You are on page 1of 29

nature cell biology

Article https://doi.org/10.1038/s41556-022-01061-0

Nucleus-exported CLOCK acetylates PRPS to


promote de novo nucleotide synthesis and
liver tumour growth

Received: 28 February 2022 Tong Liu    1,2,3,4,12, Zheng Wang1,2,12, Leiguang Ye5,6,12, Yuran Duan1,12,
Hongfei Jiang7, Haiyan He1,2, Liwei Xiao1,2, Qingang Wu1,2, Yan Xia8,
Accepted: 24 November 2022
Mengke Yang9, Ke Wu1, Meisi Yan10, Guimei Ji1, Yuli Shen1, Lei Wang1, Lin Li1,
Published online: 16 January 2023 Peixiang Zheng1, Bofei Dong1, Fei Shao    7, Xu Qian11, Rilei Yu9, Zhiren Zhang4,
Zhimin Lu    1,2  & Daqian Xu    1,2 
Check for updates

Impairment of the circadian clock is linked to cancer development. However,


whether the circadian clock is modulated by oncogenic receptor tyrosine
kinases remains unclear. Here we demonstrated that receptor tyrosine
kinase activation promotes CK2-mediated CLOCK S106 phosphorylation
and subsequent disassembly of the CLOCK–BMAL1 dimer and suppression
of the downstream gene expression in hepatocellular carcinoma (HCC)
cells. In addition, CLOCK S106 phosphorylation exposes its nuclear export
signal to bind Exportin1 for nuclear exportation. Cytosolic CLOCK acetylates
PRPS1/2 K29 and blocks HSC70-mediated and lysosome-dependent PRPS1/2
degradation. Stabilized PRPS1/2 promote de novo nucleotide synthesis and
HCC cell proliferation and liver tumour growth. Furthermore, CLOCK S106
phosphorylation and PRPS1/2 K29 acetylation are positively correlated in
human HCC specimens and with HCC poor prognosis. These findings delineate
a critical mechanism by which oncogenic signalling inhibits canonical CLOCK
transcriptional activity and simultaneously confers CLOCK with instrumental
moonlighting functions to promote nucleotide synthesis and tumour growth.

Circadian rhythms govern many physiological and metabolic functions output cycles kaput (CLOCK) and brain and muscle Arnt-like protein 1
through the internal self-sustained circadian clock, which is regulated (BMAL1, also known as ARNTL). These molecules form a tightly inter-
by interconnected negative feedback loops1. The circadian clock is twined heterodimer and bind to E-box sequences of the targeted pro-
primarily regulated by the transcription factors circadian locomotor moters to induce the transcription of many clock-controlled genes

Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of
1

Medicine, Zhejiang University, Hangzhou, China. 2Cancer Center, Zhejiang University, Hangzhou, China. 3Department of Breast Surgery, Harbin Medical
University Cancer Hospital, Harbin, China. 4NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin,
China. 5Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China. 6Harbin, China. 7The Affiliated Hospital of Qingdao
University and Qingdao Cancer Institute, Qingdao, China. 8Department of Cancer Biology, Department of Neuro-Oncology, The University of Texas MD
Anderson Cancer Center, Houston, TX, USA. 9Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean
University of China, Qingdao, China. 10Department of Pathology, Harbin Medical University, Harbin, China. 11Department of Clinical Laboratory, Cancer
Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy
of Sciences, Hangzhou, China. 12These authors contributed equally: Tong Liu, Zheng Wang, Leiguang Ye, Yuran Duan.  e-mail: zhiminlu@zju.edu.cn;
xudaqian@zju.edu.cn

Nature Cell Biology | Volume 25 | February 2023 | 273–284 273


Article https://doi.org/10.1038/s41556-022-01061-0

(CCGs), including period (encoding PER1/2/3) and cryptochrome To determine whether CK2α phosphorylates CLOCK, we per-
(encoding CRY1/2), during the day2,3. In turn, PER and CRY assemble formed an in vitro phosphorylation analysis by incubating purified
into a heterodimer and suppress CLOCK/BMAL1 activity during the GST-CK2α with purified His-CLOCK. Mass spectrometry analyses
night, thereby forming a negative feedback cycle4,5. In addition, the showed that CLOCK was phosphorylated at S106 (Extended Data
CLOCK–BMAL1 complex regulates the expression of the orphan nuclear Fig. 1f), which is evolutionarily conserved and in a phosphorylation
hormone receptors REV-ERBα/β, which in turn represses BMAL1, and consensus motif of CK2α (Extended Data Fig. 1g). This result was vali-
retinoic acid orphan receptors, which subsequently activate BMAL1, dated by mixing purified WT GST-CK2α or inactive GST-CK2α K68M
thus constituting a second feedback loop4,5. with WT His-CLOCK or His-CLOCK S106A in the presence of [γ-32P]
The functionality of the circadian clock in tumour cells was shown ATP, showing that only WT GST-CK2α phosphorylated WT His-CLOCK
to be impaired1. Bioinformatic analyses of 14 different cancer types but not His-CLOCK S106A (Fig. 1e). This phosphorylation was also
revealed that many core clock genes and CCGs were differentially detected using a specificity-validated anti-CLOCK pS106 antibody
expressed6. Experimental jet lag and genetic mutation of the cen- (Fig. 1e and Extended Data Fig. 1h–j). Consistent results showed that
tral circadian clock components BMAL1 and PER2 exacerbated lung IGF1 treatment of Huh7 cells induced phosphorylation of CLOCK S106
tumourigenesis7. In addition, mice lacking PER1 and PER2, CRY1 and and α-catenin S641, an established CK2α phosphorylation substrate14,
CRY2 or one copy of BMAL1 all showed increased spontaneous and which was abrogated by TBB treatment (Fig. 1f). In vitro GST pulldown
radiation-induced tumour development8,9. However, circadian genes experiments showed that purified His-BMAL1 bound to purified WT
may aggravate tumour progression in some instances. For instance, GST-CLOCK and GST-CLOCK S106A, whereas the presence of purified
depletion of CLOCK and BMAL1 impeded murine acute myeloid leukae- CK2α reduced the binding of His-BMAL1 to WT GST-CLOCK but not
mia cell growth and dampened glioblastoma stem cell proliferation and to GST-CLOCK S106A (Fig. 1g). In addition, CLOCK S106A expressed
brain tumour growth10,11. These findings suggest that the roles of clock in Huh7 cells was resistant to IGF1-induced dissociation of CLOCK
genes in different types of cancer are disparate and that the distinct from BMAL1 (Fig. 1h). These results indicate that IGF1-induced and
effects of core clock deficiency on tumour formation can result from CK2α-phosphorylated CLOCK S106 phosphorylation promotes the
disruption of not only their transcriptional functions but also gene disassembly of the CLOCK–BMAL1 complex.
expression-independent functions. However, whether CLOCK directly Intriguingly, immunofluorescence staining showed that CLOCK,
regulates cancer cell proliferation in response to oncogenic signalling which was originally localized in the nucleus, exhibited cytosolic accu-
in an expression- and canonical transcriptional function-independent mulation upon IGF1 treatment (Fig. 1i). These results were validated by
manner remains unclear. cell fractionation analyses (Extended Data Fig. 1k,l). Notably, expres-
In this Article, we revealed that activation of receptor tyrosine sion of CLOCK S106A (Fig. 1j), TBB treatment (Extended Data Fig. 1m)
kinases disrupted the CLOCK/BMAL1 complex and its transcriptional or the inactive CK2α K68M expression (Extended Data Fig. 1n) inhibited
activity by CK2-dependent CLOCK S106 phosphorylation. This phos- IGF1-induced cytosolic accumulation of CLOCK. These results indicate
phorylation resulted in Exportin1-mediated nuclear exportation of that IGF1-induced and CK2α-phosphorylated CLOCK S106 promotes
CLOCK, which acetylated PRPS1/2 at K29 in the cytosol and stabilized cytosolic CLOCK accumulation in HCC cells.
PRPS1/2 expression to promote de novo nucleotide synthesis and
hepatocellular carcinoma (HCC) cell proliferation. CLOCK pS106 binds to Exportin1 for nuclear exportation
Analyses of the peptide sequence adjacent to the S106 revealed a poten-
Results tial nuclear export signal (NES) (Extended Data Fig. 2a). Molecular
CLOCK S106 phosphorylation induces its nuclear exportation dynamic (MD) simulations show that CLOCK S106 phosphorylation
To determine whether CLOCK is regulated by oncogenic signalling, elicited dramatic conformation perturbation of the CLOCK–BMAL1
we treated Huh7 and Hep3B human HCC cells for 1 h with insulin-like complex (Extended Data Fig. 2b), suggesting that the S106 phospho-
growth factor 1 (IGF1), which activates IGF1 receptor (IGF1R) and is rylation introduces notable steric effects on the adjacent amino acids.
critical for HCC development12,13. Co-immunoprecipitation (IP) analyses Consistent results showed that the non-phosphorylated S106 and the
showed that IGF1 reduced CLOCK–BMAL1 heterodimerization (Fig. 1a). three adjacent Leu residues at the NES fragment of CLOCK were closely
Consistently, IGF1 treatment inhibited the transcriptional activity surrounded or covered by three highly flexible loops (loops 1, 2 and 3)
of CLOCK (Extended Data Fig. 1a) and reduced the expression of the of BMAL1 containing negatively charged D144, D231, D299 and E303,
CLOCK downstream genes Dbp and Nr1d1 in HCC cells (Fig. 1b). which are in close proximity to S106 (Fig. 2a, top, and Extended Data
Pre-treatment of Huh7 cells with MK-2206, SU6656, SP600125 Fig. 2c). CLOCK S106 phosphorylation flipped over these three loops
and TBB, which inhibited IGF1-induced activation of AKT, c-Src, to the other sides, resulting in the more solvent exposed NES frag-
c-Jun N-terminal kinase ( JNK) and CK2α, respectively (Extended ment (Fig. 2a, bottom). However, mutations of D144, D231, D299 and
Data Fig. 1b), showed that only the CK2α inhibitor TBB blocked E303 into Ala in BMAL1 (BMAL1 4Mut) conferred the resistance of the
IGF1-induced disruption of the CLOCK–BMAL1 complex (Fig. 1c). CLOCK–BMAL1 complex to CLOCK S106 phosphorylation-induced
A GST pulldown assay showed that purified bacteria-expressed conformational changes (Extended Data Fig. 2d). In addition, co-IP
GST-CLOCK interacted with purified bacteria-expressed His-CK2α analyses showed that BMAL1 4Mut was resistant to be dissociated from
(Fig. 1d). It was previously shown that ERK activated by growth fac- CLOCK upon IGF1 treatment (Fig. 2b). Consistently, a GST pulldown
tors phosphorylates CK2α at T360/S362, subsequently enhancing assay showed that purified His-BMAL1 4Mut, unlike its WT counter-
CK2α activity14. Treatment with the ERK inhibitor U0126 (Extended part, failed to dissociate from purified GST-CLOCK in the presence of
Data Fig. 1c), the expression of CK2α T360A/S362A mutant, or treat- CK2α (Extended Data Fig. 2e). These results strongly suggested that
ment with calf intestinal alkaline phosphatase (CIP) abrogated the phosphorylated and negatively charged S106 of CLOCK repels the
IGF1-induced interaction between CK2α and CLOCK (Extended negative charged amino acids in loops 1, 2 and 3 of BMAL1, leading to
Data Fig. 1d). Consistently, the presence of purified active His-ERK2 conformational change of both CLOCK and BMAL1, exposure of CLOCK
increased the binding of purified His-CLOCK to purified wild-type NES and consequently disruption of the interaction between CLOCK
(WT) GST-CK2α but not the GST-CK2α T360A/S362A mutant, and and BMAL1. In line with these results, mutation of this NES sequence
CIP treatment reduced the association between CK2α and CLOCK diminished IGF1-induced CLOCK-nuclear exportation, as shown by
(Extended Data Fig. 1e). These results indicate that IGF1-induced immunofluorescence (Fig. 2c) and immunoblotting (Extended Data
and ERK-mediated CK2α T360/S362 phosphorylation promotes Fig. 2f) analyses, further supporting that CLOCK S106 phosphoryla-
the binding of CK2α to CLOCK. tion exposes its NES for subsequent cytosolic translocation of CLOCK.

Nature Cell Biology | Volume 25 | February 2023 | 273–284 274


Article https://doi.org/10.1038/s41556-022-01061-0

a Huh7 Hep3B b c IGF1 – + + + + +


IP: IP: Hep3B MK-2206 – – + – – –
Huh7
SU6656 – – – + – –
G

G
CLOCK CLOCK Dbp Nr1d1
Ig

Ig
Dbp Nr1d1 SP600125 – – – – + –
IGF1 – – + – – + 1.5 1.5 1.5 1.5
* * ** * TBB – – – – – +

mRNA level

mRNA level
mRNA level
mRNA level
75

Relative

Relative
Relative
Relative
1.0 1.0 1.0 1.0
IP WB: BMAL1 75
IP:
100 0.5 0.5 0.5 0.5
CLOCK WB: BMAL1
WB: CLOCK 100
0 0 0 0
75 IGF1 – + IGF1 – + IGF1 – + IGF1 – + WB: CLOCK
Input WB: BMAL1 75
50 Input WB: BMAL1
WB: tubulin
50
WB: tubulin

d e f g
His-CLOCK S106A – + – + – TBB – – – – – – +
His-CK2α + + – GST-CLOCK WT – + – + –
His-CLOCK WT + – + – + IGF1 (min) 0 5 10 15 30 60 60
GST + – – GST-CLOCK S106A – – + – +
GST-CLOCK – + + GST-CK2α WT – – + + – 100 His-CK2α WT – – – + +
GST-CK2α K68M – – – – + WB: CLOCK pS106 His-BMAL1 + + + + +
37
pulldown

WB: CK2α 100 100 GST + – – – –


150
GST

32
P-CLOCK WB: CLOCK 75
100 100 WB: BMAL1

pulldown
WB: CLOCK pS106 WB: α-catenin pS641
25 100

GST
WB: GST 75 100 WB: CLOCK pS106
Input 150
37 WB: CK2α WB: α-catenin
WB: CK2α
100 50
WB: CLOCK WB: tubulin 25
WB: GST

h i j Flag-CLOCK WT S106A Input WB: BMAL1


75

Flag-CLOCK WT S106A
IGF1 – + IGF1 – + – +
IGF1 – + – + 37
100 WB: CK2α
75
Cytosolic distributive

40 WB: Flag (CLOCK)


WB: BMAL1 CLOCK ***
Cytosol
intensity (%)

IP: Flag 100


30 50
WB: tubulin
WB: CLOCK pS106 20
37
100 10
WB: CLOCK WB: PCNA
CLOCK
75 +DAPI 0 100
WB: BMAL1 IGF1 – + WB: Flag (CLOCK)
100 100
Input WB: α-catenin pS641 20 µm WCL WB: α-catenin pS641
100 100
WB: α-catenin WB: α-catenin
50 50
WB: tubulin WB: tubulin

Fig. 1 | CLOCK S106 phosphorylation induces its nuclear exportation. g, Bacterially purified GST-CLOCK WT or GST-CLOCK S106A on glutathione
a, Huh7 and Hep3B cells were treated with or without IGF1 (100 ng ml−1) for 1 h. agarose beads was incubated with or without His-CK2α in the presence of ATP
b, The mRNA expression levels of CLOCK downstream target genes were for an in vitro kinase assay. The beads were then washed with PBS five times
measured using quantitative PCR (n = 6). The data represent the mean ± s.d. and incubated with purified His-BMAL1 proteins. h, Flag-CLOCK WT or Flag-
of three independent experiments. *P < 0.001, **P < 0.0001 by two-tailed CLOCK S106A was transfected into Huh7 cells that were then stimulated with or
Student’s t-test. c, Huh7 cells were pre-treated with or without MK-2206 without IGF1 (100 ng ml−1) for 1 h. i, Huh7 cells were treated with or without IGF1
(10 μM), SU6656 (4 μM), SP600125 (25 μM) or TBB (10 μM) for 30 min before (100 ng ml−1) for 1 h. Top: immunofluorescence analyses were performed with
treatment with or without IGF1 (100 ng ml−1) for 1 h. d, A GST pulldown assay the indicated antibodies. Bottom: the cytosolic distribution intensity of CLOCK
was performed by mixing purified His-CK2α with purified GST or GST-CLOCK. is shown. Data are shown as mean ± s.d., n = 10. *P < 0.001, **P < 0.0001 by two-
Immunoblotting analyses were performed as indicated. e, In vitro kinase assays tailed Student’s t-test. j, Huh7 cells transfected with the indicated plasmids were
and autoradiography were performed by mixing purified WT GST-CK2α or GST- treated with or without IGF1 (100 ng ml−1) for 1 h. Cytosolic and whole-cell lysates
CK2α K68M protein with purified His-CLOCK WT or His-CLOCK S106A protein in were collected. All experiments were repeated three times independently with
the presence of [γ32P]-ATP. f, Huh7 cells pre-treated with or without TBB (10 μM) similar results. In a, c and e–h, IP and immunoblotting analyses were performed
for 30 min were treated with IGF1 (100 ng ml−1) for the indicated periods. with the indicated antibodies.

The NES of nuclear proteins is recognized by a nuclear export the presence of WT CK2α but not inactive His-CK2α K68M (Fig. 2e).
receptor, such as Exportin1 (also known as CRM1 or XPO1), Expor- Consistently, IGF1 treatment induced the binding of Exportin1 to
tin2 and Exportin3, which translocate proteins out of the nucleus15,16. reconstituted WT CLOCK, but not CLOCK S106A or the CLOCK NES
Co-IP analyses showed that IGF1 treatment induced the binding of mutant, in endogenous CLOCK-depleted Huh7 (Fig. 2f and Extended
Flag-Exportin1, but not Flag-Exportin2 or Flag-Exportin3, to CLOCK Data Fig. 2h) and Hep3B cells (Extended Data Fig. 2i). As expected,
(Fig. 2d), and this binding was inhibited by TBB treatment (Extended depletion of Exportin1 blocked IGF1-induced CLOCK-nuclear exporta-
Data Fig. 2g) or expression of BMAL1 4Mut (Fig. 2b). A GST pulldown tion in Huh7 cells (Fig. 2g). These results indicate that IGF1-induced and
assay showed that purified GST-Exportin1 bound to purified WT CK2α-mediated CLOCK S106 phosphorylation promotes the binding
His-CLOCK, but not CLOCK S106A or the His-CLOCK NES mutant, in of CLOCK to Exportin1 for nuclear exportation of CLOCK.

Nature Cell Biology | Volume 25 | February 2023 | 273–284 275


Article https://doi.org/10.1038/s41556-022-01061-0

a CLOCK non-phosphorylation b c Flag-rCLOCK


Flag-rBMAL1 WT + + + + – –
BMAL1 CLOCK Flag-rBMAL1 4Mut – – – – + + WT S106A NES mutant
BMAL1 shRNA + + + + + +
HA-CLOCK WT + + – – + +

Flag
HA-CLOCK S106A – – + + – –
IGF1 – + – + – +
Loop 3

+IGF1
S106 100
WB: Exportin1

Flag + DAPI
L118 75
L113 WB: Flag (rBMAL1)
IP: HA
Loop 1 L115
100
WB: CLOCK pS106 *** 20 µm
30 ***
100

Cytosolic distributive
Loop 2
WB: HA (CLOCK)

intensity (%)
20
100
WB: Exportin1
10
75
WB: BMAL1
Input 100 0
T A n t
WB: α-catenin pS641 Flag-rCLOCK W 06 ta
CLOCK S106 phosphorylation S1 mu
100 S
WB: α-catenin NE
BMAL1 CLOCK
50
d IgG IP: Flag
WB: tubulin Flag-Exportin1 + + + – – – –
Flag-Exportin2 – – – + + – –
Loop 3 Flag-Exportin3 – – – – – + +
pS106
IGF1 + – + – + – +
f Huh7 100

Flag-rCLOCK WT + + – – – – IP: Flag WB: CLOCK


Loop 1 L118
L113 Flag-rCLOCK S106A – – + + – – 100
Flag-rCLOCK NES mutant – – – – + + WB: Flag (Exportin)
L115 CLOCK shRNA + + + + + + 100
IGF1 – + – + – +
Input WB: CLOCK

Loop 2 100 50
WB: Exportin1 WB: tubulin
IP: Flag 100
WB: CLOCK pS106 g CRM1 shRNA – – + +
e His-CLOCK WT + + – – – – + 100 IGF1 – + – +
His-CLOCK S106A – – + + – – – WB: Flag (rCLOCK) 100
His-CLOCK NES mutant – – – – + + – 100
WB: CLOCK
GST-Exportin1 + + + + + + + WB: CLOCK Cytosol 50
His-CK2α WT – + – + – + – WB: tubulin
Cytosol 50
His-CK2α K68M – – – – – – + 37
WB: tubulin
WB: PCNA
pulldown

100 37
100
GST

WB: CLOCK WB: PCNA


WB: CLOCK
150
100 100
WB: GST (Exportin1) WB: Exportin1 WB: Exportin1
100 100 WCL 100
WB: CLOCK pS106 Input WB: α-catenin pS641 WB: α-catenin pS641
Input 100 100 100
WB: CLOCK WB: α-catenin WB: α-catenin

50 50
37
WB: CK2α WB: tubulin WB: tubulin

Fig. 2 | CLOCK pS106 binds to Exportin1 for nuclear exportation. a, The is shown (bottom). Data are shown as mean ± s.d., ***P < 0.0001 by one-way
conformation of the CLOCK–BMAL1 complex of the last frame extracted from ANOVA post hoc test. d, Huh7 cells transfected with the indicated plasmids
250 ns MD simulation. CLOCK structures with and without S106 phosphorylation were treated with or without IGF1 (100 ng ml−1) for 1 h. IP and immunoblotting
are shown in cyan and green, respectively. BMAL1 is shown in blue. Loop 1, analyses were performed with the indicated antibodies. e, An in vitro kinase assay
loop 2 and loop 3 of BMAL1 and the CLOCK NES residues, including L113, L115 was performed by mixing the indicated His-CLOCK and CK2α proteins in the
and L118, are shown as indicated. The red arrows indicate CLOCK S106 with and presence of ATP, followed by a GST pulldown assay by incubation with
without side-chain phosphorylation. b, Huh7 cells expressing BMAL1 GST-Exportin1 as indicated. f, Huh7 cells expressing CLOCK shRNA with
shRNA with reconstituted expression of the indicated BMAL1 proteins were reconstituted expression of Flag-rCLOCK WT, S106A or NES mutant protein
transfected with the indicated plasmids and treated with or without IGF1 were treated with or without IGF1 (100 ng ml−1) for 1 h. Cytosolic and whole-cell
(100 ng ml−1) for 1 h. IP and immunoblotting analyses were performed with the lysates were collected for IP and immunoblotting analyses as indicated. g, Huh7
indicated antibodies. c, Huh7 cells expressing CLOCK shRNA with reconstituted cells expressing CRM1 shRNA were treated with or without IGF1 (100 ng ml−1)
expression of Flag-rCLOCK WT, S106A or NES mutant protein were treated for 1 h. IP and immunoblotting analyses were performed with the indicated
with IGF1 (100 ng ml−1) for 1 h. Immunofluorescence analyses were performed antibodies. All experiments were repeated three times independently with
with the indicated antibodies. The cytosolic distribution intensity of CLOCK similar results.

Nature Cell Biology | Volume 25 | February 2023 | 273–284 276


Article https://doi.org/10.1038/s41556-022-01061-0

a Flag-rCLOCK WT + + – – – – b GST-CLOCK WT – + – + – + GST-CLOCK WT – + – + – +


Flag-rCLOCK NES mutant – – + + – – GST-CLOCK S106A – – + – + – GST-CLOCK S106A – – + – + –
Flag-rCLOCK S106A – – – – + + His-CK2α WT – – – + + + His-CK2α WT – – – + + +
CLOCK shRNA + + + + + + His-PRPS1 + + + + + + His-PRPS2 + + + + + +
IGF1 – + – + – + TBB – – – – – + TBB – – – – – +
37 37 37
WB: acetyl-lysine WB: acetyl-lysine
WB: PRPS1/2
37 37
IP: Flag 100
WB: PRPS1/2 WB: PRPS1/2
WB: CLOCK pS106
100 100
100 WB: CLOCK pS106 WB: CLOCK pS106
WB: Flag (rCLOCK)
100 100
37 WB: CLOCK WB: CLOCK
Input WB: PRPS1/2
37 37
50 WB: CK2α WB: CK2α
WB: tubulin

c d Huh7 Hep3B
Flag-PRPS1 WT + + – – Flag-PRPS2 WT + + – – C 1 2 1 C2
R- R-
C -C -
Flag-PRPS1 K29R – – + +Flag-PRPS2 K29R – – + + 9 9 9R 9R
PRPS1/2 WT K2 K2 WT K2 K2
IGF1 – + – + IGF1 – + – +
IGF1 – + – + – + – + – + – +
37 37
IP: Flag WB: PRPS1/2 K29ac IP: Flag WB: PRPS1/2 K29ac 37
WB: PRPS1/2 K29ac
37 37
37
WB: Flag (PRPS1) WB: Flag (PRPS2)
WB: PRPS1/2
100 100
WB: CLOCK pS106 100
WB: CLOCK pS106 WB: CLOCK pS106
Input 100 Input 100
100
WB: CLOCK WB: CLOCK WB: CLOCK
50 50 50
WB: tubulin WB: tubulin
WB: tubulin

e HA-PRPS1 + + + + HA-PRPS2 + + + +
f HA-PRPS1 + + + + + + HA-PRPS2 + + + + + +
Flag-rCLOCK WT + + – – Flag-rCLOCK WT + + – – Flag-rCLOCK WT + + – – – – Flag-rCLOCK WT + + – – – –
Flag-rCLOCK mutant A – – + – Flag-rCLOCK mutant A – – + – Flag-rCLOCK NES mutant – – + + – – Flag-rCLOCK NES mutant – – + + – –
Flag-rCLOCK mutant B – – – + Flag-rCLOCK mutant B – – – + Flag-rCLOCK S106A – – – – + + Flag-rCLOCK S106A – – – – + +
CLOCK shRNA + + + + CLOCK shRNA + + + + CLOCK shRNA + + + + + + CLOCK shRNA + + + + + +
IGF1 – + + + IGF1 – + + + IGF1 – + – + – + IGF1 – + – + – +
37 37 37 37
IP: HA WB: PRPS1/2 K29ac IP: HA WB: PRPS1/2 K29ac IP: HA WB: PRPS1/2 K29ac IP: HA WB: PRPS1/2 K29ac
37 37 37 37
WB: HA (PRPS1) WB: HA (PRPS2) WB: HA (PRPS1) WB: HA (PRPS2)
100 100 100 100
WB: CLOCK pS106 WB: CLOCK pS106 WB: CLOCK pS106 WB: CLOCK pS106
Input 100 Input 100 Input 100 Input 100
WB: Flag (rCLOCK) WB: Flag (rCLOCK) WB: Flag (rCLOCK) WB: Flag (rCLOCK)
50 50 50 50
WB: tubulin WB: tubulin WB: tubulin WB: tubulin

Fig. 3 | CLOCK acetylates PRPS1/2 K29 upon IGF1R activation. a, Huh7 cells and immunoblotting analyses were performed with the indicated antibodies.
expressing CLOCK shRNA with reconstituted expression of Flag-rCLOCK WT, d, Parental Huh7 and Hep3B cells and the indicated clones with knock-in
S106A or NES mutant protein were treated with or without IGF1 (100 ng ml−1) expression of PRPS1/2 K29R mutants were stimulated with or without IGF1
for 1 h. IP and immunoblotting analyses were performed with the indicated (100 ng ml−1) for 1 h. Immunoblotting analyses were performed with the
antibodies. b, Purified GST-CLOCK proteins on glutathione agarose beads were indicated antibodies. e,f, Huh7 cells expressing CLOCK shRNA with
incubated with or without His-CK2α and TBB (10 μM) in the presence of ATP for reconstituted expression of the indicated CLOCK proteins were transfected
an in vitro kinase assay. The beads were then washed with PBS (five times) and with HA-PRPS1/2 and treated with or without IGF1 (100 ng ml−1) for 1 h. IP using
incubated with or without the indicated His-PRPS1/2 proteins in the presence of limited amount of HA antibody and immunoblotting analyses were performed
acetyl-CoA. c, Huh7 cells transfected with the indicated plasmids were treated with the indicated antibodies. All experiments were repeated three times
with or without IGF1 (100 ng ml−1) for 1 h. IP using limited amount of Flag antibody independently with similar results.

CLOCK acetylates PRPS1/2 K29 upon IGF1R activation residue is K29 (Extended Data Fig. 3b), which is shared between PRPS1
To determine the cytosolic function of CLOCK, we performed mass spec- and PRPS2 and evolutionarily conserved (Extended Data Fig. 3c). IGF1
trometry analyses of CLOCK immunoprecipitates from IGF1-stimulated treatment of Huh7 cells for 1 h induced acetylation of WT Flag-PRPS1
Huh7 cells and revealed that CLOCK interacted with cytosolic phos- (Fig. 3c, left) and WT Flag-PRPS2 (Fig. 3c, right) but not Flag-PRPS1 K29R
phoribosyl pyrophosphate (PRPP) synthetase 1 (PRPS1) and PRPS2 or Flag-PRPS2 K29R, as detected by a specific anti-PRPS1/2 K29 acetyla-
(Extended Data Fig. 3a). PRPS catalyses the first and rate-limiting reac- tion antibody (Extended Data Fig. 3d,e). Consistently, knock-in expres-
tion for de novo nucleotide synthesis, converting ribose 5-phosphate to sion of PRPS1/2 K29R in HCC cells by clustered regularly interspaced
PRPP17. Co-IP analyses showed that IGF1 treatment of Huh7 cells induced short palindromic repeats (CRISPR)/Cas9 genome-editing technology
the binding of WT CLOCK, but not the CLOCK S106A or NES mutant, (Extended Data Fig. 3f–i) abrogated IGF1- (Fig. 3d) or overexpression of
to PRPS1/2, as detected by an antibody recognizing both PRPS1 and an activated IGF1R-CA mutant-induced (Extended Data Fig. 3j) PRPS1/2
PRPS2 (Fig. 3a). These results indicate that cytosolic CLOCK interacts K29 acetylation. As expected, catalytically inactive CLOCK P660A/
with PRPS1/2 upon IGF1 stimulation. Y662A/N663A (mutant A) and CLOCK G673A/S674A/V676A (mutant
CLOCK possesses intrinsic acetyltransferase activity that acety- B)18 were unable to acetylate PRPS1 or PRPS2 at K29 in vitro (Extended
lates histone, BMAL1 and argininosuccinate synthase 1 (ASS1)18–20. Data Fig. 3k), and reconstituted expression of the CLOCK (mutant
An in vitro acetylation assay showed that purified GST-CLOCK acety- A/B) mutants in endogenous CLOCK-depleted Huh7 cells (Extended
lated purified His-PRPS1 (Fig. 3b, left) and His-PRPS2 (Fig. 3b, right), as Data Fig. 3l) inhibited IGF1-induced K29 acetylation of PRPS1/2
detected by immunoblotting analyses with an anti-pan-acetylation anti- (Fig. 3e), although these mutants could still bind to PRPS1/2 (Extended
body. Mass spectrometry analyses showed that the acetylated PRPS1 Data Fig. 3m). In addition, reconstituted expression of CLOCK S106A

Nature Cell Biology | Volume 25 | February 2023 | 273–284 277


Article https://doi.org/10.1038/s41556-022-01061-0

or the His-CLOCK NES mutant, but not WT CLOCK, abrogated the K29 purified HSC70 (Fig. 4k). Notably, inclusion of CK2α-phosphorylated
acetylation of PRPS1/2 (Fig. 3f) upon IGF1 treatment. These results S protein-Flag-streptavidin-binding peptide (SFB)-tagged CLOCK
indicate that cytosolic CLOCK acetylates PRPS1/2 K29 in response to inhibited the binding of HSC70 to WT PRPS1/2 but not to PRPS1/2 K29R
IGF1R activation. (Fig. 4k). In addition, IGF1 treatment reduced the association of
HSC70 and LAMP2A with PRPS1/2 (Fig. 4l and Extended Data Fig. 4o),
CLOCK-induced PRPS1/2 acetylation inhibits their and this effect was largely abrogated by expression of PRPS1/2 K29R
degradation (Fig. 4l), CLOCK S106A, the CLOCK NES mutant and inactive CLOCK
A time-course experiment showed that IGF1 treatment increased the mutants (Extended Data Fig. 4o). These results indicate that IGF1R
PRPS1/2 K29 acetylation levels that corresponded with upregulated activation-induced and cytosolic CLOCK-mediated PRPS1/2 acetylation
PRPS1/2 expression levels (Fig. 4a,b). In addition, overexpression of inhibits the binding of HSC70 to PRPS1/2 and the subsequent CMA/
the IGF1R-CA mutant enhanced PRPS1/2 expression (Extended Data lysosomal-dependent PRPS1/2 degradation.
Fig. 4a). IGF1 treatment (Extended Data Fig. 4b) or IGF1R-CA expression
(Extended Data Fig. 4c) did not obviously alter the Prps1/2 mRNA expres- CLOCK-acetylated PRPS1/2 promote nucleotide synthesis
sion levels, suggesting a transcription-independent PRPS1/2 regulation. To determine the effect of CLOCK-mediated PRPS1/2 acetylation
Treatment of the IGF1R-CA-expressing HCC cells with cycloheximide on de novo nucleic acid synthesis, we incubated HCC cells with a
(CHX), a protein translation inhibitor that eliminates the effect of pro- limited amount of d-[6-14C] glucose. We found that IGF1 treatment
tein translation, showed that PRPS1/2 K29R had a much shorter half-life (Fig. 5a,b) or IGF1R-CA expression (Extended Data Fig. 5a,b) strongly
than its WT counterpart (Fig. 4c,d). Consistently, the expression of increased the production of glucose-derived 14C-RNA (Fig. 5a and
CLOCK S106A, the CLOCK NES mutant (Extended Data Fig. 4d,e), or the Extended Data Fig. 5a) and 14C-DNA (Fig. 5b and Extended Data Fig.
inactive CLOCK mutants (Extended Data Fig. 4f,g) accelerated PRPS1/2 5b). Reconstituted expression of CLOCK S106A, the CLOCK NES
turnover. These results indicate that CLOCK-mediated PRPS1/2 K29 mutant or the inactive CLOCK mutants (Fig. 5a,b and Extended Data
acetylation stabilizes PRPS1/2 expression. Fig. 5a,b) or knock-in expression of PRPS1/2 K29R (Fig. 5c,d and
We next treated HCC cells with the proteasome inhibitor MG132 or Extended Data Fig. 5c, d) inhibited IGF1- and IGF1R-CA-increased
14
PS341 or the lysosomal inhibitor chloroquine (CQ) or bafilomycin A1 C-RNA and 14C-DNA.
(BFA) and showed that CQ or BFA treatment enhanced PRPS1/2 expres- We next measured the incorporation of stable isotope-labelled
sion (Extended Data Fig. 4h), unveiling a role of lysosome in PRPS1/2 glucose (13C6-glucose) into purine and pyrimidine intermediates.
degradation. Analyses of the PRPS1/2 peptide sequence revealed that IGF1 treatment of Huh7 cells (Fig. 5e,f) or expression of IGF1R-CA in
30-VVTKK-34 is a consensus and evolutionarily conserved sequence these cells (Extended Data Fig. 5e,f) strongly increased the amount of
13
that can be recognized by heat shock cognate protein of 70 kDa C-labelled PRPP, purine (IMP, AMP and GMP) and pyrimidine (UMP
(HSC70) (Extended Data Fig. 4i), which mediates chaperone-mediated and CMP) intermediates; this increase was abrogated by reconsti-
autophagy (CMA) and subsequent lysosomal degradation21–23. Deple- tuted expression of CLOCK S106A, the CLOCK NES mutant or the inac-
tion of HSC70 (Fig. 4e) or lysosome-associated membrane protein-2A tive CLOCK mutants (Fig. 5e and Extended Data Fig. 5e) or knock-in
(LAMP2A) (Extended Data Fig. 4j), a receptor in the lysosomal mem- expression of PRPS1/2 K29R (Fig. 5f and Extended Data Fig. 5f). Given
brane for substrate proteins of CMA, enhanced PRPS1/2 expression in that PRPS1/2 K29 acetylation did not directly activate PRPS1/2 activ-
HCC cells, demonstrating a critical role of the CMA–lysosome pathway ity in vitro (Extended Data Fig. 5g), these results indicate that IGF1R
in PRPS1/2 degradation. activation-induced and cytosolic CLOCK-mediated PRPS1/2 stabiliza-
Co-IP analyses showed that HSC70 interacted with Flag-PRPS1/2 tion promotes de novo nucleotide synthesis.
but not the HSC70 binding motif-mutated Flag-PRPS1/2 V30A/V31A Similarly to IGF1 treatment, treatment of Huh7 cells with epidermal
mutant (Fig. 4f), which had a higher expression level and longer half-life growth factor (EGF) and fibroblast growth factor 1 (FGF1) enhanced
than its WT counterpart (Fig. 4g,h) in the cells with reconstituted CLOCK S106 phosphorylation, the cytosolic accumulation of CLOCK,
expression of Flag-PRPS1/2 (Extended Data Fig. 4k–n). In addition, and PRPS1/2 K29 acetylation (Extended Data Fig. 5h,i). Consequently,
the PRPS1/2 V30A/V31A mutation abrogated the effect of the PRPS1/2 treatment with EGF and FGF1 (Extended Data Fig. 5j) or expression
K29R mutation on PRPS1/2 degradation (Fig. 4i,j). Consistently, of activated EGFR-vIII and FGFR1 CA mutant (Extended Data Fig. 5k)
PRPS exhibited a longer half-life (~12 h) in the presence of IGF1R-CA enhanced PRPS1/2 expression. In addition, expression of the CLOCK
(Fig. 4d,j) than that in the absence of IGF1R-CA (~4.5 h) (Fig. 4h). S106A and NES mutant inhibited EGF- and FGF1-induced nucleus
These results indicate that PRPS1/2 K29 acetylation inhibits the bind- exportation of CLOCK and PRPS1/2 K29 acetylation (Extended Data
ing of HSC70 to the K29-adjacent consensus sequence of PRPS1/2 Fig. 5h,i) and decreased half-life of PRPS1/2 in HCC cells expressing
and subsequent PRPS1/2 degradation. To corroborate this finding, EGFR-vIII or FGFR1 CA (Extended Data Fig. 5l). These results indicate
we performed a GST pulldown assay and showed that both purified that activation of IGF1R, EGFR and FGFR1 shares the same mechanism
WT GST-PRPS1/2 and the GST-PRPS1/2 K29R mutant interacted with to regulate PRPS1/2.

Fig. 4 | CLOCK-induced PRPS1/2 acetylation inhibits their degradation. a,b, proteins were treated with CHX (100 μg ml−1) for the indicated periods of time.
Huh7 and Hep3B cells were stimulated with IGF1 (100 ng ml−1) for the indicated The quantification of PRPS1/2 protein levels relative to tubulin levels is shown.
time periods (a). The relative PRPS1/2 protein abundance was quantified by Data are the mean ± s.d., n = 6, **P < 0.001 by two-tailed Student’s t-test
densitometric analysis of the blots (n = 6). Data are the mean ± s.d. ***P < 0.0001 (h). ***P < 0.0001 by one-way ANOVA post hoc test (j). k, Purified SFB-CLOCK
by two-tailed Student’s t-test (b). c,d, Constitutively active IGF1R (IGF1R-CA)- proteins on streptavidin beads were incubated with or without His-CK2α in
expressing parental Huh7 and Hep3B cells and the indicated clones with knock-in the presence of ATP for an in vitro kinase assay. The washed beads were then
expression of PRPS1/2 K29R mutants were treated with CHX (100 μg ml−1) for incubated with or without the indicated GST-PRPS1/2 proteins in the presence of
the indicated periods of time (c). The quantification of PRPS1/2 protein levels acetyl-CoA. The beads were removed, and GST-PRPS1/2 was incubated with His-
relative to tubulin levels is shown. Data are the mean ± s.d., n = 6, **P < 0.001; *** HSC70 followed by a GST pulldown assay as indicated. l, Huh7 cells expressing
P < 0.0001 by one-way ANOVA post hoc test (d). e, Huh7 and Hep3B cells stably the indicated proteins were serum-starved for 12 h in the presence of lysosomal
transfected with HSC70 shRNA were collected. f, Huh7 cells expressing the inhibitor CQ (25 μM) and then treated with or without IGF1 (100 ng ml−1) for 1 h.
indicated proteins were collected for IP using limited amount of Flag antibody. All experiments were repeated three times independently with similar results. In
g–j, Huh7 cells expressing PRPS1/2 shRNA (g and h) and constitutively active a–c, g and i, IP and immunoblotting analyses were performed with the indicated
IGF1R (IGF1R-CA) (i and j) with reconstituted expression of the indicated PRPS1/2 antibodies.

Nature Cell Biology | Volume 25 | February 2023 | 273–284 278


Article https://doi.org/10.1038/s41556-022-01061-0

To examine the effect of IGF1R activation on other types of liver translocation of CLOCK, PRPS1/2 K29 acetylation (Extended Data
tumour, such as intrahepatic cholangiocarcinomas, we treated CCLP1 Fig. 6a,b) and glucose-derived amounts of 14C-RNA and 14C-DNA
or HuCCT1 intrahepatic cholangiocarcinoma cells with IGF1. IGF1 (Extended Data Fig. 6c). This induction was inhibited by expression of
treatment increased CLOCK S106 phosphorylation, the cytosolic the CLOCK S106A and NES mutant, which also decreased the half-life of

a b Huh7 Hep3B
c

protein abundance
Huh7 Hep3B Huh7 (IGF1R-CA)

protein abundance
4 4

Relative PRPS1/2
***

Relative PRPS1/2
IGF1 (h) 0 0.5 2 6 18 0 0.5 2 6 18 *** PRPS1/2 WT K29R-C1 K29R-C2
3 3
37 CHX (h) 0 3 6 12 0 3 6 12 0 3 6 12
WB: PRPS1/2 K29ac 2 2
37
37
WB: PRPS1/2 1 1 WB: PRPS1/2
50
50 0 0
WB: tubulin WB: tubulin
IGF1 (h) 0 0.5 2 6 18 0 0.5 2 6 18

d e PRPS1/2 WT
Hep3B (IGF1R-CA)
K29R-C1 K29R-C2
Huh7 (IGF1R-CA) Hep3B (IGF1R-CA)
PRPS1/2 WT PRPS1/2 WT Huh7 Hep3B CHX (h) 0 3 6 12 0 3 6 12 0 3 6 12
PRPS1/2 K29R-C1 PRPS1/2 K29R-C1 Mock + – – + – – 37
Ratio of initial protein

Ratio of initial protein

120 PRPS1/2 K29R-C2 120 PRPS1/2 K29R-C2 HSC70 shRNA1 – + – – + – WB: PRPS1/2
100 100 HSC70 shRNA2 – – + – – + 50
80 80
37 WB: tubulin
(%)

(%)

60 60 WB: PRPS1/2
40 40 *** 75
***
***
**

20 20 WB: HSC70
0 0 50
0 3 6 12 0 3 6 12 WB: tubulin
CHX chase time (h) CHX chase time (h)

f g
Flag-rPRPS1 WT V30A/V31A Flag-rPRPS2 WT V30A/V31A
CHX (h) 0 3 6 12 0 3 6 12 CHX (h) 0 3 6 12 0 3 6 12
l ag l ag 37
:F :F 37
IgG IP IgG IP WB: Flag (rPRPS1) WB: Flag (rPRPS2)
Flag-PRPS1 WT + + – Flag-PRPS2 WT + + – 50 50
Flag-PRPS1 V30A/V31A – – + Flag-PRPS2 V30A/V31A – – + WB: tubulin WB: tubulin
75 75
WB: HSC70 WB: HSC70 h Flag-rPRPS1 WT Flag-rPRPS2 WT
IP: Flag Flag-rPRPS1 V30A/V31A Flag-rPRPS2 V30A/V31A
100 IP: Flag 100

Ratio of initial protein


Ratio of initial protein

120 120
WB: LAMP2A WB: LAMP2A
37 37 100 100
WB: Flag (PRPS1) WB: Flag (PRPS2) 80 80

(%)
(%)

100 100 60 60
WB: LAMP2A WB: LAMP2A 40 40
** **
Input 75 Input 75 20 20
WB: HSC70 WB: HSC70 0 0
50 50 0 3 6 12 0 3 6 12
WB: tubulin WB: tubulin CHX chase time (h) CHX chase time (h)

i Huh7 (IGF1R-CA)
K29R/
j
Flag-rPRPS1 WT K29R V30A/V31A Huh7 (IGF1R-CA) Huh7 (IGF1R-CA)
CHX (h) 0 3 6 12 0 3 6 12 0 3 6 12
Flag-rPRPS1 WT Flag-rPRPS2 WT
37
Flag-rPRPS1 K29R Flag-rPRPS2 K29R
WB: PRPS1/2
Flag-rPRPS1 K29R/V30A/V31A Flag-rPRPS2 K29R/V30A/V31A
Ratio of initial protein

Ratio of initial protein

50 120 120
WB: tubulin 100
K29R/ 100
WT K29R 80
Flag-rPRPS2 V30A/V31A 80
(%)

(%)

60 60
CHX (h) 0 3 6 12 0 3 6 12 0 3 6 12
40 40
***
***

***
**

37 20
WB: PRPS1/2 20
0 0
50 0 3 6 12 0 3 6 12
WB: tubulin
CHX chase time (h) CHX chase time (h)

k His-CK2α WT – – + + His-CK2α WT – – + + l
SFB-CLOCK WT – + + + SFB-CLOCK WT – + + +
GST-PRPS1 WT + + + – GST-PRPS2 WT + + + –
GST-PRPS1 K29R – – – + GST-PRPS2 K29R – – – + Flag-PRPS1 WT + + – – Flag-PRPS2 WT + + – –
His-HSC70 + + + + His-HSC70 + + + + Flag-PRPS1 K29R – – + + Flag-PRPS2 K29R – – + +
75 75 IGF1 – + – + IGF1 – + – +
pulldown
pulldown

WB: HSC70 WB: HSC70 75 75


GST
GST

50 50 WB: HSC70 WB: HSC70


100
WB: PRPS1/2 K29ac WB: PRPS1/2 K29ac 100
IP: Flag WB: LAMP2A IP: Flag WB: LAMP2A
50 50 37
WB: GST (PRPS1) WB: GST (PRPS2) 37
WB: PRPS1/2 K29ac
75 75 WB: PRPS1/2 K29ac
37
WB: HSC70 WB: HSC70 37
WB: Flag (PRPS1) WB: Flag (PRPS2)
100 100 100
100
Input WB: CLOCK pS106 Input WB: CLOCK pS106 WB: LAMP2A
WB: LAMP2A
100 100 Input 75 Input 75
WB: Flag (SFB-CLOCK) WB: Flag (SFB-CLOCK) WB: HSC70 WB: HSC70
37 37 50 50
WB: CK2α WB: CK2α WB: tubulin WB: tubulin

Nature Cell Biology | Volume 25 | February 2023 | 273–284 279


Article https://doi.org/10.1038/s41556-022-01061-0

a Huh7 Hep3B b Huh7 Hep3B

** ** *
* * ** *
2.5 ** 3 ****
** 2.5 **
* **
* 2.5

DNA/total DNA

DNA/total DNA
RNA/total RNA

RNA/total RNA
radiolabelled

radiolabelled

radiolabelled

radiolabelled
2.0 2.0 2.0

Relative

Relative
2

Relative

Relative
1.5 1.5 1.5
1.0 1.0 1.0
1
0.5 0.5 0.5
0 0 0 0
Flag-rCLOCK WT + + – – – – + + – – – – Flag-rCLOCK WT + + – – – – + + – – – –
Flag-rCLOCK NES mutant – – + – – – – – + – – – Flag-rCLOCK NES mutant – – + – – – – – + – – –
Flag-rCLOCK S106A – – – + – – – – – + – – Flag-rCLOCK S106A – – – + – – – – – + – –
Flag-rCLOCK mutant A – – – – + – – – – – + – Flag-rCLOCK mutant A – – – – + – – – – – + –
Flag-rCLOCK mutant B – – – – – + – – – – – + Flag-rCLOCK mutant B – – – – – + – – – – – +
IGF1 – + + + + + – + + + + + IGF1 – + + + + + – + + + + +

c e 13
C-PRPP 13
C-IMP 13
C-AMP
Huh7 Hep3B * **
** ** **
** ** **

radiolabelled PRPP

radiolabelled AMP
3 3 ** 3.0 2.5 * 3.0 **

radiolabelled IMP
* * ***
RNA/total RNA

RNA/total RNA
radiolabelled

2.5 2.5
radiolabelled

2.0
2 2 2.0 2.0
Relative

Relative

Relative
Relative
Relative

1.5
1.5 1.5
1.0
1 1 1.0 1.0
0.5 0.5 0.5
0 0 0 0 0
PRPS1/2 WT + + – – + + – – Flag-rCLOCK WT + + – – – – + + – – – – + + – – – –
PRPS1/2 K29R – – + + – – + + Flag-rCLOCK NES mutant – – + – – – – – + – – – – – + – – –
IGF1 – + – + – + – + Flag-rCLOCK S106A – – – + – – – – – + – – – – – + – –
Flag-rCLOCK mutant A – – – – + – – – – – + – – – – – + –
Flag-rCLOCK mutant B – – – – – + – – – – – + – – – – – +
IGF1 – + + + + + – + + + + + – + + + + +

13 13 13
C-GMP C-UMP C-CMP
* ***
d Huh7 Hep3B
***
** *
* ***
**
*
*****

radiolabelled CMP
radiolabelled GMP

radiolabelled UMP
3 3 2.5 2.5 * 2.5
DNA/total DNA

DNA/total DNA

*
radiolabelled

radiolabelled

* 2.0 2.0 2.0


2 2

Relative
Relative
Relative

Relative

Relative

1.5 1.5 1.5


1.0 1.0 1.0
1 1
0.5 0.5 0.5
0 0 0 0 0
PRPS1/2 WT + + – – + + – – Flag-rCLOCK WT + + – – – – + + – – – – + + – – – –
PRPS1/2 K29R – – + + – – + + Flag-rCLOCK NES mutant – – + – – – – – + – – – – – + – – –
IGF1 – + – + – + – + Flag-rCLOCK S106A – – – + – – – – – + – – – – – + – –
Flag-rCLOCK mutant A – – – – + – – – – – + – – – – – + –
Flag-rCLOCK mutant B – – – – – + – – – – – + – – – – – +
IGF1 – + + + + + – + + + + + – + + + + +

f 13
C-PRPP 13
C-IMP 13
C-AMP 13
C-GMP 13
C-UMP 13
C-CMP
*
radiolabelled PRPP

radiolabelled AMP

3 3 3
radiolabelled GMP

radiolabelled CMP
radiolabelled UMP

3 3 3
radiolabelled IMP

** ***
** ** ***
2 2 2
Relative
Relative

Relative

2 2 2
Relative

Relative
Relative

1 1 1 1 1 1

0 0 0 0 0 0
PRPS1/2 WT + + – – + + – – + + – – + + – – + + – – + + – –
PRPS1/2 K29R – – + + – – + + – – + + – – + + – – + + – – + +
IGF1 – + – + – + – + – + – + – + – + – + – + – + – +

Fig. 5 | CLOCK-acetylated PRPS1/2 promote nucleotide synthesis. a,b, Huh7 were treated with or without IGF1 (100 ng ml−1) for 12 h, followed by metabolic
or Hep3B cells expressing CLOCK shRNA with reconstituted expression of labelling with 13C6-glucose (10 mmol l−1) for 30 min. The 13C-labelled metabolite
the indicated CLOCK proteins were treated with or without IGF1 (100 ng ml−1) intermediates of PRPP, IMP, AMP, GMP, UMP and CMP were measured by
for 12 h, followed by labelling with d-[6-14C] glucose (1 μCi, 0.01 mmol l−1) for LC–MS/MS. Data are the mean ± s.d., *P < 0.01; **P < 0.001; ***P < 0.0001 by
0.5 h. The amounts of 14C-RNA (a) and 14C-DNA (b) were measured. Data are the one-way ANOVA post hoc test. f, Parental Huh7 cells and the indicated clones
mean ± s.d., *P < 0.01; **P < 0.001 by one-way ANOVA post hoc test. c,d, Parental with knock-in expression of PRPS1/2 K29R mutants were treated with or without
Huh7 and Hep3B cells and the indicated clones with knock-in expression of IGF1 (100 ng ml−1) for 12 h, followed by metabolic labelling with 13C6-glucose
PRPS1/2 K29R mutants were treated with or without IGF1 (100 ng ml−1) for 12 h, (10 mmol l−1) for 30 min. The 13C-labelled metabolite intermediates of PRPP,
followed by labelling with d-[6-14C] glucose (1 μCi, 0.01 mmol l−1) for 0.5 h. The IMP, AMP, GMP, UMP and CMP were measured by LC–MS/MS. Data are the
amounts of 14C-RNA (c) and 14C-DNA (d) were measured. Data are the mean ± s.d., mean ± s.d., *P < 0.01; **P < 0.001; ***P < 0.0001 by one-way ANOVA post
*P < 0.01; **P < 0.001 by one-way ANOVA post hoc test. e, Huh7 cells expressing hoc test. All experiments were repeated three times independently with
CLOCK shRNA with reconstituted expression of the indicated CLOCK proteins similar results.

PRPS1/2 in HCC cells expressing IGF1R-CA (Extended Data Fig. 6d). These in L02 or THLE-2 normal hepatocytes upon IGF1 treatment, but at very
results suggest that IGF1 induces the same signalling transduction in moderate levels compared with HCC cells (Extended Data Fig. 6e). These
more than one cancer type. Notably, phosphorylation of α-catenin S641 results suggest that highly expressed IGF1, overexpressed or mutated
and CLOCK S106 and acetylation of PRPS1/2 at K29 were also increased IGF1R, highly activated ERK due to activated IGFR or other receptor

Nature Cell Biology | Volume 25 | February 2023 | 273–284 280


Article https://doi.org/10.1038/s41556-022-01061-0

a Huh7 Hep3B b Huh7 Hep3B


Flag-rCLOCK WT Flag-rCLOCK WT PRPS1/2 WT PRPS1/2 WT
Flag-rCLOCK S106A Flag-rCLOCK S106A PRPS1/2 K29R-C1 PRPS1/2 K29R-C1
Flag-rCLOCK NES mutant Flag-rCLOCK NES mutant PRPS1/2 K29R-C2 PRPS1/2 K29R-C2
60 60 60 60

Cell number
Cell number

Cell number
50 50 50 50

Cell number

***
***
***
***

**
***
***

**
(×104)
(×104)

(×104)
40 40 40 40

(×104)
30 30 30 30
20 20 20 20
10 10 10 10
0 1 2 3 0 1 2 3 0 1 2 3 0 1 2 3
Days Days Days Days

c d
Flag-rCLOCK WT S106A NES mutant PRPS1/2 WT K29R-C1 K29R-C2 *
* *

Tumour volume
1,500 1,500
*

Tumour volume
1,000

(mm3)
1,000

(mm3)
500 500

0 0
2 cm Flag-rCLOCK T A nt 2 cm PRPS1/2 T 1 2
W 106 uta W R-C -C
S m 2 9 9R
K K 2
ES
N
e Flag-rCLOCK WT S106A NES mutant f
CLOCK
pS106

PRPS1/2 WT K29R-C1 K29R-C2

PRPS1/2
K29ac
PRPS1/2
K29ac

PRPS1/2
PRPS1/2

50 µm

50 µm

Fig. 6 | CLOCK-acetylated PRPS1/2 promote HCC cell proliferation. a, Huh7 was examined 28 days after injection. The arrows indicate tumours. Tumour
and Hep3B cells expressing CLOCK shRNA with reconstituted expression of volumes were calculated (right). Data are the mean ± s.d., *P < 0.01 by one-way
the indicated CLOCK proteins were plated in complete culture medium for ANOVA post hoc test. d, Parental Huh7 cells and the indicated clones with
3 days. The cells were then collected and counted. Data are the mean ± s.d. n = 6, knock-in expression of PRPS1/2 K29R mutants were intrahepatically injected
***P < 0.0001 by one-way ANOVA post hoc test. b, Parental Huh7 and Hep3B cells into athymic nude mice (n = 6 per group). Tumour growth was examined 28 days
and the indicated clones with knock-in expression of PRPS1/2 K29R mutants were after injection. The arrows indicate tumours. Tumour volumes were calculated
plated in complete culture medium for 3 days. The cells were then collected and (right). Data are the mean ± s.d., *P < 0.01 by one-way ANOVA post hoc test.
counted. Data are the mean ± s.d., n = 6, **P < 0.001; ***P < 0.0001 by one-way e,f, IHC analyses of the indicated xenograft tumours from nude mice (n = 6)
ANOVA post hoc test. c, Huh7 cells expressing CLOCK shRNA with reconstituted were performed with the indicated antibodies. Representative staining images
expression of the indicated CLOCK proteins were intrahepatically injected into are shown. All experiments were repeated at least twice independently with
6-week-old male athymic BALB/c nude mice (n = 6 per group). Tumour growth similar results.

tyrosine kinase receptors, such as EGFR and FGFR, and overexpressed CLOCK S106A and the CLOCK NES mutant (Fig. 6a) or knock-in expres-
CK2α in cancer cells confer specific enhancement of the signalling in sion of PRPS1/2 K29R (Fig. 6b) inhibited the proliferation of HCC cells
cancer cells. Consistent results showed that CK2α expression levels expressing IGF1R. To differentiate the role of cytosolic CLOCK func-
were much higher in the HCC cells than in the normal hepatocytes tion from the transcriptional functions of CLOCK–BMAL1 complex in
(Extended Data Fig. 6e). As expected, overexpression of CK2α in L02 tumour cell proliferation, we depleted BMAL1 in HCC cells (Extended
cells increased CLOCK S106 phosphorylation and PRPS1/2 K29 acetyla- Data Fig. 7a). As expected, BMAL1 depletion inhibited the transcrip-
tion (Extended Data Fig. 6f) with correspondingly prolonged half-life tional activity of CLOCK (Extended Data Fig. 7b) and the expression
of PRPS1/2 (Extended Data Fig. 6g) upon IGF1 treatment. In contrast, of CLOCK–BMAL1 complex-regulated Dbp and Nr1d1 genes (Extended
CK2α depletion in Huh7 cells decreased IGF1-induced CLOCK S106 phos- Data Fig. 7c). Notably, these effects were recapitulated by expression
phorylation and PRPS1/2 K29 acetylation (Extended Data Fig. 6h) with of CLOCK S106D phosphorylation-mimicking mutant (Extended Data
correspondingly shortened PRPS1/2 half-life (Extended Data Fig. 6i). Fig. 7a–c), indicating that both BMAL1 depletion and CLOCK S106
These results indicate that CK2α, which is overexpressed in HCC cells, phosphorylation inhibit the transcriptional functions of CLOCK.
is critical for CLOCK S106 phosphorylation-enhanced PRPS1/2 stability. However, BMAL1 depletion did not induce CLOCK S106 phosphoryla-
tion (Extended Data Fig. 7d). Consistently, BMAL1 depletion, unlike
CLOCK-acetylated PRPS1/2 promote HCC cell proliferation CLOCK S106D expression, failed to increase the cytosolic translo-
Consistent with the effect of CLOCK-mediated PRPS1/2 acetylation on cation of CLOCK (Extended Data Fig. 7d), PRPS1/2 K29 acetylation
de novo nucleotide synthesis in HCC cells, reconstituted expression of (Extended Data Fig. 7d), PRPS1/2 stability (Extended Data Fig. 7e) and

Nature Cell Biology | Volume 25 | February 2023 | 273–284 281


Article https://doi.org/10.1038/s41556-022-01061-0

a Case 1 Case 2 c Tumour 1 Tumour 2 Tumour 3 Tumour 4


Non- tumour HCC Non-tumour HCC

pY1161
IGF1R
pY1161
IGF1R

α-Catenin
pS641
CLOCK
pS106

CLOCK
pS106
PRPS1/2
K29ac

PRPS1/2
K29ac
PRPS1/2

PRPS1/2
50 µm

b Non-tumour HCC
8 *** *** *** *** 50 µm e Low staining
Staining score

6 High staining
P < 0.01
4

Survival proportion
IGF1R pY1161
1.0
2
0.8
0 0.6
61 10
6
9a
c
1/
2 0.4
11 PS
pY pS K2
PR
0.2
F 1R CK 1 /2 0
IG O PS
CL PR 0 20 40 60 80
Survival time (months)
d R = 0.7730 R = 0.8213 R = 0.7970 Low staining
P < 0.0001 P < 0.0001 High staining
P < 0.0001
PRPS1/2 K29ac score

CLOCK pS106 score


CLOCK pS106 score

P < 0.01
8 8 8

Survival proportion
1.0 CLOCK pS106
6 6 6
0.8
4 4 4 0.6
2 2 2 0.4
0.2
0 0 0
0
0 2 4 6 8 0 2 4 6 8 0 2 4 6 8
0 20 40 60 80
IGF1R pY1161 score IGF1R pY1161 score α-Catenin pS641 score
Survival time (months)
Low staining
R = 0.8607 R = 0.7981 R = 0.8397 High staining
P < 0.0001 P < 0.0001 P < 0.0001 P < 0.01
PRPS1/2 K29ac score

Survival proportion

8 8 8 PRPS1/2 K29ac
1.0
PRPS1/2 score
PRPS1/2 score

6 6 6 0.8
4 4 4 0.6
0.4
2 2 2
0.2
0 0 0 0
0 2 4 6 8 0 2 4 6 8 0 2 4 6 8 0 20 40 60 80
CLOCK pS106 score PRPS1/2 K29ac score CLOCK pS106 score Survival time (months)

f
HSC70 Lamp2A

HSC70 Lamp2A
Ac
Limited PRPS1/2 PRPS1/2 Sufficient
nucleotide synthesis nucleotide synthesis
CLOCK

CMA-mediated CMA-mediated
degradation degradation

Exportin1

P Circadian-clock-
CK2 CLOCK related genes

Fig. 7 | CLOCK-acetylated PRPS1/2 predict human HCC aggressiveness. of human HCC samples with the indicated antibodies was scored, and correlation
a,b, IHC staining of human HCC and matched non-tumour tissue samples was analyses were performed. A Pearson correlation test was used (two-tailed). Note
performed with the indicated antibodies. Representative images of two cases that the scores for some samples overlap (d). e, Kaplan–Meier plots of the overall
are shown (a). The indicated staining scores for the indicated proteins in HCC survival rates in 90 patients with HCC grouped according to high (staining score,
and matched non-tumour liver samples (n = 30) were compared. Data are the 4–8) and low (staining score, 0–3) expression of IGFR1 pY1161, CLOCK pS106 and
mean ± s.d., *P < 0.0001 by Mann–Whitney U test compared with the non-tumour PRPS1/2 K29ac. P values were calculated using a log-rank test (two-tailed). f, A
adjacent tissue (b). c,d, Ninety human HCC samples were analysed with the schematic model showing nucleus-exported CLOCK acetylates PRPS to promote
indicated antibodies. Representative staining images are shown (c). IHC staining de novo nucleotide synthesis and tumour growth.

Nature Cell Biology | Volume 25 | February 2023 | 273–284 282


Article https://doi.org/10.1038/s41556-022-01061-0

glucose-derived amounts of 14C-RNA and 14C-DNA (Extended Data demonstrate here that activation of receptor tyrosine kinases promoted
Fig. 7f,g). Consequently, BMAL1 depletion promoted HCC cell prolifera- the binding of CK2α to CLOCK and the subsequent CK2α-mediated
tion to a lesser degree than did CLOCK S106D expression (Extended CLOCK S106 phosphorylation. This phosphorylation disassembled
Data Fig. 7h). These results indicate that both inhibition of canonical CLOCK–BMAL1 heterodimerization, leading to suppression of the
transcriptional activity of CLOCK and activation of its non-canonical CLOCK/BMAL1-mediated expression of the downstream genes in HCC
cytosolic function contribute to HCC cell proliferation. cells. Peptide sequence analyses of CLOCK revealed that an NES is adja-
Sorafenib treatment of HCC is a standard therapy24. Treatment cent to phosphorylated S106, and MD simulation analyses showed that
of HCC cells with sorafenib dose-dependently, but partially, inhibited S106 phosphorylation induced a conformational change in CLOCK and
IGF1-induced phosphorylation of ERK, α-catenin S641, and CLOCK S106 exposed its NES for binding to Exportin1, resulting in nuclear exporta-
and acetylation of PRPS1/2 K29 (Extended Data Fig. 7i). In addition, tion of CLOCK. In the cytosol, CLOCK bound and acetylated PRPS1/2 at
sorafenib treatment reduced PRPS1/2 stability in Huh7 cells express- K29, which is close to the binding consensus sequence (30-VVTKK-34)
ing IGF1R-CA (Extended Data Fig. 7j) with corresponding inhibition of of HSC70, thereby reducing the interaction of HSC70 with PRPS1/2 and
cell proliferation (Extended Data Fig. 7k). Notably, these effects were blocking HSC70-mediated CMA-lysosomal degradation of PRPS1/2.
reverted by CLOCK S106D expression (Extended Data Fig. 7i–k) which Stabilized PRPS1/2 promoted de novo nucleotide synthesis and HCC
by itself was sufficient to increase PRPS1/2 K29 acetylation (Extended cell proliferation and liver tumour growth in mice (Fig. 7f).
Data Fig. 7l), PRPS1/2 expression (Extended Data Fig. 7m) and HCC cell IGF1 expression is connected to circadian rhythms, and
proliferation (Extended Data Fig. 7n). These results suggested that the plasma IGF1 rhythms are disrupted with reduced IGF1 expression in
therapeutic effect of sorafenib on HCC could be attributed to the inhibi- Cry-deficient mice25. IGF1, in turn, entrains the circadian clock by upreg-
tion of the non-canonical function of CLOCK. However, these effects ulating PER expression through mTOR-promoted Per translation26. We
could be dampened due to overexpressed CK2α in tumour cells, which demonstrate that IGF1 elicited limited effect on CLOCK-dependent
bypasses the input from receptor protein kinase activity, compensates regulation of PRPS1/2 stability in normal cells, which was substantially
sorafenib-inhibited upstream signalling and phosphorylates CLOCK enhanced by CK2α expression that frequently occurs in tumour cells.
S106 to promote tumour cell proliferation. These findings suggest that IGF1-induced downstream signalling is
We next intrahepatically (Fig. 6c,d) and subcutaneously (Extended distinctly regulated in normal and tumour cells by cytosolic oncogenic
Data Fig. 8a,b) injected these cells into nude mice. Reconstituted proteins, leading to distinct impact on canonical and non-canonical
expression of CLOCK S106A and the CLOCK NES mutant (Fig. 6c functions of CLOCK.
and Extended Data Fig. 8a) or knock-in expression of PRPS1/2 K29R The connection of circadian rhythms to cancer has been inten-
(Fig. 6d and Extended Data Fig. 8b) inhibited orthotoptic and subcuta- sively studied by modulating core clock gene expression levels to
neous tumour growth with a corresponding decrease in Ki67 expres- intervene in the transcriptional activity of the CLOCK–BMAL1 complex1.
sion (Extended Data Fig. 8c,d) and increase in apoptosis (Extended However, a meta-analysis of studies assessing the clinicopathological
Data Fig. 8e,f). Immunohistochemistry (IHC) analyses of liver tumour and prognostic importance of the expression of PER1-3, CRY1-2, CLOCK
tissues showed that the expression of these mutants reduced the lev- and BMAL1 in 7,476 cases of different types of cancer revealed that low
els of PRPS1/2 K29 acetylation and PRPS1/2 expression (Fig. 6e,f). In expression of PER1 and PER2, but not CLOCK or others, could serve as
addition, CLOCK NES mutant expression resulted in the accumulation unfavourable indicators for cancer progression and poor overall sur-
of phosphorylated CLOCK S106 in the nucleus, whereas the CLOCK vival of some types of patients with cancer27. These findings suggested
S106A-expressing liver tumour tissues exhibited vanished staining of that the dysregulation of CLOCK at its transcriptional activity rather
CLOCK pS106 (Fig. 6e). In contrast to CLOCK S106A-elicited tumour than its expression is critical for tumour development. Here we revealed
growth inhibition, reconstituted expression of CLOCK S106D pro- a critical mechanism underlying oncogenic signalling-induced disrup-
moted tumour growth (Extended Data Fig. 8g,h). tion of the CLOCK–BMAL1 complex and its transcriptional activity in
HCC cells. Importantly, the finding of inhibited tumour growth by inhi-
CLOCK-acetylated PRPS1/2 predict human HCC bition of CLOCK-increased PRPS1/2 stability revealed the instrumental
aggressiveness role of the non-canonical and acetyltransferase functions of CLOCK in
To determine the clinical importance of CLOCK-upregulated PRPS1/2 tumour development. The positive correlation between CLOCK S106
expression, we analysed 90 human primary HCC specimens using IHC phosphorylation and PRPS1/2 K29 acetylation and poor survival of
staining with validated anti-IGF1R pY1161 to reflect the combinational patients with HCC highlighted the importance of CLOCK-mediated
inputs to IGF1R activities from overexpressed IGF1, IGF1R and oncogenic PRPS1 acetylation in the clinical aggressiveness of human HCC. Our
mutations of IGFR, anti-CLOCK pS106, anti-PRPS1/2 K29 acetylation, findings delineate a critical mechanism by which oncogenic signalling
and anti-PRPS1/2 antibodies, and the results showed that the levels disrupts CLOCK-regulated circadian rhythms and confers CLOCK with
of these protein markers were much higher in the HCC tissues than a moonlighting function in the promotion of de novo nucleotide syn-
in their adjacent normal tissues (Fig. 7a,b). In addition, the levels of thesis. These findings underscore the potential to target the cytosolic
IGF1R pY1161, α-catenin pS641, CLOCK pS106, PRPS1/2 K29 acetylation moonlighting function of CLOCK for human HCC treatment.
and PRPS1/2 expression were positively correlated with each other
(Fig. 7c,d). Notably, phosphorylation of IGF1R Y1161 and CLOCK S106 Online content
and PRPS1/2 K29 acetylation were positively associated with poor sur- Any methods, additional references, Nature Portfolio reporting sum-
vival of patients with HCC (Fig. 7e). These results indicate that IGF1R maries, source data, extended data, supplementary information,
activation-induced and cytosolic CLOCK-mediated PRPS1/2 acetylation acknowledgements, peer review information; details of author contri-
plays an instrumental role in the clinical aggressiveness of human HCC. butions and competing interests; and statements of data and code avail-
ability are available at https://doi.org/10.1038/s41556-022-01061-0.
Discussion
The transcriptional activity of CLOCK governs circadian rhythms, and References
impairment of the circadian clock has been linked to cancer develop- 1. Kinouchi, K. & Sassone-Corsi, P. Metabolic rivalry: circadian
ment1. However, whether the circadian clock is regulated by the activa- homeostasis and tumorigenesis. Nat. Rev. Cancer 20, 645–661
tion of receptor tyrosine kinases, which frequently occurs in cancer (2020).
and plays a direct role in the regulation of cancer cell proliferation in 2. Bass, J. & Takahashi, J. S. Circadian integration of metabolism and
a CLOCK transcriptional activity-independent manner, is unclear. We energetics. Science 330, 1349–1354 (2010).

Nature Cell Biology | Volume 25 | February 2023 | 273–284 283


Article https://doi.org/10.1038/s41556-022-01061-0

3. Sahar, S. & Sassone-Corsi, P. Metabolism and cancer: the 18. Doi, M., Hirayama, J. & Sassone-Corsi, P. Circadian regulator
circadian clock connection. Nat. Rev. Cancer 9, 886–896 (2009). CLOCK is a histone acetyltransferase. Cell 125, 497–508
4. Reinke, H. & Asher, G. Crosstalk between metabolism and (2006).
circadian clocks. Nat. Rev. Mol. Cell Biol. 20, 227–241 (2019). 19. Hirayama, J. et al. CLOCK-mediated acetylation of BMAL1 controls
5. Patke, A., Young, M. W. & Axelrod, S. Molecular mechanisms and circadian function. Nature 450, 1086–1090 (2007).
physiological importance of circadian rhythms. Nat. Rev. Mol. Cell 20. Lin, R. et al. CLOCK acetylates ASS1 to drive circadian rhythm of
Biol. 21, 67–84 (2020). ureagenesis. Mol. Cell 68, 198–209 e196 (2017).
6. Ye, Y. et al. The genomic landscape and pharmacogenomic 21. Liu, R. et al. Choline kinase alpha 2 acts as a protein
interactions of clock genes in cancer chronotherapy. Cell Syst. 6, kinase to promote lipolysis of lipid droplets. Mol. Cell 81,
314–328 e312 (2018). 2722–2735 e2729 (2021).
7. Papagiannakopoulos, T. et al. Circadian rhythm disruption 22. Cuervo, A. M. & Wong, E. Chaperone-mediated autophagy: roles
promotes lung tumorigenesis. Cell Metab. 24, 324–331 (2016). in disease and aging. Cell Res 24, 92–104 (2014).
8. Fu, L., Pelicano, H., Liu, J., Huang, P. & Lee, C. The circadian gene 23. Kaushik, S. & Cuervo, A. M. The coming of age of
Period2 plays an important role in tumor suppression and DNA chaperone-mediated autophagy. Nat. Rev. Mol. Cell Biol. 19,
damage response in vivo. Cell 111, 41–50 (2002). 365–381 (2018).
9. Lee, S., Donehower, L. A., Herron, A. J., Moore, D. D. & Fu, L. 24. Wong, K. M., King, G. G. & Harris, W. P. The treatment landscape
Disrupting circadian homeostasis of sympathetic signaling of advanced hepatocellular carcinoma. Curr. Oncol. Rep. 24,
promotes tumor development in mice. PLoS One 5, e10995 917–927 (2022).
(2010). 25. Chaudhari, A., Gupta, R., Patel, S., Velingkaar, N. & Kondratov, R.
10. Ruan, W., Yuan, X. & Eltzschig, H. K. Circadian rhythm as a Cryptochromes regulate IGF-1 production and signaling through
therapeutic target. Nat. Rev. Drug Discov. 20, 287–307 (2021). control of JAK2-dependent STAT5B phosphorylation. Mol. Biol.
11. Dong, Z. et al. Targeting glioblastoma stem cells through Cell 28, 834–842 (2017).
disruption of the circadian clock. Cancer Discov. 9, 1556–1573 26. Crosby, P. et al. Insulin/IGF-1 drives period synthesis to entrain
(2019). circadian rhythms with feeding time. Cell 177, 896–909 e820
12. Breuhahn, K. & Schirmacher, P. Reactivation of the insulin-like (2019).
growth factor-II signaling pathway in human hepatocellular 27. Zhang, J., Lv, H., Ji, M., Wang, Z. & Wu, W. Low circadian clock
carcinoma. World J. Gastroenterol. 14, 1690–1698 (2008). genes expression in cancers: a meta-analysis of its association
13. Xu, D. et al. The gluconeogenic enzyme PCK1 phosphorylates with clinicopathological features and prognosis. PLoS ONE 15,
INSIG1/2 for lipogenesis. Nature 580, 530–535 (2020). e0233508 (2020).
14. Ji, H. et al. EGF-induced ERK activation promotes CK2-mediated
disassociation of α-Catenin from β-Catenin and transactivation of Publisher’s note Springer Nature remains neutral with regard to
β-Catenin. Mol. Cell. 36, 547–559 (2009). jurisdictional claims in published maps and institutional affiliations.
15. Paci, G., Caria, J. & Lemke, E. A. Cargo transport through the
nuclear pore complex at a glance. J. Cell Sci. https://doi.org/ Springer Nature or its licensor (e.g. a society or other partner) holds
10.1242/jcs.247874 (2021). exclusive rights to this article under a publishing agreement with
16. Elfgang, C. et al. Evidence for specific nucleocytoplasmic the author(s) or other rightsholder(s); author self-archiving of the
transport pathways used by leucine-rich nuclear export signals. accepted manuscript version of this article is solely governed by the
Proc. Natl Acad. Sci. USA 96, 6229–6234 (1999). terms of such publishing agreement and applicable law.
17. Ben-Sahra, I., Howell, J. J., Asara, J. M. & Manning, B. D.
Stimulation of de novo pyrimidine synthesis by growth signaling © The Author(s), under exclusive licence to Springer Nature Limited
through mTOR and S6K1. Science 339, 1323–1328 (2013). 2023

Nature Cell Biology | Volume 25 | February 2023 | 273–284 284


Article https://doi.org/10.1038/s41556-022-01061-0

Methods TCAAGACTGCAGTGCAGAT-3′; LAMP2A shRNA2: 5′-GCAGTGCAG


This research complies with all relevant ethical regulations of The ATGACGACAACT-3′. BMAL1 shRNA1: 5′-CTTCTAGGCACATC
First Affiliated Hospital, Zhejiang University School of Medicine and GTGTTAT-3′; BMAL1 shRNA2: 5′-GCAGAATGTCATAGGCAAGTT-3′.
Qingdao Cancer Institute, including the Institutional Review Board CK2α shRNA1: 5′-GCACAGAAAGCTACGACTAAT-3′; CK2α shRNA2:
and Institutional Animal Care and Use Committee. 5′-TGGACAAACTGCTGCGATATG-3′. HA-EGFR-vIII was kindly provided
by Dr. Xinjian Li from CAS Key Laboratory of Infection and Immunity,
Materials CAS Center for Excellence in Biomacromolecules, Institute of Bio-
Antibodies against HA (ab18181) (for IP), c-SRC (ab16885), c-Src pY418 physics, Chinese Academy of Sciences, Beijing, China. The expression
(ab4816), CK2α pT360/S362 (ab119410), CLOCK (ab3517) (for IHC), plasmid for the Per1-driven luciferase reporter was kindly provided by
HSC70 (ab154415), IGF1R pY1161 (ab39398) and LAMP2A (ab18528) were Dr Yi Liu (School of Basic Medical Sciences, Fudan University, China).
purchased from Abcam. Antibodies against CLOCK (#5157) (for western
blot (WB)), Exportin1 (#46249), AKT (#4685), AKT pS473 (#4060), acet- Cell lines and cell culture conditions
ylated lysine (#9441), HA (#3724) (for WB), ERK1/2 (#4695), pERK1/2 Hep3B, THLE-2 and 293T were obtained from ATCC. Huh7, L02, CCLP1
(#4370), c-Jun (#9165) and c-Jun pS73 (#3270) were purchased from Cell and HuCCT1 were purchased from the Type Culture Collection of the
Signaling Technology. Anti-PCNA (#610665) antibody was obtained Chinese Academy of Sciences, Shanghai, China. Huh7, Hep3B, 293T and
from BD Biosciences. BMAL1 (NBP2-02544) antibody was purchased L02 were maintained in Dulbecco’s modified Eagle medium (DMEM)
from Novus Biologicals. Normal mouse IgG (sc-2025), normal rabbit IgG supplemented with 10% bovine calf serum (HyClone). THLE-2 was
(sc-2027), GST (sc-138), CK2α (sc373894) (for WB), PRPS1/2 (sc-376440) cultured in BEGM from Lonza, supplied with 10% foetal bovine serum
(for IHC), PRPS1/2 (sc-100822) (for IP and WB), and tubulin (sc-8035) in pre-coated dish. CCLP1 and HuCCT1 were cultured in RPMI1640 com-
antibodies were obtained from Santa Cruz Biotechnology. [γ-32P] ATP plete medium supplemented with 10% bovine calf serum (HyClone).
was obtained from PerkinElmer. Rabbit anti-Ki67 antibody (AB9260) No cell lines used in this study were found in the database of commonly
was obtained from Millipore. TBB, SP600125, sorafenib, U0126, misidentified cell lines that is maintained by the International Cell Line
SU6656, CHX, CQ, PS341, MG132, BFA, EGF, ATP, acetyl co-enzyme A, Authentication Committee and the BioSample database maintained
streptavidin magnetic beads, anti-Flag M2 agarose beads, EDTA-free by the NCBI. Glucose deprivation was carried out by washing the cells
protease inhibitor cocktail, 3× Flag peptides, mouse anti-Flag (F1804), twice with phosphate-buffered saline (PBS) and then culturing the
rabbit anti-Flag (F7425) and anti-His (SAB1305538) antibodies were pur- cells in glucose-free DMEM (Gibco). The transfection procedure was
chased from Sigma-Aldrich. MK-2206, hygromycin (400053), puromy- performed using PolyJet (SignaGen) or Lipofectamine 2000 (Invitro-
cin (540222) and G418 (345810) were purchased from EMD Biosciences. gen) according to the manufacturers’ instructions29.
FGF1 was purchased from R&D Systems. Antibodies against α-catenin
pS641 (GTX79145), α-catenin (GTX111095) and CLOCK (GTX134464) IP and immunoblotting analysis
(for immunofluorescence) were obtained from Genetex. CLOCK anti- The extraction of proteins from cultured cells was performed with a
body (A302-617A) (for IP), CK2α (A300-198A) (for WB and IP), glu- modified buffer and was followed by IP and immunoblotting using
tathione agarose, DAPI, Alexa Fluor 488 goat anti-rabbit (A11008), Alexa corresponding antibodies as described previously30. Immunoblot-
Fluor 594 goat anti-rabbit (A11012), Alexa Fluor 488 goat anti-mouse ting intensity was quantified by ImageJ (National Institutes of Health).
(A11029) and Alexa Fluor 594 goat anti-mouse antibodies (A11005) were
obtained from Thermo Fisher Scientific. Primary antibodies were used Measurement of PRPS activity
at a dilution of 1:1,000. Secondary HRP-coupled antibodies were used PRPS activity was measured as previously reported31,32. Briefly, different
at 1:5,000.Ni-NTA agarose was obtained from Qiagen. CLOCK pS106 purified PRPS1/2 (10 ng) proteins were incubated in 100 μl of reaction
blocking peptide (WKPTFL-pS-NEEFTQ) and PRPS1/2 K29ac blocking buffer (110 mM NaH2PO4, pH 7.0, 4.7 mM ribose 5-phosphate, 0.4 mM
peptide (LGLELG-Kac-VVTKKF) were synthesized by Selleck-Chem. NADH, 3.2 mM ATP, 1.8 mM phosphoenolpyruvate, 6 mM MgCl2, 31 mM
CLOCK pS106- and PRPS1/2 K29ac-specific antibodies were generated NaHCO3, 7 U of pyruvate kinase, 10 U of lactic dehydrogenase and 10 U
by Signalway Biotechnology. of myokinase) at 25 °C in a 96-well plate. The absorbance was read at
340 nm in kinetic mode for 5 min using multi-detection microplate
DNA construction and mutagenesis readers (BMG Labtech). Myokinase, pyruvate kinase, lactic dehydro-
PCR-amplified human CLOCK, PRPS1, PRPS1, CK2α, HSC70, BMAL1, genase and phosphoenolpyruvate were excessive in the system to
ERK2, IGF1R, FGFR1, EGFR, Exportin1, Exportin2, Exportin3 and LAMP2A ensure complete consumption of AMP generated by the PRPS pro-
were cloned into pcDNA3.1/hygro(+)-Flag, pcDNA3.1/hygro(+)-HA, tein. The amount of PRPS protein in the immunoprecipitates or elu-
pcDNA3.1/hygro(+)-Myc, twin Flag-streptavidin, PGEX4T-1 (GST) or ates was quantified by immunoblot using purified His-PRPS protein
pColdI vectors, as previously described28. The following mutations as a standard.
were generated using a QuikChange site-directed mutagenesis kit
(Stratagene): PRPS1/2 K29R, V30A/V31A, and K29R/V30A/V31; CLOCK GST pulldown assay
S106A, CLOCK S106D, NES mutant (L113A/L115A/L118A), mutant A Equal amounts of His-tagged purified protein (200 ng per sample) were
(P660A/Y662A/N663A) and mutant B (G673A/S674A/V676A); Ck2α incubated with 100 ng of GST fusion proteins together with glutathione
K68M and T360/S362A, IGF1R (V922E) (IGF1R-CA), Bmal1 4Mut (D144A/ agarose beads in modified binding buffer (50 mM Tris–HCl, pH 7.5; 1%
D231A/D299A/E303A), DFGFR1 (K567E) (FGFR1-CA), and short hairpin Triton X-100; 150 mM NaCl; 1 mM dithiothreitol (DTT); 0.5 mM EDTA;
RNA (shRNA)-resistant CLOCK constructs containing T723C, G726C, 100 μM phenylmethylsulfonyl fluoride; 100 μM leupeptin; 1 μM apro-
A729G and T732C; shRNA-resistant PRPS1 constructs containing T567C, tinin; 100 μM sodium orthovanadate; 100 μM sodium pyrophosphate;
C570A and G573A; shRNA-resistant PRPS2 constructs containing and 1 mM sodium fluoride). The glutathione agarose beads were then
C285A, A288C and A291G; shRNA-resistant BMAL1 constructs contain- washed four times with binding buffer and subjected to immunoblot-
ing C1110A, A1113C and G1116A. The PLKO.1 shRNA target sequences ting analysis as previously described33.
were as follows: CLOCK, 5′-CGCACACATAGGCCATCTTAT-3′; PRPS1,
5′-TGGACTTTGCCTTGATTCACA-3′; PRPS2, 5′-CCATACGCCCGACA Purification of recombinant proteins
AGATAAA-3′; and Exportin1 (CRM1), 5′-CCTGCTTTCAAGGAACAT GST-CLOCK WT, GST-CLOCK S106A, GST-CLOCK NES mutant
TTA-3′. HSC70 shRNA1: 5′-CCAAGACTTCTTCAATGGAAA-3′; HSC70 (L113A/L115A/L118A), GST-CLOCK mutant A (P660A/Y662A/N663A),
shRNA2: 5′-GCAACTGTTGAAGATGAGAAA-3′; LAMP2A shRNA1: 5′-GC GST-CLOCK mutant B (G673A/S674A/V676A), GST-Exportin1,

Nature Cell Biology


Article https://doi.org/10.1038/s41556-022-01061-0

GST-PRPS1/2 WT, GST-PRPS1/2 K29R, GST-CK2α WT, GST-CK2α K68M, extracted using 90/9/1 (v/v/v) acetonitrile/water/formic acid. Samples
GST-CK2α T360/S362A, His-PRPS1/2 WT, His-PRPS1/2 K29R, His-CLOCK were centrifuged at 17,000g for 5 min at 4 °C, and supernatants were
WT, His-CLOCK S106A, His-CLOCK NES mutant, His-ERK2, transferred to clean tubes, followed by evaporation to dryness using
His-CK2α WT, His-CK2α K68M, His-BMAL1 and His-HSC70 were nitrogen. Samples were reconstituted in 0.2% ammonium hydroxide
expressed in bacteria and purified as previously described 34. in ammonium acetate (10 mmol l−1), and then, 10 μl was injected into a
S-protein-Flag-streptavidin-binding peptide (SFB)-tagged CLOCK Thermo Scientific Vanquish liquid chromatography system containing
and the indicated mutants expressed in 293T cells were enriched by a ThermoHypercarb 100 × 3 mm 3 μm HPLC column heated to 35 °C
anti-Flag agarose beads and eluted with 3× Flag peptide. After ultra- with mobile phase A (MPA) consisting of 0.2% ammonium hydroxide
filtration and removal of the 3× Flag peptide from the eluted protein, in ammonium acetate (10 mmol l−1) and mobile phase B (MPB) consist-
a secondary streptavidin pulldown assay was performed. ing of 0.2% ammonium hydroxide in acetonitrile. With a flow rate of
0.3 ml min−1, the gradient elution programme was 0 min (0% MPB)–
CRISPR–Cas9-mediated genome editing 2.0 min (0% MPB)–15.0 min (30% MPB)–15.1 min (95% MPB)–20.0 min
Genomic mutations were introduced into cells using the CRISPR–Cas9 (95% MPB)–20.1 min (0% MPB)–25.0 min (STOP). Data were acquired
system, as described previously35. Single-guide RNAs (sgRNAs) were using a Thermo Orbitrap Fusion Tribrid Mass Spectrometer in selected
designed to target the genomic area adjacent to mutation sites in ion mode (SIM) electrospray positive mode. Peak integration and area
PRPS1/2 K29R using the CRISPR design tool (http://crispr.mit.edu/). The calculation were performed by using Thermo TraceFinder software.
annealed guide RNA oligonucleotides were inserted into a PX458 vector
(Addgene) digested with the BbsI restriction enzyme. Cells were seeded In vitro kinase assay
at 60% confluence, followed by co-transfection of sgRNAs (0.5 μg) An in vitro kinase assay was performed as previously described with
and single-stranded donor oligonucleotides (10 pmol) as a template modifications36. Briefly, purified CK2α was incubated with different
to introduce mutations. Twenty-four hours after transfection, cells bacterially purified His or GST-CLOCK (200 ng) proteins in 25 μl of
were trypsinized, diluted for single cells and seeded into 96-well plates. kinase buffer (50 mM Tris–HCl, pH 7.5; 100 mM KCl; 50 mM MgCl2; 1 mM
Genomic DNA was extracted from GFP-positive cells, followed by Na3VO4; 1 mM DTT; 5% glycerol; 0.5 mM ATP; and 10 μCi [γ-32P] ATP) at
sequencing of the PCR products spanning the mutation sites. sgRNA tar- 25 °C for 1 h. The reaction was terminated by adding SDS–PAGE loading
geting sequence for Prps1 (K29R): 5′-GGGCCTGGAGCTAGGCAAGG-3′; buffer and heating to 100 °C for 5 min. The reaction mixture was then
single-stranded donor oligonucleotide (ssODN) sequence for subjected to SDS–PAGE or autoradiography analysis.
PRPS1 (K29R): 5′- ATGCCGAATATCAAAATCTTCAGCGGCAGCTCCC
ACCAGGACTTATCTCAGAAAATTGCTGACCGCCTGGGCCTGGA In vitro acetylation assay
G C TAG G C AGAGTAGTGAC TA AGA A AT TC AG C A ACC AG GAG An in vitro acetylation reaction was performed as described previ-
ACCTGGTAAGGACCAAGTTGGGACCCTGACTAGACCTCACCTGC ously37. In brief, purified GST-CLOCK proteins on glutathione agarose
CCAGGC-3′; sgRNA targeting sequence for PRPS2 (K29R): beads were incubated with or without His-CK2α in the presence of ATP
5′-GGGCCTGGAGCTGGGCAAGG -3′; ssODN sequence for PRPS2 for an in vitro kinase assay. The beads were then washed with PBS five
(K29R): 5′- CCATGCCCAACATCGTGCTGTTCAGCGGCAGCTCG times and incubated with or without the indicated His-PRPS1/2 in 30 μl
CATCAGGACCTGTCCCAGCGCGTGGCCGACCGCCTGGGCCTG of reaction buffer (20 mM Tris–HCl (pH 8.0), 20% glycerol, 100 mM KCl,
GAGCTGGGCAGAGTAGTCACGAAGAAGTTCAGCAACCAGGAGACCAG 1 mM DTT, 0.2 mM EDTA, 10 mM Trichostatin A, 10 mM nicotinamide
GTGGGGGCCGCGCCGGCGGCCGGGCTAGGGAGAGCGCTGGGCAC-3′. and 100 mM acetyl-CoA) at 30 °C for 1 h. The reaction was terminated
The lowercase letters in the ssODN sequences indicate the mutated by adding SDS–PAGE loading buffer. The samples were then subjected
nucleotides that will replace the endogenous nucleotides in the to immunoblotting analyses.
genomic DNA of parental cells using the CRISPR–Cas9 system.
Genotyping was performed by sequencing PCR products ampli- Per1-driven luciferase reporter assay
fied from the following primers: Prps1 forward: 5′-CGGGAATGTA Luciferase reporter activity in cell lysates was measured as previ-
AGATGGCGGA-3′; Prps1 reverse: 5′-TCAGTACGCAGGTAGGCTCT-3′; ously described38. In brief, Huh7 cells were transfected with 0.1 μg
Prps2 forward: 5′-CTTCCCTACATCTAGCCGCC-3′; Prps2 reverse: of Per1-driven luciferase reporter and 0.075 μg of β-galactosidase.
5′- GGGGGAAACCTTTAGTCGCA-3′. Twenty-four hours later, the cells were stimulated with or without
IGF1 (100 ng ml−1) for 12 h. Cell lysates were measured for the activity
Influx of glucose into nucleic acids of luciferase and β-galactosidase according to the manufacturer’s
An assay of the influx of glucose to nucleic acids was performed as previ- instructions (Promega).
ously described with modifications. In brief, Huh7 or Hep3B cells were
seeded in a six-well plate and incubated in 2 ml of fresh medium spiked Apoptosis assay
with 1 μCi of d-[6-14C] glucose (0.01 mmol l−1) for 30 min, followed by For a TUNEL assay, tumour tissues were sectioned at 5 μm thickness.
washing with ice-cold PBS to remove extra medium components. RNA Apoptotic cells were counted using a Dead-End colourimetric TUNEL
or DNA was extracted, and 14C-RNA or 14C-DNA was assayed using a system (Promega) according to the manufacturer’s instructions.
scintillation counter.
Cell proliferation assay
Analysis of intermediate metabolites by LC–MS/MS A total of 2 × 105 cells suspended in 2 ml of medium were seeded in
Analysis of intermediate metabolites by liquid chromatography with six-well plates and were maintained in maintained in complete medium.
tandem mass spectrometry (LC–MS/MS) was performed as previously The cells in each well were trypsinized and counted every day after
described32. Briefly, for analysis of the incorporation of glucose carbon seeding.
(13C6-glucose) into intracellular nucleotide metabolites, extracts were
prepared and analysed by high-resolution mass spectrometry. Cells at Mass spectrometry analyses
approximately 80% confluence were seeded in 10 cm dishes in tripli- For identification of interacting proteins, a protein band visualized
cate. For glucose labelling, cells were washed with glucose-free DMEM via Coomassie blue staining was excised from an SDS–PAGE gel and
and incubated in fresh medium containing 13C6-glucose (10 mmol l−1) digested in gel in 50 mM ammonium bicarbonate buffer containing
for 30 min. Then, the cells were quickly washed with ice-cold PBS to RapiGest (Waters Corporation) overnight at 37 °C with 200 ng of
remove extra medium components. Nucleotide metabolites were modified sequencing-grade trypsin (Promega). The digested protein

Nature Cell Biology


Article https://doi.org/10.1038/s41556-022-01061-0

samples were analysed using high-sensitivity LC–MS/MS with an with the pressure coupling set at 1 atm and a constant temperature of
Orbitrap Elite mass spectrometer (Thermo Fisher Scientific). Proteins 300 K were performed for the CLOCK/BMAL1 system with and without
were identified by searching the fragment spectra against the UniProt S106 phosphorylation. MD trajectories were analysed using VMD
protein database (EMBL-EBI) using the Mascot search engine (v.2.3; (http://www.ks.uiuc.edu/), and the molecules were drawn using PyMOL
Matrix Science) with the Proteome Discoverer software program (v.1.4; (https://pymol.org/2/)).
Thermo Fisher Scientific). For detection of phosphorylation sites,
in vitro phosphorylation of CLOCK by CK2α was performed according IHC analysis and histological evaluation of human HCC
to the in vitro kinase assay protocol described above. For detection of specimens
acetylation sites, purified GST-CLOCK proteins on glutathione agarose Human HCC and adjacent matched non-tumour tissue samples were
beads were incubated with or without His-CK2α in the presence of obtained from The First Affiliated Hospital, Zhejiang University School
ATP for an in vitro kinase assay. The beads were then washed with PBS of Medicine and The Affiliated Hospital of Qingdao University. The use
five times and incubated with or without the indicated His-PRPS1/2 of human HCC samples and the relevant database was approved by the
proteins in the presence of acetyl-CoA. Then, the protein samples Research Ethics Committee of The First Affiliated Hospital, Zhejiang
were digested using trypsin or GluC and analysed using LC–MS/MS University School of Medicine and The Affiliated Hospital of Qingdao
with an Orbitrap Elite mass spectrometer as described previously34. University, complied with all relevant ethical regulations. All tissue
Mass spectrometric analysis of a tryptic fragment at m/z 887.35431 Da samples were collected in compliance with the informed consent
(−0.33 mmu/−0.37 ppm), which was matched with the +1 charged policy. Non-tumour and HCC samples from 24 males and 6 females
peptide 102-PTFLSNE-108, suggested that CLOCK S106 was phospho- with an average (±standard deviation (s.d.)) age 63.1 (±10.8) years
rylated. The XCorr score was 2.33. Mass spectrometric analysis of a were used for IHC staining to compare the protein expression between
tryptic fragment at m/z 599.87543 Da (+1.51 mmu/+2.53 ppm), which tumour and normal tissue specimen. HCC samples from 71 males and
was matched with the +2 charged peptide 23-LGLELGKVVTK-33, sug- 19 females with an average (±s.d.) age of 61.4 (±11.2) years were used
gested that PRPS K29 was acetylated. The XCorr score was 3.25. to analyse the relationship between IGFR1 pY1161, α-catenin pS641,
CLOCK pS106, PRPS1/2 K29ac, PRPS1/2 and the clinical aggressiveness
Immunofluorescence analysis of HCC. All the patient samples underwent surgical resection and were
Cells were fixed and incubated with primary antibodies at a dilution of collected in compliance with the informed consent policy. Sections of
1:100, fluorescence dye-conjugated secondary antibodies and DAPI, paraffin-embedded human HCC samples were stained with antibodies
according to standard protocols. Immunofluorescence microscopic against IGFR1 pY1161, α-catenin pS641, CLOCK pS106, PRPS1/2 K29ac or
images of the cells were obtained and viewed with an IX81 confocal PRPS1/2 and non-specific IgG as a negative control. The staining of the
microscope system (Olympus America). FV10-ASW Viewer (Version tissue sections was quantitatively scored according to the percentage
4.2b) is used to collect the immunofluorescence data. Fluorescence of positive cells and the staining intensity as described previously42.
intensity was quantified by ImageJ (National Institutes of Health). The following proportion scores were assigned to the sections: 0 if
0% of the tumour cells exhibited positive staining, 1 for 0–1% of cells
Subcellular fractionation stained, 2 for 2–10% of cells stained, 3 for 11–30% of cells stained, 4 for
Nuclear and cytosolic fractions were isolated from the cells using a 31–70% of cells stained and 5 for 71–100% of cells stained. In addition,
nuclear/cytosol fractionation kit (K266, BioVision) according to the the staining intensity was rated on a scale of 0–3: 0, no staining; 1, weak;
manufacturer’s instructions. 2, moderate; and 3, strong. The proportion and intensity scores were
then combined to obtain a total score (range, 0–8). The scores were
Real-time PCR compared with overall survival duration, defined as the time from the
Total RNA was extracted from cells and tissue specimens using TRIzol date of diagnosis to that of death or last known follow-up examination.
according to the manufacturer’s instructions (Invitrogen). A total All patients had received standard therapies after surgery.
of 1 mg per sample RNA was used for cDNA synthesis with a TaqMan
reverse transcription reagent kit (Applied Biosystems). Quantitative Animal studies
real-time PCR was carried out in a 7500 real-time PCR system (Applied Six-week-old male athymic BALB/c nude mice were housed under
Biosystems) using a SYBR Premix Ex Taq kit for real-time PCR (TaKaRa) specific-pathogen-free conditions in a 14 h light/10 h dark cycle. Ambi-
as previously reported39. GAPDH served as the normalization gene in ent temperature (21–23 °C, 50% humidity) with access to water and food
these studies. The following primers were used for qRT–PCR: ad libitum. Animal studies were approved by Research Ethics Commit-
Nr1d1, 5′-CCCTGGGAGTCTACAAGTGG-3′ and 5′-GCGATTGATGC tee and complied with the ethical regulations of The First Affiliated
GGACGAT-3′; Dbp, 5′-CCTCGAAGACATCGCTTCTC-3′ and 5′-GCCT Hospital, Zhejiang University School of Medicine and Qingdao Cancer
CGTTGTTCTTGTACCG-3′; Prps1, 5′-GATCTATTTGGCCTCTCAAA-3′ Institute. One million Huh7 cells expressing CLOCK shRNA with recon-
and 5′-CACACAGGTACACACACTTTATT-3′; Prps2, 5′-GGACCTGCA stituted expression of the indicated Flag-rCLOCK proteins or parental
TGCTTCTCAG-3′ and 5′-ATGTTTTCCCGAATCCACTG-3′; and Gapdh, Huh7 cells and the indicated clones with knock-in expression of PRPS1/2
5′-AGCCACATCGCTCAGACAC-3′ and 5′-GCCCAATACGACCAATCC-3′. K29R mutants were collected in 20 μl of DMEM with 33% Matrigel and
intrahepatically injected into the livers of 6-week-old male athymic
MD simulations BALB/c nude mice. The injections were performed as described previ-
The human CLOCK/BMAL1 complex model was built using the crystal ously31. The animals were killed 28 days after injection. The liver of each
structure of the mouse CLOCK/BMAL1 (PDB ID: 4F3L) as the template mouse was dissected and then fixed in 4% formaldehyde and embed-
using MODELLER10.1 as previously described40. In this study, 1,000 ded in paraffin. Tumour formation and phenotype were determined
homology models were created and the top model with highest dope by histologic analysis of haematoxylin-and-eosin-stained sections.
score was selected for the MD simulations. After creation of the human Tumour volume was calculated using the following formula: V = 1/2a2b
CLOCK/BMAL1 complex model, MD simulations were performed using (V, volume; a, shortest diameter; b, longest diameter). The animals
FF19SB forcefield for the proteins in AMBER20 package as previously were treated in accordance with relevant institutional and national
described41. The protein was solvated in a truncated octahedral OPC guidelines and regulations. The use of the animals was approved by
water box containing 25,534 water molecules. Three Na+ ions were the Institutional Review Board at The First Affiliated Hospital, Zheji-
added to neutralize the systems. Before production runs, the sys- ang University School of Medicine and Qingdao Cancer Institute. The
tem was minimized and equilibrated. Afterwards, 500 ns simulations maximal tumour burden permitted by ethics committee is 1,500 cm3,

Nature Cell Biology


Article https://doi.org/10.1038/s41556-022-01061-0

and the maximal tumour burden did not exceed the limit. No statistical 39. Xu, D. et al. PAQR3 modulates cholesterol homeostasis by
method was used to pre-determine the sample size. The investigators anchoring Scap/SREBP complex to the Golgi apparatus. Nat.
were not blinded to treatment allocation during experiments or to the Commun. 6, 8100 (2015).
outcome assessment. 40. Yu, R., Craik, D. J. & Kaas, Q. Blockade of neuronal α7-nAChR
by α-conotoxin ImI explained by computational scanning
Statistics and reproducibility and energy calculations. PLoS Comput. Biol. 7, e1002011
All statistical data are presented as mean ± s.d. All experiments were (2011).
repeated at least twice independently with similar results. The mean 41. Yu, R. et al. Molecular determinants conferring the
values obtained in the control and experimental groups were analysed stoichiometric-dependent activity of α-conotoxins at the human
for significant differences. Pairwise comparisons were performed using α9α10 nicotinic acetylcholine receptor subtype. J. Med. Chem. 61,
a Student’s t-test (two-tailed), one-way ANOVA post hoc test (unpaired 4628–4634 (2018).
two-tailed Student’s t-test with Bonferroni correction) or Mann–Whit- 42. Yang, W. et al. Nuclear PKM2 regulates β-catenin transactivation
ney U test. P values of less than 0.05 were considered significant. No upon EGFR activation. Nature 480, 118–122 (2011).
statistical method was used to pre-determine sample size. No data
were excluded from the analyses. Data distribution was assumed to be Acknowledgements
normal, but this was not formally tested. Unless stated otherwise, the This study was supported by grants from the Ministry of Science
experiments were not randomized. Data collection and analysis were and Technology of the People’s Republic of China (2020YFA0803300,
not performed blind to the conditions of the experiments. Z.L.; 2021YFA0805600, D.X.), the National Natural Science Foundation
of China (92157113 and 82072630, D.X.; 82173114, Z.W.; 82072903
Reporting summary and 82272872, T.L.; 82002811, M. Yan; 82188102 and 82030074,
Further information on research design is available in the Nature Port- Z.L.), the Zhejiang Natural Science Foundation Key Project
folio Reporting Summary linked to this article. (LD22H160002, D.X.; LD21H160003, Z.L.), Zhejiang Natural Science
Foundation Discovery Project (LQ22H160023, Z.W.), and the Leading
Data availability Innovative and Entrepreneur Team Introduction Program of Zhejiang
Mass spectrometry data have been deposited in ProteomeXchange with (2019R01001, Z.L.). Z.L. is the Kuancheng Wang Distinguished
the accession code PXD037738. UniProt protein database (EMBL-EBI) Chair. The authors received no specific funding for this work. We
was used for protein identification. All other data supporting the find- express our great gratitude to Dr. Hong Wang and his team from
ings of this study are available from the corresponding author on the Hangzhou Cosmos Wisdom Mass Spectrometry Center of
reasonable request. Source data are provided with this paper. Zhejiang University Medical School for their technical support in
sample analysis utilizing an integrated nanoLC–ESI–MS/MS and data
References processing platform.
28. Li, X. et al. Nucleus-translocated ACSS2 promotes gene
transcription for lysosomal biogenesis and autophagy. Mol. Cell Author contributions
66, 684–697 e689 (2017). D.X. and Z.L. conceived and designed the study and wrote the
29. Lee, J. H. et al. EGFR-phosphorylated platelet isoform of manuscript. T.L., Z.W., L.Y., Y.D., H.H., K.W., M. Yan, G.J., Y.S., L.W., L.L.,
phosphofructokinase 1 promotes PI3K activation. Mol. Cell 70, P.Z., B.D., F.S. and Z.Z. performed the experiments; H.J., M. Yang and
197–210 e197 (2018). R.Y. were involved in MD simulation analyses. Y.X., L.X., Q.W. and X.Q.
30. Lee, J. H. et al. Stabilization of phosphofructokinase 1 platelet reviewed and edited the manuscript.
isoform by AKT promotes tumorigenesis. Nat. Commun. 8, 949
(2017). Competing interests
31. Li, X. et al. A splicing switch from ketohexokinase-C to Z.L. owns shares in Signalway Biotechnology (Pearland, TX), which
ketohexokinase-A drives hepatocellular carcinoma formation. supplied rabbit antibodies that recognize CLOCK pS106 and PRPS1/2
Nat. Cell Biol. 18, 561–571 (2016). K29ac. Z.L.’s interest in this company had no bearing on its being
32. Qian, X. et al. Conversion of PRPS hexamer to monomer by chosen to supply these reagents. The remaining authors declare no
AMPK-mediated phosphorylation inhibits nucleotide synthesis in competing interests.
response to energy stress. Cancer Discov. 8, 94–107 (2018).
33. Xu, D. Q. et al. PAQR3 controls autophagy by integrating AMPK Additional information
signaling to enhance ATG14L-associated PI3K activity. EMBO J. 35, Extended data is available for this paper at https://doi.org/10.1038/
496–514 (2016). s41556-022-01061-0.
34. Qian, X. et al. KDM3A senses oxygen availability to regulate
PGC-1α-mediated mitochondrial biogenesis. Mol. Cell 76, Supplementary information The online version contains
885–895 e887 (2019). supplementary material available at https://doi.org/10.1038/s41556-
35. Qian, X. et al. PTEN suppresses glycolysis by dephosphorylating 022-01061-0.
and inhibiting autophosphorylated PGK1. Mol. Cell 76, 516–527
e517 (2019). Correspondence and requests for materials should be addressed to
36. Xu, D. et al. The protein kinase activity of fructokinase A specifies Zhimin Lu or Daqian Xu.
the antioxidant responses of tumor cells by phosphorylating p62.
Sci. Adv. 5, eaav4570 (2019). Peer review information Nature Cell Biology thanks Lars Zender,
37. Lin, S. Y. et al. GSK3-TIP60-ULK1 signaling pathway links growth Tsuyoshi Hirota and the other, anonymous, reviewer(s) for their
factor deprivation to autophagy. Science 336, 477–481 (2012). contribution to the peer review of this work.
38. Du, L. et al. β-Catenin induces transcriptional expression of PD-L1
to promote glioblastoma immune evasion. J. Exp. Med. https:// Reprints and permissions information is available at
doi.org/10.1084/jem.20191115 (2020). www.nature.com/reprints.

Nature Cell Biology


Article https://doi.org/10.1038/s41556-022-01061-0

Extended Data Fig. 1 | CLOCK S106 phosphorylation disassembles the with purified His-CLOCK. Proteins precipitated by GST pulldown assay were
CLOCK–BMAL1 complex. (b-e, h, j, k, m, n) Immunoprecipitation and incubated with or without CIP for 30 min. (f) In vitro kinase assays were
immunoblotting with the indicated antibodies was performed. All experiments performed by mixing purified His–CLOCK with or without purified GST-CK2α in
were repeated at least twice independently. Data are the mean ± SD. (a) The the presence of ATP. Mass-spectrometric analysis was performed. (g) Alignment
luciferase activity of the indicated cells expressing a Per1-driven luciferase of CLOCK to the consensus CK2α-phosphorylated substrate motif (SXXD/E). (h,
reporter was measured after IGF1 treatment (12 h) (n = 6). *P < 0.01; **P < 0.001 i) Huh7 cells expressing Flag-CLOCK were treated with or without IGF1 for 1 h (h).
by One-way ANOVA post hoc test. (b, c, m, n) The indicated cells were pretreated Immunoblotting (h) or IHC analyses of human HCC samples (i) were performed
with or without the indicated inhibitors for 30 min before IGF1 treatment for with the indicated antibodies and a CLOCK pS106-blocking peptide. ( j-l) The
30 min (b, c, m) or transfected with the indicated plasmids before IGF1 treatment indicated cells expressing the indicated plasmids (j) were treated with or without
for 1 h (n). Total cell lysates and cytosolic fractions were prepared. (d) Flag-CK2α IGF1 for 1 h ( j-l). Total cell lysates and cytosolic and nuclear fractions were
immunoprecipitated from Huh7 cells treated with or without IGF1 for 30 min prepared (k). The relative CLOCK abundance was quantified (n = 10) (l). *P < 0.05;
was incubated with or without CIP for 30 min. (e) Purified GST-CK2α was mixed ***P < 0.0001 by two-tailed Student’s t test.
with or without active His-ERK2 for in vitro kinase assay, followed by incubation

Nature Cell Biology


Article https://doi.org/10.1038/s41556-022-01061-0

Extended Data Fig. 2 | CLOCK S106 phosphorylation is required for its and blue, respectively. The red arrow indicates CLOCK S106. D144, D232, D299,
nuclear exportation. (e-i) Immunoprecipitation and immunoblotting analyses and E303 of BMAL1, which are in a close proximity with CLOCK S106, are shown.
were performed with the indicated antibodies. All experiments were repeated (e) Bacterially purified WT GST-CLOCK or GST-CLOCK S106A was incubated with
three times independently with similar results. (a) Alignment of protein or without His-CK2α in the presence of ATP for an in vitro kinase assay, followed
sequences spanning CLOCK S106 and the adjacent noncanonical NES from by incubation with the indicated His-BMAL1 proteins. (f) Huh7 cells stably
different species. (b, d) MD simulation of CLOCK bound with BMAL1 WT (b) or transfected with the indicated plasmids were treated with or without IGF1 for
BMAL1 4Mut (D144A/D231A/D299A/E303A) (d). Evolution of the backbone root- 1 h. Total cell lysates and cytosolic fractions were prepared. (g) Huh7 cells were
mean-square-deviation (RMSD) of the CLOCK-BMAL1 complex from the initial pretreated with or without TBB for 30 min before treatment with or without
frame in 250 ns MD simulations. RMSD of the CLOCK-BMAL1 complex with and IGF1 for 1 h. (h) The indicated cells expressing CLOCK shRNA with reconstituted
without CLOCK S106 phosphorylation are shown in red and black, respectively. expression of the indicated CLOCK proteins were harvested. (i) Hep3B cells
(c) The conformation of the CLOCK-BMAL1 complex of the last frame extracted expressing CLOCK shRNA with reconstituted expression of Flag-rCLOCK
from 250 ns MD simulation. CLOCK and BMAL1 structures are shown in green proteins were treated with or without IGF1 for 1 h.

Nature Cell Biology


Article https://doi.org/10.1038/s41556-022-01061-0

Extended Data Fig. 3 | CLOCK acetylates PRPS1/2 K29. (d, j, m) were performed with the indicated antibodies and a PRPS1/2 K29 acetylation-
Immunoprecipitation and immunoblotting with the indicated antibodies was blocking peptide. (f-j) Genomic DNA was extracted from two individual clones
performed. All experiments were repeated at least twice independently. (a) of the indicated cells with knock-in expression of PRPS1/2 K29R. PCR products
Huh7 cells with or without expression of Flag–CLOCK were treated with or amplified from the indicated DNA fragments were shown (f, g) and sequenced
without IGF1 for 1 h. The immunoprecipitated Flag–CLOCK was eluted with (h, i). The red line indicates the sgRNA-targeting sequence. The black line
Flag peptide and stained with Coomassie Brilliant Blue after SDS-PAGE. Mass indicates the PAM. Blue arrows indicate mutated nucleotides. A mutated amino
spectrometry-identified protein peptide hits are shown. (b) Purified GST–CLOCK acid and its WT counterpart are indicated by the solid red box (h, i). These cells
was incubated with or without His-CK2α and TBB for an in vitro kinase assay, were transfected with or without constitutively active IGF1R-CA ( j). (k) Purified
followed by incubation with or without the indicated His-PRPS1/2 proteins in GST–CLOCK were incubated with or without His-CK2α for an in vitro kinase assay,
the presence of acetyl-CoA. Mass-spectrometric analysis was performed. (c) followed by incubation with or without the indicated His-PRPS1/2 proteins in the
Alignment of protein sequences spanning PRPS1/2 K29 from different species. presence of acetyl-CoA. (l, m) Huh7 (l, m) and Hep3B (l) cells expressing CLOCK
(d, e) Huh7 cells expressing Flag-PRPS1 were treated with or without IGF1 for shRNA with reconstituted expression of the indicated CLOCK proteins were
1 h (d). Immunoblotting analyses (d) or IHC analyses of human HCC samples (e) harvested or (l) treated with or without IGF1 for 1 h (m).

Nature Cell Biology


Article https://doi.org/10.1038/s41556-022-01061-0

Extended Data Fig. 4 | Cytosolic CLOCK promotes PRPS1/2 stabilization. hoc test (e, g). (h) The indicated cells were treated with DMSO, MG132 (10 μM),
(a, d-h, j, k, l, o) Immunoprecipitation and immunoblotting with the indicated PS341 (10 μM), CQ (25 μM) or bafilomycin A1 (BFA) (10 nM) for 8 h. (i) Alignment
antibodies was performed. All experiments were repeated three times of protein sequences spanning PRPS1/2 K29 and the adjacent HSC70 recognized
independently. (a) The indicated cells were stably transfected with active motif from different species. ( j) LAMP2A shRNA was expressed in the indicated
IGF1R-CA. (b, c) Huh7 cells stimulated with IGF1 for the indicated time (b) or cells. (k-n) Huh7 cells expressing PRPS1/2 shRNA with reconstituted expression
expressing IGF1R-CA (c) were harvested. The mRNA expression levels of the of the indicated PRPS1/2 proteins were harvested for immunoblotting (k, l) or
PRPS1/2 genes were measured using qPCR (n = 6). Data are the mean ± SD. N.S., qPCR (n = 6) to measure Prps1/2 mRNA levels (m, n). Data are the mean ± SD.
not significant by One-way ANOVA post hoc test (b) or two-tailed Student’s t N.S., not significant by One-way ANOVA post hoc test. (o) Endogenous CLOCK-
test (c). (d-g) The indicated cells expressing CLOCK shRNA and active IGF1R-CA depleted Huh7 cells with reconstituted expression of the indicated CLOCK
with reconstituted expression of the indicated CLOCK protein were treated with protein were transfected with HA-PRPS1/2 and treated with or without IGF1 for
CHX for the indicated time (d, f). The quantification of PRPS1/2 levels is shown. 1 h. Immunoprecipitation using limited amount of HA antibody was performed.
Data are the mean ± SD, n = 6, **P < 0.001; ***P < 0.0001 by One-way ANOVA post

Nature Cell Biology


Article https://doi.org/10.1038/s41556-022-01061-0

Extended Data Fig. 5 | See next page for caption.

Nature Cell Biology


Article https://doi.org/10.1038/s41556-022-01061-0

Extended Data Fig. 5 | CLOCK-mediated PRPS1/2 stabilization promotes de kinase assay, followed by incubation with or without the indicated His-PRPS1/2
novo nucleotide synthesis. (g-l) Immunoprecipitation and immunoblotting proteins in the presence of acetyl-CoA. The PRPS1/2 activity was measured. (h,
with the indicated antibodies was performed. All experiments were repeated i) Huh7 cells expressing CLOCK shRNA with reconstituted expression of the
at least twice independently. (a-g) Data are the mean ± SD, n = 6, *P < 0.01; indicated Flag-rCLOCK were serum-starved for 12 h in the presence of CQ and
**P < 0.001; ***P < 0.0001; N.S., not significant by One-way ANOVA post hoc test. then treated with or without EGF (h) or FGF1 (i) for 1 h. Cytosolic and whole
(a-f) The indicated cells expressing CLOCK shRNA with reconstituted expression cell lysates were harvested. ( j) Huh7 cells were stimulated with EGF (100 ng/
of the indicated CLOCK proteins (a, b, e) or the indicated clones with knock-in ml) or FGF1 (25 ng/ml) for the indicated time. (k) Huh7 cells stably transfected
expression of PRPS1/2 K29R (c, d, f) were transfected with or without active with constitutively active EGFR-vIII or FGFR1-CA were harvested. (l) Huh7 cells
IGF1R-CA, followed by labeling with D-[6-14C] glucose for 30 min. The amounts of expressing CLOCK shRNA and constitutively active EGFR-vIII or FGFR1-CA with
14
C-RNA (a, c) and 14C-DNA (b, d) were measured. The 13C-labeled PRPP, IMP, AMP, reconstituted expression of the indicated Flag-rCLOCK were treated with CHX
GMP, UMP, and CMP were measured by LC/MS-MS (e, f). (g) Purified GST–CLOCK for the indicated time. The quantification of PRPS1/2 protein levels is shown. Data
was incubated with or without His-CK2α in the presence of ATP for an in vitro are the mean ± SD, **P < 0.001 by One-way ANOVA post hoc test.

Nature Cell Biology


Article https://doi.org/10.1038/s41556-022-01061-0

Extended Data Fig. 6 | See next page for caption.

Nature Cell Biology


Article https://doi.org/10.1038/s41556-022-01061-0

Extended Data Fig. 6 | Overexpressed CK2α in HCC cells is critical for The amounts of 14C-RNA and 14C-DNA were measured. (d) The indicated cells
CLOCK-enhanced PRPS1/2 stability. (a, b, d-l) Immunoprecipitation and expressing CLOCK shRNA and IGF1R-CA with reconstituted expression of Flag-
immunoblotting with the indicated antibodies was performed. All experiments rCLOCK were treated with CHX for the indicated time. The quantification of
were repeated three times independently. (c, d, g, i) Data are the mean ± SD. PRPS1/2 levels is shown. (e, f) The indicated cells were treated with or without
n = 6, *P < 0.01; **P < 0.001; ***P < 0.0001 by One-way ANOVA post hoc test. (a, b) IGF1 for 1 h. Whole cell lysates were harvested. (g) L02 cells transfected with the
The indicated cells expressing CLOCK shRNA with reconstituted expression of indicated plasmids were treated with CHX for the indicated time in the presence
the indicated shRNA-resistant CLOCK were constructed (a) and serum-starved or absence of IGF1. The quantification of PRPS1/2 levels is shown. (h) Huh7 cells
for 12 h in the presence of CQ before treatment with or without IGF1 for 1 h (b). transfected with the indicated shRNA were treated with or without IGF1 for 1 h. (i)
Cytosolic and whole cell lysates were harvested. (c) The indicated cells with Huh7 cells transfected with the indicated shRNA were treated with CHX for the
reconstituted expression of the indicated CLOCK proteins were treated with indicated time in the presence or absence of IGF1. The quantification of PRPS1/2
or without IGF1 for 12 h, followed by labeling with D-[6-14C] glucose for 30 min. levels is shown.

Nature Cell Biology


Article https://doi.org/10.1038/s41556-022-01061-0

Extended Data Fig. 7 | Both nuclear transcriptional activity and cytosolic labeled with D-[6-14C] glucose. The amounts of 14C-RNA (f) and 14C-DNA (g) were
function of CLOCK contribute to HCC cell proliferation. (a, d, e, i, j, l, m) measured (n = 6). (h) The indicated cells transfected with the indicated plasmids
Immunoprecipitation and immunoblotting with the indicated antibodies was and shRNA were plated in complete culture medium and counted (n = 6). (i)
performed. All experiments were repeated at least twice independently. (a-c, Huh7 cells were pretreated with or without different dose of Sorafenib for
e-g, h, j, k, n) Data are the mean ± SD. *P < 0.01; **P < 0.001; ***P < 0.0001 by One- 30 min before IGF1 treatment for 1 h. ( j, k) The indicated cells expressing CLOCK
way ANOVA post hoc test. (a-c) The indicated cells stably transfected with the shRNA and active IGF1R-CA with reconstituted expression of Flag-rCLOCK WT or
indicated plasmids and shRNA were constructed (a) and transiently expressed S106D were treated with CHX ( j) or without CHX (k) in the presence or absence
a Per1-driven luciferase reporter before IGF1 treatment for 12 h. The luciferase of Sorafenib for the indicted time. PRPS1/2 protein quantification ( j) and cell
activity is shown (n = 6). The mRNA levels of CLOCK-downstream target genes counting (k) were performed (n = 6). (l-n) The indicated cells with reconstituted
were measured using qPCR (n = 6) (c). Cytosolic and whole cell lysates from Huh7 expression of Flag-rCLOCK WT or S106D were harvested (l) or treated with CHX
cells were harvested (d). (e) Huh7 cells transfected with the indicated plasmids (m) or without CHX (n) for the indicated time. PRPS1/2 protein quantification (m)
and shRNA were treated with CHX. PRPS1/2 protein was quantified (n = 6). (f, and cell counting (n) were performed (n = 6).
g) The indicated cells transfected with the indicated plasmids and shRNA were

Nature Cell Biology


Article https://doi.org/10.1038/s41556-022-01061-0

Extended Data Fig. 8 | CLOCK-mediated PRPS1/2 stabilization promotes brown and quantified in n = 10 microscopic fields (lower). Data are the mean ± SD,
HCC cell proliferation and liver tumor growth. (a, b) Huh7 cells (1 × 106) ***P < 0.0001 by One-way ANOVA post hoc test. (g) Huh7 cells (1 × 106) expressing
expressing CLOCK shRNA with reconstituted expression of the indicated CLOCK CLOCK shRNA with reconstituted expression of the indicated CLOCK proteins
proteins (a) expressing WT PRPS1/2 or PRPS1/2 K29R knock-in mutants (b) were intrahepatically injected into athymic nude mice (n = 6 per group). The mice
were subcutaneously injected into athymic nude mice (n = 6 per group). The were euthanized and examined for tumor growth 22 days after injection. The
mice were euthanized and examined for tumor growth 28 days after injection. arrows indicate tumors (left). The tumor volumes were measured (right). Data are
Tumor volumes were calculated, and the tumors were weighed. Data are the the mean ± SD, n = 6, *P < 0.01 by two-tailed Student’s t test. (h) Huh7 cells (1 × 106)
mean ± SD, n = 7, *P < 0.01; **P < 0.001 by One-way ANOVA post hoc test. (c, d) expressing CLOCK shRNA with reconstituted expression of the indicated CLOCK
IHC analyses of the indicated tumor samples were performed with an anti-Ki67 proteins were subcutaneously injected into athymic nude mice (n = 7 per group).
antibody. Ki67-positive cells were quantified. Data are the mean ± SD, n = 10, The resulting tumors were resected 22 days after injection (left). The growth of
***P < 0.0001 by One-way ANOVA post hoc test. (e, f) TUNEL analyses of the xenografted tumors in the mice was measured (middle) and the tumors were
indicated tumor samples were performed (upper). Apoptotic cells were stained weighed (right). Data are the mean ± SD, **P < 0.001 by two-tailed Student’s t test.

Nature Cell Biology


α

α α
α

You might also like