Skip to main page content
U.S. flag

An official website of the United States government

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
[Preprint]. 2023 Nov 30:2023.02.04.527050.
doi: 10.1101/2023.02.04.527050.

Differential CpG methylation at Nnat in the early establishment of beta cell heterogeneity

Affiliations

Differential CpG methylation at Nnat in the early establishment of beta cell heterogeneity

Vanessa Yu et al. bioRxiv. .

Update in

  • Differential CpG methylation at Nnat in the early establishment of beta cell heterogeneity.
    Yu V, Yong F, Marta A, Khadayate S, Osakwe A, Bhattacharya S, Varghese SS, Chabosseau P, Tabibi SM, Chen K, Georgiadou E, Parveen N, Suleiman M, Stamoulis Z, Marselli L, De Luca C, Tesi M, Ostinelli G, Delgadillo-Silva L, Wu X, Hatanaka Y, Montoya A, Elliott J, Patel B, Demchenko N, Whilding C, Hajkova P, Shliaha P, Kramer H, Ali Y, Marchetti P, Sladek R, Dhawan S, Withers DJ, Rutter GA, Millership SJ. Yu V, et al. Diabetologia. 2024 Jun;67(6):1079-1094. doi: 10.1007/s00125-024-06123-6. Epub 2024 Mar 21. Diabetologia. 2024. PMID: 38512414 Free PMC article.

Abstract

Aims/hypothesis: Beta cells within the pancreatic islet represent a heterogenous population wherein individual sub-groups of cells make distinct contributions to the overall control of insulin secretion. These include a subpopulation of highly-connected 'hub' cells, important for the propagation of intercellular Ca2+ waves. Functional subpopulations have also been demonstrated in human beta cells, with an altered subtype distribution apparent in type 2 diabetes. At present, the molecular mechanisms through which beta cell hierarchy is established are poorly understood. Changes at the level of the epigenome provide one such possibility which we explore here by focussing on the imprinted gene neuronatin (Nnat), which is required for normal insulin synthesis and secretion.

Methods: Single cell RNA-seq datasets were examined using Seurat 4.0 and ClusterProfiler running under R. Transgenic mice expressing eGFP under the control of the Nnat enhancer/promoter regions were generated for fluorescence-activated cell (FAC) sorting of beta cells and downstream analysis of CpG methylation by bisulphite and RNA sequencing, respectively. Animals deleted for the de novo methyltransferase, DNMT3A from the pancreatic progenitor stage were used to explore control of promoter methylation. Proteomics was performed using affinity purification mass spectrometry and Ca2+ dynamics explored by rapid confocal imaging of Cal-520 and Cal-590. Insulin secretion was measured using Homogeneous Time Resolved Fluorescence Imaging.

Results: Nnat mRNA was differentially expressed in a discrete beta cell population in a developmental stage- and DNA methylation (DNMT3A)-dependent manner. Thus, pseudo-time analysis of embryonic data sets demonstrated the early establishment of Nnat-positive and negative subpopulations during embryogenesis. NNAT expression is also restricted to a subset of beta cells across the human islet that is maintained throughout adult life. NNAT+ beta cells also displayed a discrete transcriptome at adult stages, representing a sub-population specialised for insulin production, reminiscent of recently-described "βHI" cells and were diminished in db/db mice. 'Hub' cells were less abundant in the NNAT+ population, consistent with epigenetic control of this functional specialization.

Conclusions/interpretation: These findings demonstrate that differential DNA methylation at Nnat represents a novel means through which beta cell heterogeneity is established during development. We therefore hypothesise that changes in methylation at this locus may thus contribute to a loss of beta cell hierarchy and connectivity, potentially contributing to defective insulin secretion in some forms of diabetes.

Keywords: Beta cell development; Ca2+; CpG methylation; connectivity; heterogeneity; identity; imprinted genes; insulin; islet; neuronatin; type 2 diabetes.

PubMed Disclaimer

Figures

Fig. 1.
Fig. 1.. Nnat expression is a marker for late-stage beta cell differentiation during islet development.
(a-c) UMAP projection of scRNA-seq data from embryonic mouse islets (E12.5-E17.5), with cells labelled by their cellular state (a), pseudo-time (b), and corresponding Nnat expression (c). (d) Changes in the distribution of beta cell precursors from E12.5 to E17.5. (e, f) Violin plots showing the fluctuations in Ins1 (e) and Nnat (f) expression from E12.5 to adulthood in the beta cell development trajectory.
Fig. 2.
Fig. 2.. Transcriptomic analysis of islet cells at E17.5 reveal Nnat+ cells to be a more differentiated beta cell subcluster.
(a, b) Islet cells plotted in the UMAP space and coloured by Ins1 (a) and Nnat (b) expression, respectively. Individual cellular clusters are labelled. Beta cell clusters are highlighted with black circles. (c) Distribution of Nnat expression among beta cell clusters, along with the expression range used in defining Nnat+ beta cells (highlighted by the red rectangle). (d) Volcano plot showing the log2 fold change in gene expression between Nnat+ and Nnat beta cells against FDR-corrected P values. Genes with FDR < 0.1 are coloured red. Top 10 upregulated and downregulated genes are labelled by gene name. (e) Geneset enrichment analysis (GSEA) of differentially expressed genes between Nnat+ and Nnat beta cells (using GO-BP terms), showing the top 10 upregulated and downregulated processes. The normalized enrichment score (NES) is plotted along the X-axis. Circle diameters are proportional to the number of genes in each process, with the colours defining statistical significance.
Fig. 3.
Fig. 3.. A subpopulation of NNAT+ beta cells develop during the early postnatal period in mice.
(a) Representative confocal microscopy of pancreatic cryosections from wild type mice on a C57BL/6J background of developmental stages from embryonic day 17.5 (E17.5) through the postnatal (P) period into adulthood. Sections were immunostained with antibodies against endogenous neuronatin (NNAT, green) and insulin (INS, red). Nuclei are visualised with DAPI and sections from P56 mice with constituent deletion of Nnat were used as an immunostaining control. Scale bar = 100μm (inset = 10μm) (n = 4-15 mice per timepoint, Kruskal-Wallace test with Dunn’s multiple comparisons). (b) Quantification of NNAT+ beta cells from images shown in a, expressed as NNAT/INS co-positive cells as a percentage of total INS-positive cells. (c) Representative confocal microscopy of pancreatic cryosections as in a from E17.5 and postnatal (P) day 14 mice (n = 18 and n = 5 mice, respectively) immunostained with antibodies against endogenous neuronatin (NNAT, green) and GCG or SST (both grey). Scale bar = 100μm. Representative images from three independent experiments and breeding pairs. * P < 0.05, ** P < 0.01, *** P < 0.001.
Fig. 4.
Fig. 4.. NNAT is expressed in a subset of human beta cells and NNAT deficiency in human beta cells reduces insulin secretion.
(a) GSIS analysis at low (3 mM) and high (16.7 mM) glucose in human EndoC-βH1 beta cells following 72h lentiviral-mediated shRNA knockdown of neuronatin with data expressed as of insulin secreted into culture media as a percentage of total cellular insulin content (n = 8 independent cultures per group, two-way ANOVA with Sidak’s multiple corrections test). (b, c) RT-PCR (n = 8, unpaired Student’s t test) and Western blot (n = 4, Mann-Whitney U test) analysis of neuronatin expression in EndoC-βH1 beta cells after transient NNAT silencing as in a. Western blotting analysis of NNAT protein levels shown via a representative blot of two independent experiments. β-tubulin was used as a loading control. Mean values for band intensities in multiple experiments quantified by densitometry are shown below the panel, expressed relative to scramble (SCR) shRNA controls. (d) Representative confocal microscopy of human pancreatic cryosections from younger (15.6 ± 0.9 years, n = 5) and older (71.0 ± 3.9 years, n = 4) donors. Sections were immunostained with antibodies against endogenous neuronatin (NNAT, green), insulin (INS, red) and glucagon (GCG, grey). Donor sex and age are indicated in the text. Nuclei are visualised with DAPI. Scale bar = 100μm. (e, f) Quantification of NNAT beta (e) and alpha (f) cells from images shown in d, expressed as NNAT/INS or NNAT/GCG co-positive cells as a percentage of total INS-positive or GCG-positive cells. * P < 0.05, *** P < 0.001, **** P < 0.0001).
Fig. 5.
Fig. 5.. Beta cell heterogeneity of NNAT expression is associated with changes in CpG methylation at the Nnat promoter.
