Skip to main content
Top
Published in: Diabetologia 3/2013

Open Access 01-03-2013 | Article

Autonomous and self-sustained circadian oscillators displayed in human islet cells

Authors: P. Pulimeno, T. Mannic, D. Sage, L. Giovannoni, P. Salmon, S. Lemeille, M. Giry-Laterriere, M. Unser, D. Bosco, C. Bauer, J. Morf, P. Halban, J. Philippe, C. Dibner

Published in: Diabetologia | Issue 3/2013

Login to get access

Abstract

Aims/hypothesis

Following on from the emerging importance of the pancreas circadian clock on islet function and the development of type 2 diabetes in rodent models, we aimed to examine circadian gene expression in human islets. The oscillator properties were assessed in intact islets as well as in beta cells.

Methods

We established a system for long-term bioluminescence recording in cultured human islets, employing lentivector gene delivery of the core clock gene Bmal1 (also known as Arntl)-luciferase reporter. Beta cells were stably labelled using a rat insulin2 promoter fluorescent construct. Single-islet/cell oscillation profiles were measured by combined bioluminescence–fluorescence time-lapse microscopy.

Results

Human islets synchronised in vitro exhibited self-sustained circadian oscillations of Bmal1-luciferase expression at both the population and single-islet levels, with period lengths of 23.6 and 23.9 h, respectively. Endogenous BMAL1 and CRY1 transcript expression was circadian in synchronised islets over 48 h, and antiphasic to REV-ERBα (also known as NR1D1), PER1, PER2, PER3 and DBP transcript circadian profiles. HNF1A and PDX1 exhibited weak circadian oscillations, in phase with the REV-ERBα transcript. Dispersed islet cells were strongly oscillating as well, at population and single-cell levels. Importantly, beta and non-beta cells revealed oscillatory profiles that were well synchronised with each other.

Conclusions/interpretation

We provide for the first time compelling evidence for high-amplitude cell-autonomous circadian oscillators displayed in human pancreatic islets and in dispersed human islet cells. Moreover, these clocks are synchronised between beta and non-beta cells in primary human islet cell cultures.
Appendix
Available only for authorised users
Literature
2.
go back to reference Dibner C, Schibler U, Albrecht U (2010) The mammalian circadian timing system: organization and coordination of central and peripheral clocks. Annu Rev Physiol 72:517–549PubMedCrossRef Dibner C, Schibler U, Albrecht U (2010) The mammalian circadian timing system: organization and coordination of central and peripheral clocks. Annu Rev Physiol 72:517–549PubMedCrossRef
3.
go back to reference Nagoshi E, Saini C, Bauer C, Laroche T, Naef F, Schibler U (2004) Circadian gene expression in individual fibroblasts: cell-autonomous and self-sustained oscillators pass time to daughter cells. Cell 119:693–705PubMedCrossRef Nagoshi E, Saini C, Bauer C, Laroche T, Naef F, Schibler U (2004) Circadian gene expression in individual fibroblasts: cell-autonomous and self-sustained oscillators pass time to daughter cells. Cell 119:693–705PubMedCrossRef
4.
go back to reference Balsalobre A, Damiola F, Schibler U (1998) A serum shock induces circadian gene expression in mammalian tissue culture cells. Cell 93:929–937PubMedCrossRef Balsalobre A, Damiola F, Schibler U (1998) A serum shock induces circadian gene expression in mammalian tissue culture cells. Cell 93:929–937PubMedCrossRef
5.
go back to reference Kohsaka A, Laposky AD, Ramsey KM et al (2007) High-fat diet disrupts behavioral and molecular circadian rhythms in mice. Cell Metabol 6:414–421CrossRef Kohsaka A, Laposky AD, Ramsey KM et al (2007) High-fat diet disrupts behavioral and molecular circadian rhythms in mice. Cell Metabol 6:414–421CrossRef
7.
go back to reference Lamia KA, Storch KF, Weitz CJ (2008) Physiological significance of a peripheral tissue circadian clock. Proc Natl Acad Sci USA 105:15172–15177PubMedCrossRef Lamia KA, Storch KF, Weitz CJ (2008) Physiological significance of a peripheral tissue circadian clock. Proc Natl Acad Sci USA 105:15172–15177PubMedCrossRef
8.
go back to reference Turek FW, Joshu C, Kohsaka A et al (2005) Obesity and metabolic syndrome in circadian clock mutant mice. Science 308:1043–1045PubMedCrossRef Turek FW, Joshu C, Kohsaka A et al (2005) Obesity and metabolic syndrome in circadian clock mutant mice. Science 308:1043–1045PubMedCrossRef
9.
go back to reference Cho H, Zhao X, Hatori M et al (2012) Regulation of circadian behaviour and metabolism by REV-ERB-α and REV-ERB-β. Nature 485:123–127PubMedCrossRef Cho H, Zhao X, Hatori M et al (2012) Regulation of circadian behaviour and metabolism by REV-ERB-α and REV-ERB-β. Nature 485:123–127PubMedCrossRef
10.
go back to reference Delezie J, Dumont S, Dardente H et al (2012) The nuclear receptor REV-ERBα is required for the daily balance of carbohydrate and lipid metabolism. FASEB J 26:3321–3335PubMedCrossRef Delezie J, Dumont S, Dardente H et al (2012) The nuclear receptor REV-ERBα is required for the daily balance of carbohydrate and lipid metabolism. FASEB J 26:3321–3335PubMedCrossRef
11.
go back to reference Le Martelot G, Claudel T, Gatfield D et al (2009) REV-ERBalpha participates in circadian SREBP signaling and bile acid homeostasis. PLoS Biology 7:e1000181PubMedCrossRef Le Martelot G, Claudel T, Gatfield D et al (2009) REV-ERBalpha participates in circadian SREBP signaling and bile acid homeostasis. PLoS Biology 7:e1000181PubMedCrossRef
12.
go back to reference Zhang EE, Liu Y, Dentin R et al (2010) Cryptochrome mediates circadian regulation of cAMP signaling and hepatic gluconeogenesis. Nat Med 16:1152–1156PubMedCrossRef Zhang EE, Liu Y, Dentin R et al (2010) Cryptochrome mediates circadian regulation of cAMP signaling and hepatic gluconeogenesis. Nat Med 16:1152–1156PubMedCrossRef
13.
go back to reference Marcheva B, Ramsey KM, Buhr ED et al (2010) Disruption of the clock components CLOCK and BMAL1 leads to hypoinsulinaemia and diabetes. Nature 466:627–631PubMedCrossRef Marcheva B, Ramsey KM, Buhr ED et al (2010) Disruption of the clock components CLOCK and BMAL1 leads to hypoinsulinaemia and diabetes. Nature 466:627–631PubMedCrossRef
14.
go back to reference 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 54:120–124PubMedCrossRef 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 54:120–124PubMedCrossRef
15.
go back to reference Eberhard D, Lammert E (2009) The pancreatic beta-cell in the islet and organ community. Curr Opin Genet Dev 19:469–475PubMedCrossRef Eberhard D, Lammert E (2009) The pancreatic beta-cell in the islet and organ community. Curr Opin Genet Dev 19:469–475PubMedCrossRef
16.
go back to reference Englund A, Kovanen L, Saarikoski ST et al (2009) NPAS2 and PER2 are linked to risk factors of the metabolic syndrome. J Circadian Rhythms 7:5PubMedCrossRef Englund A, Kovanen L, Saarikoski ST et al (2009) NPAS2 and PER2 are linked to risk factors of the metabolic syndrome. J Circadian Rhythms 7:5PubMedCrossRef
17.
go back to reference Dupuis J, Langenberg C, Prokopenko I et al (2010) New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk. Nat Genet 42:105–116PubMedCrossRef Dupuis J, Langenberg C, Prokopenko I et al (2010) New genetic loci implicated in fasting glucose homeostasis and their impact on type 2 diabetes risk. Nat Genet 42:105–116PubMedCrossRef
18.
go back to reference Stamenkovic JA, Olsson AH, Nagorny CL et al (2012) Regulation of core clock genes in human islets. Metabolism 61:978–985PubMedCrossRef Stamenkovic JA, Olsson AH, Nagorny CL et al (2012) Regulation of core clock genes in human islets. Metabolism 61:978–985PubMedCrossRef
19.
go back to reference Bucher P, Mathe Z, Morel P et al (2005) Assessment of a novel two-component enzyme preparation for human islet isolation and transplantation. Transplantation 79:91–97PubMedCrossRef Bucher P, Mathe Z, Morel P et al (2005) Assessment of a novel two-component enzyme preparation for human islet isolation and transplantation. Transplantation 79:91–97PubMedCrossRef
20.
go back to reference Ricordi C, Lacy PE, Finke EH, Olack BJ, Scharp DW (1988) Automated method for isolation of human pancreatic islets. Diabetes 37:413–420PubMedCrossRef Ricordi C, Lacy PE, Finke EH, Olack BJ, Scharp DW (1988) Automated method for isolation of human pancreatic islets. Diabetes 37:413–420PubMedCrossRef
21.
go back to reference Parnaud G, Bosco D, Berney T et al (2008) Proliferation of sorted human and rat beta cells. Diabetologia 51:91–100PubMedCrossRef Parnaud G, Bosco D, Berney T et al (2008) Proliferation of sorted human and rat beta cells. Diabetologia 51:91–100PubMedCrossRef
22.
go back to reference Liu AC, Tran HG, Zhang EE, Priest AA, Welsh DK, Kay SA (2008) Redundant function of REV-ERBalpha and beta and non-essential role for Bmal1 cycling in transcriptional regulation of intracellular circadian rhythms. PLoS Genetics 4:e1000023PubMedCrossRef Liu AC, Tran HG, Zhang EE, Priest AA, Welsh DK, Kay SA (2008) Redundant function of REV-ERBalpha and beta and non-essential role for Bmal1 cycling in transcriptional regulation of intracellular circadian rhythms. PLoS Genetics 4:e1000023PubMedCrossRef
23.
go back to reference Salmon P, Oberholzer J, Occhiodoro T, Morel P, Lou J, Trono D (2000) Reversible immortalization of human primary cells by lentivector-mediated transfer of specific genes. Mol Ther 2:404–414PubMedCrossRef Salmon P, Oberholzer J, Occhiodoro T, Morel P, Lou J, Trono D (2000) Reversible immortalization of human primary cells by lentivector-mediated transfer of specific genes. Mol Ther 2:404–414PubMedCrossRef
24.
go back to reference Shaner NC, Campbell RE, Steinbach PA, Giepmans BN, Palmer AE, Tsien RY (2004) Improved monomeric red, orange and yellow fluorescent proteins derived from Discosoma sp. red fluorescent protein. Nat Biotechnol 22:1567–1572PubMedCrossRef Shaner NC, Campbell RE, Steinbach PA, Giepmans BN, Palmer AE, Tsien RY (2004) Improved monomeric red, orange and yellow fluorescent proteins derived from Discosoma sp. red fluorescent protein. Nat Biotechnol 22:1567–1572PubMedCrossRef
25.
go back to reference Toledo JR, Prieto Y, Oramas N, Sanchez O (2009) Polyethylenimine-based transfection method as a simple and effective way to produce recombinant lentiviral vectors. Appl Biochem Biotechnol 157:538–544PubMedCrossRef Toledo JR, Prieto Y, Oramas N, Sanchez O (2009) Polyethylenimine-based transfection method as a simple and effective way to produce recombinant lentiviral vectors. Appl Biochem Biotechnol 157:538–544PubMedCrossRef
26.
go back to reference Dibner C, Sage D, Unser M et al (2009) Circadian gene expression is resilient to large fluctuations in overall transcription rates. EMBO J 28:123–134PubMedCrossRef Dibner C, Sage D, Unser M et al (2009) Circadian gene expression is resilient to large fluctuations in overall transcription rates. EMBO J 28:123–134PubMedCrossRef
27.
go back to reference Saini C, Morf J, Stratmann M, Gos P, Schibler U (2012) Simulated body temperature rhythms reveal the phase-shifting behavior and plasticity of mammalian circadian oscillators. Genes Dev 26:567–580PubMedCrossRef Saini C, Morf J, Stratmann M, Gos P, Schibler U (2012) Simulated body temperature rhythms reveal the phase-shifting behavior and plasticity of mammalian circadian oscillators. Genes Dev 26:567–580PubMedCrossRef
28.
go back to reference Sage D, Unser M, Salmon P, Dibner C (2010) A software solution for recording circadian oscillator features in time-lapse live cell microscopy. Cell Div 5:17PubMedCrossRef Sage D, Unser M, Salmon P, Dibner C (2010) A software solution for recording circadian oscillator features in time-lapse live cell microscopy. Cell Div 5:17PubMedCrossRef
29.
go back to reference Luisier F, Blu T, Unser M (2011) Image denoising in mixed Poisson–Gaussian noise. IEEE Trans Image Process 20:696–708PubMedCrossRef Luisier F, Blu T, Unser M (2011) Image denoising in mixed Poisson–Gaussian noise. IEEE Trans Image Process 20:696–708PubMedCrossRef
30.
go back to reference Brown SA, Fleury-Olela F, Nagoshi E et al (2005) The period length of fibroblast circadian gene expression varies widely among human individuals. PLoS Biology 3:e338PubMedCrossRef Brown SA, Fleury-Olela F, Nagoshi E et al (2005) The period length of fibroblast circadian gene expression varies widely among human individuals. PLoS Biology 3:e338PubMedCrossRef
31.
go back to reference Balsalobre A, Brown SA, Marcacci L et al (2000) Resetting of circadian time in peripheral tissues by glucocorticoid signaling. Science 289:2344–2347PubMedCrossRef Balsalobre A, Brown SA, Marcacci L et al (2000) Resetting of circadian time in peripheral tissues by glucocorticoid signaling. Science 289:2344–2347PubMedCrossRef
32.
go back to reference Balsalobre A, Marcacci L, Schibler U (2000) Multiple signaling pathways elicit circadian gene expression in cultured Rat-1 fibroblasts. Curr Biol 10:1291–1294PubMedCrossRef Balsalobre A, Marcacci L, Schibler U (2000) Multiple signaling pathways elicit circadian gene expression in cultured Rat-1 fibroblasts. Curr Biol 10:1291–1294PubMedCrossRef
33.
go back to reference Yoo SH, Yamazaki S, Lowrey PL et al (2004) PERIOD2::LUCIFERASE real-time reporting of circadian dynamics reveals persistent circadian oscillations in mouse peripheral tissues. Proc Natl Acad Sci U S A 101:5339–5346PubMedCrossRef Yoo SH, Yamazaki S, Lowrey PL et al (2004) PERIOD2::LUCIFERASE real-time reporting of circadian dynamics reveals persistent circadian oscillations in mouse peripheral tissues. Proc Natl Acad Sci U S A 101:5339–5346PubMedCrossRef
34.
go back to reference Asher G, Gatfield D, Stratmann M et al (2008) SIRT1 regulates circadian clock gene expression through PER2 deacetylation. Cell 134:317–328PubMedCrossRef Asher G, Gatfield D, Stratmann M et al (2008) SIRT1 regulates circadian clock gene expression through PER2 deacetylation. Cell 134:317–328PubMedCrossRef
35.
go back to reference Miyamoto Y, Sancar A (1999) Circadian regulation of cryptochrome genes in the mouse. Brain Res Mol Brain Res 71:238–243PubMedCrossRef Miyamoto Y, Sancar A (1999) Circadian regulation of cryptochrome genes in the mouse. Brain Res Mol Brain Res 71:238–243PubMedCrossRef
36.
go back to reference Sun ZS, Albrecht U, Zhuchenko O, Bailey J, Eichele G, Lee CC (1997) RIGUI, a putative mammalian ortholog of the Drosophila period gene. Cell 90:1003–1011PubMedCrossRef Sun ZS, Albrecht U, Zhuchenko O, Bailey J, Eichele G, Lee CC (1997) RIGUI, a putative mammalian ortholog of the Drosophila period gene. Cell 90:1003–1011PubMedCrossRef
37.
go back to reference Liu AC, Welsh DK, Ko CH et al (2007) Intercellular coupling confers robustness against mutations in the SCN circadian clock network. Cell 129:605–616PubMedCrossRef Liu AC, Welsh DK, Ko CH et al (2007) Intercellular coupling confers robustness against mutations in the SCN circadian clock network. Cell 129:605–616PubMedCrossRef
38.
go back to reference Allaman-Pillet N, Roduit R, Oberson A et al (2004) Circadian regulation of islet genes involved in insulin production and secretion. Mol Cell Endocrinol 226:59–66PubMedCrossRef Allaman-Pillet N, Roduit R, Oberson A et al (2004) Circadian regulation of islet genes involved in insulin production and secretion. Mol Cell Endocrinol 226:59–66PubMedCrossRef
39.
go back to reference Geiss GK, Bumgarner RE, Birditt B et al (2008) Direct multiplexed measurement of gene expression with color-coded probe pairs. Nat Biotechnol 26:317–325PubMedCrossRef Geiss GK, Bumgarner RE, Birditt B et al (2008) Direct multiplexed measurement of gene expression with color-coded probe pairs. Nat Biotechnol 26:317–325PubMedCrossRef
40.
go back to reference Gale JE, Cox HI, Qian J, Block GD, Colwell CS, Matveyenko AV (2011) Disruption of circadian rhythms accelerates development of diabetes through pancreatic beta-cell loss and dysfunction. J Biol Rhythm 26:423–433CrossRef Gale JE, Cox HI, Qian J, Block GD, Colwell CS, Matveyenko AV (2011) Disruption of circadian rhythms accelerates development of diabetes through pancreatic beta-cell loss and dysfunction. J Biol Rhythm 26:423–433CrossRef
Metadata
Title
Autonomous and self-sustained circadian oscillators displayed in human islet cells
Authors
P. Pulimeno
T. Mannic
D. Sage
L. Giovannoni
P. Salmon
S. Lemeille
M. Giry-Laterriere
M. Unser
D. Bosco
C. Bauer
J. Morf
P. Halban
J. Philippe
C. Dibner
Publication date
01-03-2013
Publisher
Springer-Verlag
Published in
Diabetologia / Issue 3/2013
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-012-2779-7

Other articles of this Issue 3/2013

Diabetologia 3/2013 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine