Abstract N^6-methyladenosine (m^6A) is among the most abundant mRNA modifications, yet its cell-type-specific regulatory roles remain unclear. Here we show that m^6A methyltransferase-like 14 (METTL14) differentially regulates transcriptome in brown versus white adipose tissue (BAT and WAT), leading to divergent metabolic outcomes. In humans and mice with insulin resistance, METTL14 expression differs significantly from BAT and WAT in the context of its correlation with insulin sensitivity. Mettl14-knockout in BAT promotes prostaglandin secretion, improving systemic insulin sensitivity. Conversely, Mettl14-knockout in WAT triggers adipocyte apoptosis and systemic insulin resistance. m^6A-seq and RNA-seq integration revealed upregulated prostaglandin biosynthesis pathways in BAT and apoptotic pathways in WAT with Mettl14 deficiency. Stable METTL14-knockout hBAs/hWAs show METTL14-mediated m^6A promotes mRNA decay of PTGES2 and CBR1 in hBAs and TRAIL and TNFR1 in hWAs. These data shed light on the ability of m^6A to impact metabolism in a cell-type-specific manner with implications for influencing the pathophysiology of metabolic diseases. Subject terms: Insulin signalling, Apoptosis, RNA metabolism __________________________________________________________________ N6 - methyladenosine (m6 A) is a prevalent and abundant mRNA modification, yet its cell-type specific roles remains unclear. Here, the authors show that m6 A modification differentially regulates the transcriptomes of brown and white adipose tissue, resulting in distinct impacts on systemic metabolism. Introduction N^6-methyladenosine (m^6A) modification is a pivotal post-transcriptional regulatory process in mammalian cells, controlled by writers (METTL3, METTL14, WTAP), erasers (ALKBH5, FTO), and readers (YTHDC, YTHDF, IGF2BP2)^[48]1,[49]2. m^6A methylation of mRNA can affect its expression, stability, and translation efficiency, leading to altered protein abundance to determine diverse biological outcomes related to cell fate determination, organ development, cell proliferation, and/or survival^[50]1,[51]2. Indeed, this ability to modify mRNA to regulate intricate cell processes can be an advantage considering the human body comprises more than 200 distinct cell types, each with specialized roles in physiological processes. Nevertheless, m^6A has also shown divergent functions; for example, METTL14-mediated m^6A modification exhibits both oncogenic^[52]3–[53]5 and anti-tumor effects^[54]6,[55]7 across cancer types. A lingering question in the field of m^6A biology pertains to the precise molecular underpinnings by which m^6A governs cell-type-specific modulation of gene expression in a contingent fashion, thereby affording precise control over diverse cellular functionalities. Therefore, understanding cell-type-specific m^6A regulation is crucial for unraveling the complexities of cellular processes and for developing targeted therapeutic strategies for various pathophysiological states including metabolic diseases^[56]8. Brown adipose tissue (BAT) and white adipose tissue (WAT) are two major types of tissues that share some common features but also have distinct properties. BAT is specialized for energy expenditure through thermogenesis and is acknowledged as a secretory organ capable of producing various batokines that play roles in energy metabolism, inflammation, and/or whole-body insulin sensitivity^[57]9,[58]10. WAT primarily functions as an energy storage depot, accumulating excess energy in the form of triglycerides (TGs)^[59]9,[60]11,[61]12. In addition, WAT also secretes endocrine hormones that can regulate the metabolism of distal organs. Dysregulation of WAT, specifically in the context of perturbed signaling pathways involving cytokines of the tumor necrosis factor (TNF) family, underlies the initiation of cellular apoptosis and inflammation within the adipose tissue, eventually resulting in lipid spill-over, glucotoxicity, and insulin resistance^[62]13. Hence, the context-specific regulation within distinct cell types such as BAT and WAT holds relevance for the modulation of whole-body metabolism. The involvement of m^6A modification in BAT and WAT has recently emerged as a nascent yet critical realm of investigation, particularly within the scope of addressing obesity and metabolic disorders. While METTL3, METTL14, and WTAP are classified as major components of the m^6A writer complex, previous research including our own argues for unique roles for each. Some studies have investigated the impact of m^6A by focusing on METTL3 and WTAP on adipose tissue biology and function, with a particular focus on BAT or beige adipocytes^[63]14–[64]18. Our previous study indicates that Mettl14 deficiency in BAT promotes the secretion of beneficial endocrine factor(s) to protect mice from diet-induced obesity (DIO) and insulin resistance independent of BAT thermogenesis^[65]19. In this manuscript, we directly address the cell-type specific regulation involving m^6A by examining the roles of METTL14 in BAT versus WAT in the modulation of whole-body metabolism. In this study, we build on our preliminary observations of distinct expression patterns of METTL14 in BAT versus WAT from humans and mice with insulin resistance. The BAT-specific (Ucp1-cre-driven) Mettl14-knockout (M14^KO) mouse model exhibits enhanced secretion of endocrine factor(s) with improved systemic metabolic homeostasis that is independent of BAT thermogenic function. Conversely, an independent mouse model with Adipoq-cre-driven Mettl14-knockout shows white adipocyte apoptosis that consequently reduces its lipid storage capacity, impairing whole-body insulin sensitivity and energy expenditure. m^6A-MeRIP-sequencing (m^6A-seq) and RNA-sequencing (RNA-seq) in Mettl14-deficient BAT versus Mettl14-deficient WAT revealed different methylomes and transcriptomes in the two cell types. Mechanistically, Mettl14-mediated m^6A modification specifically destabilizes critical BAT transcripts associated with phospholipid metabolic processes, while in WAT, it negatively regulates TNFα and apoptosis pathway-related genes via m^6A-dependent and independent mechanisms. These findings underscore the divergent roles of METTL14 in BAT versus WAT-mediated metabolism, emphasizing cell-type and context-specific m^6A regulation as a significant consideration for addressing metabolic disorders. Results Differential abundance of METTL14 in BAT versus WAT obtained from humans or mice with insulin resistance To distinguish the m^6A methylome landscapes between BAT and WAT, we compared RNA-seq and m^6A-MeRIP-seq analyses between fully differentiated human brown adipocytes (hBAs) versus human white adipocytes (hWAs)^[66]20. To substantiate that m^6A selectively modifies transcripts expressed in hBAs and hWAs, we first filtered the transcripts unique to hWAs or hBAs and identified 25,563 transcripts that were ubiquitously expressed in both cell types (Supplementary Fig. [67]1a). Next, to specifically focus on the m^6A-modified transcripts, we intersected these 25,563 transcripts with those that are m^6A-modified, to reveal a total of 7866 m^6A-modified transcripts (Supplementary Fig. [68]1b). Principal component analysis (PCA) of the m^6A-seq data revealed a distinct separation between hBAs and hWAs (Supplementary Fig. [69]1c). About a quarter (2086) of the 7866 m^6A-modified transcripts showed comparable m^6A methylation levels between hBAs and hWAs (Supplementary Fig. [70]1d). On the other hand, 2681 and 3099 hypermethylated m^6A transcripts were exclusively identified in hBAs and hWAs, respectively (Supplementary Fig. [71]1d). These differentially m^6A-modified transcripts were notably enriched in pathways associated with VEGFA-VEGFR2, TGFβ, mRNA processing, insulin, TNFα, or apoptosis modulation signaling pathways (Supplementary Fig. [72]1e). Thus, brown and white adipocytes manifest highly distinct m^6A modification patterns that strongly align with cell-type-specific gene expression programs, pointing to functional specificity of m^6A at the cellular/tissue level. These preliminary results provided us with the rationale to explore the direct impact of m^6A modulators in these two different metabolic cell types. METTL14, as a key component of the m^6A writer complex, has been reported to exhibit pronounced cell-type-specific roles within the context of metabolism^[73]19,[74]21. For example, our previous studies revealed that m^6A hypomethylation, and more specifically METTL14 downregulation, led to impaired β-cell function and identity^[75]21. Conversely, METTL14 showed higher gene expression in BAT from humans with obesity, and BAT-specific Mettl14-knockout mice presented improved systemic insulin sensitivity^[76]19. Considering these observations, we hypothesized that METTL14 has divergent roles in the transcriptional programs to regulate the functions of brown versus white adipocytes. To investigate this hypothesis, we first examined METTL14 protein abundance in the fully differentiated hBAs and hWAs under physiological conditions. We observed a higher relative METTL14 protein level in hWAs compared to hBAs cells (Supplementary Fig. [77]1f, g). In C57BL/6 N male mice fed a chow diet (CD), METTL14 protein abundance was significantly higher in inguinal white adipose tissue (iWAT) than epididymal white adipose tissue (eWAT) or interscapular brown adipose tissue (iBAT) (Supplementary Fig. [78]1h, i). These data support the notion that METTL14 exhibits adipocyte- and adipose depot-specific expression patterns under physiological conditions. Next, we examined whether METTL14 exhibits differential expression across distinct human WATs, such as individuals with varying levels of body mass index (BMI) or insulin sensitivity (Supplementary Table [79]1). First, a comparison between lean, insulin-sensitive individuals and those with obesity but who are also insulin-sensitive revealed no significant differences in METTL14 protein abundance (Supplementary Fig. [80]1j, [81]k). Likewise, when comparing individuals who are lean and insulin-resistant to those with obesity and insulin-resistant, no significant differences were detected in the levels of METTL14 protein (Supplementary Fig. [82]1l, [83]m). This lack of distinction may be attributable to patient heterogeneity and it is possible that METTL14 abundance is not intrinsically linked to human BMI. Next, to isolate the influence of insulin sensitivity from BMI, we compared individuals with obesity who are insulin-sensitive (HOMA-IR < 2.9) to individuals with obesity who are insulin-resistant (HOMA-IR > 2.9). Remarkably, METTL14 protein level was significantly lower in both subcutaneous white adipose tissue (scWAT) and visceral white adipose tissue (vsWAT) from humans with insulin resistance (Fig. [84]1a, [85]b). Spearman correlation analysis revealed a robust negative correlation between METTL14 abundance and HOMA-IR in scWAT or vcWAT (Fig. [86]1c, [87]d). Fig. 1. METTL14 exhibits differential protein abundance in BAT or WAT from insulin-resistant humans and mice. [88]Fig. 1 [89]Open in a new tab a, b Western blots (a) and quantification (b) of METTL14 and VINCULIN in subcutaneous and visceral white adipose tissues from individuals with obesity who are insulin-sensitive or insulin-resistant (n = 11 for insulin-sensitive individuals with obesity in scWAT and n = 26 for insulin-resistant individuals with obesity in scWAT; n = 11 for insulin-sensitive individuals with obesity in vcWAT and n = 13 for insulin-resistant individuals with obesity in vcWAT). IS, insulin-sensitive; IR, insulin-resistant. scWAT from insulin-sensitive individuals with obesity was loaded in both the upper and lower panels of (a) to accommodate the insulin-resistant samples. c, d Spearman correlation between METTL14 relative abundance in scWAT (c) or vcWAT (d) and insulin sensitivity, as indicated by HOMA-IR (n = 11 for insulin-sensitive individuals with obesity in scWAT and n = 26 for insulin-resistant individuals with obesity in scWAT; n = 11 for insulin-sensitive individuals with obesity in vcWAT and n = 13 for insulin-resistant individuals with obesity in vcWAT). e, f Western blots (e) and quantification (f) of METTL14 and VINCULIN in iWAT, eWAT, and iBAT from control (ob + ) or ob/ob mice (n = 6/group). iWAT, inguinal white adipose tissue; eWAT, epididymal white adipose tissue; iBAT, interscapular brown adipose tissue. g–i Spearman correlation between METTL14 relative abundance in iWAT (g), eWAT (h), or iBAT (i) and insulin sensitivity indicated by HOMA-IR (n = 6/group). j, k Western blots (j) and quantification (k) of METTL14 and VINCULIN or GAPDH in iWAT, eWAT, and iBAT from control (db + ) or db/db mice (n = 6/group). l–n Spearman correlation between METTL14 relative abundance in iWAT (l), eWAT (m), or iBAT (n) and insulin sensitivity indicated by HOMA-IR (n = 6/group). o, p Western blots (o) and quantification (p) of METTL14 and VINCULIN or GAPDH in iWAT, eWAT, and iBAT from LFD- or HFD-fed mice (n = 6/group). q–s Spearman correlation between METTL14 relative abundance in iWAT (q), eWAT (r), or iBAT (s) and insulin sensitivity indicated by HOMA-IR (n = 6/group). All samples in each panel are biologically independent. Data are presented as means ± SEM from two independent experiments by two-tailed unpaired t-test (b, f, k, p) and one-tailed Spearman correlation (c, d, g–i, l–n, q–s). See also Supplementary Fig. [90]1. Source data are provided as a Source Data file. Consistent with the human data we detected similar changes in three different mouse models. First, we examined METTL14 protein levels in the fat tissues from the ob/ob (Lep^ob) mouse, which is a model for T2D and obesity that is characterized by hyperinsulinemia (Supplementary Fig. [91]1n), glucose intolerance (Supplementary Fig. [92]1o), and insulin resistance (Supplementary Fig. [93]1p). The METTL14 protein levels were lower in iWAT and eWAT from ob/ob mice compared to ob+ mice (Fig. [94]1e, [95]f). On the other hand, the METTL14 protein level was higher in the ob/ob-iBAT as compared to ob/+-iBAT (Fig. [96]1e, [97]f). Thus, in the ob/ob mouse model, METTL14 in WATs is positively correlated with insulin sensitivity, and its protein abundance in BAT is negatively correlated with insulin sensitivity (Fig. [98]1g–[99]i). In the db/db (Lepr^db) mouse model, which exhibits hyperinsulinemia (Supplementary Fig. [100]1q) and peripheral tissue insulin resistance (Supplementary Fig. [101]1r), METTL14 protein level was lower in iWAT and eWAT as compared to the db+ controls (Fig. [102]1j, [103]k). Conversely, elevated protein abundance of METTL14 was observed in iBAT dissected from db/db mice (Fig. [104]1j, [105]k). METTL14 displayed an inverse relationship with WAT-mediated insulin sensitivity and a positive correlation with BAT-mediated whole-body insulin sensitivity (Fig. [106]1l–[107]n). Comparable outcomes were evident in the high-fat diet (HFD)-induced obese (DIO) mice compared to the low-fat diet (LFD)-fed group which comprised lean insulin-sensitive mice (Fig. [108]1o, [109]s). The lack of a significant correlation between METTL14 protein levels in iBAT and whole-body insulin sensitivity could be related, in part, to the short duration of HFD feeding (Fig. [110]1s). Collectively, these data argue that METTL14 plays a differential role in BAT versus WAT to regulate whole-body metabolism. Mettl14 deficiency promotes contrasting modulation of BAT- versus WAT-mediated systemic insulin sensitivity in mice To comprehensively interrogate the divergent roles of METTL14-mediated m^6A modifications in BAT versus WAT and their respective effects on whole-body metabolic processes, we employed a dual approach. Specifically, we generated two distinct lines of knockout mice by breeding Mettl14-floxed (M14^fl/fl) mice^[111]21, with either a) Adipoq-cre mice^[112]22, which facilitated exploration of METTL14’s role in the overall functionality of adipose tissue, or b) Ucp1-cre mice^[113]23, enabling a focused analysis of BAT-specific Mettl14 knockout and its repercussions on systemic metabolism (knockouts verified in Supplementary Fig. [114]2a, [115]b). Both knockout-out lines displayed a typical female and male ratio and normal birth counts. Under CD feeding, both M14^fl/fl-Ucp1^cre and M14^fl/fl-Adipoq^cre male and female mice exhibited similar body weight trajectories compared to the M14^fl/fl control group (Fig. [116]2a for males, Supplementary Fig. [117]2c for females). Intriguingly, evaluation of glucose homeostasis revealed that M14^fl/fl-Ucp1^cre male and female mice displayed enhanced insulin sensitivity and improved glucose tolerance, while M14^fl/fl-Adipoq^cre mice exhibited diminished insulin sensitivity and similar glucose tolerance as the control M14^fl/fl mice (Fig. [118]2b, [119]c for males, and Supplementary Fig. [120]2d and e for females). Fig. 2. Ablation of Mettl14 in BAT or WAT results in opposite systemic metabolic phenotypes in mice. [121]Fig. 2 [122]Open in a new tab a Body weight trajectories of CD-fed male mice (n = 10/group). b Insulin tolerance test of CD-fed male mice (n = 6 for M14^fl/fl group, n = 6 for M14^fl/fl-Ucp1^cre group, and n = 9 for M14^fl/fl-Adipoq^cre group). c Glucose tolerance test of CD-fed male mice (n = 14 for M14^fl/flgroup, n = 8 for M14^fl/fl-Ucp1^cre group, and n = 9 for M14^fl/fl-Adipoq^cre group). d Body weight trajectories of high-fat diet (HFD)-fed male mice (n = 11 for M14^fl/flgroup, n = 8 for M14^fl/fl-Ucp1^cre group, and n = 11 for M14^fl/fl-Adipoq^cre group). e Insulin tolerance test of HFD-fed male mice (n = 11 for M14^fl/fl group, n = 8 for M14^fl/fl-Ucp1^cre group, and n = 11 for M14^fl/fl-Adipoq^cregroup). f Glucose tolerance test of HFD-fed male mice (n = 18 for M14^fl/fl group, n = 8 for M14^fl/fl-Ucp1^cre group, and n = 14 for M14^fl/fl-Adipoq^cre group). g Fasting glucose levels in the plasma of M14^fl/fl, M14^fl/fl-Ucp1^cre, and M14^fl/fl-Adipoq^cre male mice (n = 19 for M14^fl/fl group, n = 8 for M14^fl/fl-Ucp1^cre group, and n = 16 for M14^fl/fl-Adipoq^cre group). h Fasting insulin levels in the plasma of M14^fl/fl, M14^fl/fl-Ucp1^cre, and M14^fl/fl-Adipoq^cre male mice (n = 15 for M14^fl/fl group, n = 8 for M14^fl/fl-Ucp1^cre group, and n = 13 for M14^fl/fl-Adipoq^cre group). i Fasting adiponectin levels in the plasma of M14^fl/fl, M14^fl/fl-Ucp1^cre, and M14^fl/fl-Adipoq^cre male mice (n = 17 for M14^fl/fl group, n = 8 for M14^fl/fl-Ucp1^cre group, and n = 13 for M14^fl/fl-Adipoq^cregroup). j Representative images of the gross appearance of iBAT, iWAT, and eWAT from M14^fl/fl, M14^fl/fl-Ucp1^cre, and M14^fl/fl-Adipoq^cremale mice on HFD. k–o Representative images of H&E stained iWAT (k), eWAT (I), iBAT (m), liver (n), and muscle (o) from M14^fl/fl, M14^fl/fl-Ucp1^cre, and M14^fl/fl-Adipoq^cremale mice on HFD. All samples in each panel are biologically independent. Data are presented as means ± SEM from two independent experiments by Two-way ANOVA (a–f), and One-way ANOVA (g–i). Also see Supplementary Figs. [123]2 and [124]3. Source data are provided as a Source Data file. Upon challenging the mice with an HFD comprising 60% fat for 16 weeks, M14^fl/fl control mice exhibited the expected body weight gain and demonstrated compromised insulin sensitivity and glucose intolerance when compared to their low-fat diet (LFD) fed counterparts (data not presented). Both M14^fl/fl-Ucp1^cre and M14^fl/fl-Adipoq^cre male mice displayed significantly lower body weights compared to M14^fl/fl mice (Fig. [125]2d). M14^fl/fl-Ucp1^cre female mice showed consistently lower body weight, while M14^fl/fl-Adipoq^cre females had similar body weight when each group was compared to the control females (Supplementary Fig. [126]2f). Consistent with our earlier observations^[127]19, M14^fl/fl-Ucp1^cre mice were shielded from HFD-induced systemic insulin resistance and glucose intolerance (Fig. [128]2e, [129]f for males, Supplementary Fig. [130]2g, h for females). Conversely, M14^fl/fl-Adipoq^cre mice manifested pronounced insulin resistance, while their glucose tolerance was comparable to the controls (Fig. [131]2e, [132]f for males, Supplementary Fig. [133]2g, h for females). Additionally, M14^fl/fl-Ucp1^cre male mice exhibited substantially lower fasting glucose and insulin levels, along with elevated adiponectin levels, whereas M14^fl/fl-Adipoq^cre male mice displayed a different plasma profile characterized by a tendency towards elevated insulin and decreased adiponectin levels (Fig. [134]2g–[135]i). Notably, both KO mouse models exhibited reduced mass in iWAT and eWAT and increased mass in iBAT compared to controls; the latter was especially pronounced in M14^fl/fl-Adipoq^cre mice (Fig. [136]2j for males, and Supplementary Fig. [137]2i for females). We next examined the characteristics of the different fat mass depots and changes in the liver and skeletal muscle. Hematoxylin and eosin (H&E) staining revealed diminished adipocyte size in both iWAT and eWAT of both knockout mouse models (Fig. [138]2k, [139]l for males, Supplementary Fig. [140]2j, [141]k for females). Consistent with prior observations, M14^fl/fl-Ucp1^cre mice were protected from HFD-induced immune cell infiltration in eWAT, hypertrophy of brown adipocytes, and hepatic lipid accumulation (Fig. [142]2k–[143]n for males, Supplementary Fig. [144]2j–[145]n for females). In contrast, M14^fl/fl-Adipoq^cre mice displayed heightened immune cell infiltration in both iWAT and eWAT, brown adipocyte hypertrophy, and liver lipid accumulation (Fig. [146]2k–[147]n for males, Supplementary Fig. [148]2j–[149]n for females). Strikingly, while both M14^fl/fl M14^fl/fl-Ucp1^cre mice displayed normal skeletal muscle morphology, we observed the presence of both inter- and intra-cellular lipid accumulation in the muscle from M14^fl/fl-Adipoq^cre male mice indicating ectopic lipid deposition (Fig. [150]2o). Thus, manipulation of the expression of Mettl14 leads to contrasting regulatory roles in terms of insulin sensitivity and systemic metabolism in Ucp1^cre- and Adipoq^cre-mediated knockout mouse models. We next considered that Mettl14 was depleted in both WAT and BAT in the M14^fl/fl-Adipoq^cre mice (Supplementary Fig. [151]2a). To specifically investigate the role of Mettl14 in WAT functionality and overall metabolic control that is independent of iBAT, we surgically removed iBAT from M14^fl/fl and M14^fl/fl-Adipoq^cre mice (Supplementary Fig. [152]3a). Comparing the iBAT-depleted M14^fl/fl-Adipoq^cre mice to iBAT-depleted M14^fl/fl mice fed regular CD, revealed that the former persisted in exhibiting insulin resistance (Supplementary Fig. [153]2b). Of note, iBAT removal exacerbated insulin resistance observed in M14^fl/fl-Adipoq^cre mice as compared to iBAT-depleted M14^fl/fl mice (Supplementary Fig. [154]2b). Additionally, it was noted that in certain M14^fl/fl-Adipoq^cre mice following iBAT removal, there was a conspicuous absence of eWAT and pronounced atrophy in their iWAT, whereas iBAT-depleted M14^fl/fl mice exhibited normal gross appearance in their WAT depots (Supplementary Fig. [155]2c). These observations suggest that while WAT likely plays a dominant role in the phenotype of M14^fl/fl-Adipoq^cre mice, brown adipose tissues, including iBAT, may contribute positively, potentially acting as a metabolic sink for excess triglycerides or FFAs released from WAT. The specific contribution of other BAT depots in addition to iBAT in this context requires further investigation. Ablation of Mettl14 in BAT or WAT differentially impacts insulin sensitivity in distinct peripheral metabolic tissues Next, we examined tissue-autonomous and endocrine effects of Mettl14 ablation on whole-body insulin sensitivity by injecting mice with insulin via the vena cava and assessing changes in insulin signaling proteins (pIRβ/IGF1Rβ and pAKT[S473]) in target metabolic tissues (Fig. [156]3a). In control mice, HFD decreased insulin-stimulated (1 U/mouse) pIRβ/IGF1Rβ and pAKT[S473] in iWAT and eWAT compared to LFD (Fig. [157]3b, [158]c). Insulin resistance induced by the HFD was mitigated in M14^fl/fl-Ucp1^cre mice, evident in both iWAT and eWAT (Fig. [159]3b, [160]c). However, the insulin response was notably blunted in M14^fl/fl-Adipoq^cre mice in both iWAT and eWAT (Fig. [161]3b, [162]c). Fig. 3. Ablation of Mettl14 in BAT or WAT differentially impacts insulin sensitivity of peripheral metabolic tissues. [163]Fig. 3 [164]Open in a new tab a Diagram illustrating the timeline for collecting metabolic tissues following saline/insulin injection into the vena cava of M14^fl/fl, M14^fl/fl-Ucp1^cre, and M14^fl/fl-Adipoq^cre male mice fed either a low-fat diet (LFD) or a high-fat diet (HFD) (n = 6/group) as indicated (Created with BioRender.com). b–f Western blot analysis and quantification of insulin-stimulated phosphorylation of IRβ/IGF1Rβ and AKT[S473] in iWAT (b), eWAT (c), iBAT (d), liver (e), and muscle (f) after injection of 1U insulin into the vena cava (n = 6/groups). g Western blot analysis of METTL14 protein abundance in sgNTC-, sgM14#1-, sgM14#2-, sgM14#3-hBA preadipocytes. h, i Western blot analysis (h) and quantification (i) of insulin-stimulated phosphorylation of IRβ/IGF1Rβ and AKT[S473] in differentiated sgNTC-, sgM14#1-, sgM14#2-, sgM14#3-hBA cells (n = 6/group). j Glucose uptake of differentiated sgNTC-, sgM14#1-, sgM14#2-, sgM14#3-hBA cells treated with or without insulin stimulation (n = 4/group). k Western blot analysis of METTL14 protein abundance in sgNTC-, sgM14#1-, sgM14#2-, sgM14#3-hWA preadipocytes. l, m Western blot analysis (l) and quantification (m) of insulin-stimulated phosphorylation of IRβ/IGF1Rβ and AKT[S473] in differentiated sgNTC-, sgM14#1-, sgM14#2-, sgM14#3-hWA cells (n = 6/group). n Glucose uptake of differentiated sgNTC-, sgM14#1-, sgM14#2-, sgM14#3-hWA cells treated with or without insulin stimulation (n = 4/group). All samples in each panel are biologically independent. Data are presented as means ± SEM from two or three independent experiments by Two-way ANOVA (b–f, i, j, m, n). Also see Supplementary Fig. [165]4. Source data are provided as a Source Data file. Furthermore, M14^fl/fl-Ucp1^cre mice on HFD exhibited elevated levels of pIRβ/IGF1Rβ and pAKT[S473] in iBAT, liver, and skeletal muscle tissues (Fig. [166]3d–[167]f). Conversely, M14^fl/fl-Adipoq^cre mice displayed reduced insulin signaling in their liver and skeletal muscle, excluding iBAT (Fig. [168]3d–[169]f). These findings highlight the distinct effects of Mettl14 ablation in different adipose tissue depots and underscore its role in modulating insulin sensitivity in diverse metabolic tissues. To investigate the translational significance of our findings in murine models, we capitalized on immortalized human brown and white adipocytes and established stable METTL14-knockout cell line colonies in hBAs and hWAs utilizing a single guide RNA targeting METTL14 (sgM14) (Fig. [170]3g, [171]k, respectively). Notably, the ablation of METTL14 yielded contrasting effects on insulin sensitivity and glucose uptake capacity in fully differentiated hBAs (Fig. [172]3h–[173]j) versus hWAs (Fig. [174]3l–[175]n). These observations support the notion that METTL14 regulates insulin sensitivity in a context- and cell-type-dependent manner. Mettl14 deficiency in BAT or WAT differentially impacts food intake and energy expenditure in mice Next, we investigated whether METTL14 differentially modulates BAT- or WAT-mediated whole-body energy balance. Mettl14 deficiency utilizing Ucp1^cre exhibited an enhanced cold tolerance (Fig. [176]4a) without significant alterations in food intake (Fig. [177]4b), oxygen consumption (Fig. [178]4c), or energy expenditure (Fig. [179]4d). Conversely, M14^KO driven by Adipoq^cre resulted in a significant reduction in thermogenic capacity (Fig. [180]4a), food intake (Fig. [181]4b), oxygen consumption (Fig. [182]4e), and energy expenditure (Fig. [183]4f). Similarly, at the cellular level, METTL14 deficiency showed no significant impact on the basal (Fig. [184]4g–[185]j) or β3-adrenergic agonist-stimulated (Figs. [186]4k–[187]n) mitochondrial and glycolytic functions of hBAs, while, the absence of METTL14 in hWAs negatively regulated these functions (Fig. [188]4o–[189]r). Previous studies have reported that METTL3 positively regulates thermogenesis in both BAT-specific (Ucp1^cre driven-knockout) and beige-specific (Adipoq^cre-mediated knockout) mouse models^[190]15,[191]17. Our findings highlight the intricate and diverse roles of the m^6A writer protein, METTL14, in the regulation of thermogenesis and metabolic control in adipose tissues, and unlike METTL3, METTL14 can regulate the thermogenesis of adipose tissues in a cell-specific manner. Fig. 4. Mettl14 Deficiency Differentially Regulates Thermogenic Capacity of BAT and WAT. [192]Fig. 4 [193]Open in a new tab a Core body temperature of M14^fl/fl, M14^fl/fl-Ucp1^cre, and M14^fl/fl-Adipoq^cre mice after acute cold exposure (n = 15 for M14^fl/fl group, n = 8 for M14^fl/fl-Ucp1^cre group, and n = 6 for M14^fl/fl-Adipoq^cre group). b Daily food consumption measured by indirect calorimetry (n = 11 for M14^fl/fl group, n = 5 for M14^fl/fl-Ucp1^cre group, and n = 11 for M14^fl/fl-Adipoq^cre group). c, d Oxygen consumption (c) and energy expenditure (d) of M14^fl/fl-Ucp1^cre mice versus M14^fl/fl mice (n = 6 for M14^fl/fl group, n = 5 for M14^fl/fl-Ucp1^cre group). e, f Oxygen consumption (e) and energy expenditure (f) of M14^fl/fl-Adipoq^cre mice versus M14^fl/fl mice (n = 11/group). g–j Seahorse analysis of cellular oxygen consumption rate (OCR) (g, h, n = 5/group) and extracellular acidification rate (ECAR) (i, j, n = 5 for sgNTC, and n = 10 for sgM14) in differentiated sgNTC- and sgM14-hBA cells. k–n Seahorse analysis of cellular oxygen consumption rate (OCR) (k, l, n = 5/group) and extracellular acidification rate (ECAR) (m, n, n = 4 for sgNTC, and n = 5 for sgM14) in differentiated sgNTC- and sgM14-hBA cells stimulated with 10 µM CL-316,243 for 5 h before seahorse assay. o–r Seahorse analysis of cellular oxygen consumption rate (OCR) (o, p, n = 9 for sgNTC, and n = 10 for sgM14) and extracellular acidification rate (ECAR) (q, r, n = 5/group) in differentiated sgNTC- and sgM14-hWA cells (Data are representative of 2 independent experiments). All samples in each panel are biologically independent. Data are presented as means ± SEM from two or three independent experiments by Two-way ANOVA with Dunnett’s multiple comparisons test (a, b), Two-way ANOVA (c–g, i, k, m, o, q), and Two-tailed unpaired t-tests (h, j, l, n, p, r). Source data are provided as a Source Data file. Mettl14 Deficiency Differentially Impacts m^6A Methylome and Transcriptome in Mouse iBAT and iWAT The distinct roles of METTL14 in BAT- versus WAT-mediated whole-body metabolism prompted the hypothesis that METTL14 differentially modulates the transcriptional profiles of BAT and WAT by selectively