The induction of beige adipocytes in white adipose tissue (WAT), also known as WAT beiging, improves glucose and lipid metabolism. However, the regulation of WAT beiging at the posttranscriptional level remains to be studied. Here, we report that METTL3, the methyltransferase of N6-methyladenosine (m6A) mRNA modification, is induced during WAT beiging in mice. Adipose-specific depletion of the Mettl3 gene undermines WAT beiging and impairs the metabolic capability of mice fed with a high-fat diet. Mechanistically, METTL3-catalyzed m6A installation on thermogenic mRNAs, including Krüppel-like factor 9 (Klf9), prevents their degradation. Activation of the METTL3 complex by its chemical ligand methyl piperidine-3-carboxylate promotes WAT beiging, reduces body weight, and corrects metabolic disorders in diet-induced obese mice. These findings uncover a novel epitranscriptional mechanism in WAT beiging and identify METTL3 as a potential therapeutic target for obesity-associated diseases.

Article Highlights

  • METTL3, the methyltransferase of N6-methyladenosine (m6A) mRNA modification, is induced during WAT beiging.

  • Depletion of Mettl3 undermines WAT beiging and impairs thermogenesis.

  • METTL3-mediated m6A installation promotes the stability of Krüppel-like factor 9 (Klf9).

  • KLF9 rescues impaired beiging elicited by Mettl3 depletion.

  • Pharmaceutical activation of the METTL3 complex by its chemical ligand methyl piperidine-3-carboxylate induces WAT beiging.

  • Methyl piperidine-3-carboxylate corrects obesity-associated disorders.

  • The METTL3-KLF9 pathway may serve as a potential therapeutic target for obesity-associated diseases.

Adipose tissue is a central metabolic organ, the dysfunction of which disrupts energy homeostasis and induces the pathogenesis of metabolic disorders, such as obesity and type 2 diabetes (1). Three different types of adipocytes have been identified: white, brown, and beige (2). White adipocytes are responsible for fat storage, whereas beige and brown adipocytes mediate adaptive thermogenesis by expressing uncoupling protein 1 (UCP1) (2,3). Although beige and brown adipocytes share similar thermogenic features, they have distinct origins and molecular identities (4). Beige adipocytes originate from white adipose tissue (WAT), a process that could be induced by various environmental stimuli, such as cold exposure, exercise, and the agonists of peroxisome proliferator–activated receptor-γ (PPARγ) (2,4). The inducible feature of beige adipocytes highlights an attractive way to treat obesity-associated disorders (4,5).

The beiging process relies on the activation of a beige adipocyte–specific gene program, which is regulated by several transcriptional and/or epigenetic factors (5). Transcriptional regulators, such as PPARγ, C/EBPβ, PGC1α, and PRDM16, lie at the center of the beige adipocyte fate decision (5,6). In addition to transcriptional regulation, gene expression could be regulated at the posttranscriptional level, but the posttranscriptional regulation of the beiging process remains largely uncharacterized.

N6-methyladenosine (m6A), the most abundant modification on mRNAs, has been defined as a new layer of gene expression regulation (7). The formation of m6A is catalyzed by the RNA methyltransferase complex METTL3/METTL14/WTAP (810), and demethylation is mediated by ALKBH5 or FTO (11,12). The metabolism of m6A-modified mRNAs, including mRNA splicing, degradation, and translation, is regulated by reader proteins, including YTH domain proteins (13,14).

Evidence indicates m6A has a role in adipocyte metabolism. Epidemiologic studies demonstrate that the single nucleotide polymorphism of FTO is highly correlated with the incidence of obesity in humans (15). Depletion of Fto in mice shows obvious reduction in adipose tissue mass (16). Moreover, Fto deletion in vitro promotes thermogenesis and white-to-beige adipocyte transition (17). In contrast, METTL3 is reported to positively regulate the development and energy expenditure of brown adipose tissue (BAT) (18). Entacapone, a recently identified FTO inhibitor, dramatically enhances thermogenesis in adipose tissue (19). However, whether targeting METTL3 could improve adipose tissue metabolism remains to be explored.

In this study, we demonstrate an essential role of METTL3 in WAT beiging. METTL3-mediated m6A installation promotes the stability of mRNAs involved in thermogenesis. Pharmaceutical activation of the METTL3 complex induces WAT beiging and corrects obesity-associated disorders. These results highlight METTL3 as a promoter of the beiging process and a potential target for obesity treatment.

Mouse Experiments

Eight-week-old male mice on the C57BL/6J background were used for experiments unless mentioned otherwise. Mice were fed with either a standard chow (CD) (10% fat as kcal; cat. no. D12450J; Research Diets, Inc., New Brunswick, NJ) or a high-fat diet (HFD) (60% fat-derived calories; cat. no. D12492; Research Diets Inc.) for 8 weeks. Mice containing a loxP-flanked Mettl3 allele (Mettl3fl/fl) were provided by Ming-Han Tong (Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences). Mettl3 adipose tissue conditional knockout mice were generated by mating Mettl3fl/fl mice with Adipoq-Cre mice (The Jackson Laboratory). The primers used for genotyping are listed in Supplementary Table 1. For cold exposure, mice were individually housed in plastic cages at 4°C for 7 days. After anesthetization with 2% isoflurane, 6-week-old mice were injected with recombinant adeno-associated virus (AAV) (1.0 × 1012 vg per 20 μL PBS) into inguinal WAT (iWAT) according to the method provided by Zhu et al. (20). For compound treatment, mice were intraperitoneally injected with methyl piperidine-3-carboxylate (MP3C) (Shanghai Aladdin Biochemical Technology) at 2.5 mg/kg every 3 days for 2 weeks. The metabolic parameters of each mouse were monitored using the TSE Systems PhenoMaster (Berlin, Germany). Oxygen (O2) consumption, carbon dioxide (CO2) generation, and energy heat generation were recorded for each mouse and normalized to body weight. iWAT, epididymal WAT (eWAT), and interscapular BAT from mice were surgically removed and immediately frozen at −80°C for additional analyses.

Mice were maintained in specific pathogen-free conditions at the Animal Center of Zhejiang University. All animal studies were performed in compliance with the Guide for the Care and Use of Laboratory Animals by the Medical Experimental Animal Care Commission of Zhejiang University. All animal studies used the protocol that has been approved by the Medical Experimental Animal Care Commission of Zhejiang University (AP code ZJU20220513).

Glucose and Insulin Tolerance Tests

For the glucose tolerance test, HFD-fed mice were intraperitoneally injected with 100 mg/mL d-glucose at a dose of 0.8 g/kg body weight after overnight fasting. For the insulin tolerance test, HFD-fed mice were intraperitoneally injected with human insulin at a dose of 1.0 units/kg body weight after 4-h fasting. Glucose level was measured in tail blood at 0, 15, 30, 60, 90, and 120 min after glucose or insulin injection using a glucometer (Accu-Chek; Roche). Core body temperature was measured intrarectally at approximately 4:00 p.m.

WAT O2 Consumption

O2 consumption rates (OCRs) were measured using Clark-type oxygen electrodes (Strathkelvin Instruments). Freshly isolated tissue (∼30 mg) was minced in respiration buffer (1.5 mmol/L pyruvate, 25 mmol/L glucose, and 2% BSA) and placed in electrode chambers, and the OCR was detected (21).

Cell Culture

3T3-L1 preadipocytes, HEK293T cells, and AAV293 cells were obtained from ATCC and cultured in DMEM medium (HyClone Laboratories, Logan, UT), supplemented with 10% FBS (Thermo Fisher Scientific, Scoresby, Victoria, Australia).

Plasmid Construction

To construct shRNA-expressing plasmids, shRNA oligos were cloned to the lentiviral vector pLKO.1. The coding sequence (CDS) of Krüppel-like factor 9 (Klf9) was cloned into the pAAV-MCS plasmid. To construct and purify FLAG-tagged protein, the CDS of METTL3 or METTL16 tagged at the C terminus with FLAG was cloned into the lentivector pCDH. To construct luciferase reporters, the 3′ end of the Klf9 CDS region was first amplified from cDNA of iWAT and cloned into pGL3-control vector in frame with the luciferase gene (Promega, Madison, WI). The oligos used for plasmid construction are listed in Supplementary Table 1.

Lentivirus and AAV Packaging

The lentivirus was packaged in HEK293T cells with helper plasmids pMD2.G and psPAX2. The AAV was packaged in AAV293 cells with helper plasmids pHelper and RC. After 72-h culture, the supernatant was collected for infection.

Luciferase Reporter Assay

Klf9 luciferase reporters were cotransfected with pRL-TK into 3T3-L1 cells in a 12-well plate using Lipofectamine 2000 (Invitrogen, Waltham, MA). Cells were harvested at 24 h posttransfection. Firefly and Rinella luciferase activities were measured using the Dual-Luciferase Assay Kit (Promega). The activities of the luciferase were expressed as normalized relative light units to the Rinella internal control.

METTL3 and METTL16 Protein Production

HEK293 cells were transfected with METTL3 or METTL16 plasmids using Lipofectamine 2000. METTL3 and METTL16 were isolated by lysis of HEK293 cells at 48 h posttransfection, and the lysate was purified with anti-FLAG M2 magnetic beads (Sigma-Aldrich, St Louis, MO).

METTL3 and METTL16 Enzymatic Assay

The experiments were conducted in reaction buffer (20 mmol/L Tris-hydrochloride [pH 7.5], 1 mmol/L dithiothreitol, 0.01% Triton X-100, and 20 units/100 μL buffer RNaseOUT) (22,23). The reaction mixture contained 2.5 μmol/L unmethylated single-stranded RNA probe with a biotin tag (5′-biotin-CGUCUCGGACUCGGACUGCU-3′; Dharmacon, Inc.), 25 μmol/L S-adenosylmethionine (Yeasen Biotechnology, Shanghai, China), and 50 ng/μL purified METTL3 or METTL16 protein. Enzymatic assay reactions were transferred to streptavidin-coated 96-well plates (Beaver Biomedical Engineering, Suzhou, China) and incubated for 20 h at 21°C on shakers. The plate was incubated with rabbit anti-m6A antibody (Synaptic Systems, Göttingen, Germany) for 2 h and incubated for additional 1 h with horseradish peroxidase–conjugated secondary antibody at room temperature with slow shaking. The plate was incubated with TMB (3,3′, 5,5; -tetramethylbenzidine) chromogen solution (Biosharp Life Sciences, Hefei, China) for 30 min. The reactions were stopped with 2 mol/L hydrochloride, and expression levels of m6A were detected at 450 nm using Varioskan Flash (Thermo Fisher Scientific).

m6A Detection

mRNA was purified from total RNA by using Dynabeads Oligo(dT)25 (Invitrogen) and spotted to a Hybond-N+ membrane (GE Healthcare, Waukesha, WI), followed by ultraviolet crosslinking at ultraviolet 254 nm (0.12 J/cm2). The membrane was blocked with 5% nonfat milk in PBS and then incubated with an anti-m6A antibody. The membrane was incubated with horseradish peroxidase–conjugated secondary antibody and visualized by using enhanced chemiluminescence. Methylene blue staining was used as mRNA loading control.

RNA, m6A, and Ribosome Sequencing

Total RNA from iWAT was extracted with TRIzol reagent. The polyadenylated RNA was enriched using Dynabeads Oligo(dT)25 and fragmented into fragments ∼100 nucleotides long using RNA fragmentation reagent (cat. no. AM8740; Ambion). Fragmented RNA samples were used for library construction and high-throughput sequencing by Hangzhou KaiTai Biotechnology Co., Ltd.

For m6A sequencing (m6A-seq), fragmented RNA was incubated with m6A primary antibody–coated beads for 6 h at 4°C. The immunoprecipitation complex was digested with proteinase K at 55°C for 1 h. RNA was then extracted using TRIzol reagent and used for library construction and high-throughput sequencing.

For ribosome sequencing (ribo-seq), the iWAT of mice was homogenized in polysome lysis buffer. One of 10 clarified lysates was set aside as input. The remaining nine of 10 lysates were digested with Escherichia coli RNase I (Ambion) for 1 h. Ribosome-protected fragments were collected by sucrose density gradient ultracentrifugation and extracted using TRIzol reagent. The library was constructed with a small RNA library construction kit (New Englad Biolabs) and used for high-throughput sequencing.

Sequencing Data Analysis

For RNA sequencing (RNA-seq) and m6A-seq, adaptor and low-quality bases were trimmed by Fastp, and the trimmed reads with lengths <15 nucleotides were discarded (24). The remaining reads were mapped to the mouse reference genome (mm10) using HISAT2 (25). After counting the number of reads, the expression of transcripts was quantified as reads per kilobase per million mapped reads (RPKM). Read coverage of m6A-seq and RNA-seq on transcripts was plotted, and the m6A peaks were further identified based on the following criteria: 1) genes with sufficient expression level (RPKM >1), 2) m6A modification detected by software MACS2 (significance level ≤0.05), and 3) POI (peak reads/input reads) >2 (26).

Statistical Analysis

Statistical analysis was performed using GraphPad Prism 8 software (GraphPad Software, Inc., San Diego, CA). Throughout, data are represented as mean ± SD unless stated otherwise. Asterisks denote the level of statistical significance (*P < 0.05, **P < 0.01, ***P < 0.001, and ****P < 0.0001).

Data and Resource Availability

The data and resources that support the findings of this study are available from the corresponding author upon reasonable request. Sequencing data, including m6A-seq, ribo-seq, and RNA-seq data, are available in the Gene Expression Omnibus under accession number GSE178884 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE178884).

METTL3-Mediated m6A Reprogramming During Cold-Induced WAT Beiging

We determined the expression level of METTL3 in different types of adipose tissue. Data revealed that METTL3 is ubiquitously expressed in adipose tissue with enrichment in BAT (Supplementary Fig. 1A). Cold treatment (4°C) successfully induced the beiging of WAT, as evidenced by smaller lipid droplets and induction of thermogenesis genes, including UCP1 (Fig. 1A). Strikingly, the protein level of METTL3 increased in beige adipose tissue (Fig. 1A and B). Consistent with the upregulation of METTL3, the total m6A methylation level was significantly increased in beige adipose tissue but decreased after Mettl3 knockout (Fig. 1C), implying that METTL3-mediated m6A formation may be involved in the beiging process.

Figure 1

Reprogramming of m6A methylome during WAT beiging. A: Hematoxylin-eosin (H-E) staining and immunohistochemical staining of UCP1 and METTL3 in iWAT from mice housed at room temperature (RT) or 4°C for 7 days. Scale bar, 50 μm. B: Immunoblot analysis of METTL3 and UCP1 expression in iWAT from mice housed at room temperature or 4°C. C: Dot blot analysis of m6A level of mRNAs extracted from the indicated iWAT. Methylene blue (MB) was used for loading control. D: Metagene profiles of m6A distribution across transcripts in iWAT from mice housed at RT or 4°C (two biological replicates). E: Consensus motif of m6A sites in the above samples. F: Gene Set Enrichment Analysis analysis of gene set for positive regulation of cold induced thermogenesis. Negative normalized enrichment score (NES) indicated lower expression in iWAT from mice housed at RT (two biologic replicates). G: Top Kyoto Encyclopedia of Genes and Genomes (KEGG) terms enriched for transcripts with upregulated m6A modification during beiging (two biologic replicates). FDR, false discovery rate.

Figure 1

Reprogramming of m6A methylome during WAT beiging. A: Hematoxylin-eosin (H-E) staining and immunohistochemical staining of UCP1 and METTL3 in iWAT from mice housed at room temperature (RT) or 4°C for 7 days. Scale bar, 50 μm. B: Immunoblot analysis of METTL3 and UCP1 expression in iWAT from mice housed at room temperature or 4°C. C: Dot blot analysis of m6A level of mRNAs extracted from the indicated iWAT. Methylene blue (MB) was used for loading control. D: Metagene profiles of m6A distribution across transcripts in iWAT from mice housed at RT or 4°C (two biological replicates). E: Consensus motif of m6A sites in the above samples. F: Gene Set Enrichment Analysis analysis of gene set for positive regulation of cold induced thermogenesis. Negative normalized enrichment score (NES) indicated lower expression in iWAT from mice housed at RT (two biologic replicates). G: Top Kyoto Encyclopedia of Genes and Genomes (KEGG) terms enriched for transcripts with upregulated m6A modification during beiging (two biologic replicates). FDR, false discovery rate.

Close modal

To analyze the m6A signature, we performed m6A-seq in WAT from wild-type mice treated with or without cold exposure (Supplementary Fig. 1B). m6A-seq data revealed the expected distribution of m6A peaks enriched at the stop codon (Fig. 1D). However, the pattern of the two groups was different. The m6A peaks in start and stop codons were higher in WAT from control mice, whereas the CDS region was significantly higher in WAT from cold-treated mice (Fig. 1D and Supplementary Fig. 1C). The m6A peaks in both groups were characterized by the canonic AGACA motif (Fig. 1E) (27,28). Further analysis of m6A-seq data identified 6,199 differential m6A sites within 3,853 transcripts. A higher number of sites showed elevated m6A methylation in beige adipose tissue compared with WAT (Supplementary Fig. 1D). m6A methylation alternation also reflected cold-induced thermogenesis in Gene Set Enrichment Analysis (Fig. 1F). Furthermore, Kyoto Encyclopedia of Genes and Genomes pathway and Gene Ontology analyses of the m6A methylome revealed that the genes with upregulated m6A modification during beiging were involved in a series of metabolism- and thermogenesis-related pathways, such as oxidative phosphorylation (Cox10 and Ndufa5), fatty acid metabolism (Cpt2, Elovl6, and Scd2), and carbon metabolism (Pfkp, Phgdh, and Eno1) (Fig. 1G and Supplementary Fig. 1E). Together, these data provide an overview of METTL3-mediated m6A reprogramming during beiging and imply a role for m6A mRNA modification in this biological process.

Depletion of Mettl3 Undermines WAT Beiging

To study the function of METTL3 in adipose tissue, we generated an adipocyte-specific Mettl3 knockout mouse model under the adiponectin promoter (AdipoqCre/+: Mettl3fl/fl, termed Mettl3cKO or M3cKO) and successfully depleted Mettl3 in adipose tissue (Fig. 2A and Supplementary Fig. 2A). Compared with wild-type mice (Mettl3fl/fl, termed Mettl3CTL or M3CTL), Mettl3cKO mice had similar weights of BAT, iWAT, eWAT, and liver, as well as similar body weights (Supplementary Fig. 2B and C). Of note, daily food intake on standard CD did not change in the two groups (Supplementary Fig. 2D). Mettl3cKO mice had whiter BAT (Supplementary Fig. 2E), in line with the previous report that UCP1-Cre-driven Mettl3 depletion impairs BAT maturation and energy expenditure (18). Mature iWAT is characterized by high expression of complement factor D (Adipsin) and low expression of preadipocyte factor-1 (Pref-1). Mettl3 depletion did not alter the expression of Adipsin or Pref-1 mRNA in vivo, indicating the successful maturation of WAT (Supplementary Fig. 2F). Hematoxylin-eosin staining showed the similar size of lipid droplets in Mettl3cKO and Mettl3CTL mice (Supplementary Fig. 2G). The above data suggest that adipocyte-specific knockout of Mettl3 does not impair WAT adipogenesis.

Figure 2

METTL3 is required for WAT beiging. A: Generation of M3cKO mouse. B: Gross view of iWAT from M3CTL and M3cKO mice after 7-day cold exposure (4°C). Scale bar, 0.5 cm. C: Hematoxylin-eosin staining of iWAT from M3CTL and M3cKO mice after 7-day cold exposure. Scale bar, 50 μm. D: Immunoblot of iWAT from M3CTL and M3cKO mice after 7-day cold exposure. E: Heat map showing the mRNA level of essential genes related to the adipose tissue general marker, lipolysis and fatty acid β-oxidation (FAO), and thermogenesis in RNA-seq data from M3CTL and M3cKO mice after 7-day cold exposure (two biologic replicates). F: Quantitative RT-PCR analysis of mRNA level of thermogenic and lipolytic genes in iWAT from M3CTL and M3cKO mice after 7-day cold exposure (*P < 0.05, **P < 0.01; three independent experiments; two-tailed t test).

Figure 2

METTL3 is required for WAT beiging. A: Generation of M3cKO mouse. B: Gross view of iWAT from M3CTL and M3cKO mice after 7-day cold exposure (4°C). Scale bar, 0.5 cm. C: Hematoxylin-eosin staining of iWAT from M3CTL and M3cKO mice after 7-day cold exposure. Scale bar, 50 μm. D: Immunoblot of iWAT from M3CTL and M3cKO mice after 7-day cold exposure. E: Heat map showing the mRNA level of essential genes related to the adipose tissue general marker, lipolysis and fatty acid β-oxidation (FAO), and thermogenesis in RNA-seq data from M3CTL and M3cKO mice after 7-day cold exposure (two biologic replicates). F: Quantitative RT-PCR analysis of mRNA level of thermogenic and lipolytic genes in iWAT from M3CTL and M3cKO mice after 7-day cold exposure (*P < 0.05, **P < 0.01; three independent experiments; two-tailed t test).

Close modal

The upregulation of METTL3 during WAT beiging prompted us to study its function in this process. We found that cold exposure failed to induce beiging in Mettl3cKO mice (Fig. 2B). Histological analysis showed that the adipose tissue of Mettl3cKO mice had larger lipid droplets than that of Mettl3CTL mice (Fig. 2C). Consistently, the expression of UCP1, along with other thermogenic and lipolytic genes, was dramatically reduced in the iWAT of Mettl3cKO mice (Fig. 2D–F), just as in the BAT of Mettl3cKO mice (Supplementary Fig. 2H and I).

Depletion of Mettl3 Impairs Metabolic Capability Under HFD Feeding

The process of WAT beiging antagonizes obesity. One study showed that UCP1-Cre-driven Mettl3 knockout mice were prone to obesity (18). To further study the role of Mettl3 in the regulation of the metabolic capability of mice, we treated M3CTL and M3cKO mice with HFD feeding. Interestingly, we found that HFD-treated mice showed decreased expression of METTL3 in iWAT compared with CD-fed mice (Fig. 3A). The depletion of Mettl3 had no effect on body weight or rectal temperature under similar food intake (Fig. 3B–D). However, the BAT of M3cKO mice was whiter and larger than that of M3CTL mice (Fig. 3E and F). M3cKO mice also showed impaired glucose tolerance and insulin resistance compared with M3CTL mice (Fig. 3G and H). O2 consumption, CO2 generation, and energy heat generation were significantly reduced during light and dark cycles in M3cKO mice (Fig. 3I–K). Despite the unchanged body weight that can be attributed to the low expression of METTL3 under HFD treatment, these data imply that the depletion of Mettl3 impairs the metabolic capability of diet-induced obese mice.

Figure 3

Depletion of Mettl3 impairs the metabolic capability of obese mice. A: Immunoblot analysis of METTL3 expression in iWAT of mice treated with CD or HFD. B: Body weight of HFD-fed M3CTL and M3cKO mice (n = 6 mice in each group; two-tailed t test). C: Food intake was measured in HFD-fed M3CTL and M3cKO mice (n = 6 mice in each group; two-tailed t test). D: Rectal temperature was measured in HFD-fed M3CTL and M3cKO mice (n = 5 mice in each group; two-tailed t test). E: Gross view of iWAT and BAT from M3CTL and M3cKO mice. F: Weight of iWAT and BAT from M3CTL and M3cKO mice (**P < 0.01; n = 5 M3CTL mice and n = 6 M3cKO mice; two-tailed t test). G and H: Glucose (GTT) (G) and insulin tolerance test (ITT) (H) assays were performed in HFD-fed M3CTL and M3cKO mice (*P < 0.05, **P < 0.01; n = 6 mice in each group; two-tailed t test). IK: O2 consumption (I), CO2 generation (J), and energy heat generation (K) in M3CTL and M3cKO mice fed an HFD (***P < 0.001, ****P < 0.0001; n = 3 mice in each group; two-tailed t test). White and gray areas in the graphs indicate day and night, respectively.

Figure 3

Depletion of Mettl3 impairs the metabolic capability of obese mice. A: Immunoblot analysis of METTL3 expression in iWAT of mice treated with CD or HFD. B: Body weight of HFD-fed M3CTL and M3cKO mice (n = 6 mice in each group; two-tailed t test). C: Food intake was measured in HFD-fed M3CTL and M3cKO mice (n = 6 mice in each group; two-tailed t test). D: Rectal temperature was measured in HFD-fed M3CTL and M3cKO mice (n = 5 mice in each group; two-tailed t test). E: Gross view of iWAT and BAT from M3CTL and M3cKO mice. F: Weight of iWAT and BAT from M3CTL and M3cKO mice (**P < 0.01; n = 5 M3CTL mice and n = 6 M3cKO mice; two-tailed t test). G and H: Glucose (GTT) (G) and insulin tolerance test (ITT) (H) assays were performed in HFD-fed M3CTL and M3cKO mice (*P < 0.05, **P < 0.01; n = 6 mice in each group; two-tailed t test). IK: O2 consumption (I), CO2 generation (J), and energy heat generation (K) in M3CTL and M3cKO mice fed an HFD (***P < 0.001, ****P < 0.0001; n = 3 mice in each group; two-tailed t test). White and gray areas in the graphs indicate day and night, respectively.

Close modal

METTL3 Regulates the Expression of Thermogenic Genes at the mRNA Level

To identify METTL3-targeted m6A modifications and their function in mRNA metabolism, we performed m6A-seq, RNA-seq, and ribo-seq in beige adipose tissue from Mettl3cKO and Mettl3CTL mice (Fig. 4A and B and Supplementary Fig. 3A). We considered that m6A sites with a decreased signal after Mettl3 knockout were METTL3 direct targets. On the basis of this criterion, we identified 3,827 METTL3-targeted m6A sites located in 2,847 transcripts, which account for ∼62% of all m6A modifications in mRNAs in beige adipose tissue (Fig. 4C and Supplementary Fig. 3B). These METTL3-targeted transcripts showed a notable decrease at the mRNA level (RNA-seq) after Mettl3 knockout, compared with non-METTL3-targeted transcripts and non-m6A transcripts (Fig. 4D and E and Supplementary Fig. 3C), suggesting that m6A modification in beige adipocytes positively regulates gene expression. Specifically, the m6A distribution within UCP1 was significantly increased in beige adipose tissue but decreased after Mettl3 knockout (Supplementary Fig. 3D), which is consistent with the previous report (18). The m6A methylation levels of other thermogenic-associated genes, such as Pparg, Prdm16, and Zfp423, changed slightly in beige adipose tissue (Supplementary Fig. 3EG). Ribo-seq change correlated well with RNA-seq change, and only a small group of mRNAs were regulated at the translational level (altered in ribo-seq but not RNA-seq) (Fig. 4F). These data suggest that METTL3 regulates gene expression mainly at the mRNA level during iWAT beiging.

