numbers were significantly less than Cre^− Tregs ([108]Fig. 3H). As expected, control Foxp3-Cre.YFP^+/− females without Bmal1 deletion did not show these differences: Cre^+ cells represented 40% of total T[regs] in the gonadal fat, and the numbers of Cre^+ and Cre^− T[regs] were similar ([109]fig. S2H, [110]I). To rule out transcriptional effects of Bmal1 unrelated to its role in circadian rhythms, we performed analogous experiments on another core-clock gene, Nr1d1, encoding REV-ERBα. Like the T[reg]^Bmal1Δ strain, Foxp3-Cre.YFP^+.Nr1d1^flox/flox (hereafter referred to as T[reg]^Nr1d1Δ) mice showed normal accumulation of VAT T[regs] ([111]fig. S2J). However, donor Nr1d1 Δ T[regs] did not persist like Nr1d1 WT T[regs] did in the recipients’ VAT 12 weeks after transfer ([112]Fig. 3I, [113]J), whereas competitively transferred T[regs] of the two genotypes were maintained equally well in the spleen ([114]Fig. 3K). Since Nr1d1 Δ T[regs] showed no reduction in Bmal1 expression at ZT12 ([115]fig. S2K), consistent with the role of REV-ERBα in repression of Bmal1 expression, the loss of competitive fitness of both Bmal1 Δ and Nr1d1 Δ donor cells indicated that circadian rhythms, rather than expression of Bmal1 per se, promoted VAT-T[reg] fitness. Diurnal variation of the activation state of ST2^+ T[reg] subtypes As a preliminary step in understanding the mechanisms underlying diurnal control of VAT-T[reg] fitness, we performed single-cell (sc)RNA-seq on VAT T[regs] at ZT0 and ZT12 from T[reg]^Bmal1WT and T[reg]^Bmal1Δ animals. To permit simultaneous isolation of cell populations at ZT0 and ZT12, we housed half of the experimental animals in a room with the opposite light cycle for at least 14 days ([116]25), and verified that the animals therein indeed entrained to the new light regimen ([117]fig. S3A). Cells from the four conditions were separately hashtagged and encapsulated together. After demultiplexing, we obtained robust scRNA-seq data for over 6,500 VAT CD4^+ T cells, with an average of 1,300 genes detected per cell. Overlay of Foxp3 expression on a Uniform Manifold Approximation and Projection (UMAP) of the pooled datasets allowed us to distill the T[reg] compartment ([118]fig. S3B), consisting of over 2,000 cells. We then reclustered the T[regs], revealing five distinct subtypes ([119]Fig. 4A). The p1 ST2^+, p2 ST2^+, Tbet^+, IL18R^+ and resting clusters were distinguished by divergent and diagnostic gene expression profiles ([120]Fig. 4B). For instance, the resting cluster expressed elevated levels of Sell and Ccr7; the Tbet^+ cluster higher levels of Cxcr3 and Tbx21; and the IL18R^+ cluster elevated levels of Il18r, Cxcr3 and S1pr1. Two clusters expressed Il1rl1, encoding ST2: p1 with higher levels of Areg, Nfkbia and Junb transcripts; p2 with elevated Ly6c expression ([121]Fig. 4B). A differential density map revealed enrichment of the p1 over the p2 ST2^+ subtype in ZT0 VAT T[regs], as well an increased representation of the Il18r^+ subtype ([122]Fig. 4C). Fig. 4: Diurnal variation of activation state within the ST2^+ T[reg] population. Fig. 4: [123]Open in a new tab (A-D, G-J) scRNA-seq analysis of 21-to-23-week-old Foxp3-Cre.YFP^+ (WT) and Foxp3-Cre.YFP^+Bmal1^flox/flox (Δ) VAT T[regs] at ZT0 and ZT12. (A) Heterogeneity of VAT T[regs] visualized by UMAP. Left: a composite of all four conditions; right: disentangled plots for each condition. (B) Heatmap of top 30 differentially expressed genes of the clusters distinguished in panel A. (C) Cell-density differential between the UMAPs of ZT0 and ZT12 Bmal1 WT VAT T[regs]; frequencies of p1 and p2 subtypes of ST2^+ cells are indicated. (D) Differential RNA expression of cell-surface markers for flow cytometric distinction of p1 ST2^+ and p2 ST2^+ T[regs]. (E) Fraction of CD69^hi VAT T[regs] at ZT0 or ZT12 in B6 mice. Left: flow cytometric dot plot; right: summary data. (F) Fraction of CD69^hi splenic T[regs] at ZT0 and ZT12. (E,F) Pooled data from two independent experiments. (G) KEGG pathway analysis by EnrichR of differentially expressed genes in cells of the p1 and p2 ST2^+ clusters. (H-J) Examples of differentially expressed genes encoding activation (H), effector (I), and survival (J) molecules. ** p ≤ 0.01 by Student’s t-test. n ≥ 3 per group per experiment. UMAP, Uniform Manifold Approximation and Projection; VAT, visceral adipose tissue; Spl, spleen. Flow-cytometric gating strategies for all quantified or isolated cell-types can be found in [124]fig. S7. To validate these findings at the protein level, we searched for differentially expressed genes whose products could be detected by flow cytometry. Antibodies recognizing CD62L, ST2, IL18R, CD39, CD69 and CD25 distinguished the five T[reg] clusters defined in [125]Fig. 4A ([126]fig. S3C). For the p1 and p2 ST2^+ clusters, levels of Cd69, Cd44 and Il2ra transcripts were all higher in the p1 cluster, whereas Ly6c transcript levels were augmented in the p2 cluster ([127]Fig. 4D). However, by flow cytometry, cell-surface expression of only CD69 clearly distinguished the two subtypes of ST2^+ cells. CD69^high ST2^+ T[regs] showed a higher mean fluorescence intensity (MFI) of CD44 and CD25 staining, and a lower MFI of Ly6c staining compared with those of CD69^low ST2^+ T[regs], mirroring the scRNA-seq data ([128]fig. S3D). Consistent with the enrichment of p1 ST2^+ T[regs] at ZT0 ([129]Fig. 4C), the fraction of CD69^high cells was significantly higher within the ST2^+ T[reg] population at ZT0 compared with ZT12 ([130]Fig. 4E). In contrast, splenic T[regs] showed a constant component of CD69^high cells ([131]Fig. 4F). We then used the scRNA-seq data to explore differences between the p1 and p2 ST2^+ subtypes as well as to examine their interrelationships. Consistent with the higher CD44, CD25 and CD69 expressions, which are canonical T cell activation markers, T[regs] of the p1 ST2^+ subtype appeared to be more activated. Pathway analysis identified heightened NF-κB and TCR signaling pathways ([132]Fig. 4G), and transcripts encoding activation markers such as IκBα, NR4A1, EGR1 and JUNB were expressed at an elevated level ([133]Fig. 4H). Furthermore, p1 ST2^+ cells expressed higher levels of transcripts encoding T[reg] effector molecules such as Areg, Ctla4 and Il10 ([134]Fig. 4I), as well as transcripts encoding pro-survival molecules such as Tnfaip3, Bcl2a1b and Il10ra ([135]Fig. 4J). On the other hand, p2 ST2^+ cells expressed more Ki67 transcripts, indicative of more cells in cycle ([136]fig. S3E); thus the two subtypes might reflect a balance of activation versus proliferation. RNA-velocity analysis, which predicts the likely future state of a cell based on the ratio of spliced to unspliced transcripts ([137]26, [138]27), revealed distinct trajectories at the two time-points. At ZT12, a trajectory from the p2 toward the p1 ST2^+ subtype was predicted. That trajectory was absent at ZT0; instead, a subset of p1 ST2^+ cells exhibited a trajectory toward the p2 subtype ([139]fig. S3F). In brief, then, the VAT-Treg compartment includes multiple Treg subtypes. Clear diurnal variation was seen in the relative contributions of the p1 and p2 subtypes of ST2^+ cells. Heightened activation state of Bmal1 Δ VAT T[regs] Diurnal variations within the ST2^+ T[reg] population raised the question of whether they depended on the T[reg-]intrinsic circadian clock. Analysis of cell density differences on the UMAP reproduced from [140]Fig. 4A ([141]Fig. 5A, left) revealed an enrichment in ST2^+ T[regs], particularly the p1 ST2^+ subtype, at both ZT0 ([142]Fig. 5A, center) and ZT12 ([143]Fig. 5A, right) in Bmal1 Δ compared with Bmal1 WT T[regs]. Furthermore, Bmal1 Δ T[regs] showed an enrichment of the p1 ST2^+ subtype at both ZT0 and ZT12 ([144]Fig. 5A), in contrast to WT cells, for which we observed a diurnal switch in the dominant ST2^+ subtype ([145]Fig. 4C), suggesting that Bmal1 Δ VAT T[regs] might be constitutively activated. Indeed, population-level RNA-seq revealed heightened activation terms such as “Th1 and Th2 activation”, “TNFR2 signaling” and “IL-2 signaling”, as well as elevated expression of genes encoding cytokine common-γ-chain receptors (Il2ra, Il7r) and members of the NF-κB signaling pathway (Nfkbia, Nfkb1, Traf1) in Bmal1 Δ cells ([146]Fig. 5B and [147]fig. S4A). Consistent with the scRNA-seq data, T[reg]^Bmal1Δ mice had an elevated fractions of ST2^+ T[regs] ([148]Fig. 5C), which translated into an enrichment of the canonical VAT-T[reg] signature ([149]28) ([150]Fig. 5D). Transcripts upregulated in the p1 vs p2 ST2^+ clusters were also enriched in the Bmal1 Δ T[reg] transcriptome ([151]Fig. 5E). According to flow cytometric analysis, the fraction of CD69^high cells within the ST2^+ VAT-T[reg] population was higher in T[reg]^Bmal1Δ than T[reg]^Bmal1WT mice ([152]Fig. 5F), consistent with an enrichment of the p1 ST2^+ subtype. This enrichment was accompanied by a non-significant relative decrease in the Tbet^+ (Il18r^−) subtype in Bmal1 Δ VAT T[regs] ([153]fig. S4B). In contrast, the fraction of CD69^high T[regs] was the same in T[reg]^Bmal1WT and T[reg]^Bmal1Δ spleens. ([154]Fig. 5F). Consistent with the transcriptomic data, the MFI of CD44 staining was significantly higher and the MFI of CD25 staining trended higher in Bmal1 Δ than in Bmal1 WT VAT T[regs] ([155]Fig. 5G). These variations were not present in splenic T[regs] ([156]fig. S4C). Fig. 5: Elevated activation state of Bmal1 Δ T[regs]. Fig. 5: [157]Open in a new tab (A) Cell-density differential comparing Bmal1 WT and Bmal1 Δ VAT T[regs]at ZT0 (middle) and ZT12 (right). The left panel re-presents [158]figure 4A for convenient and accurate comparison. (B-D) Population-level RNA-seq of Bmal1 WT and Bmal1 Δ VAT T[regs] from 18-week-old male mice at ZT0. (B) Ingenuity Pathway Analysis. (C) Fraction of ST2^+ VAT T[regs] from 18–30-week-old male mice at ZT0. (D) Volcano plot overlaid with up- (red) and down- (blue) regulated VAT-T[reg] signature genes ([159]28). (E) Volcano plot overlaid with genes upregulated in the p1 ST2^+ cluster (orange) or p2 ST2^+ cluster (pink). (F) Left: flow plot. Right: summary data showing fraction of CD69^high T[regs] in VAT and spleen at ZT0 or ZT12. (G) Geometric MFI (gMFI) of CD44 (left, middle) and CD25 (right) expression at ZT0. For flow cytometric data, p: * ≤0.05, ** ≤0.01, *** ≤ 0.001 according to Student’s t-test. Th, T helper cell; TNFR2, tumor necrosis factor recptor 2; ZT, Zeitgeiber time; VAT, visceral adipose tissue; Spl, spleen. Flow-cytometric gating strategies for all quantified or isolated cell-types can be found in [160]fig. S7. Male and female mice have distinct gonadal VAT T[reg] compartments ([161]18, [162]29, [163]30). We identified five T[reg] subtypes in T[regs] isolated from the gonadal fat (gVAT) of 8-to-12-week-old female mice using the gating strategy adopted from our single-cell analysis ([164]fig. S4D). The fraction of ST2^+ cells was lower in female than male mice, consistent with prior reports ([165]29, [166]30). Similar to males, female Bmal1 Δ mice exhibited an increased fraction of gVAT ST2^+ T[regs] and a decrease in the Tbet^+ (IL18r^−) T[reg] subtype ([167]fig. S4E). However, representation of the CD69^hi p1 subtype within the ST2^+ population was unchanged in mutant T[regs] ([168]fig. S4E). A heightened activation state in the absence of Bmal1 likely also explained the preferential expansion of the Bmal1 Δ donor-derived Treg population at 9 weeks after competitive transfer. Indeed, population-level RNA-seq on sorted Bmal1 WT and mutant donor-derived cells at this time-point revealed an enrichment in activation pathways such as “TNF-alpha signaling via NF-kb” and “IL-2/Stat5 Signaling” ([169]fig. S4F). Expression of Egr1, Nfkb1, Cd44, Il2ra and Il7r were increased in Bmal1 Δ donor-derived cells ([170]fig. S4G), as they were in the non-competitive setting ([171]fig. S4A). Flow cytometric analysis confirmed the expected increase in CD44 MFI in Bmal1 Δ donor-derived cells 12 weeks after transfer ([172]fig. S4H). Thus, Bmal1 Δ T[regs] showed an enrichment of the ST2^+ subtype in both male and female mice at steady-state, and were more activated than their WT counterparts in male mice and after adoptive transfer. VAT T[regs] enforce diurnal lipolysis We next sought to identify the physiological function(s) of the cell-intrinsic clock in VAT T[regs]. When fed on lean chow, T[reg]^Bmal1Δ mice exhibited a small increase in adiposity compared with their WT littermates ([173]Fig. 6A), without significant changes in their glucose tolerance or insulin sensitivity ([174]fig. S5A). Because increased adiposity can result from reduced lipolysis, and given that the extent of adipose-tissue lipolysis is known to oscillate diurnally ([175]31), we interrogated the role of T[regs] in this process. Systemic depletion of T[regs], achieved through diphteria toxin (DT) administration to >16-week-old Foxp3-DTR^+ mice ([176]Fig. 6B), provoked a rapid decrease in epididymal VAT (eVAT) weight 3 days after DT injection ([177]Fig. 6C), and the epididymal fat pads were visibly smaller ([178]Fig. 6D), suggestive of increased lipolysis. To assess the lipolytic rate of adipose tissue, we explanted eVAT and stimulated it with norepinephrine (NE) in vitro, as described previously ([179]32, [180]33). This approach circumvents the challenge of interpreting serum glycerol levels, which depend on both lipolytic output and peripheral-organ uptake ([181]34). Explants from T[reg]-depleted animals were less sensitive to NE stimulation and resisted further lipolysis, expressed as micromolar release of glycerol per milligram per hour, while basal glycerol release were similar ([182]Fig. 6E). This diminished lipolysis in vitro likely resulted from an elevated lipolysis in vivo. For instance, injection of the β3-adrenergic agonist CL316,243 (CL) to B6 mice, which is well known to induce extensive lipolysis in vivo, also resulted in diminished lipolysis in vitro ([183]fig. S5B). Fig. 6. VAT T[regs] enforce diurnal lipolysis. Fig. 6 [184]Open in a new tab (A) Body composition of 18-to-24-week-old male mice. (B-E, G-H) lipolysis experiments in 22-to-32-week-old male Foxp3-DTR^+ mice. (B) schematic of DT injection and subsequent analyses. (C-E) Body and epididymal VAT weight (C); photo of epididymal fat pad (D); in vitro glycerol release rate of VAT explants (E) 3 days after DT injection at ZT12 (F) in vitro glycerol release rate of VAT explants isolated from T[reg]^Bmal1WT male mice stimulated with 0.1μM norepinephrine. (G-H) Fraction of p1 ST2^+ (CD69^hi) T[regs] (G); in vitro glycerol release rate of VAT explants 4 weeks after DT injection stimulated with 0.1μM norepinephrine (H). (I) in vitro glycerol release rate of VAT explants of 12-to-23-week-old T[reg]^Bmal1WT and T[reg]^Bmal1Δ male mice stimulated with 0.1μM norepinephrine pooled from two independent experiments. For in vitro lipolysis assays, each dot is one explant, two explants were taken per mouse per condition. Paired Student’s t-test between littermate pairs was performed for panel A. For other panels, p: * ≤0.05, ** ≤0.01, *** ≤ 0.001 according to Student’s t-test. DTR, diphteria toxin receptor; μM, micro-molar; mg, milligram; h, hour; NE, norepinephrine; ZT, zeitgeiber time. Flow-cytometric gating strategies for all quantified or isolated cell-types can be found in [185]fig. S7. eVAT explants taken at ZT12 underwent significantly more in vitro lipolysis in response to NE stimulation than did those taken at ZT0 ([186]Fig. 6F), indicative of a diurnal rhythm in lipolysis. However, explants from mice deficient in the ST2^+ T[reg] population, in particular the p1 subtype, did not exhibit this rhythmicity. Four weeks after DT injection, eVAT weight and VAT T[reg] numbers had normalized in DT-injected mice ([187]fig. S5C), but the fraction of p1 ST2^+ T[regs] remained low ([188]Fig. 6G). Explants from these animals exhibited constitutively low lipolysis in vitro ([189]Fig. 6H), suggesting that p1 ST2^+ T[regs] de-sensitized adipocytes to NE-induced lipolysis in vivo and was required for diurnal rhythmicity. Finally, we assessed whether adipose-tissue lipolysis was controlled by BMAL1 in T[regs]. Diurnal rhythms of lipolysis in eVAT from T[reg]^Bmal1Δ mice appeared blunted: in vitro lipolysis was elevated at both ZT0 and ZT12 after NE stimulation ([190]Fig. 6I), indicative of reduced lipolysis in vivo and consistent with the increased adiposity of T[reg]^Bmal1Δ mice ([191]Fig. 6A). Thus, BMAL1 expression enables VAT T[regs] to help enforce a diurnal rhythm in adipocyte lipolysis. Accelerated activation but loss of fitness during high-fat-diet challenge The heightened activation state of Bmal1 Δ T[regs] in lean mice prompted us to characterize these mutant cells in the context of chronic activation such as high-fat-diet (HFD)-induced obesity. Upon HFD feeding, the VAT-T[reg] compartment responds in two distinct stages ([192]35). VAT T[regs] initially undergo a period of proliferation to counter mounting levels of inflammation ([193]35) and their transcriptome shows an enrichment of the VAT-T[reg] signature ([194]28). With chronic HFD feeding, bona fide (ST2^+, KLRG1^+, GATA3^+) VAT T[regs] are greatly reduced and the remnant cells lack the canonical VAT-T[reg] signature ([195]14, [196]28, [197]35). We fed T[reg]^Bmal1WT and T[reg]^Bmal1Δ mice on a lean diet for 14 weeks to establish a normal pool of VAT T[regs], followed by HFD feeding for 4, 8 or 16 weeks ([198]Fig. 7A). Weight gain was similar between the two genotypes ([199]fig. S6A), as was the fraction and number of VAT T[regs]([200]Fig. 7B and [201]fig. S6B). But after 4 weeks of HFD, transcriptional profiling of VAT T[regs] revealed an enrichment of the VAT-T[reg] signature in the mutants’ T[reg] transcriptome ([202]Fig. 7C), associated with an elevated fraction of ST2^+ T[regs] ([203]fig. S6C), and higher cell-surface expression of CD44 and CD25 ([204]Fig. 7D). In contrast, after 16 weeks of HFD, the VAT-T[reg] signature was decreased in Bmal1 Δ compared with Bmal1 WT T[regs] ([205]Fig. 7E), suggestive of poor adaptation of Bmal1 Δ T[regs]. As a result, T[reg]^Bmal1Δ mice had a higher fraction of CD11c^+ inflammatory macrophages ([206]Fig. 7F), and a trend toward a greater accumulation of CD8^+ T cells at this time-point ([207]fig. S6D), idicative of increased VAT inflammation. In line with this notion, inflammatory pathways distinguished the eVAT depots from T[reg]^Bmal1WT and T[reg]^Bmal1Δ mice after 16 weeks of HFD, as assessed by whole-tissue RNA-seq. Pathways related to interferon, IL6 and TNFα signaling were upregulated in the VAT of T[reg]^Bmal1Δ mice, whereas metabolic pathways such as fatty acid metabolism and oxidative phosphorylation were downregulated ([208]Fig. 7G, [209]H). Glucose and insulin tolerance, assessed at 7 and 9 weeks after HFD feeding, were similar between the two genotypes ([210]fig. S6E). Thus, disruption of the cell-intrinsic clock impaired VAT T[regs]’ ability to adapt to the dysregulated VAT environment induced by HFD feeding. Fig. 7. Accelerated activation but loss of fitness in Bmal1 Δ T[regs] during HFD challenge. Fig. 7 [211]Open in a new tab T[reg]^Bmal1WT and T[reg]^Bmal1Δ mice were fed a LFD for 14 weeks followed by a HFD for indicated durations; VAT T[regs] were isolated at ZT0, schematized in (A). (B) Percent of VAT T[regs]. (C) Volcano plot of VAT-T[reg] transcriptome after 4 weeks of HFD, overlaid with VAT-T[reg] signature genes similar to [212]figure 6D. (D) gMFI of CD44 (left) and CD25 (right) staining after 4 weeks of HFD. (E) Volcano plot of VAT-T[reg] transcriptome after 16 weeks of HFD, overlaid with VAT-T[reg] signature genes. (F) Fraction of CD11c^+ macrophages. (G) Volcano plot of whole VAT transcriptome; up- or downregulated genes with p ≤ 0.05 and ≥1.5 fold change are highlighted; genes of interest are boxed. (H) Gene Set Enrichment Analysis (GSEA) of whole VAT transcriptome. For flow cytometric data, p: * ≤0.05, ** ≤0.01, *** ≤ 0.001 according to Student’s t-test. Data representative or pooled of at least two experiments. NES, normalized enrichment score; HFD, high-fat diet; wks, weeks. Flow-cytometric gating strategies for all quantified or isolated cell-types can be found in [213]fig. S7. Diurnal variation in mitochondrial electron transport chain activity To explore potential mechanisms of how BMAL1 promoted T[reg] fitness, we returned to the rhythmic pathways identified in VAT T[regs] ([214]Fig. 2E). Besides loci associated with TCR activation, we noted that the rhythmically expressed genes in VAT T[regs] encoded molecules related to several mitochondrial oxidative metabolism pathways ([215]Fig. 2E). Genes encoding components of the electron transport chain (ETC) and the tricarboxylic acid (TCA) cycle peaked between ZT20 and ZT0/24 according to JTK_CYCLE analysis ([216]Fig. 8A). We analyzed the metabolic states of VAT T[regs] at ZT0 and ZT12 to be consistent with other experiments, although analysis at ZT20 might have revealed even greater differences. Due to the very low number of VAT T[regs] in individual mice, we could not implement conventional methods such as Seahorse assays to evaluate mitochondrial metabolism. Instead, we turned to flow cytometry to assess mitochondrial oxidative metabolism by measuring the mitochondrial membrane potential (ΔΨ[m]) at steady-state and after inhibition of ATP synthase (Complex V). Catabolism of molecules such as glucose, fatty acids and amino acids ultimately feeds into the TCA cycle, where acetyl-CoA is sequentially oxidized and electrons are passed to the ETC via reduction and oxidation of NAD+/NADH. Electron transport is coupled to translocation of protons from the mitochondrial matrix into the intermembrane space. Such proton translocation generates an electrochemical gradient across the inner mitochondrial membrane, the ΔΨ[m], which is primarily used to power ATP synthesis at Complex V. While the basal ΔΨ[m] is a function of proton influx and efflux, inhibition of Complex V by oligomycin impairs proton influx to the matrix, and the resulting elevation of ΔΨ[m] is indicative of the extent of mitochondrial oxidative metabolism ([217]Fig. 8B). In other words, higher TCA cycle and ETC activity would result in a larger increase in mitochondrial membrane potential after oligomycin treatment. Fig. 8: Circadian regulation of mitochondrial electron transport chain activity. Fig. 8: [218]Open in a new tab (A) Circadian expression profiles of rhythmically expressed genes in the oxidative phosphorylation and TCA cycle pathways. RNA-seq data from [219]figure 2. (B) Schematic of mitochondrial membrane potential at baseline (left) and after inhibition of Complex V by oligomycin (right). Red color emphasizes changes after oligomycin inhibition. (C, D) Basal mitochondrial membrane potential measured by TMRE at ZT0 and ZT12 in VAT (C) and splenic (D) T[regs]. (E) VAT and splenic T[reg] responses to oligomycin. Ratio calculated as post-oligomycin gMFI/basal gMFI. (F) Basal TMRE in p1 ST2^+ (CD69^hi ST2^+) and p2 ST2^+ (CD69^lo ST2^+) VAT T[regs] at ZT0 and ZT12; (G) their response to oligomycin. (H, I) Basal mitochondrial membrane potential of Bmal1 WT and Bmal1 Δ VAT (H) and splenic (I) T[regs] at ZT0. (J) VAT and splenic T[reg] responses to oligomycin in T[reg]^Bmal1WT and T[reg]^Bmal1Δ mice at ZT0. p: * ≤0.05, ** ≤0.01 by Student’s t-test. Basal TMRE plots: representative of at least two independent experiments. Oligomycin/basal FC: pooled from at least two independent experiments. n ≥ 2 per condition per experiment. IMS, intermembrane space; oxphos, oxidative phosphorylation; TMRE, tetramethylrhodamine ethyl ester; FC, fold change. Flow-cytometric gating strategies for all quantified or isolated cell-types can be found in [220]fig. S7. At baseline, VAT T[regs] had a lower ΔΨ[m] at ZT0 than at ZT12, as measured by tetramethylrhodamine ethyl ester (TMRE) labeling ([221]Fig. 8C), unlike splenic T[regs], wherein the ΔΨ[m] remained constant ([222]Fig. 8D). The lower ΔΨ[m] at ZT0 reflected greater oxidative phosphorylation, since inhibition by 1 μM oligomycin induced a greater increase in membrane potential in VAT T[regs] at ZT0 than ZT12 ([223]Fig. 8E); in contrast, splenic T[regs] responded similarly to oligomycin at the two timepoints ([224]Fig. 8E). Within ST2^+ T[regs], the p1 subtype exhibited the most prominent diurnal changes ([225]Fig. 8F, [226]G). VAT T[regs] from T[reg]^Bmal1Δ mice exhibited a higher baseline ΔΨ[m] at ZT0 than their counterparts from Bmal1 WT littermates ([227]Fig. 8H), whereas such differences were not observed in the spleen ([228]Fig. 8I). These