Glucose Regulates m6A Methylation of RNA in Pancreatic Islets

Type 2 diabetes is characterized by chronic hyperglycemia associated with impaired insulin action and secretion. Although the heritability of type 2 diabetes is high, the environment, including blood components, could play a major role in the development of the disease. Amongst environmental effects, epitranscriptomic modifications have been recently shown to affect gene expression and glucose homeostasis. The epitranscriptome is characterized by reversible chemical changes in RNA, with one of the most prevalent being the m6A methylation of RNA. Since pancreatic β cells fine tune glucose levels and play a major role in type 2 diabetes physiopathology, we hypothesized that the environment, through variations in blood glucose or blood free fatty acid concentrations, could induce changes in m6A methylation of RNAs in pancreatic β cells. Here we observe a significant decrease in m6A methylation upon high glucose concentration, both in mice and human islets, associated with altered expression levels of m6A demethylases. In addition, the use of siRNA and/or specific inhibitors against selected m6A enzymes demonstrate that these enzymes modulate the expression of genes involved in pancreatic β-cell identity and glucose-stimulated insulin secretion. Our data suggest that environmental variations, such as glucose, control m6A methylation in pancreatic β cells, playing a key role in the control of gene expression and pancreatic β-cell functions. Our results highlight novel causes and new mechanisms potentially involved in type 2 diabetes physiopathology and may contribute to a better understanding of the etiology of this disease.

[1]  Wei Huang,et al.  Glucose Is Involved in the Dynamic Regulation of m6A in Patients With Type 2 Diabetes , 2018, The Journal of clinical endocrinology and metabolism.

[2]  Roger D. Cox,et al.  Overexpression of Fto leads to increased food intake and results in obesity , 2010, Nature Genetics.

[3]  Arne Klungland,et al.  A majority of m6A residues are in the last exons, allowing the potential for 3′ UTR regulation , 2015, Genes & development.

[4]  O. Elemento,et al.  Comprehensive Analysis of mRNA Methylation Reveals Enrichment in 3′ UTRs and near Stop Codons , 2012, Cell.

[5]  Julian J. Emmanuel,et al.  A link between FTO, ghrelin, and impaired brain food-cue responsivity. , 2013, The Journal of clinical investigation.

[6]  Miao Yu,et al.  A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation , 2013, Nature chemical biology.

[7]  Erez Y. Levanon,et al.  m6A mRNA methylation facilitates resolution of naïve pluripotency toward differentiation , 2015, Science.

[8]  Chuan He,et al.  YTHDC1 mediates nuclear export of N6-methyladenosine methylated mRNAs , 2017, eLife.

[9]  Chengqi Yi,et al.  N6-Methyladenosine in Nuclear RNA is a Major Substrate of the Obesity-Associated FTO , 2011, Nature chemical biology.

[10]  R. Kulkarni,et al.  Epigenetic modifiers of islet function and mass , 2014, Trends in Endocrinology & Metabolism.

[11]  Yang Shi,et al.  Zc3h13 Regulates Nuclear RNA m6A Methylation and Mouse Embryonic Stem Cell Self-Renewal. , 2018, Molecular cell.

[12]  Ran Elkon,et al.  Transcription Impacts the Efficiency of mRNA Translation via Co-transcriptional N6-adenosine Methylation , 2017, Cell.

[13]  Roger D. Cox,et al.  A Mouse Model for the Metabolic Effects of the Human Fat Mass and Obesity Associated FTO Gene , 2009, PLoS genetics.

[14]  L. Sussel,et al.  Epigenetic modifications and long noncoding RNAs influence pancreas development and function. , 2015, Trends in genetics : TIG.

[15]  Rajashekar Varma Kadumuri,et al.  Epitranscriptomic Code and Its Alterations in Human Disease. , 2018, Trends in molecular medicine.

[16]  R. Scharfmann,et al.  Expression and functional assessment of candidate type 2 diabetes susceptibility genes identify four new genes contributing to human insulin secretion , 2017, Molecular metabolism.

[17]  Wei Huang,et al.  Decreased N(6)-methyladenosine in peripheral blood RNA from diabetic patients is associated with FTO expression rather than ALKBH5. , 2015, The Journal of clinical endocrinology and metabolism.

[18]  Samir Adhikari,et al.  Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase , 2014, Cell Research.

[19]  Arne Klungland,et al.  ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. , 2013, Molecular cell.

[20]  S. Kwak,et al.  10-year trajectory of β-cell function and insulin sensitivity in the development of type 2 diabetes: a community-based prospective cohort study. , 2016, The lancet. Diabetes & endocrinology.

[21]  Miao Yu,et al.  A METTL 3-METTL 14 complex mediates mammalian nuclear RNA N 6-adenosine methylation , 2016 .

[22]  U. Boggi,et al.  Persistent or Transient Human β Cell Dysfunction Induced by Metabolic Stress: Specific Signatures and Shared Gene Expression with Type 2 Diabetes. , 2020, Cell reports.

[23]  P. Froguel,et al.  Cofactors As Metabolic Sensors Driving Cell Adaptation in Physiology and Disease , 2017, Front. Endocrinol..

[24]  I. Santin,et al.  A novel RT-QPCR-based assay for the relative quantification of residue specific m6A RNA methylation , 2019, Scientific Reports.

[25]  H. Jun,et al.  Fatty Acid-Induced Lipotoxicity in Pancreatic Beta-Cells During Development of Type 2 Diabetes , 2018, Front. Endocrinol..

