#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Mesenchyme-derived IGF2 is a major paracrine regulator of pancreatic growth and function


Autoři: Constanze M. Hammerle aff001;  Ionel Sandovici aff001;  Gemma V. Brierley aff001;  Nicola M. Smith aff001;  Warren E. Zimmer aff004;  Ilona Zvetkova aff001;  Haydn M. Prosser aff005;  Yoichi Sekita aff001;  Brian Y. H. Lam aff001;  Marcella Ma aff001;  Wendy N. Cooper aff001;  Antonio Vidal-Puig aff001;  Susan E. Ozanne aff001;  Gema Medina-Gómez aff006;  Miguel Constância aff001
Působiště autorů: University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrookes Hospital, Cambridge, United Kingdom aff001;  Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, United Kingdom aff002;  Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom aff003;  Department of Medical Physiology, Texas A&M Health Science Center, College Station, Texas, United States of America aff004;  The Wellcome Trust Sanger Institute, Genome Campus, Hinxton, United Kingdom aff005;  Área de Bioquímica y Biología Molecular, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, 28922-Alcorcón, Madrid, Spain aff006
Vyšlo v časopise: Mesenchyme-derived IGF2 is a major paracrine regulator of pancreatic growth and function. PLoS Genet 16(10): e32767. doi:10.1371/journal.pgen.1009069
Kategorie: Research Article
doi: https://doi.org/10.1371/journal.pgen.1009069

Souhrn

The genetic mechanisms that determine the size of the adult pancreas are poorly understood. Imprinted genes, which are expressed in a parent-of-origin-specific manner, are known to have important roles in development, growth and metabolism. However, our knowledge regarding their roles in the control of pancreatic growth and function remains limited. Here we show that many imprinted genes are highly expressed in pancreatic mesenchyme-derived cells and explore the role of the paternally-expressed insulin-like growth factor 2 (Igf2) gene in mesenchymal and epithelial pancreatic lineages using a newly developed conditional Igf2 mouse model. Mesenchyme-specific Igf2 deletion results in acinar and beta-cell hypoplasia, postnatal whole-body growth restriction and maternal glucose intolerance during pregnancy, suggesting that the mesenchyme is a developmental reservoir of IGF2 used for paracrine signalling. The unique actions of mesenchymal IGF2 are demonstrated by the absence of any discernible growth or functional phenotypes upon Igf2 deletion in the developing pancreatic epithelium. Additionally, increased IGF2 levels specifically in the mesenchyme, through conditional Igf2 loss-of-imprinting or Igf2r deletion, leads to pancreatic acinar overgrowth. Furthermore, ex-vivo exposure of primary acinar cells to exogenous IGF2 activates AKT, a key signalling node, and increases their number and amylase production. Based on these findings, we propose that mesenchymal Igf2, and perhaps other imprinted genes, are key developmental regulators of adult pancreas size and function.

Klíčová slova:

Alleles – Body weight – Gene expression – Genetically modified animals – Mouse models – Pancreas – Polymerase chain reaction – Pregnancy


Zdroje

1. Stanger BZ, Tanaka AJ, Melton DA. Organ size is limited by the number of embryonic progenitor cells in the pancreas but not the liver. Nature. 2007;445:886–891.

2. Landsman L, Nijagal A, Whitchurch TJ, Vanderlaan RL, Zimmer WE, Mackenzie TC, et al. Pancreatic mesenchyme regulates epithelial organogenesis throughout development. PLoS Biol. 2011;9:e1001143. doi: 10.1371/journal.pbio.1001143 21909240

3. Harari N, Sakhneny L, Khalifa-Malka L, Busch A, Hertel KJ, Hebrok M, et al. Pancreatic pericytes originate from the embryonic pancreatic mesenchyme. Dev Biol. 2019;449:14–20. doi: 10.1016/j.ydbio.2019.01.020 30771302

4. Hibsher D, Epshtein A, Oren N, Landsman L. Pancreatic Mesenchyme Regulates Islet Cellular Composition in a Patched/Hedgehog-Dependent Manner. Sci Rep. 2016;6:38008. doi: 10.1038/srep38008 27892540

