The TCF7L2-dependent high-voltage activated calcium channel subunit α2δ-1 controls calcium signaling in rodent pancreatic beta-cells

https://doi.org/10.1016/j.mce.2019.110673Get rights and content

Highlights

  • Tcf7l2 controls expression of Cacna2d1/α2δ-1.

  • Silencing of Cacna2d1 prevents trafficking of Cav1.2 to the plasma membrane.

  • Silencing of Cacna2d1 retains Cav1.2 in recycling endosomes.

  • Silencing of Cacna2d1 affects Ca2+ signaling and exocytosis.

  • Overexpression of α2δ-1 partially counteracts the effect of Tcf7l2 silencing.

Abstract

The transcription factor TCF7L2 remains the most important diabetes gene identified to date and genetic risk carriers exhibit lower insulin secretion. We show that Tcf7l2 regulates the auxiliary subunit of voltage-gated Ca2+ channels, Cacna2d1 gene/α2δ-1 protein levels. Furthermore, suppression of α2δ-1 decreased voltage-gated Ca2+ currents and high glucose/depolarization-evoked Ca2+ signaling which mimicked the effect of silencing of Tcf7l2. This appears to be the result of impaired voltage-gated Ca2+ channel trafficking to the plasma membrane, as Cav1.2 channels accumulated in the recycling endosomes after α2δ-1 suppression, in clonal as well as primary rodent beta-cells. This impaired the capacity for glucose-induced insulin secretion in Cacna2d1-silenced cells. Overexpression of α2δ-1 increased high-glucose/K+-stimulated insulin secretion. Furthermore, overexpression of α2δ-1 in Tcf7l2-silenced cells rescued the Tcf7l2-dependent impairment of Ca2+ signaling, but not the reduced insulin secretion. Taken together, these data clarify the connection between Tcf7l2, α2δ-1 in Ca2+-dependent insulin secretion.

Introduction

Type 2 diabetes develops when the demand for insulin exceeds what the pancreatic beta cells can deliver. Glucose-stimulated insulin secretion occurs by Ca2+-dependent exocytosis and in this respect, Ca2+ entering via voltage-gated calcium channels (VGCCs) plays a particularly important role (Rorsman et al., 2000). Pancreatic β-cells are equipped with at least six types of calcium channels, including Cav1.2, Cav1.3, Cav2.1, Cav2.2, Cav2.3 and Cav3.1 (Yang and Berggren, 2006), of which L-type calcium channels (Cav1.2 and Cav1.3) conduct ~50% of the whole Ca2+ currents (Schulla et al., 2003). Nitert et al. reported that the mRNA level of Cav1.2 exceeded that of Cav1.3 and Cav2.3 two-fold in INS-1 832/13 cells, and suggested that Cav1.2 rather than Cav1.3 is critical to glucose-stimulated insulin secretion in INS-1 832/13 cells (Nitert et al., 2008). Rorsman et al. concluded that Cav1.2 is the principal L-type Ca2+-channels subtype in mouse β-cells (Rorsman et al., 2012). VGCCs are multi-subunit proteins consisting of the pore-forming α1 subunit, which exists in ten different isoforms in mammalian genomes and is the major VGCC subunit determining the main physiological and pharmacological properties. In addition, a number of auxiliary subunits exist in the genome, each in different isoforms, which attach to the α1 pore subunit and modulate its functions. Of the auxiliary subunits, the function of the γ subunits remains largely unknown, whereas the α2δ and β subunits have been suggested to control VGCC trafficking to plasma membrane (PM), but also to influence certain of the channels’ biophysical properties (Dolphin, 2013). Accordingly, in heterologous expression systems the α2δ subunits increase the maximum current density of Cav1 and Cav2 VGCCs (Dolphin, 2003). The main mechanism has been suggested that increased expression of Cav1 and Cav2 complexes at the plasma membrane coupled to a decrease in their turnover (Canti et al., 2005; Bernstein and Jones, 2007). Interestingly, it was demonstrated in mouse that genetic ablation of α2δ-1 (the major variant in pancreatic islets) reduces Ca2+ influx via all types of functional VGCCs in the pancreatic beta cells, which resulted in reduced insulin secretion and impaired glucose tolerance, at least in male mice (Mastrolia et al., 2017). However, a detailed characterization of the cellular effects at the single beta cell level remains to be performed.

TCF7L2 (T-cell factor 7-like 2, also known as TCF4) harbors the single nucleotide polymorphism (SNP) rs7903146, which remains the most significantly common genetic variation associated with human type 2-diabetes (Grant et al., 2006). This genetic variant is linked to reduced gene expression, in particular of certain splice variants (Osmark et al., 2009), and reduced capacity for insulin secretion (Lyssenko et al., 2007), but pathophysiological role of TCF7L2 in development of type 2-diabetes is elusive. Recently, report showed that TCF7L2 is a master regulator of insulin production and processing (Zhou et al., 2014). By contrast TCF7L2 has no major influence over genes involved in the control of Ca2+ signaling and exocytosis, with one exception: Cacna2d1 gene expression is markedly reduced in Tcf7l2-silenced cells (Zhou et al., 2014). However, the functional consequences in terms of protein levels and beta cell function are still missing. Therefore, we decided to extend on these previous reports by verifying the regulatory role of the diabetes gene Tcf7l2 on α2δ-1 expression, as well as its consequences for Ca2+ signaling and insulin secretion in the beta cell.

Section snippets

Cell culture and preparation of islets, β cells

Rat-derived INS-1 832/13 cells were cultured in RPMI 1640 (HyClone, USA) medium containing 11.1 mΜ D-glucose (Sigma, USA) supplemented with 10% fetal bovine serum (Sigma, USA), 11.2 mM HEPES (HyClone, USA), 100 U/ml penicillin (HyClone, USA), 100 μg/ml streptomycin (HyClone, USA), 2 mM L-glutamine (HyClone, USA), 1 mM sodium pyruvate (HyClone, USA) and 0.05 mΜ 2-mercaptoethanol (Sigma, USA), at 37 °C in a humidified atmosphere containing 95% air and 5% CO2. The INS-1 832/13 cells were stored at

Tcf7l2 controls expression of Cacna2d1/α2δ-1

To assess the effect of Tcf7l2 on expression of Cacna2d1, INS-1 832/13 cells were first pre-treated with Tcf7l2 targeting siRNA and non-targeting siRNA for 48 h. Silencing efficiency on the mRNA level amounted 65% ± 1% (Fig. 1A). Tcf7l2 silencing resulted in a significant decrease in gene expression of Cacna2d1 (Fig. 1B). Similar results were observed in primary rat islets in which silencing of Tcf7l2 (Fig. 1C) also induced a significant decrease in the expression of Cacna2d1 (Fig. 1D), even

Discussion

Tissue distribution of the different α2δ subunit isoforms, as well as their subcellular expression is thoroughly studied. The α2δ-1 subunit is primarily expressed in excitable cells (Gong et al., 2001). In the present study, α2δ-1 exhibited significant expression in insulin-secreting β cells and was regulated by the transcription factor Tcf7l2 (Fig. 1). The limitation of lower efficacy of silencing in rat/mouse islets could be complemented by studies in conditional knockout mice in the future.

Contribution statement

E.R. and T.T. conceived the study. Y.Y, M.B, C.L, Y.Z, E.R. designed experiments, acquired and analysed data. E.R, T.T, E.Z, L.E and O.H analysed and interpreted data. Y.Y and A.K drafted the article. All authors revised the article and approved it for publishing. E.R is the guarantor of this work.

Declaration of competing interest

None.

Acknowledgements

We thank Britt-Marie Nilsson and Anna-Maria Veljanovska Ramsay for expert technical assistance. Y.Y.'s position was financed by China Scholarship Council (201306310018). Grants to E.R. supporting this project include: the Swedish Research Council (2017-01090), Swedish Diabetes Association, Diabetes Wellness Foundation Sweden, and grants for clinical research (ALF). Grants to E.Z.: Swedish Research Council (2018-03258). The study used equipment/infrastructure funded by the Knut and Alice

References (46)

  • C. Canti et al.

    The metal-ion-dependent adhesion site in the Von Willebrand factor-A domain of alpha2delta subunits is key to trafficking voltage-gated Ca2+ channels

    Proc. Natl. Acad. Sci. U. S. A.

    (2005)
  • S. Cauchi et al.

    Transcription factor TCF7L2 genetic study in the French population: expression in human beta-cells and adipose tissue and strong association with type 2 diabetes

    Diabetes

    (2006)
  • A.C. Dolphin

    G protein modulation of voltage-gated calcium channels

    Pharmacol. Rev.

    (2003)
  • R. Felix et al.

    Dissection of functional domains of the voltage-dependent Ca2+ channel alpha2delta subunit

    J. Neurosci.

    (1997)
  • J.C. Florez

    The new type 2 diabetes gene TCF7L2

    Curr. Opin. Clin. Nutr. Metab. Care

    (2007)
  • J.C. Florez et al.

    TCF7L2 polymorphisms and progression to diabetes in the Diabetes Prevention Program

    N. Engl. J. Med.

    (2006)
  • U. Fransson et al.

    Anaplerosis via pyruvate carboxylase is required for the fuel-induced rise in the ATP:ADP ratio in rat pancreatic islets

    Diabetologia

    (2006)
  • B.R. Gauthier et al.

    Synaptotagmins bind calcium to release insulin

    Am. J. Physiol. Endocrinol. Metab.

    (2008)
  • S.H. Gerber et al.

    Molecular determinants of regulated exocytosis

    Diabetes

    (2002)
  • H.C. Gong et al.

    Tissue-specific expression and gabapentin-binding properties of calcium channel alpha2delta subunit subtypes

    J. Membr. Biol.

    (2001)
  • S.F. Grant et al.

    Variant of transcription factor 7-like 2 (TCF7L2) gene confers risk of type 2 diabetes

    Nat. Genet.

    (2006)
  • C.J. Groves et al.

    Association analysis of 6,736 U.K. subjects provides replication and confirms TCF7L2 as a type 2 diabetes susceptibility gene with a substantial effect on individual risk

    Diabetes

    (2006)
  • J. Hendrich et al.

    Pharmacological disruption of calcium channel trafficking by the alpha2delta ligand gabapentin

    Proc. Natl. Acad. Sci. U. S. A.

    (2008)
  • Cited by (11)

    • The T-type calcium channel Ca<inf>V</inf>3.2 regulates insulin secretion in the pancreatic β-cell

      2022, Cell Calcium
      Citation Excerpt :

      Blockade of CaV channels leads to suppression of action potential firing and insulin release [5]. CaV channels are composed of pore-forming alpha1 subunits and the auxiliary alpha2delta, beta, and gamma subunits that regulate the function and trafficking of CaV alpha1 subunits [6–9]. The newly synthesized CaV channel subunits are properly folded in the endoplasmic reticulum (ER) to be assembled as heteromeric complexes.

    View all citing articles on Scopus
    View full text