ΔΨ[m] differences translated into a greater response to oligomycin in Bmal1 WT than Bmal1 Δ VAT T[regs], but not in the corresponding splenic T[regs] ([229]Fig. 8J). Overall, these findings illustrated VAT-specific diurnal variation in the extent of mitochondrial oxidative metabolism and suggested a role for BMAL1 in promoting ETC activity in VAT T[regs]. DISCUSSION It has been reported that the adaptive immune response is unaffected by the cell-intrinsic clock ([230]12). However, that study focused on lymphocytes within lymphoid organs, and we found that the transcriptomes of T[regs] within non-lymphoid, compared with lymphoid, tissues were enriched in pathways associated with circadian rhythms ([231]1, [232]2). In this report, we showed that genes encoding core components of the cell-intrinsic clock were rhythmically expressed in VAT, but not splenic, T[regs]. T[reg]-specific mutations of core-clock genes impacted VAT, but not splenic, T[reg] activation, metabolism and, ultimately, fitness. These differences had organismal consequences: for example, the cell-intrinsic clock enabled VAT T[regs] to enforce a diurnal rhythm in epididymal VAT lipolysis. As T[regs] refine their phenotype in various tissues, they become more activated and adapt their cellular metabolism to their specific locale ([233]1). There is past and current evidence that both of these processes fall under circadian control. T cells isolated at different ZTs are differentially sensitive to TCR stimulation ([234]36, [235]37). Murine CD8^+ T cells isolated during the day are more robustly activated after recognition of cognate antigen in vitro, and exhibit a diurnal variation in their activation after DC-mediated vaccination in vivo ([236]37). BMAL1 restrained VAT, but not splenic, T[reg] activation, similar to its role in other immunocyte populations. Inflammatory cytokine production in macrophages or IL-22 and IL-17 production in ILC3s are heightened after Bmal1 or Nr1d1 deletion ([237]38–[238]44). The brakes on cell activation imposed by BMAL1 in multiple immunocyte lineages that lack TCRs also suggest that the NF-κB signaling pathway might be a primary target of circadian regulation. Indeed, Bmal1 deletion heightens NF-κB signaling in macrophages ([239]38) and intestinal epithelial cells ([240]45). Many of the rhythmically expressed genes in VAT T[regs] belonged to metabolic pathways. Our results evidenced a clear diurnal variation in the TCA cycle and in ETC activity. Rhythms in mitochondrial ETC activity can originate from several processes. For instance, mitochondria undergo diurnal cycles of fusion and fission ([241]46–[242]48), which modulate the efficiency of ATP production. Fused mitochondria, which are more efficient at producing ATP, occur early in the day ([243]46–[244]49) and are associated with a heightened mitochondrial oxygen consumption rate and ATP production ([245]47, [246]48). In VAT T[regs], the timing of increased oxidative metabolism coincided with the period of heightened activation state to potentially meet an elevated energy demand. These variations were not present in splenic T[regs], highlighting the different metabolic needs of their VAT-T[reg] counterparts. In the absence of these and presumably other temporal modulations, VAT T[regs] lost fitness in competitive transfers, in female Bmal1 heterozygotes, and in response to HFD challenge. The reduced fitness resulted from disruption of the core-clock program rather than loss of BMAL1 expression per se, since adoptive transfer of either Bmal1 or Nr1d1 Δ T[regs] led to loss of fitness. Some of the phenotypes in T[reg]^Bmal1Δ mice might be attributable to other core-clock proteins, such as REV-ERBα, whose expression was significantly altered by loss of BMAL1. The constitutively heightened activation state in Bmal1-deficient T[regs] suggests dysregulation of the NF-κB signaling pathway, which is intimately related to apoptosis and necrosis. Indeed, the upstream regulators of NF-κB signaling serve a second, less recognized, function as cell death checkpoints, and NF-κB target genes are known to regulate cell-death (reviewed in ([247]50, [248]51)). However, the in vivo signals that result in cell death instead of pro-survival signaling by NF-κB in VAT T[regs] after competitive transfer remain undefined. Furthermore, ETC activity appeared dampened in Bmal1 Δ VAT T[regs] and might contribute to the loss of fitness. BMAL1 promotes mitochondrial oxidative metabolism ([249]42, [250]46, [251]52). An inefficient mitochondrial respiration in the absence of BMAL1 increases the production of reactive oxygen species (ROS) ([252]53) and ROS exhibits circadian rhythmicity ([253]41, [254]42, [255]47). Poorly optimizied mitochondrial function can impair T[reg] fitness and function, as has been shown in other reports ([256]54–[257]58). We uncovered a role for VAT T[regs] in regulating diurnal lipolysis of the adipose tissue, which requires BMAL1 expression in T[regs]. Norepinephrine-induced lipolysis is an important mechanism for providing nutrients during states of energy deprivation ([258]59), but excessive lipolysis recruits inflammatory infiltrates to the adipose tissue ([259]60, [260]61). Thus, adipocyte lipolysis is highly regulated. In mice, lipolysis peaks near ZT0 ([261]31), and we recapitulated these findings in vitro as VAT explants isolated at ZT0 resisted further norepinephrine stimulation, compared with those taken at ZT12. We found that ST2^+ T[regs], in particular the p1 subtype, suppressed lipolysis in lean animals, and rhythmic lipolysis was disrupted in both DT-treated Foxp3-DTR^+ mice and in T[reg]^Bmal1Δ animals. It seems likely that VAT T[regs] regulate lipolysis by controlling the inflammatory tenor of the adipose tissue, since cytokines such as TNFα induce lipolysis ([262]62, [263]63). Alternatively, T[regs] might control other VAT immunocytes that modulate sympathetic nervous signaling, such as macrophages ([264]32, [265]64). Lastly, VAT T[regs] might control the sympathetic tone directly through an unknown mechanism. That VAT T[regs] enforce diurnal lipolysis is consistent with their role in promoting metabolic homeostasis in VAT ([266]14, [267]16, [268]17). It is unclear how VAT T[regs] are entrained by circadian rhythms. Identifying specific signals that entrain them, indeed non-neuronal cells in general, remains an important goal in the field. Hormonal, neuronal and metabolic signals are drivers of some of the mechanisms (reviewed in ([269]6)). TCR activation upregulates the expression of core-clock genes such as Bmal1 and Nfil3 ([270]65, [271]66), and thus might kick-start the cell-intrinsic clock program. Metabolic cues such as insulin signaling ([272]67) might also entrain resident immunocytes. Given the diurnal lipolysis of adipose tissue, whether lipid species also provide entrainment cues to VAT T[regs] is an intriguing possibility. While we did not find most core-clock genes to be significantly rhythmic in splenic T[regs], it remains possible that a more frequent and prolonged sampling, for instance over 48 hours with a 2-hour sampling interval, might detect weak rhythmicity in the expression of other core-clock genes. Nonetheless, other studies have noted a similarly weak rhythmicity in splenocytes ([273]12, [274]13). Rhythmicity in VAT T[regs] entailed changes on the order of 2-fold, which is less than what has been observed with some cell-types, such as hepatocytes, but is similar to observations on other immunocytes ([275]68, [276]69). Lastly, due to the very low number of VAT T[regs] per mouse, we could not perform metabolomics or metabolite-tracing experiments to pinpoint the specific metabolic reactions under diurnal control. Tissue microenvironments undergo significant changes throughout the day. The elevated rhythmicity of core-clock gene expression provides a molecular mechanism for VAT T[regs] to synchronize and adapt to the changing tissue environment. An improved understanding of the diurnal behaviors of tissue T[regs] might help maximize the efficacies of T[reg]-based immunotherapeutic strategies. MATERIALS AND METHODS Study design This study aimed to establish to what extent circadian rhythms occurs in T[regs] in VAT versus the spleen, and how important it is. To that end, we performed transcriptomic profiling by population-level and single-cell RNA sequencing over circadian time, as well as cytofluorimetry-based metabolic assays in C57BL/6 and mutant littermates lacking core-clock components. Mice C57BL/6 (B6), B6.CD45.1 (stock number 002014, B6.SJL-Ptprca Pepcb/BoyJ) and Bmal1^flox/flox (stock number 007668, B6.129S4(Cg)-Arntltm1Weit/J) mice were obtained from the Jackson Laboratory. Nr1d1^flox/flox mice have been reported (University of Pennsylvania) ([277]70). Foxp3-Ires GFP.hDTR mice were obtained from A. Rudensky (Memorial Sloan Kettering Cancer Center). The B6.Foxp3-Ires GFP ([278]71), B6.Foxp3-Cre.YFP ([279]72), and VAT TCR-tg lines ([280]18) have been described. Mice lacking Bmal1 or Nr1d1 specifically in T[regs] were generated by crossing B6.Foxp3-Cre.YFP mice with the respective flox strains, and are typically referred to in the text as T[reg]^Bmal1Δ or Treg^Nr1d1Δ For transfer experiments, knock-out donor strains were generated by crossing B6.Foxp3-Cre.YFP.CD45.2^+ mice with VAT TCR-tg mice and either the Bmal1^flox/flox or Nr1d1^flox/flox strain to obtain Foxp3-Cre.YFP.CD45.2^+[.]TCR-tg^+ Bmal1^flox/flox or Nr1d1^flox/flox offspring. Female heterozygous Cre strains were generated by crossing Foxp3-Cre.YFP^+/+Bmal1^flox/flox with Bmal1^flox/flox mice. Wild-type donor strains were obtained by crossing B6.Foxp3-Cre.YFP mice with B6.CD45.1 and VAT TCR-tg mice to obtain Foxp3-CreYFP.CD45.1^+.TCR-tg^+ offspring. Recipients were generated by crossing B6.Foxp3-Cre.YFP with B6.CD45.1 to obtain Foxp3-Cre.YFP.CD45.1/2^+ offspring. Appropriate littermate controls were always used except for transfer experiments, where co-housed donors of similar age were used. All animals were housed in specific pathogen-free facilities at Harvard Medical School’s Center for Animal Resources and Comparative Medicine. All experiments were performed under protocols approved by the HMS Institutional Animal Care and Use Committee (protocol# IS00001257). Mice were housed under a strict 12h:12h light: dark cycle. In some experiments, they were entrained in a reverse light-cycle room for a minimum of 14 days with a 12h:12h dark: light cycle, which enabled simultaneous tissue collection from mice at different circadian time-points. To assess adaptation to reverse light cycle, mice were housed individually with unrestricted access to running wheels equipped with activity tracking (Columbs Instruments). Wheel revolutions were recorded in each cage every 10 seconds and cumulative wheel turns from all mice are plotted. Unless otherwise noted, weaned mice were fed a low-fat-diet chow (no.5053, Picolab Rodent Diet 20; 13% kcal fat) ad libitum. For high-fat-diet experiments, mice were fed a low-fat-diet chow (no.5053) ad libitum until 14 weeks of age, then fed a high-fat diet (Open Source Diet D12492, 60% kcal fat) ad libitum for 4, 8 or 16 weeks. Tissue preparations for flow cytometry and cell sorting Tissue preparation has been previously described with slight modifications ([281]29) Briefly, male mice were euthanized by CO[2]. Epididymal VAT and spleen were excised and placed in Dulbecco’s modified Eagle’s medium (DMEM) containing 2% fetal bovine serum (FBS) and 10mM HEPES (referred to as FACs buffer). VAT depots were minced with scissors and incubated with FACs buffer containing 1.5mg/mL collagenase type II (Sigma) at 37°C for 20 minutes. The adipocyte layer was removed after centrifugation at 420 g. Red blood cells were lysed by ACK lysing buffer (Gibco). Spleen and lymph nodes were mechanically dissociated by mashing against a 40 μM filter, followed by red blood cell removal by ACK lysis. For liver, lung and kidney preparations, excised tissues were minced with scissors and incubated in 1% FCS DMEM containing 0.5mg/mL collagenase IV (Gibco), 150 μg/mL DNase I (Sigma) at 37°C for 30–45 minutes. Digested tissues were filtered and washed. For liver and kidney, leukocytes were enriched by centrifugation (10 min at 800 × g) with 36% Percoll (GE Healthcare). For surface-antigen staining, single-cell suspensions were incubated in phosphate-buffered saline (PBS) containing live/dead stain and surface antibodies for 10 minutes on ice, followed by washes in FACs buffer. For intracellular staining, single-cell suspensions were fixed in eBioscience Fix/Perm buffer for 20–30 minutes at room temperature, followed by permeabilization in eBioscience permeabilization buffer. Cells were stained with intracellular antibodies for 30 minutes at room temperature followed by washes with permeabilization buffer. The following antibodies were used: anti-mouse -CD4 (GK1.5), -CD8α (53–6.7), -CD11b (M1/70), -CD11c (N418), -CD19 (6D5), -CD39 (Duha59), -CD45 (30-F11), CD45.1 (A20), -CD45.2 (104), CD62L (MEL-14), -CD69 (H1.2F3), -Il18r/CD218α (BG/IL18RA), -CD44 (IM7), -Il17r/CD127 (A7R34), -CD206 (C068C2), -Ly6c (HK1.4), -F4/80 (BM8) and -I-A/I-E (M5/114.15.2) all from BioLegend; -CD25 (PC61), -TCRβ (H57–597) and -Ki67 (B56) from BD bioscience; -Foxp3 (FJK-16s), -Il33R/ST2 (RMST2–2) and -Klrg1 (2F1) from Thermofisher. Live/Dead fixable viability dye yellow, near-IR or UV (Thermofisher) were used to distinguish live and dead cells. Typically T[regs] were defined as CD45^+ TCRβ^+ CD4^+ Foxp3^+. For cell sorting, T[regs] were defined as CD45^+ Tcrb^+ CD4^+ YFP^+ CD8α^− CD11b^− CD11c^− CD19^− DAPI^−, unless otherwise specified. For 5-ethynyl-2’-deoxyuridine (EdU) experiments, 40μg/g of body weight EdU (Thermofisher) was intraperitoneally injected 12 hours before sacrifice. EdU was detected using Click-iT plus Pacific Blue Flow Cytometry Assay Kit (Thermofisher) according to the manufacturer’s instructions. RNA Sequencing RNA sequencing was performed as described at [282]www.immgen.org. 1000 T[regs] were double-sorted directly into 5 μl TCL-buffer (Qiagen) containing 1% β-mercaptoethanol (Sigma). For adoptive transfer experiments, 500 cells were double-sorted. Smart-seq2 libraries were prepared and sequenced as previously described ([283]73) using the Broad Genomics Platform. Briefly, RNA was captured and purified using RNAClean XP beads (Beckman Coulter). Polyadenylated mRNA was selected using an anchored oligo(dT) primer (50 –AAGCAGTGGTATCAACGCAGAGTACT30VN-30) and converted to cDNA by reverse transcription. Limited PCR amplification was performed on the first-strand cDNA followed by Tn5 transposon-based fragmentation using the Nextera XT DNA Library Preparation Kit (Illumina). Samples were PCR amplified for 12 cycles using barcoded primers such that each sample carried a specific combination of eight base Illumina P5 and P7 barcodes for subsequent pooling and sequencing. Paired-end sequencing was performed on an Illumina NextSeq 500 using 2 × 38bp reads with no further trimming. Reads were aligned to the mouse genome (GENCODE GRCm38/mm10 primary assembly and gene annotations vM16; [284]https://www.gencodegenes.org/mouse_releases/16.html) with STAR 2.5.4a ([285]https://github.com/alexdobin/STAR/releases). Ribosomal RNA gene annotations were removed from GTF (General Transfer Format) file. Gene-level quantification was calculated by featureCounts ([286]http://subread.sourceforge.net/). Raw read counts tables were normalized by the median of ratios method with DESeq2 package from Bioconductor ([287]https://bioconductor.org/packages/release/bioc/html/DESeq2.html) and then converted to GCT and CLS format. Samples with less than 1 million uniquely mapped reads or with fewer than 8,000 genes over ten reads were considered low quality and removed, followed by further screening for contamination by using known cell-type-specific transcripts. Biological replicates with poor Pearson correlation (<0.9) were removed. Genes with less than 10 reads across samples or a high coefficient of variation (typically >0.6) were removed. Differential gene-expression analysis was performed using EdgeR ([288]74); differentially expressed genes were defined as p ≤ 0.05 according to quasi-likelihood F-test. For pathway enrichment analysis, differentially expressed genes were analyzed by Ingenuity Pathway Analysis (Qiagen, [289]https://www.qiagenbioinformatics.com/products/ingenuitypathway-ana lysis) ([290]75), Gene Set Enrichment Analysis (Subramanian et al 2005) or EnrichR ([291]76, [292]77). Enriched pathways were defined as FDR ≤0.05. In some cases, previously established signatures were tested for enrichment in our dataset, where significant enrichment was defined as p ≤0.05 according to Fisher’s exact test. For whole-tissue RNA-sequencing, RNA was extracted from VAT using the RNeasy lipid tissue mini kit following the manufacturer’s protocol (Qiagen). 2ng of RNA was added into 5 μl TCL-buffer (Qiagen) containing 1% β-mercaptoethanol (Sigma) and sequenced as above. RT-PCR RNA was extracted from 100,000 sorted splenocytes using Trizol (Invitrogen) following the manufacturer’s protocol. Complementary DNA synthesis was performed using a Maxima H Minus Reverse Transcriptase kit (Thermo Fisher Scientific). Quantitative PCR (qPCR) was run on a QuantStudio 5 Real-Time PCR System (Applied Biosystems). The expression of Bmal1 was calculated by the 2^−ΔΔCt method relative to the expression of the ribosomal subunit 36b4 (Rplp0). Primer sequences: Bmal1 AGGATCAAGAATGCAAGGGAGG (F), TGAAACTGTTCATTTTGTCCCGA (R); Rplp0 GGAGTGACATCGTCTTTAAACCCC (F), TCTGCTCCCACAATGAAGCA (R). Adoptive transfers Cells from 6-to-8-week-old male donors were pooled from spleen and peripheral lymph nodes, followed by enrichment for CD4^+ cells using Dynabeads untouched Mouse CD4 (Thermofisher). Cells were single sorted for YFP^+ T[regs]. For the competitive transfer experiments, a 1:1 mix of 250,000 donor T[regs] from wild-type or knock-out donors (500,000 total T[regs]) were resuspended in PBS and intravenously injected via the tail vein into 8-to-10-week-old male recipients. Donor and recipient mice were fed a low-fat-diet chow with 21% kcal fat (no. 5058, Picolab Rodent Diet 20) scRNA-seq 34,000 VAT and 32,000 splenic CD4^+ T cells were sorted and multiplexed using TotalSeq hashing antibodies (BioLegend). Cell encapsulation, RNA isolation, and hash library construction and sequencing were performed by the Broad Institute Genomic Services using the 10X Genomics platform. Samples were processed using 3’ v3.1 chemistry and hashed cells were loaded onto one lane. cDNA and gene expression libraries were constructed following the manufacturer’s instructions (10X Genomics). Gene expression libraries were sequenced on HiSeq X. Samples were demultiplexed using cellranger v6.0 and hash libraries were processed using cite-seq-count ([293]https://hoohm.github.io/CITE-seq-Count/) Data analysis was performed following the Seurat v3 pipeline ([294]78). Low-quality cells were removed and were defined as those with less than 700 or more than 3000 unique genes, as well as those with >10% mitochondrial gene reads. Clusters of proliferating cells were also removed. Non-CD4^+ T cell contaminants were removed using well-known cell-identity markers. Clustering, dimensionality reduction onto UMAP, differential gene expression, and visualization were performed using the built-in functions of Seurat v3. Density distributions were calculated using two-dimensional kernel density estimation. RNA velocity was performed as described ([295]27). Briefly, the spliced and unspliced counts were calculated from the original sequencing data using the RNA velocity package ([296]26). The spliced and unspliced expression matrices were then integrated into the single-cell data matrix, and the scvelo package was used to calculate the velocity vectors using the dynamical model of transcriptional dynamics and splicing kinetics. The calculated velocity vectors were then projected onto the UMAP embedding calculated during the scRNA-seq analysis. Lipolysis assays The in vitro lipolysis assay was adapted from previous studies ([297]32, [298]33, [299]79). Briefly, approximately 20mg of epididymal VAT was excised into 200μl of 1g/L glucose DMEM (Gibco) supplemented with 10 mM HEPES, 2% fatty-acid free bovine serum albumin (Sigma), and 5 μM Triacsin C (Sigma) (collectively referred to as “lipolysis buffer”), with or without norepinephrine (A0937 Sigma) for 1 hour at 37°C. Explants were then transferred into fresh lipolysis buffer with or without NE for an additional hour, and the supernatant was collected for glycerol quantification following the manufacturer’s protocol (SGA1, Zenbio). To quantify protein content, the we placed the fat pad into a 1 mL of chloroform:methanol (2:1 v/v) with 1% glacial acetic acid (extraction solution) for 1 hour at 37°C while rotating, and then incubated the material in 500μl of 0.3N NaOH with 0.1% SDS in dH[2]O (lysis buffer) overnight at 55 °C. The lysis buffer supernatant was taken for protein quantification using the Pierce BCA Assay following the manufacturer’s protocol (Thermofisher). Glycerol release rate was expressed as μM glycerol per milligram protein per hour. For induction of lipolysis in vivo, 1mg/kg CL316,243 (Sigma) was i.p-injected. Serum was collected via the tail vein using non-heparinized capillary tubes (Fisher) and was allowed to clot at room temperature for 15 minutes. After centrifugation at 2000 × g for 15 minutes, the supernatant was assayed for free fatty acid or glycerol levels following the manufacturer’s protocol (GFA-1 Zenbio). Foxp3-DTR^+ mice was i.p-injected with 20ng per g body weight of DT once per day, at ZT10-ZT12, for three consecutive days. Organismal metabolism Organismal metabolism was assessed by the glucose tolerance test (GTT) and insulin tolerance test (ITT). For the GTT, mice were fasted for 5 hours and acclimatized to the experiment room for 45 minutes, followed by intraperitoneal injection of 1g/kg body weight of glucose. Glucose levels were then measured from tail vein blood at 0, 20, 40, 60, 90 and 120 minutes after the glucose injection. For ITT, mice were fasted for 4 hours, followed by 45 minutes of acclimatization to the experiment room. They were intraperitoneally injected with 0.6 unit/kg (Humulin R U-100 from Eli Lilly) body weight, followed by glucose level measurements at 0, 20, 40, 60, 90 and 120 minutes after injection. Blood glucose levels were measured using a Contour glucose meter. Cellular metabolism Mitochondrial membrane potential was measured using tetramethylrhodamine ethyl ester perchlorate (TMRE) from Thermofisher. Single-cell suspensions were stained with surface antibodies as described above and washed in FACs buffer. Cells were then incubated in round-bottom 96-well tissue-culture plate (Corning) in 100μl of DMEM + 2% FBS + 10 mM HEPES + 2mM glutamine at 37°C for 30 minutes. Where applicable, cells were treated with 1 μM oligomycin (delivered at 3X concentration in 50 μL) at 37°C for 10 minutes. Finally, cells were stained with 100 μM TMRE (delivered at 4X in 50 μL) at 37°C for 20 minutes. Cells were spun down and resuspended in cold FACs buffer without any further washes before immediate acquisition on the flow cytometer. Statistical analyses Data are routinely shown as mean ± SD. Unless stated otherwise, statistical significance was determined by two-tailed Student’s t-test using GraphPad Prism 9.0. * p <0.05; ** p <0.01; *** p < 0001. A value of more than three standard deviations from the mean was adopted as criteria to exclude outliers. Independent replicate experiments were typically performed with at least two biological replicates per group. Circadian rhythmicity of gene expression was assessed using JTK_CYCLE ([300]80). Rhythmic genes were defined as p ≤ 0.05, gene expression ≥ 20, and period ≥ 16 hours. Time of peak expression was determined by JTK_CYCLE. Supplementary Material XiaoEtAl-FigsS1-S6 fig. S1: Expression of core-clock genes. fig. S2: Clock genes confer fitness to VAT T[regs]. fig. S3: T[reg] heterogeneity. fig. S4: Characterization of Bmal1Δ Tregs. fig. S5: VAT Tregs regulate adipocyte lipolysis. fig. S6: T[reg]^Bmal1WT and T[reg]^Bmal1Δ mice during HFD challenge fig. S7: Gating strategies [301]NIHMS1854639-supplement-XiaoEtAl-FigsS1-S6.pdf^ (736KB, pdf) XiaoEtAl_RawDataFile [302]NIHMS1854639-supplement-XiaoEtAl_RawDataFile.xlsx^ (4.7MB, xlsx) XiaoEtAl_Table_S1 Table S1: JTK analysis of VAT and splenic T[reg] transcriptome over circadian time [303]NIHMS1854639-supplement-XiaoEtAl_Table_S1.xlsx^ (4.6MB, xlsx) ACKNOWLEDGEMENTS