Skip to main content
Erschienen in: Diabetologia 10/2017

04.07.2017 | Article

MicroRNAs modulate core-clock gene expression in pancreatic islets during early postnatal life in rats

verfasst von: Cécile Jacovetti, Adriana Rodriguez-Trejo, Claudiane Guay, Jonathan Sobel, Sonia Gattesco, Volodymyr Petrenko, Camille Saini, Charna Dibner, Romano Regazzi

Erschienen in: Diabetologia | Ausgabe 10/2017

Einloggen, um Zugang zu erhalten

Abstract

Aims/hypothesis

Evidence continues to emerge detailing a fine-tuning of the regulation of metabolic processes and energy homeostasis by cell-autonomous circadian clocks. Pancreatic beta cell functional maturation occurs after birth and implies transcriptional changes triggered by a shift in the nutritional supply that occurs at weaning, enabling the adaptation of insulin secretion. So far, the developmental timing and exact mechanisms involved in the initiation of the circadian clock in the growing pancreatic islets have never been addressed.

Methods

Circadian gene expression was measured by quantitative RT-PCR in islets of rats at different postnatal ages up to 3 months, and by in vitro bioluminescence recording in newborn (10-day-old) and adult (3-month-old) islets. The effect of the microRNAs miR-17-5p and miR-29b-3p on the expression of target circadian genes was assessed in newborn rat islets transfected with microRNA antisense or mimic oligonucleotides, and luciferase reporter assays were performed on the rat insulin-secreting cell line INS832/13 to determine a direct effect. The global regulatory network between microRNAs and circadian genes was computationally predicted.

Results

We found up to a sixfold-change in the 24 h transcriptional oscillations and overall expression of Clock, Npas2, Bmal1, Bmal2, Rev-erbα, Per1, Per2, Per3 and Cry2 between newborn and adult rat islets. Synchronisation of the clock machinery in cultured islet cells revealed a delayed cell-autonomous rhythmicity of about 1.5 h in newborn compared with adult rats. Computational predictions unveiled the existence of a complex regulatory network linking over 40 microRNAs displaying modifications in their expression profiles during postnatal beta cell maturation and key core-clock genes. In agreement with these computational predictions, we demonstrated that miR-17-5p and miR-29b-3p directly regulated circadian gene expression in the maturing islet cells of 10-day-old rats.

Conclusions/interpretation

These data show that the circadian clock is not fully operational in newborn islets and that microRNAs potently contribute to its regulation during postnatal beta cell maturation. Defects in this process may have long-term consequences on circadian physiology and pancreatic islet function, favouring the manifestation of metabolic diseases such as diabetes.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat Jacovetti C, Matkovich SJ, Rodriguez-Trejo A, Guay C, Regazzi R (2015) Postnatal beta-cell maturation is associated with islet-specific microRNA changes induced by nutrient shifts at weaning. Nat Commun 6:8084CrossRefPubMedPubMedCentral Jacovetti C, Matkovich SJ, Rodriguez-Trejo A, Guay C, Regazzi R (2015) Postnatal beta-cell maturation is associated with islet-specific microRNA changes induced by nutrient shifts at weaning. Nat Commun 6:8084CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Pildes RS, Hart RJ, Warrner R, Cornblath M (1969) Plasma insulin response during oral glucose tolerance tests in newborns of normal and gestational diabetic mothers. Pediatrics 44:76–83PubMed Pildes RS, Hart RJ, Warrner R, Cornblath M (1969) Plasma insulin response during oral glucose tolerance tests in newborns of normal and gestational diabetic mothers. Pediatrics 44:76–83PubMed
3.
Zurück zum Zitat Rozzo A, Meneghel-Rozzo T, Delakorda SL, Yang SB, Rupnik M (2009) Exocytosis of insulin: in vivo maturation of mouse endocrine pancreas. Ann N Y Acad Sci 1152:53–62CrossRefPubMed Rozzo A, Meneghel-Rozzo T, Delakorda SL, Yang SB, Rupnik M (2009) Exocytosis of insulin: in vivo maturation of mouse endocrine pancreas. Ann N Y Acad Sci 1152:53–62CrossRefPubMed
4.
Zurück zum Zitat Rakshit K, Qian J, Colwell CS, Matveyenko AV (2015) The islet circadian clock: entrainment mechanisms, function and role in glucose homeostasis. Diabetes Obes Metab 17(Suppl 1):115–122CrossRefPubMedPubMedCentral Rakshit K, Qian J, Colwell CS, Matveyenko AV (2015) The islet circadian clock: entrainment mechanisms, function and role in glucose homeostasis. Diabetes Obes Metab 17(Suppl 1):115–122CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Saini C, Petrenko V, Pulimeno P et al (2016) A functional circadian clock is required for proper insulin secretion by human pancreatic islet cells. Diabetes Obes Metab 18:355–365CrossRefPubMed Saini C, Petrenko V, Pulimeno P et al (2016) A functional circadian clock is required for proper insulin secretion by human pancreatic islet cells. Diabetes Obes Metab 18:355–365CrossRefPubMed
6.
Zurück zum Zitat Perelis M, Marcheva B, Ramsey KM et al (2015) Pancreatic beta cell enhancers regulate rhythmic transcription of genes controlling insulin secretion. Science 350:aac4250CrossRefPubMedPubMedCentral Perelis M, Marcheva B, Ramsey KM et al (2015) Pancreatic beta cell enhancers regulate rhythmic transcription of genes controlling insulin secretion. Science 350:aac4250CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Pulimeno P, Mannic T, Sage D et al (2013) Autonomous and self-sustained circadian oscillators displayed in human islet cells. Diabetologia 56:497–507CrossRefPubMed Pulimeno P, Mannic T, Sage D et al (2013) Autonomous and self-sustained circadian oscillators displayed in human islet cells. Diabetologia 56:497–507CrossRefPubMed
8.
Zurück zum Zitat Schibler U, Gotic I, Saini C et al (2015) Clock-talk: interactions between central and peripheral circadian oscillators in mammals. Cold Spring Harb Symp Quant Biol 80:223–232CrossRefPubMed Schibler U, Gotic I, Saini C et al (2015) Clock-talk: interactions between central and peripheral circadian oscillators in mammals. Cold Spring Harb Symp Quant Biol 80:223–232CrossRefPubMed
9.
Zurück zum Zitat Asher G, Schibler U (2011) Crosstalk between components of circadian and metabolic cycles in mammals. Cell Metab 13:125–137CrossRefPubMed Asher G, Schibler U (2011) Crosstalk between components of circadian and metabolic cycles in mammals. Cell Metab 13:125–137CrossRefPubMed
10.
Zurück zum Zitat Dibner C, Schibler U (2015) Circadian timing of metabolism in animal models and humans. J Intern Med 277:513–527CrossRefPubMed Dibner C, Schibler U (2015) Circadian timing of metabolism in animal models and humans. J Intern Med 277:513–527CrossRefPubMed
11.
Zurück zum Zitat Duffield GE (2003) DNA microarray analyses of circadian timing: the genomic basis of biological time. J Neuroendocrinol 15:991–1002CrossRefPubMed Duffield GE (2003) DNA microarray analyses of circadian timing: the genomic basis of biological time. J Neuroendocrinol 15:991–1002CrossRefPubMed
12.
Zurück zum Zitat Panda S, Antoch MP, Miller BH et al (2002) Coordinated transcription of key pathways in the mouse by the circadian clock. Cell 109:307–320CrossRefPubMed Panda S, Antoch MP, Miller BH et al (2002) Coordinated transcription of key pathways in the mouse by the circadian clock. Cell 109:307–320CrossRefPubMed
13.
Zurück zum Zitat Mehta N, Cheng HY (2013) Micro-managing the circadian clock: the role of microRNAs in biological timekeeping. J Mol Biol 425:3609–3624CrossRefPubMed Mehta N, Cheng HY (2013) Micro-managing the circadian clock: the role of microRNAs in biological timekeeping. J Mol Biol 425:3609–3624CrossRefPubMed
14.
Zurück zum Zitat Asher G, Sassone-Corsi P (2015) Time for food: the intimate interplay between nutrition, metabolism, and the circadian clock. Cell 161:84–92CrossRefPubMed Asher G, Sassone-Corsi P (2015) Time for food: the intimate interplay between nutrition, metabolism, and the circadian clock. Cell 161:84–92CrossRefPubMed
16.
Zurück zum Zitat Ribas-Latre A, Eckel-Mahan K (2016) Interdependence of nutrient metabolism and the circadian clock system: importance for metabolic health. Mol Metab 5:133–152CrossRefPubMedPubMedCentral Ribas-Latre A, Eckel-Mahan K (2016) Interdependence of nutrient metabolism and the circadian clock system: importance for metabolic health. Mol Metab 5:133–152CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Perrin L, Loizides-Mangold U, Skarupelova S et al (2015) Human skeletal myotubes display a cell-autonomous circadian clock implicated in basal myokine secretion. Mol Metab 4:834–845CrossRefPubMedPubMedCentral Perrin L, Loizides-Mangold U, Skarupelova S et al (2015) Human skeletal myotubes display a cell-autonomous circadian clock implicated in basal myokine secretion. Mol Metab 4:834–845CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Ansari N, Agathagelidis M, Lee C, Korf HW, von Gall C (2009) Differential maturation of circadian rhythms in clock gene proteins in the suprachiasmatic nucleus and the pars tuberalis during mouse ontogeny. Eur J Neurosci 29:477–489CrossRefPubMedPubMedCentral Ansari N, Agathagelidis M, Lee C, Korf HW, von Gall C (2009) Differential maturation of circadian rhythms in clock gene proteins in the suprachiasmatic nucleus and the pars tuberalis during mouse ontogeny. Eur J Neurosci 29:477–489CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Yamazaki S, Yoshikawa T, Biscoe EW et al (2009) Ontogeny of circadian organization in the rat. J Biol Rhythm 24:55–63CrossRef Yamazaki S, Yoshikawa T, Biscoe EW et al (2009) Ontogeny of circadian organization in the rat. J Biol Rhythm 24:55–63CrossRef
20.
Zurück zum Zitat Sumova A, Bendova Z, Sladek M et al (2006) Setting the biological time in central and peripheral clocks during ontogenesis. FEBS Lett 580:2836–2842CrossRefPubMed Sumova A, Bendova Z, Sladek M et al (2006) Setting the biological time in central and peripheral clocks during ontogenesis. FEBS Lett 580:2836–2842CrossRefPubMed
22.
23.
Zurück zum Zitat Balsalobre A, Brown SA, Marcacci L et al (2000) Resetting of circadian time in peripheral tissues by glucocorticoid signaling. Science 289:2344–2347CrossRefPubMed Balsalobre A, Brown SA, Marcacci L et al (2000) Resetting of circadian time in peripheral tissues by glucocorticoid signaling. Science 289:2344–2347CrossRefPubMed
24.
Zurück zum Zitat Sladek M, Jindrakova Z, Bendova Z, Sumova A (2007) Postnatal ontogenesis of the circadian clock within the rat liver. Am J Phys Regul Integr Comp Phys 292:R1224–R1229 Sladek M, Jindrakova Z, Bendova Z, Sumova A (2007) Postnatal ontogenesis of the circadian clock within the rat liver. Am J Phys Regul Integr Comp Phys 292:R1224–R1229
25.
Zurück zum Zitat Gotoh M, Maki T, Satomi S et al (1987) Reproducible high yield of rat islets by stationary in vitro digestion following pancreatic ductal or portal venous collagenase injection. Transplantation 43:725–730CrossRefPubMed Gotoh M, Maki T, Satomi S et al (1987) Reproducible high yield of rat islets by stationary in vitro digestion following pancreatic ductal or portal venous collagenase injection. Transplantation 43:725–730CrossRefPubMed
26.
Zurück zum Zitat Hohmeier HE, Mulder H, Chen G, Henkel-Rieger R, Prentki M, Newgard CB (2000) Isolation of INS-1-derived cell lines with robust ATP-sensitive K+ channel-dependent and -independent glucose-stimulated insulin secretion. Diabetes 49:424–430CrossRefPubMed Hohmeier HE, Mulder H, Chen G, Henkel-Rieger R, Prentki M, Newgard CB (2000) Isolation of INS-1-derived cell lines with robust ATP-sensitive K+ channel-dependent and -independent glucose-stimulated insulin secretion. Diabetes 49:424–430CrossRefPubMed
27.
Zurück zum Zitat Jacovetti C, Abderrahmani A, Parnaud G et al (2012) MicroRNAs contribute to compensatory beta cell expansion during pregnancy and obesity. J Clin Invest 122:3541–3551CrossRefPubMedPubMedCentral Jacovetti C, Abderrahmani A, Parnaud G et al (2012) MicroRNAs contribute to compensatory beta cell expansion during pregnancy and obesity. J Clin Invest 122:3541–3551CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Liu AC, Tran HG, Zhang EE, Priest AA, Welsh DK, Kay SA (2008) Redundant function of REV-ERBα and beta and non-essential role for Bmal1 cycling in transcriptional regulation of intracellular circadian rhythms. PLoS Genet 4:e1000023CrossRefPubMedPubMedCentral Liu AC, Tran HG, Zhang EE, Priest AA, Welsh DK, Kay SA (2008) Redundant function of REV-ERBα and beta and non-essential role for Bmal1 cycling in transcriptional regulation of intracellular circadian rhythms. PLoS Genet 4:e1000023CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Petrenko V, Saini C, Perrin L, Dibner C (2016) Parallel measurement of circadian clock gene expression and hormone secretion in human primary cell cultures. J Vis Exp. doi:10.3791/54673/ Petrenko V, Saini C, Perrin L, Dibner C (2016) Parallel measurement of circadian clock gene expression and hormone secretion in human primary cell cultures. J Vis Exp. doi:10.​3791/​54673/​
30.
Zurück zum Zitat Qian J, Block GD, Colwell CS, Matveyenko AV (2013) Consequences of exposure to light at night on the pancreatic islet circadian clock and function in rats. Diabetes 62:3469–3478CrossRefPubMedPubMedCentral Qian J, Block GD, Colwell CS, Matveyenko AV (2013) Consequences of exposure to light at night on the pancreatic islet circadian clock and function in rats. Diabetes 62:3469–3478CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Ru Y, Kechris KJ, Tabakoff B et al (2014) The multiMiR R package and database: integration of microRNA-target interactions along with their disease and drug associations. Nucleic Acids Res 42:e133CrossRefPubMedPubMedCentral Ru Y, Kechris KJ, Tabakoff B et al (2014) The multiMiR R package and database: integration of microRNA-target interactions along with their disease and drug associations. Nucleic Acids Res 42:e133CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Su G, Morris JH, Demchak B, Bader GD (2014) Biological network exploration with Cytoscape 3. Curr Protoc Bioinformatics 47:8.13.1–8.1324CrossRef Su G, Morris JH, Demchak B, Bader GD (2014) Biological network exploration with Cytoscape 3. Curr Protoc Bioinformatics 47:8.13.1–8.1324CrossRef
33.
Zurück zum Zitat Lee J, Kim MS, Li R et al (2011) Loss of Bmal1 leads to uncoupling and impaired glucose-stimulated insulin secretion in beta-cells. Islets 3:381–388CrossRefPubMedPubMedCentral Lee J, Kim MS, Li R et al (2011) Loss of Bmal1 leads to uncoupling and impaired glucose-stimulated insulin secretion in beta-cells. Islets 3:381–388CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Sadacca LA, Lamia KA, deLemos AS, Blum B, Weitz CJ (2011) An intrinsic circadian clock of the pancreas is required for normal insulin release and glucose homeostasis in mice. Diabetologia 54:120–124CrossRefPubMed Sadacca LA, Lamia KA, deLemos AS, Blum B, Weitz CJ (2011) An intrinsic circadian clock of the pancreas is required for normal insulin release and glucose homeostasis in mice. Diabetologia 54:120–124CrossRefPubMed
35.
Zurück zum Zitat Rakshit K, Hsu TW, Matveyenko AV (2016) Bmal1 is required for beta cell compensatory expansion, survival and metabolic adaptation to diet-induced obesity in mice. Diabetologia 59:734–743CrossRefPubMedPubMedCentral Rakshit K, Hsu TW, Matveyenko AV (2016) Bmal1 is required for beta cell compensatory expansion, survival and metabolic adaptation to diet-induced obesity in mice. Diabetologia 59:734–743CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Marcheva B, Ramsey KM, Buhr ED et al (2010) Disruption of the clock components CLOCK and BMAL1 leads to hypoinsulinaemia and diabetes. Nature 466:627–631CrossRefPubMedPubMedCentral Marcheva B, Ramsey KM, Buhr ED et al (2010) Disruption of the clock components CLOCK and BMAL1 leads to hypoinsulinaemia and diabetes. Nature 466:627–631CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Kowalska E, Moriggi E, Bauer C, Dibner C, Brown SA (2010) The circadian clock starts ticking at a developmentally early stage. J Biol Rhythm 25:442–449CrossRef Kowalska E, Moriggi E, Bauer C, Dibner C, Brown SA (2010) The circadian clock starts ticking at a developmentally early stage. J Biol Rhythm 25:442–449CrossRef
38.
Zurück zum Zitat Brooks E, Canal MM (2013) Development of circadian rhythms: role of postnatal light environment. Neurosci Biobehav Rev 37:551–560CrossRefPubMed Brooks E, Canal MM (2013) Development of circadian rhythms: role of postnatal light environment. Neurosci Biobehav Rev 37:551–560CrossRefPubMed
39.
Zurück zum Zitat Petrenko V, Saini C, Giovannoni L et al (2017) Pancreatic alpha- and beta-cellular clocks have distinct molecular properties and impact on islet hormone secretion and gene expression. Genes Dev 31:383–398CrossRefPubMedPubMedCentral Petrenko V, Saini C, Giovannoni L et al (2017) Pancreatic alpha- and beta-cellular clocks have distinct molecular properties and impact on islet hormone secretion and gene expression. Genes Dev 31:383–398CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Jermendy A, Toschi E, Aye T et al (2011) Rat neonatal beta cells lack the specialised metabolic phenotype of mature beta cells. Diabetologia 54:594–604CrossRefPubMedPubMedCentral Jermendy A, Toschi E, Aye T et al (2011) Rat neonatal beta cells lack the specialised metabolic phenotype of mature beta cells. Diabetologia 54:594–604CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Froy O (2007) The relationship between nutrition and circadian rhythms in mammals. Front Neuroendocrinol 28:61–71CrossRefPubMed Froy O (2007) The relationship between nutrition and circadian rhythms in mammals. Front Neuroendocrinol 28:61–71CrossRefPubMed
43.
Zurück zum Zitat Huang J, Lu C, Chen S, Hua L, Qian R (2010) Postnatal ontogenesis of clock genes in mouse suprachiasmatic nucleus and heart. Lipids Health Dis 9:22CrossRefPubMedPubMedCentral Huang J, Lu C, Chen S, Hua L, Qian R (2010) Postnatal ontogenesis of clock genes in mouse suprachiasmatic nucleus and heart. Lipids Health Dis 9:22CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Brown SA, Schibler U (1999) The ins and outs of circadian timekeeping. Curr Opin Genet Dev 9:588–594CrossRefPubMed Brown SA, Schibler U (1999) The ins and outs of circadian timekeeping. Curr Opin Genet Dev 9:588–594CrossRefPubMed
45.
Zurück zum Zitat Saito M, Suda M, Matzuda H (1978) Postnatal development of circadian rhythms in disaccharidase activities in rat small intestine. Am J Phys 234:E500–E503 Saito M, Suda M, Matzuda H (1978) Postnatal development of circadian rhythms in disaccharidase activities in rat small intestine. Am J Phys 234:E500–E503
46.
Zurück zum Zitat Stephan FK (2002) The “other” circadian system: food as a Zeitgeber. J Biol Rhythm 17:284–292CrossRef Stephan FK (2002) The “other” circadian system: food as a Zeitgeber. J Biol Rhythm 17:284–292CrossRef
47.
48.
Zurück zum Zitat Luby MD, Hsu CT, Shuster SA et al (2012) Food anticipatory activity behavior of mice across a wide range of circadian and non-circadian intervals. PLoS One 7:e37992CrossRefPubMedPubMedCentral Luby MD, Hsu CT, Shuster SA et al (2012) Food anticipatory activity behavior of mice across a wide range of circadian and non-circadian intervals. PLoS One 7:e37992CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Carneiro BT, Araujo JF (2009) The food-entrainable oscillator: a network of interconnected brain structures entrained by humoral signals? Chronobiol Int 26:1273–1289CrossRefPubMed Carneiro BT, Araujo JF (2009) The food-entrainable oscillator: a network of interconnected brain structures entrained by humoral signals? Chronobiol Int 26:1273–1289CrossRefPubMed
50.
Zurück zum Zitat Delezie J, Dumont S, Sandu C, Reibel S, Pevet P, Challet E (2016) Rev-erbα in the brain is essential for circadian food entrainment. Sci Rep 6:29386CrossRefPubMedPubMedCentral Delezie J, Dumont S, Sandu C, Reibel S, Pevet P, Challet E (2016) Rev-erbα in the brain is essential for circadian food entrainment. Sci Rep 6:29386CrossRefPubMedPubMedCentral
Metadaten
Titel
MicroRNAs modulate core-clock gene expression in pancreatic islets during early postnatal life in rats
verfasst von
Cécile Jacovetti
Adriana Rodriguez-Trejo
Claudiane Guay
Jonathan Sobel
Sonia Gattesco
Volodymyr Petrenko
Camille Saini
Charna Dibner
Romano Regazzi
Publikationsdatum
04.07.2017
Verlag
Springer Berlin Heidelberg
Erschienen in
Diabetologia / Ausgabe 10/2017
Print ISSN: 0012-186X
Elektronische ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-017-4348-6

Weitere Artikel der Ausgabe 10/2017

Diabetologia 10/2017 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.