methylating specific target transcripts. To test this hypothesis, we conducted m^6A-MeRIP-seq and RNA-seq analyses on iBAT from M14^fl/fl and M14^fl/fl-Ucp1^cre mice, as well as iWAT from M14^fl/fl and M14^fl/fl-Adipoq^cre mice. First, a comparison of the m^6A methylome between M14^fl/fl-Ucp1^cre-iBAT versus M14^fl/fl-iBAT, and between M14^fl/fl-Adipoq^cre-iWAT versus M14^fl/fl-iWAT (comparison approach shown in Fig. [194]5a) revealed 2465 and 2838 m^6A-hypomethylated genes (Fig. [195]5b), as well as 4854 and 8007 hypermethylated genes by Mettl14 deficiency in iBAT and iWAT respectively (Fig. [196]5c). Mettl14 deficiency resulted in 1375 and 1748 m^6A-hypermethylated genes (Fig. [197]5b), as well as 2010 and 5163 m^6A-hypermethylated genes exclusively in iBAT and iWAT, respectively (Fig. [198]5b). Next, the intersection of the differentially m^6A-modified genes in M14^fl/fl-Ucp1^cre-iBAT and M14^fl/fl-Adipoq^cre-iWAT revealed 1090 (25.9%) and 2844 (28.4%) commonly hypomethylated and hypermethylated genes compared to M14^fl/fl mice, respectively (Fig. [199]5b, [200]c). Fig. 5. Mettl14 Deficiency Results in Different m^6A Methylome and Transcriptome of Mouse iBAT and iWAT. [201]Fig. 5 [202]Open in a new tab a Schematic diagram of comparison approach for analyzing RNA-seq and m^6A-seq data derived from iWAT in M14^fl/fl and M14^fl/fl-Adipoq^cre mice, and from iBAT in M14 ^fl/fl and M14^fl/fl-Ucp1^cre mice (Created with BioRender.com). b Venn diagram representation of the exclusively m^6A-hypomethylated genes in M14^fl/fl-Ucp1^cre-iBAT versus M14^fl/fl-iBAT, commonly m^6A-hypomethylated genes, and exclusively m^6A-hypomethylated genes in M14^fl/fl-Adipoq^cre-iWAT versus M14^fl/fl-iWAT, statistical analyses were performed using the Benjamin-Hochberg procedure and genes were filtered for p < 0.01. c Venn diagram representation of the exclusively m^6A-hypermethylated genes in M14^fl/fl-Ucp1^cre-iBAT versus M14^fl/fl-iBAT, commonly m^6A-hypermethylated genes, and exclusively m^6A-hypermethylated genes in M14^fl/fl-Adipoq^cre-iWAT versus M14^fl/fl-iWAT, statistical analyses were performed using the Benjamin-Hochberg procedure and genes were filtered for p < 0.01. d Venn diagram representation of the exclusively upregulated genes in M14^fl/fl-Ucp1^cre-iBAT versus M14^fl/fl-iBAT, commonly upregulated genes, and exclusively upregulated genes in M14^fl/fl-Adipoq^cre-iWAT versus M14^fl/fl-iWAT, statistical analyses were performed using the Benjamin-Hochberg procedure and genes were filtered for FDR < 0.05. e Venn diagram representation of the exclusively downregulated genes in M14^fl/fl-Ucp1^cre-iBAT versus M14^fl/fl-iBAT, commonly downregulated genes, and exclusively downregulated genes in M14^fl/fl-Adipoq^cre-iWAT versus M14^fl/fl-iWAT, statistical analyses were performed using the Benjamin-Hochberg procedure and genes were filtered for FDR < 0.05. f, g Enriched pathways of the upregulated (f) or downregulated g genes in M14^fl/fl-Ucp1^cre-iBAT versus M14^fl/fl-iBAT. h, i Enriched pathways of the upregulated (h) or downregulated (i) genes in M14^fl/fl-Adipoq^cre-iWAT versus M14^fl/fl-iWAT. j Venn diagram representation of the m^6A-hypomethylated (p < 0.01) and upregulated genes (FDR < 0.05) in M14^fl/fl-Ucp1^cre-iBAT versus M14^fl/fl-iBAT. k Top 10 enriched pathways of the m^6A-hypomethylated and differentially upregulated genes in M14^fl/fl-Ucp1^cre-iBAT versus M14^fl/fl-iBAT. l Venn diagram representation of the m^6A-hypomethylated (p < 0.01) and upregulated genes (FDR < 0.05) in M14^fl/fl-Adipoq^cre-iWAT versus M14^fl/fl-iWAT. m Top 10 enriched pathways of the m^6A-hypomethylated and differentially upregulated genes in M14^fl/fl-Adipoq^cre-iWAT versus M14^fl/fl-iWAT. p-values of pathway enrichment analysis were calculated according to the hypergeometric test based on the number of physical entities present in both the pre-defined set and user-specified list of physical entities (f–i, m). Source data are provided as a Source Data file. Similar comparisons were performed between the differentially expressed genes in M14^fl/fl-Ucp1^cre-iBAT and M14^fl/fl-Adipoq^cre-iWAT (Fig. [203]5a). Only 106 genes (2.8% of total upregulated genes) exhibited overlap between M14^fl/fl-Ucp1^cre iBAT and M14^fl/fl-Adipoq^cre iWAT (Fig. [204]5d), and 91 (1.8%) genes were commonly downregulated (Fig. [205]5e). Gene set enrichment analysis revealed significant divergence in the transcriptional programs of METTL14 between BAT and WAT (Fig. [206]5f–[207]i). Upregulated genes of M14^fl/fl-Ucp1^cre iBAT were enriched in pathways related to lipid metabolism, prostaglandin synthesis, and phospholipid biosynthetic processes (Fig. [208]5f), while downregulated genes were associated with insulin resistance, white fat cell differentiation, and apoptosis pathways (Fig. [209]5g). In contrast, M14^fl/fl-Adipoq^cre iWAT showed upregulation of pathways involving immune response, TNFα signaling, and apoptosis (Fig. [210]5h), with downregulated genes enriched in lipid metabolism, lipolysis, insulin signaling, and phospholipid metabolism pathways (Fig. [211]5i). Considering that METTL14 is responsible for installing m^6A, the depletion of METTL14 is expected to result in the hypomethylation of its target transcripts. Moreover, it is established that reduced m^6A methylation correlates with enhanced mRNA stability, as it protects the target transcripts from degradation^[212]19,[213]21,[214]24. To clarify if these transcriptomic changes were governed by METTL14-mediated m^6A modification, we intersected m^6A-hypomethylated and differentially upregulated genes. These analyses confirmed that lipid and phospholipid-related pathways were m^6A-dependently upregulated in M14^fl/fl-Ucp1^cre iBAT (Fig. [215]5j, [216]k), while apoptosis and TNFα signaling pathways were upregulated in an m^6A-dependent manner in M14^fl/fl-Adipoq^cre iWAT (Fig. [217]5l, [218]m). METTL14 selectively methylates key target transcripts in hBAs versus hWAs Given the marked cellular heterogeneity of BAT and WAT tissues^[219]25,[220]26, we harnessed hBA and hWA models to delve into the cellular-level transcriptional mechanism(s) influenced by METTL14. Using the same comparison strategy described above, the intersection of m^6A hypomethylated genes in sgM14-hBAs and sgM14-hWA cells revealed only 160 (6.3%) commonly hypomethylated and 912 (14.2%) hypermethylated genes, along with 559 (11.8%) commonly upregulated and 554 (11.9%) commonly downregulated genes (Fig. [221]6a–[222]d). Fig. 6. METTL14 Selectively Methylates Key Target Transcripts in hBA versus hWA cells. [223]Fig. 6 [224]Open in a new tab a Venn diagram representation of the exclusively m^6A-hypomethylated genes in sgM14-hBA versus sgNTC-hBA cells, commonly m^6A-hypomethylated genes, and exclusively m^6A-hypomethylated genes in sgM14-hWA versus sgNTC-hWA cells, statistical analyses were performed using the Benjamin-Hochberg procedure and genes were filtered for p < 0.01. b Venn diagram representation of the exclusively m^6A-hypermethylated genes in sgM14-hBA versus sgNTC-hBA cells, commonly m^6A-hypermethylated genes, and exclusively m^6A-hypermethylated genes in sgM14-hWA versus sgNTC-hWA cells, statistical analyses were performed using the Benjamin-Hochberg procedure and genes were filtered for p < 0.01. c Venn diagram representation of the exclusively upregulated genes in sgM14-hBA versus sgNTC-hBA cells, commonly upregulated genes, and exclusively upregulated genes in sgM14-hWA versus sgNTC-hWA cells, statistical analyses were performed using the Benjamin-Hochberg procedure and genes were filtered for FDR < 0.01. d Venn diagram representation of the exclusively downregulated genes in sgM14-hBA versus sgNTC-hBA cells, commonly downregulated genes, and