Figure 4

METTL3 regulates the expression of m6A-modified mRNAs. A: Consensus motif of m6A sites in iWAT from M3CTL and M3cKO mice after 7-day cold exposure (4°C) (two biologic replicates). B: Metagene profiles of m6A distribution across transcripts in iWAT from M3CTL and M3cKO mice after 7-day cold exposure (two biologic replicates). C: The relative m6A peak coverage for METTL3-targeted and non–METTL3-targeted transcripts in M3CTL and M3cKO iWAT from mice after 7-day cold exposure (two biological replicates). Upper and lower quartiles and median are shown for each group (****P < 0.0001; Mann-Whitney U test). D: Violin plots showing mRNA change between M3CTL and M3cKO iWAT for METTL3-targeted and non–METTL3-targeted transcripts from mice after 7-day cold exposure (two biologic replicates). Upper and lower quartiles and median are indicated for each group (Mann-Whitney U test). E: Cumulative distributions of mRNA change between M3CTL and M3cKO iWAT for METTL3-targeted and non-METTL3-targeted transcripts as in panel D (Mann-Whitney U test). F: Scatterplot showing RNA-seq and ribo-seq data of total mRNA, m6A targets, and M3 targets (two biologic replicates). POI, peak over input.

Figure 4

METTL3 regulates the expression of m6A-modified mRNAs. A: Consensus motif of m6A sites in iWAT from M3CTL and M3cKO mice after 7-day cold exposure (4°C) (two biologic replicates). B: Metagene profiles of m6A distribution across transcripts in iWAT from M3CTL and M3cKO mice after 7-day cold exposure (two biologic replicates). C: The relative m6A peak coverage for METTL3-targeted and non–METTL3-targeted transcripts in M3CTL and M3cKO iWAT from mice after 7-day cold exposure (two biological replicates). Upper and lower quartiles and median are shown for each group (****P < 0.0001; Mann-Whitney U test). D: Violin plots showing mRNA change between M3CTL and M3cKO iWAT for METTL3-targeted and non–METTL3-targeted transcripts from mice after 7-day cold exposure (two biologic replicates). Upper and lower quartiles and median are indicated for each group (Mann-Whitney U test). E: Cumulative distributions of mRNA change between M3CTL and M3cKO iWAT for METTL3-targeted and non-METTL3-targeted transcripts as in panel D (Mann-Whitney U test). F: Scatterplot showing RNA-seq and ribo-seq data of total mRNA, m6A targets, and M3 targets (two biologic replicates). POI, peak over input.

Close modal

KLF9 Rescues Impaired Beiging Elicited by Mettl3 Depletion

Metagene analysis revealed a common reduction of m6A methylation around the stop codon of transcripts in Mettl3cKO beige adipose tissue (Fig. 5A). Among the altered transcripts, we noticed that m6A modification in Klf9 mRNA showed a dramatic change (Fig. 5B). Consistently, the mRNA level of Klf9 was also dramatically reduced in Mettl3 knockout iWAT (Fig. 5C). The elevated expression of KLF9 in beige adipose tissue was also confirmed, implying a role of KLF9 in iWAT beiging (Fig. 5D and E). To decipher the functional relationship between KLF9 and METTL3, we performed rescue experiments by expressing FLAG-tagged KLF9 in Mettl3-depleted iWAT using an AAV8 system (Fig. 5F). Mettl3 knockout induced larger lipid droplets and inhibited the expression of UCP1 (Fig. 5G and H). Overexpression of KLF9 largely reversed the impaired beiging and UCP1 expression elicited by METTL3 deletion (Fig. 5G and H). These results indicate that KLF9 is a key effector in promoting beiging downstream of METTL3.

Figure 5

KLF9 rescues impaired beiging elicited by Mettl3 deficiency. A: Distribution of m6A peaks in the vicinity of stop codons of iWAT mRNAs from cold-exposed M3CTL and M3cKO mice (two biologic replicates). B: Volcano plot of m6A fold change from iWAT of cold-treated M3CTL and M3cKO mice (two biologic replicates). C: mRNA level of Klf9 in iWAT depots from cold-treated M3CTL and M3cKO mice (*P < 0.05; three independent experiments; two-tailed t test). D: Immunoblot analysis of KLF9 expression in iWAT from mice housed at room temperature or 4°C. E: mRNA level of Klf9 in iWAT from mice housed at room temperature or 4°C (*P < 0.05; three independent experiments; two-tailed t test). F: Schematic diagram of the rescue experiment using an AAV system. G: Immunoblot analysis of iWAT from M3CTL and M3cKO mice with or without KLF9 expression after 7-day cold exposure. H: Hematoxylin-eosin (H-E) staining and immunohistochemical staining of UCP1 of iWAT depots from M3CTL and M3cKO mice with or without KLF9 expression after 7-day cold exposure. Scale bar, 50 μm.

Figure 5

KLF9 rescues impaired beiging elicited by Mettl3 deficiency. A: Distribution of m6A peaks in the vicinity of stop codons of iWAT mRNAs from cold-exposed M3CTL and M3cKO mice (two biologic replicates). B: Volcano plot of m6A fold change from iWAT of cold-treated M3CTL and M3cKO mice (two biologic replicates). C: mRNA level of Klf9 in iWAT depots from cold-treated M3CTL and M3cKO mice (*P < 0.05; three independent experiments; two-tailed t test). D: Immunoblot analysis of KLF9 expression in iWAT from mice housed at room temperature or 4°C. E: mRNA level of Klf9 in iWAT from mice housed at room temperature or 4°C (*P < 0.05; three independent experiments; two-tailed t test). F: Schematic diagram of the rescue experiment using an AAV system. G: Immunoblot analysis of iWAT from M3CTL and M3cKO mice with or without KLF9 expression after 7-day cold exposure. H: Hematoxylin-eosin (H-E) staining and immunohistochemical staining of UCP1 of iWAT depots from M3CTL and M3cKO mice with or without KLF9 expression after 7-day cold exposure. Scale bar, 50 μm.

Close modal

METTL3-Dependent m6A Installation Stabilizes Klf9 mRNA

To resolve the direct regulation of METTL3 on KLF9 expression, we generated Mettl3 knockdown preadipocyte 3T3-L1 cell lines (Fig. 6A), which also showed decreased KLF9 protein. Consistent with the result in vivo, knockdown of METTL3 dramatically reduced the expression of Klf9 at the mRNA level (Fig. 6A). Furthermore, we examined the stability of Klf9 mRNA by blocking de novo RNA synthesis with actinomycin D. The half-life of Klf9 mRNA is longer in control cells than in METTL3-deficient cells (Fig. 6B), indicating that METTL3-mediated m6A modification stabilizes Klf9 mRNA.