[26]  M. Kupiec,et al.  Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq , 2012, Nature.

[27]  D R Hadden,et al.  Beta‐cell deterioration determines the onset and rate of progression of secondary dietary failure in Type 2 diabetes mellitus: the 10‐year follow‐up of the Belfast Diet Study , 1998, Diabetic medicine : a journal of the British Diabetic Association.

[28]  Beverley Balkau,et al.  Variation in FTO contributes to childhood obesity and severe adult obesity , 2007, Nature Genetics.

[29]  R. Rodriguez,et al.  Unravelling the genomic targets of small molecules using high-throughput sequencing , 2014, Nature Reviews Genetics.

[30]  J. Auwerx,et al.  Transcriptional coregulators: fine-tuning metabolism. , 2014, Cell metabolism.

[31]  Yue Huang,et al.  Targeting FTO Suppresses Cancer Stem Cell Maintenance and Immune Evasion. , 2020, Cancer cell.

[32]  Decheng Ren,et al.  Acute Deletion of METTL14 in β-cells of adult mice results in glucose intolerance. , 2019, Endocrinology.

[33]  Zhike Lu,et al.  m6A-dependent regulation of messenger RNA stability , 2013, Nature.

[34]  M. Cnop,et al.  Recent insights into mechanisms of β-cell lipo- and glucolipotoxicity in type 2 diabetes. , 2020, Journal of molecular biology.

[35]  Samie R. Jaffrey,et al.  m6A RNA methylation promotes XIST-mediated transcriptional repression , 2016, Nature.

[36]  Gang Xiao,et al.  Histone H3 trimethylation at lysine 36 guides m6A RNA modification co-transcriptionally , 2019, Nature.

[37]  Christopher E. Mason,et al.  Single-nucleotide resolution mapping of m6A and m6Am throughout the transcriptome , 2015, Nature Methods.

[38]  Chuan He,et al.  m6A mRNA Methylation Regulates Human β-Cell Biology in Physiological States and in Type 2 Diabetes , 2019, Nature Metabolism.

[39]  F. Pattou,et al.  Upgrading Pretransplant Human Islet Culture Technology Requires Human Serum Combined With Media Renewal , 2010, Transplantation.

[40]  Schraga Schwartz,et al.  Perturbation of m6A writers reveals two distinct classes of mRNA methylation at internal and 5' sites. , 2014, Cell reports.

[41]  A. Mooradian,et al.  Dyslipidemia in Type 2 Diabetes: Prevalence, Pathophysiology, and Management , 2013, Drugs.

[42]  Minoru Yoshida,et al.  RNA-Methylation-Dependent RNA Processing Controls the Speed of the Circadian Clock , 2013, Cell.

[43]  Samir Adhikari,et al.  Nuclear m(6)A Reader YTHDC1 Regulates mRNA Splicing. , 2016, Molecular cell.

[44]  Nian Liu,et al.  N 6-methyladenosine alters RNA structure to regulate binding of a low-complexity protein , 2017, Nucleic acids research.

[45]  J. Auwerx,et al.  KAT2B Is Required for Pancreatic Beta Cell Adaptation to Metabolic Stress by Controlling the Unfolded Protein Response. , 2016, Cell reports.

[46]  Chuan He,et al.  N 6 -methyladenosine Modulates Messenger RNA Translation Efficiency , 2015, Cell.

[47]  Joshua D. Jones,et al.  A molecular‐level perspective on the frequency, distribution, and consequences of messenger RNA modifications , 2020, Wiley interdisciplinary reviews. RNA.

[48]  H. S. Hundal,et al.  Chronic Effects of Palmitate Overload on Nutrient-Induced Insulin Secretion and Autocrine Signalling in Pancreatic MIN6 Beta Cells , 2011, PloS one.

[49]  D. R. Laybutt,et al.  Failure of the Adaptive Unfolded Protein Response in Islets of Obese Mice Is Linked With Abnormalities in β-Cell Gene Expression and Progression to Diabetes , 2013, Diabetes.

[50]  Weiqing Wang,et al.  m6A mRNA Methylation Controls Functional Maturation in Neonatal Murine β-Cells , 2020, Diabetes.

[51]  Chuan He,et al.  Post-transcriptional gene regulation by mRNA modifications , 2016, Nature Reviews Molecular Cell Biology.

[52]  A. Volchuk,et al.  Chronic palmitate but not oleate exposure induces endoplasmic reticulum stress, which may contribute to INS-1 pancreatic beta-cell apoptosis. , 2006, Endocrinology.

[53]  P. Froguel,et al.  Cdkn2a deficiency promotes adipose tissue browning , 2017, Molecular metabolism.

[54]  Chuan He,et al.  METTL14 is essential for β-cell survival and insulin secretion. , 2019, Biochimica et biophysica acta. Molecular basis of disease.

[55]  Douglas L Black,et al.  m6A mRNA modifications are deposited in nascent pre-mRNA and are not required for splicing but do specify cytoplasmic turnover , 2017, Genes & development.

[56]  S. Bonner-Weir,et al.  Five stages of evolving beta-cell dysfunction during progression to diabetes. , 2004, Diabetes.

[57]  Jun Liu,et al.  VIRMA mediates preferential m6A mRNA methylation in 3′UTR and near stop codon and associates with alternative polyadenylation , 2018, Cell Discovery.