exclusively downregulated genes in sgM14-hWA versus sgNTC-hWA cells, statistical analyses were performed using the Benjamin-Hochberg procedure and genes were filtered for FDR < 0.01. e, f Enriched pathways of the upregulated (e) or downregulated (f) genes in sgM14-hBA versus sgNTC-hBA cells. g, h Enriched pathways of the upregulated (g) or downregulated (h) genes in sgM14-hWAs versus sgNTC-hWA cells. i Venn diagram representation of the m^6A-hypomethylated and upregulated genes in sgM14-hWA versus sgNTC-hWA cells, statistical analyses were performed using the Benjamin-Hochberg procedure and genes were filtered for p < 0.01, and FDR < 0.05, respectively. j Top 10 enriched pathways of the m^6A-hypomethylated and differentially upregulated genes in sgM14-hWA versus sgNTC-hWA cells. k Schematic depicting experimental strategy to knockdown METTL14 in the differentiated hWA cells for m^6A- and RNA-seq (Created with BioRender.com). l Venn diagram representation of the intersection of the m^6A-hypomethylated and upregulated genes in siM14-hWA versus siNTC-hWA cells. Wildtype hWA cells were fully differentiated and transfected with siNTC or siM14 to knock down METTL14 in the mature hWA cells. Statistical analyses were performed using the Benjamin-Hochberg procedure and genes were filtered for p < 0.01 and FDR < 0.05, respectively. m Top 10 enriched pathways of the m^6A-hypomethylated and upregulated genes in siM14-hWA versus siNTC-hWA cells. n Coverage plots of m^6A peaks in TRAIL (I), TNFSF1R (m), RIP1K (n), CASP7 (o), DFFA (p) genes in sgM14-hWA versus sgNTC-hWA cells. Plotted coverages are the median of the n replicates presented. o, p Heatmaps of genes related to TNFα and apoptosis signaling pathways (o) and prostaglandins synthesis and regulation pathway (p) (filtered by FDR < 0.05) in sgM14-hBAcells versus sgNTC-hBA cells, or sgM14-hWA cells versus sgNTC-hWA cells. q A proposed model for the molecular mechanism of action that METTL14-mediated m^6A installation selectively destabilizes transcripts in brown (left panel) and white adipocytes (right panel). p-values of pathway enrichment analysis were calculated according to the hypergeometric test based on the number of physical entities present in both the pre-defined set and user-specified list of physical entities (e–h, j, m). Source data are provided as a Source Data file. In hBA cells, enrichment analyses of the DGE (differentially gene expression) pointed to elevated lipid metabolism and phospholipid processes among upregulated pathways, and apoptosis and TNFα signaling among the prominent downregulated pathways due to METTL14 deficiency (Fig. [225]6e, [226]f). Conversely, in hWA cells, METTL14 deficiency predominantly led to m^6A-dependent upregulation of TNFα and apoptosis pathways (Fig. [227]6g, i, [228]j), while resulting in the downregulation of pathways regulated by EGFR1, focal adhension-PK3K-AKT-mTOR, and insulin signaling (Fig. [229]6h). Importantly, while the stable knockout cell line enables the conduct of several phenotypical and mechanistic studies, the ablation of METTL14 in preadipocytes may also influence the differentiation process of both hWAs and hBAs. To address this potential confounding factor in adipocyte differentiation, we performed a transient knockdown of METTL14 using siRNA in fully differentiated adipocytes, (Fig. [230]6k). Consistently, METTL14 deficiency in mature hWA cells accentuated the significance of TNFα and cell death signaling pathways (Fig. [231]6l, [232]m). These findings emphasize the ability of METTL14 to exhibit divergent roles in shaping the transcriptional profiles of mature brown adipocytes to promote a favorable metabolic phenotype. Conversely, in the M14^fl/fl-Adipoq^cre model, METTL14 deficiency distinctly upregulated genes linked to TNFα and apoptosis in mature white adipocytes, contributing to adipose tissue apoptosis and insulin resistance. Thus, at both tissue and cellular levels, METTL14 deficiency leads to an upregulation of genes encoding prostaglandin synthases in BAT, and key genes related to TNFα signaling and apoptosis in WAT. As mentioned above, METTL14 deficiency is expected to induce hypomethylation in its target transcripts. Next, we conducted further analysis to explore the m^6A-hypomethylated and upregulated genes, which are considered METTL14-targeted transcripts, within the most enriched pathways of sgM14-hBAs cells and sgM14-hWA cells. Within hBA cells, METTL14 specifically destabilizes transcripts associated with prostaglandin synthesis^[233]19. In this study, we identified five transcripts—necrosis factor-related apoptosis-inducing ligand (TRAIL), TNF receptor superfamily member 1 A (TNFR1), receptor-interacting serine/threonine kinase 1 (RIP), caspase 7 (CASP7), and DNA fragmentation factor subunit alpha (DFFA), as direct targets of METTL14 in hWA cells (Fig. [234]6n). Interestingly, while METTL14 deficiency led to the upregulation of several genes within the TNFα and apoptosis pathways in hWA cells, most of these genes exhibited downregulation in sgM14-hBA cells (Fig. [235]6o). Notably, transcripts previously identified as METTL14 targets in hBA, including PTGES2, CRB1, and PGC1A, were downregulated in sgM14-hWA compared to sgNTC-hWA cells (Fig. [236]6p). These findings provide strong evidence that METTL14 regulates distinct transcriptional processes in BAT versus WAT, as summarized in Fig. [237]6q. METTL14 deficiency promotes apoptosis in mouse and human white adipocytes We next sought to validate our sequencing outcome and investigate the molecular mechanism(s) underlying the METTL14-mediated m^6A decoration of target transcripts in brown versus white adipocytes/adipose tissues. METTL14 deficiency promotes prostaglandin synthesis in brown adipocytes, as reported in our previous study^[238]19. To validate our sequencing outcome from mouse and human white adipocytes, which indicate increased apoptosis (Figs. [239]5 and [240]6), we conducted Terminal deoxynucleotidyl Transferase dUTP Nick End Labeling (TUNEL) assays on the iWAT and eWAT isolated from M14^fl/fl-Adipoq^cre mice. Our data confirmed a higher percentage of apoptotic cells in both depots (Fig. [241]7a–[242]c). Notably, apoptosis was primarily observed in PERILIPIN-positive cells (Fig. [243]7a), suggesting adipocyte-specific apoptosis induced by METTL14 deficiency. Fig. 7. METTL14 deficiency promotes apoptosis in mouse and human white adipocytes. [244]Fig. 7 [245]Open in a new tab a Representative pictures of immunofluorescence staining of apoptotic nuclei (TUNEL-stain, green), adipocytes (PERILIPIN-stain, red), and nuclei (DAPI, blue) in eWAT and iWAT sections collected from male M14^fl/fl and M14^fl/fl-Adipoq^cre mice fed with HFD (n = 7/group) (scale bar, 50 μm; insert, 2× magnification). b, c Quantification of TUNEL-positive nuclei as a percentage of total nuclei in eWAT (b) and iWAT (c) of male M14^fl/fl and M14^fl/fl-Adipoq^cre mice fed with HFD (n = 7/group). d qRT-PCR analysis of the indicated mRNAs in the iWAT of male M14^fl/fl and M14^fl/fl-Adipoq^cre mice (n = 10/group). mRNA levels were normalized to Actb. e, f Western blot analysis (e) and quantification (f) of the indicated proteins in the iWAT of male M14^fl/fl and M14^fl/fl-Adipoq^cre mice. VINCULIN was used as a loading control (n = 5/group). g, h ELISA analysis of free fatty acid (FFA) (g, n = 10 for M14^fl/fl, and n = 11 for M14^fl/fl-Adipoq^cre) and triglyceride (TG) (h, n = 10/group) levels in the serum of male M14^fl/fl and M14^fl/fl-Adipoq^cre mice on HFD. i ELISA analysis of triglyceride (TG) levels in the liver and muscle of male M14^fl/fl and M14^fl/fl-Adipoq^cre mice on HFD (left panels) and iBAT removed cohort on CD (right panels) (n = 6/group for liver and muscle from HFD-fed mice, and liver from iBAT removed mice; n = 5 for muscle from iBAT removed M14^fl/fl mice, and n = 6 for iBAT from iBAT removed M14^fl/fl-Adipoq^cre mice). j qRT-PCR analysis of the indicated mRNAs in the differentiated sgNTC-hWA and sgM14-hWA cells. mRNA levels were normalized to ACTINB mRNA (n = 4/group). k Western blot analysis of the indicated proteins in the differentiated sgNTC-hWA and sgM14-hWA cells. TUBULIN was used as a loading control (n = 3/group). l ELISA analysis of FFA (upper panels, n = 5 for sgNTC, and n = 4 for sgM14) and TG (lower panels, n = 7 for sgNTC, and n = 5 for sgM14) levels in the culture medium of differentiated sgNTC-hWAs and sgM14-hWA cells. m A proposed model illustrating the mechanism of systemic insulin resistance induced by WAT-apoptosis in the M14^fl/fl-Adipoq^cre mouse model (Created with BioRender.com). All samples in each panel are biologically independent. Data are presented as means ± SEM from two or three independent experiments by Two-tailed unpaired t-tests (b–d, f–i, j, l). Source data are provided as a Source Data file. In mouse iBAT and differentiated hBAs, METTL14-mediated m^6A installation promotes the decay of transcripts related to prostaglandin synthesis in a YTHDF2/3-dependent manner^[246]19. In iWAT, Mettl14 deficiency leads to upregulation of Trail, Dr5, Tnfα, Tnfr1, Casp3, Casp7, and Dffa at the mRNA level (Fig. [247]7d), and increased protein abundance of TRAIL and TNFA (Fig. [248]7e, [249]f). Considering the ectopic fat deposition in iBAT, liver, and muscle (Fig. [250]1m–[251]o), we wondered whether apoptosis of white adipocytes increases circulating free fatty acids (FFAs) and/or triglyceride (TG) levels, leading to iBAT hypertrophy, hepatic steatosis and/or muscle lipid accumulation. Since Mettl14 deficiency downregulated genes associated with fatty acid degradation (also referred to as lipolysis) and regulation of lipolysis in adipocyte pathways (Fig. [252]5h), it is plausible that lipolysis-induced FFA production does not contribute to ectopic lipid accumulation. Indeed, FFA levels (Fig. [253]7g) were lower while TG levels (Fig. [254]7h) were elevated in the serum of M14^fl/fl-Adipoq^cre mice compared to control mice. Moreover, the TG level was higher in the liver of HFD-fed and CD-fed iBAT-deficient M14^fl/fl-Adipoq^cre mice; as well as higher in the liver of HFD-fed Adipoq^cre mice (Fig. [255]7i). Consistent results were obtained at the cellular level, as indicated by the upregulated gene expression and protein abundance of apoptotic markers (Fig. [256]7j, [257]k, respectively), decreased levels of FFAs, and heightened TGs in the conditioned media of differentiated sgM14-hWA cells (Fig. [258]7l). Of note, the levels of both cleaved caspase 3 (cCASP3) and caspase 7 (cCASP7) were significantly increased in the sgM14-hWA cell line (Fig. [259]7k), providing further support for adipocyte-specific apoptosis. These data collectively indicate that M14^KO-WAT apoptosis-induced lipid spillover contributes to the observed ectopic lipid accumulation and consequent peripheral insulin resistance (Fig. [260]7m). METTL14-Mediated m^6A accelerates the decay of apoptosis-related transcripts in A YTHDF2/3-dependent manner in white adipocytes Next, we conducted mechanistic studies using differentiated hWA cells. First, we examined the effects of METTL14 deficiency on the stability of m^6A-targeted mRNAs (identified in Fig. [261]6n) following treatment with transcription inhibitor actinomycin D (ActD). METTL14 deficiency suppressed the degradation of TRAIL and TNFR1 mRNAs without significantly affecting the stability of ACTINB mRNA (Fig. [262]8a). These findings suggest that METTL14-mediated m^6A modification orchestrates hWA apoptosis by regulating the stability of key apoptotic transcripts. Fig. 8. METTL14-mediated m^6A promotes decay of TNFR1 and TRAIL in a YTHDF2/3 dependent manner in white adipocytes. [263]Fig. 8 [264]Open in a new tab a qRT-PCR analysis of the TRAIL and TNFR1 mRNA in differentiated sgNTC-hWA and sgM14-hWA cells after a time-course treatment with 100 μg/mL Actinomycin D (Act D). mRNA levels were normalized to ACTINB mRNA (n = 3). b Representative Western blot of YTHDF1, YTHDF2, and YTHDF3 protein levels in differentiated wildtype hWA cells transfected with siNTC, siYTHDF1, siYTHDF2, or siYTHDF3 siRNA. GAPDH was used as a loading control (n = 3). c qRT-PCR analysis of TRAIL and TNFR1 mRNA in differentiated wildtype hWA cells transfected with siNTC, siYTHDF1, siYTHDF2, or siYTHDF3 siRNA following a time-course treatment 100 μg/mL Act D. TRAIL and TNFR1 mRNA levels were normalized to ACTIN (n = 4). The p-value indicates the significance of differences between any group and the siNTC group. d, e Western blot analysis (d) and quantification (e) of METTL14 protein abundance in the differentiated hWA cells transfected with scramble, wild-type METTL14 (METTL14-WT), or mutated METTL14 at R298 (METTL14-MUT) plasmid (n = 3/group). f Relative m^6A levels in the differentiated hWA cells transfected with scramble, wild-type METTL14-WT, or METTL14-MUT plasmid (n = 3/group). g qRT-PCR analysis of TRAIL and TNFR1 mRNA in the differentiated hWA cells transfected with scramble, wild-type METTL14-WT, or METTL14-MUT plasmid after a time-course treatment with 100 μg/mL Actinomycin D (Act D). TRAIL and TNFR1 mRNA levels were normalized to ACTINB mRNA (n = 3). The p-value indicates the significance of differences between any group and the Scr group. h Scheme of experimental approach depicting M14^fl/fl mice receiving AAV8 eGFP (AAV-scramble), M14^fl/fl-Adipoq^cre mice receiving AAV8 eGFP, M14^fl/fl-Adipoq^cre mice receiving AAV8 knocking down Tnfr1 (AAV-shTnfr1), and M14^fl/fl-Adipoq^cre mice receiving AAV8 knocking down Trail (AAV-shTrail) (Created with BioRender.com). i, j Representative Western blot analysis (i) and quantification (j) of METTL14, TNFR1, and TRAIL protein levels in iWAT of the AAV-injected mice (n = 3). k Serum TG levels measured by ELISA assays (n = 5–6/group). l Intraperitoneal insulin tolerance tests of mice injected with AAVs (n = 7 for M14^fl/fl-AAV-Scr group; n = 5 for M14^fl/fl-Adipoq^cre_AAV-Scr group, n = 7 for M14^fl/fl-Adipoq^cre_AAV-shTnfr1 group, n = 8 for M14^fl/fl-Adipoq^cre_AAV-shTrail group,). The p-value indicates the significance of differences between any group and the M14^fl/fl-Adipoq^cre_AAV-Scr group. m, n Western blot analysis (m) and quantification (n) of pIRβ/IGF1Rβ and pAKT[S473] in iWAT 5-week post-AAV injections, followed by injection of 1U of insulin via the vena cava (n = 2 for non-stimulated groups; n = 4 for insulin-stimulated groups). The p-value indicates the significance of differences between any group and the M14^fl/fl-Adipoq^cre_AAV-Scr group. All samples in each panel are biologically independent. Data are presented as means ± SEM from two or three independent experiments by Two-tailed unpaired t-test (a), One-way ANOVA (c, g), and Two-way ANOVA (e, f, j, k, l, n). Source data are provided as a Source Data file. To further confirm this hypothesis, we performed independent siRNA-mediated knockdown of the m^6A reader proteins YTHDF1, YTHDF2, or YTHDF3 (Fig. [265]7i), which are known to regulate mRNA decay^[266]24,[267]27, and examined the expression and stability of TRAIL and TNFR1 mRNAs after ActD treatment in wild type hWA cells. Knockdown of YTHDF1, YTHDF2, or YTHDF3 (Fig. [268]8b) followed by ActD treatment significantly increased the stability of TRAIL and TNFR1 (Fig. [269]8c). These data collectively confirmed that METTL14-mediated m^6A installation negatively regulates the stability of TRAIL and TNFR1 mRNAs in white adipocytes. Next, to substantiate the implication that METTL14 is linked to the modulation of mRNA stability of apoptosis-associated genes, we explored this possibility with a gain-of-function model by overexpressing METTL14 in hWA cells (Fig. [270]8d, [271]e). Additionally, to investigate whether METTL14 regulates key target mRNA stability in an m^6A-dependent manner, we established a hWA cell line harboring the METTL14 R298 mutation (Fig. [272]8d, [273]e). As expected, overexpression of METTL14 increased global m^6A levels, whereas mutated METTL14 had no impact (Fig. [274]8f). Importantly, while METTL14 upregulation led to decreased stability of TRAIL and TNFR1 through m^6A hypermethylation, their stability was not affected by mutated METTL14 in hWA cells (Fig. [275]8g). These findings support that METTL14 directly regulates TRAIL and TNFR1 in an m^6A-dependent manner. Finally, to validate our identification of TRAIL and TNFR1 as drivers of systemic insulin resistance in M14^fl/fl-Adipoq^cre mice, we used AAV8 vectors to genetically knock down Trail or Tnfr1 specifically in iWAT by injecting AAV8-scramble, AAV8-shTrail, or AAV8-shTnfr1 (Fig. [276]8h). Knockdown efficiency was confirmed by reduced TRAIL or TNFR1 protein levels in iWAT (Fig. [277]8i, [278]j). Notably, knockdown of each gene significantly decreased serum triglyceride (TG) levels in M14^fl/fl-Adipoq^cre mice (Fig. [279]8k), affirming that TRAIL and TNFR1 triggered adipocyte apoptosis and subsequent lipid spillover. Additionally, depletion of either Trail or Tnfr1 partially reversed insulin resistance in M14^fl/fl-Adipoq^cre mice at both the systemic and tissue levels (Fig. [280]8l–[281]n, respectively). Discussion An accumulating body of evidence points to the importance of m^6A mRNA modifications in orchestrating pathways that underlie intricate mechanisms that contribute to metabolic disorders^[282]28. However, the current gap in our understanding pertains to the cell-type-specific regulation by m^6A within various biological domains, including metabolism. Here, we provide one example of the divergent roles of METTL14-mediated m^6A modifications in orchestrating the metabolic functions of brown versus white adipocytes. By integrating m^6A-seq and RNA-seq obtained from both mouse BAT/WAT and human brown/white adipocytes, we unveil cell-type specific m^6A methylomes and transcriptomes. Our study identifies specific transcripts through which METTL14 exerts dual regulatory roles: on one hand, negatively modulating the secretory functions of BAT while on the other simultaneously enhancing the lipid sequestration capabilities of WAT. This multifaceted role of METTL14 in shaping adipocyte-mediated metabolism adds a new layer of nuance to the field of metabolic regulation. Notably, while the M14^fl/fl-Ucp1^cre model was featured in our recent report^[283]19, the current study includes newly generated data from a different cohort of animals in this model, which is essential as it provides a critical BAT-specific knockout for comparison with the overall-adipose knockout (M14^fl/fl-Adipoq^cre) model. This approach allows us to differentiate BAT-specific effects from those in total adipose tissue. Furthermore, the consistency of findings in different cohorts of animals demonstrates the reproducibility and robustness of our experimental approach, further validating the significance of our results. A study recently reported by Kang and colleagues^[284]29 also used the M14^fl/fl-Adipoq^cre mouse model to create an adipose-specific Mettl14-knockout. While our study, as well as the study by Kang et al., observed decreased body weight, impaired cold tolerance, decreased WAT mass, and increased BAT mass, several obvious differences stand out when comparing the two reports: First, while the major findings in the Kang et al. study exclusively rely on the Mettl14^fl/fl-Adipoq^cre model, it was surprising that no information regarding the origin of Mettl14^fl/fl mice and the KO targeting strategy (e.g., which exons are floxed) was provided, which limits the comparison between our KO model and theirs. On the other hand, we and several other groups have characterized our Mettl14^fl/fl mice^[285]21,[286]30. Second, in their study, Kang et al surprisingly do not use m^6A-seq but bulk RNA-seq to determine targets that contribute to the phenotype. In contrast, we used m^6A-MeRIP- and RNA-seq as drivers to determine the mechanism(s) that underlie the phenotype. Considering METTL14’s non-m^6A-dependent effects, it is likely the approach by Kang et al. introduced bias in the identification of m^6A-targeted genes. Third, in their study, Kang et al. attribute decreased body weight, enhanced insulin sensitivity, and improved glucose tolerance to heightened cell-autonomous lipolysis resulting from Mettl14 deficiency. This conclusion is inconsistent with the large body of literature demonstrating that excessive lipolysis, particularly in white adipocytes, contributes to adipose tissue- and systemic “insulin resistance”^[287]31–[288]34. The exacerbated insulin resistance is secondary to the influx of elevated circulating FFAs into non-adipose tissues and the release of proinflammatory cytokines^[289]31, both of which interfere with insulin signaling. Kang et al. noted increased circulating FFAs, improved hepatic insulin sensitivity and steatosis in their model, while reduced Tnfα and Il6 gene expression in M14^KO adipose tissue, confounding the overall interpretation of the phenotype^[290]29. Finally, insulin usually inhibits lipolysis via Akt activation^[291]35,[292]36, yet the authors suggested enhanced insulin sensitivity and lipolysis synergy, creating interpretive challenges. In contrast, our study reveals the downregulation of lipolysis pathways in the Mettl14^fl/fl-Adipoq^cre iWAT (Fig. [293]5i), aligning with a consistent decrease in circulating FFAs (Fig. [294]7i). Mettl14 deficiency in WAT induces insulin resistance in a tissue-autonomous manner. This, in turn, leads to severe peripheral insulin resistance through adipocyte apoptosis-induced triglyceride spillover and systemic inflammation. Nevertheless, other factors, including diets, and differences in microbiome composition secondary to housing conditions should be considered when interpreting the discrepancies of the two studies. Our study identifies specific transcripts by which METTL14 negatively governs the secretory function within BAT and positively regulates the lipid sequestration capability within WAT in an m^6A-dependent manner. This cell-type-specific regulation by METTL14-mediated m^6A can be attributed to various factors. The level of m^6A modification depends on the expression and abundance of m^6A regulators including METTL14 and METTL3, the intrinsic preference of METTL3/METTL14 methyltransferase for specific nucleotide sequences, and the extrinsic regulation of methyltransferase complex activity by transcription factors, RNA-binding proteins (PBPs), RNA polymerases, histone modification, or posttranslational modifications^[295]37–[296]39. The cell-type specificity of m^6A modifications is highlighted by the differential expression and protein abundance of METTL14 across various cell types. Specifically, under physiological conditions, METTL14 exhibits significantly lower protein levels of iBAT and hBAs compared to iWAT and hWAs, respectively (Supplementary Fig. [297]1f–[298]h). Despite METTL3 being the enzyme with catalytic activity, its function is critically dependent on forming a complex with METTL14^[299]40. Therefore, the observed variability in METTL14 abundance may lead to differences in the activity of the m^6A writer complex and substrate availability. This, in turn, could result in distinct m^6A target preferences, influencing the physiological functions of brown versus