(a) Representative confocal microscopy of pancreatic cryosections from P56 mice with Nnat-driven EGFP expression from the paternal allele (NnatWT/eGFPpat) (n = 7 mice). Sections were immunostained with antibodies against EGFP (green) and insulin (INS, grey). Nuclei are visualised with DAPI. Scale bar = 50μm. (b) Separation of dispersed primary islet cells by FACS from reporter mice with insulin-driven expression of tdTomato (to label beta cells) and Nnat-driven EGFP expression from the paternal (NnatWT/eGFPpat), or maternal (NnatWT/eGFPmat) allele or wild type (NnatWT/WT) at this locus (representative image of one dispersed islet preparation, each from a single mouse) (n = 8, 3 and 3 mice, respectively, Kruskal-Wallace test with Dunn’s multiple comparisons). (c) Quantification of data in b expressed as percentage of EGFP/tdTomato co-positive primary islet cells. (d) Representative bisulphite analysis of CpG methylation at the Nnat promoter in FACS-purified islet cell populations from b (n = 3 NnatWT/eGFPpat mice with paternally expressed Nnat-driven EGFP, n = >12 clones each). Closed circles = methylated CpG, open circles = unmethylated CpG. (e, f) Quantification of data in d expressed as percentage CpG methylation across the Nnat promoter at individual CpGs (e) and across the entire Nnat promoter (f) (paired Student’s t test). (g) Schematic summarising level of CpG methylation at the Nnat promoter and gametic DMR in NNAT+ vs NNAT beta cells. Representative images from three independent experiments and breeding pairs. * P < 0.05, ** P < 0.01.
Fig. 6.
Fig. 6.. Postnatal restriction of NNAT in a subset of beta cells is at least partially driven by the de novo methyltransferase, DNMT3A.
(a) Representative confocal microscopy of pancreatic cryosections from mice with conditional deletion of DNMT3A under the control of the Pdx1 promoter (Pdx1-Cre+ Dnmt3afl/fl) vs control (Pdx1-Cre Dnmt3afl/fl) mice at postnatal (P) day 6. Sections were immunostained with antibodies against endogenous neuronatin (NNAT, green) and insulin (INS, red). (b) Quantification of NNAT+ beta cells from images shown in a, expressed as NNAT/INS co-positive cells as a percentage of total INS-positive cells. Scale bar = 50μm (n = 8 mice per genotype, unpaired Students t test, ** P < 0.01). (c) Representative confocal microscopy of pancreatic cryosections from P56 (8 week old) wild type mice on a C57BL/6J background immunostained with antibodies against endogenous neuronatin (NNAT, green) and H3K27me3 (red). Scale bar = 100μm, n = 3 mice. Nuclei are visualised with DAPI. Representative images from three independent experiments and breeding pairs.
Fig. 7.
Fig. 7.. NNAT+ adult beta cells are transcriptionally distinct and have significantly higher insulin content.
(a) Correlation matrix of differentially expressed genes between NNAT+ and NNAT beta cells as assessed by RNA-sequencing analysis (n = 4 FACS-purified populations from individual mouse islet preparations). (b) Heatmap of top 10 most differentially expressed genes reduced in NNAT+ beta cells compared with NNAT beta cells. (c, d) Gene set enrichment analysis (GSEA) showing categories significantly enriched (c) and reduced (d) in NNAT+ (vs NNAT) beta cells. (e, f) Representative confocal microscopy of pancreatic cryosections from P56 (8 week old) wild type mice on a C57BL/6J background immunostained with antibodies against endogenous neuronatin (NNAT, green) and UCN3 (red, e) or TOM20 (red, f). Scale bar = 100μm, n = 3 mice. Nuclei are visualised with DAPI. Representative images from three independent experiments and breeding pairs. (g) Insulin content assessed in NNAT+ and NNAT beta cells (* P < 0.05, n = 7 FACS-purified populations from individual mouse islet preparations, Wilcoxon matched-pairs signed rank test).
Fig. 8.
Fig. 8.. NNAT+ beta cells show unaltered glucose-induced Ca2+ dynamics but are de-enriched for highly connected ‘hub’ cells within individual islets.
(a) Ca2+ bound Cal-590 fluorescence in response to high glucose (11 mM) in NNAT and NNAT+ cells from primary islets from NnatWT/eGFPpat reporter mice expressed as normalised intensity over time (F / Fmin) (n = 61 islets total from five mice per genotype, inset shows quantification of area under the curve (AUC), P = 0.063, Wilcoxon matched-pairs signed rank test). (b) Representative cartesian map of beta cells with colour coded lines connecting cells according of the strength of coactivation (colour coded R values from 0 to 1, blue to red). Beta cells are represented by differently coloured nodes depending on their coactivity with the other beta cells, where black nodes indicate coactivity with ≥80% of the remaining beta cells, while grey and white nodes represent coactivity with ≥60% and ≥40%, respectively. Nodes circled with a solid black line indicate NNAT+ cells. (c) Representative heatmaps depicting connectivity strength (r) of all cell pairs (colour coded r values from 0 to 1, blue to yellow). (d) Log-log graphs of beta cell-beta cell connectivity distribution. NNAT+ cells are represented by green circles while NNAT cells are represented by red circles (45 islets total using primary islet preparations, each from an individual NnatWT/eGFPpat reporter mouse). (e) Categorisation of beta cells based on data from d. (f) Percentage coactivity of beta cells between all cells in identified ‘hub’ and ‘follower’ cells. (g) The proportion of cells designated as ‘hub’ vs ‘follower’ cells in both the NNAT and NNAT+ populations assessed in each of 45 islets (** P < 0.01, Wilcoxon matched-pairs signed rank test). Representative images from three independent experiments and breeding pairs.

