Gastroenterology

Gastroenterology

Volume 139, Issue 2, August 2010, Pages 609-619.e6
Gastroenterology

Basic—Liver, Pancreas, and Biliary Tract
Regulator of Calcineurin 1 Controls Growth Plasticity of Adult Pancreas

https://doi.org/10.1053/j.gastro.2010.04.050Get rights and content

Background & Aims

Growth of exocrine pancreas is regulated by gastrointestinal hormones, notably cholecystokinin (CCK). CCK-driven pancreatic growth requires calcineurin (CN), which activates Nuclear Factor of Activated T cells (NFATs), but the genetic underpinnings and feedback mechanisms that regulate this response are not known.

Methods

Pancreatic growth was stimulated by protease inhibitor (PI)-containing chow, which induces secretion of endogenous CCK. Expression profiling of PI stimulation was performed on Affymetrix 430A chips, and CN was inhibited via FK506. Exocrine pancreas-specific overexpression of CN inhibitor Regulator of Calcineurin 1 (Rcan1) was achieved by breeding elastase-Cre(estrogen receptor [ER]) transgenics with “flox-on” Rcan1 mice.

Results

CN inhibitor FK506 blocked expression of 38 genes, as confirmed by quantitative polymerase chain reaction. The CN-dependent genes were linked to growth-related processes, whereas their promoters were enriched in NFAT and NFAT/AP1 sites. Multiple NFAT targets, including Rcan1, Rgs2, HB-EGF, Lif, and Gem, were validated by chromatin immunoprecipitation. One of these, a CN feedback inhibitor Rcan1, was induced >50 fold during 1–8 hours course of pancreatic growth and strongly inhibited (>99%) by FK506. To examine its role in pancreatic growth, we overexpressed Rcan1 in an inducible, acinar-specific fashion. Rcan1 overexpression inhibited CN-NFAT signaling, as shown using an NFAT-luciferase reporter and quantitative polymerase chain reaction. Most importantly, the increase in exocrine pancreas size, protein/DNA content, and acinar proliferation were all blocked in Rcan1 overexpressing mice.

Conclusions

We profile adaptive pancreatic growth, identify Rcan1 as an important new feedback regulator, and firmly establish that CN-NFAT signaling is required for this response.

Section snippets

Materials

The PI camostat was provided by Ono Pharmaceuticals (Osaka, Japan). TaqMan reverse transcription and Expand polymerase chain reaction (PCR) kits were purchased from Roche (Basel, Switzerland). Antibodies and all other reagents are listed in Supplementary Materials and Methods.

Animals and Treatment

Animal experiments, including PI feeding and FK506 injections, are outlined in Supplementary Materials and Methods or performed as previously described.8 To overexpress Rcan1 in pancreatic acini, we bred FLAG-Rcan1 (Rcan1)

RNA Microarray Analysis of PI-Induced and CN-Dependent Genes

We performed genome-wide expression profiling of the 2-hour time point of CCK-driven pancreatic growth, focused on CN-dependent genes in particular. Pancreatic RNA was harvested from mice fasted overnight or fasted and refed PI-containing chow for 2 hours and injected with either 3 mg/kg of FK506 or vehicle then hybridized with Affymetrix 430A genechips (4 mice/group, 16 chips total), as outlined in Figure 1A, top. PI feeding led to a significant (>3-fold, q ≤ 0.08) increase in expression of 81

Discussion

Physiologic growth of the pancreas takes place in response to high protein diet, hyperphagia, pregnancy, and lactation.20 The molecular mechanisms that govern this adaptive response, however, are poorly understood. Here, we examined the expression profile of CCK-mediated pancreatic growth. We focused on CN-NFAT signaling axis, identifying several novel growth-related, NFAT-regulated genes. We also demonstrate that 1 of these genes, Rcan1, functions as a CN/NFAT-dependent feedback inhibitor that

Acknowledgments

The authors thank Linda Samuelson (University of Michigan) for CCK-deficient mice, Bradley Nelson for help with immunohistochemistry, and Scott Tomlins and the Arul Chinnayian laboratory (University of Michigan) for assistance with bioinformatics.

References (39)

  • P.M. Brannon

    Adaptation of the exocrine pancreas to diet

    Annu Rev Nutr

    (1990)
  • M. Tashiro et al.

    Calcineurin mediates pancreatic growth in protease inhibitor-treated mice

    Am J Physiol Gastrointest Liver Physiol

    (2004)
  • B.J. Wilkins et al.

    Calcineurin/NFAT coupling participates in pathological, but not physiological, cardiac hypertrophy

    Circ Res

    (2004)
  • M. Tashiro et al.

    Calcineurin mediates pancreatic growth in protease inhibitor-treated mice

    Am J Physiol Gastrointest Liver Physiol

    (2004)
  • G.T. Gurda et al.

    Cholecystokinin activates pancreatic calcineurin-NFAT signaling in vitro and in vivo

    Mol Biol Cell

    (2008)
  • M. Oh et al.

    Calcineurin is necessary for the maintenance but not embryonic development of slow muscle fibers

    Mol Cell Biol

    (2005)
  • B. Ji et al.

    Robust acinar cell transgene expression of CreErT via BAC recombineering

    Genesis

    (2008)
  • S.L. Le Page et al.

    CCK-A receptor activates RhoA through G α 12/13 in NIH3T3 cells

    Am J Physiol Cell Physiol

    (2003)
  • L. Guo et al.

    Induction of early response genes in trypsin inhibitor-induced pancreatic growth

    Am J Physiol Gastrointest Liver Physiol

    (2007)
  • Cited by (29)

    • RCAN1 is a marker of oxidative stress, induced in acute pancreatitis

      2018, Pancreatology
      Citation Excerpt :

      It has been shown to be regulated in neuronal cells by both CN-NFAT signaling, NFκB [28] and oxidative stress [29]. The specific role of Rcan1 in the pancreas is not as well studied, however overexpression of Rcan1 in pancreatic β-cells has been shown to cause hypoinsulinemia, β-cell dysfunction and diabetes [30] and CN-NFAT signaling induced Rcan1 expression in acinar cells, following damage-induced regeneration [15]. One of the most commonly used in vivo models of AP is the caerulein induction model [31], where hyper-stimulation with caerulein induces zymogen activation [32], ROS production and AP [33,34].

    • NFATc1 links EGFR signaling to induction of sox9 transcription and acinar-ductal transdifferentiation in the pancreas

      2015, Gastroenterology
      Citation Excerpt :

      The identification of Cox2 as an EGFR/NFAT target already points toward critical functions of the EGFR/NFAT cascade for the integration of inflammatory signals during carcinogenesis. In particular, during pancreatic cancer formation, NFAT proteins have been shown to play a central role in the integration of environmental signals into oncogenic transcriptional processes that mediate proliferation, cellular differentiation, and adaptation to inflammation.21,22,28,30 In this study, we showed that sustained pancreatic inflammation induced a strong up-regulation of NFATc1 in metaplastic areas of both murine and human chronic pancreatitis models.

    • Bile acids induce pancreatic acinar cell injury and pancreatitis by activating calcineurin

      2013, Journal of Biological Chemistry
      Citation Excerpt :

      Each of the inhibitors reduced the activation of our reporter system driven by the NFAT promoter, suggesting that TLCS injury is mediated by calcineurin. Further, infecting cells with an adenovirus that overexpresses an endogenous inhibitor of calcineurin, Mcip1, also known as regulator of calcineurin 1 (Rcan1) (33), showed similar results (Fig. 1F). We demonstrate that TLCS promotes transcriptional activation via a calcineurin-specific pathway.

    • Regulation of Pancreatic Protein Synthesis and Growth

      2023, The Pancreas: an Integrated Textbook of Basic Science, Medicine, and Surgery, Fourth Edition
    View all citing articles on Scopus

    Conflicts of interest The authors disclose no conflicts.

    Funding Supported by NIH grants DK 59578 (to J.A.W.) and P30 DK-34933 (Michigan Gastrointestinal Peptide Center); by Systems and Integrative Biology Training Grant (T32 GM008322; to G.T.G.) and the Medical Scientist Training Program (G.T.G.); by DK52067 (to C.D.L.), R21 DK068414 (to B.J.), DK-0077423 (to S.J.C.), and HL072016 (B.A.R.); and for use of the Morphology and Image Analysis Core of the Michigan Diabetes Research and Training Center funded by NIH5P60 DK20572.

    View full text