Exp Clin Endocrinol Diabetes 2022; 130(10): 638-651
DOI: 10.1055/a-1750-9190
Article

Valproic Acid Initiates Transdifferentiation of the Human Ductal Adenocarcinoma Cell-line Panc-1 Into α-Like Cells

1   Clinical Research Unit, Center of Internal Medicine, Medical Clinic and Polyclinic III, Justus Liebig University, Giessen, Germany
,
Naga Deepa Kandula*
1   Clinical Research Unit, Center of Internal Medicine, Medical Clinic and Polyclinic III, Justus Liebig University, Giessen, Germany
,
Stefan Günther
2   Bioinformatics and deep sequencing platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
,
Christian Helker
3   Cell Signaling and Dynamics, Department of Biology, Philipps University, Marburg, Germany
,
Undraga Schagdarsurengin
4   Epigenetics of Urogenital System, Clinic and Polyclinic of Urology, Pediatric Urology and Andrology, Justus Liebig University, Giessen, Germany
,
Thomas Linn
1   Clinical Research Unit, Center of Internal Medicine, Medical Clinic and Polyclinic III, Justus Liebig University, Giessen, Germany
› Author Affiliations
Funding General funding from the University of Giessen was employed for the realization of this study. There was no role of the funders in study design, data collection, analysis, decision to publish, or preparation of the manuscript.

Abstract

Non-mesenchymal pancreatic cells are a potential source for cell replacement. Their transdifferentiation can be achieved by triggering epigenetic remodeling through e. g. post-translational modification of histones. Valproic acid, a branched-chain saturated fatty acid with histone deacetylase inhibitor activity, was linked to the expression of key transcription factors of pancreatic lineage in epithelial cells and insulin transcription. However, the potential of valproic acid to cause cellular reprogramming is not fully understood. To shed further light on it we employed next-generation RNA sequencing, real-time PCR, and protein analyses by ELISA and western blot, to assess the impact of valproic acid on transcriptome and function of Panc-1-cells. Our results indicate that valproic acid has a significant impact on the cell cycle, cell adhesion, histone H3 acetylation, and metabolic pathways as well as the initiation of epithelial-mesenchymal transition through acetylation of histone H3 resulting in α-cell-like characteristics. We conclude that human epithelial pancreatic cells can be transdifferentiated into cells with endocrine properties through epigenetic regulation by valproic acid favoring an α-cell-like phenotype.

* These authors have contributed equally to this work and share first authorship.


Supplementary Material



Publication History

Received: 26 July 2021
Received: 16 November 2021

Accepted: 18 January 2022

Article published online:
21 April 2022

© 2022. Thieme. All rights reserved.

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Rezania A, EBruin Jennifer, Arora Payal. et al. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat Biotechnol 2014; 32: 1121-1133
  • 2 Wang P, Alvarez-Perez J-C, Felsenfeld DP. et al. A high-throughput chemical screen reveals that harmine-mediated inhibition of dyrk1a increases human pancreatic beta cell replication. Nat Med 2015; 21: 383-388
  • 3 Xu X, D’Hoker J, Stangé G. et al. Beta cells can be generated from endogenous progenitors in injured adult mouse pancreas. Cell 2008; 132: 197-207
  • 4 Al-Hasani K, Pfeifer A, Courtney M. et al. Adult duct-lining cells can reprogram into beta-like cells able to counter repeated cycles of toxin-induced diabetes. Dev cell 2013; 26: 86-100
  • 5 Zhou Qiao, Brown Juliana, Kanarek Andrew. et al. Nature 2008; 455: 627-632
  • 6 Li W, Nakanishi M, Zumsteg A. et al. In vivo reprogramming of pancreatic acinar cells to three islet endocrine subtypes. eLife 2014; 3: e01846 DOI: 10.7554/eLife.01846.
  • 7 Lee J, Sugiyama T, Liu Y. et al. Expansion and conversion of human pancreatic ductal cells into insulin-secreting endocrine cells. eLife 2013; 2: e00940 DOI: 10.7554/eLife.00940.
  • 8 Bonner-Weir S, Taneja M, Weir GC. et al. In vitro cultivation of human islets from expanded ductal tissue. Proc NatiL Acad Sci U S A 2000; 97: 7999-8004
  • 9 Jonsson J, Carlsson L, Edlund T. et al. Insulin-promoter-factor 1 is required for pancreas development in mice. Nature 1994; 371: 606-609
  • 10 Yang Y-P, Thorel F, Boyer DF. et al. Context-specific alpha- to-beta-cell reprogramming by forced pdx1 expression. Genes Dev 2011; 25: 1680-1685
  • 11 Gradwohl G, Dierich A, LeMeur M. et al. neurogenin3 is required for the development of the four endocrine cell lineages of the pancreas. Proc Natl Acad Sci U S A 2000; 97: 1607-1611
  • 12 Schwitzgebel VM, Scheel DW, Conners JR. et al. Expression of neurogenin3 reveals an islet cell precursor population in the pancreas. Development 2000; 127: 3533-3542
  • 13 Gao N, LeLay J, Vatamaniuk MZ. et al. Dynamic regulation of pdx1 enhancers by foxa1 and foxa2 is essential for pancreas development. Genes Dev 2008; 22: 3435-3448
  • 14 Kaestner KH, Katz J, Liu Y. et al. Inactivation of the winged helix transcription factor hnf3alpha affects glucose homeostasis and islet glucagon gene expression in vivo. Genes Dev 1999; 13: 495-504
  • 15 Heddad Masson M, Poisson C, Guérardel A. et al. Foxa1 and foxa2 regulate alpha-cell differentiation, glucagon biosynthesis, and secretion. Endocrinology 2014; 155: 3781-3792
  • 16 St-Onge L, Sosa-Pineda B, Chowdhury K. et al. Pax6 is required for differentiation of glucagon-producing alpha-cells in mouse pancreas. Nature 1997; 387: 406-409
  • 17 Sander M, Neubüser A, Kalamaras J. et al. Genetic analysis reveals that pax6 is required for normal transcription of pancreatic hormone genes and islet development. Genes Dev 1997; 11: 1662-1673
  • 18 Ahlgren U, Pfaff SL, Jessell TM. et al. Independent requirement for isl1 in formation of pancreatic mesenchyme and islet cells. Nature 1997; 385: 257-260
  • 19 Jonatan D, Spence JR, Method AM. et al. Sox17 regulates insulin secretion in the normal and pathologic mouse beta cell. PloS One 2014; 9: e104675
  • 20 Dassaye R, Naidoo S, Cerf ME. Transcription factor regulation of pancreatic organogenesis, differentiation and maturation. Islets 2016; 8: 13-34
  • 21 Du A, Hunter CS, Murray J. et al. Islet-1 is required for the maturation, proliferation, and survival of the endocrine pancreas. Diabetes 2009; 58: 2059-2069
  • 22 Collombat P, Hecksher-Sørensen J, Krull J. et al. Embryonic endocrine pancreas and mature beta cells acquire alpha and pp cell phenotypes upon arx misexpression. J Clin Investig 2007; 117: 961-970
  • 23 Bramswig NC, Everett LJ, Schug J. et al. Epigenomic plasticity enables human pancreatic alpha to beta cell reprogramming. J Clin Investig 2013; 123: 1275-1284
  • 24 Chen S, Borowiak M, Fox JL. et al. A small molecule that directs differentiation of human escs into the pancreatic lineage. Nat Chem Biol 2009; 5: 258-265
  • 25 Oström M, Loffler KA, Edfalk S. et al. Retinoic acid promotes the generation of pancreatic endocrine progenitor cells and their further differentiation into beta-cells. PloS one 2008; 3: e2841
  • 26 Yuan Y, Hartland K, Boskovic Z. et al. A small-molecule inducer of pdx1 expression identified by high-throughput screening. Chem Biol 2013; 20: 1513-1522
  • 27 Mosley AL, Ozcan S. Glucose regulates insulin gene transcription by hyperacetylation of histone h4. J Biol Chem 2003; 278: 19660-19666
  • 28 Mosley AL, Corbett JA, Ozcan S. Glucose regulation of insulin gene expression requires the recruitment of p300 by the beta-cell-specific transcription factor pdx-1. Mol Endocrinol 2004; 18: 2279-2290
  • 29 Francis J, Chakrabarti SK, Garmey JC. et al. Pdx-1 links histone H3-Lys-4 methylation to RNA polymerase II elongation during activation of insulin transcription. J Biol Chem 2005; 280: 36244-36253
  • 30 Haumaitre C, Lenoir O, Scharfmann R. Directing cell differentiation with small-molecule histone deacetylase inhibitors: the example of promoting pancreatic endocrine cells. Cell Cycle 2009; 8: 536-544
  • 31 Liu X, Fan D. The epithelial-mesenchymal transition and cancer stem cells: Functional and mechanistic links. Curr Pharm Des 2015; 21: 1279-1291
  • 32 Phiel CJ, Zhang F, Huang EY. et al. Histone deacetylase is a direct target of valproic acid, a potent anticonvulsant, mood stabilizer, and teratogen. J Biol Chem 2001; 276: 36734-36741
  • 33 Krämer OH, Zhu P, Ostendorff HP. et al. The histone deacetylase inhibitor valproic acid selectively induces proteasomal degradation of HDAC2. EMBO J 2003; 22: 3411-3420
  • 34 Petrakova OS, Ashapkin VV, Shtratnikova VY. et al. Valproic acid increases the hepatic differentiation potential of salivary gland cells. Acta Naturae 2015; 7: 80-92
  • 35 Helker CSM, Mullapudi S-T, Mueller LM. et al. A whole organism small molecule screen identifies novel regulators of pancreatic endocrine development. Development 2019; 146: 14
  • 36 Khan S, Jena G. Valproic acid improves glucose homeostasis by increasing beta-cell proliferation, function, and reducing its apoptosis through hdac inhibition in juvenile diabetic rat. J Biochem Mol Toxicol 2016; 30: 438-446
  • 37 Lieber M, Mazzetta J, Nelson-Rees W. et al. Establishment of a continuous tumor-cell line (panc-1) from a human carcinoma of the exocrine pancreas. Int J Cancer 1975; 15: 741-747
  • 38 Hui H, Wright C, Perfetti R. Glucagon-like peptide 1 induces differentiation of islet duodenal homeobox-1-positive pancreatic ductal cells into insulin-secreting cells. Diabetes 2001; 50: 785-796
  • 39 Misiti S, Anastasi E, Sciacchitano S. et al. 3,5,3′-triiodo-L-thyronine enhances the differentiation of a human pancreatic duct cell line (hpanc-1) towards a beta-cell-like phenotype. J Cell Physiol 2005; 204: 286-296
  • 40 Hardikar AA, Marcus-Samuels B, Geras-Raaka E. et al. Human pancreatic precursor cells secrete fgf2 to stimulate clustering into hormone-expressing islet-like cell aggregates. Proc Natl Acad Sci U S A 2003; 100: 7117-7122
  • 41 Bolger AM, Lohse M, Usadel B. Trimmomatic: A flexible trimmer for illumina sequence data. Bioinformatics 2014; 30: 2114-2120
  • 42 Dobin A, Davis CA, Schlesinger F. et al. Star: Ultrafast universal RNA-seq aligner. Bioinformatics 2013; 29: 15-21
  • 43 Liao Y, Smyth GK, Shi W. featurecounts: An efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 2014; 30: 923-930
  • 44 Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for rna-seq data with deseq2. Genome Biol 2014; 15: 550
  • 45 Chen C-M, Shih T-H, Pai T-W. et al. Gene expression rate comparison for multiple high-throughput datasets. IET Syst Biol 2013; 7: 135-142
  • 46 Haas BJ, Papanicolaou A, Yassour M. et al. De novo transcript sequence reconstruction from RNA-seq using the Trinity platform for reference generation and analysis. Nat Protoc 2013; 8: 1494-1512
  • 47 Xie C, Mao X, Huang J. et al. Kobas 2.0: A web server for annotation and identification of enriched pathways and diseases. Nucleic Acids Res 2011; 39: W316-W322
  • 48 Burgess A, Vigneron S, Brioudes E. et al. Loss of human Greatwall results in G2 arrest and multiple mitotic defects due to deregulation of the cyclin b-cdc2/pp2a balance. Proc Natl Acad Sci U S A 2010; 107: 12564-12569
  • 49 Petry SF, Sun LM, Knapp A. et al. Distinct shift in beta-cell glutaredoxin 5 expression is mediated by hypoxia and lipotoxicity both in vivo and in vitro. Front Endocrinol (Lausanne) 2018 Mar 12 9: 84 DOI: 10.3389/fendo.2018.00084.
  • 50 Jonkman JEN, Cathcart JA, Xu F. et al. An introduction to the wound healing assay using live-cell microscopy. Cell Adh Migr 2014; 8: 440-451
  • 51 Straatman K. Wound Healing Assay. University of Leicester, College of Medicine, Biological Sciences and Psychology, Advanced Imaging Facilities. 2008
  • 52 Gittes GK, Galante PE, Hanahan D. et al. Lineage-specific morphogenesis in the developing pancreas: Role of mesenchymal factors. Development 1996; 122: 439-447
  • 53 Lammert E, Cleaver O, Melton D. Induction of pancreatic differentiation by signals from blood vessels. Science 2001; 294: 564-567
  • 54 Cai Q, Brissova M, Reinert RB. et al. Enhanced expression of VEGF-A in β cells increases endothelial cell number but impairs islet morphogenesis and b cell proliferation. Dev Biol 2012; 367: 40-54
  • 55 Apelqvist A, Li H, Sommer L. et al. Notch signalling controls pancreatic cell differentiation. Nature 1999; 400: 877-881
  • 56 Seymour PA, Collin CA, Egeskov-Madsen AR. et al. Jag1 modulates an oscillatory Dll1-Notch-Hes1 signaling module to coordinate growth and fate of pancreatic progenitors. Dev Cell 52: 731-747.e8
  • 57 Adams RH, Wilkinson GA, Weiss C. et al. Roles of ephrinb ligands and ephb receptors in cardiovascular development: Demarcation of arterial/venous domains, vascular morphogenesis, and sprouting angiogenesis. Genes Dev 1999; 13: 295-306
  • 58 Villasenor A, Marty-Santos L, Dravis C. et al. EphB3 marks delaminating endocrine progenitor cells in the developing pancreas. Dev Dyn 2012; 241: 1008-1019
  • 59 Piccand J, Meunier A, Merle C. et al. Pak3 promotes cell cycle exit and differentiation of b-cells in the embryonic pancreas and is necessary to maintain glucose homeostasis in adult mice. Diabetes 2014; 63: 203-215
  • 60 Wang R, Li J, Lyte K. et al. Role for beta1 integrin and its associated alpha3, alpha5, and alpha6 subunits in development of the human fetal pancreas. Diabetes 2005; 54: 2080-2089
  • 61 Crisera CA, Kadison AS, Breslow GD. et al. Expression and role of laminin-1 in mouse pancreatic organogenesis. Diabetes 2000; 49: 936-944
  • 62 Kaido T, Yebra M, Cirulli V. et al. Regulation of human beta-cell adhesion, motility, and insulin secretion by collagen IV and its receptor alpha1beta1. J Biol Chem 2004; 279: 53762-53769
  • 63 Kantengwa S, Baetens D, Sadoul K. et al. Identification and characterization of alpha 3 beta 1 integrin on primary and transformed rat islet cells. Exp Cell Res 237: 394-402
  • 64 Cirulli V, Beattie GM, Klier G. et al. Expression and function of alpha(v)beta(3) and alpha(v)beta(5) integrins in the developing pancreas: Roles in the adhesion and migration of putative endocrine progenitor cells. J Cell Biol 2000; 150: 1445-1460
  • 65 Saleem S, Li J, Yee S-P. et al. beta1 integrin/FAL/ERK signalling pathway is essential for human fetal islet cell differentiation and survival. J Pathol 219: 182-192
  • 66 Sharon N, Vanderhooft J, Straubhaar J. et al. Wnt signaling separates the progenitor and endocrine compartments during pancreas development. Cell Reports 2019; 27: 2281-2291.e5
  • 67 Qin Y, Xiao L, Zhan XB. et al. Pdxl and its role in activating ngn3 and pax6 to induce differentiation of ipscs into islet beta cells. Genet Mol Res 2015; 14: 8892-8900
  • 68 Gomez DL, O’Driscoll M, Sheets TP. et al. Neurogenin 3 expressing cells in the human exocrine pancreas have the capacity for endocrine cell fate. PloS One 2015; 10: e0133862
  • 69 Cheng C-W, Villani V, Buono R. et al. Fasting-mimicking diet promotes ngn3-driven beta-cell regeneration to reverse diabetes. Cell 2017; 168: 775-788.e12
  • 70 Liu T, Sun L, Jiang B. et al. Lineage conversion of mouse fibroblasts to pancreatic alpha-cells. Exp Mol Med 2017; 49: e350
  • 71 Rezania A, Riedel MJ, Wideman RD. et al. Production of functional glucagon-secreting alpha-cells from human embryonic stem cells. Diabetes 2011; 60: 239-247
  • 72 Rall LB, Pictet RL, Williams RH. et al. Early differentiation of glucagon-producing cells in embryonic pancreas: A possible developmental role for glucagon. Proc Natl Acad Sci U S A 1973; 70: 3478-3482
  • 73 Ye L, Robertson MA, Hesselson D. et al. Glucagon is essential for alpha cell transdifferentiation and beta cell neogenesis. Development 2015; 142: 1407-1417
  • 74 Ben-Othman N, Vieira A, Courtney M. et al. Long-term GABA administration induces alpha cell-mediated beta-like cell neogenesis. Cell 2017; 168: 73-85.e11
  • 75 Tallant C, Marrero A, Gomis-Rüth FX. Matrix metalloproteinases: Fold and function of their catalytic domains. Biochim Biophys Acta 2010; 1803: 20-28
  • 76 Platta CS, Greenblatt DY, Kunnimalaiyaan M. et al. Valproic acid induces notch1 signaling in small cell lung cancer cells. J Surg Res 2008; 148: 31-37
  • 77 Sakamoto T, Kobayashi S, Yamada D. et al. A histone deacetylase inhibitor suppresses epithelial-mesenchymal transition and attenuates chemoresistance in biliary tract cancer. PloS One 2016; 11: e0145985
  • 78 Jiang G-M, Wang H-S, Fan Zhang F. et al. Histone deacetylase inhibitor induction of epithelial-mesenchymal transitions via up-regulation of snail facilitates cancer progression. Biochim Biophysica Acta 2013; 1833: 663-671
  • 79 Harris SL, Levine AJ. The p53 pathway: Positive and negative feedback loops. Oncogene 2005; 24: 2899-2908
  • 80 Gartel AJ, Radhakrishnan SK. Lost in transcription: p21 repression, mechanisms, and consequences. Cancer Res 2005; 65: 3980-3985
  • 81 Boone TC, Johnson MJ, Clerck YAD. et al. cDNA cloning and expression of a metalloproteinase inhibitor related to tissue inhibitor of metalloproteinases. Proc Natl Acad Sci U S A 1990; 87: 2800-2804
  • 82 Glorieux C, Huang P. Cd137 expression in cancer cells: Regulation and significance. Cancer Commun 2019; 39: 70
  • 83 Liao W-C, Yen H-R, Liao C-K. et al. DSE regulates the malignant characters of hepatocellular carcinoma cells by modulating CCL5/CCR1 axis. Am J Cancer Res 2019; 9: 347-362
  • 84 Liao W-C, Liao C-K, Tsai Y-H. et al. DSE promotes aggressive glioma cell phenotypes by enhancing HB-EGF/ErbB signaling. PloS One 2018; 13: e0198364