Skip to main content
Erschienen in: Diabetologia 9/2015

01.09.2015 | Article

Prolactin-stimulated survivin induction is required for beta cell mass expansion during pregnancy in mice

verfasst von: Yili Xu, Xiaojing Wang, Li Gao, Jiayu Zhu, Hui Zhang, Houxia Shi, Minna Woo, Xiaohong Wu

Erschienen in: Diabetologia | Ausgabe 9/2015

Einloggen, um Zugang zu erhalten

Abstract

Aims/hypothesis

Prolactin (PRL)-stimulated beta cell proliferation is critical for maternal pancreatic beta cell mass expansion during pregnancy. However, the molecular effectors of the multiple putative signalling pathways downstream of the PRL receptor (PRL-R) are still elusive. Survivin has been shown to be induced during pregnancy. The aim of the present study was to define the essential role of survivin in gestational beta cell mass expansion.

Methods

Expression of survivin was assessed in mouse islets during pregnancy and in insulinoma cells (INS-1) stimulated with PRL. Pregnant mice with targeted deletion of the survivin gene (also known as Birc5) in beta cells were assessed to determine the essential function of survivin in maternal beta cell mass expansion. INS-1 cells stimulated with PRL were used to explore the role of survivin in signalling pathways downstream of the PRL-R.

Results

Survivin was significantly upregulated in maternal islets during pregnancy. With PRL stimulation, induction of survivin expression occurred predominantly in the nucleus and was associated with cell cycle progression to S and G2/M phase. Beta cell-specific survivin-knockout pregnant mice displayed glucose intolerance, attenuated beta cell mass expansion and impaired beta cell proliferation, with significant attenuation in the increased expression of Cdk4/Ccnd1, E2f1, p53 (also known as Trp53) and p21 (also known as Cdkn1a) compared with wild-type controls during pregnancy. Targeted deletion of survivin in INS-1 cells resulted in cell cycle disturbance with an arrest in G1/S phase after PRL stimulation. Inhibitors of Akt, signal transducer and activator of transcription 5 (STAT5), PIM or extracellular signal-regulated kinase (ERK), significantly decreased the expression of survivin in PRL-stimulated INS-1 cells.

Conclusions/interpretation

Survivin directly participates in PRL-mediated beta cell proliferation via Akt, STAT5–PIM and ERK signalling pathways during pregnancy.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat Sorenson RL, Brelje TC (1997) Adaptation of islets of Langerhans to pregnancy: beta-cell growth, enhanced insulin secretion and the role of lactogenic hormones. Horm Metab Res 29:301–307PubMedCrossRef Sorenson RL, Brelje TC (1997) Adaptation of islets of Langerhans to pregnancy: beta-cell growth, enhanced insulin secretion and the role of lactogenic hormones. Horm Metab Res 29:301–307PubMedCrossRef
2.
Zurück zum Zitat Parsons JA, Brelje TC, Sorenson RL (1992) Adaptation of islets of Langerhans to pregnancy: increased islet cell proliferation and insulin secretion correlates with the onset of placental lactogen secretion. Endocrinology 130:1459–1466PubMed Parsons JA, Brelje TC, Sorenson RL (1992) Adaptation of islets of Langerhans to pregnancy: increased islet cell proliferation and insulin secretion correlates with the onset of placental lactogen secretion. Endocrinology 130:1459–1466PubMed
3.
Zurück zum Zitat Van Assche FA, Aerts L, De Prins F (1978) A morphological study of the endocrine pancreas in human pregnancy. Br J Obstet Gynaecol 85:818–820PubMedCrossRef Van Assche FA, Aerts L, De Prins F (1978) A morphological study of the endocrine pancreas in human pregnancy. Br J Obstet Gynaecol 85:818–820PubMedCrossRef
4.
Zurück zum Zitat Butler AE, Cao-Minh L, Galasso R et al (2010) Adaptive changes in pancreatic beta cell fractional area and beta cell turnover in human pregnancy. Diabetologia 53:2167–2176PubMedCentralPubMedCrossRef Butler AE, Cao-Minh L, Galasso R et al (2010) Adaptive changes in pancreatic beta cell fractional area and beta cell turnover in human pregnancy. Diabetologia 53:2167–2176PubMedCentralPubMedCrossRef
5.
Zurück zum Zitat Kim C, Newton KM, Knopp RH (2002) Gestational diabetes and the incidence of type 2 diabetes: a systematic review. Diabetes Care 25:1862–1868PubMedCrossRef Kim C, Newton KM, Knopp RH (2002) Gestational diabetes and the incidence of type 2 diabetes: a systematic review. Diabetes Care 25:1862–1868PubMedCrossRef
6.
Zurück zum Zitat Brelje TC, Stout LE, Bhagroo NV, Sorenson RL (2004) Distinctive roles for prolactin and growth hormone in the activation of signal transducer and activator of transcription 5 in pancreatic islets of Langerhans. Endocrinology 145:4162–4175PubMedCrossRef Brelje TC, Stout LE, Bhagroo NV, Sorenson RL (2004) Distinctive roles for prolactin and growth hormone in the activation of signal transducer and activator of transcription 5 in pancreatic islets of Langerhans. Endocrinology 145:4162–4175PubMedCrossRef
7.
Zurück zum Zitat Amaral ME, Cunha DA, Anhe GF et al (2004) Participation of prolactin receptors and phosphatidylinositol 3-kinase and MAP kinase pathways in the increase in pancreatic islet mass and sensitivity to glucose during pregnancy. J Endocrinol 183:469–476PubMedCrossRef Amaral ME, Cunha DA, Anhe GF et al (2004) Participation of prolactin receptors and phosphatidylinositol 3-kinase and MAP kinase pathways in the increase in pancreatic islet mass and sensitivity to glucose during pregnancy. J Endocrinol 183:469–476PubMedCrossRef
8.
Zurück zum Zitat Amaral ME, Ueno M, Carvalheira JB et al (2003) Prolactin-signal transduction in neonatal rat pancreatic islets and interaction with the insulin-signaling pathway. Horm Metab Res 35:282–289PubMedCrossRef Amaral ME, Ueno M, Carvalheira JB et al (2003) Prolactin-signal transduction in neonatal rat pancreatic islets and interaction with the insulin-signaling pathway. Horm Metab Res 35:282–289PubMedCrossRef
9.
Zurück zum Zitat Hugl SR, Merger M (2007) Prolactin stimulates proliferation of the glucose-dependent beta-cell line INS-1 via different IRS-proteins. JOP 8:739–752PubMed Hugl SR, Merger M (2007) Prolactin stimulates proliferation of the glucose-dependent beta-cell line INS-1 via different IRS-proteins. JOP 8:739–752PubMed
10.
Zurück zum Zitat Friedrichsen BN, Galsgaard ED, Nielsen JH, Moldrup A (2001) Growth hormone- and prolactin-induced proliferation of insulinoma cells, INS-1, depends on activation of STAT5 (signal transducer and activator of transcription 5). Mol Endocrinol 15:136–148PubMedCrossRef Friedrichsen BN, Galsgaard ED, Nielsen JH, Moldrup A (2001) Growth hormone- and prolactin-induced proliferation of insulinoma cells, INS-1, depends on activation of STAT5 (signal transducer and activator of transcription 5). Mol Endocrinol 15:136–148PubMedCrossRef
11.
Zurück zum Zitat Mita AC, Mita MM, Nawrocki ST, Giles FJ (2008) Survivin: key regulator of mitosis and apoptosis and novel target for cancer therapeutics. Clin Cancer Res 14:5000–5005PubMedCrossRef Mita AC, Mita MM, Nawrocki ST, Giles FJ (2008) Survivin: key regulator of mitosis and apoptosis and novel target for cancer therapeutics. Clin Cancer Res 14:5000–5005PubMedCrossRef
12.
Zurück zum Zitat Altieri DC (2008) Survivin, cancer networks and pathway-directed drug discovery. Nat Rev Cancer 8:61–70PubMedCrossRef Altieri DC (2008) Survivin, cancer networks and pathway-directed drug discovery. Nat Rev Cancer 8:61–70PubMedCrossRef
14.
Zurück zum Zitat Wu X, Wang L, Schroer S et al (2009) Perinatal survivin is essential for the establishment of pancreatic beta cell mass in mice. Diabetologia 52:2130–2141PubMedCrossRef Wu X, Wang L, Schroer S et al (2009) Perinatal survivin is essential for the establishment of pancreatic beta cell mass in mice. Diabetologia 52:2130–2141PubMedCrossRef
15.
Zurück zum Zitat Jiang Y, Nishimura W, Devor-Henneman D et al (2008) Postnatal expansion of the pancreatic beta-cell mass is dependent on survivin. Diabetes 57:2718–2727PubMedCentralPubMedCrossRef Jiang Y, Nishimura W, Devor-Henneman D et al (2008) Postnatal expansion of the pancreatic beta-cell mass is dependent on survivin. Diabetes 57:2718–2727PubMedCentralPubMedCrossRef
16.
Zurück zum Zitat Wu X, Zhang Q, Wang X et al (2012) Survivin is required for beta-cell mass expansion in the pancreatic duct-ligated mouse model. Plos One 7:e41976PubMedCentralPubMedCrossRef Wu X, Zhang Q, Wang X et al (2012) Survivin is required for beta-cell mass expansion in the pancreatic duct-ligated mouse model. Plos One 7:e41976PubMedCentralPubMedCrossRef
18.
Zurück zum Zitat Hakonen E, Ustinov J, Palgi J, Miettinen PJ, Otonkoski T (2014) EGFR signaling promotes beta-cell proliferation and survivin expression during pregnancy. Plos One 9:e93651PubMedCentralPubMedCrossRef Hakonen E, Ustinov J, Palgi J, Miettinen PJ, Otonkoski T (2014) EGFR signaling promotes beta-cell proliferation and survivin expression during pregnancy. Plos One 9:e93651PubMedCentralPubMedCrossRef
19.
Zurück zum Zitat Nguyen KT, Tajmir P, Lin CH et al (2006) Essential role of Pten in body size determination and pancreatic beta-cell homeostasis in vivo. Mol Cell Biol 26:4511–4518PubMedCentralPubMedCrossRef Nguyen KT, Tajmir P, Lin CH et al (2006) Essential role of Pten in body size determination and pancreatic beta-cell homeostasis in vivo. Mol Cell Biol 26:4511–4518PubMedCentralPubMedCrossRef
20.
Zurück zum Zitat Liadis N, Murakami K, Eweida M et al (2005) Caspase-3-dependent beta-cell apoptosis in the initiation of autoimmune diabetes mellitus. Mol Cell Biol 25:3620–3629PubMedCentralPubMedCrossRef Liadis N, Murakami K, Eweida M et al (2005) Caspase-3-dependent beta-cell apoptosis in the initiation of autoimmune diabetes mellitus. Mol Cell Biol 25:3620–3629PubMedCentralPubMedCrossRef
21.
Zurück zum Zitat Xue Y, Liu C, Xu Y et al (2010) Study on pancreatic islet adaptation and gene expression during pregnancy in rats. Endocrine 37:83–97PubMedCrossRef Xue Y, Liu C, Xu Y et al (2010) Study on pancreatic islet adaptation and gene expression during pregnancy in rats. Endocrine 37:83–97PubMedCrossRef
22.
Zurück zum Zitat Wang H, Gambosova K, Cooper ZA et al (2010) EGF regulates survivin stability through the Raf-1/ERK pathway in insulin-secreting pancreatic beta-cells. BMC Mol Biol 11:66PubMedCentralPubMedCrossRef Wang H, Gambosova K, Cooper ZA et al (2010) EGF regulates survivin stability through the Raf-1/ERK pathway in insulin-secreting pancreatic beta-cells. BMC Mol Biol 11:66PubMedCentralPubMedCrossRef
24.
Zurück zum Zitat Weinhaus AJ, Stout LE, Bhagroo NV, Brelje TC, Sorenson RL (2007) Regulation of glucokinase in pancreatic islets by prolactin: a mechanism for increasing glucose-stimulated insulin secretion during pregnancy. J Endocrinol 193:367–381PubMedCrossRef Weinhaus AJ, Stout LE, Bhagroo NV, Brelje TC, Sorenson RL (2007) Regulation of glucokinase in pancreatic islets by prolactin: a mechanism for increasing glucose-stimulated insulin secretion during pregnancy. J Endocrinol 193:367–381PubMedCrossRef
25.
Zurück zum Zitat Parsons JA, Bartke A, Sorenson RL (1995) Number and size of islets of Langerhans in pregnant, human growth hormone-expressing transgenic, and pituitary dwarf mice: effect of lactogenic hormones. Endocrinology 136:2013–2021PubMed Parsons JA, Bartke A, Sorenson RL (1995) Number and size of islets of Langerhans in pregnant, human growth hormone-expressing transgenic, and pituitary dwarf mice: effect of lactogenic hormones. Endocrinology 136:2013–2021PubMed
26.
Zurück zum Zitat Freemark M, Avril I, Fleenor D et al (2002) Targeted deletion of the PRL receptor: effects on islet development, insulin production, and glucose tolerance. Endocrinology 143:1378–1385PubMedCrossRef Freemark M, Avril I, Fleenor D et al (2002) Targeted deletion of the PRL receptor: effects on islet development, insulin production, and glucose tolerance. Endocrinology 143:1378–1385PubMedCrossRef
27.
Zurück zum Zitat Huang C, Snider F, Cross JC (2009) Prolactin receptor is required for normal glucose homeostasis and modulation of beta-cell mass during pregnancy. Endocrinology 150:1618–1626PubMedCrossRef Huang C, Snider F, Cross JC (2009) Prolactin receptor is required for normal glucose homeostasis and modulation of beta-cell mass during pregnancy. Endocrinology 150:1618–1626PubMedCrossRef
28.
Zurück zum Zitat Zhang H, Zhang J, Pope CF et al (2010) Gestational diabetes mellitus resulting from impaired beta-cell compensation in the absence of FoxM1, a novel downstream effector of placental lactogen. Diabetes 59:143–152PubMedCentralPubMedCrossRef Zhang H, Zhang J, Pope CF et al (2010) Gestational diabetes mellitus resulting from impaired beta-cell compensation in the absence of FoxM1, a novel downstream effector of placental lactogen. Diabetes 59:143–152PubMedCentralPubMedCrossRef
29.
Zurück zum Zitat Gianani R, Campbell-Thompson M, Sarkar SA et al (2010) Dimorphic histopathology of long-standing childhood-onset diabetes. Diabetologia 53:690–698PubMedCrossRef Gianani R, Campbell-Thompson M, Sarkar SA et al (2010) Dimorphic histopathology of long-standing childhood-onset diabetes. Diabetologia 53:690–698PubMedCrossRef
30.
Zurück zum Zitat Hasel C, Bhanot UK, Heydrich R, Strater J, Moller P (2005) Parenchymal regression in chronic pancreatitis spares islets reprogrammed for the expression of NFκB and IAPs. Lab Invest 85:1263–1275PubMedCrossRef Hasel C, Bhanot UK, Heydrich R, Strater J, Moller P (2005) Parenchymal regression in chronic pancreatitis spares islets reprogrammed for the expression of NFκB and IAPs. Lab Invest 85:1263–1275PubMedCrossRef
31.
Zurück zum Zitat Liggins C, Orlicky DJ, Bloomquist LA, Gianani R (2003) Developmentally regulated expression of survivin in human pancreatic islets. Pediatr Dev Pathol 6:392–397PubMedCrossRef Liggins C, Orlicky DJ, Bloomquist LA, Gianani R (2003) Developmentally regulated expression of survivin in human pancreatic islets. Pediatr Dev Pathol 6:392–397PubMedCrossRef
33.
Zurück zum Zitat Chen J, Jackson PK, Kirschner MW, Dutta A (1995) Separate domains of p21 involved in the inhibition of Cdk kinase and PCNA. Nature 374:386–388PubMedCrossRef Chen J, Jackson PK, Kirschner MW, Dutta A (1995) Separate domains of p21 involved in the inhibition of Cdk kinase and PCNA. Nature 374:386–388PubMedCrossRef
34.
Zurück zum Zitat Goubin F, Ducommun B (1995) Identification of binding domains on the p21Cip1 cyclin-dependent kinase inhibitor. Oncogene 10:2281–2287PubMed Goubin F, Ducommun B (1995) Identification of binding domains on the p21Cip1 cyclin-dependent kinase inhibitor. Oncogene 10:2281–2287PubMed
35.
Zurück zum Zitat LaBaer J, Garrett MD, Stevenson LF et al (1997) New functional activities for the p21 family of CDK inhibitors. Genes Dev 11:847–862PubMedCrossRef LaBaer J, Garrett MD, Stevenson LF et al (1997) New functional activities for the p21 family of CDK inhibitors. Genes Dev 11:847–862PubMedCrossRef
36.
Zurück zum Zitat Cheng M, Olivier P, Diehl JA et al (1999) The p21(Cip1) and p27(Kip1) CDK ‘inhibitors’ are essential activators of cyclin D-dependent kinases in murine fibroblasts. EMBO J 18:1571–1583PubMedCentralPubMedCrossRef Cheng M, Olivier P, Diehl JA et al (1999) The p21(Cip1) and p27(Kip1) CDK ‘inhibitors’ are essential activators of cyclin D-dependent kinases in murine fibroblasts. EMBO J 18:1571–1583PubMedCentralPubMedCrossRef
37.
Zurück zum Zitat Li F, Ackermann EJ, Bennett CF et al (1999) Pleiotropic cell-division defects and apoptosis induced by interference with survivin function. Nat Cell Biol 1:461–466PubMedCrossRef Li F, Ackermann EJ, Bennett CF et al (1999) Pleiotropic cell-division defects and apoptosis induced by interference with survivin function. Nat Cell Biol 1:461–466PubMedCrossRef
38.
Zurück zum Zitat Ezhevsky SA, Nagahara H, Vocero-Akbani AM, Gius DR, Wei MC, Dowdy SF (1997) Hypo-phosphorylation of the retinoblastoma protein (pRb) by cyclin D:Cdk4/6 complexes results in active pRb. Proc Natl Acad Sci U S A 94:10699–10704PubMedCentralPubMedCrossRef Ezhevsky SA, Nagahara H, Vocero-Akbani AM, Gius DR, Wei MC, Dowdy SF (1997) Hypo-phosphorylation of the retinoblastoma protein (pRb) by cyclin D:Cdk4/6 complexes results in active pRb. Proc Natl Acad Sci U S A 94:10699–10704PubMedCentralPubMedCrossRef
39.
Zurück zum Zitat Resnitzky D, Reed SI (1995) Different roles for cyclins D1 and E in regulation of the G1-to-S transition. Mol Cell Biol 15:3463–3469PubMedCentralPubMed Resnitzky D, Reed SI (1995) Different roles for cyclins D1 and E in regulation of the G1-to-S transition. Mol Cell Biol 15:3463–3469PubMedCentralPubMed
40.
Zurück zum Zitat Le TN, Elsea SH, Romero R, Chaiworapongsa T, Francis GL (2013) Prolactin receptor gene polymorphisms are associated with gestational diabetes. Genet Test Mol Biomarkers 17:567–571PubMedCentralPubMedCrossRef Le TN, Elsea SH, Romero R, Chaiworapongsa T, Francis GL (2013) Prolactin receptor gene polymorphisms are associated with gestational diabetes. Genet Test Mol Biomarkers 17:567–571PubMedCentralPubMedCrossRef
41.
Zurück zum Zitat Kalin TV, Ustiyan V, Kalinichenko VV (2011) Multiple faces of FoxM1 transcription factor: lessons from transgenic mouse models. Cell Cycle 10:396–405PubMedCentralPubMedCrossRef Kalin TV, Ustiyan V, Kalinichenko VV (2011) Multiple faces of FoxM1 transcription factor: lessons from transgenic mouse models. Cell Cycle 10:396–405PubMedCentralPubMedCrossRef
42.
Zurück zum Zitat Wang IC, Chen YJ, Hughes D et al (2005) Forkhead box M1 regulates the transcriptional network of genes essential for mitotic progression and genes encoding the SCF (Skp2-Cks1) ubiquitin ligase. Mol Cell Biol 25:10875–10894PubMedCentralPubMedCrossRef Wang IC, Chen YJ, Hughes D et al (2005) Forkhead box M1 regulates the transcriptional network of genes essential for mitotic progression and genes encoding the SCF (Skp2-Cks1) ubiquitin ligase. Mol Cell Biol 25:10875–10894PubMedCentralPubMedCrossRef
43.
Zurück zum Zitat Gannon M, Shiota C, Postic C, Wright CV, Magnuson M (2000) Analysis of the Cre-mediated recombination driven by rat insulin promoter in embryonic and adult mouse pancreas. Genesis 26:139–142PubMedCrossRef Gannon M, Shiota C, Postic C, Wright CV, Magnuson M (2000) Analysis of the Cre-mediated recombination driven by rat insulin promoter in embryonic and adult mouse pancreas. Genesis 26:139–142PubMedCrossRef
44.
Zurück zum Zitat Brouwers B, de Faudeur G, Osipovich AB et al (2014) Impaired islet function in commonly used transgenic mouse lines due to human growth hormone minigene expression. Cell Metab 20:979–990PubMedCrossRef Brouwers B, de Faudeur G, Osipovich AB et al (2014) Impaired islet function in commonly used transgenic mouse lines due to human growth hormone minigene expression. Cell Metab 20:979–990PubMedCrossRef
Metadaten
Titel
Prolactin-stimulated survivin induction is required for beta cell mass expansion during pregnancy in mice
verfasst von
Yili Xu
Xiaojing Wang
Li Gao
Jiayu Zhu
Hui Zhang
Houxia Shi
Minna Woo
Xiaohong Wu
Publikationsdatum
01.09.2015
Verlag
Springer Berlin Heidelberg
Erschienen in
Diabetologia / Ausgabe 9/2015
Print ISSN: 0012-186X
Elektronische ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-015-3670-0

Weitere Artikel der Ausgabe 9/2015

Diabetologia 9/2015 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.