Figure 6

METTL3-dependent m6A promotes the stability of Klf9 mRNA. A: Protein and mRNA levels of Klf9 expression in 3T3-L1 cells with or without METTL3 knockdown (****P < 0.0001; three independent experiments; two-tailed t test). B: Remaining mRNA level of Klf9 after actinomycin D treatment in control or METTL3 knockdown cells. C: m6A distribution within Klf9 transcript in different samples (two biologic replicates; *indicates the predicted m6A peak). D: Schematic of luciferase constructs with the predicted m6A sites in the CDS and 3′UTR of Klf9 mRNA. E: Luciferase activity and mRNA level of the constructs as indicated in panel D in 3T3-L1 cells (**P < 0.01; three independent experiments; two-tailed t test). RT, room temperature.

Figure 6

METTL3-dependent m6A promotes the stability of Klf9 mRNA. A: Protein and mRNA levels of Klf9 expression in 3T3-L1 cells with or without METTL3 knockdown (****P < 0.0001; three independent experiments; two-tailed t test). B: Remaining mRNA level of Klf9 after actinomycin D treatment in control or METTL3 knockdown cells. C: m6A distribution within Klf9 transcript in different samples (two biologic replicates; *indicates the predicted m6A peak). D: Schematic of luciferase constructs with the predicted m6A sites in the CDS and 3′UTR of Klf9 mRNA. E: Luciferase activity and mRNA level of the constructs as indicated in panel D in 3T3-L1 cells (**P < 0.01; three independent experiments; two-tailed t test). RT, room temperature.

Close modal

Two m6A peaks were identified in Klf9 mRNA, lying in the CDS region near stop codon and the 3′ untranslated region (UTR), respectively. The m6A peak in the 3′UTR (1816nt-2199nt of the transcript ENSMUST00000036884) showed dynamic change, with an increase in cold-treated iWAT and a decrease in Mettl3cKO iWAT (Fig. 6C). To examine whether the m6A modification in this region regulates Klf9 stability, we cloned each of the m6A peaks to a luciferase reporter (Fig. 6D). Erasing m6A by silencing METTL3 decreased Klf9-3′UTR luciferase activity but not Klf9-CDS luciferase activity (Fig. 6E), suggesting that the m6A modification in 3′UTR is involved in the stabilization of Klf9 mRNA. Collectively, METTL3-dependent m6A modification in the 3′UTR mediates the stabilization of Klf9 mRNA, which is an essential prerequisite for KLF9 expression.

Pharmaceutical Activation of METTL3 Promotes Beiging

We proposed that activation of m6A methyltransferase activity using small-molecule chemicals may induce beiging. Intriguingly, a former study identified methyl piperidine-3-carboxylate (MP3C) as a small-molecule agonist targeting the METTL3-METTL14-WTAP complex (23). Indeed, we confirmed that intraperitoneal injection of MP3C increased the mRNA m6A methylation level of adipose tissue (Fig. 7A and B). The in vitro addition of MP3C increased the activity of METTL3 in a dose-dependent manner but had almost no effect on another SAM-dependent m6A methyltransferase, METTL16 (Fig. 7C). Mice treated with MP3C showed smaller lipid droplets, elevated UCP1 expression, and increased OCR in iWAT (Fig. 7D–F). The MP3C treatment significantly improved the O2 consumption, CO2 generation, and energy heat generation of mice (Fig. 7G–I). These data indicate that this agonist promotes beiging and thermogenesis.

Figure 7

METTL3 agonist promotes WAT beiging. A: Chemical structure of methyl piperidine-3-carboxylate. B: m6A level of mRNAs in iWAT from mice treated with or without MP3C (*P < 0.05; three independent experiments; two-tailed t test). C: Percentage of methylation as compared with the control (CTL) (no MP3C added). D: Hematoxylin-eosin (H-E) staining of iWAT from mice treated with or without MP3C. Scale bar, 50 μm. E: Immunoblot analysis of UCP1, KLF9, and METTL3 expression in iWAT from mice treated with or without MP3C. F: OCR of iWAT from mice treated with or without MP3C (*P < 0.05; three independent experiments; two-tailed t test). GI: O2 consumption (G), CO2 generation (H), and energy heat generation (I) of mice treated with or without MP3C (*P < 0.05, ***P < 0.001, ****P < 0.0001; n = 4 mice in each group; two-tailed t test). White and gray areas in the graphs indicate day and night, respectively.

Figure 7

METTL3 agonist promotes WAT beiging. A: Chemical structure of methyl piperidine-3-carboxylate. B: m6A level of mRNAs in iWAT from mice treated with or without MP3C (*P < 0.05; three independent experiments; two-tailed t test). C: Percentage of methylation as compared with the control (CTL) (no MP3C added). D: Hematoxylin-eosin (H-E) staining of iWAT from mice treated with or without MP3C. Scale bar, 50 μm. E: Immunoblot analysis of UCP1, KLF9, and METTL3 expression in iWAT from mice treated with or without MP3C. F: OCR of iWAT from mice treated with or without MP3C (*P < 0.05; three independent experiments; two-tailed t test). GI: O2 consumption (G), CO2 generation (H), and energy heat generation (I) of mice treated with or without MP3C (*P < 0.05, ***P < 0.001, ****P < 0.0001; n = 4 mice in each group; two-tailed t test). White and gray areas in the graphs indicate day and night, respectively.

Close modal

Pharmaceutical Activation of METTL3 Corrects Obesity-Associated Metabolic Defects

We proposed that MP3C might regulate obesity-associated metabolic dysfunction. To that end, we treated diet-induced obese mice with the agonist. Notably, MP3C hampered the body weight accumulation of HFD-fed mice, despite comparable daily food intake (Fig. 8A–C). The weight of iWAT from MP3C-treated mice was smaller than that from control mice, but the weight of BAT did not change (Fig. 8D and E). Strikingly, the MP3C-treated mice fed an HFD had more ameliorated glucose intolerance and insulin resistance than their control littermates (Fig. 8F and G). The MP3C treatment of obese mice significantly upregulated O2 consumption, CO2 generation, and energy heat generation (Fig. 8H–J). Thus, MP3C is considered a potential molecule targeting the m6A writer complex for curing metabolic diseases (Fig. 8K).

Figure 8

METTL3 agonist corrects metabolic disorders. A: Gross view of HFD-fed mice treated with or without MP3C. B: Body weight of HFD-fed mice treated with or without MP3C (*P < 0.05, ***P < 0.001; n = 7 mice in each group; two-tailed t test). C: Daily food intake by HFD-fed mice treated with or without MP3C (n = 7 mice in each group; two-tailed t test). D: Gross view of iWAT and BAT from mice as in panel A. Scale bar, 0.5 cm. E: Weight of iWAT and BAT from HFD-fed mice treated with or without MP3C (*P < 0.05; n = 7 mice in control [CTL] group and n = 4 mice in MP3C-treated group; two-tailed t test). F and G: Glucose (GTT) (F) and insulin tolerance test (ITT) (G) assays were performed in HFD-fed mice treated with or without MP3C (*P < 0.05, **P < 0.01; n = 7 mice in CTL group and n = 4 mice in MP3C-treated group; two-tailed t test). HJ: O2 consumption (H), CO2 generation (I), and energy heat generation (J) of HFD-fed mice treated with or without MP3C (*P < 0.05, **P < 0.01, ****P < 0.0001; n = 4 mice in each group; two-tailed t test). White and gray areas in the graphs indicate day and night, respectively. K: Proposed model illustrating the role of METTL3 in WAT beiging.

Figure 8

METTL3 agonist corrects metabolic disorders. A: Gross view of HFD-fed mice treated with or without MP3C. B: Body weight of HFD-fed mice treated with or without MP3C (*P < 0.05, ***P < 0.001; n = 7 mice in each group; two-tailed t test). C: Daily food intake by HFD-fed mice treated with or without MP3C (n = 7 mice in each group; two-tailed t test). D: Gross view of iWAT and BAT from mice as in panel A. Scale bar, 0.5 cm. E: Weight of iWAT and BAT from HFD-fed mice treated with or without MP3C (*P < 0.05; n = 7 mice in control [CTL] group and n = 4 mice in MP3C-treated group; two-tailed t test). F and G: Glucose (GTT) (F) and insulin tolerance test (ITT) (G) assays were performed in HFD-fed mice treated with or without MP3C (*P < 0.05, **P < 0.01; n = 7 mice in CTL group and n = 4 mice in MP3C-treated group; two-tailed t test). HJ: O2 consumption (H), CO2 generation (I), and energy heat generation (J) of HFD-fed mice treated with or without MP3C (*P < 0.05, **P < 0.01, ****P < 0.0001; n = 4 mice in each group; two-tailed t test). White and gray areas in the graphs indicate day and night, respectively. K: Proposed model illustrating the role of METTL3 in WAT beiging.

Close modal

A series of transcriptional and epigenetic regulators have been characterized in WAT beiging. In this study, we demonstrate that the m6A writer METTL3 is induced in beige adipocytes and required for WAT beiging. Moreover, METTL3-mediated m6A modification posttranscriptionally regulates the expression of thermogenic mRNAs. Importantly, the agonist of METTL3, methyl piperidine-3-carboxylate, shows great potential for promoting beiging and reversing obesity-associated disorders.

In our study, we used Adipoq-Cre for the deletion of the Mettl3 gene, which induces Mettl3 knockout in mature WAT and BAT, avoiding the possible effect on embryonic development. Using this knockout mouse, we demonstrated that in addition to BAT thermogenesis, METTL3 plays an essential role in WAT beiging under cold treatment. A recent study reported that UCP1-Cre-driven Mettl3 knockout inhibits BAT maturation and thermogenesis (18). It should be noted that UCP1 is activated in both brown and beige adipocytes. Therefore, the phenotype observed in UCP1-Cre-driven Mettl3 deletion mice may be attributed to the abnormality in both WAT and BAT. Moreover, we demonstrated that MP3C, an activator of METTL3-METTL14-WTAP, combats obesity-associated disorders in mice. The weight of WAT rather than BAT was reduced under MP3C treatment, implying that the effects of MP3C occur largely through beige adipose tissue, although the function of BAT is not excluded.

During WAT beiging, not only does the total m6A level increases, but the landscape of m6A methylome is also remodeled. Profiling of m6A revealed that most of the metabolism-related mRNAs were dynamically harbored with m6A modifications during the process of beiging. Loss of Mettl3 impaired proper m6A modification in metabolic mRNAs, leading to decreased mRNA levels. These findings point to a model in which mRNA transcription and m6A-regulated mRNA stabilization orchestrate a regulatory network required for beiging.

It has been reported that KLF9 is induced by cold treatment and β-adrenergic agonists in adipose tissue by an unknown mechanism (29). We demonstrate that the expression of KLF9 relies heavily on the stabilization of its mRNA by m6A modifications. Multiple m6A sites were identified in Klf9 mRNA, but only the m6A in the 3′UTR dynamically responded to cold treatment and Mettl3 deletion. Luciferase assays confirmed the regulatory effect of this m6A modification on Klf9 expression. Of note, the sequences flanking the functional m6A sites are conserved between human and mouse, implying an evolutionary principle for regulation. Overexpression of KLF9 rescues impaired beiging by Mettl3 depletion, indicating that KLF9 functionally mediates METTL3-promoted adipose thermogenesis. Consistent with our findings, adipose-specific Klf9 transgenic mice display enhanced energy expenditure and are resistant to HFD-induced obesity (29). Moreover, a genome-wide association study also indicated that common variants at the KLF9 gene are associated with BMI (30). These data indicate an essential role of KLF9 in the control of adipose energy homeostasis; however, the detailed mechanism requires further investigation.

The thermogenic activity of m6A provides an attractive therapeutic target for obesity and other metabolic disorders. Indeed, the FTO inhibitor entacapone, which increases total m6A level, shows great potential in activating thermogenesis in adipose tissue, reducing body weight, and lowering the blood glucose level in obese mice (19). Here, we report that methyl piperidine-3-carboxylate, a small-molecule agonist of the METTL3-METTL14-WTAP complex (23), can also promote the thermogenesis of adipose tissue. Mice treated with this agonist showed activation of beige adipose tissue, along with reduced body weight and improved glucose metabolism. The MP3C treatment significantly improved energy consumption and energy expenditure, including upregulated O2 consumption, CO2 generation, and energy heat generation. In conclusion, the results indicate that m6A has therapeutic potential in regulating metabolic disorders.

This article contains supplementary material online at https://doi.org/10.2337/figshare.23060858.

R.X., S.Y., and X.Z. contributed equally.

Acknowledgments. The authors are grateful to Dr. Ming-Han Tong (Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences) for providing Mettl3fl/fl mice and Dr. Chi Luo (Zhejiang University) for the critical reading of the manuscript.

Funding. This work was supported by grants from the National Natural Science Foundation of China (82100926 to S.W. and 82073110 and 81672847 to X.G.), the Natural Science Foundation of Zhejiang Province, China (LQ19H120012 to K.L.), and the Zhejiang Youth Talent Support Program (ZJWR0308085 to X.G.).

