Endocrinol Metab.  2014 Sep;29(3):379-387. 10.3803/EnM.2014.29.3.379.

Effect of Resveratrol, a SIRT1 Activator, on the Interactions of the CLOCK/BMAL1 Complex

Affiliations
  • 1Department of Biological Sciences and Brain Research Center for 21st Frontier Program in Neuroscience, Seoul National University College of Natural Sciences, Seoul, Korea. kyungjin@snu.ac.kr
  • 2Department of Brain and Cognitive Sciences, Seoul National University College of Natural Sciences, Seoul, Korea.

Abstract

BACKGROUND
In mammals, the CLOCK/BMAL1 heterodimer is a key transcription factor complex that drives the cyclic expression of clock-controlled genes involved in various physiological functions and behavioral consequences. Recently, a growing number of studies have reported a molecular link between the circadian clock and metabolism. In the present study, we explored the regulatory effects of SIRTUIN1 (SIRT1), an NAD+-dependent deacetylase, on CLOCK/BMAL1-mediated clock gene expression.
METHODS
To investigate the interaction between SIRT1 and CLOCK/BMAL1, we conducted bimolecular fluorescence complementation (BiFC) analyses supplemented with immunocytochemistry assays. BiFC experiments employing deletion-specific mutants of BMAL1 were used to elucidate the specific domains that are necessary for the SIRT1-BMAL1 interaction. Additionally, luciferase reporter assays were used to delineate the effects of SIRT1 on circadian gene expression.
RESULTS
BiFC analysis revealed that SIRT1 interacted with both CLOCK and BMAL1 in most cell nuclei. As revealed by BiFC assays using various BMAL1 deletion mutants, the PAS-B domain of BMAL1 was essential for interaction with SIRT1. Activation of SIRT1 with resveratrol did not exert any significant change on the interaction with the CLOCK/BMAL1 complex. However, promoter analysis using Per1-Luc and Ebox-Luc reporters showed that SIRT1 significantly downregulated both promoter activities. This inhibitory effect was intensified by treatment with resveratrol, indicating a role for SIRT1 and its activator in CLOCK/BMAL1-mediated transcription of clock genes.
CONCLUSION
These results suggest that SIRT1 may form a regulatory complex with CLOCK/BMAL1 that represses clock gene expression, probably via deacetylase activity.

Keyword

Circadian clocks; CLOCK/BMAL1 heterodimer; SIRT1; BiFC analysis; Resveratrol

MeSH Terms

Cell Nucleus
Circadian Clocks
Complement System Proteins
Fluorescence
Gene Expression
Immunohistochemistry
Luciferases
Mammals
Metabolism
Transcription Factors
Complement System Proteins
Luciferases
Transcription Factors

Figure

  • Fig. 1 SIRTUIN1 interacts with CLOCK and BMAL1 in cell nuclei. (A) Schematic diagram of Venus-based bimolecular fluorescence complementation (BiFC) constructs. (B) Expression of SIRT1-N-terminal of Venus (VN) was detected by sodium dodecyl sulfate polyacrylamide gel electrophoresis separation followed by immunoblotting with an anti-green fluorescent protein (GFP) antibody (arrows). (C) Results from BiFC analysis and immunocytochemical experiments (ICC). COS7 cells were coexpressed with SIRT1-VN and either CLOCK-C-terminal of Venus (VC) or BMAL1-VC. Cells were immunostained with anti-GFP, anti-CLOCK, and anti-BMAL1 antibodies (red) to detect SIRT1-VN, CLOCK-VC, and BMAL1-VC, respectively. Nuclei were visualized with 4',6-diamidino-2-phenylindole (DAPI; blue). The images were acquired by fluorescence microscopy using specific filter sets for yellow fluorescent protein and red fluorescent protein. Scale bar=10 µm.

  • Fig. 2 Identification of the SIRTUIN1 (SIRT1)-binding domain of BMAL1. (A) Schematic diagram of mouse BMAL1. (B) Binding of SIRT1 to BMAL1 deletion mutants was analyzed by bimolecular fluorescence complementation (BiFC). COS7 cells were transfected with plasmids encoding SIRT1-N-terminal of Venus (VN) and BMAL1-C-terminal of Venus (VC) wildtype (WT) or its various mutants (Δnuclear localization sequence [NLS], BMAL1 without a functional nuclear localization signal; Δbasic-helix-loop-helix [bHLH], encoding BMAL1 Δ71 to 140; ΔPer-Arnt-Sim [PAS]-A, BMAL1 Δ210 to 320; ΔPAS-B, BMAL1 Δ350 to 480; Δtranscriptional activation domain [TAD], BMAL1 Δ553 to 625). The images were captured by fluorescence imaging microscopy using specific filter sets for yellow fluorescent protein and red fluorescent protein. Scale bar=10 µm. ICC, immunocytochemical; DAPI, 4',6-diamidino-2-phenylindole.

  • Fig. 3 Interaction of SIRTUIN1 (SIRT1) with CLOCK and BMAL1 upon resveratrol treatment. (A) COS7 cells were transfected with expression plasmids encoding SIRT1-N-terminal of Venus (VN) and either CLOCK-C-terminal of Venus (VC) or BMAL1-VC. Twelve hours after transfection, cells were treated with 100 µM resveratrol for 6 hours and then imaged. Scale bar=20 µm. (B) Bimolecular fluorescence complementation (BiFC) signals from control and resveratrol-stimulated groups were compared with signals from 4',6-diamidino-2-phenylindole (DAPI)-stained cells. Values are expressed as mean±SEM % (n=10 fields for each group). Veh, vehicle; RSV, resveratrol; YFP, yellow fluorescent protein.

  • Fig. 4 The effects of resveratrol on Ebox-Luc and Per1-Luc transcriptional activities. NIH3T3 cells expressing wild-type CLOCK, BMAL1, and either (A) Ebox-Luc or (B) Per1-Luc promoters were treated with increasing amounts of resveratrol (10 and 100 µM) to determine transcriptional activity. Luciferase activities in cell extracts were analyzed and normalized by cotransfected pRL-TK activity in each sample. To examine the effects of SIRTUIN1 (SIRT1) and resveratrol on the transcriptional activity of (C) Ebox-Luc and (D) Per1-Luc, SIRT1 was expressed with or without CLOCK and BMAL1 in NIH3T3 cells and incubated in resveratrol (10 and 100 µM). Values are expressed as mean±SEM in arbitrary units (AU), where the mean activity of the empty vector was set at 1 (n=3). aP<0.01 vs. vehicle-treated group.


Reference

1. Son GH, Chung S, Kim K. The adrenal peripheral clock: glucocorticoid and the circadian timing system. Front Neuroendocrinol. 2011; 32:451–465.
2. Dibner C, Schibler U, Albrecht U. The mammalian circadian timing system: organization and coordination of central and peripheral clocks. Annu Rev Physiol. 2010; 72:517–549.
3. Takahashi JS, Hong HK, Ko CH, McDearmon EL. The genetics of mammalian circadian order and disorder: implications for physiology and disease. Nat Rev Genet. 2008; 9:764–775.
4. Preitner N, Damiola F, Lopez-Molina L, Zakany J, Duboule D, Albrecht U, Schibler U. The orphan nuclear receptor REV-ERBalpha controls circadian transcription within the positive limb of the mammalian circadian oscillator. Cell. 2002; 110:251–260.
5. Sato TK, Panda S, Miraglia LJ, Reyes TM, Rudic RD, McNamara P, Naik KA, FitzGerald GA, Kay SA, Hogenesch JB. A functional genomics strategy reveals Rora as a component of the mammalian circadian clock. Neuron. 2004; 43:527–537.
6. Kaasik K, Lee CC. Reciprocal regulation of haem biosynthesis and the circadian clock in mammals. Nature. 2004; 430:467–471.
7. Rutter J, Reick M, McKnight SL. Metabolism and the control of circadian rhythms. Annu Rev Biochem. 2002; 71:307–331.
8. Tu BP, McKnight SL. Metabolic cycles as an underlying basis of biological oscillations. Nat Rev Mol Cell Biol. 2006; 7:696–701.
9. Nakahata Y, Kaluzova M, Grimaldi B, Sahar S, Hirayama J, Chen D, Guarente LP, Sassone-Corsi P. The NAD+-dependent deacetylase SIRT1 modulates CLOCK-mediated chromatin remodeling and circadian control. Cell. 2008; 134:329–340.
10. Asher G, Gatfield D, Stratmann M, Reinke H, Dibner C, Kreppel F, Mostoslavsky R, Alt FW, Schibler U. SIRT1 regulates circadian clock gene expression through PER2 deacetylation. Cell. 2008; 134:317–328.
11. Nakahata Y, Sahar S, Astarita G, Kaluzova M, Sassone-Corsi P. Circadian control of the NAD+ salvage pathway by CLOCK-SIRT1. Science. 2009; 324:654–657.
12. Ramsey KM, Yoshino J, Brace CS, Abrassart D, Kobayashi Y, Marcheva B, Hong HK, Chong JL, Buhr ED, Lee C, Takahashi JS, Imai S, Bass J. Circadian clock feedback cycle through NAMPT-mediated NAD+ biosynthesis. Science. 2009; 324:651–654.
13. Blander G, Guarente L. The Sir2 family of protein deacetylases. Annu Rev Biochem. 2004; 73:417–435.
14. Dali-Youcef N, Lagouge M, Froelich S, Koehl C, Schoonjans K, Auwerx J. Sirtuins: the 'magnificent seven', function, metabolism and longevity. Ann Med. 2007; 39:335–345.
15. Bordone L, Motta MC, Picard F, Robinson A, Jhala US, Apfeld J, McDonagh T, Lemieux M, McBurney M, Szilvasi A, Easlon EJ, Lin SJ, Guarente L. Sirt1 regulates insulin secretion by repressing UCP2 in pancreatic beta cells. PLoS Biol. 2006; 4:e31.
16. Brunet A, Sweeney LB, Sturgill JF, Chua KF, Greer PL, Lin Y, Tran H, Ross SE, Mostoslavsky R, Cohen HY, Hu LS, Cheng HL, Jedrychowski MP, Gygi SP, Sinclair DA, Alt FW, Greenberg ME. Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science. 2004; 303:2011–2015.
17. Motta MC, Divecha N, Lemieux M, Kamel C, Chen D, Gu W, Bultsma Y, McBurney M, Guarente L. Mammalian SIRT1 represses forkhead transcription factors. Cell. 2004; 116:551–563.
18. Howitz KT, Bitterman KJ, Cohen HY, Lamming DW, Lavu S, Wood JG, Zipkin RE, Chung P, Kisielewski A, Zhang LL, Scherer B, Sinclair DA. Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature. 2003; 425:191–196.
19. Lagouge M, Argmann C, Gerhart-Hines Z, Meziane H, Lerin C, Daussin F, Messadeq N, Milne J, Lambert P, Elliott P, Geny B, Laakso M, Puigserver P, Auwerx J. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1alpha. Cell. 2006; 127:1109–1122.
20. Baur JA, Pearson KJ, Price NL, Jamieson HA, Lerin C, Kalra A, Prabhu VV, Allard JS, Lopez-Lluch G, Lewis K, Pistell PJ, Poosala S, Becker KG, Boss O, Gwinn D, Wang M, Ramaswamy S, Fishbein KW, Spencer RG, Lakatta EG, Le Couteur D, Shaw RJ, Navas P, Puigserver P, Ingram DK, de Cabo R, Sinclair DA. Resveratrol improves health and survival of mice on a high-calorie diet. Nature. 2006; 444:337–342.
21. Vetterli L, Maechler P. Resveratrol-activated SIRT1 in liver and pancreatic beta-cells: a Janus head looking to the same direction of metabolic homeostasis. Aging (Albany NY). 2011; 3:444–449.
22. Lee Y, Lee J, Kwon I, Nakajima Y, Ohmiya Y, Son GH, Lee KH, Kim K. Coactivation of the CLOCK-BMAL1 complex by CBP mediates resetting of the circadian clock. J Cell Sci. 2010; 123(Pt 20):3547–3557.
23. Kwon I, Lee J, Chang SH, Jung NC, Lee BJ, Son GH, Kim K, Lee KH. BMAL1 shuttling controls transactivation and degradation of the CLOCK/BMAL1 heterodimer. Mol Cell Biol. 2006; 26:7318–7330.
24. Son GH, Chung S, Choe HK, Kim HD, Baik SM, Lee H, Lee HW, Choi S, Sun W, Kim H, Cho S, Lee KH, Kim K. Adrenal peripheral clock controls the autonomous circadian rhythm of glucocorticoid by causing rhythmic steroid production. Proc Natl Acad Sci U S A. 2008; 105:20970–20975.
25. Huang N, Chelliah Y, Shan Y, Taylor CA, Yoo SH, Partch C, Green CB, Zhang H, Takahashi JS. Crystal structure of the heterodimeric CLOCK:BMAL1 transcriptional activator complex. Science. 2012; 337:189–194.
26. Kornmann B, Schaad O, Bujard H, Takahashi JS, Schibler U. System-driven and oscillator-dependent circadian transcription in mice with a conditionally active liver clock. PLoS Biol. 2007; 5:e34.
27. Panda S, Antoch MP, Miller BH, Su AI, Schook AB, Straume M, Schultz PG, Kay SA, Takahashi JS, Hogenesch JB. Coordinated transcription of key pathways in the mouse by the circadian clock. Cell. 2002; 109:307–320.
28. Bass J, Takahashi JS. Circadian integration of metabolism and energetics. Science. 2010; 330:1349–1354.
29. Masri S, Sassone-Corsi P. The circadian clock: a framework linking metabolism, epigenetics and neuronal function. Nat Rev Neurosci. 2013; 14:69–75.
30. Luo J, Nikolaev AY, Imai S, Chen D, Su F, Shiloh A, Guarente L, Gu W. Negative control of p53 by Sir2alpha promotes cell survival under stress. Cell. 2001; 107:137–148.
31. Picard F, Kurtev M, Chung N, Topark-Ngarm A, Senawong T, Machado De Oliveira R, Leid M, McBurney MW, Guarente L. Sirt1 promotes fat mobilization in white adipocytes by repressing PPAR-gamma. Nature. 2004; 429:771–776.
Full Text Links
  • ENM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr