m6A-mRNA methylation regulates cardiac gene expression and cellular growth

Vivien Kmietczyk, Eva Riechert, Laura Kalinski, Etienne Boileau, Ellen Malovrh, Brandon Malone, Agnieszka Gorska, Christoph Hofmann, Eshita Varma, Lonny Jürgensen, Verena Kamuf-Schenk, Janine Altmüller, Rewati Tappu, Martin Busch, Patrick Most, Hugo A Katus, Christoph Dieterich, Mirko Völkers, Vivien Kmietczyk, Eva Riechert, Laura Kalinski, Etienne Boileau, Ellen Malovrh, Brandon Malone, Agnieszka Gorska, Christoph Hofmann, Eshita Varma, Lonny Jürgensen, Verena Kamuf-Schenk, Janine Altmüller, Rewati Tappu, Martin Busch, Patrick Most, Hugo A Katus, Christoph Dieterich, Mirko Völkers

Abstract

Conceptually similar to modifications of DNA, mRNAs undergo chemical modifications, which can affect their activity, localization, and stability. The most prevalent internal modification in mRNA is the methylation of adenosine at the N6-position (m6A). This returns mRNA to a role as a central hub of information within the cell, serving as an information carrier, modifier, and attenuator for many biological processes. Still, the precise role of internal mRNA modifications such as m6A in human and murine-dilated cardiac tissue remains unknown. Transcriptome-wide mapping of m6A in mRNA allowed us to catalog m6A targets in human and murine hearts. Increased m6A methylation was found in human cardiomyopathy. Knockdown and overexpression of the m6A writer enzyme Mettl3 affected cell size and cellular remodeling both in vitro and in vivo. Our data suggest that mRNA methylation is highly dynamic in cardiomyocytes undergoing stress and that changes in the mRNA methylome regulate translational efficiency by affecting transcript stability. Once elucidated, manipulations of methylation of specific m6A sites could be a powerful approach to prevent worsening of cardiac function.

Conflict of interest statement

The authors declare that they have no conflict of interest.

© 2019 Kmietczyk et al.

Figures

Figure S1.. Validation of m 6 A-IPs.
Figure S1.. Validation of m6A-IPs.
(A) Enrichment of m6A mRNA after m6A-IP shown by m6A mRNA dot blot of input RNA, flow through/supernatant, and RNA immunoprecipitated (IP) with m6A antibody (m6A Ab) or IgG antibody (IgG Ab) as control. (B) Enrichment of m6A mRNA after m6A-IP shown by qPCR on Nde1 and Drd1, previously shown to be unmethylated or methylated, respectively. (C) Immunoblot for Mettl3 in human DCM samples showing a trend to increased Mettl3 expression. + denotes a positive control sample (Hela cell lysate). (D) mRNA expression of m6A machinery in healthy and DCM cardiac samples analyzed by qPCR. P < 0.05 by one-tailed t test (n = 3 per group). (E) Enrichment of DCM-specific GDPG and GJD3 or control-specific ZNF324, HNRPHR3, and HSPA1A m6A mRNA after m6A-IP shown by qPCR (n = 3, **P < 0.005, ***P < 0.0005 by one-way ANOVA). (F) Gene ontology (GO) term enrichment analysis for the 1,595 DCM-specific methylated mRNAs for molecular functions.
Figure 1.. The m6A mRNA methylome differs…
Figure 1.. The m6A mRNA methylome differs between healthy and failing human heart tissue.
(A) Percentage of m6A in mRNA from DCM and control samples measured by m6A ELISA (n = 6 control and n = 10 DCM, *P = 0.0476 by t test). (B) mRNA expression of m6A machinery in healthy and DCM cardiac samples analyzed by normalized read counts from RNA-seq from myocardial biopsies (n = 33 DCM and n = 24 control). (C) Venn diagram showing overlap of healthy and DCM m6A mRNA methylome analyzed by sequencing of m6A immunoprecipitated mRNA. (D) Gene Ontology (GO) term enrichment analysis for biological processes on the 1,595 DCM-specific methylated mRNAs (high m6A) and mRNAs not enriched in IP (low m6A). The width of the bars represents the significance (–log10 (adjusted P-value, Fisher’s exact test)) of the respective GO term enrichment. (E) Volcano plot of sequencing of m6A immunoprecipitated mRNA data. Significant enriched transcripts are shown in green. Transcripts involved in transcription (GO term: transcription and DNA-templated are shown in red). Examples of enriched transcripts in DCM hearts are indicated. (F) IGV plots of sequencing reads after m6A-IP for DCM-specific methylated GJD3 transcript and control-specific ZNF324B transcript.
Figure S2.. Gain and loss of function…
Figure S2.. Gain and loss of function of Mettl3 in in vitro and in vivo lead to changes cell size and gene expression changes.
(A) Left: immunofluorescence of neonatal (top) and adult rat cardiomyocytes (bottom); actin antibody (red), Mettl3 (green), and nuclei (blue). Right: immunofluorescence of neonatal (top) and adult rat cardiomyocytes (bottom); actin antibody (red), Fto (green), and nuclei (blue). Bar graph: 20 μM. (B) Knockdown of Mettl3 and Fto in NRVMs demonstrated by immunoblot. (C) Relative m6A content (n-fold change) in mRNA in NRVMs after Mettl3 or Fto knockdown by siRNA measured by m6A ELISA (n = 3, P < 0.05 by one-way ANOVA). (D) Cell surface of NRVMs with Mettl3 and Fto knockdown compared with siScr with and without PE for 24 h stimulation (n = 3, ****P < 0.0001 by one way ANOVA). (E) mRNA expression of Nppa and Nppb in NRVMs after Mettl3 or Fto KD compared with siScr with and without PE stimulation (n = 3, **P < 0.005, ***P < 0.0005 by one-way ANOVA). (F) Overexpression of Mettl3 and enzymatically inactive mutant in NRVMs in neonatal cardiomycytes by adenoviral infection; actin antibody (red), flag (green), and nuclei (blue). Bar graph: 20 μM. (G) Cardiac overexpression of Mettl3 in mice demonstrated by immunoblot (top) and immunofluorescence (bottom); actin antibody (red), Mettl3 antibody (green), and nuclei (blue). Bar graph: 40 μM. (H) Percentage of m6A in mRNA in NRVMs after Mettl3 overexpression in vivo measured by m6A ELISA (n = 3). *P < 0.05 by t test. (I) mRNA expression of Nppa and Nppb of either control virus or Mettl3-injected mouse hearts after either sham or TAC surgery in 10-wk-old mice (n = 6–14,*P < 0.05, **P < 0.005).
Figure 2.. m 6 A affects cell…
Figure 2.. m6A affects cell growth and cardiac function.
(A) Percentage of m6A in mRNA in NRVMs after Mettl3 or Mettl3 enzymatically inactive mutant overexpression measured by m6A ELISA (n = 7 for control and n = 8 for Mettl3 WT and Mettl3 mutant). ****P < 0.0001 by one-way ANOVA followed by Bonferroni's post hoc comparisons. (B) Cell surface of NRVMs overexpressing Mettl3 or inactive mutant compared with control virus with and without PE stimulation (n = 3 independent biological experiments with >69 cells for each group *P < 0.0047, ****P < 0.0001 by one-way ANOVA). (C) Gross morphology of either control virus or Mettl3-injected mouse hearts after either sham or TAC surgery in 10-wk-old mice. (D) Ratio of LV weight to tibia length of control and Mettl3 OE mice (n = 10–12, ***P < 0.001, ****P < 0.0001 by one-way ANOVA). (E) Immunofluorescence of heart sections WGA (green), sarcomeric actin (red), and nuclei blue. Bar graph: 20 μM. (F) Cell surface area measurement from WGA staining (n = 4 animals per group and 200–300 cells in total, *P < 0.05, ****P < 0.0001 by one-way ANOVA). (G) Masson trichrome staining on paraffin heart sections. Bar graph: 80 μM. (H) Quantification of fibrotic area from Masson trichrome staining (n = 4–10, *P < 0.05 by one-way ANOVA). (I) mRNA expression of Col1a1 analyzed by qPCR (n = 6–14, ****P < 0.0001 by one-way ANOVA).
