Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

NADP modulates RNA m6A methylation and adipogenesis via enhancing FTO activity

An Author Correction to this article was published on 01 September 2023

This article has been updated

Abstract

Metabolism is often regulated by the transcription and translation of RNA. In turn, it is likely that some metabolites regulate enzymes controlling reversible RNA modification, such as N6-methyladenosine (m6A), to modulate RNA. This hypothesis is at least partially supported by the findings that multiple metabolic diseases are highly associated with fat mass and obesity-associated protein (FTO), an m6A demethylase. However, knowledge about whether and which metabolites directly regulate m6A remains elusive. Here, we show that NADP directly binds FTO, independently increases FTO activity, and promotes RNA m6A demethylation and adipogenesis. We screened a set of metabolites using a fluorescence quenching assay and NADP was identified to remarkably bind FTO. In vitro demethylation assays indicated that NADP enhances FTO activity. Furthermore, NADP regulated mRNA m6A via FTO in vivo, and deletion of FTO blocked NADP-enhanced adipogenesis in 3T3-L1 preadipocytes. These results build a direct link between metabolism and RNA m6A demethylation.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: MARTFQ screening reveals that NADP directly binds FTO.
Fig. 2: NADP increases FTO activity in vitro.
Fig. 3: Intracellular NADP modulates RNA m6A methylation through FTO.
Fig. 4: The NADP/FTO axis increases adipogenesis in 3T3-L1 cells.
Fig. 5: NADP modulates RNA m6A methylation in vivo through FTO.

Similar content being viewed by others

Data availability

All sequencing data have been deposited to NCBI under accession no. PRJNA546061. Source Data are provided with this paper.

Change history

References

  1. Wu, J., Ocampo, A. & Belmonte, J. C. I. Cellular metabolism and induced pluripotency. Cell 166, 1371–1385 (2016).

    Article  CAS  PubMed  Google Scholar 

  2. Schulze, A. & Harris, A. L. How cancer metabolism is tuned for proliferation and vulnerable to disruption. Nature 491, 364–373 (2012).

    Article  CAS  PubMed  Google Scholar 

  3. Semenza, G. L. Hypoxia-inducible factors in physiology and medicine. Cell 148, 399–408 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Serganov, A. & Patel, D. J. Ribozymes, riboswitches and beyond: regulation of gene expression without proteins. Nat. Rev. Genet. 8, 776–790 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Thorleifsson, G. et al. Genome-wide association yields new sequence variants at seven loci that associate with measures of obesity. Nat. Genet. 41, 18–24 (2009).

    Article  CAS  PubMed  Google Scholar 

  6. Frayling, T. M. Genome-wide association studies provide new insights into type 2 diabetes aetiology. Nat. Rev. Genet. 8, 657–662 (2007).

    Article  CAS  PubMed  Google Scholar 

  7. Dominissini, D. et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature 485, 201–206 (2012).

    Article  CAS  PubMed  Google Scholar 

  8. Shi, H., Wei, J. & He, C. Where, when and how: context-dependent functions of RNA methylation writers, readers and erasers. Mol. Cell 74, 640–650 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Nilsen, T. W. Molecular biology. Internal mRNA methylation finally finds functions. Science 343, 1207–1208 (2014).

    Article  CAS  PubMed  Google Scholar 

  10. Jia, G. et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat. Chem. Biol. 7, 885–887 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Zheng, G. et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol. Cell 49, 18–29 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Fu, Y. et al. FTO-mediated formation of N6-hydroxymethyladenosine and N6-formyladenosine in mammalian RNA. Nat. Commun. 4, 1798 (2013).

    Article  PubMed  Google Scholar 

  13. Gerken, T. et al. The obesity-associated FTO gene encodes a 2-oxoglutarate-dependent nucleic acid demethylase. Science 318, 1469–1472 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Xiao, W., Wang, R. S., Handy, D. E. & Loscalzo, J. NAD(H) and NADP(H) redox couples and cellular energy metabolism. Antioxid. Redox Signal. 28, 251–272 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Lu, W., Wang, L., Chen, L., Hui, S. & Rabinowitz, J. D. Extraction and quantitation of nicotinamide adenine dinucleotide redox cofactors. Antioxid. Redox Signal. 28, 167–179 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Nakahata, Y., Sahar, S., Astarita, G., Kaluzova, M. & Sassone-Corsi, P. Circadian control of the NAD+ salvage pathway by CLOCK-SIRT1. Science 324, 654–657 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Gorres, K. L. & Raines, R. T. Prolyl 4-hydroxylase. Crit. Rev. Biochem. Mol. Biol. 45, 106–124 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Yin, R. et al. Ascorbic acid enhances Tet-mediated 5-methylcytosine oxidation and promotes DNA demethylation in mammals. J. Am. Chem. Soc. 135, 10396–10403 (2013).

    Article  CAS  PubMed  Google Scholar 

  19. Lomenick, B. et al. Target identification using drug affinity responsive target stability (DARTS). Proc. Natl Acad. Sci. USA 106, 21984–21989 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Martinez Molina, D. et al. Monitoring drug target engagement in cells and tissues using the cellular thermal shift assay. Science 341, 84–87 (2013).

    Article  PubMed  Google Scholar 

  21. Han, Z. et al. Crystal structure of the FTO protein reveals basis for its substrate specificity. Nature 464, 1205–1209 (2010).

    Article  CAS  PubMed  Google Scholar 

  22. Tao, R. et al. Genetically encoded fluorescent sensors reveal dynamic regulation of NADPH metabolism. Nat. Methods 14, 720–728 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Canto, C. et al. The NAD+ precursor nicotinamide riboside enhances oxidative metabolism and protects against high-fat diet-induced obesity. Cell Metab. 15, 838–847 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Chowdhry, S. et al. NAD metabolic dependency in cancer is shaped by gene amplification and enhancer remodelling. Nature 569, 570–575 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Jackson, R. M., Griesel, B. A., Gurley, J. M., Szweda, L. I. & Olson, A. L. Glucose availability controls adipogenesis in mouse 3T3-L1 adipocytes via up-regulation of nicotinamide metabolism. J. Biol. Chem. 292, 18556–18564 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Hoxhaj, G. et al. Direct stimulation of NADP+ synthesis through Akt-mediated phosphorylation of NAD kinase. Science 363, 1088–1092 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Tian, W. N. et al. Importance of glucose-6-phosphate dehydrogenase activity for cell growth. J. Biol. Chem. 273, 10609–10617 (1998).

    Article  CAS  PubMed  Google Scholar 

  28. Poulain, L. et al. High mTORC1 activity drives glycolysis addiction and sensitivity to G6PD inhibition in acute myeloid leukemia cells. Leukemia 31, 2326–2335 (2017).

    Article  CAS  PubMed  Google Scholar 

  29. Jiang, P. et al. p53 regulates biosynthesis through direct inactivation of glucose-6-phosphate dehydrogenase. Nat. Cell Biol. 13, 310–316 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Du, W. et al. TAp73 enhances the pentose phosphate pathway and supports cell proliferation. Nat. Cell Biol. 15, 991–1000 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Huang, Y. et al. Small-molecule targeting of oncogenic FTO demethylase in acute myeloid leukemia. Cancer Cell 35, 677–691 e610 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Peng, S. et al. Identification of entacapone as a chemical inhibitor of FTO mediating metabolic regulation through FOXO1. Sci. Transl. Med. 11, eaau7116 (2019).

    Article  PubMed  Google Scholar 

  33. Liu, J. et al. N6-methyladenosine of chromosome-associated regulatory RNA regulates chromatin state and transcription. Science 367, 580–586 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zhao, X. et al. FTO-dependent demethylation of N6-methyladenosine regulates mRNA splicing and is required for adipogenesis. Cell Res. 24, 1403–1419 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Fischer, J. et al. Inactivation of the Fto gene protects from obesity. Nature 458, 894–898 (2009).

    Article  CAS  PubMed  Google Scholar 

  36. Lian, C. G. et al. Loss of 5-hydroxymethylcytosine is an epigenetic hallmark of melanoma. Cell 150, 1135–1146 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Vissers, M. C., Bozonet, S. M., Pearson, J. F. & Braithwaite, L. J. Dietary ascorbate intake affects steady state tissue concentrations in vitamin C-deficient mice: tissue deficiency after suboptimal intake and superior bioavailability from a food source (kiwifruit). Am. J. Clin. Nutr. 93, 292–301 (2011).

    Article  CAS  PubMed  Google Scholar 

  38. Yang, Q. et al. Pharmacological inhibition of BMK1 suppresses tumor growth through promyelocytic leukemia protein. Cancer Cell 18, 258–267 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Song, C. et al. PML recruits TET2 to regulate DNA modification and cell proliferation in response to chemotherapeutic agent. Cancer Res. 78, 2475–2489 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Huang, Y. et al. Meclofenamic acid selectively inhibits FTO demethylation of m6A over ALKBH5. Nucleic Acids Res. 43, 373–384 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. Sulkowska, A. Interaction of drugs with bovine and human serum albumin. J. Mol. Struct. 614, 227–232 (2002).

    Article  CAS  Google Scholar 

  42. Zhu, Y. et al. LC-MS-MS quantitative analysis reveals the association between FTO and DNA methylation. PLoS ONE 12, e0175849 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Dominissini, D., Moshitch-Moshkovitz, S., Salmon-Divon, M., Amariglio, N. & Rechavi, G. Transcriptome-wide mapping of N6-methyladenosine by m6A-seq based on immunocapturing and massively parallel sequencing. Nat. Protoc. 8, 176–189 (2013).

    Article  CAS  PubMed  Google Scholar 

  44. Cui, X. et al. MeTDiff: a novel differential RNA methylation analysis for MeRIP-Seq Data. IEEE/ACM Trans. Comput. Biol. Bioinform. 15, 526–534 (2018).

    CAS  Google Scholar 

  45. Wang, X. et al. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature 505, 117–120 (2014).

    Article  PubMed  Google Scholar 

  46. Ryu, K. W. et al. Metabolic regulation of transcription through compartmentalized NAD. biosynthesis. Science 360, eaan5780 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Church, C. D., Berry, R. & Rodeheffer, M. S. Isolation and study of adipocyte precursors. Methods Enzymol. 537, 31–46 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Nadeem, A. et al. Glucose-6-phosphate dehydrogenase inhibition attenuates acute lung injury through reduction in NADPH oxidase-derived reactive oxygen species. Clin. Exp. Immunol. 191, 279–287 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank C. He at the University of Chicago for providing information for the expression and purification of FTO and ALKBH5. This study was supported by grants from the National Natural Science Foundation of China (NSFC nos. 81872310 and 81622040 to Q.Y.). This study was also supported by LiaoNing Revitalization Talents Program (XLYC1802058 to Q.Y.).

Author information

Authors and Affiliations

Authors

Contributions

Q.Y. and S.-C.Y. designed the experiments and wrote the paper. L.W., C.S. and N.W. performed most experiments. C.S. performed bioinformatics analyses. N.W. and S.L. raised the mice and performed the Oil red O staining and triglyceride assays. Q.L. expressed and purified the FTO and ALKBH5 proteins. N.W. and Z.S. maintained cells and performed immunoblotting analysis. K.W. constructed the plasmids.

Corresponding authors

Correspondence to Shi-Cang Yu or Qingkai Yang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 MARTFQ screening reveals that NADP directly binds FTO.

a, Schematic showing MARTFQ assay. b, 3D structure of FTO protein containing tryptophan residues. The data for the FTO 3D structure were derived from NCBI accession code: 3LFM (Protein Data Bank). Red dots represent tryptophan residues in FTO protein. c, Coomassie staining of in vitro purified FTO protein. d, 3D structure of FTO. The data of FTO 3D structure were derived from accession code: 3LFM (Protein Data Bank). N terminal: blue; C terminal: red; 2KG: green. e, Coomassie staining of in vitro purified 6XHis-FTO-NT and 6XHis-FTO-CT proteins. f, Quenching of the fluorescence of FTO-NT (left) and FTO-CT (right) proteins by NADPH at different molar ratios. g, The modified Stern–Volmer curves for estimating the K values of FTO-NT (18.5 x 103 M-1) and FTO-CT (30.2 x 103 M-1). h, Immunoblotting analyses of the cell lysates from HEK293 cells transfected with pCDNA3 Flag-FTO (WT), pCI-Neo Flag-FTO-NT or pCI-Neo Flag-FTO-CT plasmid. i,j, DARTS (i) and CETSA (j) analyses of the abilities of NADPH to bind FTO-NT and FTO-CT in HEK293 cells transfected with pCI-Neo Flag-FTO-NT or pCI-Neo Flag-FTO-CT plasmid. Immunoblotting was performed using anti-Flag antibody.

Source data

Extended Data Fig. 2 NADP increases the FTO activity in vitro.

a, Schematic showing the in vitro m6A demethylation assay. b, Base ion mass transitions of A and m6A in standard compounds and mRNAs at different collision energies. c, Kinetic curves of FTO in the presence of 2 mM Vc. d, MS analyses of m6A in the post-reaction mixtures as described in Fig. 2a. e,f, Dot blotting analyses of m6A in the FTO post-reaction mixtures as described in Fig. 2a (e) and Fig. 2b (f).

Extended Data Fig. 3 NADP was not stoichiometrically consumed in the reaction catalyzed by FTO.

a, MS analyses of the compounds as noted in the post-reaction mixtures containing compound as noted. NADPH, NADP, NADH, NAD or Vc at 10 µM was separately added into the mixtures (10 µM ssRNA, 0.5 µM FTO, 300 µM Fe2+ and 300 µM 2KG). Boiled FTO was used as the negative control. Resultant mixtures were incubated at 37 °C for 90 min without heatin. b, Demethylation of m6A in the FTO reaction mixtures containing varied concentration of Vc or DHA for 10 min. c, Demethylation of m6A in the FTO reaction mixtures incubated for varied times in the presence of 0.5 mM of Vc or DHA. d, Coomassie staining of in vitro purified ALKBH5 protein. e, Kinetic curves of m6A ssRNA for ALKBH5 in the presence of 2 mM Vc. f, MS analyses of m6A in the ALKBH5 post-reaction mixtures described in Fig. 2c. g, Demethylation of m6A in the ALKBH5 reaction mixtures containing high concentrations of the cofactor as noted.

Extended Data Fig. 4 Intracellular NADP modulates RNA m6A methylation through FTO.

a,b, Total concentration (a) and reduced/oxidized ratio (b) of NADP in the cells. c,d, Total concentration (c) and reduced/oxidized ratio (d) of NAD in the cells. e, The NADP and NAD levels in the cells treated with 300 µM NADPH, NADP+, NADH or NAD+ for 4 hrs. f,g, Total concentration and reduced/oxidized ratio of NADP and NAD in the fourth fat pads (f) and livers (g) of mice treated with HFD for 6 hrs or IP injection of glucose for 4 hrs. h, The NADP and NAD levels in liver of mice described in (g). i, The Vc levels in the cells described in Fig. 3c. j,k, Dot blot analyses of DNA 5hmdC (j) and RNA m6A (k) levels in the cells treated with 300 µM Vc or NADPH for 4 hrs.

Source data

Extended Data Fig. 5 NADP modulates RNA m6A methylation via FTO.

a, Schematic showing the synthesis of NADP and NAD. b, Protein levels in HEK293 cells treated as described in Fig. 3e. c, The NADP and NAD levels in the cells treated as described in Fig. 3e. d, Protein levels in HEK293 cells treated as described in Fig. 3g. e, The NADP and NAD levels in the cells treated as described in Fig. 3g. f, The NADP and NAD levels in the cells treated as described in Fig. 3i. g, The m6A levels in the cells treated with NADPH and/or 6AN for 6 hrs. h, Protein levels in the cells treated with siRNA as noted. i, The m6A levels in HEK293 cells treated as described in Fig. 3j.

Source data

Extended Data Fig. 6 Identification of the NADP\FTO-regulated m6A methylated genes by anti-m6A MeRIP-seq.

a, Schematic showing anti-m6A MeRIP-seq of control (Ctrl) and knockdown (KD) 3T3-L1 cells treated with 6AN for 8 hrs or NADPH for 4 hrs. b,c, Volcano plots of p value versus FC of 6AN/Mock (b) and NADPH/Mock (c) in Ctrl and KD cells. Red plot: substantially upregulated peak (SUP) with p < 0.05 and FC > 1.5; green plot: substantially downregulated peak (SDP) with p < 0.05 and FC < 0.66. d,e, Volcano plots of p value versus FC in CDS, 3’UTR and 5’UTR regions of 6AN/Mock (d) and NADPH/Mock (e) in Ctrl and KD cells. f, Ratio of SUP number versus SDP number in Ctrl and KD cells treated with 6AN or NADPH. g, Ratio of SUP/SDP in CDS and 5’UTR. h, The distribution curves of treatment/Mock peaks in Ctrl and KD cells.

Extended Data Fig. 7 Enrichment and validation analyses of NFRM genes.

a, Venn diagram of 265 NFRM genes showing the overlap of the significant peaks in Ctrl and KD cells treated with 6AN or NADPH. b, Protein-protein interaction network analysis of NFRM genes. The molecular complex detection (MCODE) algorithm was applied to identify densely connected network components using Metascape (metascape.org). Identified MCODE networks are gathered and shown by Cytoscape (www.cytoscape.org). c, Top 5 protein complexes with significant p value of enrichment. d,e, Reactome (d) and GO (e) enrichment analyses of NFRM genes. f, The normalized ΔCt levels of 17 selected genes in 3T3-L1 cells treated with Mock, FB23-2 (FTO inhibitor), NADPH or NADPH+FB23-2 for 4 hrs. The ΔCt values were normalized to Gapdh. g,h, The qRT-PCR analyses of anti-Fto PAR-CLIP (g) and anti-m6A MeRIP (h) RNAs. The RNA level in IgG IP was taken as 1. i,j, The relative RNA levels in 3T3-L1 cells treated with actinomycin D for 3 (i) or 6 (j) hrs. 3T3-L1 cells were treated with Mock, FB23-2, NADPH or NADPH+FB23-2 for 4 hrs. Then actinomycin D was applied to stop transcription. The RNA level in the Mock cells treated with actinomycin D for 0 h was taken as 1.

Source data

Extended Data Fig. 8 NADP\FTO axis increases adipogenesis in 3T3-L1 cells.

a,b, Oil Red O staining of 3T3-L1 cells on day 6 (a) and day 8 (D8) (b). 3T3-L1 cells were treated with siRNAi against control, Fto, Alkbh5 or Mettl3 on day -2 (D-2). After 2 days, 3T3-L1 preadipocytes were induced to differentiate by MDI cocktail on D0. Resultant cells were stained by Oil Red O. Scale bar = 100 μm. c, The NADP and NAD levels in control and Fto knockdown 3T3-L1 cells treated with NADPH. d, Oil Red O staining and triglyceride analyses of 3T3-L1 cells pretreated with or without siFto were treated with 300 μM of NADPH or NADP+ prior to induction. e, Oil Red O staining and triglyceride analyses of 3T3-L1 cells treated with NADPH, NADP+, and/or FB23-2 (20 μM) prior to induction. f, Schematic showing the treatment of FTO inhibitor for the time as noted. g, Oil Red O staining and triglyceride analyses of 3T3-L1 cells treated as described in (f). h, Oil Red O staining and triglyceride analyses of 3T3-L1 cells treated with 6AN, NADPH and/or FB23-2 for 6 hrs prior to induction.

Source data

Extended Data Fig. 9 The impact of NADP for adipogenesis is dependent on the catalytic activity of FTO.

a, The NADP and NAD levels in control and Fto knockdown 3T3-L1 cells treated with 0.5 μM insulin and/or 500 μM 6AN for 6 hrs. b, The m6A levels in control and Fto knockdown 3T3-L1 cells treated with insulin and/or 6AN for 6 hrs. c, The m6A levels in 3T3-L1 cells treated with FB23-2, insulin and/or 6AN as noted for 6 hrs. d, Oil Red O staining and triglyceride analyses of 3T3-L1 cells treated with FB23-2, insulin and/or 6AN prior to induction. e, Immunoblotting analyses of the cell lysates from mouse Fto knockdown 3T3-L1 cells infected with adenovirus encoding human wild type (Ad FTO-WT) or catalytic inactive mutant (Ad FTO-Mut) FTO. f, The m6A levels in control and Fto knockdown 3T3-L1 cells treated with adenoviral particles and NADPH. The 3T3-L1 cells were treated with siRNAs and adenoviruses as noted. The resultant cells were treated with NADPH for 6 hrs. g,h, Oil Red O staining and triglyceride analyses of 3T3-L1 cells treated with siRNAs, adenoviral particles, NADPH (g) or insulin (h) as noted.

Source data

Extended Data Fig. 10 NADP modulates RNA m6A methylation in vivo through FTO.

a, PCR analyses of the genomic DNA samples from WT and KO mice using the primers as described in Supplementary Table 1. b, Representative images of WT and KO mice at different ages. c, Body weights of male WT and KO mice at different ages. WT mice, n = 14; KO mice, n = 11. d, Haematoxylin and eosin staining of white adipose tissue (WAT) of WT and KO mice treated with HFD for 12 weeks. e, The total concentrations of Vc, NAD and NADP in the livers and fourth fat pads of mice. n = 6 mice. f-h, The Vc (f), NAD(P) (g) and 5mdC (h) levels in the fat pads described in Fig. 5g. i-m, The Vc (i), NAD(P) (j), 5hmdC (k), 5mdC (l) and m6A (m) levels in the liver as described in Fig. 5g.

Source data

Supplementary information

Supplementary Information

Supplementary Fig. 1, Tables 1–7 and unprocessed western blots for Supplementary Fig. 1.

Reporting Summary

Supplementary Data 1

Statistical source data for Supplementary Fig. 1.

Source data

Source Data Fig. 1

Unprocessed western blots and/or gels.

Source Data Fig. 3

Unprocessed western blots and/or gels.

Source Data Fig. 3

statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Unprocessed western blots and/or gels.

Source Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 5

statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 9

statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, L., Song, C., Wang, N. et al. NADP modulates RNA m6A methylation and adipogenesis via enhancing FTO activity. Nat Chem Biol 16, 1394–1402 (2020). https://doi.org/10.1038/s41589-020-0601-2

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-020-0601-2

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing