Efficient differentiation of AR42J cells towards insulin-producing cells using pancreatic transcription factors in combination with growth factors

https://doi.org/10.1016/j.mce.2012.02.024Get rights and content

Abstract

The AR42J-B13 rat pancreatic acinar cell line was used to identify pancreatic transcription factors and exogenous growth factors (GFs) that might facilitate the reprogramming of exocrine cells into islets. Adenoviruses were used to induce exogenous expression of the pancreatic transcription factors (TFs) Pdx1, MafA, Ngn3 and Pax4. Individually Pdx1, MafA and Pax4 had no effect on the expression of endocrine markers, whilst adeno-Ngn3 on its own increased the expression of Pax4, Ngn3 and NeuroD. In combination the four TFs had a significant effect on the expression of insulin 1 and 2 that was associated with a change in cell morphology from a rounded to a spindle-like shape. Amongst a range of growth factors, Betacellulin and Nicotinamide were shown to enhance the effects of the four TFs. The presence of adeno-Pax4 in the differentiation cocktail was important in limiting the expression of glucagon and in generating glucose sensitive insulin secretion. Further experiments asked whether the adenoviral TFs could be replaced by protein transduction domain (PTD)-containing TFs. The results showed that the PTD-TFs could mimic in part the effects of the adeno-TFs, but the resultant cells did not undergo the important morphological change associated with differentiation to endocrine lineages and levels of endogenous markers were very much lower. In summary, the results describe a cocktail of four TFs and two GFs that can be used to induce formation of glucose sensitive insulin secreting cells from ARJ42 cells, and demonstrate that it would be difficult to replace adenoviral transduction with PTD-TFS.

Highlights

► The AR42J-B13 rat exocrine cell line can be induced towards insulin-secreting lineages. ► Four pancreatic transcription factors in combination with Betacellulin and Nicotinamide generated insulin producing cells. ► Pax4 reduced glucagon levels and enhanced sensitivity of the cells to glucose. ► Reprogramming efficiency with PTD-TFs was lower than with adenoviral expression.

Introduction

The major therapeutic challenges in type 1 diabetes are, in the short term avoiding unexpected episodes of hypoglycaemia and in the long term avoiding the late complications of the disease that affect eye, kidney, and peripheral nerve function. Recent successes in islet transplantation (Shapiro et al., 2000), albeit limited (Ryan et al., 2005), have raised hope these challenges might be met. However, the dependence on the rare availability of cadaveric tissue has stimulated a drive towards generating an alternative source of islets. An understanding of the transcription factors that control the developing pancreas (Gittes, 2009) has provided protocols that reproducibly generate pancreatic progenitors from embryonic stem cells which can develop into functional β-cells when engrafted in mice (Kroon et al., 2008). At the same time the discovery that cells of one tissue type could be reprogrammed into those of other tissues (Takahashi and Yamanaka, 2006) has led to new therapeutic options. Thus acinar (Zhou et al., 2008) and liver (Yechoor et al., 2009, Yatoh et al., 2007) cells, which are developmentally closely related to pancreatic endocrine cells, have been reprogrammed to β-cells through a specific combination of pancreatic transcription factors that include Pdx1, Ngn3, NeuroD and MafA. Pdx1 is present in early progenitors that give rise to all cells of the pancreas, Ngn3 directs cells towards an endocrine fate, while later expression of NeuroD, MafA, and Pdx1 in a second wave of expression, act as β-cell differentiation factors (Bernardo et al., 2008). Because of the risk of inflammation and resultant pancreatitis in vivo reprogramming of acinar tissue using viral vectors is unrealistic for treatment of type 1 diabetes, however, the in vitro reprogramming of exocrine tissue may be of therapeutic value.

AR42J is a rat cell line that was originally derived from a chemically induced pancreatic tumour (Longnecker et al., 1979). AR42J cells can be induced to differentiate towards hepatocyte (Wallace et al., 2010) and endocrine pancreatic lineages (Mashima et al., 1996a). Here we used these cells in an attempt to optimise conditions for the efficient reprogramming of pancreatic exocrine cells towards functional pancreatic endocrine cells. Our strategy was to infect the cells with adenoviruses containing pancreatic transcription factors (TFs) and to measure effects on phenotypic markers that are characteristic of pancreatic endocrine lineages. An additional aim was to determine whether protein transduction domain (PTD) mediated uptake of TFs might provide a better alternative to the use of adenoviruses. The most commonly used PTD is the 11 amino acid peptide derived from the TAT protein of the TAT/HIV-1 transactivator protein (Gump and Dowdy, 2007). Recombinant proteins that have been engineered to contain the TAT sequence are able to directly cross the cell membrane, and enter the nucleus of a variety of cell types (Manceur et al., 2007, Krosl et al., 2005).

Section snippets

Cell culture

AR42J-B13, a sub clone of ARJ42 that was selected on the basis of its enhanced capability to differentiate into insulin-secreting cells (Mashima et al., 1996a), was cultured in DMEM supplemented with 10% Foetal Calf Serum (Invitrogen, Paisley, UK). Cells were plated at a density of 5 × 106 cells per well of 6 well-culture tissue dishes (Greiner, Stonehouse, UK). For adenoviral infection, the cells were incubated with adenoviruses at a multiplicity of infection of 100, for 4 h at 37 °C before

Results

Adenoviruses containing Pdx1, MafA, Ngn3 and Pax4 were used to reprogramme AR42J-B13 cells towards endocrine lineages. Efficient expression of each of these transcription factors (TFs) was detected by immunocytochemistry in ∼80% of the cells three days after infection (Fig. 1). Cells that were not infected with the adenoviruses contained low levels of endogenous Pdx1 (but no GFP) and no detectable levels of the other three TFs (Fig. 1B). The presence of Pdx1 in AR42J-B13 cells has been

Discussion

The major finding of this study was that AR42J-B13 exocrine cells could be reprogrammed into cells that expressed and secreted insulin in response to glucose using four transcription factors, namely Pdx1, MafA, Ngn3 and Pax4, in combination with two factors, Betacellulin and Nicotinamide. The reprogrammed cells also expressed transcription factors such as NeuroD, MafA, Pax4 and Ngn3 that are typically expressed in pancreatic endocrine cells and adopted a spindle-shaped morphology that was more

References (37)

  • J. Wang et al.

    The concerted activities of Pax4 and Nkx2.2 are essential to initiate pancreatic beta-cell differentiation

    Dev. Biol.

    (2004)
  • V. Yechoor et al.

    Neurogenin3 is sufficient for transdetermination of hepatic progenitor cells into neo-islets in vivo but not transdifferentiation of hepatocytes

    Dev. Cell

    (2009)
  • A. Alidibbiat et al.

    Inability to process and store proinsulin in transdifferentiated pancreatic acinar cells lacking the regulated secretory pathway

    J. Endocrinol.

    (2008)
  • E. Akinci et al.

    Reprogramming of pancreatic exocrine cells towards a beta cell character using Pdx1,Ngn3 and MafA

    Biochem. J.442

    (2012)
  • M. Asfari et al.

    Establishment of 2-mercaptoethanol-dependent differentiated insulin-secreting cell lines

    Endocrinology

    (1992)
  • L. Baeyens et al.

    In vitro generation of insulin-producing beta cells from adult exocrine pancreatic cells

    Diabetologia

    (2005)
  • K. Docherty

    Reprogramming Towards Pancreatic Beta Cells

  • H. Docherty et al.

    Relative contribution of PDX-1, MafA and E47/beta 2 to the regulation of the human insulin promoter

    Biochem. J.

    (2005)
  • Cited by (24)

    • Emergence of synthetic mRNA: In vitro synthesis of mRNA and its applications in regenerative medicine

      2018, Biomaterials
      Citation Excerpt :

      Non-exocrine cells, such as AR42J rat pancreatic tumor cells, were transfected with mRNAs encoding three pancreatic transcription factors (Neurogenin3, Pdx1, and MafA) for the derivation of insulin secreting cells (Fig. 9a). AR42J cells can differentiate into insulin secreting cells upon treatment with viral vectors [47,191]. The researchers optimized the transdifferentiation protocol with 5-Aza-2′-deoxycytidine (5Aza2DC), which facilitate DNA demethylation and enhances the accessibility of transcription factors to genomic DNA.

    • The Human Endocrine Pancreas: New Insights on Replacement and Regeneration

      2016, Trends in Endocrinology and Metabolism
      Citation Excerpt :

      The latter observation could be explained by the fact that the newly created β cells remained isolated and did not cluster to form islets, as β cell communication is essential to stimulate glucose-mediated insulin secretion [36]. More recently, the above results were confirmed in vitro using the AR42J acinar cell line [37–39]. Although signs of rudimentary endocrine differentiation were observed in these studies, the resulting cells were not glucose responsive.

    • Reprogramming of Pancreatic Exocrine Cells AR42J Into Insulin-producing Cells Using mRNAs for Pdx1, Ngn3, and MafA Transcription Factors

      2016, Molecular Therapy Nucleic Acids
      Citation Excerpt :

      Limited endogenous expression of MafA, which was significantly under-expressed in comparison with the native β-cells, can be explained by insufficient induction of Nkx6.1, Pax6, and Isl1 transcription factors that all positively regulate the MafA expression.33,36,37,38 Previous studies have reported the reprogramming of exocrine cells into insulin-producing cells using adenoviral vectors.11,12,13,14,15,36 Although adenoviral vectors are highly efficient in the delivery and expression of introduced genes, the application of this process is limited by the potential for insertional mutagenesis,18,19 and by the prolonged persistence in infected cells that does not allow modulation of the reprogramming process.46

    • Beta Cell Transplantation and Regeneration

      2015, Endocrinology: Adult and Pediatric
    View all citing articles on Scopus
    1

    Present address: Department of Cell Biology and Medical Genetics, Kunming Medical College, Kunming 650500, China.

    View full text