Figure S3.. Decrease of m6a levels in…
Figure S3.. Decrease of m6a levels in murine hearts after TAC surgery is associated with differential regulation of translation.
(A) Percentage of m6A in mRNA in mice hearts 2 d after TAC or sham surgery (n = 5–6, *P < 0.05 by t test). (B) Venn diagram showing overlap between m6A-containing transcripts between sham- and TAC-operated mice 2 d after surgery. (C) RT-PCR and immunoblots for Mettl3 in mice after sham or 2 d post TAC surgery. n = 3–4, *P < 0.05 by t test). (D) Enrichment of Gene Ontology (GO) terms in the group of m6A-enriched genes (sham conditions) for biological processes. Colors depict the log fold change (logFC) of individual genes within the GO category and numbers behind the bars correspond to the number of genes within the GO category. (E) Cumulative fraction of mRNAs relative to their fold change of RNA-seq and mRNAs relative to their fold change of Ribo-seq between all transcripts and methylated transcripts 2 d after TAC surgery (Kolmogorov–Smirnov test P < 0.01). (F) GO term enrichment analysis for the TAC-specific methylated mRNAs for biological processes and cellular compartments. (G) Overexpression of Mettl3 in HL-1 cells shown by immunoblotting. (H) List of top translationally regulated transcripts by Mettl3 overexpression in HL-1 cells. (I) Enrichment of m6A-mRNA after m6A-IP shown by qPCR of Arhgef3 and Myl2 after different time points post TAC surgery. <0.05 by t test. n = 3.
Figure 3.. m 6 A affects translation…
Figure 3.. m6A affects translation efficiency.
(A) Scatter blot of fold change of RNA-seq and Ribo-seq (sham/TAC) from murine hearts 2 d after TAC surgery. All transcripts (gray), enriched in m6A-IP (blue). (B) Box plots of fold change (sham/TAC) of Ribo-seq and RNA-seq from murine hearts 2 d after TAC surgery for methylated transcripts in sham-operated mice (sham only m6A) or post TAC surgery (TAC only m6A). *P < 0.05 by t test. (C) Volcano blot of Ribo-seq data from HL-1 cells overexpressing Mettl3. (D) GO term enrichment analysis (biological process) for Mettl3 target mRNAs, which are identified as regulated in Ribo-seq. The width of the bars represents the significance (–log10 (adjusted P-value, Fisher’s exact test)) of the respective GO term enrichment. Colors depict the log fold change (logFC) of individual genes within the GO category. (E) Cumulative fraction of mRNAs relative to their fold change of Ribo-seq and RNA-seq (sham/TAC) between all transcripts and Mettl3-regulated transcripts 2 d after TAC surgery (Kolmogorov–Smirnov test, P < 0.001).
Figure 4.. m 6 A affects expression…
Figure 4.. m6A affects expression of target genes by regulating transcript stability.
(A) Polysome profile of control and Mettl3-overexpressing cardiomyocytes (HL-1 cells). (B) RT-PCR (left panel) and Ribo-seq counts (right panel) for Myl2 and Arhgef3 in HL-1 cells (*P < 0.05 after EdgeR analysis for the Ribo-seq quantification, n = 3). (C) Percentage of total transcript abundance of Arhgef3 and Myl2 in mRNPs, 80s, and polyribosomes (*P < 0.05 by t test; n = 2 independent experiments). (D) Enrichment of m6A-mRNA after m6A-IP shown by qPCR of Arhgef3 and Myl2 after Mettl3 overexpression. *P < 0.01 by t test; n = 3 independent experiments. (E) mRNA stability of Mettl3 targets Arhgef3, Polr3d, and Myl2 analyzed by qPCR upon actinomycin D treatment. (F, G) Immunoblots and (G) quantification for Arhgef3 and Myl2 in HL-1 cells after confirming the Mettl3 overexpression. P < 0.05 by t test; n = 3 independent experiments. (H, I) Immunoblots and (I) quantification for Arhgef3 and Myl2 in mice after sham or TAC surgery cells after Mettl3 overexpression. *P < 0.05 by one-way ANOVA (n = 3 for each group; SE, short exposure; LE, long exposure).

References

    1. Alarcón CR, Lee H, Goodarzi H, Halberg N, Tavazoie SF (2015) N6-methyladenosine marks primary microRNAs for processing. Nature 519: 482–485. 10.1038/nature14281
    1. Boissel S, Reish O, Proulx K, Kawagoe-Takaki H, Sedgwick B, Yeo GSH, Meyre D, Golzio C, Molinari F, Kadhom N, et al. (2009) Loss-of-function mutation in the dioxygenase-encoding FTO gene causes severe growth retardation and multiple malformations. Am J Hum Genet 85: 106–111. 10.1016/j.ajhg.2009.06.002
    1. Bui AL, Horwich TB, Fonarow GC (2011) 2006 National Hospital Discharge Survey. Nat Rev Cardiol 8: 1–20. 10.1038/nrcardio.2010.165
    1. Choi J, Ieong KW, Demirci H, Chen J, Petrov A, Prabhakar A, O’Leary SE, Dominissini D, Rechavi G, Soltis SM, et al. (2016) N6-methyladenosine in mRNA disrupts tRNA selection and translation-elongation dynamics. Nat Struct Mol Biol 23: 110–115. 10.1038/nsmb.3148
    1. Claycomb WC, Lanson NA, Stallworth BS, Egeland DB, Delcarpio JB, Bahinski A, Izzo NJ (1998) HL-1 cells: A cardiac muscle cell line that contracts and retains phenotypic characteristics of the adult cardiomyocyte. Proc Natl Acad Sci USA 95: 2979–2984. 10.1073/pnas.95.6.2979
    1. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras TR (2013) STAR: Ultrafast universal RNA-seq aligner [supplementary data]. Bioinformatics 29: 15–21. 10.1093/bioinformatics/bts635 Available at:
    1. Dodt M, Roehr J, Ahmed R, Dieterich C (2012) FLEXBAR—flexible barcode and adapter processing for next-generation sequencing platforms. Biology (Basel) 1: 895–905. 10.3390/biology1030895. Available at:
    1. Dominissini D, Moshitch-Moshkovitz S, Schwartz S, Salmon-Divon M, Ungar L, Osenberg S, Cesarkas K, Jacob-Hirsch J, Amariglio N, Kupiec M, et al. (2012) Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature 485: 201–206. 10.1038/nature11112
    1. Dorn LE, Lasman L, Chen J, Xu X, van Berlo JH, Accornero F (2018) The m 6 A mRNA methylase METTL3 controls cardiac homeostasis and hypertrophy. Circulation 139: 533–545. 10.1161/CIRCULATIONAHA.118.036146
    1. Doroudgar S, Völkers M, Thuerauf DJ, Khan M, Mohsin S, Respress JL, Wang W, Gude N, Müller OJ, Wehrens XHT, et al. (2015) Hrd1 and ER-associated protein degradation, ERAD, are critical elements of the adaptive ER stress response in cardiac myocytes. Circ Res 117: 536–546. 10.1161/circresaha.115.306993
    1. Eimre M, Paju K, Pelloux S, Beraud N, Roosimaa M, Kadaja L, Gruno M, Peet N, Orlova E, Remmelkoor R, et al. (2008) Distinct organization of energy metabolism in HL-1 cardiac cell line and cardiomyocytes. Biochim Biophys Acta 1777: 514–524. 10.1016/j.bbabio.2008.03.019 Available at: . Accessed February 1, 2019.
    1. Fu Y, Dominissini D, Rechavi G, He C (2014) Gene expression regulation mediated through reversible m6A RNA methylation. Nat Rev Genet 15: 293–306. 10.1038/nrg3724
    1. Geula S, Moshitch-Moshkovitz S, Dominissini D, Mansour AAF, Kol N, Salmon-Divon M, Hershkovitz V, Peer E, Mor N, Manor YS, et al. (2015) m6A mRNA methylation facilitates resolution of naïve pluripotency toward differentiation. Science 347: 1002–1006. 10.1126/science.1261417
    1. Gilbert WV, Bell TA, Schaening C (2016) Messenger RNA modifications: Form, distribution, and function. Science 352: 1408–1412. 10.1126/science.aad8711
    1. Gulati P, Avezov E, Ma M, Antrobus R, Lehner P, O’Rahilly S, Yeo GSH (2014) Fat mass and obesity-related (FTO) shuttles between the nucleus and cytoplasm. Biosci Rep 34: 621–628. 10.1042/bsr20140111
    1. Han P, Li W, Lin CH, Yang J, Shang C, Nurnberg ST, Jin KK, Xu W, Lin CY, Lin CJ, et al. (2014) A long noncoding RNA protects the heart from pathological hypertrophy. Nature 514: 102–106. 10.1038/nature13596
    1. He C. (2010) Grand challenge commentary: RNA epigenetics? Nat Chem Biol 6: 863–865. 10.1038/nchembio.482 Available at: . Accessed January 31, 2019.
    1. Ingolia NT. (2016) Ribosome footprint profiling of translation throughout the genome. Cell 165: 22–33. 10.1016/j.cell.2016.02.066
    1. Jia G, Fu Y, Zhao X, Dai Q, Zheng G, Yang Y, Yi C, Lindahl T, Pan T, Yang YG, et al. (2011) N6-Methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat Chem Biol 7: 885–887. 10.1038/nchembio.687
    1. Khanna N, Fang Y, Yoon MS, Chen J (2013) XPLN is an endogenous inhibitor of mTORC2. Proc Natl Acad Sci U S A 110: 15979–15984. 10.1073/pnas.1310434110
    1. Koitabashi N, Kass DA (2012) Reverse remodeling in heart failure-mechanisms and therapeutic opportunities. Nat Rev Cardiol 9: 147–157. 10.1038/nrcardio.2011.172
    1. Liu J, Yue Y, Han D, Wang X, Fu Y, Zhang L, Jia G, Yu M, Lu Z, Deng X, et al. (2014) A METTL3–METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol 10: 93–95. 10.1038/nchembio.1432 Available at: . Accessed February 5, 2019.
