1 title page - theranostics

42
1 / 42 Title page 1 Full title: PLAGL2 promotes the proliferation and migration of gastric cancer cells via USP37- 2 mediated deubiquitination of Snail1 3 List of authors: Liang Wu 1 *, Ning Zhao 1 *, Zili Zhou 1 , Jinhuang Chen 2 , Shengbo Han 1 , Xudan 4 Zhang 1 , Haijun Bao 1 , Wenzheng Yuan 3 and Xiaogang Shu 1 5 *These authors contributed equally to this work. 6 Liang Wu: [email protected]; Ning Zhao: [email protected]; 7 Zili Zhou: [email protected]; Jinhuang Chen: [email protected]; 8 Shengbo Han: [email protected]; Xudan Zhang: [email protected]; 9 Haijun Bao: [email protected]; Wenzheng Yuan: [email protected]; 10 Xiaogang Shu: [email protected]. 11 12 Authors’ institutional affiliations: 13 1 Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong 14 University of Science and Technology, Jiefang Road No. 1277, Wuhan 430022, China. 15 2 Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong 16 University of Science and Technology, Jiefang Road No. 1277, Wuhan 430022, China. 17 3 Department of Gastrointestinal Surgery II, Renmin Hospital of Wuhan University, Zhang Zhidong 18 Road No. 99 Wuhan 430060, China. 19 20 Corresponding author: 21 Name: Xiaogang Shu 22

Upload: others

Post on 15-Nov-2021

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: 1 Title page - Theranostics

1 / 42

Title page 1

Full title: PLAGL2 promotes the proliferation and migration of gastric cancer cells via USP37-2

mediated deubiquitination of Snail1 3

List of authors: Liang Wu1*, Ning Zhao1*, Zili Zhou1, Jinhuang Chen2, Shengbo Han1, Xudan 4

Zhang1, Haijun Bao1, Wenzheng Yuan3 and Xiaogang Shu1 5

*These authors contributed equally to this work. 6

Liang Wu: [email protected]; Ning Zhao: [email protected]; 7

Zili Zhou: [email protected]; Jinhuang Chen: [email protected]; 8

Shengbo Han: [email protected]; Xudan Zhang: [email protected]; 9

Haijun Bao: [email protected]; Wenzheng Yuan: [email protected]; 10

Xiaogang Shu: [email protected]. 11

12

Authors’ institutional affiliations: 13

1 Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong 14

University of Science and Technology, Jiefang Road No. 1277, Wuhan 430022, China. 15

2 Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong 16

University of Science and Technology, Jiefang Road No. 1277, Wuhan 430022, China. 17

3 Department of Gastrointestinal Surgery II, Renmin Hospital of Wuhan University, Zhang Zhidong 18

Road No. 99 Wuhan 430060, China. 19

20

Corresponding author: 21

Name: Xiaogang Shu 22

Page 2: 1 Title page - Theranostics

2 / 42

E-mail: [email protected] 23

Phone: +8613971537678 24

Fax: 027-85726935 25

Address: Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, 26

Huazhong University of Science and Technology, Jiefang Road No. 1277, Wuhan, Hubei Province, 27

China. 28

29

30

31

32

33

34

35

36

37

38

39

40

41

42

43

44

Page 3: 1 Title page - Theranostics

3 / 42

Abstract 45

Rationale: PLAGL2 (pleomorphic adenoma gene like-2), a zinc finger PLAG transcription 46

factor, is aberrantly expressed in several malignant tumors. However, the biological roles of 47

PLAGL2 and its underlying mechanism in gastric cancer (GC) remain unclear. Methods: A series 48

of experiments in vitro and in vivo were conducted to reveal the role of PLAGL2 in GC progression. 49

Results: The data revealed that PLAGL2 promotes GC cell proliferation, migration, invasion, and 50

EMT in vitro and in vivo. Mechanistically, we demonstrated the critical role of PLAGL2 in the 51

stabilization of snail family transcriptional repressor 1 (Snail1) and promoting Snail1-mediated 52

proliferation and migration of GC cells. PLAGL2 activated the transcription of deubiquitinase 53

USP37, which then interacted with and deubiquitinated Snail1 protein directly. In addition, GSK-54

3β-dependent phosphorylation of Snail1 protein is essential for USP37-mediated Snail1 55

deubiquitination regulation. Conclusions: In general, PLAGL2 promotes the proliferation and 56

migration of GC cells through USP37-mediated deubiquitination of Snail1 protein. This work 57

provided potential therapeutic targets for GC treatment. 58

59

60

Key words: PLAGL2, USP37, Snail1, GC, deubiquitination 61

62

63

64

65

66

Page 4: 1 Title page - Theranostics

4 / 42

Introduction 67

GC has been estimated to be the fifth most frequently diagnosed cancer, and the third leading 68

cause of cancer death worldwide in 2018, with about 1,000,000 new cases and 783,000 deaths [1]. 69

In particular, nearly more than half of the new cases occur in East Asia [1]. Despite significant 70

advances in the diagnosis and treatment of GC in the past decade, GC survival rates have not 71

increased significantly. Metastasis, accounting for up to 90% of cancer-related deaths, is still the 72

most incomprehensible part of cancer progression [2]. However, little is known about the detailed 73

molecular mechanisms involved in GC tumorigenesis and metastasis. 74

PLAGL2 is a highly similar homologue to PLAG1 structurally and functionally. The PLAG 75

transcription factors specifically bind to the GRGGC(N)6-8RGGK consensus sequences in the 76

target genes promoter to regulate its transcription [3]. We previously clarified that PLAGL2 is 77

implicated in the pathogenesis of Hirschsprung disease and colorectal cancer [4,5]. PLAGL2 plays 78

a carcinogenic role and has been reported in salivary gland tumors [6], colorectal cancer [7], and 79

lung adenocarcinoma [8]. PLAGL2 promotes the epithelial-mesenchymal transition (EMT) and 80

mediates colorectal cancer metastasis by β-catenin-dependent regulation of ZEB1[5]. PLAGL2 also 81

enhances Wnt6 expression at the transcriptional level and promotes the progression of colorectal 82

cancer [9]. In addition, the PLAGL2-EGFR-HIF-1/2α signalling loop induces the malignant 83

phenotype of liver cancer and the chemotherapy resistance of erlotinib [10]. Although growing 84

evidence has showed that PLAGL2 functions as a dominant oncogene in gastrointestinal cancers 85

[7,11,12], the exact role of PLAGL2 and the underlying mechanism in GC remain largely unknown. 86

Metastasis is a major contributor to the mortality of cancer patients. Accumulating evidence 87

indicates that the EMT initiates the metastatic progression of tumors [13]. Therefore, a more 88

Page 5: 1 Title page - Theranostics

5 / 42

comprehensive understanding of the EMT molecular mechanism will provide important insights to 89

overcome cancer metastasis. It is well known that Snail1 plays a central role in inducing EMT. 90

Snail1 triggers EMT by directly repressing E-cadherin expression and activating mesenchymal gene 91

expression, such as fibronectin. Consistent with its function, Snail1 expression in human tumors is 92

positively correlated with the tumor grade and enhanced invasiveness and metastasis [14,15]. 93

Therefore, it is critical to explore the exact molecular mechanisms, responsible for the aberrant 94

expression of Snail1 in cancer. 95

Snail1 is a highly unstable protein that is subjected to the proteasome-ubiquitination 96

degradation pathway. The E3-ubiquitin ligases transfer ubiquitin (Ub) from an E2 ubiquitin-97

conjugating enzyme to the substrate proteins with a lysine residue or a growing Ub chain. Multiple 98

E3 ligases, including Skp-Culin-Fbox(SCF) E3 ligases [16,17], ubiquitin specific peptidase 99

27X(USP27X) [18], USP1 [19], USP37 [20,21], deubiquitinase3 (DUB3) [22] and A20 [23], 100

modulates Snail1 stability in various biological contexts. Snail1 proteasome-ubiquitination 101

degradation, which is enhanced by β-TrCP1/FBXW1, requires phosphorylation of Snail1 by GSK3β 102

