Skip to main content
Top
Published in: Diabetologia 4/2016

01-04-2016 | Article

Bmal1 is required for beta cell compensatory expansion, survival and metabolic adaptation to diet-induced obesity in mice

Authors: Kuntol Rakshit, Tu Wen Hsu, Aleksey V. Matveyenko

Published in: Diabetologia | Issue 4/2016

Login to get access

Abstract

Aims/hypothesis

Obesity and consequent insulin resistance are known risk factors for type 2 diabetes. A compensatory increase in beta cell function and mass in response to insulin resistance permits maintenance of normal glucose homeostasis, whereas failure to do so results in beta cell failure and type 2 diabetes. Recent evidence suggests that the circadian system is essential for proper metabolic control and regulation of beta cell function. We set out to address the hypothesis that the beta cell circadian clock is essential for the appropriate functional and morphological beta cell response to insulin resistance.

Methods

We employed conditional deletion of the Bmal1 (also known as Arntl) gene (encoding a key circadian clock transcription factor) in beta cells using the tamoxifen-inducible CreERT recombination system. Upon adulthood, Bmal1 deletion in beta cells was achieved and mice were exposed to either chow or high fat diet (HFD). Changes in diurnal glycaemia, glucose tolerance and insulin secretion were longitudinally monitored in vivo and islet morphology and turnover assessed by immunofluorescence. Isolated islet experiments in vitro were performed to delineate changes in beta cell function and transcriptional regulation of cell proliferation.

Results

Adult Bmal1 deletion in beta cells resulted in failed metabolic adaptation to HFD characterised by fasting and diurnal hyperglycaemia, glucose intolerance and loss of glucose-stimulated insulin secretion. Importantly, HFD-induced beta cell expansion was absent following beta cell Bmal1 deletion indicating impaired beta cell proliferative and regenerative potential, which was confirmed by assessment of transcriptional profiles in isolated islets.

Conclusion/interpretation

Results of the study suggest that the beta cell circadian clock is a novel regulator of compensatory beta cell expansion and function in response to increased insulin demand associated with diet-induced obesity.
Appendix
Available only for authorised users
Literature
1.
go back to reference Yki-Jarvinen H (1995) Role of insulin resistance in the pathogenesis of NIDDM. Diabetologia 38:1378–1388CrossRefPubMed Yki-Jarvinen H (1995) Role of insulin resistance in the pathogenesis of NIDDM. Diabetologia 38:1378–1388CrossRefPubMed
2.
go back to reference Polonsky KS, Given BD, Van Cauter E (1988) Twenty-four-hour profiles and pulsatile patterns of insulin secretion in normal and obese subjects. J Clin Invest 81:442–448CrossRefPubMedPubMedCentral Polonsky KS, Given BD, Van Cauter E (1988) Twenty-four-hour profiles and pulsatile patterns of insulin secretion in normal and obese subjects. J Clin Invest 81:442–448CrossRefPubMedPubMedCentral
3.
go back to reference Polonsky KS, Given BD, Hirsch L et al (1988) Quantitative study of insulin secretion and clearance in normal and obese subjects. J Clin Invest 81:435–441CrossRefPubMedPubMedCentral Polonsky KS, Given BD, Hirsch L et al (1988) Quantitative study of insulin secretion and clearance in normal and obese subjects. J Clin Invest 81:435–441CrossRefPubMedPubMedCentral
4.
go back to reference Kahn SE (2003) The relative contributions of insulin resistance and beta-cell dysfunction to the pathophysiology of type 2 diabetes. Diabetologia 46:3–19CrossRefPubMed Kahn SE (2003) The relative contributions of insulin resistance and beta-cell dysfunction to the pathophysiology of type 2 diabetes. Diabetologia 46:3–19CrossRefPubMed
5.
go back to reference Bergman RN, Phillips LS, Cobelli C (1981) Physiologic evaluation of factors controlling glucose tolerance in man: measurement of insulin sensitivity and beta-cell glucose sensitivity from the response to intravenous glucose. J Clin Invest 68:1456–1467CrossRefPubMedPubMedCentral Bergman RN, Phillips LS, Cobelli C (1981) Physiologic evaluation of factors controlling glucose tolerance in man: measurement of insulin sensitivity and beta-cell glucose sensitivity from the response to intravenous glucose. J Clin Invest 68:1456–1467CrossRefPubMedPubMedCentral
6.
go back to reference Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, Butler PC (2003) Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes 52:102–110CrossRefPubMed Butler AE, Janson J, Bonner-Weir S, Ritzel R, Rizza RA, Butler PC (2003) Beta-cell deficit and increased beta-cell apoptosis in humans with type 2 diabetes. Diabetes 52:102–110CrossRefPubMed
7.
go back to reference Saisho Y, Butler AE, Manesso E, Elashoff D, Rizza RA, Butler PC (2013) beta-cell mass and turnover in humans: effects of obesity and aging. Diabetes Care 36:111–117CrossRefPubMedPubMedCentral Saisho Y, Butler AE, Manesso E, Elashoff D, Rizza RA, Butler PC (2013) beta-cell mass and turnover in humans: effects of obesity and aging. Diabetes Care 36:111–117CrossRefPubMedPubMedCentral
8.
go back to reference Kahn SE (2001) Clinical review 135: the importance of beta-cell failure in the development and progression of type 2 diabetes. J Clin Endocrinol Metab 86:4047–4058PubMed Kahn SE (2001) Clinical review 135: the importance of beta-cell failure in the development and progression of type 2 diabetes. J Clin Endocrinol Metab 86:4047–4058PubMed
10.
go back to reference Brown SA (2014) Circadian clock-mediated control of stem cell division and differentiation: beyond night and day. Development 141:3105–3111CrossRefPubMed Brown SA (2014) Circadian clock-mediated control of stem cell division and differentiation: beyond night and day. Development 141:3105–3111CrossRefPubMed
11.
go back to reference Rakshit K, Thomas AP, Matveyenko AV (2014) Does disruption of circadian rhythms contribute to beta-cell failure in type 2 diabetes? Curr Diab Rep 14:474CrossRefPubMedPubMedCentral Rakshit K, Thomas AP, Matveyenko AV (2014) Does disruption of circadian rhythms contribute to beta-cell failure in type 2 diabetes? Curr Diab Rep 14:474CrossRefPubMedPubMedCentral
12.
go back to reference Takahashi JS, Hong HK, Ko CH, McDearmon EL (2008) The genetics of mammalian circadian order and disorder: implications for physiology and disease. Nat Rev Genet 9:764–775CrossRefPubMedPubMedCentral Takahashi JS, Hong HK, Ko CH, McDearmon EL (2008) The genetics of mammalian circadian order and disorder: implications for physiology and disease. Nat Rev Genet 9:764–775CrossRefPubMedPubMedCentral
13.
go back to reference Gekakis N, Staknis D, Nguyen HB et al (1998) Role of the CLOCK protein in the mammalian circadian mechanism. Science 280:1564–1569CrossRefPubMed Gekakis N, Staknis D, Nguyen HB et al (1998) Role of the CLOCK protein in the mammalian circadian mechanism. Science 280:1564–1569CrossRefPubMed
14.
go back to reference Lee C, Etchegaray JP, Cagampang FR, Loudon AS, Reppert SM (2001) Posttranslational mechanisms regulate the mammalian circadian clock. Cell 107:855–867CrossRefPubMed Lee C, Etchegaray JP, Cagampang FR, Loudon AS, Reppert SM (2001) Posttranslational mechanisms regulate the mammalian circadian clock. Cell 107:855–867CrossRefPubMed
15.
16.
go back to reference Rudic RD, McNamara P, Curtis AM et al (2004) BMAL1 and CLOCK, two essential components of the circadian clock, are involved in glucose homeostasis. PLoS Biol 2, e377CrossRefPubMedPubMedCentral Rudic RD, McNamara P, Curtis AM et al (2004) BMAL1 and CLOCK, two essential components of the circadian clock, are involved in glucose homeostasis. PLoS Biol 2, e377CrossRefPubMedPubMedCentral
17.
go back to reference Silva CM, Sato S, Margolis RN No time to lose: workshop on circadian rhythms and metabolic disease. Genes Dev 24: 1456-1464 Silva CM, Sato S, Margolis RN No time to lose: workshop on circadian rhythms and metabolic disease. Genes Dev 24: 1456-1464
18.
go back to reference Lin YC, Hsiao TJ, Chen PC (2009) Persistent rotating shift-work exposure accelerates development of metabolic syndrome among middle-aged female employees: a five-year follow-up. Chronobiol Int 26:740–755CrossRefPubMed Lin YC, Hsiao TJ, Chen PC (2009) Persistent rotating shift-work exposure accelerates development of metabolic syndrome among middle-aged female employees: a five-year follow-up. Chronobiol Int 26:740–755CrossRefPubMed
19.
go back to reference Pan A, Schernhammer ES, Sun Q, Hu FB (2011) Rotating night shift work and risk of type 2 diabetes: two prospective cohort studies in women. PLoS Med 8, e1001141CrossRefPubMedPubMedCentral Pan A, Schernhammer ES, Sun Q, Hu FB (2011) Rotating night shift work and risk of type 2 diabetes: two prospective cohort studies in women. PLoS Med 8, e1001141CrossRefPubMedPubMedCentral
20.
go back to reference Suwazono Y, Dochi M, Oishi M, Tanaka K, Kobayashi E, Sakata K (2009) Shiftwork and impaired glucose metabolism: a 14-year cohort study on 7104 male workers. Chronobiol Int 26:926–941CrossRefPubMed Suwazono Y, Dochi M, Oishi M, Tanaka K, Kobayashi E, Sakata K (2009) Shiftwork and impaired glucose metabolism: a 14-year cohort study on 7104 male workers. Chronobiol Int 26:926–941CrossRefPubMed
21.
go back to reference Buxton OM, Cain SW, O'Connor SP et al (2012) Adverse metabolic consequences in humans of prolonged sleep restriction combined with circadian disruption. Sci Transl Med 4:129ra143CrossRef Buxton OM, Cain SW, O'Connor SP et al (2012) Adverse metabolic consequences in humans of prolonged sleep restriction combined with circadian disruption. Sci Transl Med 4:129ra143CrossRef
22.
go back to reference Lee J, Moulik M, Fang Z et al (2013) Bmal1 and beta-cell clock are required for adaptation to circadian disruption, and their loss of function leads to oxidative stress-induced beta-cell failure in mice. Mol Cell Biol 33:2327–2338CrossRefPubMedPubMedCentral Lee J, Moulik M, Fang Z et al (2013) Bmal1 and beta-cell clock are required for adaptation to circadian disruption, and their loss of function leads to oxidative stress-induced beta-cell failure in mice. Mol Cell Biol 33:2327–2338CrossRefPubMedPubMedCentral
23.
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 466:627–631CrossRefPubMedPubMedCentral 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
24.
go back to reference 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
25.
go back to reference Storch KF, Paz C, Signorovitch J et al (2007) Intrinsic circadian clock of the mammalian retina: importance for retinal processing of visual information. Cell 130:730–741CrossRefPubMedPubMedCentral Storch KF, Paz C, Signorovitch J et al (2007) Intrinsic circadian clock of the mammalian retina: importance for retinal processing of visual information. Cell 130:730–741CrossRefPubMedPubMedCentral
26.
go back to reference Dor Y, Brown J, Martinez OI, Melton DA (2004) Adult pancreatic beta-cells are formed by self-duplication rather than stem-cell differentiation. Nature 429:41–46CrossRefPubMed Dor Y, Brown J, Martinez OI, Melton DA (2004) Adult pancreatic beta-cells are formed by self-duplication rather than stem-cell differentiation. Nature 429:41–46CrossRefPubMed
27.
go back to reference Ernst J, Bar-Joseph Z (2006) STEM: a tool for the analysis of short time series gene expression data. BMC Bioinforma 7:191CrossRef Ernst J, Bar-Joseph Z (2006) STEM: a tool for the analysis of short time series gene expression data. BMC Bioinforma 7:191CrossRef
28.
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
30.
go back to reference Matveyenko AV, Gurlo T, Daval M, Butler AE, Butler PC (2009) Successful versus failed adaptation to high-fat diet-induced insulin resistance: the role of IAPP-induced beta-cell endoplasmic reticulum stress. Diabetes 58:906–916CrossRefPubMedPubMedCentral Matveyenko AV, Gurlo T, Daval M, Butler AE, Butler PC (2009) Successful versus failed adaptation to high-fat diet-induced insulin resistance: the role of IAPP-induced beta-cell endoplasmic reticulum stress. Diabetes 58:906–916CrossRefPubMedPubMedCentral
32.
go back to reference Meier JJ, Bhushan A, Butler PC (2006) The potential for stem cell therapy in diabetes. Pediatr Res 59:65R–73RCrossRefPubMed Meier JJ, Bhushan A, Butler PC (2006) The potential for stem cell therapy in diabetes. Pediatr Res 59:65R–73RCrossRefPubMed
33.
34.
go back to reference Grechez-Cassiau A, Rayet B, Guillaumond F, Teboul M, Delaunay F (2008) The circadian clock component BMAL1 is a critical regulator of p21WAF1/CIP1 expression and hepatocyte proliferation. J Biol Chem 283:4535–4542CrossRefPubMed Grechez-Cassiau A, Rayet B, Guillaumond F, Teboul M, Delaunay F (2008) The circadian clock component BMAL1 is a critical regulator of p21WAF1/CIP1 expression and hepatocyte proliferation. J Biol Chem 283:4535–4542CrossRefPubMed
35.
go back to reference Matsuo T, Yamaguchi S, Mitsui S, Emi A, Shimoda F, Okamura H (2003) Control mechanism of the circadian clock for timing of cell division in vivo. Science 302:255–259CrossRefPubMed Matsuo T, Yamaguchi S, Mitsui S, Emi A, Shimoda F, Okamura H (2003) Control mechanism of the circadian clock for timing of cell division in vivo. Science 302:255–259CrossRefPubMed
36.
go back to reference Chen Y, Xu J, Borowicz S, Collins C, Huo D, Olopade OI (2011) c-Myc activates BRCA1 gene expression through distal promoter elements in breast cancer cells. BMC Cancer 11:246CrossRefPubMedPubMedCentral Chen Y, Xu J, Borowicz S, Collins C, Huo D, Olopade OI (2011) c-Myc activates BRCA1 gene expression through distal promoter elements in breast cancer cells. BMC Cancer 11:246CrossRefPubMedPubMedCentral
37.
go back to reference Hunt T, Sassone-Corsi P (2007) Riding tandem: circadian clocks and the cell cycle. Cell 129:461–464CrossRefPubMed Hunt T, Sassone-Corsi P (2007) Riding tandem: circadian clocks and the cell cycle. Cell 129:461–464CrossRefPubMed
38.
go back to reference Mateyak MK, Obaya AJ, Sedivy JM (1999) c-Myc regulates cyclin D-Cdk4 and -Cdk6 activity but affects cell cycle progression at multiple independent points. Mol Cell Biol 19:4672–4683CrossRefPubMedPubMedCentral Mateyak MK, Obaya AJ, Sedivy JM (1999) c-Myc regulates cyclin D-Cdk4 and -Cdk6 activity but affects cell cycle progression at multiple independent points. Mol Cell Biol 19:4672–4683CrossRefPubMedPubMedCentral
39.
go back to reference Abraham RT (2001) Cell cycle checkpoint signaling through the ATM and ATR kinases. Genes Dev 15:2177–2196CrossRefPubMed Abraham RT (2001) Cell cycle checkpoint signaling through the ATM and ATR kinases. Genes Dev 15:2177–2196CrossRefPubMed
40.
go back to reference Sancar A, Lindsey-Boltz LA, Kang TH, Reardon JT, Lee JH, Ozturk N (2010) Circadian clock control of the cellular response to DNA damage. FEBS Lett 584:2618–2625CrossRefPubMedPubMedCentral Sancar A, Lindsey-Boltz LA, Kang TH, Reardon JT, Lee JH, Ozturk N (2010) Circadian clock control of the cellular response to DNA damage. FEBS Lett 584:2618–2625CrossRefPubMedPubMedCentral
41.
go back to reference 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 20:979–990CrossRefPubMed 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 20:979–990CrossRefPubMed
42.
go back to reference Montgomery MK, Hallahan NL, Brown SH et al (2013) Mouse strain-dependent variation in obesity and glucose homeostasis in response to high-fat feeding. Diabetologia 56:1129–1139CrossRefPubMed Montgomery MK, Hallahan NL, Brown SH et al (2013) Mouse strain-dependent variation in obesity and glucose homeostasis in response to high-fat feeding. Diabetologia 56:1129–1139CrossRefPubMed
43.
go back to reference Kroenke CH, Spiegelman D, Manson J, Schernhammer ES, Colditz GA, Kawachi I (2007) Work characteristics and incidence of type 2 diabetes in women. Am J Epidemiol 165:175–183CrossRefPubMed Kroenke CH, Spiegelman D, Manson J, Schernhammer ES, Colditz GA, Kawachi I (2007) Work characteristics and incidence of type 2 diabetes in women. Am J Epidemiol 165:175–183CrossRefPubMed
44.
go back to reference Mikuni E, Ohoshi T, Hayashi K, Miyamura K (1983) Glucose intolerance in an employed population. Tohoku J Exp Med 141(Suppl):S251–S256CrossRef Mikuni E, Ohoshi T, Hayashi K, Miyamura K (1983) Glucose intolerance in an employed population. Tohoku J Exp Med 141(Suppl):S251–S256CrossRef
Metadata
Title
Bmal1 is required for beta cell compensatory expansion, survival and metabolic adaptation to diet-induced obesity in mice
Authors
Kuntol Rakshit
Tu Wen Hsu
Aleksey V. Matveyenko
Publication date
01-04-2016
Publisher
Springer Berlin Heidelberg
Published in
Diabetologia / Issue 4/2016
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-015-3859-2

Other articles of this Issue 4/2016

Diabetologia 4/2016 Go to the issue
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

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discuss last year's major advances in heart failure and cardiomyopathies.