Similar articles

References

    1. Rutter GA, Pullen TJ, Hodson DJ, Martinez-Sanchez A (2015) Pancreatic beta-cell identity, glucose sensing and the control of insulin secretion. Biochem J 466(2): 203–218. 10.1042/BJ20141384 - DOI - PubMed
    1. Marchetti P, Suleiman M, De Luca C, et al. (2020) A direct look at the dysfunction and pathology of the beta cells in human type 2 diabetes. Semin Cell Dev Biol 103: 83–93. 10.1016/j.semcdb.2020.04.005 - DOI - PubMed
    1. Salinno C, Cota P, Bastidas-Ponce A, Tarquis-Medina M, Lickert H, Bakhti M (2019) beta-Cell Maturation and Identity in Health and Disease. Int J Mol Sci 20(21). 10.3390/ijms20215417 - DOI - PMC - PubMed
    1. Arda HE, Benitez CM, Kim SK (2013) Gene regulatory networks governing pancreas development. Dev Cell 25(1): 5–13. 10.1016/j.devcel.2013.03.016 - DOI - PMC - PubMed
    1. Avrahami D, Li C, Zhang J, et al. (2015) Aging-Dependent Demethylation of Regulatory Elements Correlates with Chromatin State and Improved beta Cell Function. Cell Metab 22(4): 619–632. 10.1016/j.cmet.2015.07.025 - DOI - PMC - PubMed

Publication types