Cancer-associated Fibroblast promoted LncRNA DNM3OS ......fold change of DNM3OS expression in...

13
Translational Cancer Mechanisms and Therapy Cancer-associated Fibroblastpromoted LncRNA DNM3OS Confers Radioresistance by Regulating DNA Damage Response in Esophageal Squamous Cell Carcinoma Hongfang Zhang 1 , Yuhui Hua 2 , Zhenzhen Jiang 1 , Jing Yue 1 , Ming Shi 3 , Xiaoli Zhen 1 , Xiaoyan Zhang 1 , Ling Yang 1 , Rongjing Zhou 4 , and Shixiu Wu 1 Abstract Purpose: Our study aimed to investigate whether CAF (cancer-associated broblasts) were involved in long noncod- ing RNAs (lncRNA)-regulated radioresponse in esophageal squamous cell carcinoma (ESCC). Experimental Design: By use of lncRNAs PCR array, 38 lncRNAs were screened in esophageal cancer cells and in normal esophageal epithelial cells Het-1A. LncRNA DNM3OS was detected in tumor tissues of patients with ESCC and in matched normal esophageal epithelial tissues by qRT-PCR analysis and in situ hybridization assay. The association of DNM3OS and tumor radioresistance was investigated in vitro and in vivo. The inuences of DNM3OS on DNA damage response (DDR) was investigated by Western blotting, immu- nouorescence imaging, and comet assay. The mechanisms by which CAFs promoted DNM3OS expression was investigated by kinase inhibitors' screening, luciferase assay, and chromatin immunoprecipitation. Results: Among the 38 lncRNAs tested, DNM3OS was found to have a much higher expression level in esophageal cancer cells than in Het-1A. In tumor tissues of 16 patients with ESCC, the expression level of DNM3OS showed an average increase of 6.3429-fold compared with that in matched nor- mal tissues. DNM3OS conferred signicant radioresistance in vitro and in vivo by regulating DDR. CAFs promoted the expression of DNM3OS with a 39.2554-fold and 38.3163-fold increase in KYSE-30 and KYSE-140, respectively. CAFs pro- moted the expression of DNM3OS in a PDGFb/PDGFRb/ FOXO1 signaling pathwaydependent manner. FOXO1, a transcription factor downstream of PDGFb/PDGFRb signaling pathway, initiated the transcription of DNM3OS by binding to DNM3OS promoter. Conclusions: Our study highlighted CAF-promoted DNM3OS as an attractive target to reverse tumor radioresis- tance in ESCC. Introduction The 5-year survival rate of patients with esophageal squamous cell carcinoma (ESCC) treated with radiotherapy, one major curative treatment choice for patients with ESCC, is less than 20% due to radioresistance (1). Accumulating evidences have suggested that tumor microenvironment, especially its major component cancer-associated broblasts (CAF), were closely associated with tumor initiation and progression (2, 3). The complicated interaction between tumor cells and their host microenvironment has important inuences on their biological behavior. Tumor cells promote the growth and survival of stromal broblasts; in parallel, the microenvironment potentiates the activation of signaling pathways that are involved in proliferation, invasion, and survival of tumor cells (46). Our previous study revealed that the cross-talk of esophageal cancer cells and CAFs induced TGFb1 expression in an autocrine/paracrine signaling loop, which resulted in the development of chemoresistance (7). Furthermore, we found CAF-secreted CXCL1 conferred ESCC- signicant radioresistance in vitro and in vivo by regulating DNA damage response (DDR) in a reactive oxygen speciesdependent manner (8). Long noncoding RNAs (lncRNA) are a group of transcripts comprising more than 200 nucleotides and lacking protein- coding potential (9). LncRNAs exerted their functions through chromatin modication, transcriptional regulation, and post- transcriptional regulation. Aberrant expressions of lncRNAs were associated with human diseases such as cancer (10). LncRNAs were involved in many cellular biological processes including cell-cycle progression, apoptosis, and pluripotency of stem cells. Recent studies showed that lncRNAs were involved in DDR by sensing DNA damage, transducing damage signals, repairing damaged DNA, activating cell-cycle checkpoints, and inducing apoptosis (11). Our study aimed to investigate wheth- er lncRNAs were involved in DNA damage repair following irradiation in ESCC and whether CAFs regulated the expres- sions of those functional lncRNAs in esophageal cancer cells. By screening lncRNAs PCR array, we found lncRNA DNM3OS 1 Hangzhou Cancer Institution, Hangzhou Cancer Hospital, Hangzhou, China. 2 Department of Pharmacy, Hangzhou Cancer Hospital, Hangzhou, China. 3 School of Life Science and Technology, Harbin Institute of Technology, Harbin, China. 4 Department of Pathology, Hangzhou Cancer Hospital, Hangzhou, China. Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). H. Zhang and Y. Hua contributed equally to this article. Corresponding Author: Shixiu Wu, Hangzhou Cancer Hospital, Yanguan Lane 34, Hangzhou, Zhejiang 310002, China. Phone: 8657-1860-62281; Fax: 8657- 1560-06191; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-18-0773 Ó2018 American Association for Cancer Research. Clinical Cancer Research www.aacrjournals.org 1989 on October 27, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst November 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0773

Transcript of Cancer-associated Fibroblast promoted LncRNA DNM3OS ......fold change of DNM3OS expression in...

Page 1: Cancer-associated Fibroblast promoted LncRNA DNM3OS ......fold change of DNM3OS expression in esophageal cancer cells including KYSE-30, KYSE-180, KYSE-150, KYSE-140, and KYSE-450

Translational Cancer Mechanisms and Therapy

Cancer-associated Fibroblast–promoted LncRNADNM3OS Confers Radioresistance by RegulatingDNA Damage Response in Esophageal SquamousCell CarcinomaHongfang Zhang1, Yuhui Hua2, Zhenzhen Jiang1, Jing Yue1, Ming Shi3, Xiaoli Zhen1,Xiaoyan Zhang1, Ling Yang1, Rongjing Zhou4, and Shixiu Wu1

Abstract

Purpose: Our study aimed to investigate whether CAF(cancer-associated fibroblasts) were involved in long noncod-ing RNAs (lncRNA)-regulated radioresponse in esophagealsquamous cell carcinoma (ESCC).

Experimental Design: By use of lncRNAs PCR array, 38lncRNAs were screened in esophageal cancer cells and innormal esophageal epithelial cells Het-1A. LncRNADNM3OSwas detected in tumor tissues of patients with ESCC and inmatched normal esophageal epithelial tissues by qRT-PCRanalysis and in situ hybridization assay. The association ofDNM3OS and tumor radioresistance was investigated in vitroand in vivo. The influences of DNM3OS on DNA damageresponse (DDR) was investigated by Western blotting, immu-nofluorescence imaging, and comet assay. Themechanisms bywhich CAFs promoted DNM3OS expression was investigatedby kinase inhibitors' screening, luciferase assay, and chromatinimmunoprecipitation.

Results: Among the 38 lncRNAs tested, DNM3OS wasfound to have a much higher expression level in esophagealcancer cells than inHet-1A. In tumor tissues of 16patientswithESCC, the expression level of DNM3OS showed an averageincrease of 6.3429-fold compared with that in matched nor-mal tissues. DNM3OS conferred significant radioresistancein vitro and in vivo by regulating DDR. CAFs promoted theexpression ofDNM3OSwith a 39.2554-fold and 38.3163-foldincrease in KYSE-30 and KYSE-140, respectively. CAFs pro-moted the expression of DNM3OS in a PDGFb/PDGFRb/FOXO1 signaling pathway–dependent manner. FOXO1, atranscription factor downstreamof PDGFb/PDGFRb signalingpathway, initiated the transcription ofDNM3OSby binding toDNM3OS promoter.

Conclusions: Our study highlighted CAF-promotedDNM3OS as an attractive target to reverse tumor radioresis-tance in ESCC.