5. Christofori G, Naik P, Hanahan D. A second signal supplied by insulin-like growth factor II in oncogene-induced tumorigenesis. Nature. 1994;369:414–418. doi: 10.1038/369414a0 7910953

6. Stewart CE, Rotwein P. Insulin-like growth factor-II is an autocrine survival factor for differentiating myoblasts. J Biol Chem. 1996;271:11330–11338. doi: 10.1074/jbc.271.19.11330 8626686

7. Petrik J, Arany E, McDonald TJ, Hill DJ. Apoptosis in the pancreatic islet cells of the neonatal rat is associated with a reduced expression of insulin-like growth factor II that may act as a survival factor. Endocrinology. 1998;139:2994–3004. doi: 10.1210/endo.139.6.6042 9607811

8. Bendall SC, Stewart MH, Menendez P, George D, Vijayaragavan K, Werbowetski-Ogilvie T, et al. IGF and FGF cooperatively establish the regulatory stem cell niche of pluripotent human cells in vitro. Nature. 2007;448:1015–1021. doi: 10.1038/nature06027 17625568

9. Ziegler AN, Levison SW, Wood TL. Insulin and IGF receptor signalling in neural-stem-cell homeostasis. Nat Rev Endocrinol. 2015;11:161–170. doi: 10.1038/nrendo.2014.208 25445849

10. Ferrón SR, Radford EJ, Domingo-Muelas A, Kleine I, Ramme A, Gray D, et al. Differential genomic imprinting regulates paracrine and autocrine roles of IGF2 in mouse adult neurogenesis. Nat Commun. 2015;6:8265. doi: 10.1038/ncomms9265 26369386

11. Youssef A, Han VK. Low Oxygen Tension Modulates the Insulin-Like Growth Factor-1 or -2 Signalling via Both Insulin-Like Growth Factor-1 Receptor and Insulin Receptor to Maintain Stem Cell Identity in Placental Mesenchymal Stem Cells. Endocrinology. 2016;157:1163–1174. doi: 10.1210/en.2015-1297 26760116

12. Youssef A, Aboalola D, Han VK. The Roles of Insulin-Like Growth Factors in Mesenchymal Stem Cell Niche. Stem Cells Int. 2017;2017:9453108. doi: 10.1155/2017/9453108 28298931

13. Ziegler AN, Feng Q, Chidambaram S, Testai JM, Kumari E, Rothbard DE, et al. Insulin-like Growth Factor II: An Essential Adult Stem Cell Niche Constituent in Brain and Intestine. Stem Cell Rep. 2019;12:816–830.

14. DeChiara TM, Robertson EJ, Efstratiadis A. Parental imprinting of the mouse insulin-like growth factor II gene. Cell 1991;64:849–859. doi: 10.1016/0092-8674(91)90513-x 1997210

15. Constância M, Kelsey G, Reik W. Resourceful imprinting. Nature. 2004;432:53–57. doi: 10.1038/432053a 15525980

16. Ideraabdullah FY, Vigneau S, Bartolomei MS. Genomic imprinting mechanisms in mammals. Mutat Res. 2008;647:77–85. doi: 10.1016/j.mrfmmm.2008.08.008 18778719

17. Gicquel C, Rossignol S, Cabrol S, Houang M, Steunou V, Barbu V, et al. Epimutation of the telomeric imprinting center region on chromosome 11p15 in Silver-Russell syndrome. Nat Genet. 2005;37:1003–1007. doi: 10.1038/ng1629 16086014

18. Weksberg R, Shen DR, Fei YL, Song QL, Squire J. Disruption of insulin-like growth factor 2 imprinting in Beckwith-Wiedemann syndrome. Nat Genet. 1993;5:143–150. doi: 10.1038/ng1093-143 8252039

19. Efstratiadis A. Genetics of mouse growth. Int J Dev Biol. 1998;42:955–976. 9853827

20. Devedjian JC, George M, Casellas A, Pujol A, Visa J, Pelegrín M, et al. Transgenic mice overexpressing insulin-like growth factor-II in beta cells develop type 2 diabetes. J Clin Invest. 2000;105:731–740. doi: 10.1172/JCI5656 10727441

21. Casellas A, Mallol C, Salavert A, Jimenez V, Garcia M, Agudo J, et al. Insulin-like Growth Factor 2 Overexpression Induces β-Cell Dysfunction and Increases Beta-cell Susceptibility to Damage. J Biol Chem. 2015;290:16772–16785. doi: 10.1074/jbc.M115.642041 25971976

22. Modi H, Jacovetti C, Tarussio D, Metref S, Madsen OD, Zhang FP, et al. Autocrine Action of IGF2 Regulates Adult β-Cell Mass and Function. Diabetes. 2015;64:4148–4157. doi: 10.2337/db14-1735 26384384

23. Petrik J, Pell JM, Arany E, McDonald TJ, Dean WL, Reik W, et al. Overexpression of insulin-like growth factor-II in transgenic mice is associated with pancreatic islet cell hyperplasia. Endocrinology. 1999;140:2353–2363. doi: 10.1210/endo.140.5.6732 10218989

24. Hill DJ, Strutt B, Arany E, Zaina S, Coukell S, Graham CF. Increased and persistent circulating insulin-like growth factor II in neonatal transgenic mice suppresses developmental apoptosis in the pancreatic islets. Endocrinology. 2000;141:1151–1157. doi: 10.1210/endo.141.3.7354 10698192

25. Kido Y, Nakae J, Hribal ML, Xuan S, Efstratiadis A, Accili D. Effects of mutations in the insulin-like growth factor signalling system on embryonic pancreas development and beta-cell compensation to insulin resistance. J Biol Chem. 2002;277:36740–36747. doi: 10.1074/jbc.M206314200 12101187

26. Srinivas S, Watanabe T, Lin CS, William CM, Tanabe Y, Jessell TM, et al. Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Dev Biol. 2001;1:4. doi: 10.1186/1471-213x-1-4 11299042

27. Verzi MP, Stanfel MN, Moses KA, Kim BM, Zhang Y, Schwartz RJ, et al. Role of the homeodomain transcription factor Bapx1 in mouse distal stomach development. Gastroenterology. 2009;136:1701–1710. doi: 10.1053/j.gastro.2009.01.009 19208343

28. Kawaguchi Y, Cooper B, Gannon M, Ray M, MacDonald RJ, Wright CV. The role of the transcriptional regulator Ptf1a in converting intestinal to pancreatic progenitors. Nat Genet. 2002;32:128–134. doi: 10.1038/ng959 12185368

29. Herrera PL. Adult insulin- and glucagon-producing cells differentiate from two independent cell lineages. Development. 2000;127:2317–2322. 10804174

30. Kisanuki YY, Hammer RE, Miyazaki J, Williams SC, Richardson JA, Yanagisawa M. Tie2-Cre transgenic mice: a new model for endothelial cell-lineage analysis in vivo. Dev Biol. 2001;230:230–242. doi: 10.1006/dbio.2000.0106 11161575

31. Prosser HM, Koike-Yusa H, Cooper JD, Law FC, Bradley A. A resource of vectors and ES cells for targeted deletion of microRNAs in mice. Nat Biotechnol. 2011;29:840–845. doi: 10.1038/nbt.1929 21822254

32. Srivastava M, Hsieh S, Grinberg A, Williams-Simons L, Huang SP, Pfeifer K. H19 and Igf2 monoallelic expression is regulated in two distinct ways by a shared cis acting regulatory region upstream of H19. Genes Dev. 2000;14:1186–1195. 10817754

33. Ghosh P, Dahms NM, Kornfeld S. Mannose 6-phosphate receptors: new twists in the tale. Nat Rev Mol Cell Biol. 2003;4:202–212. doi: 10.1038/nrm1050 12612639

34. Lowe ME, Kaplan MH, Jackson-Grusby L, D'Agostino D, Grusby MJ. Decreased neonatal dietary fat absorption and T cell cytotoxicity in pancreatic lipase-related protein 2-deficient mice. J Biol Chem. 1998;273:31215–31221. doi: 10.1074/jbc.273.47.31215 9813028

35. D'Agostino D, Cordle RA, Kullman J, Erlanson-Albertsson C, Muglia LJ, Lowe ME. Decreased postnatal survival and altered body weight regulation in procolipase-deficient mice. J Biol Chem. 2002;277:7170–7177. doi: 10.1074/jbc.M108328200 11751900

36. Le Y, Zhou Y, Iribarren P, Wang J. Chemokines and chemokine receptors: their manifold roles in homeostasis and disease. Cell Mol Immunol. 2004;1:95–104. 16212895

37. Grady T, Liang P, Ernst SA, Logsdon CD. Chemokine gene expression in rat pancreatic acinar cells is an early event associated with acute pancreatitis. Gastroenterology. 1997;113:1966–1975. doi: 10.1016/s0016-5085(97)70017-9 9394737

38. Apelqvist A, Ahlgren U, Edlund H. Sonic hedgehog directs specialised mesoderm differentiation in the intestine and pancreas. Curr Biol. 1997;7:801–804. doi: 10.1016/s0960-9822(06)00340-x 9368764

39. Bhushan A, Itoh N, Kato S, Thiery JP, Czernichow P, Bellusci S, et al. Fgf10 is essential for maintaining the proliferative capacity of epithelial progenitor cells during early pancreatic organogenesis. Development. 2001;128:5109–5117. 11748146

40. Kawahira H, Scheel DW, Smith SB, German MS, Hebrok M. Hedgehog signalling regulates expansion of pancreatic epithelial cells. Dev Biol. 2005;280:111–121. doi: 10.1016/j.ydbio.2005.01.008 15766752

41. Jonckheere N, Mayes E, Shih HP, Li B, Lioubinski O, Dai X, et al. Analysis of mPygo2 mutant mice suggests a requirement for mesenchymal Wnt signalling in pancreatic growth and differentiation. Dev Biol. 2008;318:224–235. doi: 10.1016/j.ydbio.2008.03.014 18452912

42. Ahnfelt-Rønne J, Ravassard P, Pardanaud-Glavieux C, Scharfmann R, Serup P. Mesenchymal bone morphogenetic protein signalling is required for normal pancreas development. Diabetes. 2010;59:1948–1956. doi: 10.2337/db09-1010 20522595

43. Ludwig CU, Menke A, Adler G, Lutz MP. Fibroblasts stimulate acinar cell proliferation through IGF-I during regeneration from acute pancreatitis. Am J Physiol. 1999;276:G193–G198. doi: 10.1152/ajpgi.1999.276.1.G193 9886995

44. Lu Y, Herrera PL, Guo Y, Sun D, Tang Z, LeRoith D, et al. Pancreatic-specific inactivation of IGF-I gene causes enlarged pancreatic islets and significant resistance to diabetes. Diabetes. 2004;53:3131–3141. doi: 10.2337/diabetes.53.12.3131 15561943

45. Varrault A, Gueydan C, Delalbre A, Bellmann A, Houssami S, Aknin C, et al. Zac1 regulates an imprinted gene network critically involved in the control of embryonic growth. Dev Cell. 2006;11:711–722. doi: 10.1016/j.devcel.2006.09.003 17084362

46. Burns JL, Hassan AB. Cell survival and proliferation are modified by insulin-like growth factor 2 between days 9 and 10 of mouse gestation. Development. 2001;128:3819–3830. 11585807

47. Thorvaldsen JL, Duran KL, Bartolomei MS. Deletion of the H19 differentially methylated domain results in loss of imprinted expression of H19 and Igf2. Genes Dev. 1998;12:3693–3702. doi: 10.1101/gad.12.23.3693 9851976

48. Keniry A, Oxley D, Monnier P, Kyba M, Dandolo L, Smits G, et al. The H19 lincRNA is a developmental reservoir of miR-675 that suppresses growth and Igf1r. Nat Cell Biol. 2012; 14:659–665. doi: 10.1038/ncb2521 22684254

49. Okamoto T, Katada T, Murayama Y, Ui M, Ogata E, Nishimoto I. A simple structure encodes G protein-activating function of the IGF-II/mannose 6-phosphate receptor. Cell 1990;62:709–717. doi: 10.1016/0092-8674(90)90116-v 2167177

50. Leighton PA, Ingram RS, Eggenschwiler J, Efstratiadis A, Tilghman SM. Disruption of imprinting caused by deletion of the H19 gene region in mice. Nature. 1995;375:34–39. doi: 10.1038/375034a0 7536897

51. Wang ZQ, Fung MR, Barlow DP, Wagner EF. Regulation of embryonic growth and lysosomal targeting by the imprinted Igf2/Mpr gene. Nature. 1994;372:464–467. doi: 10.1038/372464a0 7984240

52. Wylie AA, Pulford DJ, McVie-Wylie AJ, Waterland RA, Evans HK, Chen YT, et al. Tissue-specific inactivation of murine M6P/IGF2R. Am J Pathol. 2003;162:321–328. doi: 10.1016/S0002-9440(10)63823-0 12507915

53. Schwenk F, Baron U, Rajewsky K. A cre-transgenic mouse strain for the ubiquitous deletion of loxP-flanked gene segments including deletion in germ cells. Nucleic Acids Res. 1995;23:5080–5081. doi: 10.1093/nar/23.24.5080 8559668

54. Feil R, Walter J, Allen ND, Reik W. Developmental control of allelic methylation in the imprinted mouse Igf2 and H19 genes. Development. 1994;120:2933–2943. 7607083

55. Constância M, Angiolini E, Sandovici I, Smith P, Smith R, Kelsey G, et al. Adaptation of nutrient supply to fetal demand in the mouse involves interaction between the Igf2 gene and placental transporter systems. Proc Natl Acad Sci USA. 2005;102:19219–19224. doi: 10.1073/pnas.0504468103 16365304

56. Simmons DG, Rawn S, Davies A, Hughes M, Cross JC. Spatial and temporal expression of the 23 murine Prolactin/Placental Lactogen-related genes is not associated with their position in the locus. BMC Genomics. 2008;9:352. doi: 10.1186/1471-2164-9-352 18662396

57. Feroze-Merzoug F, Berquin IM, Dey J, Chen YQ. Peptidylprolyl isomerase A (PPIA) as a preferred internal control over GAPDH and beta-actin in quantitative RNA analyses. Biotechniques. 2002;32:776–778. doi: 10.2144/02324st03 11962599

58. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25:402–408. doi: 10.1006/meth.2001.1262 11846609

59. Kim D, Pertea G, Trapnell C, Pimentel H, Kelley R, Salzberg SL. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 2013;14:R36. doi: 10.1186/gb-2013-14-4-r36 23618408

60. Trapnell C, Williams BA, Pertea G, Mortazavi A, Kwan G, van Baren MJ, et al. Transcript assembly and quantification by RNA-seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat Biotechnol. 2010;28:511–515. doi: 10.1038/nbt.1621 20436464

61. Supek F, Bošnjak M, Škunca N, Šmuc T. REVIGO summarizes and visualizes long lists of gene ontology terms. PLoS One. 2011;6:e21800. doi: 10.1371/journal.pone.0021800 21789182

62. Gout J, Pommier RM, Vincent DF, Kaniewski B, Martel S, Valcourt U, et al. Isolation and culture of mouse primary pancreatic acinar cells. J Vis Exp. 2013;13:78.

63. Sandovici I, Georgopoulou A, Hufnagel AS, Schiefer SN, Santos F, Hoelle K, et al. Fetus-derived IGF2 matches placental development to fetal demand. bioRxiv. 2019; https://doi.org/10.1101/520536.


Článek vyšel v časopise

PLOS Genetics


2020 Číslo 10
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

Svět praktické medicíny 1/2024 (znalostní test z časopisu)
nový kurz

Koncepce osteologické péče pro gynekology a praktické lékaře
Autoři: MUDr. František Šenk

Sekvenční léčba schizofrenie
Autoři: MUDr. Jana Hořínková

Hypertenze a hypercholesterolémie – synergický efekt léčby
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Význam metforminu pro „udržitelnou“ terapii diabetu
Autoři: prof. MUDr. Milan Kvapil, CSc., MBA

Všechny kurzy
Kurzy Podcasty Doporučená témata Časopisy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#