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. R.X. and S.Y. performed most experiments. R.X. and X.G. designed the experiments, performed the data analysis, and wrote the manuscript. X.Z., Y.G., Y.Q., J.H., Z.C., and K.L. assisted in the animal experiments. X.G. and S.W. conceived the project. All authors discussed the results and edited the manuscript. X.G. and S.W. are the guarantors of this work and, as such, had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

1.
Richard
AJ
,
White
U
,
Elks
CM
,
Stephens
JM
.
Adipose tissue: physiology to metabolic dysfunction
. In
Endotext
.
South Dartmouth, MA
,
MDText.com, Inc.
,
2000
, pp.
1
3
2.
Wu
J
,
Boström
P
,
Sparks
LM
, et al
.
Beige adipocytes are a distinct type of thermogenic fat cell in mouse and human
.
Cell
2012
;
150
:
366
376
3.
Shabalina
IG
,
Petrovic
N
,
de Jong
JM
,
Kalinovich
AV
,
Cannon
B
,
Nedergaard
J
.
UCP1 in brite/beige adipose tissue mitochondria is functionally thermogenic
.
Cell Rep
2013
;
5
:
1196
1203
4.
Harms
M
,
Seale
P
.
Brown and beige fat: development, function and therapeutic potential
.
Nat Med
2013
;
19
:
1252
1263
5.
Inagaki
T
,
Sakai
J
,
Kajimura
S
.
Transcriptional and epigenetic control of brown and beige adipose cell fate and function
.
Nat Rev Mol Cell Biol
2016
;
17
:
480
495
6.
Cohen
P
,
Levy
JD
,
Zhang
Y
, et al
.
Ablation of PRDM16 and beige adipose causes metabolic dysfunction and a subcutaneous to visceral fat switch
.
Cell
2014
;
156
:
304
316
7.
Fu
Y
,
Dominissini
D
,
Rechavi
G
,
He
C
.
Gene expression regulation mediated through reversible m6A RNA methylation
.
Nat Rev Genet
2014
;
15
:
293
306
8.
Bokar
JA
,
Shambaugh
ME
,
Polayes
D
,
Matera
AG
,
Rottman
FM
.
Purification and cDNA cloning of the AdoMet-binding subunit of the human mRNA (N6-adenosine)-methyltransferase
.
RNA
1997
;
3
:
1233
1247
9.
Liu
J
,
Yue
Y
,
Han
D
, et al
.
A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation
.
Nat Chem Biol
2014
;
10
:
93
95
10.
Ping
XL
,
Sun
BF
,
Wang
L
, et al
.
Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase
.
Cell Res
2014
;
24
:
177
189
11.
Jia
G
,
Fu
Y
,
Zhao
X
, et al
.
N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO
.
Nat Chem Biol
2011
;
7
:
885
887
12.
Zheng
G
,
Dahl
JA
,
Niu
Y
, et al
.
ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility
.
Mol Cell
2013
;
49
:
18
29
13.
Wang
X
,
Lu
Z
,
Gomez
A
, et al
.
N6-methyladenosine-dependent regulation of messenger RNA stability
.
Nature
2014
;
505
:
117
120
14.
Wang
X
,
Zhao
BS
,
Roundtree
IA
, et al
.
N(6)-methyladenosine modulates messenger RNA translation efficiency
.
Cell
2015
;
161
:
1388
1399
15.
Frayling
TM
,
Timpson
NJ
,
Weedon
MN
, et al
.
A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity
.
Science
2007
;
316
:
889
894
16.
Fischer
J
,
Koch
L
,
Emmerling
C
, et al
.
Inactivation of the Fto gene protects from obesity
.
Nature
2009
;
458
:
894
898
17.
Wu
R
,
Chen
Y
,
Liu
Y
, et al
.
m6A methylation promotes white-to-beige fat transition by facilitating Hif1a translation
.
EMBO Rep
2021
;
22
:
e52348
18.
Wang
Y
,
Gao
M
,
Zhu
F
, et al
.
METTL3 is essential for postnatal development of brown adipose tissue and energy expenditure in mice
.
Nat Commun
2020
;
11
:
1648
19.
Peng
S
,
Xiao
W
,
Ju
D
, et al
.
Identification of entacapone as a chemical inhibitor of FTO mediating metabolic regulation through FOXO1
.
Sci Transl Med
2019
;
11
:
eaau7116
20.
Zhu
Y
,
Gao
Y
,
Tao
C
, et al
.
Connexin 43 mediates white adipose tissue beiging by facilitating the propagation of sympathetic neuronal signals
.
Cell Metab
2016
;
24
:
420
433
21.
Hatting
M
,
Rines
AK
,
Luo
C
, et al
.
Adipose tissue CLK2 promotes energy expenditure during high-fat diet intermittent fasting
.
Cell Metab
2017
;
25
:
428
437
22.
Li
F
,
Kennedy
S
,
Hajian
T
, et al
.
A radioactivity-based assay for screening human m6A-RNA methyltransferase, METTL3-METTL14 complex, and demethylase ALKBH5
.
J Biomol Screen
2016
;
21
:
290
297
23.
Selberg
S
,
Blokhina
D
,
Aatonen
M
, et al
.
Discovery of small molecules that activate RNA methylation through cooperative binding to the METTL3-14-WTAP complex active site
.
Cell Rep
2019
;
26
:
3762
3771.e5
24.
Chen
S
,
Zhou
Y
,
Chen
Y
,
Gu
J
.
fastp: an ultra-fast all-in-one FASTQ preprocessor
.
Bioinformatics
2018
;
34
:
i884
i890
25.
Kim
D
,
Paggi
JM
,
Park
C
,
Bennett
C
,
Salzberg
SL
.
Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype
.
Nat Biotechnol
2019
;
37
:
907
915
26.
Zhang
Y
,
Liu
T
,
Meyer
CA
, et al
.
Model-based analysis of ChIP-Seq (MACS)
.
Genome Biol
2008
;
9
:
R137
27.
Dominissini
D
,
Moshitch-Moshkovitz
S
,
Schwartz
S
, et al
.
Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq
.
Nature
2012
;
485
:
201
206
28.
Meyer
KD
,
Saletore
Y
,
Zumbo
P
,
Elemento
O
,
Mason
CE
,
Jaffrey
SR
.
Comprehensive analysis of mRNA methylation reveals enrichment in 3′ UTRs and near stop codons
.
Cell
2012
;
149
:
1635
1646
29.
Fan
H
,
Zhang
Y
,
Zhang
J
, et al
.
Cold-inducible Klf9 regulates thermogenesis of brown and beige fat
.
Diabetes
2020
;
69
:
2603
2618
30.
Okada
Y
,
Kubo
M
,
Ohmiya
H
, et al.;
GIANT Consortium
.
Common variants at CDKAL1 and KLF9 are associated with body mass index in east Asian populations
.
Nat Genet
2012
;
44
:
302
306
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/journals/pages/license.