MicroRNA-29a is up-regulated in beta-cells by glucose and decreases glucose-stimulated insulin secretion

https://doi.org/10.1016/j.bbrc.2012.08.082Get rights and content

Abstract

Chronically elevated levels of glucose impair pancreatic beta-cell function while inducing beta-cell proliferation. MicroRNA-29a (miR-29a) levels are increased in several tissues in diabetic animals and mediate decreased insulin-stimulated glucose-transport of adipocytes. The aim was to investigate the impact of glucose on miR-29a levels in INS-1E beta-cells and in human islets of Langerhans and furthermore to evaluate the impact of miR-29a on beta-cell function and proliferation. Increased glucose levels up-regulated miR-29a in beta-cells and human and rat islets of Langerhans. Glucose-stimulated insulin-secretion (GSIS) of INS-1E beta-cells was decreased by forced expression of miR-29a, while depletion of endogenous miR-29a improved GSIS. Over-expression of miR-29a increased INS-1E proliferation. Thus, miR-29a up-regulation is involved in glucose-induced proliferation of beta-cells. Furthermore, as depletion of miR-29a improves beta-cell function, miR-29a is a mediator of glucose-induced beta-cell dysfunction. Glucose-induced up-regulation of miR-29a in beta-cells could be implicated in progression from impaired glucose tolerance to type 2 diabetes.

Highlights

► MicroRNA-29a (miR-29a) levels are increased by glucose in human and rat islets and INS-1E cells. ► miR-29a increases proliferation of INS-1E beta-cells. ► Forced expression of miR-29a decreases glucose-stimulated insulin secretion (GSIS). ► Depletion of beta-cell miR-29a improves GSIS. ► miR-29a may be a mediator of glucose toxicity in beta-cells.

Introduction

Type 2 diabetes mellitus is a complex metabolic disorder involving two core defects: insulin resistance and beta-cell dysfunction, both of which are present in pre-diabetic states and act in concert to progressively exacerbate glucose intolerance. Elevated plasma glucose levels (post-prandially or chronically) lead to decreased glucose-stimulated insulin secretion (GSIS) and beta-cell dysfunction [1]. The mechanisms underlying glucose-induced beta-cell dysfunction, or glucose toxicity, are incompletely understood, but involve beta-cell exhaustion from continued insulin release as well as mitochondrial dysfunction [2], [3], [4], however, an increased glucose level is also a powerful beta-cell mitogen [5].

MicroRNAs (miRNAs) are regulators of gene expression at the post-transcriptional level and function by partially binding to the 3′untranslated region (UTR) of their target gene transcripts either mediating transcript degradation or translational inhibition [6], [7], [8]. Several miRNAs have been implicated in beta-cell function: miR-9, miR-30d, miR-124, miR-133a and miR-375 are involved in glucose-dependent regulation of insulin transcription and/or insulin release in beta-cells by targeting beta-cell transcription factors and/or transcripts involved in insulin exocytosis [9], [10], [11], [12], [13], [14], [15]. Furthermore, fatty acid induced increase in miR-34a levels may cause beta-cell dysfunction [16].

miR-29a is up-regulated by glucose in skeletal muscle, liver and white adipose tissue, where it leads to insulin resistance [17], [18], [19]. We hypothesized that miR-29a could be similarly regulated by glucose in pancreatic beta-cells and mediate glucose-induced dysfunction. Thus, the aim of this study was to determine the glucose-dependent regulation of miR-29a in beta-cells, as well as effects on GSIS.

Our results indicate that glucose-mediated up-regulation of miR-29a in beta-cells mediates beta-cell dysfunction and increased beta-cell proliferation, while inhibition of miR-29a improves GSIS. Thus, the up-regulation of miR-29a by glucose could be a link between glucose-induced proliferation and beta-cell dysfunction.

Section snippets

Cell culture and nucleofection

INS-1E cells (gift from Claes Wollheim, Geneva, Switzerland) were cultured in RPMI as described previously [20]. Cells (4 × 106) were nucleofected using a Nucleofector (Amaxa, Lonza, Copenhagen, Denmark) with miR-29a LNA knock-down or scrambled LNA oligonucleotide (Exiqon, Vedbaek, Denmark), or with miRIDIAN miR-29a mimic or the miRIDIAN negative control #2 (Dharmacon, ThermoFisher Scientific, Slangerup, Denmark). Nucleofected cells were seeded in poly-lysine treated culture plates, and 24 h

Regulation of miR-29a levels by glucose in human and rat islets of Langerhans and INS-1E cells

The expression levels of miR-29a in human islets of Langerhans and INS-1E cells treated with low (5 mM) or high glucose (11 mM and 20 mM respectively) for 48 h were assessed by real-time RT-Q-PCR or northern blotting. The expression levels of mature miR-29a in INS-1E cells (Fig. 1A–C), rat and human pancreatic islets (Fig. 1D–E) increased 1.7 ± 0.1 (P < 0.001), 1.5 ± 0.3 (P < 0.05) and 1.23 ± 0.06-fold (P < 0.05), respectively, in response to increased glucose levels, and in INS-1E cells miR-29a up-regulation

Discussion

Prolonged exposure of beta-cells to high levels of glucose decreases GSIS [2], [30]. The current results show that over-expression of miR-29a, up-regulated by glucose in human islets of Langerhans and in INS-1E beta-cells, decreased GSIS markedly in INS-1E cells that normally exhibit a robust insulin secretion response. Inhibition of miR-29a increases GSIS, suggesting that endogenous miR-29a exert a tonic inhibition on GSIS. Even though inhibition of miR-29a increased GSIS in cells cultured in

Acknowledgments

We are very grateful for the skilled technical assistance of Kirsten Olesen, Roskilde University and Vibeke Nielsen, Novo Nordisk A/S. LTD and JHN have been and TRC and ML are employed by and own stocks in Novo Nordisk A/S, a pharmaceutical company selling diabetes products. These studies were supported by the Danish Research Council for Technology and Production and the Danish microRNA Consortium.

References (34)

  • D.P. Bartel et al.

    Micromanagers of gene expression: the potentially widespread influence of metazoan microRNAs

    Nat. Rev. Genet.

    (2004)
  • T.M. Rana

    Illuminating the silence: understanding the structure and function of small RNAs

    Nat. Rev. Mol. Cell Biol.

    (2007)
  • L. Song et al.

    MicroRNAs and cell differentiation in mammalian development birth defects

    Res. C. Embryo. Today

    (2006)
  • P. Lovis et al.

    Regulation of the expression of components of the exocytotic machinery of insulin-secreting cells by microRNAs

    Biol. Chem.

    (2008)
  • M.N. Poy et al.

    A pancreatic islet-specific microRNA regulates insulin secretion

    Nature

    (2004)
  • M.N. Poy et al.

    miR-375 maintains normal pancreatic a- and b-cell mass

    Proc. Natl. Acad. Sci. USA

    (2009)
  • R.G. Fred et al.

    High glucose suppresses human islet insulin biosynthesis by inducing miR-133a leading to decreased polypyrimidine tract binding protein-expression

    PLoS One

    (2010)
  • Cited by (91)

    • Comparing the effect of cinnamaldehyde and metformin on expression of MiR320 and MiR26-b in insulin resistant 3T3L1 adipocytes

      2021, Phytomedicine Plus
      Citation Excerpt :

      MiRNAs are novel biomarkers for predicting IR and Diabetes. MiR320, 122, 223 and 29a are up-regulated and miR26b, 21 and 138 are down-regulated in hyperglycemia and insulin resistance (Bagge et al., 2012; Chen et al., 2014; Chuang et al., 2015;Flowers et al., 2015; Ling et al., 2009; Naghiaee, 2020b, 2020c; Udesen et al., 2020; Xu et al., 2015). In our study after making 3T3L1 adipocytes insulin resistance, we determined content of expression of miR320 and miR26-b compared to non-IR adipocytes.

    • Role of microRNA in pancreatic beta cell function

      2021, International Review of Cell and Molecular Biology
      Citation Excerpt :

      Since Granuphilin mediates the docking of secretory granules onto the plasma membrane, but it inhibits the release of insulin (Kato et al., 2006), the increased expression of miR-9 and miR-29 further enhance granuphilin-dependent inhibition of insulin secretion. Interestingly, miR-29 upregulation is observed in the islets of non-obese diabetic (NOD) mice, the murine model of type 1 diabetes, and it is induced by pro-inflammatory cytokines (Roggli et al., 2012) and high glucose (Bagge et al., 2012), in both murine and human islets. Moreover, in high glucose-treated rodent beta cells, the upregulation of miR-29 reduces Syntaxin1A expression and insulin secretion (Bagge et al., 2013).

    View all citing articles on Scopus
    View full text