IntroductionThe 5-year survival rate of patients with esophageal squamous

cell carcinoma (ESCC) treated with radiotherapy, one majorcurative treatment choice for patients with ESCC, is less than20% due to radioresistance (1). Accumulating evidences havesuggested that tumor microenvironment, especially its majorcomponent cancer-associated fibroblasts (CAF), were closelyassociated with tumor initiation and progression (2, 3). Thecomplicated interaction between tumor cells and their hostmicroenvironment has important influences on their biologicalbehavior. Tumor cells promote the growth and survival of stromal

fibroblasts; in parallel, the microenvironment potentiates theactivation of signaling pathways that are involved in proliferation,invasion, and survival of tumor cells (4–6). Our previous studyrevealed that the cross-talk of esophageal cancer cells and CAFsinduced TGFb1 expression in an autocrine/paracrine signalingloop, which resulted in the development of chemoresistance (7).Furthermore, we found CAF-secreted CXCL1 conferred ESCC-significant radioresistance in vitro and in vivo by regulating DNAdamage response (DDR) in a reactive oxygen species–dependentmanner (8).

Long noncoding RNAs (lncRNA) are a group of transcriptscomprising more than 200 nucleotides and lacking protein-coding potential (9). LncRNAs exerted their functions throughchromatin modification, transcriptional regulation, and post-transcriptional regulation. Aberrant expressions of lncRNAswere associated with human diseases such as cancer (10).LncRNAs were involved in many cellular biological processesincluding cell-cycle progression, apoptosis, and pluripotency ofstem cells. Recent studies showed that lncRNAs were involvedin DDR by sensing DNA damage, transducing damage signals,repairing damaged DNA, activating cell-cycle checkpoints, andinducing apoptosis (11). Our study aimed to investigate wheth-er lncRNAs were involved in DNA damage repair followingirradiation in ESCC and whether CAFs regulated the expres-sions of those functional lncRNAs in esophageal cancer cells. Byscreening lncRNAs PCR array, we found lncRNA DNM3OS

1Hangzhou Cancer Institution, Hangzhou Cancer Hospital, Hangzhou, China.2Department of Pharmacy, Hangzhou Cancer Hospital, Hangzhou, China.3School of Life Science and Technology, Harbin Institute of Technology, Harbin,China. 4Department of Pathology, Hangzhou Cancer Hospital, Hangzhou, China.

Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

H. Zhang and Y. Hua contributed equally to this article.

Corresponding Author: Shixiu Wu, Hangzhou Cancer Hospital, Yanguan Lane34, Hangzhou, Zhejiang 310002, China. Phone: 8657-1860-62281; Fax: 8657-1560-06191; E-mail: [email protected]

doi: 10.1158/1078-0432.CCR-18-0773

�2018 American Association for Cancer Research.

ClinicalCancerResearch

www.aacrjournals.org 1989

on October 27, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0773

Page 2: Cancer-associated Fibroblast promoted LncRNA DNM3OS ......fold change of DNM3OS expression in esophageal cancer cells including KYSE-30, KYSE-180, KYSE-150, KYSE-140, and KYSE-450

conferred significant radioresistance by regulating DDR inESCC. More importantly, we found CAFs significantly promot-ed the expression of DNM3OS in esophageal cancer cells byactivating the PDGFb/PDGFbR/FOXO1 signaling pathway.

Materials and MethodsIsolation and culture of CAFs

To isolate stromal CAFs, tumor tissues were obtained with thepermission of patients with ESCC who had not been treated withany preoperative chemoradiotherapy before esophagectomy. Theuse of tumor tissues was in accordance with the guidelines of theCommittees for Ethical Review of Research at Hangzhou CancerHospital (Hangzhou, China). The isolation of CAFs in ESCC wasaccomplished according to the method described in our previousstudy (8). CAFs were cultured in RPMI1640medium supplemen-ted with 10% FBS.

Preparation of the conditioned mediumCAFs were seeded into 75-cm2 culture flask. Forty-eight hours

later, when the cells were grown at approximately 80% conflu-ence, the culture medium was collected and centrifuged at 3,000rpm at 4�C for 30 minutes. The supernatant was collected asconditioned medium and kept at �80�C until use. The condi-tionedmedium fromCAFswas defined as CAFsmedium. Normalmedium was fresh RPMI1640 medium with 10% FBS.

Culture of ESCC cellsThe human ESCC cells KYSE-30, KYSE-140, KYSE-150, KYSE-

180, and KYSE-450, as well as normal esophageal epithelial cellsHet-1A were kindly provided by Dr. Xiaonan Sun of ZhejiangUniversity Medical College, Zhejiang University (Hangzhou, Chi-na) in2015. The radioresistant esophageal cancer cells KYSE-150Rhad been established from the parental KYSE-150 cells by mul-tiple fractionated irradiations (12). Cells were cultured inRPMI1640 medium (Gibco, Life Technologies Inc.) supplemen-ted with 10% FBS (Gibco, Life Technologies Inc.) at 37�C in 5%

CO2/95% air. All of the cells were authenticated and routinelytested for Mycoplasma contamination with the MycoAlert Myco-plasma Detection Kit (Lonza). The use of cells was no longer than2 months after thawing early-passage cells.

Animals and tumor tissues of patients with ESCCSix-week-old female BALB/c nude mice were purchased from

Vital River and maintained under standard conditions in Exper-imental Animal Center in Zhejiang Chinese Medicine University(Zhejiang, China). All of animal protocols in our study were inaccordance with the institutional animal welfare guidelines ofZhejiang Chinese Medicine University (Zhejiang, China). Tumortissues of patientswith ESCCused inour studywere collectedwiththe informed consent of each patient. All of human studies in ourstudy were in accordance with Declaration of Helsinki and theguidelines of the Committee for Ethical Review of Research atHangzhou Cancer Hospital (Hangzhou, China).

In vivo tumor experimentsTo develop xenograft tumors in mice, in vitro growing KYSE-30

cells and KYSE-150R cells transfected with lentivirus vectorscontaining DNM3OS siRNA-1 or scrambled siRNA were har-vested by treatment with trypsin-EDTA, washed with ice-cold PBStwice, and implanted into the right flanks of female BALB/c nudemice (1.0� 105 cells in 100 mL PBS). When xenograft tumors hadreached a mean diameter of approximately 0.5 cm, the mice wererandomly assigned into different groups (7 mice in each group).Tumorswere treatedwith fractionated irradiation at a total dose of12 Gy, once every 2 days with 2 Gy per fraction for 12 days(experimental group). The tumors without exposure to irradia-tion were chosen as a control. Each animal was earmarked andfollowed individually throughout the experiments. Tumor vol-ume (mm3)was calculated using the following formula:V (mm3)¼ A (mm) � B (mm)2/2, where A and B were the longest andwidest diameter of tumor, respectively, andmeasured every 2 dayswith a caliper. The day when fractionated irradiation was deliv-ered for thefirst timewasdefined as day 1 and the treatment endedon day 12. When the experiment ended, all of the mice weresacrificed according to the institutional guidelines and the tumorswere resected and weighted. The tumor inhibition rates (IR) werecalculated using the following equation: IR ¼ 100% � (meantumor weight of control group � mean tumor weight of exper-imental group)/mean tumor weight of control group.

Statistical analysisAll of the experiments in our study were independently per-

formed in triplicate and the data were presented as mean � SD.Statistical analyses of 138 patients were performed with x2 good-ness-of-fit test and the others with Student t test unless otherwisespecified using SPSS software 16.0. Differences were consideredstatistically significant at a level of P < 0.05.

ResultsLncRNA DNM3OS was highly expressed in esophageal cancercells by screening lncRNAs PCR array

Because lncRNAs were involved in tumor initiation and pro-gression, our study investigatedwhich lncRNAshadhigher expres-sion levels in esophageal cancer cells than in normal esophagealepithelial cells Het-1A. By screening lncRNA PCR array, wedetected 38 lncRNAs expressions in three esophageal cancer cells

Translational Relevance

Radiotherapy is one of major curative treatment modalitiesfor patients with esophageal squamous cell carcinoma(ESCC). However, the 5-year survival rate of patients withESCC treated with radiotherapy is less than 20% due toradioresistance. By screening long noncoding RNAs (lncRNA)PCR array, our study identified that lncRNA DNM3OS had amuchhigher expression level in esophageal cancer cells than innormal esophageal epithelial cells Het-1A. DNM3OSwas alsohighly expressed in tumor tissues of ESCC patients than inmatched normal esophageal epithelial tissues. CAFs, as onemajor component of tumormicroenvironment, promoted theexpressionofDNM3OS in esophageal cancer cells in a PDGFb/PDGFRb/FOXO1 signaling pathway–dependent manner.CAF-promoted DNM3OS conferred significant radioresis-tance in vitro and in vivo by regulating DNA damage response.These findings highlighted CAF-promoted DNM3OS as anattractive therapeutic target to reverse tumor radioresistance inESCC and might provide novel insights into CAF-inducedtumor radioresistance.

Zhang et al.

Clin Cancer Res; 25(6) March 15, 2019 Clinical Cancer Research1990

on October 27, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0773

Page 3: Cancer-associated Fibroblast promoted LncRNA DNM3OS ......fold change of DNM3OS expression in esophageal cancer cells including KYSE-30, KYSE-180, KYSE-150, KYSE-140, and KYSE-450

including KYSE-30, KYSE-150, and KYSE-180, which were oftenadopted in the literature (13–15), and in normal esophagealepithelial cells Het-1A. As shown in Fig. 1A, among the lncRNAstested, we found lncRNADNM3OShad amuch higher expressionlevel in esophageal cancer cells than in normal esophageal epi-thelial cells Het-1A. In KYSE-30 cells, the expression level ofDNM3OS showed a 39.4553-fold increase compared with thatin Het-1A (Fig. 1B). In KYSE-180 cells, the expression level ofDNM3OS showed a 28.5365-fold increase compared with that inHet-1A (Fig. 1B). In KYSE-150 cells, DNM3OS exhibited a35.8426-fold increase compared with that in Het-1A (Fig. 1B).Furthermore, DNM3OSwas also highly expressed in other esoph-ageal cancer cells compared with that in Het-1A (Fig. 1B). BecauseDNM3OS was highly expressed in esophageal cancer cells, wethen investigated its pathologic roles. We compared the expres-sion of DNM3OS in parental KYSE-150 cells and radioresistantKYSE-150R cells, which had been established by multiple frac-tionated irradiations and used in our previous study (12). The

result showed that the expression level of DNM3OS exhibited a4.3169-fold increase in radioresistant KYSE-150R cells comparedwith that in parental KYSE-150 cells (Fig. 1B), suggesting thatDNM3OS was possibly associated with tumor radioresistance.

LncRNA DNM3OS was highly expressed in tumor tissues ofpatients with ESCC and associated with tumor stage

To further define the association of DNM3OS and esophagealcancer, we examined the expression of DNM3OS in tumor tissuesand matched normal esophageal epithelial tissues of 16 patientswith primary ESCC who had not been treated with any chemor-adiotherapy before surgery. As shown in Fig. 2A, by use of in situhybridization assay, we found DNM3OS had a much higherexpression level in tumor tissues of patients with ESCC than inmatched normal esophageal epithelial tissues. By qRT-PCR anal-ysis, DNM3OSwas found to be expressedwith an average 6.3429-fold increase in tumor tissues compared with that in matchednormal esophageal epithelial tissues (Fig. 2B). We further

A

KYSE-150/Het-1A

KYSE-30/Het-1A

KYSE-180/Het-1A

14

12

10

8

6

4

2

45

40

35

30

25

20

15

10

5

0

Fo

ld c

han

ge

of

DN

M3O

S e

xpre

ssio

nB

KYSE-30/Het-1A

KYSE-180/Het-1A

KYSE-150/Het-1A

KYSE-140/Het-1A

KYSE-450/Het-1A

KYSE-150R/KYSE-150

Figure 1.

The screening of lncRNAs PCR array revealed DNM3OSwas highly expressed in esophageal cancer cells.A, The expressions of 38 lncRNAs in esophageal cancer cellsincluding KYSE-150, KYSE-30, and KYSE-180 compared with that in normal esophageal epithelial cells Het-1A by qRT-PCR analysis, and the heatmapof these lncRNAs expressionswas generated with the log2-transformed data. Blue (red) coloring indicates expressions levels of the genes (log2 transformed).B, Thefold change of DNM3OS expression in esophageal cancer cells including KYSE-30, KYSE-180, KYSE-150, KYSE-140, and KYSE-450 compared with that innormal esophageal epithelial cells Het-1A, and in radioresistant KYSE-150R cells compared with that in parental KYSE-150 cells by qRT-PCR analysis.

CAF-promoted DNM3OS Confers Radioresistance

www.aacrjournals.org Clin Cancer Res; 25(6) March 15, 2019 1991

on October 27, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0773

Page 4: Cancer-associated Fibroblast promoted LncRNA DNM3OS ......fold change of DNM3OS expression in esophageal cancer cells including KYSE-30, KYSE-180, KYSE-150, KYSE-140, and KYSE-450

investigated the clinical significance of DNM3OS in patients withESCC. By use of in situ hybridization assay, we detected theexpression of DNM3OS in tumor tissues of 138 patients withESCC with the patients' information shown in SupplementaryTable S1. The results showed that the expression level ofDNM3OSin tumor tissueswas significantly associatedwith tumor stage (P¼0.018), suggesting that DNM3OS was closely associated withtumor progression.

LncRNADNM3OS conferred significant radioresistance in vitroand in vivo

By clonogenic survival assay, we investigated the effect ofDNM3OSon the radiosensitivity of esophageal cancer cells.WhenDNM3OS was stably downregulated by transfection with lenti-virus vectors containing DNM3OS siRNA-1, the radiosensitivityof KYSE-30 cells and KYSE-140 cells was significantly increased(Fig. 3A and B; Supplementary Fig. S1). The dose enhancement

ratios (DER10) defined as the ratio of doses required to achieve10% surviving fraction for indicated cells were determined. TheDER10 for KYSE-30 cells without or with siRNA-1 transfectionwas3.4492 (Fig. 3A). The DER10 for KYSE-140 cells without or withsiRNA-1 transfection was 2.589 (Fig. 3B). Because DNM3OS wasinvolved in the radioresponse of KYSE-30 cells and KYSE-140cells, we then investigated whether irradiation affected the expres-sion of DNM3OS in these cells. As shown in Supplementary Fig.S2, 24 hours after exposure to irradiation, the expression level ofDNM3OS showed an increase of 1.0396-fold and 3.7442-fold inKYSE-30 cells and in KYSE-140 cells, respectively, suggesting thatirradiation had differential influences on DNM3OS expression incancer cells, and irradiation-induced high DNM3OS expressionmay also be involved in the radioresponse of KYSE-140 cells. Inaddition toKYSE-30 cells andKYSE-140 cells, we also investigatedthe radiosensitivity of KYSE-150 cells, KYSE-150R cells, KYSE-450cells, and Het-1A cells in which the expression levels of DNM3OS

DAPIA

B

DNM3OS Merge HE

No

rmal

Can

cer

14

12

10

8

6

4

2

0

Fo

ld c

han

ge

of

DN

M3O

Sex

pre

ssio

n

Patient 1Patient 2

Patient 3Patient 4

Patient 5Patient 6

Patient 7Patient 8

Patient 9

Patient 10

Patient 11

Patient 12

Patient 13

Patient 14

Patient 15

Patient 16

Figure 2.

DNM3OS was highly expressed in tumor tissues of patients with ESCC. A, One representative result of DNM3OS expression in tumor tissues of patients with ESCCcompared with that in matched normal esophageal epithelial tissues by in situ hybridization assay. B, The fold change of DNM3OS expression in tumor tissues of 16patients with ESCC compared with that in matched normal esophageal epithelial tissues by qRT-PCR analysis.

Zhang et al.

Clin Cancer Res; 25(6) March 15, 2019 Clinical Cancer Research1992

on October 27, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0773

Page 5: Cancer-associated Fibroblast promoted LncRNA DNM3OS ......fold change of DNM3OS expression in esophageal cancer cells including KYSE-30, KYSE-180, KYSE-150, KYSE-140, and KYSE-450

KYSE-140

Control

siRNA-1 DER10 = 2.589

Dose (Gy)0 1 2 3 4 5 6

Dose (Gy)1 2 3 4 5 6

0 1 2 3 4 5 6

1 4 7 10 13 17 20 23 26Days

Control

siRNA-1

Tum

or

inh

ibit

ion

rat

e (%

)

90

80

70

60

50

40

30

20

10

0

HP = 0.0318

Tum

or

volu

me

(mm

3 )F 1,400

1,200

1,000

800

600

400

200

0

siRNA-1+IR

Control+IRControl

siRNA-1

KYSE-150KYSE-150R+siRNA-1

KYSE-150+siRNA-1KYSE-150R

0.01

0.1

Su

rviv

al f

ract

ion

D 1

0.1

Su

rviv

al f

ract

ion

C 1

Su

rviv

al f

ract

ion

B

0.01

0.1

1

0.01

Su

rviv

al f

ract

ion

A

0.1

1

Control

siRNA-1DER10 = 3.4492

0

Dose (Gy)1 2 3 4 5 60

Dose (Gy)1 2 3 4 5 60

KYSE-150KYSE-150R

KYSE-450Het-1A

0.01

0.1

Su

rviv

al f

ract

ion

E1

KYSE-150KYSE-150R+siRNA-2KYSE-150+siRNA-2

KYSE-150R0.01

1.6

1.4

1.2

1

0.8

0.6

0.4

0.2

0

Tum

or

wei

gh

t (g

)

G

Control

Control+IRsiRNA-1

siRNA-1+IR

P = 0.0261

P = 0.152

KYSE-30

Figure 3.

DNM3OS conferred significant radioresistance in vitro and in vivo. Radiation survival curves are shown for KYSE-30 cells (A) and KYSE-140 cells (B) which weretransfected with lentivirus vectors containing DNM3OS siRNA-1 or scrambled siRNA (Control). � , P < 0.05, using ANOVA. The DER10 was defined as the ratioof doses required to achieve 10% surviving fraction for cells without or with siRNA transfection. C,Radiation survival curves are shown for KYSE-150 cells, KYSE-150Rcells, KYSE-450 cells, and Het-1A cells. � , P < 0.05, using ANOVA. D and E, Radiation survival curves are shown for KYSE-150 cells and KYSE-150R cells withor without siRNA transfection. � , P < 0.05, using ANOVA. F, The growth curve of xenograft tumors implanted with KYSE-30 cells which were transfected withlentivirus vectors containing DNM3OS siRNA-1 or scrambled siRNA (Control). The day when fractionated irradiation (FIR) was delivered for the first time wasdefined as day 1 and the treatment ended on day 12. G, The weight of xenograft tumors in F at the end of experiment. H, The tumor inhibition rate of fractionatedirradiation in xenograft tumors transfected with DNM3OS siRNA-1 or scrambled siRNA (Control).

CAF-promoted DNM3OS Confers Radioresistance

www.aacrjournals.org Clin Cancer Res; 25(6) March 15, 2019 1993

on October 27, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0773

Page 6: Cancer-associated Fibroblast promoted LncRNA DNM3OS ......fold change of DNM3OS expression in esophageal cancer cells including KYSE-30, KYSE-180, KYSE-150, KYSE-140, and KYSE-450

were significantly different. As shown in Fig. 3C, the expressionlevels of DNM3OS were negatively correlated with the radiosen-sitivity. KYSE-150R cells with the highest expression level ofDNM3OS showed the most radioresistance, whereas Het-1A cellswith the lowest expression level of DNM3OS were the mostradiosensitive. The DER10 was 3.1742, 19.9795, 6.2943, and3.3456 for KYSE-150R versus KYSE-150, KYSE-150R versusHet-1A, KYSE-150 versus Het-1A, and KYSE-450 versus Het-1A,respectively.When lentivirus vectors containingDNM3OS siRNA-1 or siRNA-2 were transfected, the radiosensitivity of KYSE-150cells and KYSE-150R cells was both significantly increased (Fig.3D and E; Supplementary Fig. S1). TheDER10 for KYSE-150R cellswithout or with DNM3OS siRNA-1/siRNA-2 transfection was2.6399 and 2.6165, respectively. The DER10 for KYSE-150 cellswithout or with DNM3OS siRNA-1/siRNA-2 transfection was4.1698 and 5.0445, respectively. These results suggested thatDNM3OS conferred esophageal cancer cells significant radiore-sistance in vitro.

Our study further investigated the effect of DNM3OS on theradiosensitivity in vivo by the establishment of xenograft tumormodels in BALB/c nudemice. As shown in Fig. 3F, when lentivirusvectors containing DNM3OS siRNA-1 were transfected, KYSE-30xenograft tumors grew slower than control group after fraction-ated irradiation (Fig. 3F). At the end of experiment, the tumorswere weighted (Fig. 3G). The tumor IR of fractionated irradiationin siRNA-transfected groupwas 76.5900%, whichwas significant-ly higher than 47.0330% in control group (Fig. 3H). We alsoinvestigated the radiosensitivity of KYSE-150R xenograft tumorsin which DNM3OS was stably downregulated by transfectionwith DNM3OS siRNA. As shown in Supplementary Fig. S3A,when lentivirus vectors containing DNM3OS siRNA-1 were trans-fected, KYSE-150R xenograft tumors grew slower than controlgroup after fractionated irradiation. At the end of experiment, thetumorswereweighted (Supplementary Fig. S3B) and the tumor IRwas calculated. As shown in Supplementary Fig. S3C, the tumor IRof fractionated irradiation in siRNA-transfected group was signif-icantly higher than control group. These results suggested thatDNM3OS conferred esophageal cancer cells significant radiore-sistance in vivo.

LncRNADNM3OS regulated cellularDDR in esophageal cancercells

Because DDR directly regulates cell fate in response to deathstimuli, our study investigated whether DDR was involved inDNM3OS-regulated radioresponse. DNA double-strand breaks(DSB) are the major form of cellular damage induced by irradi-ation. Herein, we detected the expression of gH2AX, a marker ofDSBs following irradiation inKYSE-30 cells and inKYSE-140 cells,whichwere transfectedwithDNM3OS siRNAor scrambled siRNA(negative) before the exposure to irradiation. As shown in Sup-plementary Figs. S4 and S5, when DNM3OS siRNA-1 was trans-fected, KYSE-30 cells and KYSE-140 cells showed increasedgH2AX expression following irradiation, suggesting that the inhi-bition of DNM3OS expression enhanced irradiation-inducedDNA damage. As shown in Fig. 4A and B and SupplementaryFig. S6, whenDNM3OS siRNA-1 was transfected, KYSE-30, KYSE-140, and KYSE-150R exhibited increased expressions of DNAdamage markers gH2AX and cleaved PARP, but decreased expres-sions of DNA repair proteins including p-ATM, Rad50, p-Chk2,Ku80, MRE11, NBS1, DNA-PKcs, and P53 following irradiation.These results suggested that the inhibition of DNM3OS expres-

sion enhanced irradiation-induced DNA damage, but attenuatedDNA repair response. By the comet assay, we also confirmedDNM3OS regulated DDR in KYSE-30 cells and KYSE-140 cells. Asshown in Fig. 4C andD,whenDNM3OS siRNA-1was transfected,the DNA repair ability of KYSE-30 cells and KYSE-140 cells wassignificantly inhibited following irradiation. These results sug-gested that DNM3OS regulated DDR in esophageal cancer cells.

CAFs significantly increased the expression of lncRNADNM3OS in a PDGFb/PDGFbR/FOXO1 signaling pathway–dependent manner in esophageal cancer cells

CAFs, as one major component of tumor microenvironment,were involved in tumor radioresponse as evidenced by our pre-vious study (8). In this study, we investigated whether CAFsregulated the expression of DNM3OS in esophageal cancer cells.When KYSE-30 cells and KYSE-140 cells were cultured in theconditioned medium (CM) of CAFs for 48 hours, the expressionof DNM3OS was significantly upregulated, with a 39.2554-foldand 38.3163-fold increase, respectively, compared with that cul-tured in normalmedium (Fig. 5A).We then investigated the effectof CAFs-induced high DNM3OS expression on the radiosensitiv-ity of KYSE-30 cells and KYSE-140 cells. As shown in Fig. 6, afterthe treatment with CAFCM for 48 hours, KYSE-30 cells and KYSE-140 cells displayed significant radioresistance comparedwith thatcultured in normal medium. The DER10 was 3.3015 for KYSE-30cells cultured in CAFmedium versus cultured in normalmedium.The DER10 was 1.8368 for KYSE-140 cells cultured in CAF medi-umversus cultured innormalmedium. In contrast,whenKYSE-30cells and KYSE-140 cells were transfected with DNM3OS siRNAbefore the treatment with CAF CM, their increase of radioresis-tancewas partially reversed. TheDER10was 2.6687 and3.2615 forKYSE-30 cells without or with DNM3OS siRNA-1/siRNA-2 trans-fection before the treatment with CAF CM, respectively. TheDER10 was 1.7127 and 1.7268 for KYSE-140 cells without orwith DNM3OS siRNA-1/siRNA-2 transfection before the treat-mentwithCAFCM, respectively. These results suggested thatCAFsconferred esophageal cancer cells significant radioresistance bypromoting DNM3OS expression.

We then explored the molecular mechanisms by which CAFspromoted DNM3OS expression in esophageal cancer cells. Asshown in Fig. 5B andC and Supplementary Table S2, by screening14 kinds of small-molecule kinase inhibitors that target multiplecritical kinases, we found crenolanib, an inhibitor of PDGFRa/bsignaling, had potent ability of inhibiting DNM3OS expression.When KYSE-30 cells and KYSE-140 cells were cultured in CAFsCM,which contained crenolanib, the expression ofDNM3OSwasdecreased to 42.19% and 37.15%, respectively, compared withcells cultured in CAF CM without crenolanib (Fig. 5B and C). Wethen examined the expressions of PDGFRa, PDGFRb, PDGFa,and PDGFb in KYSE-30 cells and in KYSE-140 cells, which werecultured in CAFs CM for 48 hours. The results showed that theexpressions of PDGFRb andPDGFbwere highly upregulated,witha 6.5812-fold and 8.1062-fold increase, respectively, in KYSE-30cells, which were cultured in CAF CM for 48 hours (Fig. 5D).Similarly, PDGFRb andPDGFbwere expressedwith a 5.1694-foldand 6.4225-fold increase, respectively, in KYSE-140 cells, whichwere cultured in CAF CM for 48 hours (Fig. 5E).

To further confirm whether the PDGFb/PDGFRb signalingpathway was associated with CAF-induced DNM3OS upregula-tion, we detected the expression ofDNM3OS inKYSE-30 cells andKYSE-140 cells, which were transfected with PDGFRb or PDGFb

Zhang et al.

Clin Cancer Res; 25(6) March 15, 2019 Clinical Cancer Research1994

on October 27, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0773

Page 7: Cancer-associated Fibroblast promoted LncRNA DNM3OS ......fold change of DNM3OS expression in esophageal cancer cells including KYSE-30, KYSE-180, KYSE-150, KYSE-140, and KYSE-450

Neg+IRNeg

siRNA-1+IR

siRNA-1

Neg+IRNeg

siRNA-1+IR

siRNA-1

Neg+IRNeg

siRNA-2+IR

siRNA-2

Neg+IRNeg

siRNA-2+IR

siRNA-2

KYSE-30 KYSE-140 B KYSE-30 KYSE-140

16 kDa

89 kDa

469 kDa

86 kDa

81 kDa

95 kDa

53 kDa

350 kDa

62 kDa

153 kDa

37 kDa

16 kDa

89 kDa

469 kDa

86 kDa

81 kDa

95 kDa

53 kDa

350 kDa

62 kDa

153 kDa

37 kDa

gH2AX

Cleaved-PARP

DNA-PKs

Ku80

MRE11

NBS1

p53

p-ATM

p-Chk2

Rad50

GAPDH

Neg siRNA-1

0 G

y6

Gy K

YS

E-3

0

0 G

y6

Gy KY

SE

-140

20181614121086420

Tail

DN

A (

%)

14

12

10

8

6

4

2

0

Tail

DN

A (

%)

NegsiRNA

siRNA+6 Gy

Neg+6 Gy

KYSE-30P = 0.0146

KYSE-140P = 0.0253

Neg

siRNA-1

siRNA-1+6 Gy

Neg+6 Gy

A

DC

Figure 4.

DNM3OS regulated cellular DDR in esophageal cancer cells. A and B, The expressions of DDR proteins including gH2AX, cleaved-PARP, DNA-PKcs, Ku80, MRE11,NBS1, p53, p-ATM, p-Chk2, and Rad50 24 hours following 6 Gy of irradiation in KYSE-30 cells and in KYSE-140 cells, which were transfected with DNM3OS siRNA-1/siRNA-2 or scrambled siRNA (negative, Neg) by Western blotting analysis. GAPDH was used as a loading control. C, Irradiation-induced DNA damage inKYSE-30 cells and in KYSE-140 cells whichwere transfected with DNM3OS siRNA-1 or scrambled siRNA (negative, Neg) by the comet assay.D,Quantitative analysisof irradiation-induced DNA damage in C by the tail moment in the comet assay.

CAF-promoted DNM3OS Confers Radioresistance

www.aacrjournals.org Clin Cancer Res; 25(6) March 15, 2019 1995

on October 27, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0773

Page 8: Cancer-associated Fibroblast promoted LncRNA DNM3OS ......fold change of DNM3OS expression in esophageal cancer cells including KYSE-30, KYSE-180, KYSE-150, KYSE-140, and KYSE-450

45

40

35

30

25

20

15

10

5

0

45

40

35

30

25

20

15

10

5

0

45

40

35

30

25

20

15

10

5

0

45

40

35

30

25

20

15

10

5

0

KYSE-140 KYSE-140+CAFs KYSE-30+CAFsKYSE-30

Fold

cha

nge

of D

NM

3OS

exp

ress

ion

Fold

cha

nge

of D

NM

3OS

exp

ress

ion

Fold

cha

nge

of g

enes

exp

ress

ions

Fold

cha

nge

of D

NM

3OS

exp

ress

ion

Fold

cha

nge

of g

enes

exp

ress

ions

Fold

cha

nge

of D

NM

3OS

exp

ress

ion

Fold

cha

nge

of D

NM

3OS

exp

ress

ion

Fold

cha

nge

of D

NM

3OS

exp

ress

ion

1.2

1

0.8

0.6

0.4

0.2

0

KYSE-30

KYSE-30

KYSE-30

KYSE-30

KYSE-140

KYSE-30+CAFs

KYSE-140

KYSE-140

KYSE-140

KYSE-140+CAFs

Contro

lIco

tinib

Crenola

nibLY

2157

299

GW50

74SB59

0885

Ki8751

OSI-906

CHIR-98

014

SP6001

25BGJ3

98LY

2940

02

MK-2206

2HCI

Go698

3VX-70

2

Contro

lIco

tinib

Crenola

nibLY

2157

299

GW50

74SB59

0885

Ki8751

OSI-906

CHIR-98

014

SP6001

25BGJ3

98LY

2940

02

MK-2206

2HCI

Go698

3VX-70

2

1.2

1

0.8

0.6

0.4

0.2

0

9

8

7

6

5

4

3

2

1

0PDGFRα PDGFRβ PDGFβPDGFα

PDGFRα PDGFRβ PDGFβPDGFα

PDGFRα PDGFRβ PDGFβPDGFα

PDGFRα PDGFRβ PDGFβPDGFα

7

6

5

4

3

2

1

0

ControlControl+siRNAControl+CAFsControl+CAFs+siRNA

ControlControl+siRNAControl+CAFsControl+CAFs+siRNA

Control

Control+CAFsControl+FOXO1 siRNA

Control+CAFs+FOXO1 siRNA

A

C

E

G H

F

D

B

Figure 5.

CAFs promoted the expression of DNM3OS in esophageal cancer cells in a PDGFb/PDGFRb/FOXO1 signaling pathway–dependent manner. A, qRT-PCR analysis ofDNM3OS expression in KYSE-140 cells and in KYSE-30 cells that were cultured in normal medium or CAFs medium for 48 hours. �� , P < 0.01; ##, P < 0.01,compared with KYSE-140 cells and KYSE-30 cells that were cultured in normal medium, respectively. B and C, qRT-PCR analysis of DNM3OS expression in KYSE-30cells and KYSE-140 cells that were cultured in medium with or without kinase inhibitors. The cells without kinase inhibitor treatment were used as a control.� ,P<0.05, comparedwith control group.D andE,qRT-PCR analysis of the expressions of PDGFRa, PDGFRb, PDGFa, and PDGFb in KYSE-30 cells and KYSE-140 cellswhich were cultured in normal medium or CAFs medium for 48 hours. � , P < 0.05; �� , P < 0.01, compared with KYSE-30 cells or KYSE-140 cells that werecultured in normal medium for 48 hours. F andG, qRT-PCR analysis of DNM3OS expression in KYSE-30 cells and KYSE-140 cells that were transfected with indicatedsiRNA before the treatment with CAFs medium for 48 hours. The cells that were transfected with scrambled siRNA were used as a control. � , P < 0.05;#, P <0.05, comparedwith control group and control group cultured in CAFsmedium, respectively.H, qRT-PCR analysis of DNM3OS expression in KYSE-30 cells andKYSE-140 cells that were transfected with FOXO1 siRNA before the treatment with normal medium or CAFs medium for 48 hours. The cells which weretransfected with scrambled siRNA were used as a control. � , P < 0.05; #, P < 0.05, compared with control group and control group cultured in CAFs medium,respectively.

Zhang et al.

Clin Cancer Res; 25(6) March 15, 2019 Clinical Cancer Research1996

on October 27, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0773

Page 9: Cancer-associated Fibroblast promoted LncRNA DNM3OS ......fold change of DNM3OS expression in esophageal cancer cells including KYSE-30, KYSE-180, KYSE-150, KYSE-140, and KYSE-450

siRNAbefore the treatment with CAFCM. As shown in Fig. 5F andSupplementary Fig. S7, when KYSE-30 cells were transfected withPDGFRb siRNAbefore the treatmentwithCAFCM, the increase ofDNM3OS expression was decreased from 39.106-fold to 11.732-fold compared with cells transfected with scramble siRNA. WhenKYSE-30 cells were transfected with PDGFb siRNA before thetreatment with CAF CM, the increase of DNM3OS expression wasdecreased from 39.106-fold to 15.642-fold compared with cellstransfected with scramble siRNA (Fig. 5F; Supplementary Fig. S7).WhenKYSE-140 cellswere transfectedwithPDGFRb siRNAbeforethe treatment with CAF CM, the increase of DNM3OS expressionwas decreased from 38.204-fold to 15.282-fold compared withcells transfected with scramble siRNA (Fig. 5G; SupplementaryFig. S7). When KYSE-140 cells were transfected with PDGFbsiRNA before the treatment with CAF CM, the increase ofDNM3OS expression was decreased from 38.204-fold to14.518-fold compared with cells transfected with scramble siRNA(Fig. 5G; Supplementary Fig. S7). However, transfection withsiRNA against PDGFRa or PDGFa had no significant reversaleffects on CAF-induced DNM3OS upregulation in KYSE-30 cellsand in KYSE-140 cells (Fig. 5F and G; Supplementary Fig. S7).

There were several transcription factors downstream of thePDGFb/PDGFRb signaling pathway (16). By use of promoterprediction tool eGPMiner, FOXO1, a transcription factor down-stream of the PDGFb/PDGFRb signaling pathway, was predictedto have high affinity with two regions in the DNM3OS promoter.Our previous study had confirmed the expression of FOXO1 wasobviously increased at protein level in esophageal cancer cells,which were cultured in CAF CM (7). Herein, we investigatedwhether FOXO1 was involved in CAF-induced DNM3OS upre-

gulation. When KYSE-30 cells were transfected with FOXO1siRNA before the treatment with CAF CM, the increase ofDNM3OS expression was decreased from 38.162-fold to19.261-fold compared with cells transfected with scrambledsiRNA (Fig. 5H). When KYSE-140 cells were transfected withFOXO1 siRNA before the treatment with CAF CM, the increaseof DNM3OS expression was decreased from 37.251-fold to14.902-fold compared with cells transfected with scrambledsiRNA (Fig. 5H). These results suggested that FOXO1 wasinvolved in CAF-induced DNM3OS upregulation. By chromatinimmunoprecipitation (ChIP) assay, we analyzedwhether FOXO1regulated the transcription of DNM3OS by binding to its pro-moter region. As shown in Fig. 7A and B, FOXO1 exhibited strongability to bind to two regions in the DNM3OS promoter in KYSE-30 cells and in KYSE-140 cells. We further investigated whetherFOXO1 initiated the transcription of DNM3OS when binding tothe promoter regions of DNM3OS using luciferase reporter geneassay. As shown in Fig. 7C, when FOXO1 cDNA was transfected,the transcription activity of DNM3OS promoter in HEK293 cellswas obviously increased, suggesting FOXO1 could initiate thetranscription of DNM3OS upon binding to its promoter regions.Together, these results suggested that CAFs promoted DNM3OSexpression in a PDGFb/ PDGFRb/FOXO1 signaling pathway–dependent manner in esophageal cancer cells.

DiscussionDuring the past few years, oneof themost importantfindings of

the genomics era of biology is that less than 2% of the humangenome is coding sequences, whereas more than 90% of the

1

0.1

0.01

1

0.1

0.01

1

0.1

0.01

1

0.1

0.010 1 2 3 4 5 6

0 1 2 3 4 5 6 0 1 2 3 4 5 6

0 1 2 3 4 5 6Dose (Gy)

Dose (Gy) Dose (Gy)

Dose (Gy)

Sur

viva

l fra

ctio

nS

urvi

val f

ract

ion

Sur

viva

l fra

ctio

nS

urvi

val f

ract

ion

KYSE-30+siRNA-1

KYSE-30+siRNA-1+CAFs

KYSE-30KYSE-30+CAFs

KYSE-30+siRNA-2

KYSE-30+siRNA-2+CAFs

KYSE-30KYSE-30+CAFs

KYSE-140+siRNA-1

KYSE-140+siRNA-1+CAFs

KYSE-140KYSE-140+CAFs

KYSE-140+siRNA-2

KYSE-140+siRNA-2

KYSE-140KYSE-140+CAFs

A

C D

B

Figure 6.

CAF-induced DNM3OS upregulation significantly increased the radioresistance of esophageal cancer cells. Radiation survival curves are shown for KYSE-30cells (A and B) and KYSE-140 cells (C and D) that were transfected with lentivirus vectors containing DNM3OS siRNA or scrambled siRNA (Control) before thetreatment with normal medium or CAFs medium for 48 hours. � , P < 0.05, using ANOVA.

CAF-promoted DNM3OS Confers Radioresistance

www.aacrjournals.org Clin Cancer Res; 25(6) March 15, 2019 1997

on October 27, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0773

Page 10: Cancer-associated Fibroblast promoted LncRNA DNM3OS ......fold change of DNM3OS expression in esophageal cancer cells including KYSE-30, KYSE-180, KYSE-150, KYSE-140, and KYSE-450

genome is transcribed into nonprotein-coding RNA (17). Thissuggests those nonprotein-coding RNAs such as lncRNAs, andmiRNAs exist mainly as regulatory RNAs in human. LncRNAs areendogenous RNAs with more than 200 nucleotides in length.These RNAs control many cellular functions by interaction withchromatin-modifying complex, regulation of transcriptionalenhancers, and posttranscriptional regulation (18). LncRNAswere found to be involved in DDR by regulating the expressionsof DDR relevant genes, and the inhibition of lncRNAs expressions

improved the radiosensitivity of some cancers (19–22).Our studyinvestigated whether lncRNAs were involved in the radioresponsein esophageal cancer cells for the first time. By PCR screening of 38cancer-related lncRNAs, we found DNM3OS had a much higherexpression level in esophageal cancer cells and tumor tissues ofpatients with ESCC than in normal esophageal epithelial cellsHet-1A andmatched normal esophageal epithelial tissues, respec-tively. Furthermore, we showed thatDNM3OShad amuch higherexpression level in radioresistant KYSE-150R cells than in parental

−352~ −364

−352~ −364

−959~ −971

−959~ −971

KYSE-30

KYSE-140

DNM3OS

DNM3OS

M

M M

M1% In

put

1% In

put

IgG IgGHiston

H3

Histon

H3

FOXO1

FOXO1

p-FOXO1

1% In

put

IgG Histon

H3

FOXO1

p-FOXO1

1% In

put

IgG Histon

H3

FOXO1

p-FOXO1

p-FOXO1

7

6

5

4

3

2

1

0

Rel

ativ

e lu

cife

rase

act

ivity

GV238

GV238-DNM3OS

GV238-DNM3OS+GV141

GV238-DNM3OS+GV141-FOXO1

GV238+GV141

GV238+GV141-FOXO1

A

B

C

Figure 7.

FOXO1 regulated the expression of DNM3OS by binding tothe promoter as a transcription factor. A and B, The bindingactivity of FOXO1 and p-FOXO1 to two regions (�352 ��364;�951��971) in the DNM3OS promoter was assessedby ChIP assay. The antibodies against IgG and Histone H3were used as a negative and positive control, respectively.C,Relative luciferase activity in HEK293 cells transfected withplasmids of DNM3OS promoter (GV238-DNM3OS), emptyGV238 vector, GV141-FOXO1, or empty GV141 vector, aloneor in their combinations.

Clin Cancer Res; 25(6) March 15, 2019 Clinical Cancer Research1998

Zhang et al.

on October 27, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0773

Page 11: Cancer-associated Fibroblast promoted LncRNA DNM3OS ......fold change of DNM3OS expression in esophageal cancer cells including KYSE-30, KYSE-180, KYSE-150, KYSE-140, and KYSE-450

KYSE-150 cells, suggesting that DNM3OS may be involved intumor radioresponse.

Loss-of-function studies showed that the downregulation ofDNM3OS expression significantly enhanced the radiosensitivityof esophageal cancer cells both in vitro and in vivo. DNA DSBs arethe major form of irradiation-induced DNA damage (23). Fol-lowing irradiation, gH2AX, a marker of DNA DSBs, is highlyexpressed and initiates the activation of DDR in cancer cells.gH2AX interacts with theMRE11-RAD50–NBS1 (MRN) complex,leading to the activation of ataxia telangiectasia–mutated (ATM)kinase and/or ataxia telangiectasia andRad3-related (ATR) kinase.ATM and ATR kinases phosphorylate their downstream targetgenes such as BRCA1, p53, Chk1, and Chk2 to induce cell-cyclearrest and/or cell apoptosis (24). Our study revealed that thedownregulation of DNM3OS expression significantly increasedirradiation-induced DNA damage, but inhibited DNA repairresponse by decreasing the expressions of DNA repair proteins.Therefore, we proposed that DNM3OS conferred ESCC radio-resistance possibly through regulating DDR.

The members of tumor microenvironment, such as CAFs,transfer energy and biomass to support tumor cells growth andsurvival in response to death stimulus (3). In parallel, tumorcells facilitate the transformation of normal fibroblasts intoCAFs, which in turn enhance the malignant ability of tumorcells (4, 25). Our previous studies revealed that CAFs wereinvolved in tumor chemoradioresponse by secretion of che-mokines and cytokines (7, 8). We found that the cross-talkbetween CAFs and tumor cells caused the expressions of CXCL1and TGFb1 in an autocrine/paracrine signaling loop. Thesefindings suggest that the crosstalk between CAFs and tumorcells exerted significant influences on the expression patterns ofgenes. Previous studies focused on the pathologic roles oflncRNAs that were expressed in tumor cells or CAFs. However,whether CAFs regulated the expressions of functional lncRNAsin tumor cells has never been studied. Our study revealed thatCAFs upregulated the expression of lncRNA DNM3OS in KYSE-30 cells and KYSE-140 cells with an increase of 39.2554-foldand 38.3163-fold, respectively, suggesting that CAFs were themajor reason for the high expression of DNM3OS in esoph-ageal cancer cells.

By screening 14 types of kinase inhibitors that target manycritical kinases, we found crenolanib, an inhibitor of PDGFRa/bsignaling pathway, significantly attenuated CAF-inducedDNM3OS upregulation in esophageal cancer cells. We furtherexplored the direct evidence that the PDGFRa/b signaling path-

way was involved in CAF-induced DNM3OS upregulation. Uponinhibition of PDGFb or PDGFRb expression in tumor cells bytransfection with specific siRNAs, CAF-induced DNM3OS upre-gulation was significantly attenuated, suggesting that the PDGFb/PDGFRb pathway played a direct role in CAF-induced DNM3OSupregulation. FOXO1, a transcription factor downstream ofPDGFb/PDGFRb signaling pathway, activated the transcriptionof DNM3OS by binding to the promoter regions of DNM3OS.These results suggested that CAFs promoted the expression ofDNM3OS in esophageal cancer cells in a PDGFb/ PDGFRb/FOXO1 signaling pathway–dependent manner. Taken together,our study highlighted CAF-promoted DNM3OS as an attractivetherapeutic target to reverse radioresistance in ESCC and thesefindings might provide novel insights into CAF-induced tumorradioresistance.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: S. Wu, H. ZhangDevelopment of methodology: Y. Hua, M. Shi, X. Zheng, R. ZhouAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): H. Zhang, Z. Jiang, J. YueAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): Y. Hua, M. Shi, R. Zhou, L. YangWriting, review, and/or revision of the manuscript: S. Wu, H. ZhangAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): M. Shi, R. Zhou

AcknowledgmentsThisworkwasfinancially supported byNationalNatural Science Foundation

ofChina (nos. 81872477 and 81672994), Zhejiang Provincial Analysis and TestScientific Research of China (no. 2018C37041), Zhejiang Provincial NaturalSciences Foundation of China (no. LZ15H220001), Zhejiang Provincial Med-ical Scientific Research Foundation of China (nos. 2015KYB325 and WZJ-ZJ-1628), Hangzhou City Medical Scientific Research Foundation of ZhejiangProvince, China (no. 2015Z04), and Hangzhou City Scientific TechnologyResearch Foundation of Zhejiang Province, China (nos. 20150733Q64 and20172016A04).

The costs of publication of this articlewere defrayed inpart by the payment ofpage charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

ReceivedMarch 8, 2018; revised September 15, 2018; acceptedNovember 15,2018; published first November 21, 2018.

References1. Minsky BD, Pajak TF, Ginsberg RJ, Pisansky TM, Martenson J, Komaki

R, et al. INT 0123 (Radiation Therapy Oncology Group 94-05) phaseIII trial of combined-modality therapy for esophageal cancer: high-dose versus standard-dose radiation therapy. J Clin Oncol 2002;20:1167–74.

2. Ostman A, Augsten M. Cancer-associated fibroblasts and tumor growth -bystanders turning into key players. Curr Opin Genet Dev 2009;19:67–73.

3. Martinez-OutschoornUE, LisantiMP, Sotgia F. Catabolic cancer-associatedfibroblasts transfer energy and biomass to anabolic cancer cells, fuelingtumor growth. Semin Cancer Biol 2014;25:47–60.

4. Yang Z, Yang X, Xu S, Jin P, Li X, Wei X, et al. Reprogramming of stromalfibroblasts by SNAI2 contributes to tumor desmoplasia and ovarian cancerprogression. Mol Cancer 2017;16:163.

5. Glentis A,Oertle P,Mariani P,ChikinaA, ElMarjouF, AttiehY, et al. Cancer-associated fibroblasts induce metalloprotease-independent cancer cellinvasion of the basement membrane. Nat Commun 2017;8:924.

6. Sun Y, Campisi J, Higano C, Beer TM, Porter P, Coleman I, et al. Treatment-induced damage to the tumormicroenvironment promotes prostate cancertherapy resistance through WNT16B. Nat Med 2012;18:1359–68.

7. ZhangHF, Xie CH, Yue J, Jiang ZZ, Zhou RJ, Xie RF, et al. Cancer-associatedfibroblasts mediated chemoresistance by a FOXO1/TGF1 signaling loop inesophageal squamous cell carcinoma. Mol Carcinog 2017;56:1150–63.

8. ZhangHF, Yue J, JiangZZ, ZhouRJ, Xie RF, XuYP, et al. CAF-secretedCXCL1conferred radioresistance by regulating DNA damage response in a ROS-dependentmanner in esophageal squamous cell carcinoma. CellDeathDis2017;8:e2790.

www.aacrjournals.org Clin Cancer Res; 25(6) March 15, 2019 1999

CAF-promoted DNM3OS Confers Radioresistance

on October 27, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0773

Page 12: Cancer-associated Fibroblast promoted LncRNA DNM3OS ......fold change of DNM3OS expression in esophageal cancer cells including KYSE-30, KYSE-180, KYSE-150, KYSE-140, and KYSE-450

9. Marchese FP, Raimondi I, HuarteM. Themultidimensionalmechanisms oflong noncoding RNA function. Genome Biol 2017;18:206.

10. Hosono Y, Niknafs YS, Prensner JR, Iyer MK, Dhanasekaran SM, Mehra R,et al. Oncogenic role of THOR, a conserved cancer/testis long non-codingRNA. Cell 2017;171:1559–72.

11. Shen L, Wang Q, Liu R, Chen Z, Zhang X, Zhou P, et al. LncRNA lnc-RIregulates homologous recombination repair of DNA double-strand breaksby stabilizing RAD51 mRNA as a competitive endogenous RNA. NucleicAcids Res 2018;46:717–29.

12. Jing Z, Gong L, Xie CY, Zhang L, Su HF, Deng X, et al. Reverse resistance toradiation in KYSE-150R esophageal carcinoma cell after epidermal growthfactor receptor signal pathway inhibition by cetuximab. Radiother Oncol2009;93:468–73.

13. Sutter AP, Hopfner M, Huether A, Maaser K, Scherubl H. Targeting theepidermal growth factor receptor by erlotinib (Tarceva) for the treatment ofesophageal cancer. Int J Cancer 2006;118:1814–22.

14. Al-Taie OH, Graf T, Illert B, Katzenberger T, Mork H, Kraus MR, et al.Differential effects of PPARgamma activation by the oral antidiabetic agentpioglitazone in Barrett's carcinoma in vitro and in vivo. J Gastroenterol2009;44:919–29.

15. Yang C, Zheng S, Liu T, LiuQ, Dai F, Zhou J, et al. Down-regulatedmiR-26apromotes proliferation, migration, and invasion via negative regulation ofMTDH in esophageal squamous cell carcinoma. FASEB J 2017;31:2114–22.

16. Farooqi AA, Siddik ZH. Platelet-derived growth factor (PDGF) signalling incancer: rapidly emerging signalling landscape. Cell Biochem Funct 2015;33:257–65.

17. Tehrani SS, Karimian A, Parsian H, Majidinia M, Yousefi B. Multiplefunctions of long non-coding RNAs in oxidative stress, DNA damageresponse and cancer progression. J Cell Biochem 2018;119:223–36.

18. Schmitt AM, ChangHY. Long noncoding RNAs in cancer pathways. CancerCell 2016;29:452–63.

19. Wang XY, Li M, Wang ZQ, Han SC, Tang XH, Ge YX, et al. Silencing of longnoncoding RNAMALAT1 by miR-101 and miR-217 inhibits proliferation,migration, and invasion of esophageal squamous cell carcinoma cells. JBiol Chem 2015;290:3925–35.

20. Shen YM, Liu YY, Sun T, Yang W. LincRNA-p21 knockdown enhancesradiosensitivity of hypoxic tumor cells by reducing autophagy throughHIF-1/Akt/mTOR/P70S6K pathway. Exp Cell Res 2017;358:188–98.

21. Jiao Y, LiuC, Cui FM, Xu JY, Tong J,Qi XF, et al. Long intergenic non-codingRNA induced by X-ray irradiation regulates DNA damage response signal-ing in the human bronchial epithelial BEAS-2B cell line. Oncol Lett2015;9:169–76.

22. Lai Y, Chen Y, Lin YH, Ye L. Down-regulation of LncRNA CCAT1 enhancesradiosensitivity via regulating miR-148b in breast cancer. Cell Biol Int2018;42:227–36.

23. Srivastava M, Raghavan SC. DNA double-strand break repair inhibitors ascancer therapeutics. Chem Biol 2015;22:17–29.

24. HosoyaN,MiyagawaK. TargetingDNAdamage response in cancer therapy.Cancer Sci 2014;105:370–88.

25. Fang T, LvHW, LvGS, Li T,WangCZ,HanQ, et al. Tumor-derived exosomalmiR-1247-3p induces cancer-associated fibroblast activation to foster lungmetastasis of liver cancer. Nat Commun 2018;9:191.

Clin Cancer Res; 25(6) March 15, 2019 Clinical Cancer Research2000

Zhang et al.

on October 27, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0773

Page 13: Cancer-associated Fibroblast promoted LncRNA DNM3OS ......fold change of DNM3OS expression in esophageal cancer cells including KYSE-30, KYSE-180, KYSE-150, KYSE-140, and KYSE-450

2019;25:1989-2000. Published OnlineFirst November 21, 2018.Clin Cancer Res   Hongfang Zhang, Yuhui Hua, Zhenzhen Jiang, et al.   Esophageal Squamous Cell CarcinomaRadioresistance by Regulating DNA Damage Response in

ConfersDNM3OSpromoted LncRNA −Cancer-associated Fibroblast

  Updated version

  10.1158/1078-0432.CCR-18-0773doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://clincancerres.aacrjournals.org/content/suppl/2018/11/20/1078-0432.CCR-18-0773.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/25/6/1989.full#ref-list-1

This article cites 25 articles, 2 of which you can access for free at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://clincancerres.aacrjournals.org/content/25/6/1989To request permission to re-use all or part of this article, use this link

on October 27, 2020. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst November 21, 2018; DOI: 10.1158/1078-0432.CCR-18-0773