You are on page 1of 17

RESEARCH ARTICLE COLLECTION: CANCER PROTEOMICS

Deciphering the Clinical Significance and Kinase


Functions of GSK3α in Colon Cancer by Proteomics
and Phosphoproteomics
Authors
Li Gao, Ying Lu, Hai-Ning Chen, Zhigui Li, Meng Hu, Rou Zhang, Xiuxuan Wang, Zhiqiang Xu,
Yanqiu Gong, Rui Wang, Dan Du, Shan Hai, Shuangqing Li, Dan Su, Yuan Li, Heng Xu,
Zong-Guang Zhou, and Lunzhi Dai

Correspondence Graphical Abstract


lunzhi.dai@scu.edu.cn

In Brief
GSK3α and GSK3β are distinct
isoforms, with GSK3β studied in
cancer development while
GSK3α was considered
redundant. Yet, our research has
revealed that GSK3α, rather than
GSK3β, is significantly correlated
with colon cancer patients'
survival. Integrative In vitro and in
cell line phosphoproteomics
identified 156 phosphosites
specifically regulated by GSK3α.
Notably, phosphorylation of
THRAP3S248 remarkably
enhanced colon cancer cell
migration. These findings
suggest that GSK3α may have
unique and important roles in
colon cancer development and
progression.

Highlights

• High levels of GSK3α predict poor prognosis of colon cancer patients.


• Identification of 156 phosphosites specifically regulated by GSK3α, not GSK3β.
• THRAP3 has been identified as a phospho-substrate of GSK3α.
• GSK3α-mediated phosphorylation of THRAP3 at S248 promotes cancer cell migration.

2023, Mol Cell Proteomics 22(5), 100545


© 2023 THE AUTHORS. Published by Elsevier Inc on behalf of American Society for Biochemistry and
Molecular Biology. This is an open access article under the CC BY license (http://creativecommons.org/
licenses/by/4.0/).
https://doi.org/10.1016/j.mcpro.2023.100545
RESEARCH ARTICLE COLLECTION: CANCER PROTEOMICS

Deciphering the Clinical Significance and


Kinase Functions of GSK3α in Colon Cancer by
Proteomics and Phosphoproteomics
Li Gao1,‡, Ying Lu1,‡, Hai-Ning Chen2,‡ , Zhigui Li3,‡, Meng Hu1,‡, Rou Zhang1,
Xiuxuan Wang1, Zhiqiang Xu1, Yanqiu Gong1, Rui Wang4, Dan Du4, Shan Hai1,
Shuangqing Li1, Dan Su1, Yuan Li5, Heng Xu1, Zong-Guang Zhou2,5, and Lunzhi Dai1,*

GSK3α and GSK3β are two GSK3 isoforms with 84% deaths (2). Since the first approval of cetuximab and bev-
overall identity and 98% identity in their catalytic domains. acizumab in 2004, a great number of agents targeting the ki-
GSK3β plays important roles in the pathogenesis of can- nases EGFR and VEGFR have entered the clinic (3). Glycogen
cer, while GSK3α has long been considered a functionally synthase kinase GSK3 is a key kinase in the Wnt/β–catenin
redundant protein of GSK3β. Few studies have specifically
signaling pathway (4). It is responsible for the phosphorylation
investigated the functions of GSK3α. In this study, unex-
of β-catenin, which subsequently triggers the ubiquitination and
pectedly, we found that the expression of GSK3α, but not
GSK3β, was significantly correlated with the overall sur- degradation of β-catenin. Adenomatous polyposis coli muta-
vival of colon cancer patients in 4 independent cohorts. To tions may inactivate GSK3 and lead to activation of the Wnt/
decipher the roles of GSK3α in colon cancer, we profiled β–catenin signaling pathway, which is an initiation factor of
the phosphorylation substrates of GSK3α and uncovered more than 80% of spontaneous CRCs (5). GSK3 is a multi-
156 phosphosites from 130 proteins specifically regulated tasking molecule and serves as either a tumor promoter or tu-
by GSK3α. A number of these GSK3α-mediated phos- mor suppressor in cancers (6, 7). To date, in addition to
phosites have never been reported before or have been β-catenin (8), more than 40 phospho-substrates of GSK3, such
incorrectly identified as substrates of GSK3β. Among as TSC2 (9), SMAD1 (10), GLI3 (11), SUFU (12), cyclin D1 (13),
them, the levels of HSF1S303p, CANXS583p, MCM2S41p, Snail (14), IκB (15), and NICD1 (16), have been described.
POGZS425p, SRRM2T983p, and PRPF4BS431p were signifi-
GSK3 has two isoforms, GSK3α and GSK3β, encoded by
cantly correlated with the overall survival of colon cancer
GSK3A and GSK3B, respectively. The two GSK3 isoforms have
patients. Further pull-down assays identified 23 proteins,
such as THRAP3, BCLAF1, and STAU1, showing strong 84% overall identity and 98% identity in their catalytic domains
binding affinity to GSK3α. The interaction between (17). GSK3β can shuttle between the nucleus and cytoplasm,
THRAP3 and GSK3α was verified by biochemical experi- while GSK3α is usually found in the cytoplasm due to its glycine-
ments. Notably, among the 18 phosphosites of THRAP3, rich N terminus. However, calcium or calpain products can
phosphorylation at S248, S253, and S682 is specifically promote GSK3α nuclear accumulation (18). The activities of
mediated by GSK3α. Mutation of S248 to D (S248D), which GSK3α and GSK3β are modulated by their phosphorylation (19,
mimics the effect of phosphorylation, obviously increased 20). Despite tremendous progress in the study of GSK3 func-
cancer cell migration and the binding affinity to proteins tions, most previous studies focused on GSK3β or failed to
related to DNA damage repair. Collectively, this work not distinguish the functional differences between GSK3α and
only discloses the specific function of GSK3α as a kinase
GSK3β. Thus far, only a few studies have specifically examined
but also suggests GSK3α as a promising therapeutic
GSK3α, including the functional elucidation of GSK3α in organ
target for colon cancer.
aging (21), ESC differentiation (22), atherosclerosis and hepatic
steatosis (23), and hippocampal N-methyl-D-aspartate
Colorectal cancer (CRC) is the third most common cancer receptor-dependent long-term depression (24). However, the
and has the second highest mortality rate worldwide (1). Distant clinical significance and specific functions of GSK3α as a kinase
metastasis is responsible for approximately 90% of CRC in cancer have been largely ignored.

From the 1National Clinical Research Center for Geriatrics and General Practice Ward/International Medical Center Ward, General Practice
Medical Center, State Key Laboratory of Biotherapy, 2Colorectal Cancer Center, Department of General Surgery, 3Department of Gastroin-
testinal Surgery, 4Advanced Mass Spectrometry Center, Research Core Facility, Frontiers Science Center for Disease-related Molecular
Network, and 5Institute of Digestive Surgery, West China Hospital, Sichuan University, Chengdu, China

These authors contributed equally to this work.
* For correspondence: Lunzhi Dai, lunzhi.dai@scu.edu.cn.

Mol Cell Proteomics (2023) 22(5) 100545 1


© 2023 THE AUTHORS. Published by Elsevier Inc on behalf of American Society for Biochemistry and Molecular Biology.
This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/). https://doi.org/10.1016/j.mcpro.2023.100545
Clinical Significance and Functions of GSK3α in Colon Cancer

In this study, we screened the key kinases that affect the at 37 ◦ C overnight. The trypsin was then inactivated by heating at 95

survival of colon cancer patients and may be potential targets. C for 2 min. The digested peptides and the common reference
sample were labeled by TMT10plex reagents (Thermo Fisher Scienti-
Based on our proteome and immunostaining data and publicly
fic). After quenching with hydroxylamine, the colon cancer samples
available proteome data from 4 independent colon cancer and the reference sample in the same batch were combined. The
cohorts (25, 26), we showed that GSK3α, but not GSK3β, was combined peptides were desalted by reversed-phase C18 SPE col-
able to predict the prognosis of colon cancer patients. To umns (Phenomenex, 10 mg) and separated into 120 fractions by
decipher the roles of GSK3α in colon cancer, we profiled the reversed-phase HPLC (RP-HPLC) at a flow rate of 1 ml/min. These
fractions were combined into 15 fractions before MS analysis.
in vitro and cellular phospho-substrates of GSK3α and iden-
For phosphoproteomics of colon cancer tissues, 70 paired tumors
tified 156 phosphosites specifically regulated by GSK3α but and DNTs were divided into 18 TMT-labeled batches with common
not GSK3β. Further pull-down assays identified 23 proteins, reference samples. Forty micrograms of digested peptides from each
such as THRAP3, BCLAF1, and STAU1, with strong binding sample were labeled with TMT reagents. The combined TMT-labeled
affinity specific to GSK3α. Notably, among the 18 phospho- peptides were separated into 15 fractions by C18 SPE columns
(Phenomenex, 100 mg) and then combined into 5 fractions, which
sites of THRAP3, phosphorylation at S248, S253, and S682 is
were further used for phosphopeptide enrichment.
specifically mediated by GSK3α in vitro and in cell lines. Mu-
tation of S248 to D (S248D) to mimic the effect of its phos- Enrichment of Phosphopeptides
phorylation obviously increased the migratory ability of cancer PureCube Fe-nitrilotriacetic acid (NTA)-agarose beads were used
cells and the binding affinity to proteins related to DNA for phosphopeptide enrichment and purchased from Cube Biotech.
damage repair (DDR). The desalted peptides were dissolved in loading buffer (85% aceto-
nitrile (ACN), 0.1% TFA) and then incubated with Fe-NTA agarose
beads at 25 ◦ C for 1 h. After incubation, the agarose beads were
washed 4 times with washing buffer (80% ACN, 0.1% TFA). Then, the
EXPERIMENTAL PROCEDURES
phosphopeptides were eluted from the agarose beads by elution
Declaration of Helsinki Ethical Principles buffer (40% ACN, 15% ammonium hydroxide). The eluted peptides
were acidized with 10% TFA and dried. The samples were desalted
This work was conducted according to the Declaration of Helsinki
and further analyzed by LC-MS/MS.
ethical principles. Paired colon cancer samples from 78 treatment-
naïve patients were obtained from West China Hospital. The tissues LC-MS/MS Analysis
were quickly frozen in liquid nitrogen after surgery and stored in a −80
◦ For proteomics profiling of colon cancer tissues, desalted peptides
C refrigerator until use. The pathological sections were processed by
the Department of Pathology of West China Hospital, a laboratory of each fraction were analyzed using an EASY-nanoLC 1200 system
accredited by the College of American Pathologists. Both colon (Thermo Fisher Scientific) coupled to an Orbitrap Exploris 480 mass
cancer tissues used for omics and colon cancer tissues used for spectrometer (Thermo Fisher Scientific). The peptides were separated
immunohistochemistry were obtained with informed consent and the in the analytical column with a 65 min gradient with 4% to 100% buffer
approval of the Ethics Committee of Biology Research, West China B (80% ACN, 0.1% formic acid) at a flow rate of 300 nl/min. Under the
Hospital at Sichuan University (Permission number: 2020(374)). Clin- data-dependent acquisition mode, MS spectra were acquired from
ical information, including age, sex, tumor stage, and follow-up in- 350 to 1800 m/z with a resolution of 60,000 at m/z = 200. The
formation, was collected. normalized automatic gain control (AGC) target (%) was set as 300%.
For the MS2 analysis, a 0.7 m/z isolation window and normalized
Protein Extraction collision energy (NCE) of 36% were selected, with exclusion of pre-
cursor ions with charge states of z = 1 or 8 or unassigned charge
Tissue and cell samples were lysed in radioimmunoprecipitation states.
assay buffer (50 mM Tris–HCl, 150 mM NaCl, 1% NP-40, 0.5% so-
For phosphoproteomics of colon cancer tissues, the enriched
dium deoxycholate, protease inhibitor, phosphatase inhibitor, pH =
phosphopeptides were analyzed by an EASY-nanoLC 1200 system
7.5). Tissue samples were homogenized with a gentleMACS Dis-
coupled to a Q Exactive HF-X mass spectrometer. The flow rate of the
sociator (Miltenyi Biotec GmbH) using the procedure “protein 01. 01”. analytical column was 330 nl/min with a 65 min gradient from 6% to
The crude lysates were sonicated (model JY92-IIN, 227.5 W, 3 s on
100% buffer B (80% ACN, 0.1% formic acid). For the full MS scans,
10 s off, 5 min) and then centrifuged at 20,000g and 4 ◦ C for 30 min.
ions with m/z ranging from 350 to 1600 were acquired under data-
The supernatants were collected and the protein concentrations were dependent acquisition mode by orbitrap with a resolution of 60,000
measured.
at m/z = 200. For the MS2 scans, the top 20 most intense parent ions
Peptide Digestion and TMT Labeling were selected with an isolation window of 0.6 m/z and fragmented
with stepped NCE values of 25% and 31%.
A tandem mass tag (TMT)-based strategy was used for quantitative For cellular phosphoproteomics analysis, the enriched phospho-
proteomics and phosphoproteomics analysis of tumors and distant peptides were desalted with ZipTip (Merck Millipore) and analyzed by
normal tissues (DNTs). For proteomics, 78 paired tumors and DNTs an EASY-nLC 1200 system coupled to a Q Exactive HF-X mass
were divided into 21 TMT-labeled batches with common reference spectrometer (Thermo Fisher Scientific). The flow rate of the analytical
samples. Fifty micrograms of protein lysates were reduced with column was 330 nl/min with a 90 min gradient from 12% to 100%
10 mM tris (2-carboxyethyl) phosphine (Sigma) at 56 ◦ C for 1 h and buffer B (80% ACN, 0.1% formic acid). For the full MS, ions with m/z
alkylated with 20 mM iodoacetamide in the dark at room temperature ranging from 350 to 1800 m/z were acquired by orbitrap resolution of
for 30 min. Subsequently, the protein lysates were precipitated with 60,000 at m/z = 200. For the MS2 scans, the top 20 most intense
methanol/chloroform/water and the precipitate was resuspended in parent ions were selected with an isolation window of 1.6 m/z and
50 mM triethylammonium bicarbonate (Sigma) and digested with MS fragmented with stepped NCE values of 25% and 27%. The maximum
sequencing grade trypsin (Promega, 1:50 enzyme-to-substrate ratio) injection time for MS2 was 64 ms and the dynamic exclusion time was

2 Mol Cell Proteomics (2023) 22(5) 100545


Clinical Significance and Functions of GSK3α in Colon Cancer

50 s. For in vitro phosphoproteomics analysis, the enriched phos- then the phosphoproteome data of 18 batches were combined. The
phopeptides were desalted with ZipTip (Merck Millipore) and analyzed phosphosites with missing values in less than 50% of the samples
by an EASY-nanoLC 1000 coupled to a Q Exactive Plus mass spec- were used for subsequent data analysis. Kaplan–Meier survival curves
trometer (Thermo Fisher Scientific). The flow rate of the analytical (log-rank test) and Cox proportional hazard models for OS analyses
column was 330 nl/min with a 65 min gradient from 10% to 90% buffer were carried out to identify the most important phosphosites. The filter
B (80% ACN, 0.1% formic acid). For the full MS, ions with m/z ranging criteria for important phosphosites were as follows: log-rank p value
from 350 to 1800 were acquired by an orbitrap resolution of 60,000 at <0.05, Cox p value <0.05, and HR > 1 or <1. The functions of phos-
m/z = 200. For the MS2 scans, the top 20 most intense parent ions phoproteins were analyzed by Metascape. For processing the output
were selected for MS2 analysis and fragmented with stepped NCE datasheets of phosphoproteome data in vitro or in cell lines, the
values of 25% and 27%. The maximum injection time for MS2 was contaminant and reverse hits were first removed. The phosphopep-
64 ms and the dynamic exclusion time was 50 s. tides with localization probabilities greater than 0.75 were assigned to
For analysis of coimmunoprecipitated proteins, the digested pep- reliable identification. The phosphosites with ratio (GSK3α/Control)
tides were desalted with ZipTip (Merck Millipore) and analyzed by an >1.5 and p value <0.05 (Student’s t test) or only detected in the
EASY-nanoLC 1000 coupled to a Q Exactive Plus mass spectrometer GSK3α-treated group and p value <0.05 (Student’s t test) were defined
(Thermo Fisher Scientific). Peptides were separated in the analytical as phosphosites significantly upregulated by GSK3α in vitro. Similarly,
column with a 65 min gradient from 10% to 90% buffer B (80% ACN, phosphosites with ratio (GSK3A-KO/WT) <0.667 and p value <0.05
0.1% formic acid) at a flow rate of 330 nl/min. For the full MS, the mass (Student’s t test) or only detected in the control group and p value
range was from 350 to 1800 m/z with a resolution of 60,000 at <0.05 (Student’s t test) were defined as phosphosites significantly
200 m/z. The AGC value was set at 3e6, with a maximum fill time of regulated by GSK3α in the cell lines. The functions of phosphoproteins
20 ms. For the MS/MS scans, the top 20 most intense parent ions were analyzed by Metascape.
were selected with an isolation window of 1.6 m/z and fragmented For processing the output table of coimmunoprecipitation prote-
with an NCE of 27%. The AGC was set at 1e6, with a maximum fill time ome data, the contaminant and reverse hits were first removed. Pro-
of 64 ms and a resolution of 17,500. teins with ≥2 unique peptides were selected for data analysis. Based
on the label-free quantification intensities, the quantified proteins with
MS Data Searching ratio (GSK3α/control) >1.5 or ratio (GSK3β/control) >1.5 (Student’s
The proteomics data were analyzed with MaxQuant (version 1.6, t test, p < 0.05) or only identified in the GSK3α or GSK3β over-
https://www.maxquant.org) and searched against the Swiss-Prot hu- expression groups (p < 0.05, Student’s t test) were supposed to be the
man protein sequence database (20,431 protein sequences, updated binding proteins of GSK3α or GSK3β. For the binding proteins of
on 2019/07/16). The proteolytic enzyme was trypsin/p and two missed THRAP3 mutants, the proteins with foldchanges >1.5 (Student’s t test,
cleavage sites were allowed. Oxidation of methionine and acetylation p < 0.05) or exclusively identified in that group were considered the
of the protein N terminus was set as variable modifications. Cysteine binding proteins of the corresponding THRAP3 mutant. The functions
carbamidomethylation was set as a fixed modification. The maximum of binding proteins were analyzed by Metascape.
peptide mass was set to 12,000 Da and the minimum amino acid
length was set to 6. The first search mass tolerance was set to 20 ppm Cell Lines and Cell Culture
and the main search peptide tolerance was set to 4.5 ppm. The mass
The HEK293T and human colon cancer cell lines HCT116 and RKO
tolerance for fragment ions was set at 0.5 Da. The phosphoproteomics
were grown in Dulbecco’s modified Eagle’s medium and the DLD-
data were searched with the same database and similar parameters.
1 cell line was grown in RPMI-1640 medium. The medium was sup-
Phosphorylation at S, T, or Y residues was also set as a variable
plemented with 10% fetal bovine serum (FBS) (FSP500, ExCell Bio),
modification. For label-free phosphoproteomics, the “match between
100 U/ml penicillin and 100 μg/ml streptomycin (15,140–122, Gibco).
runs” was enabled.

Bioinformatics and Statistical Analysis Western Blotting Analysis


For processing the output datasheets of proteome data of colon The protein samples were separated by 10% or 12% SDS-PAGE and
cancer tissues, the contaminant and reverse hits were first removed. The transferred to polyvinylidene fluoride membranes (ISEQ00010, Milli-
proteins with two or more unique peptides were kept. The total protein pore). After blocking with 4% nonfat milk, the polyvinylidene fluoride
intensity of each sample as well as the reference sample was then membranes were incubated with primary antibody (1:2000) at 4 ◦ C
normalized in each batch. The protein intensity of the sample was divided overnight and then incubated with HRP secondary antibody (1:10,000)
by that of the reference sample in the same batch and then the proteome at room temperature for 1 h. Antibodies against the following proteins
data of 21 batches were combined. The proteins with missing values in were used: GAPDH (60004-1-Ig, Proteintech), GSK3α (R1312–1,
less than 50% of the samples were used for subsequent data analysis. HUABIO), GSK3β (ET1607–71, HUABIO), Flag (66008-4-Ig, Pro-
Kaplan–Meier survival curves (log-rank test) and the Cox proportional teintech), Myc (R1208–1, HUABIO), THRAP3 (19744-1-AP, Pro-
hazard model for overall survival (OS) analyses were implemented to teintech), E-cadherin (ET1607–75, HUABIO), N-cadherin (22018-1-AP,
identify the most important kinases. The above analyses were achieved Proteintech), and vimentin (10366-1-AP, Proteintech).
through the “survival” and “survminer” packages in R. The filter criteria
for kinase biomarkers were as follows: log-rank p value <0.05, Cox p Measurement of Kinase Activity
value <0.05 (for samples in all stages) or <0.1 (for samples in each tumor-
node-metastasis [TNM] stage), and HR ≥ 2 or ≤0.5. Human GSK3A (1–483 aa) and GSK3B (1–420 aa) were cloned into
For processing the output datasheets of phosphoproteome data of the pGEX-6P1 vector and transduced into Escherichia coli BL21-Gold
colon cancer tissues, the contaminant and reverse hits were first (DE3) cells. The recombinant proteins GST-GSK3α and GST-GSK3β
removed. The phosphopeptides with localization probabilities greater were extracted by lysis buffer A (50 mM Tris–HCl, 150 mM NaCl,
than 0.75 were assigned to reliable identification. The total phos- 2.5 mM DTT, 1 mM EGTA, 1 mM EDTA, 1 mM PMSF, 25% glycerol,
phopeptide intensity of each sample in the same batch was normal- pH = 7.5), and lysis buffer B (50 mM Na3PO4, 200 mM NaCl 2 mM
ized to the same level. The phosphopeptide intensity of the sample DTT, 1 mM PMSF, 25% glycerol, pH = 7.0), respectively. The re-
was divided by that of the reference sample in the same batch and combinant proteins were purified by GST affinity resin (GE Healthcare).

Mol Cell Proteomics (2023) 22(5) 100545 3


Clinical Significance and Functions of GSK3α in Colon Cancer

The kinase activity was measured by a Kinase-Lumi assay Kit THRAP3S253A-antisense: 5′ -CAGACTGGAGGGGTGCTTTGAGAG
(S0150M, Beyotime). Five micrograms of extracted HCT116 protein as CTGGGCT-3′
the substrate was mixed with 0, 0.01, 0.02, 0.05, 0.1, 0.2, 0.5, and THRAP3S253D-sense: 5′ -AGCCCAGCTCTCAAAGATCCCCTCC
1.0 μg of recombinant GST-GSK3α protein. The mixture was added to AGTCTGTG-3′
the kinase reaction buffer (20 mM Tris–HCl, 10 mM MgCl2, 5 mM DTT, THRAP3S253D-antisense: 5′ -CACAGACTGGAGGGGATCTTTGAGA
25 μM ATP, pH = 7.5) and incubated at 30 ◦ C for 0, 1, 2, or 3 h. The GCTGGGCT-3′
reaction products were mixed with the Kinase-Lumi detection reagent THRAP3S248/253A-sense: 5′ -AGCGAGCACCAGCTCTCAAAGCA
and the fluorescence was detected by Multikinetic microplate reader CCCCTCCA-3′
(Biotech). THRAP3S248/253A-antisense: 5′ -TGGAGGGGTGCTTTGAGAGCTGG
TGCTCGCT-3′
In Vitro Kinase Assays THRAP3S248/253D-sense: 5′ -AGCGAGATCCAGCTCTCAAAGAT
CCCCTCCA-3′
Two hundred micrograms of extracted proteins from HCT116 cells THRAP3S248/253D-antisense: 5′ -TGGAGGGGATCTTTGAGAGCT
were mixed with the recombinant GST-GSK3α protein and kinase GGATCTCGCT-3′
reaction buffer (20 mM Tris–HCl, 10 mM MgCl2, 5 mM DTT, and 25 μM
ATP, pH = 7.5) and incubated at 30 ◦ C for 2 h. A reaction without Colony Formation Assay
recombinant GST-GSK3α was used as a control. The experimental
For the colony formation assay, empty vector-transduced cells, and
and control assays were repeated three times. The phosphopeptides
GSK3A-KD (or KO) cells were seeded into 6-well plates and incubated
of each sample were enriched by Fe-NTA agarose beads and
for approximately 10 days. The cells were fixed with 4% para-
analyzed by LC-MS/MS.
formaldehyde at room temperature for 30 min and then stained with
1% (w/v) crystal violet. Colonies were imaged and counted by ImageJ
Generation of Stable Colon Cancer Cell Lines (https://imagej.net).
GSK3A- or THRAP3-knockdown (KD) HCT116 or DLD-1 cell lines Cell Migration Assay
were generated using a CRISPR/Cas9 system. There were two guide
RNAs targeting the human GSK3A gene and two gRNAs targeting the HCT116 cells (2 × 105) or DLD-1 cells (1 × 105) were plated in an
human THRAP3 gene. The HCT116 and DLD-1 cells after knockdown 8.0 μm, 24-well cell culture insert (353,097, Corning Life Science) in
of the target genes were screened by puromycin (ST551, Beyotime). 200 μl of medium containing 1% FBS serum, with medium containing
To obtain GSK3A-KO HCT116 cells, a limiting dilution strategy was 10% FBS at the bottom of the insert. The HCT116 and DLD-1 cells
used to obtain monoclonal cells. The gRNA sequence was as follows: were incubated for 24 h and 20 h, respectively. Then, the cells were
GSK3A-gRNA1-sense: 5′ -CACCGGGCGGAAAGGCATCTGTCG-3′ fixed with 4% paraformaldehyde at room temperature for 30 min and
GSK3A-gRNA1-antisense: 5′ -AAACCGACAGATGCCTTTCCG stained with 1% (w/v) crystal violet. The migrated cells were imaged
CCC-3′ and counted by ImageJ.
GSK3A-gRNA2-sense: 5′ -CACCGGCACTAGCTTCCCGCCGCC-3′
GSK3A-gRNA2-antisense: 5′ -AAACGGCGGCGGGAAGCTAGT Immunostaining Assays
GCC-3′ A colon cancer tissue microarray was obtained from the West China
THRAP3-gRNA1-sense: 5′ -CACCGCGAGAGAGAGATCGGCTTC-3′ Hospital Biobank of Sichuan University. Immunohistochemistry was
THRAP3-gRNA1-antisense: 5′ -AAACGAAGCCGATCTCTCTC performed using an anti-GSK3α antibody (R13121, HUABIO). The
TCGC-3′ expression level of GSK3α in each spot in the microarray was evalu-
THRAP3-gRNA2-sense: 5′ -CACCGCTCTCGTTCAAGGAAGCGC-3′ ated by Image-Pro Plus 6.0 software (https://www.meyerinst.com/
THRAP3-gRNA2-antisense: 5′ -AAACGCGCTTCCTTGAACGA- brand/mediacybernetics).
GAGC-3′
To reconstruct the GSK3A and THRAP3 mutants in GSK3A and Coimmunoprecipitation
THRAP3-KD HCT116 cell lines, respectively, we inserted the com-
plementary DNA into a vector (pCDH-CMV-MCS-EF1) and abrogated Cells were harvested 48 h after transfection in cold PBS and lysed
the binding of gRNAs by altering the protospacer adjacent motif (PAM) with lysis buffer (25 mM Tris–HCl, 150 mM NaCl, 1% NP-40, 5%
sequence with the following primers: glycerol and protease inhibitor, phosphatase inhibitor, pH = 7.4) on ice
GSK3A-PAM1-sense: 5′ -AGGCATCTGTCGGAGCCATGGGT-3′ for 30 min and centrifuged at 20,000g. GSK3α-Flag, GSK3β-Flag, and
GSK3A-PAM1-antisense: 5′ -ACCCATGGCTCCGACAGATGCCT-3′ Flag-THRAP3 were pulled down with anti-Flag magnetic beads (HY-
THRAP3-PAM2-sense: 5′ -TTCAAGGAAGCGCAGACTGAGTTCT K0207, MedChemExpress), and THRAP3 proteins were pulled down
AGGT-3′ with an anti-THRAP3 antibody (19744-1-AP, Proteintech). The inter-
THRAP3-PAM2-antisense: 5′ -ACCTAGAACTCAGTCTGCGCTT acting proteins were eluted by elution buffer (50 mM Tris–HCl, 2%
CCTTGAA-3 ′ SDS, 10% glycerol, and 1% β-mercaptoethanol, pH = 6.8) and heated
To construct the THRAP3 mutants, 6 paired primers were used and at 95 ◦ C for 5 min. The eluted products were analyzed by Western
designed as follows: blots or LC-MS/MS.
THRAP3S248A-sense: 5′ -AGTCCTCGGGAGCGAGCCCCAGCTCTC Experimental Design and Statistical Rationale
AAAA-3′
THRAP3S248A-antisense: 5′ -TTTTGAGAGCTGGGGCTCGCTCCC For the proteomics of colon cancer tissues, 156 samples were
GAGGACT-3′ divided into 21 batches with a common reference sample and 12
THRAP3S248D-sense: 5′ -AGTCCTCGGGAGCGAGACCCAGCTCTC quality control (QC) samples were used to monitor the stability of the
AAAA-3′ machine. For the phosphoproteomics of colon cancer tissues, 140
THRAP3S248D-antisense: 5′ -TTTTGAGAGCTGGGTCTCGCTCCCG samples were divided into 18 batches with a common reference
AGGACT-3′ sample. To ensure strict analysis, proteins quantified in more than
THRAP3S253A-sense: 5′ -AGCCCAGCTCTCAAAGCACCCCTCCAG 50% of samples with ≥2 unique peptides or phosphosites quantified
TCTGT-3′ in more than 50% of samples with localization probability ≥0.75 were

4 Mol Cell Proteomics (2023) 22(5) 100545


Clinical Significance and Functions of GSK3α in Colon Cancer

used for subsequent data analysis. The significance of differences cancer. Notably, in the tumors of colon cancer patients
between groups was calculated by the Wilcoxon test. The significance grouped into high-GSK3α based on the best cutoff (Fig. 1B),
of survival analysis was calculated by the log-rank test and the best
the expression differences of GSK3α between tumors and
cutoff was applied. For phosphoproteomics of GSK3α in vitro or in cell
lines, each experiment had 3 replicates. The phosphosites that were DNTs were much more significant (Fig. 1C), although some
identified in at least 4 of 6 samples or only identified in at least 2 of 3 stages were no longer significant after TNM staging, probably
samples in the control or experimental group were used for difference due to the limited number of samples. The survival correlation
analysis by Student’s t test. For the identification of binding proteins of result was confirmed in the Clinical Proteomic Tumor Analysis
GSK3α-Flag or GSK3β-Flag, each experiment had 3 replicates. The
Consortium (CPTAC) cohort (Fig. 1D) and the Cancer Genome
proteins that were identified in at least 4 of 6 samples or only identified
in at least 2 of 3 samples in the control or experimental group with ≥2 Atlas (TCGA) cohort (Fig. 1E). Notably, no significant correla-
unique peptides were used for difference analysis by Student’s t test. tion was found between the levels of GSK3β in tumors and the
For the identification of binding proteins of THRAP3 mutants prognosis of colon cancer patients (Fig. 1F). Collectively,
(THRAP3S248A, THRAP3S248D, THRAP3S253A, and THRAP3S253D), each these results indicate that GSK3α is an important kinase in
experiment had 4 replicates. Proteins identified in more than 50% of
colon cancer and that high expression of GSK3α predicts
samples or only identified in the control or experimental group were
used for difference analysis by Student’s t test. unfavorable OS in colon cancer patients.

Further Validation of the Clinical Significance of GSK3α and


RESULTS Functional Studies
Kinome Analysis Reveals That GSK3α, but Not GSK3β, To further confirm the clinical significance of GSK3α, we next
Correlates With the OS of Colon Cancer Patients performed immunohistochemical straining of colon cancer tis-
Paired tumors and DNTs collected from 78 treatment-naive sue microarrays from another independent colon cancer cohort
colon cancer patients were subjected to TMT-based prote- using a specific anti-GSK3α antibody (supplemental Fig. S3, A
omics (supplemental Fig. S1A) (27). The 156 samples were and B and supplemental Table S2). As expected, high levels of
divided into 21 batches with a common reference sample and GSK3α were also significantly correlated with unfavorable
12 QC samples were used to monitor the stability of the ma- prognosis in colon cancer patients, whether in 111 tumors of
chine. The correlation coefficients of the 12 QC samples different stages or in tumors of stage II, III, and IV alone
ranged from 0.90 to 1.00 (supplemental Fig. S1B). In addition, (supplemental Fig. S3C). To reveal the functions of GSK3α,
correlation analysis of the common reference samples in silencing of GSK3A in DLD-1 and HCT116 colon cell lines was
different batches also indicated high reproducibility of the data carried out by a CRISPR/Cas9 system. The colony formation
acquisition (supplemental Fig. S1C). Principal component abilities (supplemental Fig. S4, A and B) and the migratory
analysis showed that these samples were well separated into abilities (Figs. 2A and S4C) of HCT116 and DLD-1 cells were
DNT and tumor groups (supplemental Fig. S1D), without an significantly reduced when GSK3A was silenced, while the
obvious batch effect (supplemental Fig. S1E). phenotypes were restored by re-expressing GSK3A (Figs. 2B
Next, we screened the kinases significantly correlated with and S4D). Immunoblotting detection of epithelial-mesenchymal
the OS of colon cancer patients. Out of the 5322 proteins with transition markers showed increased E-cadherin and reduced
2 or more unique peptides that were identified in more than vimentin and N-cadherin expression after GSK3A silencing
50% of samples, 158 kinases were identified. Based on (Figs. 2C and S4E). The expression of these markers could be
Kaplan–Meier survival analysis and Cox proportional hazards recovered by restoring GSK3α expression (Fig. 2D), consistent
regression modeling in all 78 tumors, the levels of 63 kinases with the observed cell phenotypes. Collectively, these results
were found to be significantly correlated with the prognosis of suggest that the high expression of GSK3α in tumors is
colon cancer patients. The clinical significance of the 63 ki- disastrous and may promote colon cancer malignancy.
nases was further analyzed separately in the tumors of each
Identification of In Vitro Phospho-Substrates Catalyzed by
TNM stage. As a result, out of the 63 kinases, 0 kinases in
GSK3α
stage I tumors, 25 kinases in stage II tumors, 32 kinases in
stage III tumors, and 4 kinases in stage IV tumors were found To clarify the downstream phospho-signals regulated by
to be significantly associated with the OS of colon cancer GSK3α, we next performed in vitro and in vivo phosphopro-
patients (supplemental Table S1). Overlapping analysis teomic analyses. Although a number of GSK3 phospho-
revealed that only GSK3α and MAP2K1 could predict the substrates have been reported (28), the substrates
prognosis of colon cancer patients in stages II-IV (Fig. 1A). specifically regulated by GSK3α remain unknown (29). To
However, the hazard ratios of MAP2K1 in stage II-IV patients identify the phospho-substrates of GSK3α in vitro, recombi-
were inconsistent (supplemental Fig. S2). In contrast, high nant hGSK3α was expressed in E. coli. expression system and
levels of GSK3α in tumors were significantly positively asso- purified. The in vitro activity of GST-GSK3α was tested by
ciated with poor prognosis in colon cancer, whether in 74 measuring the consumption of ATP, which indicated the good
tumors of different stages or in tumors of stage II, III, and IV enzymatic activity of recombinant GST-GSK3α (Fig. 3A). Then,
alone (Fig. 1B), indicating that GSK3α is a key kinase in colon the purified GST-GSK3α and ATP were incubated with the

Mol Cell Proteomics (2023) 22(5) 100545 5


Clinical Significance and Functions of GSK3α in Colon Cancer

FIG. 1. GSK3α is an important kinase significantly correlated with colon cancer prognosis. A, the workflow of screening the critical
kinases in colon cancer based on the proteome data. B, survival analysis of colon cancer patients with different levels of GSK3α in all tumors or
in tumors of different TNM stages (stage II, stage III, and stage IV) based on the proteome data. Of the 78 patients, GSK3α was detected in 74
patients. The p value was calculated by the log-rank test and the best cutoff was applied. C, relative GSK3α expression between tumors and
DNTs in the low-GSK3α and high-GSK3α subgroups and the relative levels of GSK3α in four TNM stages in the high-GSK3α subgroup based on
the proteome data. The low-GSK3α and high-GSK3α subgroups were divided based on the best cutoff of OS in Fig. 1B. The significance of
differences between different groups was calculated by the Wilcoxon test, *p < 0.05, ***p < 0.001. D and E, survival analysis of colon cancer
patients with different GSK3α expression levels in tumors based on the proteome data of the CPTAC cohort (D) and TCGA cohort (E). The p value
was calculated by the log-rank test and the best cutoff was selected. F, survival analysis of colon cancer patients with different levels of GSK3β
in all tumors or in tumors of different TNM stages (stage II, stage III, and stage IV) based on the proteome data. The p value was calculated by the
log-rank test and the best cutoff was applied. OS, overall survival.

extracted whole cell lysate. After tryptic digestion, the phos- phosphosites increased by 1.5-fold from 843 proteins in
phopeptides were enriched with Fe-NTA agarose beads and response to GST-GSK3α treatment (localization probability of
analyzed by MS (Fig. 3B). In total, we identified 1236 ≥0.75 and Student’s t test, p < 0.05. 451 phosphosites only

6 Mol Cell Proteomics (2023) 22(5) 100545


Clinical Significance and Functions of GSK3α in Colon Cancer

FIG. 2. Effects of GSK3α on cell phenotypes. A and B, representative images of HCT116 colon cancer cell line migration after GSK3A-KO (A)
and GSK3A re-expression (B). The scale bar represents 100 μm. Each assay was repeated three times. The significance of differences between
different groups was calculated by paired t test, *p < 0.05, **p < 0.01. C and D, immunoblots of GSK3α, E-cadherin, N-cadherin, and vimentin in
HCT116 cells in response to GSK3A-KO (C) or re-expression (D).

identified upon GSK3α treatment were not displayed in the Identification of GSK3α-Mediated Phospho-Substrates in
volcano plot.) (Fig. 3, C and D and supplemental Table S3). The Colon Cancer Cell Lines
distribution analysis showed that most phosphosites regulated
In vitro phosphorylation assays may not represent in vivo
by GSK3α in vitro mainly occurred on serine residues (Fig. 3E).
regulation. Therefore, by using WT and GSK3A-KO colon
Motif analysis of GSK3α-mediated phosphosites showed that
cancer cell lines, we screened phospho-substrates exclusively
proline residues were preferred at the +1 and +2 positions
regulated by GSK3α in cell lines with three biological repeats.
(Fig. 3F). Approximately 13% of phosphosites might be primed
Principal component analysis showed that the WT and
by serine phosphorylation at the +4 position (Fig. 3G) (29). In
GSK3A-KO samples could be clearly separated based on the
addition, based on a reported algorithm (30), global phos-
phosphoproteome data (Fig. 4A). A total of 9501 phosphosites
phorylation state change (ΔPs) analysis of phosphoproteins
(localization probability ≥0.75) on 3541 proteins were identi-
was performed. The phosphorylation state of 441 proteins,
fied, of which 8069 phosphosites on 3255 proteins were
including SRRM2, THRAP3, MKI67, BCLAF1, and AHNAK,
quantified (Fig. 4B). Compared with the GSK3A-KO group, a
was significantly increased upon GSK3α treatment (hyper-
total of 988 phosphosites were increased by 1.5-fold in the
phosphorylated, ΔPs > 1.0) (Fig. 3H).
WT group (Student’s t test, p < 0.05) (Fig. 4C and
To compare the phospho-substrates of GSK3α with those
supplemental Table S4), including 225 phosphosites only
of GSK3β in vitro, we then carried out the kinase reaction with
identified in the WT group that were not displayed in the vol-
active recombinant GST-GSK3β in vitro in duplicate
cano plot. Similar to the in vitro results, serine modifications
(supplemental Fig. S5A). Pearson’s correlation coefficient of
accounted for the vast majority (Fig. 4D). By analyzing the ΔPs
the phosphoproteome between the two repeats was 0.683
of phosphoproteins, we identified 420 proteins with signifi-
(p < 0.0001) (supplemental Fig. S5B). A total of 571 phos-
cantly downregulated total phosphorylation levels in the
phosites, which had ratio (GSK3β/Control) > 1.5 in duplicate or
GSK3A-KO group (Fig. 4E). Overlapping analysis identified
were only identified in both GSK3β-treated groups, were the
161 GSK3α-mediated phosphosites in vitro and in cell lines, of
most reliable phospho-substrates of GSK3β in vitro
which 156 were specifically regulated by GSK3α but not
(supplemental Fig. S5C and supplemental Table S3). Motif
GSK3β (Fig. 4F). The 156 phosphosites belonged to 130
analysis of GSK3β-mediated phosphosites showed that
proteins. Further biological process and molecular function
GSK3β preferred to phosphorylate the threonine residues
analyses of the 130 phosphoproteins showed that RNA
compared with GSK3α (supplemental Figs. S5D and 3F).
binding, protein binding, cadherin binding, RNA splicing,
Moreover, proline residues were preferred at the +1 positions
mRNA processing, DNA repair, and transcriptional regulation
of the phosphosites and some phosphorylation events were
were most enriched (Fig. 4G). Collectively, we reveal 156
also primed by serine phosphorylation at the +4 position
phosphosites specifically regulated by GSK3α.
(supplemental Fig. S5D). Unexpectedly, only 49 phosphosites
were common substrates of GSK3α and GSK3β in vitro,
The Clinical Significance of GSK3α-Mediated
indicating the huge difference between GSK3α and GSK3β as
Phosphorylation in Colon Cancer
kinases (supplemental Fig. S5E). Collectively, these in vitro
enzymatic results suggest significant functional differences To analyze the clinical significance of 156 phosphosites
between GSK3β and GSK3α as kinases. specifically regulated by GSK3α. We analyzed the

Mol Cell Proteomics (2023) 22(5) 100545 7


Clinical Significance and Functions of GSK3α in Colon Cancer

FIG. 3. Profiling in vitro phospho-substrates of GSK3α. A, measurement of recombinant GSK3α activity. Equal amounts of protein extracts
were mixed with different amounts of recombinant GSK3α and the remaining amount of ATP in the reaction system was detected at different
reaction times to measure GSK3α activity. B, flowchart of quantitative in vitro phosphoproteomics for GSK3α. Recombinant GST-GSK3α was
added to the experimental group, while blank buffer was added to the control group. Each group had three repeats. C, numbers of identified and
quantified phosphosites and their corresponding proteins in the in vitro phosphoproteome of GSK3α. D, Venn diagram depicting the identified
phosphosites in the GSK3α-treated group and control group, and the volcano plot indicates the expression differences of overlapping phos-
phosites. The dots in red and blue indicate the significantly changed phosphosites with foldchanges greater than 1.5 (Student’s t test, p < 0.05).
The phosphosites that are identified in at least 4 of 6 samples or only identified in at least 2 of 3 samples in the control or experimental group are
included in the statistics. The phosphosites that were increased 1.5-fold or exclusively identified in the GSK3α-treated group are suggested to
be GSK3α-regulated phosphosites in vitro. E, distribution of GSK3α-regulated phosphosites in vitro. F, motif analysis of GSK3α-regulated
phosphosites in vitro using iceLogo. G, frequency of amino acids at the +4 position of the GSK3α-regulated phosphosites. H, global ΔPs
analysis of phosphoproteins using in vitro phosphoproteome data of GSK3α. ΔPs means the phosphorylation change value for an individual
protein that is calculated by the sum of log2-transformed foldchange of all significantly changed phosphosites between the experimental and
control groups (Student’s t test, p < 0.05). Blue represents ΔPs < −1, and red represents ΔPs >1. PCA, Principal component analysis.

phosphoproteome data of 70 colon cancer patients (27). phosphoproteins, and localization analysis of 10 phosphopro-
Overlapping analysis led to 34 GSK3α-regulated phosphosites teins showed that these proteins mainly localized in the nucleus
that were identified in more than 50% of tumors (Fig. 5A). We (Fig. 5D) and were involved in DNA repair, DNA binding, RNA
found that 11 out of the 34 phosphosites, such as THRAP3S682p binding, ATP binding, apoptotic process, and positive regula-
and THRAP3S248p, were increased in tumors (ratio [T/DNT] >1.2 tion of transcription by RNA polymerase II (Fig. 5C). Survival
and Wilcoxon test, p < 0.05) (Fig. 5B and supplemental analysis showed that high levels of 5 phosphosites, including
Table S5). The 11 phosphosites were derived from 10 HSF1S303p, CANXS583p, MCM2S41p, POGZS425p, and

8 Mol Cell Proteomics (2023) 22(5) 100545


Clinical Significance and Functions of GSK3α in Colon Cancer

FIG. 4. Profiling the cellular phospho-substrates of GSK3α. A, PCA of the control and experimental groups based on the phosphopro-
teome data. The control and experimental groups were WT and GSK3A-KO HCT116 cell lines, respectively. Each group had three repeats. B,
numbers of identified and quantified phosphosites and their corresponding phosphoproteins in the phosphoproteomes of GSK3A-KO and WT
cell lines. C, Venn diagram depicting the identified phosphosites in the GSK3A-KO and WT groups, and the volcano plot indicates the
expression differences of overlapping phosphosites between the GSK3A-KO and WT groups. Red and blue dots indicate the significantly
changed phosphosites with foldchanges greater than 1.5 (Student’s t test, p < 0.05). The phosphosites that are identified in at least 4 of 6
samples or only identified in at least 2 of 3 samples in the GSK3A-KO or WT group are included in the statistics. The phosphosites that were
increased 1.5-fold or exclusively identified in the WT group are suggested as GSK3α-regulated phosphosites in cells. D, distribution of GSK3α-
regulated phosphosites in cells. E, global ΔPs analysis of phosphoproteins using the phosphoproteome data of GSK3A-KO and WT cell lines.
ΔPs indicates the phosphorylation change value for an individual protein that is calculated by the sum of the log2-transformed foldchange of all
significantly changed phosphosites between the GSK3A-KO and WT groups (Student’s t test, p < 0.05). Blue represents ΔPs < −1, and red
represents ΔPs >1. F, overlap analysis of phosphosites regulated by GSK3α in vitro and by GSK3α in cell lines and by GSK3β in vitro. G,
heatmap (left) showing the 156 overlapping phosphosites and bubble chart (right) showing the enriched pathways using the corresponding 130
phosphoproteins. The filled color of the circle represents the p value and the size of the circle indicates the fold enrichment of each pathway.

Mol Cell Proteomics (2023) 22(5) 100545 9


Clinical Significance and Functions of GSK3α in Colon Cancer

FIG. 5. The clinical significance of GSK3α-regulated phospho-substrates in colon cancer. A, overlap analysis of GSK3α-regulated
phosphosites and the phosphosites identified in more than 50% of colon cancer patients. B, volcano plot displaying the changes in 34 over-
lapping phosphosites between the tumors and DNTs. The size of the circle indicates the p value (Wilcoxon test), and the filled color of the circle
indicates the ratio(tumor/DNT). C, chord diagrams showing the top 3 Gene Ontology Molecular Function (GO–MF) and Gene Ontology Biological
Process (GO–BP) pathways enriched using the 10 phosphoproteins corresponding to the 11 phosphosites. D, the cellular localization of the 11
phosphosites significantly upregulated in tumors. E, forest plot showing the effect of 6 phosphosites on the prognosis of colon cancer patients.
The p values were calculated using the Cox proportional hazard model. DNT, distant normal tissue.

SRRM2T983p, were significantly correlated with poor prognosis, 156 phosphosites specifically regulated by GSK3α, 9 phos-
while high levels of PRPF4BS431p predicted good prognosis phosites were derived from THRAP3, BCLAF1, and STAU1.
(Fig. 5E). Among the 9 phosphosites, 4 sites, including THRAP3S248p,
THRAP3S682p, BCLAF1S512p, and BCLAF1S531p, were also
The Potential Roles of THRAP3S248 Phosphorylation identified in the phosphoproteomics analysis of paired tissues
We speculated that of the 156 phosphosites specifically from 70 colon cancer patients (supplemental Table S5 and
regulated by GSK3αand their corresponding proteins might Fig. 5A). However, none of them showed a significant corre-
have different binding affinities to GSK3α. To identify the lation with OS in our cohort of 70 colon cancer patients.
protein substrates most tightly bound to GSK3α, we overex- Interestingly, of the 4 phosphosites, the levels of THRAP3S248p
pressed GSK3A-Flag and GSK3B-Flag vectors in the 293T and BCLAF1S531p were significantly correlated with OS in
cell line (Fig. 6, A–C) and performed pull-down experiments another colon cancer cohort termed the mCRC cohort (32)
with an anti-Flag antibody (31). The binding proteins of GSK3α (Fig. 6, G and H), in which more metastatic colon cancer pa-
and GSK3β were analyzed by MS (Fig. 6D). As a result, 23 tients were included (Fig. 6I).
proteins specifically bound to GSK3α but not GSK3β (Fig. 6E THRAP3 is a protein with multiple phosphorylation sites. Of
and supplemental Table S6). Of the 23 proteins, 3 proteins, the 18 identified phosphosites in this work, phosphorylation at
including THRAP3, BCLAF1, and STAU1, were phosphopro- S248, S253, and S682 was specifically mediated by GSK3α
teins specifically regulated by GSK3α (Fig. 6F). Notably, of the in vitro and in cell lines (Fig. 6J). Because THRAP3S248p but

10 Mol Cell Proteomics (2023) 22(5) 100545


Clinical Significance and Functions of GSK3α in Colon Cancer

FIG. 6. GSK3α but not GSK3β specifically binds to THRAP3. A–C, Western blotting analysis of the overexpression of the bait proteins
GSK3α-Flag and GSK3β-Flag. D, flowchart for the identification of GSK3α- and GSK3β-interacting proteins by affinity purification and MS

Mol Cell Proteomics (2023) 22(5) 100545 11


Clinical Significance and Functions of GSK3α in Colon Cancer

FIG. 7. THRAP3S248p increases cancer cell migration and its binding affinity to proteins related to DNA damage repair. A, Western
blotting analysis of the THRAP3 levels in THRAP3-KD HCT116 cells. B, Western blotting analysis of the levels of THRAP3S248A, THRAP3S248D,
THRAP3S253A, and THRAP3S253D after overexpressing the corresponding vectors. C, representative images of the migration assay using
THRAP3-KD HCT116 cells after overexpressing THRAP3S248A and THRAP3S248D. The scale bar represents 100 μm. Each assay was repeated
three times. The p value was calculated using paired t test. **p < 0.01. D, thirty-seven differentially bound proteins between THRAP3S248A and
THRAP3S248D mutants and their subcellular localization (left). The enriched pathways of the 37 proteins by GO analysis are shown on the right.

not BCLAF1S531p was significantly increased in tumors performed. The results showed that GSK3α but not GSK3β
compared with DNTs (Fig. 5B), the function of THRAP3S248p could be pulled down by THRAP3 (Fig. 6, L and M). To exclude
was further investigated. Based on the evidence from the the influence of the tag on the pull-down results, we used an
mCRC cohort (32), we assumed that THRAP3S248p might play anti-THRAP3 antibody to enrich endogenous GSK3. As ex-
a role in the process of tumor metastasis. To reveal the pected, GSK3α, but not GSK3β, was specifically enriched by
function of THRAP3S248p, the binding of THRAP3 to GSK3α the anti-THRAP3 antibody (Fig. 6N).
was first confirmed by immunoblotting (Fig. 6K). Retro-pull- Because the S248 and S253 residues of THRAP3 are very
down of GSK3α and GSK3β by THRAP3 was also close in the sequence, the function of THRAP3S253p in colon

analysis. E, Venn diagram showing the overlap between GSK3α-binding proteins (ratio (GSK3α/Control) >1.5, Student’s t test, p < 0.05) and
GSK3β-binding proteins (ratio (GSK3β/Control) >1.5, Student’s t test, p < 0.05). F, Venn diagram showing the overlap between the 23 GSK3α-
specific binding proteins and the 130 proteins containing the 156 phosphosites specifically regulated by GSK3α. G and H, survival analysis of
colon cancer patients based on the levels of THRAP3S248p (G) and BCLAF1S531p (H) using the phosphoproteome data of the mCRC cohort. I,
statistical analysis of colon cancer patients in different TNM stages in our cohort and mCRC cohort. J, eighteen phosphosites on THRAP3
identified in vitro and in the HCT116 cell line. The red balls indicate the phosphosites on THRAP3 upregulated after adding GSK3α in vitro. The
blue balls indicate the decreased phosphosites on THRAP3 after knocking out GSK3A. The length of the vertical line indicates the relative
foldchange. The longer the line is, the greater the change. The dark yellow outer circle indicates significant changes at that site (Student’s t test,
p < 0.05). K, immunoblots of the binding proteins of GSK3α-myc using anti-THRAP3 antibody. L–N, immunoblots of the binding proteins of Flag-
THRAP3 using anti-GSK3α (L) and anti-GSK3β (M) antibodies. N, immunoblots of THRAP3 coimmunoprecipitated proteins using anti-GSK3α
and anti-GSK3β antibodies. THRAP3 was enriched using an anti-THRAP3 antibody. Enrichment by IgG was used as a control. CRC, Colorectal
cancer.

12 Mol Cell Proteomics (2023) 22(5) 100545


Clinical Significance and Functions of GSK3α in Colon Cancer

cancer was also investigated for comparison. To disclose the GSK3α as a kinase but also suggests GSK3α as a promising
functions of THRAP3S248p and THRAP3S253p in colon cancer, therapeutic target for colon cancer.
we silenced the THRAP3 gene in the HCT116 cell line GSK3α and GSK3β have almost identical sequences in the
(Fig. 7A) and then overexpressed two pairs of THRAP3 mu- kinase domain but are distinct at the C terminus and N ter-
tants, Flag-THRAP3S248A and Flag-THRAP3S248D, as well as minus. All available inhibitors are known to inhibit both iso-
Flag-THRAP3S253A and Flag-THRAP3S253D, in the THRAP3- forms but many studies erroneously attribute the inhibitory
KD HCT116 cell line (Fig. 7B). Of these mutants, Flag- effect to specific inhibition of GSK3β while ignoring effects on
THRAP3S248A and Flag-THRAP3S253A mimicked the effect of GSK3α (33). In addition, clinical trials of dual GSK3α/β in-
nonphosphorylation, and Flag-THRAP3S248D and Flag- hibitors have not proven successful (4). We showed that
THRAP3S253D mimicked the effect of phosphorylation. As a GSK3α and GSK3β differ greatly in their binding proteins
result, we did not observe a significant difference in clono- (Fig. 6E) and phospho-substrates (Fig. 4F) in colon cancer
genic growth capability between cancer cell lines over- cells, although we cannot exclude the possibility that the
expressing THRAP3S248A and THRAP3S248D mutants phospho-substrates of GSK3α and GSK3β may differ in
(supplemental Fig. S6A). However, compared with different cell types. GSK3α was more closely associated with
THRAP3S248A, THRAP3S248D mimicking phosphorylation was colon cancer progression and unfavorable outcomes than
able to enhance the migration of HCT116 cells (Fig. 7C), GSK3β (Figs. 1, A and B and 1F). Therefore, we assume that
consistent with our assumption that THRAP3S248p is closely specific inhibition or targeted degradation of GSK3α by
linked to tumor metastasis. It should be mentioned that both proteolysis-targeting chimeras may be an effective strategy
THRAP3S253D and THRAP3S248/253D had no effect on cancer for the treatment of colon cancer (34, 35).
cell migration (supplemental Fig. S6, B–D). A number of known GSK3 phospho-substrates have been
We next performed pull-down experiments to identify the more accurately identified as substrates of GSK3β or GSK3α.
THRAP3S248p-binding proteins and identified 37 proteins that For example, we have confirmed that DYPSL2T509p (36–38),
tended to bind to THRAP3S248D and 0 proteins that tended to UNGT60p (39), PHF6S155p (29), and RPS14T133p (29) are com-
bind to THRAP3S248A (Fig. 7D and supplemental Table S7). mon substrates of GSK3α and GSK3β. FOXK1S428p (40) and
Enrichment analysis showed that these proteins were involved PKMT328p (41) are specific substrates of GSK3α and GSK3β,
in many biological pathways, such as double-strand break respectively. In addition, we also obtained some results that
repair (p = 1.19E-10) and cadherin binding (p = 7.85E-03) are inconsistent with previous reports. Some phosphosites,
(Fig. 7D). In contrast, 17 and 25 proteins preferred to bind to such as DPYSL3T509p (38), MAPRE1S155p (42), BCLAF1S531p
THRAP3S253D and THRAP3S253A, respectively (supplemental (43), and RPTORS859p (44), are GSK3α-specific substrates
Fig. S6E and supplemental Table S7). The functions of based on our work but these phosphosites were incorrectly
THRAP3S253D- and THRAP3S253A-binding proteins were assigned to GSK3β substrates in the PhosphoSitePlus data-
different from those of THRAP3S248D-binding proteins base (www.phosphosite.org). The reason for these results
(supplemental Fig. S6E). Collectively, the above results show may be that the investigators used GSK3 inhibitors but clas-
the potential roles of THRAP3S248p in regulating colon cancer sified the affected phosphosites as substrates of GSK3β. In
cell migration and DDR. addition, we also found that some phosphosites previously
clearly reported as substrates of GSK3β can also be regulated
by GSK3α in vitro, such as SRFS224p (45) and SOX9T236p (46).
DISCUSSION
The different cell types used may also be an important reason
In summary, we investigated the kinase functions of GSK3α for these results. Collectively, these results indicate that
and its specific phospho-substrates in colon cancer. Several clarification of the substrate specificity of GSK3α and GSK3β
new achievements have been gained. First, we demonstrated may help to obtain the correct conclusions.
that high expression of GSK3α, but not GSK3β, was signifi- Comparing the in vitro phosphoproteome data (Fig. 3C) to
cantly correlated with unfavorable outcomes in colon cancer the cell line phosphoproteome data (Fig. 4B), it was observed
patients from four independent cohorts. Second, we identified that many more phosphosites and phosphoproteins were
156 phospho-substrates specifically regulated by GSK3α, but identified in the cell lines, indicating that the phosphorylation
not GSK3β, in vitro and in colon cancer cell lines. Furthermore, events are more complex in cells than in vitro. Moreover,
a number of phospho-substrates of GSK3α have been iden- secondary phosphorylation reactions in the cells can further
tified in tissues from colon cancer patients and some of them increase the complexity. For example, some identified phos-
show a significant correlation with the OS of colon cancer phoproteins, which are substrates of GSK3α, are also kinases,
patients. Third, we investigated the potential roles of such as CDK1, CDK11B, and MAPK1. GSK3α-mediated
THRAP3S248p, a phosphosite regulated by GSK3α, in colon phosphorylation of these kinases may further phosphorylate
cancer and showed that THRAP3S248p may function in the downstream protein substrates, which are substrates of
regulation of colon cancer cell migration and DDR. Collec- downstream kinases rather than direct substrates of GSK3α.
tively, this work not only discloses the specific function of Notably, the number of phosphosites regulated by GSK3α

Mol Cell Proteomics (2023) 22(5) 100545 13


Clinical Significance and Functions of GSK3α in Colon Cancer

was greater in the in vitro enzymatic assays (Fig. 4F). Previous the dataset identifiers PXD038081 (the phosphoproteome
studies have suggested that kinases may be less specific data of the 70 paired colon cancer tissues) and PXD03883
in vitro than in vivo (47), which could lead to more differential 0 (other omics data except the phosphoproteome data of
phosphorylation reactions in vitro, consistent with our the 70 paired colon cancer tissues).The annotated spectra has
observations. been deposited to the MS-Viewer with the unique search key
THRAP3, also called TRAP150 and BCLAF2, is a member of for each batch of colon cancer proteomic data provided in the
the BRCA1-interacting RNA splicing complex involved in supplemental Table S1, colon cancer phosphoproteomic data
promoting pre-mRNA splicing and subsequent transcript provided in the supplemental Table S5, in vitro phosphopro-
stability in response to DNA damage (48, 49). THRAP3 defi- teomic data of GSK3α, and GSK3β provided in the
ciency or site mutations increase sensitivity to DNA damaging supplemental Table S3, in cell lines phosphoproteomic data of
agents (50), which induce ATR-associated phosphorylation on GSK3α provided in the supplemental Table S4, coimmuno-
multiple serine residues of THRAP3, such as S210, S211, precipitation proteomic data of GSK3α and GSK3β provided in
S399, S406, and S408 (51). THRAP3 also regulates mRNA the supplemental Table S6, and coimmunoprecipitation pro-
export to the cytoplasm and many of the transcripts exported teomic data of THRAP3 mutant (THRAP3S248A and
by THRAP3 are associated with DDR (50). In this work, we THRAP3S248D, THRAP3S253A and THRAP3S253D) provided in
showed that THRAP3S248p, THRAP3S253p, and THRAP3S682p the supplemental Table S7.
are GSK3α-specific phosphosites, and THRAP3S248p may also
be linked to the DDR process (Fig. 7D). In addition, we also Supplemental data — This article contains supplemental
revealed the roles of THRAP3S248p in regulating cancer cell data.
migration. Although THRAP3S248p and THRAP3S253p are very
close in sequence, no synergistic effect in regulating cancer Acknowledgments — This work was supported by the Na-
cell migration was observed (supplemental Fig. S6D). tional Key R&D Program of China (No. 2018YFC2000305 and
In addition to THRAP3, we also identified many novel 2022YFA1303200), National Natural Science Foundation of
phosphosites specifically regulated by GSK3α. However, most China (No. 82073221, 31870826, and 82073246), National
of their functions are unknown. For example, BCLAF1 is a Clinical Research Center for Geriatrics of West China Hospital
BCL-2-associated transcriptional repressor and can induce (No. Z20201002 and Z2021JC005) and West China Hospital
cell apoptosis. SRSF10 is a key regulator of BCLAF1 pre- 135 project (No. ZYYC20007, ZYYC20003, ZYGD20006, and
mRNA splicing. High SRSF10 expression in CRC samples 2016105).
increases the inclusion of BCLAF1 exon 5a, which is associ-
ated with higher CRC grade (52). Phosphorylation is critical to Author contributions — L. D. methodology; H.-N. C., Y. L., S.
regulate the functions of BCLAF1. BCLAF1S290p is also a H., S. L., H. X., Z.-G. Z., and L. D. supervision; L. D. writing-
known phosphosite involved in the regulation of the DNA original draft; L. G., Y. L., M. H., R. Z., X. W., Z. X., Y. G., R.
damage response (53). DNA-PKC-mediated phosphorylation W., D. D., and D. S. investigation; H.-N. C., Z. L., Y. L., S. H., S.
of BCLAF1Y150 and BCLAF1S151 is located in an RS domain L., H. X., Z.-G. Z. resources.
and drives BCLAF1 to the nuclear envelope (54). GSK3 has
been predicted to be the kinase of BCLAF1 but it is not clear Conflict of Interest — There is no competing interest.
whether GSK3α or GSK3β plays a role (43). Moreover, it is also
unclear which sites on BCLAF1 are regulated by GSK3. Here, Abbreviations — The abbreviations used are: ACN, acetoni-
we confirmed that GSK3α can regulate BCLAF1 phosphory- trile; AGC, automatic gain control; cDNA,
lation and identified five unambiguous phosphosites mediated complementary DNA; CRC, Colorectal cancer; DDR, DNA
by GSK3α, which may help to clarify the functions of these damage repair; DNT, distant normal tissue; KD, knockdown;
phosphosites. NCE, normalized collision energy; OS, overall survival; PAM,
protospacer adjacent motif; QC, quality control; RIPA, radio-
There are still some limitations in this work. First, the roles of
immunoprecipitation assay; TMT, tandem mass tag; TNM,
GSK3α-regulated phosphosites remain to be disclosed,
tumor-node-metastasis.
especially those whose levels are significantly correlated with
the OS of colon cancer patients. Second, the question of how Received December 17, 2022, and in revised form, March 30, 2023
THRAP3S248p responds to DNA damage via its binding pro- Published, MCPRO Papers in Press, April 8, 2023, https://doi.org/
10.1016/j.mcpro.2023.100545
teins or binding nucleic acids requires further investigation.

REFERENCES
DATA AVAILABILITY
1. Mauri, G., Vitiello, P. P., Sogari, A., Crisafulli, G., Sartore-Bianchi, A., Mar-
All MS raw data and the output tables have been deposited soni, S., et al. (2022) Liquid biopsies to monitor and direct cancer treat-
ment in colorectal cancer. Br. J. Cancer 127, 394–407
to the ProteomeXchange Consortium (http://proteomecentral. 2. Wei, Q., Ye, Z., Zhong, X., Li, L., Wang, C., Myers, R. E., et al. (2017)
proteomexchange.org) via the iProX partner repository, with Multiregion whole-exome sequencing of matched primary and metastatic

14 Mol Cell Proteomics (2023) 22(5) 100545


Clinical Significance and Functions of GSK3α in Colon Cancer

tumors revealed genomic heterogeneity and suggested polyclonal atherosclerosis and hepatic steatosis in high fat diet-fed low density li-
seeding in colorectal cancer metastasis. Ann. Oncol. 28, 2135–2141 poprotein receptor-deficient mice. Am. J. Pathol. 184, 3394–3404
3. Xie, Y.-H., Chen, Y.-X., and Fang, J.-Y. (2020) Comprehensive review of tar- 24. Draffin, J. E., Sánchez-Castillo, C., Fernández-Rodrigo, A., Sánchez-Sáez,
geted therapy for colorectal cancer. Signal. Transduct. Target. Ther. 5, 22 X., Ávila, J., Wagner, F. F., et al. (2021) GSK3α, not GSK3β, drives hip-
4. Kingwell, K. (2018) Flipping the switch for selective GSK3 inhibition. Nat. pocampal NMDAR-dependent LTD via tau-mediated spine anchoring.
Rev. Drug Discov. 17, 314 EMBO J. 40, e105513
5. Liu, J., Xiao, Q., Xiao, J., Niu, C., Li, Y., Zhang, X., et al. (2022) Wnt/β-catenin 25. Gong, Y. Q., Liu, Y., Wang, T., Li, Z. G., Gao, L., Chen, H. N., et al. (2021)
signalling: function, biological mechanisms, and therapeutic opportu- Age-associated proteomic signatures and potential clinically actionable
nities. Signal. Transduct. Target Ther. 7, 3 targets of colorectal cancer. Mol. Cell Proteomics 20, 100115
6. Zhou, W., Wang, L., Gou, S. M., Wang, T. L., Zhang, M., Liu, T., et al. (2012) 26. Vasaikar, S., Huang, C., Wang, X., Petyuk, V. A., Savage, S. R., Wen, B.,
ShRNA silencing glycogen synthase kinase-3 beta inhibits tumor growth et al. (2019) Proteogenomic analysis of human colon cancer reveals new
and angiogenesis in pancreatic cancer. Cancer Lett. 316, 178–186 therapeutic opportunities. Cell 177, 1035–1049.e19
7. Farago, M., Dominguez, I., Landesman-Bollag, E., Xu, X., Rosner, A., Car- 27. Zhang, R., Hu, M., Chen, H. N., Wang, X., Xia, Z., Liu, Y., et al. (2023)
diff, R. D., et al. (2005) Kinase-inactive glycogen synthase kinase 3beta Phenotypic heterogeneity analysis of APC-mutant colon cancer by pro-
promotes Wnt signaling and mammary tumorigenesis. Cancer Res. 65, teomics and phosphoproteomics identifies RAI14 as a key prognostic
5792–5801 determinant in East Asians and Westerners. Mol. Cell Proteomics 225,
8. Liu, C., Li, Y., Semenov, M., Han, C., Baeg, G.-H., Tan, Y., et al. (2002) 100532
Control of β-catenin phosphorylation/degradation by a dual-kinase 28. McCubrey, J. A., Steelman, L. S., Bertrand, F. E., Davis, N. M., Sokolosky,
mechanism. Cell 108, 837–847 M., Abrams, S. L., et al. (2014) GSK-3 as potential target for therapeutic
9. Inoki, K., Ouyang, H., Zhu, T., Lindvall, C., Wang, Y., Zhang, X., et al. (2006) intervention in cancer. Oncotarget 5, 2881–2911
TSC2 integrates Wnt and energy signals via a coordinated phosphory- 29. Shinde, M. Y., Sidoli, S., Kulej, K., Mallory, M. J., Radens, C. M., Reicherter,
lation by AMPK and GSK3 to regulate cell growth. Cell 126, 955–968 A. L., et al. (2017) Phosphoproteomics reveals that glycogen synthase
10. Fuentealba, L. C., Eivers, E., Ikeda, A., Hurtado, C., Kuroda, H., Pera, E. M., kinase-3 phosphorylates multiple splicing factors and is associated with
et al. (2007) Integrating patterning signals: Wnt/GSK3 regulates the alternative splicing. J. Biol. Chem. 292, 18240–18255
duration of the BMP/Smad1 signal. Cell 131, 980–993 30. Wang, Z., Ma, J., Miyoshi, C., Li, Y., Sato, M., Ogawa, Y., et al. (2018)
11. Trnski, D., Sabol, M., Gojević, A., Martinić, M., Ozretić, P., Musani, V., et al. Quantitative phosphoproteomic analysis of the molecular substrates of
(2015) GSK3β and Gli3 play a role in activation of Hedgehog-Gli pathway sleep need. Nature 558, 435–439
in human colon cancer — targeting GSK3β downregulates the signaling 31. Ino, Y., Arakawa, N., Ishiguro, H., Uemura, H., Kubota, Y., Hirano, H., et al.
pathway and reduces cell proliferation. Biochim. Biophys. Acta 1852, (2016) Phosphoproteome analysis demonstrates the potential role of
2574–2584 THRAP3 phosphorylation in androgen-independent prostate cancer cell
12. Chen, Y., Yue, S., Xie, L., Pu, X. H., Jin, T., and Cheng, S. Y. (2011) Dual growth. Proteomics 16, 1069–1078
Phosphorylation of suppressor of fused (Sufu) by PKA and GSK3beta 32. Li, C., Sun, Y. D., Yu, G. Y., Cui, J. R., Lou, Z., Zhang, H., et al. (2020) In-
regulates its stability and localization in the primary cilium. J. Biol. Chem. tegrated omics of metastatic colorectal cancer. Cancer cell 38, 734–747.
286, 13502–13511 e739
13. Diehl, J. A., Cheng, M., Roussel, M. F., and Sherr, C. J. (1998) Glycogen 33. Beurel, E., Grieco, S. F., and Jope, R. S. (2015) Glycogen synthase
synthase kinase-3beta regulates cyclin D1 proteolysis and subcellular kinase-3 (GSK3): regulation, actions, and diseases. Pharmacol. Ther.
localization. Genes Dev. 12, 3499–3511 148, 114–131
14. Zhou, B. P., Deng, J., Xia, W., Xu, J., Li, Y. M., Gunduz, M., et al. (2004) Dual 34. Qu, L., Li, S., Ji, L., Luo, S., Ding, M., Yin, F., et al. (2021) Discovery of PT-65
regulation of Snail by GSK-3beta-mediated phosphorylation in control of as a highly potent and selective Proteolysis-targeting chimera degrader
epithelial-mesenchymal transition. Nat. Cell Biol. 6, 931–940 of GSK3 for treating Alzheimer's disease. Eur. J. Med. Chem. 226,
15. Takada, Y., Fang, X., Jamaluddin, M. S., Boyd, D. D., and Aggarwal, B. B. 113889
(2004) Genetic deletion of glycogen synthase kinase-3beta abrogates 35. Békés, M., Langley, D. R., and Crews, C. M. (2022) PROTAC targeted
activation of IkappaBalpha kinase, JNK, Akt, and p44/p42 MAPK but protein degraders: the past is prologue. Nat. Rev. Drug Discov. 21, 181–
potentiates apoptosis induced by tumor necrosis factor. J. Biol. Chem. 200
279, 39541–39554 36. Yoshimura, T., Kawano, Y., Arimura, N., Kawabata, S., Kikuchi, A., and
16. Foltz, D. R., Santiago, M. C., Berechid, B. E., and Nye, J. S. (2002) Glycogen Kaibuchi, K. (2005) GSK-3beta regulates phosphorylation of CRMP-2 and
synthase kinase-3beta modulates notch signaling and stability. Curr. Biol. neuronal polarity. Cell 120, 137–149
12, 1006–1011 37. Liu, F., Laguesse, S., Legastelois, R., Morisot, N., Ben Hamida, S., and
17. Cohen, P., and Frame, S. (2001) The renaissance of GSK3. Nat. Rev. Mol. Ron, D. (2017) mTORC1-dependent translation of collapsin response
Cell Biol. 2, 769–776 mediator protein-2 drives neuroadaptations underlying excessive
18. Azoulay-Alfaguter, I., Yaffe, Y., Licht-Murava, A., Urbanska, M., Jaworski, J., alcohol-drinking behaviors. Mol. Psych. 22, 89–101
Pietrokovski, S., et al. (2011) Distinct molecular regulation of glycogen 38. Cole, A. R., Causeret, F., Yadirgi, G., Hastie, C. J., McLauchlan, H.,
synthase kinase-3alpha isozyme controlled by its N-terminal region: McManus, E. J., et al. (2006) Distinct priming kinases contribute to dif-
functional role in calcium/calpain signaling. J. Biol. Chem. 286, 13470– ferential regulation of collapsin response mediator proteins by glycogen
13480 synthase kinase-3 in vivo. J. Biol. Chem. 281, 16591–16598
19. Matsuda, T., Zhai, P., Maejima, Y., Hong, C., Gao, S., Tian, B., et al. (2008) 39. Baehr, C. A., Huntoon, C. J., Hoang, S. M., Jerde, C. R., and Karnitz, L.
Distinct roles of GSK-3alpha and GSK-3beta phosphorylation in the heart M. (2016) Glycogen synthase kinase 3 (GSK-3)-mediated phosphory-
under pressure overload. Proc. Natl. Acad. Sci. U. S. A. 105, 20900– lation of uracil N-glycosylase 2 (UNG2) facilitates the repair of
20905 floxuridine-induced DNA lesions and promotes cell survival. J. Biol.
20. McCubrey, J. A., Steelman, L. S., Bertrand, F. E., Davis, N. M., Abrams, S. Chem. 291, 26875–26885
L., Montalto, G., et al. (2014) Multifaceted roles of GSK-3 and Wnt/ 40. He, L., Gomes, A. P., Wang, X., Yoon, S. O., Lee, G., Nagiec, M. J., et al.
β-catenin in hematopoiesis and leukemogenesis: opportunities for ther- (2018) mTORC1 promotes metabolic reprogramming by the suppression
apeutic intervention. Leukemia 28, 15–33 of GSK3-dependent Foxk1 phosphorylation. Mol. Cell 70, 949–960.e944
21. Zhou, J., Freeman, T. A., Ahmad, F., Shang, X., Mangano, E., Gao, E., et al. 41. Xu, Q., Tu, J., Dou, C., Zhang, J., Yang, L., Liu, X., et al. (2017) HSP90
(2013) GSK-3α is a central regulator of age-related pathologies in mice. J. promotes cell glycolysis, proliferation and inhibits apoptosis by regulating
Clin. Invest. 123, 1821–1832 PKM2 abundance via Thr-328 phosphorylation in hepatocellular carci-
22. Chen, X., Wang, R., Liu, X., Wu, Y., Zhou, T., Yang, Y., et al. (2017) noma. Mol. Cancer 16, 178
A chemical-genetic approach reveals the distinct roles of GSK3α and 42. Berges, R., Denicolai, E., Tchoghandjian, A., Baeza-Kallee, N., Honore, S.,
GSK3β in regulating embryonic stem cell fate. Dev. Cell 43, 563–576.e564 Figarella-Branger, D., et al. (2018) Proscillaridin A exerts anti-tumor ef-
23. Banko, N. S., McAlpine, C. S., Venegas-Pino, D. E., Raja, P., Shi, Y., Khan, fects through GSK3β activation and alteration of microtubule dynamics in
M. I., et al. (2014) Glycogen synthase kinase 3α deficiency attenuates glioblastoma. Cell Death Dis. 9, 984

Mol Cell Proteomics (2023) 22(5) 100545 15


Clinical Significance and Functions of GSK3α in Colon Cancer

43. Linding, R., Jensen, L. J., Ostheimer, G. J., van Vugt, M. A., Jørgensen, C., 49. Meng, X., Yang, S., and Camp, V. J. A. (2020) The interplay between the
Miron, I. M., et al. (2007) Systematic discovery of in vivo phosphorylation DNA damage response, RNA processing and extracellular vesicles. Front.
networks. Cell 129, 1415–1426 Oncol. 9, 1538
44. Stretton, C., Hoffmann, T. M., Munson, M. J., Prescott, A., Taylor, P. M., 50. Vohhodina, J., Barros, E. M., Savage, A. L., Liberante, F. G., Manti, L.,
Ganley, I. G., et al. (2015) GSK3-mediated raptor phosphorylation sup- Bankhead, P., et al. (2017) The RNA processing factors THRAP3 and
ports amino-acid-dependent mTORC1-directed signalling. Biochem. J. BCLAF1 promote the DNA damage response through selective mRNA
470, 207–221 splicing and nuclear export. Nucl. Acids Res. 45, 12816–12833
45. Li, C. L., Sathyamurthy, A., Oldenborg, A., Tank, D., and Ramanan, N. 51. Beli, P., Lukashchuk, N., Wagner, S. A., Weinert, B. T., Olsen, J. V., Baskcomb,
(2014) SRF phosphorylation by glycogen synthase kinase-3 pro- L., et al. (2012) Proteomic investigations reveal a role for RNA processing
motes axon growth in hippocampal neurons. J. Neurosci. 34, 4027– factor THRAP3 in the DNA damage response. Mol. Cell 46, 212–225
4042 52. Zhou, X., Li, X., Cheng, Y., Wu, W., Xie, Z., Xi, Q., et al. (2014) BCLAF1 and
46. Hong, X., Liu, W., Song, R., Shah, J. J., Feng, X., Tsang, C. K., et al. (2016) its splicing regulator SRSF10 regulate the tumorigenic potential of colon
SOX9 is targeted for proteasomal degradation by the E3 ligase FBW7 in cancer cells. Nat. Commun. 5, 4581
response to DNA damage. Nucl. Acids Res. 44, 8855–8869 53. Liu, J., Li, J., Sun, Z., Duan, Y., Wang, F., Wei, G., et al. (2021) Bcl-2-
47. Olsen, J. V., Blagoev, B., Gnad, F., Macek, B., Kumar, C., Mortensen, P., associated transcription factor 1 Ser290 phosphorylation mediates DNA
et al. (2006) Global, in vivo, and site-specific phosphorylation dynamics in damage response and regulates radiosensitivity in gastric cancer. J.
signaling networks. Cell 127, 635–648 Transl. Med. 19, 339
48. Savage, K. I., Gorski, J. J., Barros, E. M., Irwin, G. W., Manti, L., Powell, A. 54. Lee, Y. Y., Yu, Y. B., Gunawardena, H. P., Xie, L., and Chen, X. (2012)
J., et al. (2014) Identification of a BRCA1-mRNA splicing complex BCLAF1 is a radiation-induced H2AX-interacting partner involved in
required for efficient DNA repair and maintenance of genomic stability. γH2AX-mediated regulation of apoptosis and DNA repair. Cell Death Dis.
Mol. Cell 54, 445–459 3, e359

16 Mol Cell Proteomics (2023) 22(5) 100545

You might also like