    1. Malone B, Atanassov I, Aeschimann F, Li X, Großhans H, Dieterich C (2017) Bayesian prediction of RNA translation from ribosome profiling. Nucleic Acids Res 45: 2960–2972. 10.1093/nar/gkw1350
    1. Mathiyalagan P, Adamiak M, Mayourian J, Sassi Y, Liang Y, Agarwal N, Jha D, Zhang S, Kohlbrenner E, Chepurko E, et al. (2018) FTO-dependent m6A regulates cardiac function during remodeling and repair. Circulation 118: 33794 10.1161/CIRCULATIONAHA.118.033794
    1. Mauer J, Luo X, Blanjoie A, Jiao X, Grozhik AV, Patil DP, Linder B, Pickering BF, Vasseur JJ, Chen Q, et al. (2017) Reversible methylation of m6Am in the 5′ cap controls mRNA stability. Nature 541: 371–375. 10.1038/nature21022
    1. McCarthy DJ, Chen Y, Smyth GK (2012) Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic Acids Res 40: 4288–4297. 10.1093/nar/gks042
    1. Meder B, Haas J, Sedaghat-Hamedani F, Kayvanpour E, Frese K, Lai A, Nietsch R, Scheiner C, Mester S, Bordalo DM, et al. (2017) Epigenome-wide association study identifies cardiac gene patterning and a novel class of biomarkers for heart failure. Circulation 136: 1528–1544. 10.1161/circulationaha.117.027355
    1. Meyer KD, Jaffrey SR (2014) The dynamic epitranscriptome: N6-methyladenosine and gene expression control. Nat Rev Mol Cell Biol 15: 313–326. 10.1038/nrm3785
    1. Meyer KD, Patil DP, Zhou J, Zinoviev A, Skabkin MA, Elemento O, Pestova TV, Qian SB, Jaffrey SR (2015) 5′ UTR m6A promotes cap-independent translation. Cell 163: 999–1010. 10.1016/j.cell.2015.10.012
    1. Meyer KD, Saletore Y, Zumbo P, Elemento O, Mason CE, Jaffrey SR (2012) Comprehensive analysis of mRNA methylation reveals enrichment in 3′ UTRs and near stop codons. Cell 149: 1635–1646. 10.1016/j.cell.2012.05.003
    1. Molinie B, Wang J, Lim KS, Hillebrand R, Lu ZX, Van Wittenberghe N, Howard BD, Daneshvar K, Mullen AC, Dedon P, et al. (2016) M6A-LAIC-seq reveals the census and complexity of the m6A epitranscriptome. Nat Methods 13: 692–698. 10.1038/nmeth.3898 Available at: . Accessed October 23, 2018.
    1. Most P, Pleger ST, Völkers M, Heidt B, Boerries M, Weichenhan D, Löffler E, Janssen PML, Eckhart AD, Martini J, et al. (2004) Cardiac adenoviral S100A1 gene delivery rescues failing myocardium. J Clin Invest 114: 1550–1563. 10.1172/jci200421454
    1. Mudd JO, Kass DA (2008) Tackling heart failure in the twenty-first century. Nature 451: 919–928. 10.1038/nature06798
    1. Papait R, Greco C, Kunderfranco P, Latronico MVG, Condorelli G (2013) Epigenetics: A new mechanism of regulation of heart failure? Basic Res Cardiol 108: 361 10.1007/s00395-013-0361-1 Available at: .
    1. Rockman HA, Ross RS, Harris AN, Knowlton KU, Steinhelpert ME, Fieldt LJ, Ross J, Chien KR, Steinhelper ME, Field LJ (1991) Segregation of atrial-specific and inducible expression of an atrial natriuretic factor transgene in an in vivo murine model of cardiac hypertrophy. Proc Natl Acad Sci USA 88: 8277–8281. 10.1073/pnas.88.18.8277
    1. Sanz E, Yang L, Su T, Morris DR, McKnight GS, Amieux PS (2009) Cell-type-specific isolation of ribosome-associated mRNA from complex tissues. Proc Natl Acad Sci USA 106: 13939–13944. 10.1073/pnas.0907143106
    1. Schafer S, Adami E, Heinig M, Rodrigues KEC, Kreuchwig F, Silhavy J, Van Heesch S, Simaite D, Rajewsky N, Cuppen E, et al. (2015) Translational regulation shapes the molecular landscape of complex disease phenotypes. Nat Commun 6: 7200 10.1038/ncomms8200
    1. Schöller E, Weichmann F, Treiber T, Ringle S, Treiber N, Flatley A, Feederle R, Bruckmann A, Meister G (2018) Interactions, localization, and phosphorylation of the m6A generating METTL3–METTL14–WTAP complex. RNA 24: 499–512. 10.1261/rna.064063.117
    1. Sciarretta S, Forte M, Frati G, Sadoshima J (2018) New insights into the role of mtor signaling in the cardiovascular system. Circ Res 122: 489–505. 10.1161/circresaha.117.311147
    1. Sheikh F, Lyon RC, Chen J (2015) Functions of myosin light chain-2 (MYL2) in cardiac muscle and disease. Gene 569: 14–20. 10.1016/j.gene.2015.06.027 Available at: . Accessed February 4, 2019.
    1. Shi H, Wang X, Lu Z, Zhao BS, Ma H, Hsu PJ, Liu C, He C (2017) YTHDF3 facilitates translation and decay of N6-methyladenosine-modified RNA. Cell Res 27: 315–328. 10.1038/cr.2017.15
    1. Shi H, Zhang X, Weng YL, Lu Z, Liu Y, Lu Z, Li J, Hao P, Zhang Y, Zhang F, et al. (2018) m6A facilitates hippocampus-dependent learning and memory through YTHDF1. Nature 563: 249–253. 10.1038/s41586-018-0666-1
    1. Siede D, Rapti K, Gorska AA, Katus HA, Altmüller J, Boeckel JN, Meder B, Maack C, Völkers M, Müller OJ, et al. (2017) Identification of circular RNAs with host gene-independent expression in human model systems for cardiac differentiation and disease. J Mol Cell Cardiol 109: 48–56. 10.1016/j.yjmcc.2017.06.015
    1. Slobodin B, Han R, Calderone V, Vrielink JAFO, Loayza-Puch F, Elkon R, Agami R (2017) Transcription impacts the efficiency of mRNA translation via co-transcriptional N6-adenosine methylation. Cell 169: 326–337.e12. 10.1016/j.cell.2017.03.031
    1. Tan FL, Moravec CS, Li J, Apperson-Hansen C, McCarthy PM, Young JB, Bond M (2002) The gene expression fingerprint of human heart failure. Proc Natl Acad Sci USA 99: 11387–11392. 10.1073/pnas.162370099
    1. Völkers M, Doroudgar S, Nguyen N, Konstandin MH, Quijada P, Din S, Ornelas L, Thuerauf DJ, Gude N, Friedrich K, et al. (2014) PRAS40 prevents development of diabetic cardiomyopathy and improves hepatic insulin sensitivity in obesity. EMBO Mol Med 6: 57–65. 10.1002/emmm.201303183
    1. Völkers M, Konstandin MH, Doroudgar S, Toko H, Quijada P, Din S, Joyo A, Ornelas L, Samse K, Thuerauf DJ, et al. (2013) Mechanistic target of rapamycin complex 2 protects the heart from ischemic damage. Circulation 128: 2132–2144. 10.1161/circulationaha.113.003638
    1. Volkers M, Toko H, Doroudgar S, Din S, Quijada P, Joyo AY, Ornelas L, Joyo E, Thuerauf DJ, Konstandin MH, et al. (2013) Pathological hypertrophy amelioration by PRAS40-mediated inhibition of mTORC1. Proc Natl Acad Sci USA 110: 12661–12666. 10.1073/pnas.1301455110
    1. Vu LP, Pickering BF, Cheng Y, Zaccara S, Nguyen D, Minuesa G, Chou T, Chow A, Saletore Y, Mackay M, et al. (2017) The N6 -methyladenosine (m6A)-forming enzyme METTL3 controls myeloid differentiation of normal hematopoietic and leukemia cells. Nat Med 23: 1369–1376. 10.1038/nm.4416
    1. Wang X, Zhao BS, Roundtree IA, Lu Z, Han D, Ma H, Weng X, Chen K, Shi H, He C (2015) N6-methyladenosine modulates messenger RNA translation efficiency. Cell 161: 1388–1399. 10.1016/j.cell.2015.05.014 Available at: . Accessed February 4, 2019.
    1. Wei J, Liu F, Lu Z, Fei Q, Ai Y, He PC, Shi H, Cui X, Su R, Klungland A, et al. (2018) Differential m6A, m6Am, and m1A demethylation mediated by FTO in the cell nucleus and cytoplasm. Mol Cell 71: 973–985.e5. 10.1016/j.molcel.2018.08.011

Source: PubMed

Подписаться