overexpressed nedd8 as a potential therapeutic target in

18
Cancer Biol Med 2021. doi: 10.20892/j.issn.2095-3941.2020.0484 ORIGINAL ARTICLE Overexpressed NEDD8 as a potential therapeutic target in esophageal squamous cell carcinoma Jingrong Xian 1,2,3,4* , Shiwen Wang 1,2,3,4* , Yanyu Jiang 2 , Lihui Li 2 , Lili Cai 2 , Ping Chen 5 , Yue Liu 1,2,3,4 , Xiaofei Zeng 6 , Guoan Chen 6 , Chen Ding 7,8 , Robert M. Hoffman 9,10 , Lijun Jia 2 , Hu Zhao 1,3,4 , Yanmei Zhang 1,3,4 1 Department of Laboratory Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; 2 Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; 3 Research Center on Aging and Medicine, Fudan University, Shanghai 200040, China; 4 Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040, China; 5 Department of Basic Science of Oncology, College of Basic Medical Sciences, Zhengzhou University, Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou 450001, China; 6 School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; 7 State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institutes of Biomedical Sciences, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200032, China; 8 State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang 453007, China; 9 Department of Surgery, University of California, San Diego 92101, USA; 10 Anticancer Inc., San Diego 92101, USA ABSTRACT Objective: The hyperactivated neddylation pathway plays an important role in tumorigenesis and is emerging as a promising anticancer target. We aimed to study whether NEDD8 (neural precursor cell expressed, developmentally down-regulated 8) might serve as a therapeutic target in esophageal squamous cell carcinoma (ESCC). Methods: The clinical relevance of NEDD8 expression was evaluated by using The Cancer Genome Atlas (TCGA) database and tissue arrays. NEDD8-knockdown ESCC cells generated with the CRISPR/Cas9 system were used to explore the anticancer effects and mechanisms. Quantitative proteomic analysis was used to examine the variations in NEDD8 knockdown-induced biological pathways. The cell cycle and apoptosis were assessed with fluorescence activated cell sorting. A subcutaneous-transplantation mouse tumor model was established to investigate the anticancer potential of NEDD8 silencing in vivo. Results: NEDD8 was upregulated at both the mRNA and protein expression levels in ESCC, and NEDD8 overexpression was associated with poorer overall patient survival (mRNA level: P = 0.028, protein level: P = 0.026, log-rank test). Downregulation of NEDD8 significantly suppressed tumor growth both in vitro and in vivo. Quantitative proteomic analysis revealed that downregulation of NEDD8 induced cell cycle arrest, DNA damage, and apoptosis in ESCC cells. Mechanistic studies demonstrated that NEDD8 knockdown led to the accumulation of cullin-RING E3 ubiquitin ligases (CRLs) substrates through inactivation of CRLs, thus suppressing the malignant phenotype by inducing cell cycle arrest and apoptosis in ESCC. Rescue experiments demonstrated that the induction of apoptosis after NEDD8 silencing was attenuated by DR5 knockdown. Conclusions: Our study elucidated the anti-ESCC effects and underlying mechanisms of NEDD8 knockdown, and validated NEDD8 as a potential target for ESCC therapy. KEYWORDS NEDD8; esophageal squamous cell carcinoma; cullin-RING E3 ubiquitin ligases; apoptosis; anticancer target Introduction Esophageal carcinoma is a highly malignant gastrointes- tinal cancer accounting for almost half a million deaths per year worldwide 1 . Esophageal squamous cell carcinoma (ESCC) is the most common esophageal carcinoma histo- logical subtype, and its incidence and mortality rates are ris- ing rapidly 2 . The first-line treatment for ESCC is currently *These authors contributed equally to this work. Correspondence to: Yanmei Zhang and Hu Zhao E-mail: [email protected] and [email protected] ORCID ID: https://orcid.org/0000-0002-4315-8516 and https://orcid. org/0000-0001-9728-9856 Received August 20, 2020; accepted December 24, 2020. Available at www.cancerbiomed.org ©2021 Cancer Biology & Medicine. Creative Commons Attribution-NonCommercial 4.0 International License

Upload: others

Post on 31-Jan-2022

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Overexpressed NEDD8 as a potential therapeutic target in

Cancer Biol Med 2021. doi: 10.20892/j.issn.2095-3941.2020.0484

ORIGINAL ARTICLE

Overexpressed NEDD8 as a potential therapeutic target in esophageal squamous cell carcinoma

Jingrong Xian1,2,3,4*, Shiwen Wang1,2,3,4*, Yanyu Jiang2, Lihui Li2, Lili Cai2, Ping Chen5, Yue Liu1,2,3,4, Xiaofei Zeng6, Guoan Chen6, Chen Ding7,8, Robert M. Hoffman9,10, Lijun Jia2, Hu Zhao1,3,4, Yanmei Zhang1,3,4

1Department of Laboratory Medicine, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, China; 2Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; 3Research Center on Aging and Medicine, Fudan University, Shanghai 200040, China; 4Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai 200040, China; 5Department of Basic Science of Oncology, College of Basic Medical Sciences, Zhengzhou University, Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou 450001, China; 6School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; 7State Key Laboratory of Genetic Engineering, Human Phenome Institute, Institutes of Biomedical Sciences, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200032, China; 8State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, College of Life Science, Institute of Biomedical Science, Henan Normal University, Xinxiang 453007, China; 9Department of Surgery, University of California, San Diego 92101, USA; 10Anticancer Inc., San Diego 92101, USA

ABSTRACT Objective: The hyperactivated neddylation pathway plays an important role in tumorigenesis and is emerging as a promising

anticancer target. We aimed to study whether NEDD8 (neural precursor cell expressed, developmentally down-regulated 8) might

serve as a therapeutic target in esophageal squamous cell carcinoma (ESCC).

Methods: The clinical relevance of NEDD8 expression was evaluated by using The Cancer Genome Atlas (TCGA) database and

tissue arrays. NEDD8-knockdown ESCC cells generated with the CRISPR/Cas9 system were used to explore the anticancer effects

and mechanisms. Quantitative proteomic analysis was used to examine the variations in NEDD8 knockdown-induced biological

pathways. The cell cycle and apoptosis were assessed with fluorescence activated cell sorting. A subcutaneous-transplantation mouse

tumor model was established to investigate the anticancer potential of NEDD8 silencing in vivo.

Results: NEDD8 was upregulated at both the mRNA and protein expression levels in ESCC, and NEDD8 overexpression was associated

with poorer overall patient survival (mRNA level: P = 0.028, protein level: P = 0.026, log-rank test). Downregulation of NEDD8

significantly suppressed tumor growth both in vitro and in vivo. Quantitative proteomic analysis revealed that downregulation

of NEDD8 induced cell cycle arrest, DNA damage, and apoptosis in ESCC cells. Mechanistic studies demonstrated that NEDD8

knockdown led to the accumulation of cullin-RING E3 ubiquitin ligases (CRLs) substrates through inactivation of CRLs, thus

suppressing the malignant phenotype by inducing cell cycle arrest and apoptosis in ESCC. Rescue experiments demonstrated that

the induction of apoptosis after NEDD8 silencing was attenuated by DR5 knockdown.

Conclusions: Our study elucidated the anti-ESCC effects and underlying mechanisms of NEDD8 knockdown, and validated NEDD8

as a potential target for ESCC therapy.

KEYWORDS NEDD8; esophageal squamous cell carcinoma; cullin-RING E3 ubiquitin ligases; apoptosis; anticancer target

Introduction

Esophageal carcinoma is a highly malignant gastrointes-

tinal cancer accounting for almost half a million deaths

per year worldwide1. Esophageal squamous cell carcinoma

(ESCC) is the most common esophageal carcinoma histo-

logical subtype, and its incidence and mortality rates are ris-

ing rapidly2. The first-line treatment for ESCC is currently

*These authors contributed equally to this work.Correspondence to: Yanmei Zhang and Hu ZhaoE-mail: [email protected] and [email protected] ID: https://orcid.org/0000-0002-4315-8516 and https://orcid.org/0000-0001-9728-9856Received August 20, 2020; accepted December 24, 2020.Available at www.cancerbiomed.org©2021 Cancer Biology & Medicine. Creative Commons Attribution-NonCommercial 4.0 International License

Page 2: Overexpressed NEDD8 as a potential therapeutic target in

2 Xian et al. Therapeutic potential of NEDD8 in ESCC

surgery in combination with neoadjuvant chemotherapy and

radiotherapy; however, patients have poor prognosis, with a

5-year survival of 10%–15%2,3. Therefore, an effective thera-

peutic target for ESCC is urgently needed.

Neddylation is a protein posttranslational modification that

covalently conjugates the ubiquitin-like molecule NEDD8

(neural precursor cell-expressed developmentally down-

regulated) to substrate proteins through a series of enzymatic

actions4. The cullin-RING E3 ubiquitin ligases (CRLs), the best

characterized substrates of neddylation, affect many dynamic

cellular processes, including the cell cycle, DNA damage, apop-

tosis, and tumorigenesis5-8. Moreover, members of the neddyl-

ation pathway, including NEDD8 and neddylation enzymes,

are overexpressed in several types of cancers, and inhibition of

the overexpressed neddylation pathway members may serve as

a novel anticancer strategy9-12.

MLN4924 (Pevonedistat/TAK924) is a pharmaceutical inhib-

itor of NEDD8-activating enzyme E1 (NAE)13. Recently, a

variety of phase II/III clinical trials of MLN4924 performed

on human hematological malignancies and solid tumors have

shown the efficacy of this treatment14. However, mutations in

UBA3 (a subunit of NAE E1), the target of MLN4924, have

been found to lead to drug resistance15,16. Additionally, Zhou

et al. have reported that MLN4924 stimulates tumor sphere

formation, thus suggesting potential tumor promotion by

MLN492417,18. The shortcomings of MLN4924 in cancer treat-

ment underscore the need to identify other potential therapeutic

targets against the neddylation pathway for anticancer therapy.

MLN4924 suppresses ESCC cell growth by targeting NAE

E119,20. However, whether other alternative targets against the

neddylation pathway might be used in ESCC therapy warrants

further exploration. In the present study, we demonstrate that

overexpression of NEDD8 negatively correlates with overall

survival in patients with ESCC. Furthermore, genetic down-

regulation of NEDD8 profoundly suppresses ESCC tumor

growth by triggering cell cycle arrest, DNA damage, and apop-

tosis. Our study validates NEDD8 as an alternative therapeutic

target against the neddylation pathway in ESCC therapy.

Materials and methods

Bioinformatics analysis of The Cancer Genome Atlas (TCGA) datasets

The mRNA expression level of NEDD8 and corresponding

clinicopathological data for esophageal carcinoma patients

in the TCGA esophageal carcinoma dataset were collected

from the UCSC xena website (http://xena.ucsc.edu/welcome-

to-ucsc-xena/) and the web-portal UALCAN21. RNA-seq

data for NAE and UBC12, as well as NEDD8 gene mutations

and amplifications, were downloaded from the TCGA eso-

phageal carcinoma cohort. Survival was analyzed with the

Kaplan-Meier method and compared with the log-rank test

in Statistical Program for Social Sciences software (SPSS)

Version 16.0. The association between copy number vari-ation (CNV) frequencies and NEDD8 gene expression was

calculated with standard ANOVA and Tukey’s honest signifi-

cant difference (HSD) tests.

Immunohistochemistry and evaluation of human ESCC tissue arrays

Human ESCC tissue arrays, which purchased from Shanghai

Outdo Biotech Co. Ltd. (Shanghai, China), were immunohis-

tochemically stained with antibody to NEDD8 (Cell Signaling,

Boston, MA). The tissue array sections were dehydrated

and subjected to peroxidase blocking, then incubated with

anti-NEDD8 at room temperature. The tissue array sections

were then stained with a GTVisionTM III Detection System/

Mo&Rb (Gene tech Company Limited) and counterstained

with hematoxylin. The histologic evaluation was based on

calculation of the percentage of positive tumor cells and the

staining intensity, as described previously22. The detailed clin-

icopathologic characteristics of patients with ESCC are pre-

sented in Supplementary Table S1. This study was approved by

the ethics committee of Huadong Hospital, Fudan University

(approval No. 2016K007).

Cell culture and generation of NEDD8-knockdown cells

The human esophageal epithelial cell line HET-1A and the

human ESCC cell lines Kyse30, Kyse150, Kyse510, Kyse450

and EC1 were cultured in Dulbecco’s modified Eagle’s medium

(Hyclone, Logan, UT, USA) containing 10% fetal bovine serum

(Biochrom AG, Berlin, Germany) and 1% penicillin-strep-

tomycin solution (Gibco, USA) at 37 °C under 5% CO2.

Negative control and NEDD8-knockdown cells were estab-

lished by infecting target cells with lentivirus particles pack-

aged by HEK293T cells, which were co-transfected with the

vector lenti-guide-puro (4.0 μg), and the packaging plasmids

AGP091 (3.0 μg) and AGP090 (1.2 μg), with PEI (Polyfectine)

Page 3: Overexpressed NEDD8 as a potential therapeutic target in

Cancer Biol Med Vol xx, No x Month 2021 3

reagent. Two different small guide RNA (sgRNA) oligonucle-

otides against NEDD8 were inserted into the lenti-guide-puro

vector (4.0 μg). The human sgRNA sequences for NEDD8

were as follows: 5′-ACCTGACTCACCTTGTCTGT-3′ and

5′-GAAGATGCTAATTAAAGTGA-3′. Targeted cells were

seeded 1 day before infection with lentiviral supernatant

along with 10 μg/mL polybrene (Sigma-Aldrich, St. louis,

MO). Infected cells were selected with 2 μg/mL puromycin

(Invitrogen, Carlsbad, CA) for 3 days.

RNA extraction and real-time PCR

Cells were harvested, and total RNA was extracted with an

Ultrapure RNA Kit from CWbiotech. Purified RNA (1.0 mg)

was reverse transcribed with PrimeScript RT Master

(Takara) according to the manufacturer’s instructions.

Real-time polymerase chain reaction (PCR) was performed

with SYBR Green Real-time PCR Master Mix (Applied

Biosystems) on an ABI 7900 thermocycler (Thermo Fisher

Scientific) according to the manufacturer’s instructions.

For each sample, the mRNA abundance was normalized

to that of β-actin. Sequences of primers were as follows:

β-actin: forward, 5′-CCGTTGCCCTGAGGCTCTTT-3′, reverse, 5′-CCTTCTGCATCCTGTCAGCAA-3′; NEDD8:

forward, 5′-AGACGCTGACCGGAAAGGA-3′, reverse,

5′-TCATCATTCATCTGCTTGCCAC-3′.

Western blotting and cycloheximide (CHX)-chase analysis

For western blotting, cell lysates (30 mg) were loaded on SDS-

PAGE gels and transferred onto nitrocellulose membranes

(Millipore), which were then incubated with the indicated

primary antibodies overnight. Corresponding secondary anti-

bodies were incubated with the membranes for 1 h, and the

membranes were then photographed with a Tanon 5200 visu-

alizer (Shanghai, China). Primary antibodies to the following

proteins were used: UBC12, UBA3, cullin1, cullin2, cullin5,

p21, NOXA (Abcam), NAE, cullin3, NEDD8, cullin4A, p27,

Wee1, p-H3, ORC1, CDT1, p-H2AX, t-H2AX, ATF4, CHOP,

DR5, cleaved-PARP, cleaved-caspase 8, and cleaved-caspase

3 (Cell Signaling, Boston, MA); cullin4B (Protein Tech); and

β-actin (Protein Tech). For CHX-chase assays, cells were

treated with 50 μg/mL CHX (Sigma) for the indicated times,

and the band density in western blots was quantified in Image

J software.

ATP-Lite cell viability assays

Cells were seeded in 96-well plates at a density of 1.5 × 103 cells

per well in triplicate and cultured for 72 h. Cell viability was

determined with ATP-Lite luminescence assays (PerkinElmer,

Norwalk, CT, USA) according to the manufacturer’s protocol.

Colony formation assays

Cells were seeded into 6-well plates (300 cells/well) in tripli-

cate, then incubated at 37 °C for 14 days. The 6-well plates were

washed with cold phosphate-buffered saline 3 times and fixed

with 4% paraformaldehyde at room temperature for 15 min.

Colonies on the plates were stained with 0.1% crystal violet at

room temperature for 30 min and then photographed under

an inverted microscope (Olympus, Tokyo, Japan). Colonies

with more than 50 cells were counted.

Cell migration and invasion assays

Cells were prewashed twice and seeded at a density of 5 × 104

cells per well in a 24-well Transwell polycarbonate filter plate

(8 μm pore size; Corning, Lowell, MA). The upper Transwell

chambers contained 200 μL serum-free medium, and the lower

chambers contained 600 μL medium with 10% fetal bovine

serum. In the invasion assays, a Matrigel polycarbonate mem-

brane (Corning) was placed in the upper Transwell chamber.

After 24 h incubation at 37 °C, the upper Transwell chambers

were fixed in 4% paraformaldehyde for 20 min and stained

with 0.1% crystal violet for 30 min. Cells on the outside of

the top chambers were photographed and counted under an

inverted microscope (Olympus, Tokyo, Japan).

LC-MS/MS analysis and MS quantification

LC-MS/MS analysis and label-free based MS quantification

of proteins in Kyse450 cells were performed as previously

described23. A Firmiana proteomics workstation was used to

process the raw MS data. Kyse450 lysates were digested into

peptides with trypsin, then subjected to MS analysis on a

Fusion Lumos instrument (Thermo Fisher Scientific). Raw

MS data were used to interrogate the NCBI human Refseq

protein database (released on 04-07-2013, 32,015 entries) with

the Mascot search engine (version 2.3, Matrix Science Inc.)

with a false discovery rate < 1% at the peptide and protein

level. The intensity-based label-free quantification (iBAQ)

Page 4: Overexpressed NEDD8 as a potential therapeutic target in

4 Xian et al. Therapeutic potential of NEDD8 in ESCC

algorithm was used for protein quantification. The iBAQ value

was transferred into a fraction of total protein iBAQ amount

per experiment (FOT). Then the FOT values were multiplied

by 106 and log10 transformed to obtain the FOT values for low

abundance proteins.

Flow cytometric analysis of the cell cycle and apoptosis

Cells were harvested with 0.25% trypsin without EDTA, then

washed twice with cold phosphate-buffered saline. Cells used

for cell cycle analysis were fixed in pre-cooled 70% ethanol at

−20 °C overnight. Afterward, cells were stained with propid-

ium iodide (PI, 36 μg/mL; Sigma, St. Louis, MO, USA) con-

taining RNase A (10 μg/mL; Sigma, St. Louis, MO, USA) in the

dark at 37 °C for 15 min, and detected with flow cytometry

(BD FACSVerse™). Cells used in apoptosis assays were stained

with Annexin V-fluorescein isothiocyanate (FITC) and PI

with an Annexin V-FITC Apoptosis Detection Kit (Beckman

Coulter) according to the manufacturer’s protocol, then

subjected to flow cytometric analysis. Data were analyzed in

FlowJo 7.6 software.

Gene silencing with siRNA

Kyse450 cells with NEDD8 knockdown were transfected

with the following siRNA oligonucleotides (synthesized by

GenePharma, Shanghai, China) and Lipofectamine 2000

(Invitrogen, Carlsbad, CA, USA) reagent: DR5-1: 5′-AAGACC

CUUGUGCUCGUUGUC-3′; DR5-2: 5′-CAGCCGUAGUCU

UGAUUGUTT-3′; NOXA-1: 5′-GGUGCACGUUUCAUCAA

UUUGTT-3′; NOXA-2: 5′-CCGGCAGAAACUUCUGAAUTT-3′; control: 5′-UUCUCCGAACGUGUCACGUTT-3′.

Subcutaneous-transplantation tumor model

NC (negative control) or NEDD8-knockdown cells (5 × 106

cells per mouse) were subcutaneously injected into 5-week-

old BAL b/c female nude mice purchased from the Shanghai

Lingchang Biotechnology Co., Ltd (Shanghai, China). Tumor

size was measured with calipers at the indicated time points

and calculated as (length × width2)/2. Mice were sacrificed

at the end of the study, and tumor tissues were harvested,

photographed, and weighed. Protein expression levels of the

tumor tissues were evaluated with immunoblotting analysis

with the indicated specific antibodies. Animal experiments

were performed in accordance with the National Guidelines

for Experimental Animal Welfare, with approval from the

Institutional Animal Care and Use Committee of Fudan

University (approval No. 202011008Z).

Statistical analysis

The numerical results are presented as means ± standard

deviations. Statistical significance for the comparison of para-

meters between 2 groups was evaluated with Student’s t test

in GraphPad Prism5 software (GraphPad Software, Inc., San

Diego, CA, USA). P < 0.05 was considered statistically signifi-

cant, and n.s. denotes not significant. For all tests, 3 levels of

significance (*P < 0.05, **P < 0.01, and ***P < 0.001) were

used.

Results

NEDD8 overexpression is predictive of poor overall survival in patients with ESCC

To evaluate the clinical relevance of NEDD8 expression in

esophageal carcinoma, we used the TCGA database to deter-

mine the levels of NEDD8 transcripts in esophageal carcinoma.

As shown in Figure 1A, the mRNA expression of NEDD8 was

significantly elevated in esophageal carcinoma and correlated

with nodal metastasis (normal vs. N0, P = 2.5627E-04; nor-

mal vs. N1, P = 8.0956E-07; normal vs. N2, P = 5.5654E-08;

normal vs. N3, P = 1.5798E-03; N0 vs. N1, P = 9.8279E-04;

N0 vs. N2, P = 9.1616E-04; N0 vs. N3, P = 5.7208E-01). The

NEDD8 mRNA expression in 2 histologic subtypes of esoph-

ageal carcinoma was significantly higher than that in normal

tissues (normal vs. EAC: P = 1.5926E-05; normal vs. ESCC:

P = 3.3506E-09) (Figure 1B). We further determined the

association between the CNV frequencies and NEDD8 gene

expression by standard ANOVA and Tukey’s HSD tests. CNV

gain was significantly associated with NEDD8 gene expression

in ESCC (P = 8.647E-04; Supplementary Figures S1A and

S1B). These results indicated that the upregulation of NEDD8

mRNA expression in ESCC might be partially explained by the

CNV gain. Furthermore, Kaplan-Meier analysis showed that

patients with ESCC with higher NEDD8 expression had lower

overall survival rates (P = 0.028, log-rank test; Figure 1C).

Spearman tests indicated a statistically positive correlation

Page 5: Overexpressed NEDD8 as a potential therapeutic target in

Cancer Biol Med Vol xx, No x Month 2021 5

0

0

Tumor tissues

×10

×200

Adjacent tissues300

200

100

0 0

0.2

0.4

0.6

0.8

1

NED

D8

IHC

scor

e

Cum

ulat

ive

surv

ival

0.25

0.5

0.75

1Low expressionHigh expression

Low expressionHigh expression

Normal(n = 11)

P = 0.028n = 40

n = 40

500

ESCC ESCC

Tumor Adjacent 0 20 40 60 80

P < 0.001

P = 0.026n = 73

n = 22

NEDD8

1,000

Time (days)

Time (months)

NEDD8

NEDD8

NAE

UBC12

1,500 2,000

Normal(n = 11)

Adenocarcinoma(n = 89)

Squamous cellcarcinoma(n = 95)

N0(n = 76)

***

******

*** ***

***

***

**

N1(n = 69)

Expression of NEDD8 in esophageal cancerbased on nodal metastasis status

Expression of NEDD8 in esophageal cancerbased on tumor histology

Correlation between NEDD8 and neddylation enzymes in ESCC

Spearman’scorrelation

coefficient rho

0.3115 4.66E-03

0.3933 2.83E-04

P value

n.s.

N2(n = 12)

N3(n = 8)

50

Cum

ulat

ive

surv

ival

Tran

scrip

t per

mill

ion

100

150

200

250

300

350

Tran

scrip

t per

mill

ion

A

C

E F G

D

B

400

350

300

250

200

150

100

50

0

Figure 1 Overexpression of NEDD8 is predictive of poor overall survival in patients with ESCC. (A) The mRNA expression level of NEDD8 was higher in esophageal carcinoma than in normal esophageal tissues. N0, N1, N2, and N3 denote nodal metastasis status. (B) The mRNA level of NEDD8 was higher in ESCC and EAC than in normal esophageal tissues. (C) Kaplan-Meier curve analysis was performed to determine overall survival in patients with ESCC according to the NEDD8 mRNA expression data in the TCGA RNA-Seq database. (D) The correlation between NEDD8 and neddylation enzymes (NAE and UBC12) was analyzed in ESCC with the Spearman test. (E, F) A human ESCC tissue array was immunohistochemically stained with antibody specific to NEDD8 (E), and the difference in expression of NEDD8 in the ESCC tissues was calculated according to the histologic evaluation of tumors and adjacent normal tissues (F). Scale bar for ×10 images, 500 μm; Scale bar for ×200 images, 25 μm. (G) Kaplan-Meier curves based on NEDD8 protein expression in patients with ESCC. **P < 0.01, ***P < 0.001, n.s. = not significant.

Page 6: Overexpressed NEDD8 as a potential therapeutic target in

6 Xian et al. Therapeutic potential of NEDD8 in ESCC

between NEDD8 and the neddylation enzymes NAE and

UBC12 (P < 0.01; Figure 1D), thus suggesting that NEDD8

expression and the neddylation pathway were positively cor-

related in ESCC.

In addition, we determined the NEDD8 protein expression

in human ESCC tissue arrays through IHC. The NEDD8 pro-

tein expression was higher in ESCC tissues than in adjacent

normal tissues (Figure 1E). Advanced histologic evaluation

showed that the NEDD8 expression was significantly elevated

in patient ESCC tissues (P < 0.001; Figure 1F). Furthermore,

Kaplan-Meier analysis showed that patients with ESCC with

high NEDD8 expression had poorer overall survival than those

with low expression (P = 0.026, log-rank test; Figure 1G and

Supplementary Table S1). Moreover, we detected the mRNA and protein levels of NEDD8 in the human esophageal epi-thelial cell line HET-1A and the 5 ESCC cell lines Kyse30, Kyse150, Kyse450, Kyse510 and EC1. The mRNA and protein levels of NEDD8 were higher in ESCC cell lines than in HET-1A (Supplementary Figure S2A and S2B). The

above findings demonstrated that NEDD8 is overexpressed in

ESCC and is associated with poorer overall survival, thus high-

lighting NEDD8 as a potential anti-ESCC target.

NEDD8 knockdown suppresses the malignant phenotype of ESCC cells

To investigate the inhibitory effects of NEDD8 knockdown on

the malignant phenotype of ESCC, we silenced NEDD8 with

the CRISPR/Cas9 system in the ESCC cell lines Kyse450 and

EC1 (Figure 2A). NEDD8 knockdown significantly inhibited

the proliferation of these 2 ESCC cell lines (Figure 2B and Supplementary Figure S3). The colony formation abilities

of these 2 cell lines were significantly repressed after NEDD8

knockdown (Figure 2C and 2D). Moreover, the Transwell

migration and invasion abilities were significantly impaired

in NEDD8-knockdown Kyse450 and EC1 cells (Figure 2E

and 2F). These results demonstrated that downregulation of

NEDD8 suppresses ESCC cell proliferation and survival, thus

indicating that NEDD8 is necessary to maintain the malignant

phenotype of ESCC cells.

NEDD8 knockdown triggers multiple tumor-suppressive processes in ESCC cells

To explore the mechanisms underlying the NEDD8 knock-

down-induced cell malignant phenotype arrest, we performed

quantitative proteomic analysis in NEDD8-knockdown

Kyse450 cells. Gene ontology (GO) analysis indicated that

several biological processes associated with cell survival—

including negative regulation of cell cycle process, regulation

of cell cycle arrest, extrinsic apoptotic signaling pathway via

death domain receptors, cellular response to DNA damage

stimulus, and DNA damage checkpoint—were significantly

elevated in NEDD8-knockdown ESCC cells (Figure 3A and

3B). The regulation of G2/M transition in the mitotic cell

cycle, protein neddylation, regulation of mitotic cell cycle

phase transition, positive regulation of protein ubiquitin ligase

activity and negative regulation of endothelial cell apoptotic

process were significantly inhibited (Figure 3C and 3D). These

findings were further supported by PI staining and fluores-

cence activated cell sorting (FACS) analysis, which showed

that NEDD8 knockdown significantly increased the cell

populations in the G2/M phase in both Kyse450 and EC1 cell

lines (Figure 3E–3G). These results suggested that NEDD8

knockdown triggers DNA damage, cell cycle arrest, and apop-

tosis, thereby suppressing ESCC cell growth.

NEDD8 knockdown triggers G2 phase cell cycle arrest due to the accumulation of CRL substrates in ESCC cells

Because CRL E3 ligases are the best characterized substrates

of neddylation, we determined the neddylation levels of cull-

ins (the essential components of CRLs) after NEDD8 knock-

down7. Immunoblotting analysis showed that downregulation

of NEDD8 significantly decreased the neddylation levels of

cullin1, 2, 3, 4A, 4B, and 5 (Figure 4A). However, NEDD8 knock-

down had no effect on the expression levels of NAE1, UBA3,

and UBC12 in both Kyse450 and EC1 cells (Supplementary

Figure S4). Furthermore, the cell cycle inhibitors p21, p27, and

Wee1, substrates of CRL E3 ligases, significantly accumulated

after NEDD8 knockdown, whereas phosphorylated histone

H3 (p-H3, ser10) was sharply downregulated (Figure 4B).

These results were consistent with our above findings indicat-

ing that NEDD8-knockdown ESCC cells were arrested in G2

phase and did not enter M phase (Figure 3E–3G). To explore

whether NEDD8 knockdown prolonged the half-lives of these

cell cycle inhibitors as a result of CRLs inactivation, we used

CHX to analyze the protein half-lives of p21, p27, and Wee1

after NEDD8 knockdown. As shown in Figure 4C and 4D,

the half-lives of p21, p27, and Wee1 increased after NEDD8

knockdown. These results indicated that NEDD8 knockdown

Page 7: Overexpressed NEDD8 as a potential therapeutic target in

Cancer Biol Med Vol xx, No x Month 2021 7

Kyse450A

C

E F

B

D

EC1

NEDD8

NEDD80

50

100

***

******

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

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

*****

***

******

150

0

50

100

150

NC KD1

Kyse450

Kyse450

Kyse450 Kyse450EC1 EC1

Kyse450

EC1

Kyse450

EC1

150

100

50

0

150

100

50

0

Kyse450

EC1

EC1

ATP-lite

EC1

KD2 NC KD1 KD2

NEDD8 NC KD1 KD2 NC KD1 KD2

NEDD8 NC KD1 KD2 NC KD1 KD2 NEDD8 NC KD1 KD2 NC KD1 KD2

β-actin

NED

D8

conj

ugat

edpr

otei

ns

Perc

enta

ge (%

)Pe

rcen

tage

(%)

Perc

enta

ge (%

)

Perc

enta

ge (%

)

NC

NC NEDD8-KD1 NEDD8-KD2

NC NEDD8-KD1

Migration

Migration Invasion

NEDD8-KD2 NC NEDD8-KD1 NEDD8-KD2

NC

Colony formationColony formation

Invasion

KD1 KD1KD2 KD2

Figure 2 NEDD8 knockdown suppresses the malignant phenotype of ESCC cells. (A) Global protein neddylation levels were suppressed in NEDD8-knockdown Kyse450 and EC1 cells. Immunoblotting was used to analyze the global protein neddylation levels after NEDD8 knockdown, with β-actin as a loading control. (B) Cell viability analysis with ATP-Lite assays after NEDD8 knockdown. (C, D) Colony formation in NEDD8-knockdown Kyse450 and EC1 cells, determined by crystal violet staining and counting. Representative images are shown. (E, F) NEDD8-knockdown Kyse450 and EC1 cells were used to determine the migration and invasion abilities, as described in the Materials and Methods. Representative images are shown; scale bar = 200 µm. Average values with standard deviations of triplicate experiments are shown. NC, negative control; KD, NEDD8 knockdown; **P < 0.01, ***P < 0.001.

Page 8: Overexpressed NEDD8 as a potential therapeutic target in

8 Xian et al. Therapeutic potential of NEDD8 in ESCC

0NEDD8

Perc

enta

ge (%

)Co

unt

Perc

enta

ge (%

)

NC

50K

EC1

Kyse

450

0

30

60

90

120

0

40

20

60

80

100

0

40

20

60

80

100

0

50

100

150

Negative regulation of cell cycle process

Regulation of G2/M transition ofmitotic cell cycle

Regulation of mitotic cell cycle phase transitionPositive regulation of ubiquitin protein

ligase activityNegative regulation of endothelial cell

apoptotic process

Protein neddylation

Regulation of cell cycle arrestExtrinsic apoptotic signaling pathway

via death domain receptorsCellular response to DNA damage stimulus

DNA damage checkpoint

0 1

GO_biological_processTerm

Negative regulation of cell cycleprocess (GO:0010948)

Regulation of G2/M transition ofmitotic cell cycle (GO:0010389)

Protein neddylation (GO:0045116)

Regulation of mitotic cell cyclephase transition (GO:1901990)

Positive regulation of ubiquitinprotein ligase activity

(GO:1904668)Negative regulation of endothelial

cell apoptotic process(GO:2000352)

1.32E-03

7.09E-05

4.35E-04

5.23E-04

1.83E-03

1.04E-02

17.2549

18.0122

17.9949

8.0180

8.9012

16.6661

16.6366

16.3987

54.3193

24.4086

2.54E-03

5.99E-03

6.48E-03

2.94E-02

Regulation of cell cycle arrest(GO:0071156)

Extrinsic apoptotic signalingpathway via death domain

receptors (GO:0008625)Cellular response to DNA damage

stimulus (GO:0006974)DNA damage checkpoint

(GO:0000077)

Combinedscore

P value

Term Combinedscore

P valueGO_biological_process

–Log(P value)2 3

–Log(P value)0 1 2 3 4

0

50

100

150

0

50

100

150

200

100K 200K

G2:22.27% G2:33.29% G2:35.80%

G2:28.1% G2:40.26% G2:39.74%

250K 50K 100K

PI

200K 250K 50K 100K 200K 250K

50K 100K

NC NEDD8-KD1 NEDD8-KD2

200K 250K 50K 100K 200K 250K 50K 100K 200K 250K

28.1% 40.26% 39.74% 22.27% 33.29% 35.80%

KD1

Kyse450F

E

C

A B

D

G EC1

G1 S G2/M G1 S G2/M

KD2 NEDD8 NC KD1 KD2

50

100

150

0

50

100

150

Figure 3 NEDD8 knockdown results in G2 phase cell cycle arrest. (A, B) Gene ontology (GO) analysis based on proteomics analysis was used to determine upregulated processes. (C, D) Downregulated biological processes were shown. (E–G) PI staining and FACS analysis were used to analyze the cell cycle profiles after NEDD8 depletion in Kyse450 and EC1 cells. NEDD8 knockdown induced ESCC cells arrest in G2 phase (presented in blue in E). The percentage of each cell cycle phase is shown in F and G.

Page 9: Overexpressed NEDD8 as a potential therapeutic target in

Cancer Biol Med Vol xx, No x Month 2021 9

NEDD8A

C

B

D

NEDD8

p27

p21

Wee1

p-H3

NC KD1 KD2 NC KD1 KD2NEDD8-cullin1

NEDD8-cullin2

NEDD8-cullin3

NEDD8-cullin4A

NEDD8-cullin4B

NEDD8-cullin5

NED

D8

conj

ugat

edpr

otei

nsRe

lativ

e pr

otei

n le

vel

CHX(h)

p27

p21

Wee1

cullin1

cullin2

cullin3

cullin4A

cullin4B

cullin5

β-actin

β-actin

1.5

1.0

0.5

0.0

Rela

tive

prot

ein

leve

l

1.5

1.0

0.5

0.0

Rela

tive

prot

ein

leve

l

Rela

tive

prot

ein

leve

l

1.5

1.0

0.5

0.0

1.5

1.0

0.5

0.0

Rela

tive

prot

ein

leve

l

1.5

1.0

0.5

0.0

Rela

tive

prot

ein

leve

l

1.5

1.0

0.5

0.00 2 4

Time (h)6 8

0 2 4Time (h)

6 8 0 2 4Time (h)

6 8 0 2 4Time (h)

6 8

0 2 4Time (h)

6 8 0 2 4Time (h)

6 8

NED

D8

conj

ugat

edpr

otei

ns

CHX(h)

p27

p21

Wee1

β-actin

β-actin

NC

NC0 1.5 3 6 0 1.5 3 6 0 1.5 3 6 0 1.5 3 6 0 1.5 3 6 0 1.5 3 6

NEDD8-KD1 NEDD8-KD2 NC NEDD8-KD1 NEDD8-KD2

KD1Kyse450

Kyse450 EC1

EC1

Kyse450

Kyse450-p27

EC1-p27 EC1-p21 EC1-Wee1

NCN8-KD1N8-KD2

NCN8-KD1N8-KD2

NCN8-KD1N8-KD2

NCN8-KD1N8-KD2

NCN8-KD1N8-KD2

NCN8-KD1N8-KD2

Kyse450-p21 Kyse450-Wee1

EC1

KD2 NC KD1 KD2

Figure 4 NEDD8 knockdown blocks the degradation of CRL substrates. (A) NEDD8 knockdown suppressed neddylation of cullin1, 2, 3, 4A, 4B, and 5 in Kyse450 and EC1 cells. (B) NEDD8 knockdown induced the accumulation of p27, p21, and Wee1, and a decrease in p-H3 in 2 ESCC cell lines. (C, D) The half-lives of p27, p21, and Wee1 were prolonged after NEDD8 knockdown in Kyse450 and EC1 cells. Cells were treated with CHX to block protein synthesis for the indicated times and then subjected to immunoblotting of p27, p21, and Wee1, with β-actin as a loading control. The protein levels of p27, p21, and Wee1 were quantified in comparison with β-actin, through intensity calculations in Image J software. NC, Negative control; KD, NEDD8 knockdown.

Page 10: Overexpressed NEDD8 as a potential therapeutic target in

10 Xian et al. Therapeutic potential of NEDD8 in ESCC

NEDD8 NC KD1 KD2 NC KD1 KD2

102

102 103

NC NEDD8-KD1

NEDD8

NEDD8-KD1 NEDD8-KD2

NEDD8 NC KD1 KD2 NC KD1 KD2

ATF4

CHOP

DR5

NOXA

C-CASP8

C-CASP3

C-PARP

β-actin

0

5

10

40

30

20

10

NC

NEDD8

NEDD8

C-CASP8

C-PARP

NOXA

C-CASP3

C-PARP

β-actin β-actin

DR5

NED

D8

conj

ugat

edpr

otei

ns

NED

D8

conj

ugat

edpr

otei

ns

NC KD1 KD2 NC KD1 KD2 NC KD1 KD2

NC KD1 KD2 NC KD1 KD2 NC KD1 KD2

NEDD8-KD1 NEDD8-KD2n.s.

n.s.n.s.

n.s.

NC

siControl

siControl siNOXA-1 siNOXA-2

siDR5-1

*** ***

**

siDR5-20

40

30

20

10

0

15

20

NC KD1 KD2 NC KD1 KD2

NEDD8-KD2Kyse450

30

20

10

***

*********

EC1

104 105

102 103 104 105 102 103 104 105 102 103 104 105

102 103 104 105 102 103 104 105

103

104

105

102

103

104

105

102

103

104

105

102

103

104

105

102

103

104

105

102

103

104

105

PI

Apop

tosi

s (%

)

EC1

Apop

tosi

s (%

)Ap

opto

sis

(%)

Kyse

450

Kyse450A

C

F

D

E

G

BEC1

CDT1

ORC1

p-H2AX

t-H2AX

Kyse450

Kyse450

Kyse450

siControl siDR5-1 siDR5-2

siControl siNOXA-1 siNOXA-2

Kyse450

Kyse450

EC1

Annexin-V FITC

β-actin

Figure 5 NEDD8 knockdown triggers DR5-dependent apoptosis in ESCC cells. (A) The expression levels of the DNA damage response proteins CDT1, ORC1, and phosphorylated/total H2AX in NEDD8-knockdown Kyse450 and EC1 cells were determined by immuno-blotting, with β-actin as a loading control. (B) NEDD8 knockdown triggered apoptosis of ESCC cells, as determined by Annexin V–FITC/PI double-staining analysis. (C) NEDD8 knockdown induced accumulation of ATF4, CHOP, DR5, NOXA, cleaved-caspase 8, cleaved-caspase 3, and cleaved-PARP, as assessed by immunoblotting. (D) Downregulation of DR5 rescued apoptotic induction in NEDD8-knockdown Kyse450 cells. NEDD8-knockdown Kyse450 cells transfected with siControl or siDR5 and then subjected to Annexin V–FITC/PI double-staining analysis. (E) NOXA knockdown had no rescue effect on apoptotic induction in NEDD8-knockdown Kyse450 cells. siControl or siNOXA were transfected

Page 11: Overexpressed NEDD8 as a potential therapeutic target in

Cancer Biol Med Vol xx, No x Month 2021 11

induces G2 phase cell cycle arrest, owing to the accumulation

of the CRLs substrates p21, p27, and Wee1.

NEDD8 knockdown induces DNA damage and activates DR5-dependent apoptosis in ESCC cells

Our aforementioned GO analysis suggested that DNA dam-

age effects and the apoptosis response were activated after

NEDD8 knockdown (Figure 3A–3D). Therefore, we exam-

ined the DNA damage effects and apoptosis response in

NEDD8-knockdown ESCC cells. As shown in Figure 5A,

the DNA replication factor Cdt1 (CDT1), origin recognition

complex subunit 1 (OCR1), and DNA damage marker phos-

phorylated H2AX accumulated after NEDD8 knockdown

in both Kyse450 and EC1 cells. These results suggested that

downregulation of NEDD8 induced DNA re-replication

stress and subsequent DNA damage. NEDD8 knockdown also

resulted in a shrunken morphology of apoptotic cells in both

Kyse450 and EC1 cell lines (data not shown). Consistently, the

Annexin V-positive cell populations significantly increased

after NEDD8 knockdown in both cell lines, thus indicating

that NEDD8 knockdown increased the apoptotic response of

ESCC cells (Figure 5B). Further mechanistic studies showed

that NEDD8 knockdown induced accumulation of the CRLs

substrate activating transcription factor 4 (ATF4) and the

important apoptosis mediator phorbol-12-myristate-13-ace-

tate-induced protein 1 (NOXA), as well as the classical apop-

totic hallmarks cleaved-caspase 3 and cleaved-PARP (Figure

5C). After ATF4 accumulation, the transcription factor C/

EBP-homologous protein (CHOP), a classical downstream

target of ATF4, was activated and further induced the expres-

sion of DR5, which in turn activated cleaved-caspase 8 and

triggered extrinsic apoptosis (Figure 5C). To further explore

the mechanisms underlying the apoptosis induced by NEDD8

knockdown, we knocked down NOXA and DR5 with siRNA

silencing. Only DR5 knockdown resulted in a partial rescue of

apoptosis induced by NEDD8 knockdown (Figure 5D and 5F,

Supplementary Figure S5). In contrast, NOXA knockdown

did not rescue the apoptotic induction in NEDD8-knockdown

Kyse450 cells (Figure 5E and 5G). These results indicated that

NEDD8 deficiency induces DNA damage and triggers DR5-

dependent extrinsic apoptosis in ESCC cells.

NEDD8 knockdown suppresses ESCC tumor growth in vivo

To investigate the therapeutic potential of NEDD8 silencing

in vivo, we established a subcutaneous-transplantation tumor

model using NEDD8-knockdown EC1 cells. Downregulation

of NEDD8 significantly inhibited tumor growth, according

to the tumor growth curve (P < 0.001; Figure 6A). Moreover,

compared with the control group mice, which developed

100% (7/7) large tumors, only 71.4% (5/7) of the NEDD8-

knockdown group mice developed small tumors (Figure 6B);

these results were further supported by tumor weight assess-

ment (P < 0.001; Figure 6C). We further addressed the poten-

tial mechanisms underlying the antitumor activity of NEDD8

knockdown in vivo. Downregulation of NEDD8 profoundly

inhibited cullin neddylation, thus indicating the inactivation

of CRLs. Consequently, the CRLs substrates p27, p21, and

Wee1 clearly accumulated in the NEDD8-knockdown group

(Figure 6D). Collectively, our results indicated that knock-

down of NEDD8 leads to CRLs inactivation and the accumu-

lation of tumor-suppressive CRL substrates, thus inhibiting

tumor growth in vivo.

Discussion

ESCC is one of the deadliest digestive system cancers world-

wide24. However, effective therapeutic strategies for ESCC are

lacking25,26. Therefore, developing novel and effective anti-

ESCC therapeutic targets is urgently needed. The neddylation

pathway is overactivated in multiple human cancers, and inhi-

bition of the neddylation pathway by MLN4924 has provided

an attractive anticancer therapeutic strategy27. However, drug

resistance and potential tumor promotion of MLN4924 have

led to a demand for new neddylation pathway anticancer tar-

gets15,28. In the present study, we demonstrated that NEDD8

was elevated in ESCC, and patients with high expression of

into Kyse450 cells, which were subjected to Annexin V-FITC/PI double-staining analysis. The cartogram showed no significance between 2 matching groups. (F) Kyse450 cells with siControl or siDR5 were subjected to immunoblotting for cleaved-caspase 8, cleaved-caspase 3, cleaved-PARP, and DR5, with β-actin as a loading control. (G) Kyse450 cells with siControl or siNOXA were subjected to immunoblotting of cleaved-PARP and NOXA, with β-actin as a loading control. Average values with standard deviations of triplicate experiments are shown. NC, negative control, KD, NEDD8 knockdown; *P < 0.05, ***P < 0.001, n.s. = not significant.

Page 12: Overexpressed NEDD8 as a potential therapeutic target in

12 Xian et al. Therapeutic potential of NEDD8 in ESCC

0

0 10 20

Time (days)

EC1A

C

E

B

D

EC1

EC1

NC

1 2 3 4 1 2 3 4

NEDD8-KD

EC1

***

***

Tum

or v

olum

e (m

m3 )

Tum

or w

eigh

t (g)

NC

NEDD8-KD

30

1 cm

300

1.0

0.8

0.6

0.4

0.2

0.0

CRISPR/Cas9

CRLs(Cullin1, 2, 3, 4A, 4B, 5)

N8

NEDD8

NC NEDD8-KD

p21 p27

Wee1

ORC1

DR5 NOXA

CDT1

600

900

1,200

1,500

No. 1 2 3 4 5 6 7

NC

NEDD8-KD

NEDD8-cullin2cullin2

p27

p21

Wee1

G2

G1

S

M

Tumorgrowth

G2/M arrest

DNA damage

Apoptosis

NED

D8

conj

ugat

edpr

otei

ns

β-actin

Figure 6 NEDD8 knockdown suppresses ESCC tumor growth in vivo. The subcutaneous-transplantation tumor model was established by using NEDD8-knockdown EC1 cells. (A) Tumor size was measured with calipers at the indicated time points and converted to a tumor growth curve. (B) Tumor tissues were harvested and photographed on the day of sacrifice. Scale bar = 1 cm. (C) The tumor weight was measured. (D) Proteins extracted from tumor tissues were analyzed by immunoblotting against cullin2, p27, p21, Wee1, and NEDD8, with β-actin as a loading control. (E) Working model in which NEDD8 knockdown suppresses the tumor growth of ESCC. NC, negative control, KD, NEDD8 knockdown; ***P < 0.001.

Page 13: Overexpressed NEDD8 as a potential therapeutic target in

Cancer Biol Med Vol xx, No x Month 2021 13

NEDD8 had poorer overall survival. We determined the poten-

tial of NEDD8 as an alternative therapeutic target for ESCC

therapy. Our results showed that downregulation of NEDD8

activated a series of biological tumor-suppressive processes.

Furthermore, NEDD8 silencing displayed anticancer effects

in vitro and in vivo, thus confirming NEDD8 as a potential

effective therapeutic target for ESCC (Figure 6E).

A previous study has reported that NEDD8 is upregulated

in multiple cancers, and NEDD8 overexpression is associated

with poor prognosis29. In the present study, we showed that

the mRNA and protein expression levels of NEDD8 were sig-

nificantly elevated in ESCC, and overexpressed NEDD8 nega-

tively correlated with overall survival in patients with ESCC,

thus indicating that NEDD8 may be a useful biomarker for

ESCC. In addition, NEDD8 knockdown not only obviously

suppressed ESCC cell proliferation but also profoundly inhib-

ited the migration and invasion abilities of ESCC cells. In vivo

experiments further showed that downregulation of NEDD8

significantly repressed ESCC tumor growth. Therefore, our

study suggests that NEDD8 may be a promising therapeutic

target and provides a scientific basis for the development of

specific inhibitors targeting NEDD8.

Mass spectrometry analysis identified 1219 proteins up-

regulated > 2-fold and 1108 proteins down-regulated < 2-fold

in NEDD8-knockdown ESCC cells. On the basis of our quanti-

tative proteomic analysis results, we demonstrated that down-

regulation of NEDD8 affects the cell cycle. Mechanistically, we

found that NEDD8 knockdown inactivates CRLs and induces

the accumulation of cell cycle inhibitor proteins (p21, p27,

and Wee1) in ESCC cells. Accordingly, cell cycle progression is

blocked. Proteomic analysis also indicated that downregulation

of NEDD8 induced extrinsic apoptosis in ESCC cells. Indeed,

knockdown NEDD8 induced extrinsic apoptosis via the accu-

mulation of DR5, owing to the upregulation of ATF4, a sub-

strate of CRL1, and its downstream target transcription factor

CHOP30. Further rescue experiments suggested that NEDD8

deficiency induced extrinsic apoptosis in a manner dependent

on DR5 in ESCC cells. Therefore, our results demonstrated

that genetic downregulation of NEDD8 has anti-ESCC effects

and mechanisms similar to those of MLN4924, and confirmed

NEDD8 as a therapeutic target in ESCC.

Conclusions

Overall, our findings not only validate NEDD8 as a promising

therapeutic target against ESCC but also provide a basis for

NEDD8 inhibitor development. We believe that the develop-

ment of novel anti-ESCC strategies targeting NEDD8 would

shed new light on ESCC treatment.

Grant support

This work was supported by grants from the National Natural

Science Foundation of China (grant No. 81602072, 81902380,

81820108022, and 81625018), Innovation Program of Shanghai

Municipal Education Commission (grant No. 2019-01-07-

00-10-E00056), Program of Shanghai Academic/Technology

Research Leader (grant No. 18XD1403800), National High

Technology Research and Development Program of China

(grant No. 2015AA021107-019), Scientific Research Project

of Shanghai Science and Technology Commission (grant No.

18411960600), Shanghai Technological Innovation Action

Projects (grant No. 18411950800), Shanghai ‘Rising Stars of

Medical Talent’ Youth Development Program, Outstanding

Youth Medical Talents, 2018, and the Shanghai Sailing Program

(grant No. 17YF1405000).

Conflict of interest statement

No potential conflicts of interest are disclosed.

References

1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre L, Jemal A.

Global cancer statistics 2018: Globocan estimates of incidence and

mortality worldwide for 36 cancers in 185 countries. CA Cancer J

Clin. 2018; 68: 394-424.

2. Huang F, Yu S. Esophageal cancer: risk factors, genetic association,

and treatment. Asian J Surg. 2018; 41: 210-5.

3. Rustgi A, El-Serag H. Esophageal carcinoma. N Engl J Med. 2014;

371: 2499-509.

4. van der Veen A, Ploegh H. Ubiquitin-like proteins. Annu Rev

Biochem. 2012; 81: 323-57.

5. Bosu D, Kipreos E. Cullin-ring ubiquitin ligases: global regulation

and activation cycles. Cell Div. 2008; 3: 7.

6. Deshaies R, Emberley E, Saha A. Control of cullin-ring ubiquitin

ligase activity by nedd8. Subcell Biochem. 2010; 54: 41-56.

7. Zhao Y, Sun Y. Cullin-ring ligases as attractive anti-cancer targets.

Curr Pharm Design. 2013; 19: 3215-25.

8. Brockmann M, Doengi M, Einsfelder U, Koerber N, Refojo D, Stein

V. Neddylation regulates excitatory synaptic transmission and

plasticity. Sci Rep. 2019; 9: 17935.

9. Li L, Kang J, Zhang W, Cai L, Wang S, Liang Y, et al. Validation of

NEDD8-conjugating enzyme UBC12 as a new therapeutic target in

lung cancer. Ebiomedicine. 2019; 45: 81-91.

Page 14: Overexpressed NEDD8 as a potential therapeutic target in

14 Xian et al. Therapeutic potential of NEDD8 in ESCC

10. Zhao B, Gao C, Shi D, Mao J, Zhao J, Guo L, et al. Knockdown of

Nedd8-conjugating enzyme UBE2M suppresses the proliferation

and induces the apoptosis of intrahepatic cholangiocarcinoma

cells. Oncol Rep. 2019; 42: 2670-9.

11. Fotouhi O, Kjellin H, Juhlin C, Pan Y, Vesterlund M, Ghaderi M,

et al. Proteomics identifies neddylation as a potential therapy target

in small intestinal neuroendocrine tumors. Oncogene. 2019; 38:

6881-97.

12. Zhang W, Liang Y, Li L, Wang X, Yan Z, Dong C, et al. The Nedd8-

activating enzyme inhibitor MLN4924 (TAK-924/Pevonedistat)

induces apoptosis via c-Myc-Noxa axis in head and neck squamous

cell carcinoma. Cell Prolif. 2019; 52: e12536.

13. Zhou L, Jiang Y, Luo Q, Li L, Jia L. Neddylation: a novel modulator

of the tumor microenvironment. Mol Cancer. 2019; 18: 77.

14. Swords R, Coutre S, Maris M, Zeidner J, Foran J, Cruz J, et al.

Pevonedistat, a first-in-class NEDD8-activating enzyme inhibitor,

combined with azacitidine in patients with AML. Blood. 2018; 131:

1415-24.

15. Milhollen M, Thomas M, Narayanan U, Traore T, Riceberg J,

Amidon B, et al. Treatment-emergent mutations in NAEbeta confer

resistance to the NEDD8-activating enzyme inhibitor MLN4924.

Cancer cell. 2012; 21: 388-401.

16. Verma S, Singh A, Mishra A. Molecular dynamics investigation on

the poor sensitivity of A171T mutant NEDD8-activating enzyme

(NAE) for MLN4924. J Biomol Struct Dyn. 2014; 32: 1064-73.

17. Zhou Q, Li H, Li Y, Tan M, Fan S, Cao C, et al. Inhibiting

neddylation modification alters mitochondrial morphology and

reprograms energy metabolism in cancer cells. JCI Insight. 2019; 4:

e121582.

18. Zhou X, Tan M, Nyati M, Zhao Y, Wang G, Sun Y. Blockage of

neddylation modification stimulates tumor sphere formation

in vitro and stem cell differentiation and wound healing in vivo.

Proc Natl Acad Sci U S A. 2016; 113: E2935-44.

19. Lin S, Shang Z, Li S, Gao P, Zhang Y, Hou S, et al. Neddylation

inhibitor MLN4924 induces G(2) cell cycle arrest, DNA damage

and sensitizes esophageal squamous cell carcinoma cells to

cisplatin. Oncol Lett. 2018; 15: 2583-9.

20. Chen P, Hu T, Liang Y, Li P, Chen X, Zhang J, et al. Neddylation

inhibition activates the extrinsic apoptosis pathway through

ATF4-CHOP-DR5 axis in human esophageal cancer cells. Clin

Cancer Res. 2016; 22: 4145-57.

21. Chandrashekar D, Bashel B, Balasubramanya S, Creighton C,

Ponce-Rodriguez I, Chakravarthi B, et al. Ualcan: a portal for

facilitating tumor subgroup gene expression and survival analyses.

Neoplasia. 2017; 19: 649-58.

22. Wang S, Xian J, Li L, Jiang Y, Liu Y, Cai L, et al. NEDD8-conjugating

enzyme UBC12 as a novel therapeutic target in esophageal squamous

cell carcinoma. Signal Transduct Target Ther. 2020; 5: 123.

23. Ni X, Tan Z, Ding C, Zhang C, Song L, Yang S, et al. A region-

resolved mucosa proteome of the human stomach. Nat Commun.

2019; 10: 39.

24. Ajani J, D’Amico T, Bentrem D, Chao J, Corvera C, Das P, et al.

Esophageal and esophagogastric junction cancers, version 2.2019,

nccn clinical practice guidelines in oncology. J Natl Compr Canc

Netw. 2019; 17: 855-83.

25. Yang H, Liu H, Chen Y, Zhu C, Fang W, Yu Z, et al. Neoadjuvant

chemoradiotherapy followed by surgery versus surgery alone

for locally advanced squamous cell carcinoma of the esophagus

(NEOCRTEC5010): A phase iii multicenter, randomized, open-

label clinical trial. J Clin Oncol. 2018; 36: 2796.

26. Abnet C, Arnold M, Wei W. Epidemiology of esophageal squamous

cell carcinoma. Gastroenterology. 2018; 154: 360-73.

27. Zhou L, Zhang W, Sun Y, Jia L. Protein neddylation and its

alterations in human cancers for targeted therapy. Cell Signal. 2018;

44: 92-102.

28. Zhou Q, Sun Y. MLN4924: additional activities beyond neddylation

inhibition. Mol Cell Oncol. 2019; 6: e1618174.

29. Jiang Y, Cheng W, Li L, Zhou L, Liang Y, Zhang W, et al. Effective

targeting of the ubiquitin-like modifier NEDD8 for lung

adenocarcinoma treatment. Cell Biol Toxicol. 2020; 36: 349-64.

30. Han J, Back S, Hur J, Lin Y, Gildersleeve R, Shan J, et al. ER-stress-

induced transcriptional regulation increases protein synthesis

leading to cell death. Nat Cell Biol. 2013; 15: 481-90.

Cite this article as: Xian J, Wang S, Jiang Y, Li L, Cai L, Chen P, et al.

Overexpressed NEDD8 as a potential therapeutic target in esophageal

squamous cell carcinoma. Cancer Biol Med. 2021; x: xx-xx. doi: 10.20892/j.

issn.2095-3941.2020.0484

Page 15: Overexpressed NEDD8 as a potential therapeutic target in

Cancer Biol Med Vol xx, No x Month 2021 15

Supplementary materials

A

B

Gene CNV gain CNV loss

NEDD8 12/95 (12.60%) 8/95 (8.42%)

Copy number variation frequenciesof NEDD8 in ESCC

17.0

17.5

18.0

18.5

19.0

19.5

Loss Neutral Gain

Log2

(NEDD

8 F

PKM

−U

Q +

1)

Loss vs. Neutral P = 2.532E–01Gain vs. Neutral P = 8.647E–04

Figure S1 The copy number variation (CNV) frequencies of NEDD8 in ESCC, calculated from the TCGA database. (A) The CNV gain for NEDD8 in ESCC was 12.60% and the CNV loss for NEDD8 was 8.42%. (B) The association between CNV and NEDD8 gene expression was calculated with standard ANOVA and Tukey’s HSD tests.

A

β-actin

NED

D8

conj

ugat

edpr

otei

ns

HET

-1A

EC1

Kyse

30

Kyse

150

Kyse

450

Kyse

510B

0

1

2

3

4

5

HET-1A

Kyse51

0

Kyse30

Kyse15

0

Kyse45

0EC

1

Rela

tive

expr

essi

on o

fNEDD8

mRN

A

******

***

***

*

Figure S2 The mRNA and protein levels of NEDD8 are higher in ESCC cell lines than in non-cancerous cells. (A) mRNA and (B) protein expres-sion levels of NEDD8 in HET-1A, Kyse30, Kyse150, Kyse450, Kyse510, and EC1 cell lines, determined with real-time PCR and immunoblotting, respectively. *P < 0.05, ***P < 0.001.

Page 16: Overexpressed NEDD8 as a potential therapeutic target in

16 Xian et al. Therapeutic potential of NEDD8 in ESCC

Kyse450

EC1

NC NEDD8-KD1 NEDD8-KD2

Morphology

100 μm 100 μm 100 μm

100 μm 100 μm 100 μm

Figure S3 NEDD8 knockdown suppresses the growth of the Kyse450 cells and EC1 cells. Representative micrographs of NEDD8-knockdown Kyse450 and EC1 cells are shown; scale bar = 100 µm.

β-actin

NC KD1 KD2 NC KD1 KD2

Kyse450 EC1

NED

D8

conj

ugat

edpr

otei

ns

UBA3

NAE1

UBC12

NEDD8

Figure S4 NEDD8 knockdown does not affect the protein levels of neddylation enzymes. The expression levels of NAE1, UBA3, and UBC12 in NEDD8-knockdown ESCC cells were determined by immunoblotting.

Page 17: Overexpressed NEDD8 as a potential therapeutic target in

Cancer Biol Med Vol xx, No x Month 2021 17

PI

Annexin-V FITC

siD

R5-1

siCo

ntro

l

1.94%

0.97%

NC NEDD8-KD1 NEDD8-KD2

105

102

102 103 104 105

102 103 104 105

102 103 104 105 102 103 104 105

102 103 104 105 102 103 104 105

102 103 104 105

102 103 104 105 102 103 104 105

103

104

105

102

103

104

105

102

103

104

105

102

103

104

105

102

103

104

105

102

103

104

105

102

103

104

105

102

103

104

105

102

103

104si

DR5

-2

6.46%

10.3%

5.33%

18.6%

1.35%

0.51%

2.52%

5.76%

5.15%

7.31%

2.21%

0.74%

3.99%

5.74%

6.36%

6.15%

Figure S5 Downregulation of DR5 rescues the apoptosis induced by NEDD8 knockdown in Kyse450 cells. NEDD8-knockdown Kyse450 cells transfected with siControl or siDR5 were subjected to Annexin V–FITC/PI double-staining analysis.

Page 18: Overexpressed NEDD8 as a potential therapeutic target in

18 Xian et al. Therapeutic potential of NEDD8 in ESCC

Table S1 Clinicopathologic parameters according to the expression of NEDD8

Variable   Overall No.

  

NEDD8   P

Low, n   High, n

Age (n = 95)         0.956

< 60 years   22   5   17  

≥ 60 years   73   17   56  

Sex (n = 95)         0.256

Male   77   16   61  

Female   18   6   12  

Tumor (n = 88)        0.052

T1   3   2   1  

T2   19   1   18  

T3   64   17   47  

T4   2   0   2  

Node (n = 95)         0.820

N0   47   9   38  

N1   28   8   20  

N2   16   4   12  

N3   4   1   3  

TNM (n = 88)         0.704

I   6   2   4  

II   39   7   32  

III   37   10   27  

IV   6   1   5