523559

research-article2014

JBRXXX10.1177/0748730414523559Journal of Biological RhythmsSchneider et al. / HSP90 Regulates BMAL1 Stability

HSP90 Affects the Stability of BMAL1 and Circadian Gene Expression Rebecca Schneider,* René M. Linka,* and Hans Reinke*,†,1 *University of Düsseldorf, Medical Faculty, Institute of Clinical Chemistry and Laboratory Diagnostics, Düsseldorf, Germany, and †IUF—Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany Abstract  The mammalian circadian clock comprises a system of interconnected transcriptional and translational feedback loops. Proper oscillator function requires the precisely timed synthesis and degradation of core clock proteins. Heat shock protein 90 (HSP90), an adenosine triphosphate (ATP)dependent molecular chaperone, has important functions in many cellular regulatory pathways by controlling the activity and stability of its various client proteins. Despite accumulating evidence for interplay between the heat shock response and the circadian system, the role of HSP90 in the mammalian core clock is not known. The results of this study suggest that inhibition of the ATP-dependent chaperone activity of HSP90 impairs circadian rhythmicity of cultured mouse fibroblasts whereby amplitude and phase of the oscillations are predominantly affected. Inhibition of HSP90 shortened the half-life of BMAL1, which resulted in reduced cellular protein levels and blunted expression of rhythmic BMAL1-CLOCK target genes. Furthermore, the HSP90 isoforms HSP90AA1 and HSP90AB1, and not HSP90B1-GRP94 or TRAP1, are responsible for maintaining proper cellular levels of BMAL1 protein. In summary, these findings provide evidence for a model in which cytoplasmic HSP90 is required for transcriptional activation processes by the positive arm of the mammalian circadian clock. Keywords  heat shock protein 90 (HSP90), mammalian circadian clock, BMAL1, protein stability, clock gene expression, heat shock response

Molecular chaperones are essential for the maintenance of cellular homeostasis in mammals. Initially, they were identified as components of protection systems against proteotoxic stressors such as elevated temperature or heavy-metal exposure. After a proteotoxic event, chaperones bind to denatured proteins, assist in their refolding, and prevent protein aggregation (Gidalevitz et al., 2011). Subsequent studies revealed that also under nonstress conditions, individual chaperones perform a wide range of cellular

functions. Heat shock protein 90 (HSP90), for example, regulates the activity, stability, and subcellular localization of a large number of client proteins to which it binds in a selective manner together with associated cofactors (Trepel et al., 2010). Circadian clocks enable organisms to synchronize behavior and physiology to the light-dark cycle and thereby to anticipate periodically recurring events in their environment. The mammalian molecular transcriptional-translational clock that is ticking

1.  To whom all correspondence should be addressed: Hans Reinke, Institute of Clinical Chemistry and Laboratory Diagnostics, University of Düsseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany; e-mail: [email protected]. JOURNAL OF BIOLOGICAL RHYTHMS, Vol 29 No. 2, April 2014 87­–96 DOI: 10.1177/0748730414523559 © 2014 The Author(s)

87 Downloaded from jbr.sagepub.com at HOWARD UNIV UNDERGRAD LIBRARY on January 31, 2015

88  JOURNAL OF BIOLOGICAL RHYTHMS / April 2014

autonomously in almost all body cells can be considered to be centered on a negative feedback loop of Period (Per1 and Per2) and Cryptochrome (Cry1 and Cry2) gene transcription. Expression of genes in the negative feedback loop is activated by the essential activator proteins BMAL1 and CLOCK, and in addition retinoic acid–related orphan receptor activators and REV-ERB repressors stabilize the core circadian oscillator by interlocking the expression of the activator component Bmal1 with components of the negative feedback loop (Preitner et al., 2002; Brown et al., 2012). Light serves as a resetting signal for the central oscillator in the suprachiasmatic nucleus (SCN) in the hypothalamus, which synchronizes peripheral oscillators in the rest of the body (Reppert and Weaver, 2002). Body temperature cycles, however, are a strong resetting cue for peripheral clocks (Brown et al., 2002). Accumulating evidence suggests that the temperature-sensitive heat shock response system, the primary driver of inducible chaperone gene expression, is involved in the regulation of the core circadian oscillator. Mice lacking HSF1, the master regulator of the heat shock response, have a longer free-running period than their wild-type littermates (Reinke et al., 2008). In line with this observation, pharmacological inhibition of the heat shock pathway lengthens the circadian period of SCN cells and interferes with phase entrainment and temperature compensation in peripheral clocks of mice (Buhr et al., 2010). Genetic knockdown studies revealed subsequently that phase entrainment of cultured mouse fibroblasts by physiological temperature cycles requires HSF1 but not its close homologue, HSF2 (Saini et al., 2012). These findings raise the question of how components of the heat shock pathway interact with the molecular oscillator. One possibility is that HSF1 directly binds to regulatory regions of core clock genes and regulates their transcriptional rate. One study reported binding of HSF1 close to E-boxes and physical interaction of HSF1 with BMAL1 and CLOCK after heat shock at the Per2 promoter in mouse (Tamaru et al., 2012). A similar mechanism seems to play a role in the adaptation of mice to cold temperature (Chappuis et al., 2013). Alternatively, HSF1 might affect the circadian oscillator via the expression of designated HSF1 target genes such as Hsp90, which themselves have a wide range of biological functions (Li and Buchner, 2013). Indeed, HSP90 was found to directly regulate the core circadian oscillator in Arabidopsis thaliana by stabilization of the F-box protein ZEITLUPE (Kim et al., 2011). Loss of HSP90 reduced the cellular levels of the ZEITLUPE protein and lengthened the circadian period of the plants. The function of HSP90 in circadian clocks, however, seems to vary widely in

different organisms. In contrast to its function in the core clock mechanism in A. thaliana, HSP90 is involved in connecting the circadian clock of Drosophila melanogaster to behavioral output functions but seems to be dispensable in the core oscillator mechanism in the flies (Hung et al., 2009). So far, no function for HSP90 in the mammalian circadian clock has been reported. Here, we show that inhibition of HSP90 by various small molecules from different substance classes interfered with the circadian clockwork in cultured mouse fibroblasts. Blocking HSP90 function altered predominantly the phase and amplitude of the circadian oscillations, and at the same time, the expression levels of clock genes that are dependent on transcriptional activation by the BMAL1-CLOCK complex were strongly affected. This effect might at least in part be mediated by a shorter half-life and as a result reduced protein levels of BMAL1 that were observed after treatment of cells with HSP90 inhibitors or after small interfering RNA (siRNA)-mediated knockdown of the main cytoplasmic HSP90 isoforms HSP90AA1 and HSP90AB1. This study has therefore identified HSP90 as a regulator of BMAL1 stability and, as a consequence, of BMAL1-dependent transcriptional activation in the mammalian circadian clock. Materials and Methods Cell Culture and Real-Time Bioluminescence Monitoring NIH3T3-Bmal1-luciferase cells stably expressing a Bmal1-luciferase reporter gene (Nagoshi et al., 2004) were grown in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal calf serum (FCS) and 1% penicillin-streptomycin (P/S) (Life Technologies, Darmstadt, Germany). Confluent cells were trypsinized, and 5 × 105 cells were seeded into 35 mm culture dishes 1 day before real-time bioluminescence monitoring was initiated. Cellular clocks were synchronized with either 50% horse serum for 2 h or 100 nM dexamethasone for 30 min. After synchronization, the medium was replaced with phenol red-free DMEM (Life Technologies) that was supplemented with 10% FCS and 100 µM luciferin containing GA (geldanamycin), 17-AAG (17-allylamino-17-demethoxygeldanamycin), 17-AEP-GA (17-[2-(pyrrolidin1-yl)ethyl]amino-17-demethoxygeldanamycin; InvivoGen, Toulouse, France), or radicicol (Sigma, Taufkirchen, Germany). Real-time bioluminescence was monitored in a light-tight incubator using photomultiplier tube detector assemblies (LumiCycle, Actimetrics, Wilmette, USA). The phase, amplitude,

Downloaded from jbr.sagepub.com at HOWARD UNIV UNDERGRAD LIBRARY on January 31, 2015

Schneider et al. / HSP90 REGULATES BMAL1 STABILITY  89

and period length of circadian oscillations were determined with LumiCycle Analysis software. RNA Analyses For analyses of clock gene expression, NIH3T3Bmal1-luciferase cells were treated with 17-AEP-GA for 3 days. After synchronization with 50% horse serum for 2 h, the medium was replaced by 3 mL phenol-red free medium supplemented with 10% FCS, 1% P/S, 100 µM luciferin, and 800 nM 17-AEP-GA. Real-time bioluminescence was monitored, and cells were harvested after 48, 52, 56, 60, 64, or 68 h. Total RNA was isolated by Trizol-chloroform extraction and reverse-transcribed using the Quantitect Reverse Transcription Kit (Qiagen, Hilden, Germany). A quantitative reverse transcription polymerase chain reaction was carried out with the LightCycler480II detection system using the LightCycler480 Probes Master Mix (Roche, Penzberg, Germany) and specific primers and probes (Suppl. Table S1). Protein Degradation Analyses To generate cells expressing C-terminally Myctagged BMAL1, complementary DNA (cDNA) of Bmal1 (corresponding to NM_007489.3) was cloned into the MluI and ApaI sites of a derivative of the pMC vector (Linka et al., 2007) that was modified to provide C-terminal fusion to a cMyc-epitope tag linked by a Gly-Pro-Pro-Pro-Gly spacer. NIH3T3 cells were transfected with pMC3-Bmal1-cMyc using Effectene Transfection Reagent (Qiagen). Cells were cultured in DMEM supplemented with 10% FCS, 1% P/S, and 100 µg/mL hygromycin, and stable clones were selected. For analysis of protein stability, 3.75 × 105 cells were plated into 60 mm tissue culture dishes. After 24 h, the medium was replaced with medium containing 800 nM 17-AEP-GA. Cells were incubated for 1 day with 800 nM 17-AEP-GA, and then treated with 10 µg/mL cycloheximide (Sigma) for 2, 4, or 8 h. Cells were harvested, and whole-cell protein extracts were prepared by suspending the cells in an equal volume of 2 × NUN buffer (50 mM HEPES NaOH pH 7.6; 600 mM NaCl; 2 M urea; 2% NP-40; 2 mM DTT; protease inhibitor cocktail [Roche]). 50 µg protein lysate was separated by 8% SDS-polyacrylamide gel electrophoresis under reducing conditions and transferred to PVDF membranes (Millipore, Schwalbach, Germany). Membranes were probed with anti-cMyc– horseradish peroxidase (Miltenyi, Bergisch Gladbach, Germany) and anti-U2AF65 (Sigma), and they were developed using the enhanced chemiluminescence detection method (Amersham Biosciences, Frankfurt, Germany).

Quantification of Protein Half-Life We assumed that protein degradation followed first-order decay kinetics. The measured protein intensity data (denoted by N) was initially log-transformed. The decay rate constant (k) was calculated using the RGP function from Excel (Microsoft). From the decay rate constant, the half-life (T1/2) was calculated: (N = N0e−kt; ln(N) – ln(N0) = –kt; T1/2 = ln(2) / k). siRNA-Mediated Knockdown For siRNA-mediated knockdown experiments, 2 × 105 cells were seeded into 35 mm culture dishes. After 24 h incubation, cells were transfected with 10 nM or 25 nM ON-Targetplus smartpool siRNA (Thermo Scientific, Schwerte, Germany) targeting BMAL1 or HSP90 isoforms. ON-TARGETplus non-targeting siRNA was used as a control. Transfection was performed according to the manufacturer’s instructions. Cells were harvested after 96 h, and proteins were analyzed by immunoblotting using anti-BMAL1 (Santa Cruz Biotechnology, Heidelberg, Germany), anti-HSP90 (Abcam, Cambridge, UK), or antiU2AF65 antibody (Sigma). Cell Viability Analyses For analyses of cell viability, trypan blue dye exclusion staining was performed. Cells were harvested by trypsinization. After centrifugation, cells were suspended in 0.5 to 1 mL medium. Cells were mixed with an equal amount of trypan blue stain (0.4%) and analyzed in a Countess cell-counting chamber slide (Invitrogen, Carlsbad, USA). Cell count and viability were determined using the Countess automated cell counter. Results Inhibition of HSP90 Alters the Function of the Circadian Clock in Mouse Fibroblasts The effect of small-molecule-mediated inhibition of HSP90 on the cellular circadian clock was analyzed in NIH3T3 mouse fibroblasts stably expressing the rhythmically transcribed Bmal1-luciferase reporter gene (Nagoshi et al., 2004). Cellular HSP90 activity was inhibited by treating fibroblast cultures with the benzoquinone ansamycin 17-AEP-GA, a low-toxic geldanamycin analog with high stability in aqueous solution, and a Kd of 0.4 µM for binding to HSP90 in vitro (Tian et al., 2004). Incubation of cells at 0.6 µM 17-AEP-GA resulted in a considerable

Downloaded from jbr.sagepub.com at HOWARD UNIV UNDERGRAD LIBRARY on January 31, 2015

90  JOURNAL OF BIOLOGICAL RHYTHMS / April 2014 Schneider et al., Figure 1

Bmal1-luciferase reporter with regard to the phase shift and ampli2500 2.5E-03 Bmal1 tude (Fig. 1E), which supports the H2O idea that the reporter faithfully mim0.6µM 17-AEP-GA 1500 2.0E-03 icked the transcriptional changes 500 1.5E-03 characteristic for the endogenous Bmal1 gene and, by implication, the 1.0E-03 -500 changes of the entire clock 5.0E-04 -1500 machinery. To exclude that the observed 0E+00 -2500 44 48 52 56 60 64 68 72 changes in circadian clock parame0 24 48 72 96 120 Time after synchronization [hours] Time after synchronization [hours] ters resulted from idiosyncratic offtarget effects of one particular HSP90 inhibitor, two more benzoquinone B C D ansamycins, geldanamycin and 24 27 1200 ns ** *** 17-AAG, as well as the structurally 20 1000 distinct HSP90 inhibitor radicicol 26 (Schulte et al., 1998) were compared 16 800 25 to 17-AEP-GA in the same assay. Due 12 600 to the known cytotoxicity of these 24 substances (Guo et al., 2008), cell 8 400 death was analyzed in all samples by 23 4 200 dye exclusion staining. Figure 2 shows that the rhythmic expression 0 22 0 of Bmal1-luciferase was impaired by any of the inhibitors in a dose-dependent manner, suggesting that HSP90 as the common molecular target of all substances plays a role in the cirFigure 1.  Parameters of the circadian transcriptional oscillator after inhibition of heat shock protein 90 (HSP90). (A) Circadian oscillations in synchronized NIH3T3 mouse cadian clock mechanism. Again, fibroblasts stably expressing luciferase (luc) driven by the Bmal1 promoter (NIH3T3- changes in amplitude and phase Bmal1-luc) treated with 0.6 µM 17-AEP-GA (17-[2-(pyrrolidin-1-yl)ethyl]amino-17-dewere observed under all conditions, methoxygeldanamycin; gray) or solvent (black). Quantification of (B) amplitude, (C) and a complete inhibition of HSP90 phase, and (D) period length of NIH3T3-Bmal1-luc cells treated with 0.6 µM 17-AEP-GA might even lead to a lengthening of (gray) or solvent (black) (n = 8). (E) Endogenous Bmal1 messenger RNA (mRNA) levels in NIH3T3-Bmal1-luc cells treated with 0.8 µM 17-AEP-GA (gray) compared to solventthe circadian period, as was observed treated cells (black) (n = 2). Error bars indicate standard deviations. **p < 0.005; ***p < with high concentrations of geldana0.0005 (2-sided t-test). mycin analogs (Fig. 2A-2C). Moreover, while geldanamycin and impairment of circadian transcriptional oscillation 17-AAG caused cell death at higher concentrations, compared to the solvent-treated sample (Fig. 1A). To effects on the circadian clock were also seen under quantify the effect of HSP90 inhibition on the cellular nontoxic conditions with all substances (Suppl. Fig. S1). circadian clock, eight biological replicates were anaTaken together, chemical inhibition of HSP90 by lyzed for changes in clock parameters on treatment small molecules from different substance classes with 17-AEP-GA. Incubation with the HSP90 inhibiaffects the cellular circadian clock whereby the amplitor reduced the amplitude of the oscillations on avertude and phase of transcriptional oscillations are preage by about 30% (untreated 921 ± 122, 17-AEP-GA dominantly affected. 649 ± 173; Fig. 1B). The phase of the circadian clock was delayed by 2.75 h compared to untreated control Loss of HSP90 Activity Shortens samples (untreated 13.1 ± 1.1 h, 17-AEP-GA 15.8 ± the Half-Life of BMAL1 1.1 h; Fig. 1C). The circadian period length did not differ significantly between the 17-AEP-GA treated samples and the untreated controls under these conHSP90 controls the levels of many cellular proditions (untreated 24.9 ± 0.7 h, 17-AEP-GA 25.4 ± 0.8 teins, either of client proteins by direct interaction or h; Fig. 1D). Importantly, at day 3 after inhibition of indirectly via changing the activity or stability of regHSP90, the expression levels of endogenous Bmal1 ulators of protein half-lives (Trepel et al., 2010). messenger RNA (mRNA) were similar to those of the Protein degradation is also an important regulatory E

A

PG

H 2O

17 -A E

A

PG

H 2O

17 -A E

A

PG

17 -A E

H 2O

amplitude

phase [hours]

period [hours]

Luciferase [counts/min]

relative mRNA expression

A

Downloaded from jbr.sagepub.com at HOWARD UNIV UNDERGRAD LIBRARY on January 31, 2015

Schneider et al. / HSP90 REGULATES BMAL1 STABILITY  91 Schneider et al., Figure 2

2010), we supposed that BMAL1 protein is a molecDMSO DMSO ular target of HSP90 and 0.4µM GA 0.4µM 17-AAG 0.8µM GA 0.8µM 17-AAG that altered expression of 1000 1000 1.2µM GA 1.2µM 17-AAG Bmal1 mRNA on inhibition of HSP90 (Fig. 1E) is a con0 0 sequence rather than the cause for reduced BMAL1 -1000 -1000 levels. Moreover, Bmal1 is expressed with high circa-2000 -2000 0 24 48 72 96 120 0 24 48 72 96 120 dian amplitude in cells and Time after synchronization [hours] Time after synchronization [hours] tissues, whereas BMAL1 C D protein levels are rather 2000 2000 constant throughout the EtOH H2O 4µM Radicicol day (Asher et al., 2008), 0.4µM 17-AEP-GA 8µM Radicicol 0.8µM 17-AEP-GA 1000 1000 which emphasizes the 12µM Radicicol 1.2µM 17-AEP-GA importance of posttransla0 0 tional regulation for the control of BMAL1 levels. -1000 -1000 Therefore, the stability of BMAL1 was analyzed in the presence or absence of -2000 -2000 0 24 48 72 96 120 0 24 48 72 96 HSP90 activity by blocking Time after synchronization [hours] Time after synchronization [hours] protein translation and determining the kinetics of Figure 2. Effects of structurally distinct heat shock protein 90 (HSP90) inhibitors on the cellular BMAL1 protein degradacircadian clock. Circadian oscillations in synchronized NIH3T3-Bmal1-luciferase cells treated with tion. Indeed, 17-AEP-GA various concentrations of (A) geldanamycin (GA), (B) 17-allylamino-17-demethoxygeldanamycin (17-AAG), (C) 17-[2-(pyrrolidin-1-yl)ethyl]amino-17-demethoxygeldanamycin (17-AEP-GA), or (D) treatment led to a faster radicicol. decline of BMAL1 protein levels in the cells compared to the untreated control samples (Fig. 3B and 3E), mechanism in the mammalian circadian clockwork. suggesting that HSP90 controls BMAL1 stability. For example, the stabilities of BMAL1 (Sahar et al., To corroborate this result in an independent experi2010; Zhang et al., 2012b; Ma et al., 2013), mental system, a mouse fibroblast cell line stably Cryptochromes (Godinho et al., 2007; Lamia et al., expressing cMyc-tagged BMAL1 was created, and the 2009), and Period proteins (Reischl et al., 2007; Asher half-life of BMAL1 was determined under the same et al., 2008) codetermine core clock parameters such conditions. Similar to the faster degradation rate that as amplitude and period length and are connected to was observed for endogenous BMAL1 in cells treated the metabolic control of circadian rhythmicity. with HSP90 inhibitor (Fig. 3B and 3E), also BMAL1BMAL1 and HSP90 have been found to interact as in cMyc levels expressed from the noncircadian cytovitro translated proteins (Hogenesch et al., 1997), and megalovirus promoter (Asher et al., 2008) declined knockdown of BMAL1 disrupts the cellular oscillator considerably faster in the absence of functional HSP90 (Baggs et al., 2009; Wallach et al., 2013), which compared to the degradation rate in untreated control prompted us to analyze the effect of HSP90 inhibitors cells (Fig. 3C and 3F). In summary, these findings proon cellular BMAL1 levels. We found the total levels of vide evidence that HSP90 regulates the cellular BMAL1 protein in cells treated with HSP90 inhibitor amount of BMAL1 and that reduced protein levels to be considerably lower compared to the levels in can be explained at least in part with a shorter half-life untreated cells (Fig. 3A). The amount of the slowest of BMAL1 in the absence of functional HSP90. migrating protein band, potentially representing a hyperphosphorylated form of BMAL1, was most strongly reduced after inhibition of HSP90, suggestThe Main Cytoplasmic HSP90 Isoform Is Required ing that dephosphorylation of BMAL1 might precede for the Stabilization of BMAL1 its degradation. Quantification of four independent experiments revealed a significant reduction of about As mentioned, chemical inhibitors can have side 40% of BMAL1 protein levels in 17-AEP-GA-treated effects on unrelated target proteins. To pinpoint the cells (Fig. 3D). Due to the well-established function of observed changes in BMAL1 protein stability to the HSP90 in the control of protein stability (Trepel et al., A

B

Luciferase [counts/min]

2000

Luciferase [counts/min]

Luciferase [counts/min]

Luciferase [counts/min]

2000

Downloaded from jbr.sagepub.com at HOWARD UNIV UNDERGRAD LIBRARY on January 31, 2015

92  JOURNAL OF BIOLOGICAL RHYTHMS / April 2014 Schneider et al., Figure 3 A 17-AEP-GA

-

+

0

2

-

+

-

4

8

0

+

-

+

BMAL1 U2AF65

B

H2O CHX [h]

17-AEP-GA

2

4

8

BMAL1 U2AF65

C

H2O

CHX [h]

0

2

17-AEP-GA

4

8

0

2

4

NIH3T3

8

BMAL1-cMyc U2AF65

150

*

100 50

17 H -A 2 EP O -G A

0

200

relative BMAL1 levels

200

relative BMAL1 levels

F

E

150

ns

150

100 50

* ** **

0

0

*

*

2 4 6 8 CHX [hours]

relative BMAL1-cMyc levels

D

100

50

0

* 0

2 4 6 8 CHX [hours]

Figure 3.  Cellular levels and stability of BMAL1 after inhibition of heat shock protein 90 (HSP90). (A) Cellular levels of BMAL1 protein determined by immunoblotting in four independent experiments in NIH3T3 cells treated with 0.8 µM 17-AEP-GA. U2AF65 protein levels were analyzed as a control for equal sample loading. Gel cuts are indicated by dotted lines. (B) Time course of BMAL1 and U2AF65 protein degradation in NIH3T3 cells treated with 0.8 µM 17-AEP-GA after inhibition of protein synthesis by cycloheximide (CHX) for the indicated duration. The 0-h (CHX) lanes are identical with the last two lanes in (A). (C) Time course of BMAL1 and U2AF65 protein degradation in NIH3T3 cells stably expressing BMAL1-cMyc treated with 0.8 µM 17-AEP-GA after inhibition of protein synthesis by cycloheximide (CHX) for the indicated duration. The most rightward lane shows untransfected NIH3T3 cells that do not express BMAL1-cMyc. (D) Quantification of BMAL1 protein levels from (A). (E) Quantification of BMAL1 protein levels from (B). Protein half-life H2O 42 h, and 17-AEP-GA 16 h. Asterisks above gray data points refer to significance levels of 17-AEP-GA versus H2O. The asterisk below the gray data points refers to the significance level of 17-AEP-GA t = 0 versus t = 8. (F) Quantification of BMAL1-cMyc protein levels from (C) normalized to the respective 0 h (CHX) value. Protein half-life H2O 7 h, and 17-AEP-GA 3 h. (n = 3). Error bars indicate standard deviations. *p < 0.05; **p < 0.005 (2-sided t-test).

activity of HSP90, single proteins were targeted by siRNA against all HSP90 variants that are known to be inhibited by geldanamycin analogs and radicicol.

The main cytoplasmic isoform of HSP90 is expressed from two highly similar genes, Hsp90aa1 and Hsp90ab1. Both genes were knocked down simultaneously, and as a control Bmal1 was targeted directly by a specific siRNA. Figure 4A shows that knockdown of Hsp90aa1 and Hsp90ab1 led to a significant reduction in total cellular BMAL1 levels at the higher siRNA concentration, which was almost as strong as when Bmal1 was targeted directly (Fig. 4B). HSP90 exists in several isoforms that accumulate in different cellular compartments. The main cytoplasmic HSP90 variants are HSP90AA1 and HSP90AB1, which have been shown to be localized in the nucleus as well (Collier and Schlesinger, 1986). HSP90B1-GRP94 is mainly targeted to the lumen of the endoplasmatic reticulum, where it plays a role in the unfolded protein response (Lee, 1981). The HSP90 isoform TRAP1 is a largely mitochondrial protein with functions in mitochondrial integrity, apoptosis, and oxidative stress (Altieri et al., 2012). Importantly, however, interactions with exclusively cytoplasmic proteins have been described for all HSP90 isoforms, and subpools of all proteins seem to exist in the cytoplasm and/or the nucleus (Welch et al., 1983; Chen et al., 1996; Csermely et al., 1998). Moreover, all HSP90 isoforms are well-established regulators of protein stability (Chavany et al., 1996; Landriscina et al., 2010) and have therefore the potential to control cellular BMAL1 levels. Only downregulation of cytoplasmic HSP90 by simultaneous siRNA-mediated knockdown of the highly similar proteins HSP90AA1 and HSP90AB1, however, lowered the cellular levels of BMAL1 protein (Fig. 4C). The high concentration of Trap1-specific siRNA was highly toxic, and therefore the data were excluded from the analysis. Taken together, these experiments showed that despite the fact that HSP90 is a highly abundant protein, its downregulation reduced the cellular amount of BMAL1 very efficiently (Fig. 4B), suggesting that the stability of BMAL1 is dependent on HSP90. Inhibition of HSP90 Affects the Expression of BMAL1-CLOCK Target Genes Finally, we asked if inhibition of HSP90 affects the expression of designated BMAL1-CLOCK target genes and analyzed mRNA levels of several core clock genes in synchronized mouse fibroblasts after inhibition of HSP90. Figure 5 shows that peak mRNA levels of strongly BMAL1-CLOCK-dependent genes such as Nr1d1 (Rev-Erb alpha) (Ripperger, 2006), Dbp (Ripperger and Schibler, 2006), and Per2 (Yoo et al., 2005) were considerably blunted under these conditions (Fig. 5A-5C). In particular, the maximal expression of Nr1d1 68 h after synchronization was reduced more than 8-fold (Fig. 5A). Expression of Clock,

Downloaded from jbr.sagepub.com at HOWARD UNIV UNDERGRAD LIBRARY on January 31, 2015

Schneider et al. / HSP90 REGULATES BMAL1 STABILITY  93 Schneider et al., Figure 4

inhibition of HSP90 interferes with circadian rhythmicity in cultured * 150 cells. Treatment of mouse fibroblasts 10 25 10 25 siRNA] [nM 10 25 ** with different geldanamycin derivaHSP90 100 tives or radicicol altered the phase BMAL1 and amplitude of the circadian clock. * Importantly, changes of clock param50 U2AF65 eters were observed in the absence of cell death in confluent cell cultures 0 C Hsp90aa1 non- Bmal1 Hsp90 excluding cytotoxicity and cell cycle Hsp90ab1 Trap1 Hsp90b1 targeting arrest, two common global side effects siRNA [25nM] siRNA] [nM 10 25 10 25 10 of HSP90 inhibition (Srethapakdi et HSP90 al., 2000), as confounding factors. It should be pointed out that the BMAL1 observed effects on transcriptional * U2AF65 oscillations do not necessarily reflect the endpoint of HSP90 inhibition. HSP90 activity might not have been Figure 4.  Small interfering RNA (siRNA)-mediated knockdown of heat shock pro- fully abolished under any of these conditions, and higher concentrations tein (HSP90) isoforms. (A) HSP90, BMAL1, and U2AF65 protein levels in NIH3T3 cells transfected with nontargeting siRNA or siRNAs for Bmal1 or Hsp90aa1-Hsp90ab1. might lead to a total breakdown of Arrows indicate specific protein bands; the asterisk marks a protein band that is non- rhythmicity. The observed lengthenspecifically detected by the BMAL1-specific antibody. (B) Quantification of BMAL1 ing of circadian period at higher protein levels after knockdown of Bmal1 or Hsp90aa1-Hsp90ab1 from (A) (n = 3). (C) HSP90, BMAL1, and U2AF65 protein levels in NIH3T3 cells transfected with siRNAs inhibitor concentrations was indicafor Hsp90aa1-Hsp90ab1, Hsp90b1, or Trap1. Error bars indicate standard deviations. *p < tive of such an effect. Due to the cyto0.05; **p < 0.005 (2-sided t-test). toxicity of HSP90 inhibitors at elevated concentrations, however, it however, was unchanged after treatment with will be difficult to address this point in a comprehen17-AEP-GA (Fig. 5D), which shows that the mechasive manner. nism by which HSP90 regulates the cellular circadian Second, in the presence of HSP90 inhibitors, we clock does not affect all components of the oscillator found considerably reduced mRNA levels, particumechanism. Taken together, our findings provide evilarly of clock genes that strongly depend on BMAL1dence for a model in which HSP90 is required for the CLOCK for transcriptional activation such as Nr1d1 stabilization of BMAL1 and thereby guarantees (Ripperger, 2006). On the level of gene expression, proper transcriptional oscillation of BMAL1-CLOCKthis effect might result from a decrease in transcripdependent clock genes. tional activation (e.g., due to diminished BMAL1CLOCK activity; Ripperger and Schibler, 2006) or an increase in transcriptional repression (e.g., due to Discussion elevated levels of PER; Asher et al., 2008). In fact, when HSP90 activity was blocked, we observed a In this study, we set out to analyze the function of shorter protein half-life of the transcriptional activaHSP90 in the circadian clockwork in cultured mammator BMAL1 and, as a consequence, reduced cellular lian cells. HSP90 seemed to be an obvious candidate levels of BMAL1. It has been shown before in U2OS for a chaperone regulator of the circadian clock, since it cells that diminished levels of BMAL1 lead to lower is expressed in a highly circadian manner in mouse circadian amplitudes and complete loss of BMAL1 to (Kornmann et al., 2007), and interplay between HSP90 arrhythmicity (Baggs et al., 2009), supporting the and circadian clocks has been demonstrated in other idea that BMAL1 is an important target of HSP90 in model organisms (Hung et al., 2009; Kim et al., 2011). the core oscillator mechanism, yet the contribution of Moreover, HSP90 can be efficiently inhibited by smallother mechanisms cannot be excluded at this point. molecule inhibitors, most of which compete to its Finally, gene-specific knockdown revealed that loss N-terminal ATPase domain, which is essential for all of HSP90AA1 and HSP90AB1 has the same effect on known functions of HSP90 (Nadeau et al., 1993). cellular BMAL1 levels as chemical HSP90 inhibitors. The results of this study provide evidence from varCytoplasmic HSP90 has therefore been identified as ious experimental approaches that HSP90 plays a role a regulator of BMAL1 stability. in the BMAL1-CLOCK-containing positive arm of the Future work has to reveal how HSP90 affects mammalian circadian clock. First, we found that BMAL1-CLOCK-dependent processes on the non-targeting

Bmal1

Hsp90aa1 Hsp90ab1

B

relative BMAL1 levels

A

Downloaded from jbr.sagepub.com at HOWARD UNIV UNDERGRAD LIBRARY on January 31, 2015

94  JOURNAL OF BIOLOGICAL RHYTHMS / April 2014 Schneider et al., Figure 5

O-GlcNAcylated,which blocks ubiquitination and thereby degradation 3.0E-03 7.0E-03 Dbp Nr1d1 (Ma et al., 2013). Inhibition of HSP90 2.5E-03 5.6E-03 has been shown to destabilize 2.0E-03 O-linked β-N-acetylglucosamine 4.2E-03 1.5E-03 transferase (OGT) (Zhang et al., 2.8E-03 1.0E-03 2012a), and the resulting decrease in 1.4E-03 5.0E-04 O-GlcNAcylation levels might pro0E+00 0E+00 mote the degradation of BMAL1. 44 48 52 56 60 64 68 72 44 48 52 56 60 64 68 72 The protein levels of BMAL1 are also Time after synchronization [hours] Time after synchronization [hours] controlled via phosphorylationD C mediated degradation through the 2.5E-03 1.0E-03 Clock Per2 kinase GSK-3β (Spengler et al., 2009; 2.0E-03 8.0E-04 Sahar et al., 2010), and the maturation of GSK-3β is sensitive to geldan1.5E-03 6.0E-04 amycin-mediated inhibition of 1.0E-03 4.0E-04 HSP90 (Lochhead et al., 2006). 5.0E-04 2.0E-04 Of further interest is the question of 0E+00 0E+00 how HSF1 and HSP90 work together 44 48 52 56 60 64 68 72 44 48 52 56 60 64 68 72 in the regulation of the circadian Time after synchronization [hours] Time after synchronization [hours] clock. One obvious possibility is that HSF1 regulates the circadian expresFigure 5.  Messenger RNA (mRNA) levels of core clock genes after inhibition of heat sion of Hsp90. Since HSF1 is required shock protein (HSP90). Expression of core clock genes in NIH3T3-Bmal1-luc cells for inducible transcriptional activatreated with 0.8 µM 17-AEP-GA (gray) or solvent (black). (A) Nr1d1, (B) Dbp, (C) Per2, tion of Hsp90 after heat shock (Xiao et and (D) Clock (n = 2). Error bars indicate standard deviations. al., 1999) and both the circadian activity of HSF1 and rhythmic expression molecular level. A direct interaction between BMAL1 of Hsp90 can be driven by systemic cues (Kornmann et and HSP90 has been reported with in vitro translated al., 2007; Reinke et al., 2008), HSF1 can be considered a proteins (Hogenesch et al., 1997). BMAL1 and bona fide regulator of circadian Hsp90 expression. CLOCK have been proposed to be kamikaze activaFinally, these findings open the question of what tors that are primed for degradation when they are roles the interplay between HSP90 and the circadian bound to chromatin (Stratmann et al., 2012). clock might play in mammalian homeostasis and disInhibition of HSP90 might accelerate this degradaease. HSP90AA1 has recently been identified in silico tion process and thereby lead to reduced mRNA synas a major hub in the human circadian protein-prothesis of BMAL1-CLOCK target genes. In support of tein interactome (Wallach et al., 2013). Its proposed such a model would be the reported interaction rhythmic interactions with other cellular proteins between BMAL1-CLOCK and HSF1 (Tamaru et al., inside and outside of the core clock mechanism are 2012), since HSF1 interacts with HSP90 and might supposed to contribute significantly to the temporal recruit HSP90 to the BMAL1-CLOCK complex. organization of cellular physiology. Systemically HSP90 might also associate with BMAL1 in the cytodriven diurnal expression of HSP90 (Kornmann et al., plasm, alone or in a complex with CLOCK, and pro2007) could be involved in temperature entrainment tect it from proteasomal degradation or affect nuclear or amplitude regulation of the circadian clock by import (Kwon et al., 2006). A potential interaction timed stabilization of BMAL1 during the circadian between BMAL1 and HSP90, however, might be cycle. A similar mechanism involving the PKCgvery weak or transient, and we did not succeed in mediated stabilization of BMAL1 has recently been demonstrating such an interaction in mouse liver shown to regulate the entrainment of the circadian nuclear extracts, which should contain the majority clock by timely restricted feeding (Zhang et al., of BMAL1 (Kwon et al., 2006) and significant 2012b). The requirement for HSP90 in maintaining amounts of HSP90 (Collier and Schlesinger, 1986), or proper cellular levels of BMAL1, however, adds to whole-cell extracts from cultured fibroblasts (data the complexity of using HSP90 inhibitors in cancer not shown). Therefore, the alternative that HSP90 therapy. Downregulation of BMAL1 accelerates regulates BMAL1 via an indirect mechanism should tumor growth (Zeng et al., 2010), and inhibition of also be considered. Several regulatory mechanisms HSP90 would further promote this process, providhave been described that influence the cellular leving another example for the often ambivalent role of els of BMAL1 protein. BMAL1 is rhythmically HSP90 in physiological and pathological processes. relative mRNA expression

B

relative mRNA expression

relative mRNA expression

relative mRNA expression

A

Downloaded from jbr.sagepub.com at HOWARD UNIV UNDERGRAD LIBRARY on January 31, 2015

Schneider et al. / HSP90 REGULATES BMAL1 STABILITY  95

Acknowledgments The cDNA of Bmal1 was kindly provided by Aziz Sancar. The authors thank Gabriele Schoder for excellent technical assistance and Christian Mielke for valuable comments on the manuscript. This work was supported by the Deutsche Forschungsgemeinschaft (SFB 728, RE 3046/2-1) and the Human Frontier Science Program (CDA00009/2009-C).

Conflict of Interest Statement The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

NOTE Supplementary online material is available on the journal’s website at http://jbr.sagepub.com/supplemental.

References Altieri DC, Stein GS, Lian JB, and Languino LR. (2012) TRAP-1, the mitochondrial Hsp90. Biochim Biophys Acta 1823(3):767-773. Asher G, Gatfield D, Stratmann M, Reinke H, Dibner C, Kreppel F, Mostoslavsky R, Alt FW, and Schibler U. (2008) SIRT1 regulates circadian clock gene expression through PER2 deacetylation. Cell 134(2):317-328. Baggs JE, Price TS, DiTacchio L, Panda S, Fitzgerald GA, and Hogenesch JB. (2009) Network features of the mammalian circadian clock. PLoS Biol 7(3):e52. Brown SA, Kowalska E, and Dallmann R. (2012) (Re)inventing the circadian feedback loop. Dev Cell 22(3):477-487. Brown SA, Zumbrunn G, Fleury-Olela F, Preitner N, and Schibler U. (2002) Rhythms of mammalian body temperature can sustain peripheral circadian clocks. Curr Biol 12(18):1574-1583. Buhr ED, Yoo SH, and Takahashi JS. (2010) Temperature as a universal resetting cue for mammalian circadian oscillators. Science 330(6002):379-385. Chappuis S, Ripperger JA, Schnell A, Rando G, Jud C, Wahli W, and Albrecht U. (2013) Role of the circadian clock gene Per2 in adaptation to cold temperature. Mol Metab 2(3):184-193. Chavany C, Mimnaugh E, Miller P, Bitton R, Nguyen P, Trepel J, Whitesell L, Schnur R, Moyer J, and Neckers L. (1996) p185erbB2 binds to GRP94 in vivo: dissociation of the p185erbB2/GRP94 heterocomplex by benzoquinone ansamycins precedes depletion of p185erbB2. J Biol Chem 271(9):4974-4977.

Chen CF, Chen Y, Dai K, Chen PL, Riley DJ, and Lee WH. (1996) A new member of the hsp90 family of molecular chaperones interacts with the retinoblastoma protein during mitosis and after heat shock. Mol Cell Biol 16(9):4691-4699. Collier NC and Schlesinger MJ. (1986) The dynamic state of heat shock proteins in chicken embryo fibroblasts. J Cell Biol 103(4):1495-1507. Csermely P, Schnaider T, Soti C, Prohaszka Z, and Nardai G. (1998) The 90-kDa molecular chaperone family: structure, function, and clinical applications: a comprehensive review. Pharmacol Ther 79(2):129-168. Gidalevitz T, Prahlad V, and Morimoto RI. (2011) The stress of protein misfolding: from single cells to multicellular organisms. Cold Spring Harbor Persp Biol 3(6). doi:10.1101/cshperspect.a009704 Godinho SI, Maywood ES, Shaw L, Tucci V, Barnard AR, Busino L, Pagano M, Kendall R, Quwailid MM, Romero MR, et al. (2007) The after-hours mutant reveals a role for Fbxl3 in determining mammalian circadian period. Science 316(5826):897-900. Guo W, Reigan P, Siegel D, and Ross D. (2008) Enzymatic reduction and glutathione conjugation of benzoquinone ansamycin heat shock protein 90 inhibitors: relevance for toxicity and mechanism of action. Drug Metab Dispos 36(10):2050-2057. Hogenesch JB, Chan WK, Jackiw VH, Brown RC, Gu YZ, Pray-Grant M, Perdew GH, and Bradfield CA. (1997) Characterization of a subset of the basic-helix-loop-helixPAS superfamily that interacts with components of the dioxin signaling pathway. J Biol Chem 272(13):8581-8593. Hung HC, Kay SA, and Weber F. (2009) HSP90, a capacitor of behavioral variation. J Biol Rhythms 24(3):183-192. Kim TS, Kim WY, Fujiwara S, Kim J, Cha JY, Park JH, Lee SY, and Somers DE. (2011) HSP90 functions in the circadian clock through stabilization of the client F-box protein ZEITLUPE. Proc Natl Acad Sci U S A 108(40):16843-16848. Kornmann B, Schaad O, Bujard H, Takahashi JS, and Schibler U. (2007) System-driven and oscillator-dependent circadian transcription in mice with a conditionally active liver clock. PLoS Biol 5(2):e34. Kwon I, Lee J, Chang SH, Jung NC, Lee BJ, Son GH, Kim K, and Lee KH. (2006) BMAL1 shuttling controls transactivation and degradation of the CLOCK/BMAL1 heterodimer. Mol Cell Biol 26(19):7318-7330. Lamia KA, Sachdeva UM, DiTacchio L, Williams EC, Alvarez JG, Egan DF, Vasquez DS, Juguilon H, Panda S, Shaw RJ, et al. (2009) AMPK regulates the circadian clock by cryptochrome phosphorylation and degradation. Science 326(5951):437-440. Landriscina M, Laudiero G, Maddalena F, Amoroso MR, Piscazzi A, Cozzolino F, Monti M, Garbi C, Fersini A, Pucci P, et al. (2010) Mitochondrial chaperone Trap1 and the calcium binding protein Sorcin interact and protect cells against apoptosis induced by antiblastic agents. Cancer Res 70(16):6577-6586.

Downloaded from jbr.sagepub.com at HOWARD UNIV UNDERGRAD LIBRARY on January 31, 2015

96  JOURNAL OF BIOLOGICAL RHYTHMS / April 2014 Lee AS. (1981) The accumulation of three specific proteins related to glucose-regulated proteins in a temperaturesensitive hamster mutant cell line K12. J Cell Physiol 106(1):119-125. Li J and Buchner J. (2013) Structure, function and regulation of the hsp90 machinery. Biomed J 36(3):106-117. Linka RM, Porter AC, Volkov A, Mielke C, Boege F, and Christensen MO. (2007) C-terminal regions of topoisomerase IIalpha and IIbeta determine isoform-specific functioning of the enzymes in vivo. Nucleic Acids Res 35(11):3810-3822. Lochhead PA, Kinstrie R, Sibbet G, Rawjee T, Morrice N, and Cleghon V. (2006) A chaperone-dependent GSK3beta transitional intermediate mediates activation-loop autophosphorylation. Mol Cell 24(4):627-633. Ma Y, Luo H, Guan WJ, Zhang H, Chen C, Wang Z, and Li JD. (2013) O-GlcNAcylation of BMAL1 regulates circadian rhythms in NIH3T3 fibroblasts. Biochem Biophys Res Commun 431(3):382-387. Nadeau K, Das A, and Walsh CT. (1993) Hsp90 chaperonins possess ATPase activity and bind heat shock transcription factors and peptidyl prolyl isomerases. J Biol Chem 268(2):1479-1487. Nagoshi E, Saini C, Bauer C, Laroche T, Naef F, and Schibler U. (2004) Circadian gene expression in individual fibroblasts: cell-autonomous and self-sustained oscillators pass time to daughter cells. Cell 119(5):693-705. Preitner N, Damiola F, Lopez-Molina L, Zakany J, Duboule D, Albrecht U, and Schibler U. (2002) The orphan nuclear receptor REV-ERBalpha controls circadian transcription within the positive limb of the mammalian circadian oscillator. Cell 110(2):251-260. Reinke H, Saini C, Fleury-Olela F, Dibner C, Benjamin IJ, and Schibler U. (2008) Differential display of DNAbinding proteins reveals heat-shock factor 1 as a circadian transcription factor. Genes Dev 22(3):331-345. Reischl S, Vanselow K, Westermark PO, Thierfelder N, Maier B, Herzel H, and Kramer A. (2007) Beta-TrCP1mediated degradation of PERIOD2 is essential for circadian dynamics. J Biol Rhythms 22(5):375-386. Reppert SM and Weaver DR. (2002) Coordination of circadian timing in mammals. Nature 418(6901):935-941. Ripperger JA. (2006) Mapping of binding regions for the circadian regulators BMAL1 and CLOCK within the mouse Rev-erbalpha gene. Chronobiol Int 23(1-2):135-142. Ripperger JA and Schibler U. (2006) Rhythmic CLOCKBMAL1 binding to multiple E-box motifs drives circadian Dbp transcription and chromatin transitions. Nat Genet 38(3):369-374. Sahar S, Zocchi L, Kinoshita C, Borrelli E, and SassoneCorsi P. (2010) Regulation of BMAL1 protein stability and circadian function by GSK3beta-mediated phosphorylation. PLoS ONE 5(1):e8561. Saini C, Morf J, Stratmann M, Gos P, and Schibler U. (2012) Simulated body temperature rhythms reveal the phaseshifting behavior and plasticity of mammalian circadian oscillators. Genes Dev 26(6):567-580.

Schulte TW, Akinaga S, Soga S, Sullivan W, Stensgard B, Toft D, and Neckers LM. (1998) Antibiotic radicicol binds to the N-terminal domain of Hsp90 and shares important biologic activities with geldanamycin. Cell Stress Chaperones 3(2):100-108. Spengler ML, Kuropatwinski KK, Schumer M, and Antoch MP. (2009) A serine cluster mediates BMAL1dependent CLOCK phosphorylation and degradation. Cell Cycle 8(24):4138-4146. Srethapakdi M, Liu F, Tavorath R, and Rosen N. (2000) Inhibition of Hsp90 function by ansamycins causes retinoblastoma gene product-dependent G1 arrest. Cancer Res 60(14):3940-3946. Stratmann M, Suter DM, Molina N, Naef F, and Schibler U. (2012) Circadian Dbp transcription relies on highly dynamic BMAL1-CLOCK interaction with E boxes and requires the proteasome. Mol Cell 48(2):277-287. Tamaru T, Hattori M, Honda K, Benjamin I, Ozawa T, and Takamatsu K. (2012) Synchronization of circadian Per2 rhythms and HSF1-BMAL1:CLOCK interaction in mouse fibroblasts after short-term heat shock pulse. PLoS ONE 6(9):e24521. Tian ZQ, Liu Y, Zhang D, Wang Z, Dong SD, Carreras CW, Zhou Y, Rastelli G, Santi DV, et al. (2004) Synthesis and biological activities of novel 17-aminogeldanamycin derivatives. Bioorg Med Chem 12(20):5317-5329. Trepel J, Mollapour M, Giaccone G, and Neckers L. (2010) Targeting the dynamic HSP90 complex in cancer. Nature Reviews Cancer 10(8):537-549. Wallach T, Schellenberg K, Maier B, Kalathur RK, Porras P, Wanker EE, Futschik ME, and Kramer A. (2013) Dynamic circadian protein-protein interaction networks predict temporal organization of cellular functions. PLoS Genet 9(3):e1003398. Welch WJ, Garrels JI, Thomas GP, Lin JJ, and Feramisco JR. (1983) Biochemical characterization of the mammalian stress proteins and identification of two stress proteins as glucose- and Ca2+-ionophore-regulated proteins. J Biol Chem 258(11):7102-7111. Yoo SH, Ko CH, Lowrey PL, Buhr ED, Song EJ, Chang S, Yoo OJ, Yamazaki S, Lee C, and Takahashi JS. (2005) A noncanonical E-box enhancer drives mouse Period2 circadian oscillations in vivo. Proc Natl Acad Sci U S A 102(7):2608-2613. Zeng ZL, Wu MW, Sun J, Sun YL, Cai YC, Huang YJ, and Xian LJ. (2010) Effects of the biological clock gene Bmal1 on tumour growth and anti-cancer drug activity. J Biochem (Tokyo) 148(3):319-326. Zhang F, Snead CM, and Catravas JD. (2012a) Hsp90 regulates O-linked -N-acetylglucosamine transferase: a novel mechanism of modulation of protein O-linked -N-acetylglucosamine modification in endothelial cells. Amer J Physiol 302(12):C1786-1796. Zhang L, Abraham D, Lin ST, Oster H, Eichele G, Fu YH, and Ptacek LJ. (2012b) PKC participates in food entrainment by regulating BMAL1. Proc Natl Acad Sci U S A 109(50):20679-20684.

Downloaded from jbr.sagepub.com at HOWARD UNIV UNDERGRAD LIBRARY on January 31, 2015

HSP90 affects the stability of BMAL1 and circadian gene expression.

The mammalian circadian clock comprises a system of interconnected transcriptional and translational feedback loops. Proper oscillator function requir...
799KB Sizes 1 Downloads 3 Views