[24]. Although Snail1 functions have been well explored during tumor progression and metastasis, 103

it remains unknown how Snail1 avoids ubiquitination and degradation in GC. 104

In this study, we demonstrated that PLAGL2 promoted the proliferation and migration of GC 105

cells via USP37-mediated ubiquitination of Snail1 and revealed the precise regulatory mechanism 106

of PLAGL2-mediated Snail1 stabilization. We clarified the enhanced PLAGL2 expression in GC 107

tissues and elucidated the crucial effects of PLAGL2 on the proliferation, migration, and invasion 108

of GC cells. Besides, we showed that PLAGL2 directly activates USP37 expression, which targets 109

Snail1 to proteasome-ubiquitination degradation. More importantly, the identification of such a 110

Page 6: 1 Title page - Theranostics

6 / 42

PLAGL2-USP37-Snail1 axis that regulates Snail1 ubiquitination and determination of the detailed 111

mechanism will provide a potential novel therapeutic strategy for GC tumorigenesis and metastasis. 112

113

114

115

116

117

118

119

120

121

122

123

124

125

126

127

128

129

130

131

132

Page 7: 1 Title page - Theranostics

7 / 42

Methods 133

Patients and specimens 134

In total, 49 pairs of GC primary specimens were gathered from patients who had not been 135

treated with radiotherapy or chemotherapy before the surgery. Two pathologists confirmed the 136

diagnosis of GC in each case. This study was following the Declaration of Helsinki and approved 137

by the Ethics Committee of Tongji Medical College, Huazhong University of Science and 138

Technology. 139

140

Cell culture and reagents 141

The GC cell lines SGC7901, BGC803, AGS, MKN45, BGC823, MGC803, HGC27 and the 142

gastric mucosal epithelial cell lines GES-1 were obtained from American Type Culture Collection 143

(ATCC, Manassas, VA, USA). SGC7901, AGS, BGC823, MKN45, BGC803, MGC803, HGC27, 144

and GES-1 cells were cultured in RPMI-1640 medium (Gibco, Grand Island, NY, USA). All media 145

were supplemented with 10% fetal bovine serum (FBS), 100 U/mL penicillin (Sigma-Aldrich, St 146

Louis, MO, USA), and 100 mg/mL streptomycin (Sigma-Aldrich). Cycloheximide (CHX), 147

proteasome inhibitor MG132, chloroquine diphosphate (CQ), and GSK-3β inhibitor CHIR-99021 148

were bought from Selleck Chemicals (Houston, TX, USA). Alkaline phosphatase (CIP) was 149

obtained from Roche (Roche Applied Science, Indianapolis, IN, USA). The GSK-3β inhibitor LiCl 150

was obtained from Sigma Aldrich. 151

152

Western blotting (WB) and immunoprecipitation (IP) analysis 153

WB and IP analyses were performed as described previously [25]. Detailed antibody 154

Page 8: 1 Title page - Theranostics

8 / 42

information is provided in Table S1. 155

156

In vivo ubiquitination assay 157

The indicated plasmids or siRNAs were transiently transfected into HEK293T or GC cells. The 158

ubiquitination assay was performed as previously described [25]. 159

160

Quantitative real-time polymerase chain reaction (qRT-PCR) 161

Total RNA from cells or specimens was extracted using TRIzol reagent (TaKaRa, Kyoto, 162

Japan). According to the manufacturer's instructions, the qRT-PCR assay was performed using the 163

SYBR Green PCR kit (TaKaRa). The primer sequences are given in Table S2. 164

165

Cell proliferation assay 166

Cell proliferation assays were performed with CCK-8 reagents (Dojondo Laboratories, 167

Kumamoto, Japan) according to the manufacturer's instructions. A total of 2 × 103 cells were plated 168

into 96-well plates. Then, the cells were incubated with CCK8 for 2 h at scheduled time points. 169

170

Colony formation assay 171

GC cells were plated in 6-well plates and cultured in the corresponding medium for 2 weeks. 172

The culture medium was changed every 3 days. Cell colonies were stained with 1% crystal violet 173

and then counted. 174

175

Cell cycle ananlysis 176

Page 9: 1 Title page - Theranostics

9 / 42

For cell cycle analysis, cells were harvested and washed with cold PBS, and fixed with 75% 177

cold ethanol. Before analysis with a BD FACS flow cytometer, the cells were incubated with 178

propidium iodide (PI) (Invitrogen, Gaithersburg, MD, USA) for 30 min. 179

180

Cell migration and invasion assays 181

Wound-healing and Transwell assays were used to measure the migration and invasion of cells. 182

These assays were performed as previously described [26]. 183

184

Immunofluorescence (IF) assay 185

The IF assay was carried out, as described previously [25]. Primary antibodies specific for 186

USP37 (1:100) (Proteintech, Rosemont, IL, USA) and Snail1 (1:100) (Santa Cruz Biotechnology, 187

Dallas, Texas, USA) were applied. Fluorescence images were acquired. 188

189

Immunohistochemistry (IHC) 190

IHC analysis was conducted as described elsewhere [27]. According to the staining area scores 191

and staining intensity, the quantification of IHC staining results was evaluated as previously 192

described [27]. 193

194

Transfection 195

Human Lenti-shPLAGL2-GFP (PLAGL2 shRNA 5′-GACCCATGATCCTAACAAA-3′, 196

Genechem, Shanghai, China), Lenti-PLAGL2-Flag (OBiO Technology, Shanghai, China) and their 197

controls were utilized in our study. To establish stable cell lines, 5 × 104 cells were seeded in 6-well 198

Page 10: 1 Title page - Theranostics

10 / 42

plates. After lentivirus infection, single-cell clonal isolates were maintained in a culture medium 199

with 5 μg/mL puromycin (Sigma-Aldrich) for 2 weeks. The transfection efficiency was determined 200

by WB and qRT-PCR analyses. USP38, DUSP18, His-USP37, His-USP37-C350S, Flag-Ubiquitin, 201

MYC-Ubiquitin, HA-Snail1, USP37 promoter (wild-type, mut1, mut2 and mut3), different deletion 202

mutants of HA-Snail1, wild-type Snail1 (Snail1-WT) and Snail1 mutant Snail1-6SA were cloned 203

into the pcDNA3 vector. Short interfering RNAs (siRNAs) for Snail1 (siSnail1), USP37 (siUSP37), 204

GSK-3β(siGSK-3β) and their controls were synthesized by RiboBio (Guangzhou, China). The 205

targeted sequences were as follows: siSnail1, 5′-CCTAGTCAGCCACCTTTAA-3′, siUSP37#1, 5′-206

CAGACACTATGGAAACTGA-3′, siUSP37#2, 5′-AAGCGTGGTTTACTTACAA-3′ and siGSK-207

3β, 5′-GGAGTGCACA TATGGCAAT-3′. Transfections of plasmids and siRNAs were performed 208

using Lipofectamine 3000 (Invitrogen). Effective transfection was confirmed by WB. 209

210

Chromatin immunoprecipitation (ChIP) 211

The ChIP assay was carried out using the ChIP kit (Cell Signaling Technology, Danvers, MA, 212

USA). All experimental procedures were performed following the kit instructions. The ChIP DNA 213

was amplified by qRT-PCR; the products of qRT-PCR were electrophoresed on a 2% agarose gel. 214

The primers for the USP37 promoter region sequence were as follows: forward primer (5’-215

GAAACTGTTGGTCAGCGCAA-3’), reverse primer (5’-CTCCTAATTCCCGGTGTCGG-3’). 216

Measurement of promoter-reporter activity. 217

GC cells and HEK293T cells were transfected with the designated USP37 promoter reporters 218

combined with the PLAGL2 siRNA or PLAGL2 plasmid. The cotransfection of the β-actin/Renilla 219

luciferase reporter gene was used to normalize USP37 promoter activity. According to the 220

Page 11: 1 Title page - Theranostics

11 / 42

manufacturer's instructions, the luciferase activity was quantified using a dual luciferase assay 221

system (Promega, Madison, WI, USA). 222

223

Xenograft assay 224

The mouse experiments were approved by the Institutional Animal Care and Use Committee 225

of Tongji Medical College, Huazhong University of Science and Technology. BALB/c male nude 226

mice (5 weeks old) were purchased from Beijing Vital River (Beijing, China). For the tumorigenesis 227

assay, 6×106 cells in 200 μL of PBS, were implanted subcutaneously into the groin of the mice (6 228

mice for each group). Tumor size was calculated with the following formula: V= 0.5× (length) × 229

(width)2. All mice were sacrificed 4 weeks after inoculation. For metastasis assay, 4×106 cells 230

suspended in 150 μL of PBS, were injected into the tail veins of the mice (7 mice for each group), 231

and all mice were killed after 6 weeks. The metastatic nodules were counted by two observers and 232

then confirmed by hematoxylin-eosin (HE) staining. 233

234

Statistical analysis 235

All in vitro experiments were performed three times or more. The results are presented as the 236

mean ± SD. The statistical analyses were completed by GraphPad Prism 8 or SPSS 20.0 software. 237

The data analysis was conducted using Student’s t-test for comparisons between groups. The χ2 test 238

was utilized to evaluate the association between the target genes expression and clinicopathological 239

features. Pearson's correlation test assessed correlation analysis. Differences were considered 240

significant at * p<0.05, ** p<0.01, and *** p<0.001. n.s: no significance. 241

242

Page 12: 1 Title page - Theranostics

12 / 42

Results 243

PLAGL2 is highly expressed in GC 244

To clarify the role of PLAGL2 in GC tumorigenesis, we first examined the PLAGL2 expression 245

profiles in clinical GC specimens and human GC cell lines. PLAGL2 expression in most GC cell 246

lines was markedly higher than its expression in gastric mucosal epithelial cell line GES-1(Figure 247

1A). Significantly, higher levels of PLAGL2 mRNA and protein were observed in primary GC 248

tumors than in matched paraneoplastic tissues (Figure 1B-D), which is consistent with the data 249

obtained from the Oncomine database [28] (Figure 1E) and GEPIA database [29] (Figure 1F). IHC 250

analysis further showed that PLAGL2 was highly expressed in most GC samples with distant 251

metastasis than those without distant metastasis (Figure 1G). Associations between PLAGL2 252

expression and clinicopathological parameters are shown in Table 1. Notably, high PLAGL2 253

expression was significantly linked to tumor invasion depth (P = 0.033) and lymph node metastasis 254

(P = 0.003). Data from the Kaplan-Meier Plotter database [30] showed that patients with high 255

PLAGL2 levels displayed poor overall survival (Figure 1H). Overall, these results suggest that 256

PLAGL2 is highly expressed in GC patients and is related to poor prognosis. 257

258

PLAGL2 promotes the proliferation, migration, and invasion of GC cells 259

To explore the oncogenic role of PLAGL2 in GC, we established stable PLAGL2 knockdown 260

(SGC7901-shRNA) and overexpression (AGS-PLAGL2) cells. Transfection efficiency was 261

confirmed by WB and qRT–PCR (Figure 2A and Figure S1A). The CCK-8 assay demonstrated that 262

silencing PLAGL2 significantly inhibited cell proliferation, whereas enforced PLAGL2 expression 263

enhanced the growth of GC cells (Figure 2B). Additionally, colony formation assays indicated that 264

Page 13: 1 Title page - Theranostics

13 / 42

silencing of PLAGL2 expression resulted in much smaller and fewer colonies, whereas higher 265

PLAGL2 expression promoted clonogenicity (Figure 2C). The cell cycle analysis results showed 266

that PLAGL2 decreased the G0G1 fraction and increased the S and G2M fractions (Figure S1B). 267

Additionally, stable PLAGL2 depletion decreased the expression levels of vital cell proliferation 268

regulatory proteins (cyclinD1, cyclin E, and CDK4), and enhanced p27kip1(a key cell cycle 269

inhibitor) expression (Figure 2D and Figure S1C). To extend the in vitro results, we explored the 270

effect of PLAGL2 on tumor growth using a xenograft model. The volume and weight of the 271

shPLAGL2 group were significantly reduced compared to the control. The volume and weight of 272

tumors formed by cells with higher PLAGL2 expression were dramatically increased than the 273

matched group (Figure 2E-F). Furthermore, the expression levels of cell cycle regulatory proteins 274

and p27kip1 in tumors formed by cells with stable PLAGL2 depletion are consistent with the results 275

of WB in PLAGL2 knockdown cells (Figure S1D-E). In general, these results indicate that PLAGL2 276

is a regulator of proliferation in GC. 277

Next, we explored the metastatic potential of GC cells with different PLAGL2 expression 278

levels. As shown in Figure 2G-J, PLAGL2 enhanced the migration and invasion of GC cells in vitro. 279

Given that matrix metalloproteinase (MMP) family proteins and EMT-related proteins participate 280

in tumor cell migration and invasion, we examined the transcript and protein level of MMP9, N-281

cadherin, Vimentin, and E-cadherin in cells with modified PLAGL2 expression levels. As shown in 282

Figure 2K and Figure S1C, SGC7901-shRNA cells displayed a higher expression level of E-283

cadherin and a lower level of N-cadherin, Vimentin, and MMP9 than SGC7901-NC cells. In contrast, 284

PLAGL2 overexpression in AGS cells triggered the expression of MMP9, N-cadherin and Vimentin, 285

accompanied by a concomitant decrease in E-cadherin. Furthermore, the EMT-related proteins were 286

Page 14: 1 Title page - Theranostics

14 / 42

investigated by IHC analysis (Figure S1D-E), which was conducted with mouse tumors formed by 287

the corresponding cells. An experimental lung metastasis assay was performed to assess the in vivo 288

role of PLAGL2 in tumor metastasis (Figure 2L-M). The lung weight and number of metastatic 289

nodules increased significantly in tumor xenografts that overexpress PLAGL2 (Figure 2N-O). 290

Together, these findings indicate the crucial role of PLAGL2 in GC metastasis and invasion. 291

292

PLAGL2 stabilizes Snail1 protein by inhibiting its ubiquitination 293

Snail1 plays a particularly crucial role in the proliferation and migration of GC cells, thus, we 294

examined whether PLAGL2 exerts oncogenic effects on the proliferation and migration of GC cells 295

in a Snail1-dependent manner. To examine the possible role of PLAGL2 in regulating Snail1 296

expression, we detected Snail1 protein levels in cells with different PLAGL2 expression levels. The 297

depletion of PLAGL2 significantly inhibited Snail1 protein expression in SGC7901 cells, and 298

enhanced PLAGL2 expression, which considerably elevated Snail1 protein levels in the AGS cells 299

(Figure 3A). Notably, no statistically significant changes in the Snail1 mRNA levels were observed 300

(Figure S2A), suggesting that PLAGL2 modulates Snail1 expression post-transcriptionally. 301

Similarly, ectopic PLAGL2 expression in HEK293T cells dramatically elevated exogenous Snail1 302

protein levels in a dose-dependent manner (Figure 3B). Besides, PLAGL2 expression levels were 303

positively correlated with Snail1 protein levels in GC samples (Figure 3C and Figure S2B). We 304

observed a linear correlation between PLAGL2 and Snail1 protein levels in clinical GC specimens, 305

and this linear correlation was not observed between PLAGL2 and Snail1 mRNA levels (Figure 3D-306

E). We speculate that PLAGL2 might regulate the degradation pathway of Snail1 protein, and that 307

the autophagolysosome pathway and ubiquitin-proteasome pathway are the main pathways for 308

Page 15: 1 Title page - Theranostics

15 / 42

protein degradation. We treated GC cells with ubiquitin-proteasome pathway inhibitor MG132 and 309

autophagolysosome pathway inhibitor chloroquine diphosphate (CQ), and found that MG132 could 310

reverse PLAGL2 regulation of Snail1 protein to a certain extent (Figure 3F). To examine whether 311

PLAGL2 regulates the Snail1 protein stability, we detected the abundance of Snail1 in GC cells 312

treated with CHX. Overexpression of PLAGL2 in the AGS cells significantly increased the stability 313

and half-life of Snail1. Accordingly, the silencing of PLAGL2 in SGC7901 cells significantly 314

destabilized the Snail1 protein (Figure 3G). We further explored the role of PLAGL2 in Snail1 315

ubiquitination. In the presence or absence of MG132, enforced PLAGL2 expression inhibited Snail1 316

ubiquitination in the AGS cells (Figure S2C), whereas silencing PLAGL2 promoted Snail1 317

ubiquitination (Figure 3H-I). The above data together show that PLAGL2 stabilizes the Snail1 318

protein by inhibiting its ubiquitination. 319

To further explore whether PLAGL2 induces the proliferation, migration, and invasion of GC 320

cells in a Snail1-dependent manner, we utilized either the specific Snail1 siRNA or plasmid to 321

conduct studies in SGC7901 and AGS cells. The effect of Snail1 siRNA and plasmid on the 322

proliferation, migration, invasion, and EMT markers was also examined. Snail1 siRNAs impaired 323

cell proliferation in both AGS-vector and AGS-PLAGL2 cells (Figure 3J-K and Figure S2D). 324

Conversely, the Snail1 plasmid promoted cell proliferation in both SGC7901-NC and SGC7901-325

shRNA cells (Figure 3J-K). A Transwell assay was then conducted to explore the role of Snail1 326

siRNA in GC cell motility. The results showed that PLAGL2 overexpression increased GC cell 327

motility, whereas Snail1 siRNA blocked this function (Figure 3L and Figure S2E). Moreover, the 328

regulation of proliferation and metastasis-related genes caused by PLAGL2 depletion was impaired 329

by the Snail1 plasmid, whereas the regulation resulting from PLAGL2 overexpression could be 330

Page 16: 1 Title page - Theranostics

16 / 42

opposed by Snail1 siRNA (Figure 3M and Figure S2F-G). 331

332

USP37 interacts with and deubiquitinates Snail1 directly. 333

PLAGL2 mainly serves as a transcription factor and unlikely acts as a deubiquitinase to directly 334

mediate Snail1 ubiquitination. Therefore, we speculate that some DUBs might be responsible for 335

Snail1 ubiquitination induced by PLAGL2. The mRNA microarray analysis was conducted using 336

total RNA isolated from SGC7901 NC and SGC7901 shRNA cells(n=3), and we screened three 337

DUBs from the differentially expressed genes (Figure 4A). We then investigated the influence of 338

the DUBs on Snail1 ubiquitination. Among these three DUBs, only USP37 significantly diminished 339

the ubiquitination of Snail1, suggesting that USP37 may be a potential deubiquitinase of Snail1 340

(Figure 4B). USP37 protein expression was positively correlated with Snail1 protein levels in GC 341

cell lines and GC tissues (Figure 4C). We then examined the effect of USP37 on Snail protein 342

expression. As shown in Figure 4D and Figure S3A, depletion of USP37 in SGC7901 and HEK293T 343

cells significantly reduced Snail1 protein expression, whereas enhanced USP37 expression in AGS 344

and HEK293T cells dramatically upregulated the Snail1 protein levels. As expected, USP37 did not 345

affect the mRNA levels of Snail1(Figure S3B). We further investigated the interaction between 346

USP37 and Snail1. USP37 and Snail1 proteins are mainly colocalized in the nucleus(Figure 4E). 347

Also, the reciprocal co-immunoprecipitation analysis demonstrated that endogenous USP37 and 348

Snail1 bound to each other in SGC7901 and AGS cells (Figure 4F). To map the binding protein 349

domains for Snail1 interacting with USP37, we constructed a series of HA-Snail1 truncations and 350

found that the serine-rich domain (SRD) within Snail1 is required for its interaction with 351

USP37(Figure 4G-H). To examine whether USP37 regulates Snail1 protein stability, we detected 352

Page 17: 1 Title page - Theranostics

17 / 42

the abundance of Snail1 in GC cells treated with CHX. USP37 considerably increased the stability 353

and half-life of Snail1 in the AGS cells. Accordingly, the silencing of USP37 in SGC7901 cells 354

significantly destabilized the Snail1 protein (Figure S3C-D). We also assessed the impact of USP37 355

on Snail1 ubiquitination. Overexpression of USP37 significantly reduced the endogenous 356

ubiquitination of Snail1 in the AGS cells. Accordingly, silencing endogenous USP37 promoted 357

Snail1 ubiquitination in the presence or absence of MG132 (Figure 4I-J). Moreover, overexpression 358

of USP37-C350S, a DUB dead mutant USP37 (USP37DD)[31], did not alter Snail1 ubiquitination 359

(Figure 4K). Given that the SRD domain is essential for the interaction between USP37 and Snail1, 360

we evaluated its effect on Snail1 ubiquitination in HEK293T cells. As expected, ΔSRD and Δ1-151 361

could not be deubiquitinated by USP37 (Figure 4L). Therefore, the SRD domain is also crucial for 362

USP37-mediated Snail1 deubiquitination and the interaction between USP37 and Snail1. Overall, 363

these results demonstrate that USP37 interacts with and deubiquitinates Snail1 directly. 364

365

USP37 targets Snail1 for degradation in a GSK-3β phosphorylation-dependent manner 366

As we know, the proper phosphorylation of target proteins by specific kinases is critical for 367

DUBs to identify their substrates for subsequent ubiquitination and degradation [32]. To explore 368

whether the interaction between Snail1 and USP37 requires Snail1 phosphorylation, we utilized CIP 369

to remove phosphate groups from the substrate proteins. Strikingly, Co-IP experiments showed that 370

the interaction between USP37 and Snail1 was strongly impeded by CIP intervention (Figure 5A). 371

Since GSK-3β is the major kinase involved in Snail1 phosphorylation in the SRD region, the GSK-372

3β inhibitor LiCl was used to assess GSK-3β inhibition in the interaction of USP37 and Snail1. As 373

shown in Figure 5B, LiCl treatment also significantly disrupted the USP37 and Snail1 interaction. 374

Page 18: 1 Title page - Theranostics

18 / 42

Thus, we constructed the Snail1 mutant Snail1-6SA, and its six putative GSK-3β phosphorylation 375

sites within the SRD domain were mutated from Ser to Ala [33]. Snail1-6SA demonstrated a 376

significantly decreased interaction with USP37 as expected (Figure 5C). The specific GSK-3β 377

siRNAs were further used to investigate the role of GSK-3β in the interaction between Snail1 and 378

the USP37 protein. Co-IP results showed that GSK-3β siRNAs diminished approximately 60% of 379

the interaction of USP37 and Snail1 (Figure 5D). Furthermore, we confirmed the role of GSK-3β 380

in the USP37-mediated stabilization of Snail1. As shown in Figure 5E, USP37 remarkably elevated 381

the protein level of wild-type Snail1 but had no significant impact on Snail1-6SA. Notably, the 382

Snail1-6SA exhibited higher stabilization than wild-type Snail1 with CHX treatment (Figure 5F). 383

However, Snail1-6SA was strongly resistant to USP37-mediated Snail1stabilization (Figure 5G). 384

Intervention with GSK-3β inhibitors CHIR-99021 or LiCl also displayed a strong resistance to 385

USP37-mediated stabilization of Snail1(Figure 5H-I). Besides, silencing of endogenous GSK-3β 386

impeded USP37-mediated stabilization of Snail1 in AGS cells (Figure 5J). These results consistently 387

support that GSK-3β-dependent phosphorylation of Snail1 is vital for its USP37-mediated 388

stabilization. 389

390

PLAGL2 modulates Snail1 stability by activating USP37 transcription 391

We sought to identify whether PLAGL2 serves as a transcription factor, directly regulating the 392

transcriptional level of USP37 in GC. First, we investigated the PLAGL2, and USP37 mRNA levels 393

in GC by utilizing the GEPIA database. We observed a linear correlation between PLAGL2 and 394

USP37 mRNA levels. The USP37 mRNA levels were also linearly correlated with PLAGL2 mRNA 395

levels in clinical GC specimens (Figure 6A). Moreover, silencing PLAGL2 decreased the mRNA 396

Page 19: 1 Title page - Theranostics

19 / 42

level of USP37 in SGC7901 cells, whereas PLAGL2 overexpression increased the USP37 mRNA 397

level in AGS cells (Figure 6B). This finding was further confirmed at the protein level in cells with 398

different PLAGL2 expression levels (Figure 6B). Given that PLAGL2 can specifically bind to the 399

GRGGC(N)6-8RGGK consensus sequences in the promoter, one potential PLAGL2 transcriptional 400

binding site was identified in the USP37 promoter region. The ChIP assay revealed that PLAGL2 401

was bound to the USP37 promoter region (Figure 6C). To further determine the transcriptional 402

regulation of USP37 by PLAGL2, we constructed promoter-reporter plasmids, that harbored wild-403

type and mutations of the PLAGL2 binding site (Figure 6D). These promoter-reporter plasmids 404

were transfected into cells with different PLAGL2 expression levels, and the luciferase activity was 405

subsequently detected. Results indicated that PLAGL2 can significantly increase wild-type 406

promoter activity but demonstrated no effect on mutant promoters (Figure 6E), and the core and G-407

cluster were found to be critical for PLAGL2 binding. 408

We further clarified whether PLAGL2 regulated Snail1 protein expression in a USP37-409

dependent manner, and we utilized either the specific USP37 siRNA or plasmid to conduct studies 410

in SGC7901 and AGS cells. After cotransfection with the USP37-plasmid, the expression level of 411

Snail1 protein was significantly restored, and the expression level of Snail1 protein between 412

SGC7901-NC and SGC7901-shRNA did not show statistically significant differences (Figure 6F). 413

The expression of Snail1 mRNA in cotransfected cells was detected by qRT-PCR. The results 414

showed that compared with the corresponding control group, there was no significant difference in 415

the level of Snail1 mRNA among the above groups (Figure S4A). To further explore whether 416

PLAGL2 regulated the expression of Snail1 protein in a USP37-dependent manner, it mediated the 417

proliferation, migration, and invasion of GC cells. CCK8 experiments and Transwell migration and 418

Page 20: 1 Title page - Theranostics

20 / 42

invasion experiments were performed on the cells after cotransfection. After cotransfection with 419

USP37-plasmid, the cell proliferation, migration, and invasion abilities were significantly restored, 420

and after cotransfection with USP37-siRNAs, the cell proliferation, migration, and invasion abilities 421

were significantly inhibited (Figure 6G-H and Figure S4B-D). We found that after cotransfection of 422

USP37-plasmid, there was no significant difference in the expression levels of cell cycle regulatory 423

genes and EMT-related genes between SGC7901-NC and SGC7901-shRNA (Figure 6J-I and Figure 424

S4E-F). After cotransfection of USP37-siRNA, there was no significant difference in the expression 425

levels of cell cycle regulatory genes and EMT-related genes between AGS-vector and AGS-426

PLAGL2 (Figure 6J-I and Figure S4E-F). These results collectively suggest that PLAGL2 427

transcriptionally activates USP37 expression to increase Snail1 protein levels, which in turn 428

mediates the proliferation, migration, and invasion of GC cells. (Figure 7A). 429

430

431

432

433

434

435

436

437

438

439

440

Page 21: 1 Title page - Theranostics

21 / 42

Discussion 441

PLAGL2 mainly acts as a transcription factor that has been implicated in the progression of 442

multiple types of tumors. In our study, we observed that PLAGL2 is upregulated in GC cells and 443

GC specimens. Subsequent in vitro and in vivo experiments showed that overexpression of 444

PLAGL2 significantly promotes cancer cell proliferation and metastasis, indicating the carcinogenic 445

role of PLAGL2 in regulating GC progression. Consistent with our results, recent studies have 446

revealed that PLAGL2 serves as an oncogene to induce lung cancer8, colorectal cancer [5,7,12], 447

and breast cancer [34]. Of note, we found that the underlying mechanism by which PLAGL2 448

functions as a promoter for GC proliferation and migration is by stabilizing Snail1. We also 449

determined that USP37 interacts with and deubiquitinates Snail1 directly, which is transcriptionally 450

activated by PLAGL2. 451

As an essential transcription factor, Snail1 plays a crucial role in proliferation, metastasis, 452

tumor recurrence, and metabolism [14]. Snail1 is strictly controlled at the transcriptional, 453

translational, and posttranslational levels [35]. The Snail1 protein levels change rapidly in response 454

to cellular stress, mostly dependent on protein stability regulation [16]. Enhanced Snail1 expression 455

leads to several cancers, including GC. Herein, we determined that PLAGL2 is a crucial and novel 456

regulator that upregulates the Snail1 protein by inhibiting the ubiquitin-proteasome pathway, and 457

this is one of our key findings. PLAGL2 overexpression significantly increased Snail1 protein levels 458

to promote GC cell proliferation and metastasis. Although PLAGL2 does not function as a 459

deubiquitinase to impede Snail1 ubiquitination, PLAGL2 transcriptionally activates USP37, which 460

finally interacts with and deubiquitinates Snail1 directly. 461

The deubiquitinase USP37 is involved in the tumorigenesis process, specifically cell cycle 462

Page 22: 1 Title page - Theranostics

22 / 42

progression [36-38], EMT [39], chemosensitivity [39] and cell self-renewal [39]. USP37 was 463

initially identified as an effective modulator of G1/S transition [36,38]. Previous studies reported 464

that USP37 increased the Warburg effect and cell viability by repressing ubiquitin-mediated 465

degradation of c-Myc [31]. Moreover, overexpression of USP37 in breast cancer promoted stemness, 466

cell invasion, and chemoresistance by deubiquitinating the glioma-associated oncogene 1 protein 467

[39]. In the past year, two other deubiquitinating enzymes, USP27X and USP1, have been reported 468

to impede ubiquitin-mediated degradation of Snail1 in various biological contexts [18,19]. USP27X 469

modulates tumor chemoresistance and invasion through deubiquitination and stabilization of 470

Snail1[18]. USP1 is a Snail1 deubiquitinase and increases metastatic ability and chemoresistance 471

[19]. In our study, Snail1 was identified as a new substrate of USP37 in GC, which is an important 472

finding. Overexpression of USP37 in SGC7901-shRNA cells restores Snail1 protein levels, which 473

shows that the USP37-Snail1 axis plays a vital role in PLAGL2-mediated Snail1 stabilization. 474

Generally, proper phosphorylation of substrates is vital for the DUB-mediated proteasomal 475

degradation pathway. In terms of the overall intracellular environment, Snail1 6SA protein is more 476

stable than Snail1-WT protein. However, in cells with high expression of USP37, phosphorylated 477

Snail1 promotes the mutual binding of USP37 and Snail1, and the ability of USP37 to bind to 478

phosphorylated Snail1 is significantly enhanced. USP37 exerts its powerful deubiquitinating 479

enzyme function, and the stability of Snail1-WT Significantly higher than Snail1 6SA. It further 480

shows that USP37 regulation of Snail1 protein stability requires the participation of GSK-3β-481

mediated phosphorylation. Moreover, USP37 exerts a strong deubiquitination and stabilization 482

effect on the phosphorylated Snail1 protein. These results further revealed that the SRD domain of 483

Snail1 is responsible for USP37-mediated Snail1 deubiquitination and GSK-3β-induced 484

Page 23: 1 Title page - Theranostics

23 / 42

phosphorylation and could be a potent therapeutic target for GC. In this study, we demonstrated that 485

SRD is required for its interaction with USP37. In addition, Snail1 phosphorylated by GSK-3β, is 486

easier to combine with USP37. Phosphorylated Snail1, combined with USP37 is more stable. 487

However, the Snail1 mutant, Snail1-6SA, is much more stable than wild type Snail1. Liu et al. found 488

that Snail1 phosphorylated by GSK-3β, interacts with SPSB3, showing lower stability [33]. Perhaps, 489

the phosphorylation of Snail1 mainly promotes its binding to E3 ligases. Phosphorylated Snail1 490

binds to deubiquitinating enzymes, which will increase its stability, whereas binding to the ubiquitin 491

enzyme will accelerate its degradation. 492

In summary, our study reveals a PLAGL2-USP37-Snail1 axis representing a critical 493

mechanism in the proliferation and migration of GC cells (Figure 7A). Hence, blocking this axis 494

may be a potent therapeutic strategy against GC. 495

496

Abbreviations 497

PLAGL2: Pleomorphic adenoma gene like-2; USP37: ubiquitin-specific peptidase 37; qRT-498

PCR: Real-time quantitative polymerase chain reaction; IF: Immunofluorescence; ChIP Chromatin 499

immunoprecipitation; Snail1: snail family transcriptional repressor 1; GC: gastric cancer; EMT: 500

epithelial-mesenchymal transition; HE: Hematoxylin and eosin; CHX: cycloheximide; CQ: 501

chloroquine diphosphate. 502

503

Supplementary Material 504

Supplementary figures and tables. 505

506

Page 24: 1 Title page - Theranostics

24 / 42

Acknowledgments 507

We thank AJE (American Journal Experts) for proofreading this paper (the verification code: 508

E3EF-5B4E-CE91-3329-3253). We would like to thank Mingyang Gao, Jinyuan Cui, and Zhibo Liu 509

for their generous and excellent help, Panpan Huang for flow cytometry analysis. This work was 510

supported by the National Natural Science Foundation of China (NSFC) (Grant numbers: 81271199). 511

512

Contributions 513

L.W. and X.G.S. conceived the study and supervised and coordinated all aspects of the work; 514

L.W. and N.Z. designed the research, prepared the figures and tables, and wrote the paper; L.W., 515

N.Z., Z.L.Z., J.H.C., S.B.H., and X.D.Z. designed and performed the experiments, interpreted the 516

data; and H.J.B. and W.Z.Y. contributed with analytical tools. 517

518

Ethics approval and consent to participate 519

Ethics approval was granted by the Ethics Committee of Tongji Medical College, Huazhong 520

University of Science and Technology. According to the Tongji Medical College Animal Care and 521

Use Guidelines, animal experiments were performed. Moreover, the Ethics Committee approved 522

the experimental protocols of Tongji Medical College. Written informed consent was obtained from 523

all patients. 524

525

Conflict of interest 526

The authors declare that they have no conflict of interest. 527

528

Page 25: 1 Title page - Theranostics

25 / 42

References 529

1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer statistics 2018: 530

GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA 531

Cancer J Clin. 2018;68(6):394-424. 532

2. Chaffer CL, Weinberg RA. A perspective on cancer cell metastasis. Science. 533

2011;331(6024):1559-64. 534

3. Landrette SF, Madera D, He F, Castilla LH. The transcription factor PlagL2 activates Mpl 535

transcription and signaling in hematopoietic progenitor and leukemia cells. Leukemia. 536

2011;25(4):655-62. 537

4. Wu L, Yuan W, Chen J, Zhou Z, Shu Y, Ji J, et al. Increased miR-214 expression suppresses 538

cell migration and proliferation in Hirschsprung disease by interacting with PLAGL2. Pediatr Res. 539

2019;86(4):460-70. 540

5. Wu L, Zhou Z, Han S, Chen J, Liu Z, Zhang X, et al. PLAGL2 promotes epithelial-541

mesenchymal transition and mediates colorectal cancer metastasis via beta-catenin-dependent 542

regulation of ZEB1. Br J Cancer. 2020;122(4):578-89. 543

6. Astrom AK, Voz ML, Kas K, Roijer E, Wedell B, Mandahl N, et al. Conserved mechanism of 544

PLAG1 activation in salivary gland tumors with and without chromosome 8q12 abnormalities: 545

identification of SII as a new fusion partner gene. Cancer Res. 1999;59(4):918-23. 546

7. Li D, Lin C, Li N, Du Y, Yang C, Bai Y, et al. PLAGL2 and POFUT1 are regulated by an 547

evolutionarily conserved bidirectional promoter and are collaboratively involved in colorectal 548

cancer by maintaining stemness. EBioMedicine. 2019;45:124-38. 549

8. Yang YS, Yang MC, Weissler JC. Pleiomorphic adenoma gene-like 2 expression is associated 550

Page 26: 1 Title page - Theranostics

26 / 42

with the development of lung adenocarcinoma and emphysema. Lung Cancer. 2011;74(1):12-24. 551

9. Li N, Li D, Du Y, Su C, Yang C, Lin C, et al. Overexpressed PLAGL2 transcriptionally 552

activates Wnt6 and promotes cancer development in colorectal cancer. Oncol Rep. 2019;41(2):875-553

84. 554

10. Hu W, Zheng S, Guo H, Dai B, Ni J, Shi Y, et al. PLAGL2-EGFR-HIF-1/2alpha signaling loop 555

promotes HCC progression and Erlotinib insensitivity. Hepatology. 2020, doi: 10.1002/hep.31293 556

11. Liu B, Lu C, Song YX, Gao P, Sun JX, Chen XW, et al. The role of pleomorphic adenoma 557

gene-like 2 in gastrointestinal cancer development, progression, and prognosis. Int J Clin Exp Pathol. 558

2014;7(6):3089-100. 559

12. Wang YP, Guo PT, Zhu Z, Zhang H, Xu Y, Chen YZ, et al. Pleomorphic adenoma gene like-2 560

induces epithelial-mesenchymal transition via Wnt/beta-catenin signaling pathway in human 561

colorectal adenocarcinoma. Oncol Rep. 2017;37(4):1961-70. 562

13. Jing Y, Han Z, Zhang S, Liu Y, Wei L. Epithelial-Mesenchymal Transition in tumor 563

microenvironment. Cell Biosci. 2011;1:29. 564

14. Kaufhold S, Bonavida B. Central role of Snail1 in the regulation of EMT and resistance in 565

cancer: a target for therapeutic intervention. J Exp Clin Cancer Res. 2014;33:62. 566

15. Wang Y, Shi J, Chai K, Ying X, Zhou BP. The Role of Snail in EMT and Tumorigenesis. Curr 567

Cancer Drug Targets. 2013;13(9):963-72. 568

16. Diaz VM, Vinas-Castells R, Garcia de Herreros A. Regulation of the protein stability of EMT 569

transcription factors. Cell Adh Migr. 2014;8(4):418-28. 570

17. Diaz VM, de Herreros AG. F-box proteins: Keeping the epithelial-to-mesenchymal transition 571

(EMT) in check. Semin Cancer Biol. 2016;36:71-9. 572

Page 27: 1 Title page - Theranostics

27 / 42

18. Lambies G, Miceli M, Martinez-Guillamon C, Olivera-Salguero R, Pena R, Frias CP, et al. 573

TGFbeta-Activated USP27X Deubiquitinase Regulates Cell Migration and Chemoresistance via 574

Stabilization of Snail1. Cancer Res. 2019;79(1):33-46. 575

19. Sonego M, Pellarin I, Costa A, Vinciguerra GLR, Coan M, Kraut A, et al. USP1 links platinum 576

resistance to cancer cell dissemination by regulating Snail stability. Sci Adv. 2019;5(5):eaav3235. 577

20. Cai J, Li M, Wang X, Li L, Li Q, Hou Z, et al. USP37 Promotes Lung Cancer Cell Migration 578

by Stabilizing Snail Protein via Deubiquitination. Front Genet. 2019;10:1324. 579

21. Xiao Z, Chang L, Kim J, Zhang P, Hang Q, Yap S, et al. USP37 is a SNAI1 deubiquitinase. 580

Am J Cancer Res. 2019;9(12):2749-59. 581

22. Wu Y, Wang Y, Lin Y, Liu Y, Wang Y, Jia J, et al. Dub3 inhibition suppresses breast cancer 582

invasion and metastasis by promoting Snail1 degradation. Nat Commun. 2017;8:14228. 583

23. Lee JH, Jung SM, Yang KM, Bae E, Ahn SG, Park JS, et al. A20 promotes metastasis of 584

aggressive basal-like breast cancers through multi-monoubiquitylation of Snail1. Nat Cell Biol. 585

2017;19(10):1260-73. 586

24. Frias A, Lambies G, Vinas-Castells R, Martinez-Guillamon C, Dave N, Garcia de Herreros A, 587

et al. A Switch in Akt Isoforms Is Required for Notch-Induced Snail1 Expression and Protection 588

from Cell Death. Mol Cell Biol. 2015;36(6):923-40. 589

25. Ma L, Lin K, Chang G, Chen Y, Yue C, Guo Q, et al. Aberrant Activation of beta-Catenin 590

Signaling Drives Glioma Tumorigenesis via USP1-Mediated Stabilization of EZH2. Cancer Res. 591

2019;79(1):72-85. 592

26. Du L, Xing Z, Tao B, Li T, Yang D, Li W, et al. Both IDO1 and TDO contribute to the 593

malignancy of gliomas via the Kyn-AhR-AQP4 signaling pathway. Signal Transduct Target Ther. 594

Page 28: 1 Title page - Theranostics

28 / 42

2020;5(1):10. 595

27. Gou HF, Chen XC, Zhu J, Jiang M, Yang Y, Cao D, et al. Expressions of COX-2 and VEGF-C 596

in gastric cancer: correlations with lymphangiogenesis and prognostic implications. J Exp Clin 597

Cancer Res. 2011;30:14. 598

28. Rhodes DR, Yu J, Shanker K, Deshpande N, Varambally R, Ghosh D, et al. ONCOMINE: a 599

cancer microarray database and integrated data-mining platform. Neoplasia. 2004;6(1):1-6. 600

29. Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z. GEPIA: a web server for cancer and normal 601

gene expression profiling and interactive analyses. Nucleic Acids Res. 2017;45(W1):W98-W102. 602

30. Gyorffy B, Lanczky A, Eklund AC, Denkert C, Budczies J, Li Q, et al. An online survival 603

analysis tool to rapidly assess the effect of 22,277 genes on breast cancer prognosis using microarray 604

data of 1,809 patients. Breast Cancer Res Treat. 2010;123(3):725-31. 605

31. Pan J, Deng Q, Jiang C, Wang X, Niu T, Li H, et al. USP37 directly deubiquitinates and 606

stabilizes c-Myc in lung cancer. Oncogene. 2015;34(30):3957-67. 607

32. Cardozo T, Pagano M. The SCF ubiquitin ligase: insights into a molecular machine. Nat Rev 608

Mol Cell Biol. 2004;5(9):739-51. 609

33. Liu Y, Zhou H, Zhu R, Ding F, Li Y, Cao X, et al. SPSB3 targets SNAIL for degradation in 610

GSK-3beta phosphorylation-dependent manner and regulates metastasis. Oncogene. 611

2018;37(6):768-76. 612

34. Xu B, Zhang X, Wang S, Shi B. MiR-449a suppresses cell migration and invasion by targeting 613

PLAGL2 in breast cancer. Pathol Res Pract. 2018;214(5):790-5. 614

35. Mladinich M, Ruan D, Chan CH. Tackling Cancer Stem Cells via Inhibition of EMT 615

Transcription Factors. Stem Cells Int. 2016;2016:5285892. 616

Page 29: 1 Title page - Theranostics

29 / 42

36. Huang X, Summers MK, Pham V, Lill JR, Liu J, Lee G, et al. Deubiquitinase USP37 is 617

activated by CDK2 to antagonize APC(CDH1) and promote S phase entry. Mol Cell. 618

2011;42(4):511-23. 619

37. Das CM, Taylor P, Gireud M, Singh A, Lee D, Fuller G, et al. The deubiquitylase USP37 links 620

REST to the control of p27 stability and cell proliferation. Oncogene. 2013;32(13):1691-701. 621

38. Burrows AC, Prokop J, Summers MK. Skp1-Cul1-F-box ubiquitin ligase (SCF(betaTrCP))-622

mediated destruction of the ubiquitin-specific protease USP37 during G2-phase promotes mitotic 623

entry. J Biol Chem. 2012;287(46):39021-9. 624

39. Qin T, Li B, Feng X, Fan S, Liu L, Liu D, et al. Abnormally elevated USP37 expression in 625

breast cancer stem cells regulates stemness, epithelial-mesenchymal transition and cisplatin 626

sensitivity. J Exp Clin Cancer Res. 2018;37(1):287. 627

628

629

630

631

632

633

634

635

636

637

638

Page 30: 1 Title page - Theranostics

30 / 42

Table 1. Clinicopathological analysis of PLAGL2 expression in GC. 639

640

641

642

643

644

645

646

647

648

649

650

651

652

653

654

655

656

657

658

659

660

Parameters n PLAGL2

+ - P-value

Age(years)

<60 31 26 5 0.149

≥60 18 11 7

Gender

Male 30 24 6 0.564

Female 19 13 6

Size of tumor

<3cm 20 12 8 0.079

≥3cm 29 25 4

Differentiation

Well-moderate 23 15 8 0.214

Poor 26 22 4

T Stages

T1–T2 18 10 8 0.033*

T3–T4 31 27 4

Metastasis

N Stages

N0 14 6 8 0.003*

N1-3 35 31 4

M Stages

M0 34 24 10 0.398

M1 15 13 2

Page 31: 1 Title page - Theranostics

31 / 42

Figure legends 661

Figure 1. PLAGL2 is highly expressed in GC. (A) WB and qRT–PCR analyses of PLAGL2 662

expression in human GC cell lines and GES-1. (B) WB and qRT–PCR analyses of PLAGL2 663

expression levels in clinical GC specimens. T, GC tissue; N, paired normal tissues. (C) Quantitative 664

analysis of relative PLAGL2 expression in 49 paired GC tissues. (D)Representative IHC images of 665

the PLAGL2 expression levels in paired GC tissues. Scale bars, 100μm. (E) A meta-analysis of the 666

PLAGL2 gene expression derived from the Oncomine database. (F) A box plot reflecting the 667

PLAGL2 expression in GC specimens and normal specimens was obtained from the GEPIA 668

database (G) Representative IHC images of the PLAGL2 expression levels in primary GC tumors 669

without metastasis vs. GC tumors with metastasis. Scale bars, 100μm. (H) Kaplan-Meier survival 670

analysis of overall survival based on PLAGL2 expression in GC patients. 671

672

Figure 2. PLAGL2 promotes the proliferation, migration, and invasion of GC cells. (A) WB 673

analysis of PLAGL2 expression in SGC7901 transfected with Lenti-shPLAGL2 and AGS 674

transfected with Lenti-PLAGL2. (B-C) The cell proliferation was examined by CCK8 (B) and 675

colony formation assays (C) in SGC7901 and AGS cells. Scale bars, 1cm. (D) WB analysis of the 676

expression level of crucial cell cycle regulatory proteins. (E) Representative images of the 677

corresponding xenograft. The growth curve of the tumors in the different groups. (F) The weight of 678

tumors in different groups. (G-H) The cell migration was examined by the wound-healing assay. 679

Pictures were taken at 0 and 48 h, respectively. Scale bars, 500μm. (I-J) The migration and invasion 680

capacities of GC cells were also evaluated with Transwell assays. Scale bars, 200μm. (K) WB 681

analysis of the expression level of EMT-related proteins. (L) Representative images of visible lung 682

Page 32: 1 Title page - Theranostics

32 / 42

metastases. The metastatic nodules were indicated with arrows. (M) Representative images of the 683

corresponding HE staining. Scale bars, 200μm. (N) Numbers of metastatic nodules. (O) The ratio 684

of lung weight/body weight. 685

686

Figure 3. PLAGL2 stabilizes Snail1 protein by inhibiting its ubiquitination. (A) WB analysis 687

of Snail1 expression of in PLAGL2 knockdown and overexpression cells. (B) WB analysis of 688

exogenous Snail1 in HEK293T cells co-expressing PLAGL2 and Snail1. (C) WB analysis of protein 689

levels of PLAGL2 and Snail1 in clinical GC specimens. (D) The correlation between PLAGL2 690

protein and Snail1 protein in GC tissues. (E) The correlation between PLAGL2 mRNA and Snail1 691

mRNA in GC tissues. (F) WB analysis of Snail1 level in PLAGL2 knockdown cells treated with 692

MG132 and CQ. (G) CHX chase analysis of Snail1 protein half-life in PLAGL2 knockdown 693

SGC7901 cell and PLAGL2 overexpression AGS cell. (H-I) Ubiquitination assays of endogenous 694

Snail1 in the lysates from AGS cells transfected with Flag-PLAGL2 (H) or AGC7901 cells stably 695

expressing PLAGL2 shRNA (I). (J-K) The stable PLAGL2 knockdown (SGC7901-shRNA) cell 696

was transfected with Snail1 plasmid, and stable overexpression (AGS-PLAGL2) cell was 697

transfected with the Snail1 siRNA. The role of Snail1 in PLAGL2-induced proliferation was 698

examined by CCK8 (J) and colony formation assays(K). (L) Transwell assays detected the effect 699

of Snail1 on PLAGL2-induced migration. Scale bars, 200μm(l). (M) WB analysis of the expression 700

level of EMT-related proteins and critical cell cycle regulatory proteins in cotransfected SGC7901 701

and AGS cells. 702

703

Figure 4. USP37 interacts with and deubiquitinates Snail1 directly. (A) Microarray analysis for 704

Page 33: 1 Title page - Theranostics

33 / 42

mRNAs was performed with RNA extracted from SGC7901 NC and SGC7901 shRNA cells. (B) 705

Ubiquitination assays of endogenous Snail1 in HEK293T cell, which was cotransfected with HA-706

Snail1, Flag-Ubi, and one of the three DUBs (USP37, USP38, and DUSP18). (C) WB analysis of 707

protein levels of USP37 and Snail1 in clinical GC specimens and human GC cell lines. (D) WB 708

analysis of protein levels of Snail1 and USP37 in SGC7901 cells transfected with two independent 709

USP37 siRNAs and AGS cells expressing USP37 plasmid. (E) The representative images of IF 710

staining of Snail1 (green) and USP37 (red) were shown in SGC7901 and AGS cells. Scale bars, 711

50μm. (F) Reciprocal Co-IP and WB assays indicated the interaction between endogenous USP37 712

and Snail1 in AGS and SGC7901 cells. (G) Schematic diagram of the Snail1 full-length and deletion 713

mutant plasmid. (H) HEK293T cells were cotransfected with USP37-His and the full length or 714

truncation mutants of HA-Snail1. Cell lysates were immunoprecipitated with anti-HA antibody, and 715

its production was analyzed by WB analysis with anti-His antibody (I-J) Ubiquitination assays of 716

endogenous Snail1 in the lysates from AGS cells transfected with the USP37 plasmid (I) or 717

AGC7901 cells transfected with the USP37 siRNA (J). (K) Ubiquitination assays of endogenous 718

Snail1 in the lysates from HEK293T cells, which were cotransfected with the His-USP37-WT or 719

His-USP37-DD, HA-Snail1 and Flag-Ub expressing plasmids. (L) Ubiquitination assays of 720

endogenous Snail1 in the lysates from HEK293T cells, which were cotransfected with USP37-His 721

and the full length or truncation mutants of HA-Snail1. 722

723

Figure 5. USP37 targets Snail1 for degradation in a GSK-3β phosphorylation-dependent 724

manner. (A) Co-IP and western blot assays showed the mutual binding ability between USP37 and 725

Snail1 in HEK293T cells treated either with or without CIP. (B) Co-IP and western blot assays 726

Page 34: 1 Title page - Theranostics

34 / 42

showed the mutual binding ability between USP37 and Snail1 in HEK293T cells treated either with 727

or without LiCl (C) Co-IP and western blot assays showed the mutual binding ability between 728

USP37 and Snail1 in HEK293T cells cotransfected with His-USP37 and Wild-type Snail1 or Snail1-729

6SA mutant. (D) Co-IP and western blot assays showed the mutual binding ability between USP37 730

and Snail1 in HEK293T cells cotransfected either with or without si GSK3β (E) The WB analysis 731

reflecting the expression regulation of USP37 on Wild-type Snail1 and Snail1-6SA mutants in 732

HEK293T cells. (F) Pulse-chase assays of wild-type Snail1 or Snail1-6SA mutant in HEK293T cells. 733

(G) Pulse-chase assays of Snail1 in HEK293T cells cotransfected with USP37 and Wild-type Snail1 734

or Snail1-6SA mutant. (H) Pulse-chase assays of Snail1 in HEK293T cells, which were transfected 735

with His-USP37 plasmid and were treated either with or without LiCl. (I) Pulse-chase assays of 736

Snail1 in HEK293T cells, which were transfected with His-USP37 plasmid and were treated either 737

with or without CHIR-99021. (J) Pulse-chase assays of Snail1 in AGS cells, which were 738

cotransfected with His-USP37 plasmid and siGSK-3β. 739

740

Figure 6. PLAGL2 modulates Snail1 stability by activating USP37 transcription. (A) A 741

significant positive correlation between PLAGL2 mRNA and USP37 mRNA in GC tissues could be 742

observed both in GEPIA database(left) and our data(right). (B) WB and qRT–PCR analyses of 743

Snail1expression in PLAGL2 knockdown SGC7901 cell and PLAGL2 overexpression AGS cell. 744

(C) The ChIP DNA was amplified by qRT-PCR, and then the products of qRT-PCR were 745

electrophoresed on a 2% agarose gel. (D) The schematic representation of USP37 promoter. The 746

sequences of wild-type and mutant PLAGL2 binding sites were indicated. (E) Luciferase activity 747

assays were performed in PLAGL2 knockdown SGC7901 cell and PLAGL2 overexpression AGS 748

Page 35: 1 Title page - Theranostics

35 / 42

cell, which were then transfected with wild type (USP37-WT) or mutant-type (USP37-mut) USP37 749

promoter-reporter plasmids. (F) WB of Snail1 protein level in SGC7901 cell cotransfected with 750

Lenti-shPLAGL2 and USP37 plasmid and AGS cell cotransfected with Lenti-PLAGL2 and USP37 751

siRNA. (G-H) The stable PLAGL2 knockdown (SGC7901-shRNA) cell was transfected with 752

USP37 plasmid, and stable overexpression (AGS-PLAGL2) cell was transfected with the USP37 753

siRNA. The role of USP37 in PLAGL2-induced proliferation was examined by CCK8 (G). 754

Transwell assays detected the effect of USP37 on PLAGL2-induced migration. Scale bars, 755

200μm(H). (I) WB analysis of the expression level of EMT-related proteins in cotransfected 756

SGC7901 and AGS cells. (J) The qRT–PCR analysis of the expression level of EMT-related genes 757

in cotransfected SGC7901 and AGS cells. 758

759

Figure 7. Schematic diagram of the molecular mechanism of PLAGL2 promoting 760

proliferation and migration of GC cells. (A) Schematic diagram of the molecular mechanism of 761

PLAGL2 promoting proliferation and migration of GC cells. High expression of PLAGL2 762

transcriptionally activates USP37 expression to protect Snail1 from degradation by the proteasome. 763

764

765

766

767

768

769

770

Page 36: 1 Title page - Theranostics

36 / 42

Figure 1. 771

772

773

774

775

776

777

778

779

780

781

782

783

Page 37: 1 Title page - Theranostics

37 / 42

Figure 2. 784

785

786

787

788

789

790

Page 38: 1 Title page - Theranostics

38 / 42

Figure 3. 791

792

793

794

795

796

797

Page 39: 1 Title page - Theranostics

39 / 42

Figure 4. 798

799

800

801

802

803

804

805

Page 40: 1 Title page - Theranostics

40 / 42

Figure 5. 806

807

808

809

810

811

812

813

814

815

816

817

Page 41: 1 Title page - Theranostics

41 / 42

Figure 6. 818

819

820

821

822

823

824

825

826

827

828

Page 42: 1 Title page - Theranostics

42 / 42

Figure 7. 829

830