16
Cancer Therapy: Preclinical RNF126 as a Biomarker of a Poor Prognosis in Invasive Breast Cancer and CHEK1 Inhibitor Efcacy in Breast Cancer Cells Xiaosong Yang 1,2 , You Pan 1,2 , Zhaojun Qiu 1 , Zhanwen Du 1 , Yao Zhang 1 , Pengyan Fa 1 , Shashank Gorityala 3 , Shanhuai Ma 1 , Shunqiang Li 4 , Ceshi Chen 5 , Hongbing Wang 6 , Yan Xu 3,7 , Chunhong Yan 8 , Keri Ruth 7,9 , Zhefu Ma 2,10 , and Junran Zhang 1,11 Abstract Purpose: (i) To investigate the expression of the E3 ligase, RNF126, in human invasive breast cancer and its links with breast cancer outcomes; and (ii) to test the hypothesis that RNF126 determines the efcacy of inhibitors targeting the cell-cycle check- point kinase, CHEK1. Experimental Design: A retrospective analysis by immunohis- tochemistry (IHC) compared RNF126 staining in 110 invasive breast cancer and 78 paired adjacent normal tissues with clini- copathologic data. Whether RNF126 controls CHEK1 expression was determined by chromatin immunoprecipitation and a CHEK1 promoter driven luciferase reporter. Staining for these two proteins by IHC using tissue microarrays was also conducted. Cell killing/replication stress induced by CHEK1 inhibition was evaluated in cells, with or without RNF126 knockdown, by MTT/ colony formation, replication stress biomarker immunostaining and DNA ber assays. Results: RNF126 protein expression was elevated in breast cancer tissue samples. RNF126 was associated with a poor clinical outcome after multivariate analysis and was an independent predictor. RNF126 promotes CHEK1 transcript expression. Crit- ically, a strong correlation between RNF126 and CHEK1 proteins was identied in breast cancer tissue and cell lines. The inhibition of CHEK1 induced a greater cell killing and a higher level of replication stress in breast cancer cells expressing RNF126 com- pared to RNF126 depleted cells. Conclusions: RNF126 protein is highly expressed in invasive breast cancer tissue. The high expression of RNF126 is an inde- pendent predictor of a poor prognosis in invasive breast cancer and is considered a potential biomarker of a cancer's respon- siveness to CHEK1 inhibitors. CHEK1 inhibition targets breast cancer cells expressing higher levels of RNF126 by enhancing replication stress. Clin Cancer Res; 24(7); 162943. Ó2018 AACR. Introduction Ionizing radiation (IR) and most chemotherapies damage DNA as a major part of their mechanism of action. These remain standard therapies for all types of breast cancers, the most com- mon cancer affecting women and the second most common cause of death due to cancers (1). The choice of radiotherapy or che- motherapy for breast cancer is currently made according to clinical factors. However, a subtype of breast cancer may be intrinsically resistant due to an upregulation of the DNA damage response (DDR), a major mechanism antagonizing DNA damage caused by radiotherapy/chemotherapy and involving cell-cycle checkpoints and DNA repair. This would result in a breast cancer patient subtype receiving unnecessary, aggressive treatments with mini- mal benet. In addition, resistance to radiotherapy/chemotherapy may lead to tumor recurrence that can cause considerable mor- bidity, the dissemination of disease and an increased probability of mortality due to breast cancer (2, 3). Thus, there is an important need for identifying patients who are more likely to fail therapy and to improve treatment plans for those patients. By conducting large-scale proling of cellular survival after exposure to radiation in a diverse collection of 533 genetically annotated human tumor cell lines, including breast cancer cell lines, a recent study dem- onstrated a broad variation in the response to radiotherapy, and perhaps also chemotherapy (4), as a result of genetic alterations. Thus, it is critical to identify patients whose breast cancer subtypes 1 Department of Radiation Oncology, School of Medicine, Case Western Reserve University, Cleveland, Ohio. 2 Department of Breast Surgery, The First Afliated Hospital, Sun Yat-sen University, Guangzhou, China. 3 Department of Chemistry, Cleveland State University, Cleveland, Ohio. 4 Division of Oncology Breast Oncology Section, Washington University Medical School, St. Louis, Missouri. 5 Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China. 6 Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland. 7 Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio. 8 Georgia Cancer Center, Augusta University, Augusta, Georgia. 9 Depart- ment of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio. 10 Department of Breast Surgery & Plastic Surgery, Cancer Hospital of China Medical University, Shenyang, China. 11 Department of Radi- ation Oncology, The Ohio State University, Columbus, Ohio. Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). X. Yang and Y. Pan contributed equally to this article. Corresponding Authors: Junran Zhang, Case Western Reserve University, 10900 Euclid Avenue, BRB 323, Cleveland, OH 44106. Phone: 216-368-1140; Fax: 216-368-1142; E-mail: [email protected]; and Zhefu Ma, Sun Yat-sen University, 2nd Zhong Shan Rd., # 58, Guangzhou 510080, China. E-mail: [email protected] doi: 10.1158/1078-0432.CCR-17-2242 Ó2018 American Association for Cancer Research. Clinical Cancer Research www.aacrjournals.org 1629 Research. on September 25, 2020. © 2018 American Association for Cancer clincancerres.aacrjournals.org Downloaded from Published OnlineFirst January 11, 2018; DOI: 10.1158/1078-0432.CCR-17-2242

RNF126 as a Biomarker of a Poor Prognosis in Invasive Breast … · Cleveland State University, Cleveland, Ohio. 4Division of Oncology Breast Oncology Section, Washington University

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: RNF126 as a Biomarker of a Poor Prognosis in Invasive Breast … · Cleveland State University, Cleveland, Ohio. 4Division of Oncology Breast Oncology Section, Washington University

Cancer Therapy: Preclinical

RNF126 as a Biomarker of a Poor Prognosis inInvasive Breast Cancer and CHEK1 InhibitorEfficacy in Breast Cancer CellsXiaosong Yang1,2, You Pan1,2, Zhaojun Qiu1, Zhanwen Du1, Yao Zhang1, Pengyan Fa1,Shashank Gorityala3, Shanhuai Ma1, Shunqiang Li4, Ceshi Chen5, Hongbing Wang6,Yan Xu3,7, Chunhong Yan8, Keri Ruth7,9, Zhefu Ma2,10, and Junran Zhang1,11

Abstract

Purpose: (i) To investigate the expression of the E3 ligase,RNF126, in human invasive breast cancer and its links with breastcancer outcomes; and (ii) to test the hypothesis that RNF126determines the efficacy of inhibitors targeting the cell-cycle check-point kinase, CHEK1.

Experimental Design: A retrospective analysis by immunohis-tochemistry (IHC) compared RNF126 staining in 110 invasivebreast cancer and 78 paired adjacent normal tissues with clini-copathologic data. Whether RNF126 controls CHEK1 expressionwas determined by chromatin immunoprecipitation and aCHEK1 promoter driven luciferase reporter. Staining for thesetwo proteins by IHC using tissuemicroarrays was also conducted.Cell killing/replication stress induced by CHEK1 inhibition wasevaluated in cells, with or without RNF126 knockdown, by MTT/colony formation, replication stress biomarker immunostainingand DNA fiber assays.

Results: RNF126 protein expression was elevated in breastcancer tissue samples. RNF126was associated with a poor clinicaloutcome after multivariate analysis and was an independentpredictor. RNF126 promotes CHEK1 transcript expression. Crit-ically, a strong correlation between RNF126 and CHEK1 proteinswas identified in breast cancer tissue and cell lines. The inhibitionof CHEK1 induced a greater cell killing and a higher level ofreplication stress in breast cancer cells expressing RNF126 com-pared to RNF126 depleted cells.

Conclusions: RNF126 protein is highly expressed in invasivebreast cancer tissue. The high expression of RNF126 is an inde-pendent predictor of a poor prognosis in invasive breast cancerand is considered a potential biomarker of a cancer's respon-siveness to CHEK1 inhibitors. CHEK1 inhibition targets breastcancer cells expressing higher levels of RNF126 by enhancingreplication stress. Clin Cancer Res; 24(7); 1629–43. �2018 AACR.

IntroductionIonizing radiation (IR) andmost chemotherapies damageDNA

as a major part of their mechanism of action. These remainstandard therapies for all types of breast cancers, the most com-mon cancer affectingwomen and the secondmost common causeof death due to cancers (1). The choice of radiotherapy or che-motherapy for breast cancer is currentlymade according to clinicalfactors. However, a subtype of breast cancer may be intrinsicallyresistant due to an upregulation of the DNA damage response(DDR), amajormechanism antagonizingDNAdamage caused byradiotherapy/chemotherapy and involving cell-cycle checkpointsand DNA repair. This would result in a breast cancer patientsubtype receiving unnecessary, aggressive treatments with mini-mal benefit. In addition, resistance to radiotherapy/chemotherapymay lead to tumor recurrence that can cause considerable mor-bidity, the dissemination of disease and an increased probabilityofmortality due to breast cancer (2, 3). Thus, there is an importantneed for identifying patients who are more likely to fail therapyand to improve treatment plans for those patients. By conductinglarge-scale profiling of cellular survival after exposure to radiationin a diverse collection of 533 genetically annotated human tumorcell lines, including breast cancer cell lines, a recent study dem-onstrated a broad variation in the response to radiotherapy, andperhaps also chemotherapy (4), as a result of genetic alterations.Thus, it is critical to identify patients whose breast cancer subtypes

1Department of Radiation Oncology, School of Medicine, Case Western ReserveUniversity, Cleveland, Ohio. 2Department of Breast Surgery, The First AffiliatedHospital, Sun Yat-sen University, Guangzhou, China. 3Department of Chemistry,Cleveland State University, Cleveland, Ohio. 4Division of Oncology BreastOncology Section, Washington University Medical School, St. Louis, Missouri.5Key Laboratory of Animal Models and Human Disease Mechanisms of ChineseAcademy of Sciences and Yunnan Province, Kunming Institute of Zoology,Kunming, Yunnan, China. 6Department of Pharmaceutical Sciences, Universityof Maryland School of Pharmacy, Baltimore, Maryland. 7Case ComprehensiveCancer Center, School of Medicine, CaseWestern Reserve University, Cleveland,Ohio. 8Georgia Cancer Center, Augusta University, Augusta, Georgia. 9Depart-ment of Pharmacology, School of Medicine, Case Western Reserve University,Cleveland, Ohio. 10Department of Breast Surgery & Plastic Surgery, CancerHospital of China Medical University, Shenyang, China. 11Department of Radi-ation Oncology, The Ohio State University, Columbus, Ohio.

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

X. Yang and Y. Pan contributed equally to this article.

Corresponding Authors: Junran Zhang, Case Western Reserve University,10900 Euclid Avenue, BRB 323, Cleveland, OH 44106. Phone: 216-368-1140;Fax: 216-368-1142; E-mail: [email protected]; and Zhefu Ma, Sun Yat-senUniversity, 2nd Zhong Shan Rd., # 58, Guangzhou 510080, China. E-mail:[email protected]

doi: 10.1158/1078-0432.CCR-17-2242

�2018 American Association for Cancer Research.

ClinicalCancerResearch

www.aacrjournals.org 1629

Research. on September 25, 2020. © 2018 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 11, 2018; DOI: 10.1158/1078-0432.CCR-17-2242

Page 2: RNF126 as a Biomarker of a Poor Prognosis in Invasive Breast … · Cleveland State University, Cleveland, Ohio. 4Division of Oncology Breast Oncology Section, Washington University

are intrinsically resistant to radiotherapy/chemotherapy and toexplore new approaches to target their cancers.

RNF126 is a ring E3 ligase. Recent studies have suggested thatRNF126 may have broad functions by targeting a variety ofproteins for degradation; these may range from a role in endo-somal sorting to the BAG6-dependent quality control of mislo-calized proteins (5–9). RNF126 also promotes the proliferation ofbreast cancer by ubiquitinating CDKN1A and targeting it fordegradation (10). By promoting nonhomologous end joining(NHEJ) and homologous recombination (HR; refs. 11, 12)RNF126 promotes the repair of DNA double-stranded breaks(DSB), the most dangerous type of DNA damage and that canbe caused by endogenous and exogenous sources such as repli-cation stress, radiotherapy and chemotherapeutic drugs. RNF126promotes NHEJ via the ubiquitination of Ku80 (11). Interesting-ly, we recently reported that RNF126 facilitates HR by promotingthe expression of BRCA1 in amanner independent of its E3 ligaseactivity but dependent on its interaction with E2F1 (12). Amember of the family of E2F transcription factors, E2F1, isrequired for the expression of genes involved in a wide range ofcellular processes, including cell-cycle progression, DNA replica-tion, DNA repair, differentiation, and apoptosis. Consistent withits role in HR and NHEJ, RNF126 expression is associated withresistance to ionizing radiation (IR) and PARP inhibition (12) asboth pathways are required to repair DSBs caused by IR and/or aPARP inhibitor. Thus, RNF126 appears to be associated with adiverse set of cellular processes in which its E3 ligase activity mayor may not be involved. RNF126 has a close relative, BCA2, thatshares 46%overall amino acid identity, and75% identity in RINGdomains. Although BCA2 is highly expressed and is a prognosticbiomarker in breast cancer (13–15), the pattern of RNF126protein expression and its association with outcomes of breastcancer have not yet been evaluated.

The cell-cycle checkpoint kinase, ataxia telangiectasia mutated,and Rad3-related kinase (ATR) and its key downstream effector,CHEK1, can be activated by RPA (replication protein A)-coatedelongated ssDNA. The ATR/CHEK1 pathway prevents the entry ofcells with damaged or incompletely replicated DNA into mitosiswhen cells are challenged by DNA-damaging agents, such as IR orchemotherapeutic drugs, the major modalities used to treat

cancers. This regulation is particularly evident in cells with adefective G1 checkpoint, a common feature of cancer cells, owingto mutations in TP53. In addition, ATR and CHEK1 suppressreplication stress by inhibiting excess origin firing, particularly incells with activated oncogenes (16–18). Thus, ATR and CHEK1inhibitors have been developed and are currently used either assingle agents, or pairedwith radiotherapy or a variety of genotoxicchemotherapies in preclinical and clinical studies. AlthoughCHEK1 inhibitors were initially thought to enhance the effectsof radiotherapy and genotoxic drugs, particularly in TP53-defi-cient cells (19–21), recent preclinical studies suggest that CHEK1inhibitorsmay function as signal agents since cancer cells aremorereliant on ATR/CHEK1 for survival (17). Indeed, ATR/CHEK1inhibition specifically target cancer cells expressingMYC, CYCLINE, and H-RAS (18, 22–25). In addition, we recently also reportedthat CHEK1 inhibitors, as single agents, have antitumor activityin radioresistant breast cancer by enhancing replication stress(24). Radioresistant breast cancer cells carry high levels ofC-MYC/CDC25A/C-SRC/H-RAS/E2F1 oncogenes and ATR/CHEK1/BRCA1/CtIP DDR proteins, indicating that upregulationof DDR proteins, including cell-cycle checkpoint proteins andoncogenes, may be characteristic of being targeted by CHEK1inhibitors (24). As we have demonstrated that RNF126 binds toE2F1 (12), a transcription regulator that controls the expression ofseveral hundred genes, including oncogenes andCHEK1 (26–29),we hypothesize that RNF126 promotes CHEK1 expression andthat cells expressing high levels of RNF126 may be targeted byCHEK1 inhibitors. Thus, the aims of the current study are toinvestigate the clinical significance of RNF126 expression in breastcancer, and to determine the role of RNF126 inpromotingCHEK1expression,withparticular attention to its influence on the efficacyof CHEK1 inhibitors.

Here, we demonstrate that RNF126 protein is highly expressedin invasive breast cancer and is associated with a poor prognosis.Critically, RNF126 is an independent factor for a poor prognosis.We also reveal that RNF126 controls CHEK1 expression via adirect interaction with E2F1. A strong correlation betweenRNF126 and CHEK1 protein expression exists in both breastcancer tissues and cell lines. Treatment with a CHEK1 inhibitorled to increased cell killing by enhancing replication stress in cellsexpressing a higher level of RNF126. Our studies suggest thatRNF126 is a potential biomarker for the poor prognosis ofinvasive breast cancer and for the efficacy of CHEK1 inhibitors.

Materials and MethodsPatients and specimen collection

Two cohorts of breast cancer samples were included. The firstcohort consisted of 110 paraffin-embedded tumor tissues frompatients with invasive breast cancer, as well as 78 paired adjacentnormal tissues as negative controls. The samples were taken fromThe First Affiliated Hospital of Sun Yat-sen University fromJanuary 1, 2004 to December 31, 2006. The patients in this studyall had primary operable invasive breast cancer and were underthe care of a single surgeon, Z. Ma. The most common histologicbreast cancer type was invasive ductal carcinoma that comprised88.18% (97/110) of cases, with the other 13 cases being invasivelobular, medullary, ormucinous carcinomas. Treatment includedmastectomy or local excision, with or without adjuvant systemicchemotherapy and/or radiotherapy. Follow-up involved clinicalreviews at 6-monthly intervals for the first 5 years, and then

Translational Relevance

We have previously reported that RNF126 expression isassociatedwith resistance to radiotherapy andPARP inhibition.However, RNF126 protein expression in human tumors and itsassociation with the outcomes of patients with breast cancerhave not been evaluated. CHEK1 inhibitors are currently inclinical trials but a specific target-based biomarker to identifytreatment responsive populations does not exist, which maysignificantly reduce the efficacy of such agents. Our study willbe the first step in designing clinical trials that considerRNF126 status in the selection of patients with breast cancerfor treatment with CHEK1 inhibitor-associated clinical inves-tigations. Also, the identification of biomarker to guide the useof CHEK1 inhibitors will significantly improve the efficacy ofsuch agents. Moreover, our results may improve the survival ofpatients with breast cancer and high RNF126 expression, giventhis suggests such patients generally have a poor prognosis.

Yang et al.

Clin Cancer Res; 24(7) April 1, 2018 Clinical Cancer Research1630

Research. on September 25, 2020. © 2018 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 11, 2018; DOI: 10.1158/1078-0432.CCR-17-2242

Page 3: RNF126 as a Biomarker of a Poor Prognosis in Invasive Breast … · Cleveland State University, Cleveland, Ohio. 4Division of Oncology Breast Oncology Section, Washington University

annually. All samples were confirmed histologically by twopathologists. Histologic diagnosis was determined according toAmerican Joint Commission on Cancer Staging (AJCC) criteria.Conventional pathologic data were collected retrospectively andcharacteristics of primary breast cancers are summarized in Sup-plementary Table S1.A second cohort that consistedof 67 invasivebreast cancer cases was prepared for TMA analysis of the expres-sion of both RNF126 and CHEK1. The patients of the secondcohort were treated at the same hospital between January 1, 2014and April 1, 2015.

All patients were female and signed informed consent forms.The protocol was approved by the ethics committee of The FirstAffiliated Hospital of Sun Yat-sen University (application ID:[2016]060). Inclusive criteria were: (i) all patients had unilateralinvasive breast cancer and underwent either a radical mastectomyor modified radical mastectomy. Adjacent normal breast tissueswere selected from an area more than 5 cm from the edge of thetumor and were confirmed by two pathologists. (ii) Patients whoreceived preoperative radiotherapy, chemotherapy, hormonaltherapy, or any other anticancer therapy before resection wereexcluded. (iii) adjuvant treatments such as chemotherapy, radio-therapy, or endocrine therapy were chosen based on the patient'scondition after surgery in accordance with the relevant TheNational Comprehensive Cancer Network guidelines (NCCN).(iv) All patients were followed up with medical appointments orby telephone. Cancer recurrence, metastasis, or death were endevents. The follow-up deadline was April 18, 2016.

IHCFull tissue sections of 110 paraffin embedded invasive breast

cancer and 78 normal tissues were processed for IHC staining ofRNF126. TMA blocks was constructed containing 67 invasivebreast cancer. Serial 4-mm sections were cut from the TMAblocks for both RNF126 and CHEK1 staining. Antigen retrieval,blocking procedures, and a modified ImmunoMax methodwere used as described previously (30). In brief, slides wereheated to 60�C and then deparaffinized in xylene. The slideswere rehydrated in descending alcohol concentrations. Antigenretrieval was performed by incubating slides in a retrievalsolution of citrate buffer. Hydrogen peroxide was added toblock endogenous peroxidase activity to decrease unwantedbackground staining. Primary antibody (ab183102, 1:100;Abcam; 25887-1-AP, 1:150; Proteintech) was added at anoptimum dilution. Negative controls were performed by thesubstitution of primary antibody with phosphate-bufferedsaline (PBS). To guarantee consistent IHC evaluation, slidesfrom a reference tumor previously determined as positive wereincluded in each staining procedure.

IHC scoringEvaluations of staining reactionswere performed in accordance

with the immunoreactive score (IRS) proposed by Remmele andStegner: IRS¼ staining intensity (SI)� percentage of positive cells(PP). Staining intensity was marked as nongranulated (0); lowgrade (light yellow; 1); moderate (brownish yellow; 2); or strong(reddish brown; 3). The PP was scored as negative (<5%; 0); weak(5%–10%;1);moderate (11%–50%; 2); strong (51%–80%;3); orvery strong (>81%; 4). Specimens scoring beyond 3 were consid-ered positive overexpression (31). All slides were independentlyevaluated by two pathologists blind to patients and their corre-sponding clinical information.

Cell lines, infections, transfections, and CHEK1 inhibitorsMCF7, MDA-MB-231, SKBR3, MDA-MB-361, MCF10A,

HCC202, ZR751, T47D, MDA-MB-468, HCC1187, HCC1569,HCC70, BT549, HCC1143, BT474, HCC38, and HCC1954 werecultured in DMEM (Invitrogen) supplemented with 10% FBS(Gibco/Thermo Fisher Scientific), in a humidified atmospherecontaining 5% CO2 at 37�C. The shRNA of RNF126 was pur-chased from Sigma-Aldrich. Full-length wild-type RNF126 andRING-domain mutated (C229A/C232A) RNF126 have beendescribed previously (10). The CHEK1-promoter reporter was agift from Dr. Pier Paolo Pandolfi (Beth Israel Deaconess CancerCenter, Boston, MA). All DNA-plasmid transfections were per-formed using Lipofectamine 2000 according to the manufac-turer's recommendations (Invitrogen). Flag-RNF126 construct,the full-length RNF126 fragment, has been described previously(12). Two CHEK1 inhibitors were used in this study, includingLY2603618 from ApexBio (A8638) and AZD7762 from Selleck-chem (S1532).

MTT and colony formation assaysFor the MTT assay, cells were plated into 96-well plates and

incubated overnight. Cells were then exposed to various doses ofCHEK1 or ATR inhibitors for 72 hours. MTT (20 mL of 5 mg/mL)was added to each well and cells incubated for a further 3.5 hoursin an incubator. MTT solvent was added after removing themedium and the cells in plates were agitated on an orbital shakerfor 15 minutes. The absorbance was read at 590 nm with areference filter of 620 nm. For clonogenic survival assays, cellsplated into petri dishes (60 mm � 15 mm) were exposed tovarious doses of CHEK1 or ATR inhibitors for 24 hours, and thenreplaced with fresh medium. After 13–15 days of incubation at37�C, the cellswere stainedusingGiemsa. Thenumber of colonies(>50 cells) was counted.

Quantitative reverse transcription-PCRQuantitative reverse transcription-PCR (qRT-PCR) was con-

ducted as described previously (12). Total RNAwas isolated usingan RNeasy Kit (Qiagen). Experiments were carried out in triplicatefor each data point. Reactions were performed using SYBR Greenmix and a MyiQ real-time PCR detection system (Bio-Rad).Relative mRNA levels were calculated using the comparative Ct

method (DCt).GAPDH forward/reverse primers: 50-CTCTGCTCCTCCTGTT-

CGAC-30/50-TTAAAAGCAGCCCTGGTGAC-30.CHEK1 forward/reverse primers: 50-CCAGATGCTCAGA-

GATTCTTCCA-30/ 50-TGTTCAACAAACGCTCACGATTA-30.E2F1 forward/reverse primers: 50-GTGGACTCTTCGGA-

GAACTT-30/50-TGTTCTCCTCCTCAGAAGTG-30.CYCLIN E forward/reverse primers: 50-TTTCTTGAGCAA-

CACCCTC-30/50-TGTCACATACGCAAACTGG-30.RNF126 forward/reverse primers: 50-TATCGAGGAGCTTCCG-

GAAGAGA-30/50-AAAGCAAACTGTCCGTAGCCCT-30.

Chromatin immunoprecipitation assayChromatin immunoprecipitation (ChIP) was performed using

a Simple ChIP Enzymatic Chromatin IP kit (#9002, Cell SignalingTechnology). In brief, 5 � 107 cells were fixed with 1% finalconcentration of formaldehyde for 10 minutes at room temper-ature. The formaldehyde was quenched by adding 125 mmol/Lglycine for 5 minutes at room temperature. Cells were washedwith cold PBS containing a protease inhibitor cocktail and were

RNF126 Determines CHEK1 Inhibitor Use in Breast Cancer

www.aacrjournals.org Clin Cancer Res; 24(7) April 1, 2018 1631

Research. on September 25, 2020. © 2018 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 11, 2018; DOI: 10.1158/1078-0432.CCR-17-2242

Page 4: RNF126 as a Biomarker of a Poor Prognosis in Invasive Breast … · Cleveland State University, Cleveland, Ohio. 4Division of Oncology Breast Oncology Section, Washington University

then lysed with cold Buffer A. Collected pellets were resuspendedin coldBuffer B and then treatedwith 7mLofmicrococcal nucleasefor 30 minutes at 37�C. Digested chromatin was sonicated andpurified according to kit instructions. Chromatin (10 mg) wasincubated with the following antibodies: 2 mg E2F1 (Santa CruzBiotechnology, sc193), 2 mg H3 (provided by the kit), and 2 mgIgG (provided by the kit). The primers used to amplify the regionscontaining the putative consensus DNA-binding sites of RNF126in the CHEK1 promoter by PCR were as follows: forward50-AGCACTCTGCTTCACCGACT-30, reverse 50-CTGGGCCCAAA-TATGAAGTG-30.

Immunofluorescence analysisImmunofluorescence assays were performed as described pre-

viously (24). Cells growing on slideswerefixed directly in 3%–4%paraformaldehyde. For unextractable CDC45 staining, cells wereextracted for 5 minutes on ice with 0.5% Triton X-100 in cyto-skeletal (CSK) buffer (10 mmol/L PIPES, 300 mmol/L sucrose,100 mmol/L NaCl, 3 mmol/L MgCl2; pH ¼ 6.8) supplementedwith 1mmol/L phenylmethylsulfonyl fluoride, 0.5 mmol/L sodi-um vanadate, and proteasome inhibitor for 10 minutes at 4�C.Then, extracted cells were fixed with 3%–4% paraformaldehyde.The cells were permeabilized for 10 minutes with PBS containing0.5% Triton X-100 for 15minutes at room temperature, followedby blocking with 1% BSA, and then incubated with primaryantibodies. The bound antibodies were revealed with goat anti-mouse IgG Alexa Fluor 594 and chicken anti-rabbit IgG AlexaFluor 488. Slides were viewed at 1,000� magnification with aNIKON 90i fluorescence microscope (photometric cooled monoCCD camera).

ImmunoblottingCellular extracts were prepared by resuspending cells in RIPA

lysis buffer and proteins were resolved by 5%, 12%, or 15%SDS-PAGE. For chromatin CDC45 isolation, chromatin-boundproteins were prepared according to a previous publication (32).In brief, 3 � 106 cells were resuspended in 200 mL of buffer A[10 mmol/L HEPES (pH 7.9), 10 mmol/L KCl, 1.5 mmol/LMgCl2, 0.34 mol/L sucrose, 10% glycerol, 1 mmol/L dithiothrei-tol, and protease inhibitor mixture (Roche MolecularBiochemicals)]. Triton X-100 was added to a final concentrationof 0.1%, and the cellswere incubated for 10minutes on ice.Nucleiwere collected in the pellet (P1) by low speed centrifugation(1,500 � g, 4 minutes, 4�C). The supernatant (S1) was furtherclarified by high speed centrifugation (13,000 � g, 10 minutes,4�C) to remove cell debris and insoluble aggregates. Nuclei (P1)werewashed oncewith buffer A and then lysed in 200 mL of bufferB (3 mmol/L EDTA, 0.2 mmol/L EGTA, 1 mmol/L DTT, andprotease inhibitor mixture). After 10-minute incubation on ice,soluble nuclear proteins (S2) were separated from chromatin bycentrifugation (2,000 � g, 4 minutes). Insoluble chromatin (P2)waswashedonce in buffer B and centrifuged again under the sameconditions. The final chromatin pellet (P3) was resuspendedin 30 mL Laemmli buffer and sonicated for 30 seconds in asonicator using a microtip at 25% amplitude. The fractionedchromatin-bound protein was denatured by boiling the samplefor 5–10 minutes, and analyzed by immunoprecipitation.

Dual-luciferase assaysDual-luciferase assays were conducted as described previously

(33) Cell extracts were prepared according to the instructions of

the manufacturer and assayed in a TD-20/20 luminometer(Turner Designs) using the Dual-Luciferase Reporter assay System(E1910, Promega). Briefly, cells were cotransfected with vectorcontrol, RNF126-WT, RNF126-Df, or RNF126-C229A/C232A andCHEK1-reporter vector in the ratios of 10:1 for 6 hours, thenreplaced with fresh medium and continually cultured for addi-tional 48 hours. The cells were washed with PBS, and thenlysed with PLB reagent. Finally, the lysate were detected witha TD-20/20 luminometer according to the manufacturer'sinstructions.

Cell-cycle analysisCell-cycle analyses were conducted as described previously

(24). Cells were collected and fixed with cold 70% ethanol.Approximately 106 cells/mL were incubated for 30 minutes withstaining solution containing RNase A (10 mg/mL, Sigma), andpropidium iodide (20 mg/mL, Sigma) for 30 minutes. The DNAcontent was measured by flow cytometry.

DNA fiber assaysDNA fiber assays were performed as published with

some modifications (24, 34). Cells were pulse-labeled with50 mmol/L IdU (Sigma-Aldrich, I7125) for 40 minutes and thenpulse-labeled with 200 mmol/L CldU (Sigma–Aldrich #C6891)for 40 minutes in the presence or absence of CHEK1 inhibitor. Atthe end of the CldU pulse, cell suspensions (2.5 mL) were mixedwith 7.5 mL of lysis buffer [0.5% SDS, 200 mmol/L Tris-HCl(pH 7.4), 50 mmol/L EDTA]. Each mixture was dropped on thetop of an uncoated regular glass slide. Slides were inclined at 25�

to spread the suspension on the glass. Once dried, DNA spreadswere fixed by incubation for 10 minutes in a 3:1 solution ofmethanol-acetic acid. The slides were dried and placed in pre-cooled 70%ethanol at 4�C for at least 1 hour. DNAwas denaturedwith 2.5 mol/L HCl for 30 minutes at 37�C. The slides wereblocked in 1% BSA in PBS for 30 minutes at room temperatureand then incubated with mouse anti-BrdUrd antibody (BD Bios-ciences, #347580) at a 1:200 dilution and rat anti-CldU antibody(Abcam, #ab6326) at a 1:400 dilution. The slides were incubatedwith secondary fluorescent antibodies [goat anti-mouse IgG[HþL] Alexa Fluor 594 secondary antibody (A-11032, 1:400;Thermo Fisher Scientific); or chicken anti-rabbit IgG [HþL] AlexaFluor 488 secondary antibody [A-21441, 1:400]; Thermo FisherScientific]. Replication fibers were viewed at 1,000 � magnifica-tion on a NIKON 90i fluorescence microscope (photometriccooled mono CCD camera; Nikon). Signals were measured usingImageJ software (NCI/NIH), with some modifications madespecifically to measure DNA fibers.

AntibodiesPrimary antibodies used forWestern blots were against: BRCA1

(Clone D-9, 1:200; Santa Cruz Biotechnology); RPA2 (CloneNA18, 1:100; Calbiochem/EMD Millipore); E2F1 (Clone KH95,1:200; Santa Cruz Technology); b-Actin (Clone AC-74, 1:50,000;Sigma-Aldrich); CHEK1 (G-4, 1:200; Santa Cruz Biotechnology);phospho-CHEK1 antibody (#2344, CHEK1-pSer317, 1:500; CellSignaling Technology); phospho-CHEK1 antibody (#133D3,CHEK1-p345, 1:500; Cell Signaling Technology); E2F1 (cloneKH95 sc-251, 1:500; Cell Signaling Technology); CDC45 (G-12sc55569, 1:200; Santa Cruz Biotechnology); g-H2AX (ser139JBC301,1:500;Millipore clone);p-RPA2(S4/S8; rabbitpolyclonal,BL647, 1: 1000; Bethyl Laboratories), CDC25A (clone DCS-120,

Yang et al.

Clin Cancer Res; 24(7) April 1, 2018 Clinical Cancer Research1632

Research. on September 25, 2020. © 2018 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 11, 2018; DOI: 10.1158/1078-0432.CCR-17-2242

Page 5: RNF126 as a Biomarker of a Poor Prognosis in Invasive Breast … · Cleveland State University, Cleveland, Ohio. 4Division of Oncology Breast Oncology Section, Washington University

1:100; Thermo Fisher Scientific); ORC2 (sc13238, 1:200; SantaCruzBiotechnology);CDK2(610146,1:200;BDBiosciences); andCYCLIN E (sc247, 1:200; Santa Cruz Biotechnology); PARP(65995,1:400; BD Biosciences); CASPASE 9 (M044232,1:1000;BD Biosciences); cleaved CASPASE 9 (D35427, 1:500; Calbio-chem);CASPASE8 (M043764,1:250;BDBiosciences);CASPASE7(SC56063,1:500; Santa Cruz Biotechnology); CASPASE 3(#9665,1:500; Cell Signaling Technology); cleaved CASPASE 8(#2008,1:500; Upstate Biotechnology/Thermo Fisher Scientific);cleaved CASPASE 6 (D35426,1:500; Calbiochem); and cleavedCASPASE 3 (76658,1:100; BD Biosciences). Secondary antibodieswere goat anti-mouse IgG-horseradish peroxidase (HRP)–conjugated (#7076S, 1:1,000; Cell Signaling Technology), goatanti-rabbit IgG-HRP–conjugated (#7074S, 1:1000; Cell SignalingTechnology), and donkey anti-goat IgG-HRP–conjugated (A2216,1:1,000; Santa Cruz Biotechnology).

The primary antibodies used for immunofluorescencewere against: gH2AX (clone JBW301, 1:500; Millipore); RPA2(S4/S8; A300-245A, 1:500; Bethyl Laboratories); CDC45 (H-300clone, SC20685, 1:50; Santa Cruz Biotechnology); phospho-HISTONE H3 (Ser10; #9706, 1:100; Cell Signaling Technology);goat anti-mouse IgG (HþL) Alexa Fluor 594 secondary antibody(A-11032, 1:400; Thermo Fisher Scientific); and chicken anti-rabbit IgG (HþL) Alexa Fluor 488 secondary antibody(A-21441, 1:400; Thermo Fisher Scientific).

Cell line authenticationMCF7 and MDA-MB-231, the two major cell lines used in this

study, were authenticated via Short Tandem Repeat profiling byGenetica DNA Laboratories (a LabCorp brand) using a PowerPlex16HS amplification kit (Promega Corporation) and GeneMapperID v3.2.1 software (Applied Biosystems). The authentication ofeach cell linewas confirmed by a 100%match to the reference STRprofile of the respective cell lines from the ATCC.

Statistical analysisStatistical analyses were undertaken using the statistical soft-

ware package, R version 3.3.4 and stata12.0 (StataCorp). Com-parisons between RNF126 staining and various existing prognos-tic factors were performed using a x2 test and logistic regression.Analyses of cumulative survival probability were performed usingthe Kaplan–Meier method and differences between groups weretested by log-rank test.Multivariate analysiswas undertaken usingthe Cox proportional hazard regression model. The effects ofCHEK1 inhibition on DNA repair recruitment/foci, DSB forma-tion, and replication dynamics were examined using t test (twogroups) or ANOVA (more than two groups). Tukey honest sig-nificant difference (HSD) test was further used to compare thedifference between groups. Correlation analysis was examinedusing Spearman rank correlation.

ResultsRNF126 is highly expressed in invasive breast cancer and is anindependent predictive marker for a poor prognosis

To determine RNF126 protein expression in cases of invasivebreast cancer, we collected 110 early-stage operable primaryinvasive breast cancer specimens and 78 adjacent normal tissuesfor study. All patients were female. The clinicopathologic featuresof patients with breast cancer enrolled in this study are shown inSupplementary Table S1. RNF126 expressionwas detected by IHC

(Fig. 1AandB). Because of the lack of any study todefinepositivityaccording the expression level of RNF126, we determinedRNF126 staining in tissues in accordancewith an immunoreactivescore (IRS) proposed by Remmele and Stegner (31). Of allsamples, 55.45% (61 cases) of tumors were positive for RNF126staining while 44.55% (49 cases) showed negative staining. Incomparison, only 7.69% (6 cases) of adjacent tissue samplesshowed positive immunoreactivity to RNF126 and 92.31%(72 cases) displayed negative staining. Thus, the difference inRNF126 immunoreactivity between tumor samples and adjacenttissues was significant (x2 ¼ 45.3894, P < 0.001; Fig. 1A). Rep-resentative RNF126 staining in both normal and tumor tissues isshown in Fig. 1B. RNF126 staining was found in both the nucleusand cytoplasm of cancer cells, a result consistent with a previousreport (10) and that of our unpublished data showing thatRNF126 is located in both the cytoplasm and nucleus of culturedcancer cells. In addition, RNF126 protein expression was furthercompared with several clinicopathologic variables in breast can-cer, such as age, TNM stage, histologic grade, menstruation status,andmolecular subtypes (Supplementary Table S2).With regard tocases with luminal A tumors, 68.75% (11/16), as well as 58.49%(31/53) of cases with luminal B tumors, which were both positivefor ER/PR, displayed positive RNF126 staining, whereas only42.86% (9/21) of cases with triple-negative breast cancer and50.00% (10/20) of cases with HER2-enriched tumors were pos-itive for RNF126 staining. Nevertheless, differences in RNF126expression between triple-negative, HER2, and hormone recep-tor–positive tumors were not statistically significant (c2¼ 2.9327P ¼ 0.402). In addition, logistic regression analysis was alsoestablished to measure the relationship between RNF126 expres-sion and clinicopathologic parameters, including patient age,TNM stage, histologic grade, menstruation, and molecular sub-types. In thismultivariable regression analysis, theORswere 1.57,1.07, 1.03, 0.64, and 0.67, respectively. The P values for allparameters were more than 0.05 (Fig. 1C), indicating thatRNF126 expression had no obvious relationship with thesewell-known clinicopathologic factors.

Next, using recurrence, metastasis, or deaths as endpoints thatreflect a low cumulative survival probability and poor prognosis,Kaplan–Meier plots for negative versus positive RNF126 expres-sion showed that RNF126–positive staining was associated with apoor prognosis (log-rank test, P ¼ 0.003; Fig. 1D). The medianfollow-up was 102 months (range 14–145 months). To deter-mine whether RNF126 expression was associated with outcomesin the group of patients who received adjuvant therapies, 90patients who received adjuvant chemotherapy after surgical resec-tion based on RNF126 staining of their tumors were sorted intosubgroups. Patients who showed RNF126-positive staining ofbreast cancer tumors displayed a lower cumulative survival prob-ability compared with patients who had negative RNF126 stain-ing of breast cancer tumors (log-rank test, P ¼ 0.001; Fig. 1E),indicating that RNF126-positive staining was associated with apoor outcome in the group of patients who received adjuvantchemotherapy. Finally, we used aCox proportional hazardmodelto determine the prognostic value of RNF126. RNF126 immu-noreactivity, patient's age, TNM stage, histologic grade, menstru-ation, and molecular subtypes were chosen as risk variables sinceall are potential factors affecting a low cumulative survival prob-ability of breast cancer. HRs are indicated in Fig. 1F. TheHR valuesfor RNF126 immunoreactivity and TNM stage were 7.3 (P ¼0.009) and 3.8 (P ¼ 0.002), respectively. This indicates that in

RNF126 Determines CHEK1 Inhibitor Use in Breast Cancer

www.aacrjournals.org Clin Cancer Res; 24(7) April 1, 2018 1633

Research. on September 25, 2020. © 2018 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 11, 2018; DOI: 10.1158/1078-0432.CCR-17-2242

Page 6: RNF126 as a Biomarker of a Poor Prognosis in Invasive Breast … · Cleveland State University, Cleveland, Ohio. 4Division of Oncology Breast Oncology Section, Washington University

multivariate analyses, RNF126 expression and TNM stage are twoindependent factors related to a poor outcome in patients withinvasive breast cancer (Fig. 1F). Thus, high RNF126 expression isassociated with a poor prognosis and is an independent predictorof a poor prognosis in breast cancer.

RNF126 facilitates expressionof theCHEK1 gene via interactionwith E2F1

That RNF126 is associated with a poor prognosis highlights theclinical significance of this protein in breast cancer. However, aspecific inhibitor of RNF126 is not currently available. Studyingthe role of RNF126 in the regulation of CHEK1 expression willprovide new opportunities for therapeutic intervention in breastcancer. RNF126 knockdown by two shRNAs targeting different

regions ofRNF126 led todecreasedCHEK1protein levels inMCF7(Fig. 2A) and MDA-MB-231 cells (Fig. 2B). Of note, downregula-tion of RNF126 was tolerated well by MCF7 and MDA-MB-231cells, without a significant alteration in cell-cycle profiles beingobserved (ref. 12; Supplementary Fig. S1). Thus, the decreasedexpression of CHEK1 in cells depleted of RNF126 was not causedby cell-cycle changes in our experimental conditions. However, ithas been suggested that RNF126 knockdown can cause cell arrest(10). This discrepancy may be caused by differences in themagnitude of RNF126 knockdown. RNF126 likely regulatesCHEK1 at the transcriptional level because RNF126 depletionresulted in a decrease inCHEK1mRNA in both cell lines (Fig. 2C).In accordance with these results, overexpression of Flag-RNF126led to increased CHEK1 protein (Fig. 2D and E) andmRNA levels

Figure 1.

RNF126 high expression was associated with poor outcomes in patients with breast cancer and was an independent predictive marker for a poor prognosis.A, The percentage of invasive breast cancer tumors with RNF126-positive staining was elevated, compared with that of adjacent regions (x2 test, P < 0.001).B, Representative RNF126 staining detected by IHC in adjacent normal and invasive breast cancer tissues. Anti-RNF126 antibody (ab183102) was used.Adjacent normal tissues were collected 5 cm away from the edge of tumors. Specimens were surgically removed before patients were exposed to anyneoadjuvant treatment. C, Logistic regression analysis of RNF126 expression and clinicopathologic parameters. RNF126 expression had no obvious relationshipwith the indicated clinicopathologic variables. D, Kaplan–Meier survival analysis in patients with invasive breast cancer. Increased RNF126 expressioncorrelates with a lower probability of cumulative survival. Recurrence, metastasis, or death were the final events (n ¼ 110). E, Kaplan–Meier survival analysis inpatients with invasive breast cancer who received adjuvant chemotherapy. RNF126-positive staining was also associated with a poor prognosis in patientswith a subtype of invasive breast tumor who received chemotherapy (n ¼ 90). F, Expression of RNF126 was an independent predictor of a poor prognosis.Multivariate analyses of RNF126 expression and clinicopathologic parameters in a Cox proportional hazards model are indicated. RNF126-positive staining wasan independent factor related to patients' poor outcomes (HR, 95% CI: 95% confidence interval).

Yang et al.

Clin Cancer Res; 24(7) April 1, 2018 Clinical Cancer Research1634

Research. on September 25, 2020. © 2018 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 11, 2018; DOI: 10.1158/1078-0432.CCR-17-2242

Page 7: RNF126 as a Biomarker of a Poor Prognosis in Invasive Breast … · Cleveland State University, Cleveland, Ohio. 4Division of Oncology Breast Oncology Section, Washington University

(Fig. 2F) in both MCF7 and MDA-MB-231 cells. Most important-ly, the E3 ligase activity of RNF126 appears to be dispensable forthe regulation of CHEK1 expression because the expression of avalidated RNF126 E3 ligase mutant (RNF126 C229A/C232A;ref. 10) retained the ability to increase CHEK1 proteinexpression in both MCF7 and MDA-MB-231 cells (Fig. 2G andH). This result was consistent with our previous study where E3ligase activity of RNF126 was found not to be required for BRCA1expression (12).

We next investigated whether interaction between RNF126 andE2F1 was required for controlling CHEK1 expression by using aCHEK1 promoter–driven luciferase reporter (35), given that pro-motion of E2F1 mediated–transactivation by RNF126 depends

on the direct interaction of these two proteins via a 185–195 (f)region in RNF126. Flag-RNF126 overexpression increasedluciferase activity, indicating that RNF126 promotes transactiva-tion of the CHEK1 promoter. In contrast, RNF126-Df overexpres-sion failed to induce a significant increase in luciferase activity butreduced luciferase activity compared with control cells in bothMCF7 and MDA-MB-231 cell lines (Supplementary Fig. S2A),indicating that RNF126-Df expression may interfere with thefunction of endogenous RNF126. These results are consistentwith our previous report suggesting that a RNF126-Df mutantlacking an association with E2F1 leads to a loss of function ofRNF126 in promoting the E2F1-mediated transactivation ofBRCA1; it also has a dominant-negative effect (12). This result

Figure 2.

RNF126 facilitated CHEK1 expression. A and B, RNF126 knockdown by shRNAs led to decreased expression of CHEK1 protein in MCF7 (A) and MDA-MB-231 cells (B;top). Band intensities of RNF126 and CHEK1 protein expression in cells, with or without RNF126 depletion, were quantified using ImageJ software, and normalized tob-actin. n ¼ 3 (bottom; MCF7: P1 ¼ 0.040, P2 ¼ 0.014, P3 ¼ 0.016, P4 ¼ 0.013; MDA-MB-231: P1 ¼ 0.012, P2 ¼ 0.002, P3 ¼ 0.021, P4 ¼ 0.024). C, RNF126 andCHEK1 mRNA levels in MCF7 or MDA-MB-231 cells, with or without RNF126 knockdown by shRNAs. n ¼ 3 (one-way ANOVA, P1 ¼ 0.002, P2 ¼ 0.002, P3 ¼ 0.012,P4 ¼ 0.007, P5 ¼ 0.007, P6 ¼ 0.005, P7 ¼ 0.014, P8 ¼ 0.005). D and E, Flag-RNF126 overexpression resulted in increased CHEK1 protein expression in MCF7and MDA-MB-231 cells. CHEK1 protein band intensities were quantified using ImageJ software, and normalized to b-actin. n ¼ 3. D, One of three independentexperiments is presented in E. F, The level of CHEK1 mRNA expression in MCF7 or MDA-MB-231 cells, with or without Flag-RNF126WT overexpression; n ¼ 3(paired t test). G and H, The expression of an E3 ligase mutant of RNF126 did not affect CHEK1 protein expression. MCF7 or MDA-MB-231 cells were transfected withcontrol vector, Flag-RNF126-WT, or E3 ligase-deficient RNF126 (Flag-RNF126-C229A/C232A) plasmids and levels of CHEK1 protein were then detected byWestern blotting. RNF126 and CHEK1 protein band intensitieswere quantified using ImageJ software, and normalized to b-actin; n¼ 3 (one-wayANOVA, P1¼0.037,P2 ¼ 0.008, P3 ¼ 0.001, P4 ¼ 0.001, P5 ¼ 0.023, P6 ¼ 0.004, P7 ¼ 0.008, P8 ¼ 0.013). G, One of three independent experiments is presented in H.

RNF126 Determines CHEK1 Inhibitor Use in Breast Cancer

www.aacrjournals.org Clin Cancer Res; 24(7) April 1, 2018 1635

Research. on September 25, 2020. © 2018 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 11, 2018; DOI: 10.1158/1078-0432.CCR-17-2242

Page 8: RNF126 as a Biomarker of a Poor Prognosis in Invasive Breast … · Cleveland State University, Cleveland, Ohio. 4Division of Oncology Breast Oncology Section, Washington University

was further supported by a chromatin immunoprecipitation(ChIP) assay showing that RNF126 overexpression enhanced theenrichment of E2F1 on the CHEK1 promoter; however, RNF126-Df overexpression reduced the binding of E2F1 protein to theCHEK1 promoter (Supplementary Fig. S2B). Moreover, thedecreased expression of CHEK1 at both mRNA and protein levelswas observed in cells expressing RNF126-Df compared withcontrol cells, whereas increased CHEK1 mRNA (SupplementaryFig. S2C) and protein expression (Supplementary Fig. S2D) wasfound in cells expressing Flag-RNF126-WT. Again, in support ofthe idea that the E3 ligase activity of RNF126 is dispensable for theregulation of CHEK1 expression, the expression of RNF126C229A/C232A led to increased luciferase activity of CHEK1(Supplementary Fig. S2A), enrichment of RNF126 at a promoterofCHEK1 (Supplementary Fig. S2B), and elevatedCHEK1 expres-sion at both mRNA (Supplementary Fig. S2C) and protein (Sup-plementary Fig. S2D) levels, similar to that observed in cellsexpressing wild-type Flag-RNF126. In addition to CHEK1,RNF126 also promoted the expression of CYCLIN E, anotherdownstream factor of E2F in both MCF7 and MDA-MB-231 cells(Supplementary Fig. S3A and S3B). Thus, we conclude that byinteracting with E2F1, RNF126 promoted CHEK1 expression atthe mRNA transcription level.

Correlation of RNF126 and CHEK1 protein expressionNextwewere interested indetermining any association between

RNF126 and CHEK1 in breast cancer tissues. We assessed immu-noreactive staining of these two proteins by analyzing a secondcohort of breast cancer cases that consisted of samples from 67patientswith early-stage primary invasivebreast cancer prepared astissue microarrays (TMA; n ¼ 67). Both RNF126 and CHEK1staining were determined by IHC using TMA and quantified byIRS scores. Of note, CHEK1 immunoreactivity was predominantlylocated in the cytoplasmandwas granular in appearance, althoughnuclear staining was also observed. CHEK1–positive staining wasfound in 94.59% (35/37) of RNF126-positive staining breastcancer samples. CHEK1 staining was negative in 80% (24/30) ofRNF126-negative stainingbreast cancer samples. The expressionofRNF126 in tissues was related to that of CHEK1 (x2 ¼ 38.82, P <0.001, Cram�er V ¼ 0.7612; Fig. 3A). Representative staining ofthese two proteins is shown in Fig. 3B. Thus, in invasive breastcancer, there was a strong and statistically significant correlationbetween RNF126 and CHEK1 protein expression.

In addition, we further analyzed the expression of RNF126 andCHEK1 proteins in a panel of 16 human mammary carcinoma–derived cell lines that comprised: luminal A, ERþ breast cancer(MCF7, ZR-75-1 and T47D), luminal B (MDA-MB-361, BT474),HER2þ breast cancer (HCC202, SK-BR-3, HCC1569) and triple-negative breast cancer (MDA-MB-231, HCC1143, HCC1954,HCC38, HCC1187, HCC70, BT549, MDA-MB-468) cells. We setMCF10A, a normal immortalized breast epithelial cell line, as acontrol. The expression of RNF126 and CHEK1 proteins wasdetermined byWestern blotting. Band intensities were quantifiedusing ImageJ software, and normalized to b-actin (n ¼ 3; Fig. 3Cand D). RNF126 expression was increased in a large majority ofbreast cancer cell lines when compared with a MCF10A cell lineused as a control. The highest level of RNF126 protein was foundin highly tumorigenic MDA-MB-231 cells (Fig. 3C and D). Cor-respondingly, CHEK1 protein expression was also relatively highin these cells. The cell lines, BT474 and ZR751, showed lower orundetectable levels of RNF126 and CHEK1 protein expression

compared with MDA-MB-231 cells (Fig. 3C and D). Therefore, apositive correlation between RNF126 and CHEK1 proteinexpression was observed in breast cancer cell lines (Fig. 3E;correlation coefficient of Spearman rank correlation, r ¼ 0.682,P ¼ 0.004), which is consistent with observations from breastcancer tissues (Fig. 3A). In addition, we also measured RNF126andCHEK1mRNAby qRT-PCR (n¼ 3; Fig. 3F). Levels of RNF126protein essentially paralleled mRNA levels of CHEK1 (Fig. 3G;Spearman rank correlation, r ¼ 0.532, P ¼ 0.034). This result isconsistent with Fig. 2 and Supplementary Fig. S2 showing thatRNF126 promoted CHEK1mRNA expression. However, interest-ingly, RNF126 protein and mRNA transcripts did not correlate intested breast cancer cell lines (Fig. 3H; Spearman rank correlation,r ¼ 0.300, P ¼ 0.259), indicating that the high expression ofRNF126 may not be a consequence of transcriptional regulation.Similarly, a corelationship between the expression of RNF126protein andCYCLINEmRNAexisted (Spearman rank correlation,r¼0.624,P¼0.009; Supplementary Fig. S3D) that alignswith theresult described in Supplementary Fig. S3A and S3B whereRNF126 facilitated the expression of CYCLIN E at both mRNAand protein levels (Supplementary Fig. S3A and S3B). Thus, weconcluded that RNF126 andCHEK1 protein expression positivelycorrelated in both breast cancer tissue and cell lines.

CHEK1 inhibition by pharmacologic CHEK1 inhibitors ismore effective against breast cancer cells expressing ahigher level of RNF126

We chose two pairs of breast cancer cell lines showing higher(MDA-MB-231, and MDA-MB-468), or lower/undetectableRNF126 expression (BT474, ZR751) for a 3-(4,5-dimethylthia-zol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay(Fig. 4A). LY2603618, one of the first highly selective and potentCHEK1 inhibitors, was used in this study. We found that the twocell lines expressing higher levels of RNF126 were more sensitiveto LY2603618 compared with the cells showing lower RNF126expression, suggesting that RNF126 expression may determinesensitivity toCHEK1 inhibitors (Fig. 4A). To confirm these results,we determined the effect of RNF126 knockdown by RNF126shRNA#1 (Fig. 4B and C) or #2 (Supplementary Fig. S4A andS4B) on the efficacy of LY2603618. LY2603618 exposure resultedin more killing of parental cells MCF7 and MDA-MB-231, com-pared with the corresponding cells with RNF126 knockdown byshRNA inMTT (Fig. 4B; Supplementary Fig. S4A and S4B) and/orcolony-forming assays (Fig. 4C). Treatment with a second CHEK1inhibitor, AZD7762, also decreased RNF126-expressing cell num-bers compared with both MCF7 (Supplementary Fig. S4C) andMDA-MB-231 (Supplementary Fig. S4D) cells with knockeddown RNF126, as determined by MTT assay. CHEK1 inhibitionwasmonitored bymeasuring protein levels ofCHEK1-p-S345 andits downstream factor CDC25A (Fig. 4D). Thus, we concludedthat RNF126 depletion abrogated CHEK1-inhibited cell killing.

The reduction in cell viability was accompanied by an increasein CHEK1 inhibitor–induced apoptosis as determined bymeasur-ing optimal biomarkers of apoptosis, such as cleaved CASPASE 3,6, 7, 8, 9, as well as PARP. Cleaved PARP and cleaved CASPASE 7increased in LY2603618 treated MCF7 (Supplementary Fig. S5A)and MDA-MB-231cells (Supplementary Fig. S5B), whereas anobvious increase was not found in corresponding cells withRNF126 depletion under the same conditions. Interestingly,cleaved CASPASE 8 was observed in MDA-MB-231 cells withintact RNF126, but a reduced effect on cleaved CASPASE 8 was

Yang et al.

Clin Cancer Res; 24(7) April 1, 2018 Clinical Cancer Research1636

Research. on September 25, 2020. © 2018 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 11, 2018; DOI: 10.1158/1078-0432.CCR-17-2242

Page 9: RNF126 as a Biomarker of a Poor Prognosis in Invasive Breast … · Cleveland State University, Cleveland, Ohio. 4Division of Oncology Breast Oncology Section, Washington University

observed in RNF126–depleted cells (Supplementary Fig. S5B).However, cleaved CASPASE 8 was not seen in MCF7 cells (Sup-plementary Fig. S5A). The differences in the response of apoptosisproteins in MCF7 andMDA-MB-231 cells may be due to differen-ces in basal levels of apoptotic proteins. For instance, CASPASE 3was absent in MCF7 cells, whereas CASPASE 3 was present inMDA-MB-231 cells (36). Interestingly, according to immunoflu-orescence (IF) results, we found that as a single agent, CHEK1inhibition by LY2603618 did not increase the rate of mitotic cellsin MCF7 cells, with or without RNF126 knockdown. Instead,CHEK1 inhibition resulted in a decrease in the proportion ofmitotic cells in MCF7 (Supplementary Fig. S5C) and MDA-MB-231 cell lines (Supplementary Fig. S5D), but not in cell linesshowing RNF126 depletion (representative staining of mitoticcells, as determined by IF of p-HISTONE H3, is shown in Sup-plementary Fig. S5E). Thismay be explained by the fact that whenRNF126 is intact, DNA damage induced by CHEK1 inhibitiontriggers ATR activity and G2–M arrest, preventing cells fromentering the next stage. However, in cells with depleted RNF126,

lessDNAdamage is induced byCHEK1 inhibition. An insufficientamount of DNA damage may trigger a G2–M checkpoint byCHEK1 inhibition. Thus, we conclude that CHEK1 inhibitionwas more effective in cells expressing higher levels of RNF126.Of note, a similar result was also seen with an ATR inhibitor. ATRinhibition by AZD6738 was more toxic in MCF7 (SupplementaryFig. S6A and S6B) and MDA-MB-231 cells (Supplementary Fig.S6C and S6D) with intact RNF126, compared with cells withRNF126 knocked down by RNF126 shRNA #1 (SupplementaryFig. S6A and S6C) or #2 (Supplementary Fig. S6B and S6D). Theseresults support thenotion thatATRhas a similar function to that ofCHEK1 in terms of suppressing oncogenic stress/checkpoints/HR.

CHEK1 inhibitionupregulates replication stress, particularly incells showing higher expression of RNF126

We next determined the extent of replication stress followingCHEK1 inhibition in cells, with or without RNF126 knockdown.We analyzed foci of phosphorylated RPA2 (p-RPA2), amarker forreplication stress, in response to exogenous DNA-damaging

Figure 3.

Correlation of RNF126 and CHEK1 protein expression. A, Coexpression of RNF126 and CHEK1 proteins was analyzed by tissue microarrays (TMAs; n ¼ 67).B, Typical immunostaining patterns for serial sections of the same tumor for RNF126 and CHEK1. TMA immunostaining was visualized with 3,30-diaminobenzidinesubstrate following probing with antibodies against RNF126 (ab183102, 1:100, Abcam) and CHEK1 (25887-1-AP, 1:150, Proteintech). C, The expression ofRNF126, CHEK1 and CYCLIN E proteins in a panel of 16 human breast cancer–derived cell lines by Western blotting. Normal primary cultured MCF10A cells wereused as a control. Four cell lines chosen for the toxicity assay are labeled either in blue (lower expression) or yellow (higher expression).D,Band intensities of RNF126and CHEK1 protein expression in breast cancer cell lines were quantified using ImageJ software, and normalized to b-actin. n ¼ 3. E, Positive correlationbetween RNF126 and CHEK1 proteins in breast cancer cell lines (Spearman rank correlation, r ¼ 0.682, P ¼ 0.004). F, The mRNA expression of RNF126 andCHEK1 in a panel of breast cancer cell lines was detected by quantitative real-time PCR; n ¼ 3. G, RNF126 protein levels paralleled CHEK1 mRNA levels (Spearmanrank correlation, r ¼ 0.532, P ¼ 0.034). H, RNF126 protein and mRNA transcripts did not correlate in tested breast cancer cell lines (Spearman rank correlation,r ¼ 0.300, P ¼ 0.259).

RNF126 Determines CHEK1 Inhibitor Use in Breast Cancer

www.aacrjournals.org Clin Cancer Res; 24(7) April 1, 2018 1637

Research. on September 25, 2020. © 2018 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 11, 2018; DOI: 10.1158/1078-0432.CCR-17-2242

Page 10: RNF126 as a Biomarker of a Poor Prognosis in Invasive Breast … · Cleveland State University, Cleveland, Ohio. 4Division of Oncology Breast Oncology Section, Washington University

agents by immunofluorescence staining. A more profoundincrease in the proportion of cells with p-RPA2 foci was observedin MCF7 cells compared with cells depleted of RNF126 byRNF126shRNA #1 (Fig. 5A). The greater increase in p-RPA2 fociin LY2603618-treatedMCF7 cells were also confirmed byWesternblot analysis (Fig. 5B). In addition, LY2603618 treatment led to agreater increase in gH2AX foci (Fig. 5C) and protein levels (Fig.5D) in parental MCF-7 cells compared with MCF-7 cells withRNF126knockdown.Ofnote, althoughCHEK1 inhibition causedan increase in CHEK1-p-S345 in cells depleted of RNF126, theextent was much less than that seen in cells with intact RNF126.This result supported our hypothesis that CHEK1 inhibition leadsto a reduced amount of DNA damage in RNF126-depleted cells. Asimilar resultwas observed in parentalMDA-MB-231 cells (Fig. 5Eand F), and in MCF7 and MDA-MB-231 cells treated with thesecond CHEK1 inhibitor, AZD7762 (Supplementary Fig. S7A andS7B). These results suggest that CHEK1 inhibition suppresses theproliferation of breast cancer cells expressing higher levels ofRNF126. A similar result was also seen using a second RNF126shRNA #2 (Supplementary Fig. S7C and S7D). RNF126 knock-down by RNF126 shRNA #2 abrogated CHEK1 inhibition–induced replication stress in both MCF7 and MDA-MB-231 cells.

CHEK1 inhibition disrupts dynamics of replication forks,particularly in cells expressing higher levels of RNF126

Deregulated origin firing contributes to oncogene-inducedreplication stress (37). We next determined how treatment withCHEK1 inhibitor affects the initiation of DNA replication by

analyzing DNA fiber spreads, according to the protocol illus-trated in Fig. 6A and our previous publication (24). Thepercentage of new origins increased when cells were treatedwith LY2603618 in both parental MCF7 cells and MCF7 cellswith RNF126 knockdown (Fig. 6B). However, the magnitude ofthe increase was greater in parental cells compared with cellswith RNF126 knockdown. A similar result was seen inMDA-MB-231 cells (Fig. 6C). CHEK1 is involved in controllingreplication initiation via regulating CDC45 (38), a protein thatis implicated in initiation rather than elongation processes. Wenext measured the amount of CDC45 in a nonextractablechromatin fraction. LY2603618 treatment caused a remarkableincrease in the amount of nonextractable CDC45 protein incontrol cells compared with RNF126-depleted cells (Fig. 6D),although overall CDC45 levels were comparable (Fig. 6D). Theeffect of CHEK1 inhibition on chromatin loading of CDC45was further confirmed by IF assay (Fig. 6E). As a result of analteration in replication initiation, the elongation ratio wasdecreased when CHEK1 activity was inhibited, particularly inMCF7 cells and MDA-MB-231 with intact RNF126 (Fig. 6F andG). Representative DNA fiber staining is presented in Supple-mentary Fig. S8. Cumulatively, the results presented in Fig. 6suggest that CHEK1 inhibition led to a greater increase inreplication initiation and a decrease in replication speed, par-ticularly in cells with higher RNF126 expression. This result wasconsistent with the results described in Fig. 5 where CHEK1inhibition caused greater replication stress in cells with RNF126expression compared with RNF126-depleted cells.

Figure 4.

CHEK1 inhibition by LY2603618 in parental cells compared with cells depleted of RNF126. A, An MTT assay following CHEK1 inhibition by LY2603618 in breastcancer cells with higher RNF126 expression versus breast cancer cells with lower RNF126 expression. Cells were treated with various concentrations of LY2603618for 72 hours; n ¼ 3 (two-way ANOVA, PBT474 vs. MDA-MB-231 < 0.001; PBT474 vs. MDA-MB-468 < 0.001; PZR751 vs. MDA-MB-231 P < 0.001; PZR751 vs. MDA-MB-468 < 0.001).B, MTT assay for observing the effect of the CHEK1 inhibitor, LY2603618, on MCF7 and MDA-MB-231 cell proliferation. Cells were treated with variousconcentrations of LY2603618 for 72 hours; n¼ 3 (two-wayANOVA).C,Clonogenic survival following CHEK1 inhibition by LY2603618 inMCF7 andMDA-MB-231 cells;n ¼ 3 (two-way ANOVA). D, CHEK1 inhibition was monitored by measuring levels of CHEK1 p-S345 and CDC25A by Western blots. Cells were treated withvarious concentrations of LY2603618 for 8 hours. The representative result from three independent experiments is presented.

Yang et al.

Clin Cancer Res; 24(7) April 1, 2018 Clinical Cancer Research1638

Research. on September 25, 2020. © 2018 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 11, 2018; DOI: 10.1158/1078-0432.CCR-17-2242

Page 11: RNF126 as a Biomarker of a Poor Prognosis in Invasive Breast … · Cleveland State University, Cleveland, Ohio. 4Division of Oncology Breast Oncology Section, Washington University

DiscussionHigh RNF126 expression and invasive breast cancer

The biological functions of RNF126 have been exploredrecently (6, 8, 10–12). However, to date, a report determiningRNF126 expression in human cancers is lacking. Our resultssuggest that RNF126 protein was highly expressed in invasivebreast cancer (Fig. 1). Although the mechanism contributing toincreased RNF126 expression is not clear, the data obtainedfrom a panel of breast cancer cell lines suggest levels of RNF126protein and mRNA transcripts are not correlated (Fig. 3). Thus,it is postulated that the increased RNF126 protein measured inbreast cancer tissues may not necessarily be a consequence of analteration in RNF126 mRNA transcripts. In addition, RNF126-positive staining appears to be slightly higher in the ERþ cohortcompared with the other cohorts, such as the triple-negativesubtype. However, this difference did not reach statisticalsignificance. Thus, a further study with a larger number of

invasive breast cancer cases is required. The RNF126 gene mapsto chromosome 19p13.3, which is a commonly deleted regionin ovarian cancer (39–41). Interestingly, a genome-wide studyof breast cancer also detected a high frequency of loss ofheterozygosity (LOH) in the 19p13 genomic region (42). It isnot clear whether the LOH of 19p13 in breast cancer led to thedecreased RNF126 expression observed. However, it is likelythat RNF126 may be a context-dependent signaling moleculeand that the expression of RNF126 in cancer may be contingenton the biological context. The high expression of RNF126 inbreast cancer suggests that RNF126 may contribute to breastcancer development, although the molecular mechanismsbehind this are, as yet, unclear.

In this study, we demonstrate that RNF126 expression isassociated with a poor prognosis, such recurrence, metastasis, ordeaths, in patients with invasive breast cancer (Fig. 1). Neverthe-less, the relationship between RNF126 protein expression and

Figure 5.

CHEK1 inhibition enhanced replication stress, particularly in cellswith RNF126 expression.A andC, The proportion of cellswith foci of phosphorylated RPA2 (p-RPA2,green; A) or gH2AX (red; C) in MCF7 cells, with or without RNF126 knockdown (left). Cells were treated with LY2603618 (5 mmol/L) for the indicated timesand then subjected to immunofluorescence staining. Representative foci of p-RPA2 or gH2AX are indicated (right); n ¼ 3 (two-way ANOVA). B and D, CHEK1inhibition by LY2603618 (5mmol/L) led to a greater increase in levels of p-RPA2 and gH2AX in parentalMCF7 cells comparedwithMCF7 cellswith RNF126 knockdownas determined by Western blot analysis. E, CHEK1 inhibition by LY2603618 (5 mmol/L) led to a greater increase in the levels of p-RPA2 and gH2AX proteins inparental MDA-MB-231 cells, compared with MDA-MB-231 cells with RNF126 knockdown, as determined by Western blotting. F, The proportion of cells with fociof p-RPA2 (left) or gH2AX (right) in MDA-MB-231 cells, with or without RNF126 knockdown, as determined by immunofluorescence; n ¼ 3 (two-way ANOVA).

RNF126 Determines CHEK1 Inhibitor Use in Breast Cancer

www.aacrjournals.org Clin Cancer Res; 24(7) April 1, 2018 1639

Research. on September 25, 2020. © 2018 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 11, 2018; DOI: 10.1158/1078-0432.CCR-17-2242

Page 12: RNF126 as a Biomarker of a Poor Prognosis in Invasive Breast … · Cleveland State University, Cleveland, Ohio. 4Division of Oncology Breast Oncology Section, Washington University

each end point of a poor prognosis needs to be investigatedfurther. Our most significant finding is that high RNF126 expres-sion is an independent predictor for a poorer patient prognosis,which is independent from established prognostic markers suchas patients' age, TNM stage, histologic grade, menstruation, andmolecular subtypes (Fig. 1). Further analysis using adjuvantchemotherapy as a stratification criterion suggested that patientswith RNF126-positive breast cancer tumors had a significantlylower cumulative survival probability compared with those withRNF126-negative tumors (Fig. 1). Although conclusions fromourobservations are limited due to the small number of patients whoreceived adjuvant therapies (n ¼ 90), the differences in survivalprobabilities are striking and suggest that RNF126 expressionlevels may influence the response to adjuvant therapies. As DSBrepair proteins have been suggested to play an important role inthe cellular response to chemotherapy as well as to radiotherapy,the role of RNF126 in the repair of DSBs by promoting HR and

NHEJ may contribute to its poor prognosis. The association ofRNF126 with a poor prognosis in breast cancer highlights theclinical significance of this protein.

Higher expression of RNF126 as a biomarker for determiningCHEK1 inhibitor use

In our study, we identify a relationship between RNF126 andCHEK1 by demonstrating that RNF126 promotes E2F1-medi-ated expression of CHEK1 transcripts (Fig. 2), which is consis-tent with our previous publication that outlined how RNF126promoted the activity of the transcriptional factor, E2F1 (12).Breast cancer tumors expressing higher levels of RNF126 oftenshow elevated CHEK1 protein expression in both breast cancertissues and cell lines (Fig. 3). Most importantly, a correlationbetween RNF126 protein levels and CHEK1 transcripts in breastcancer cell lines was also observed, supporting our finding thatRNF126 promotes CHEK1 expression at transcriptional levels

Figure 6.

CHEK1 inhibition disrupted dynamics of replication forks, particularly in cells expressing RNF126. A, Schematic of DNA fiber analysis (left) in MCF7 cells. Redtracks, IdU; Green tracks, CldU. B and C, CHEK1 inhibition by LY2603618 (5 mmol/L) increased the rate of replication initiation, particularly in cells with intactRNF126, comparedwith cells depleted of RNF126. The frequencyof neworiginswas calculated as the number of green signals (b) divided by the total of green (b) plusgreen/red signals (aþb; right). (B, MCF7; C, MDA-MB-231, n ¼ 3; one-way ANOVA.) D, CHEK1 inhibition led to an increase in nonextractable CDC45 protein,particularly in parental cells with RNF126 compared with cells with RNF126 knockdown, as determined by Western blotting. ORC2 was used as a loading control.E, Measurement of CDC45 chromatin loading by immunostaining after pre-extraction of cells with detergent. Cells presenting with CDC45 staining wereconsidered positive; n ¼ 3 (two-way ANOVA). F and G, CHEK1 inhibition induced a greater decrease in replication fork speeds in MCF7 (F) and MDA-MB-231cells (G) compared with corresponding cells with RNF126 knockdown. The CIdu/Idu ratio was used to determine elongation (n ¼ 3, one-way ANOVA).H, Model for targeting breast cancer cells expressing RNF126 by CHEK1 inhibitors.

Yang et al.

Clin Cancer Res; 24(7) April 1, 2018 Clinical Cancer Research1640

Research. on September 25, 2020. © 2018 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 11, 2018; DOI: 10.1158/1078-0432.CCR-17-2242

Page 13: RNF126 as a Biomarker of a Poor Prognosis in Invasive Breast … · Cleveland State University, Cleveland, Ohio. 4Division of Oncology Breast Oncology Section, Washington University

(Fig. 2). Nevertheless, the positive relationship betweenRNF126 protein and CHEK1 transcripts needs to be verifiedin breast tumor tissues in future.

It is well established that ATR/CHEK1 suppress oncogene-induced replication stress. Cancer cells often harbor some degreeof replication stress due to oncogene activities, which can be lethalto cells. Thus, they often upregulate ATR and CHEK1 activity tomediate survival because ATR/CHEK1 suppress replication stressto an intolerable level by the suppression of replication initiationand/or promoting HR (24, 43, 44). In support of this concept,increased ATR/CHEK1 expression was frequently observed in avariety of cancer cells, including lung cancer, ovarian cancer, headneck cancer, triple-negative breast cancer, neuroblastoma, T-cellacute lymphoblastic leukemia, acute myeloid leukemia, andhepatocellular carcinoma. Although the biological significanceof the correlation between RNF126 and CHEK1 expressionremains unknown, it may be related to the inhibition of replica-tion stress by CHEK1 in cells expressing high levels of RNF126.Thus, increased CHEK1 protein expression in RNF126-positivebreast cancer cells is likely related to the suppression of replicationstress because RNF126 also promotes oncogene expression suchas CYCLIN E (Supplementary Fig. S3), an oncogene that causesreplication stress. It is most likely that RNF126-positive breastcancer upregulates oncogenes in addition to CHEK1, renderingcells dependent on ATR/CHEK1 for survival. Indeed, CHEK1inhibition causes greater killing in cells expressing RNF126,whereas a lesser effect was found in cells with RNF126 depletion.Thus, in addition to MYC, CYCLIN E, and H-RAS that have beenreported to affect the outcome of CHEK1 or ATR inhibitors,RNF126 is also a potential factor determining the efficacy ofCHEK1 inhibitors (Fig. 6H). Using RNF126 expression as abiomarker for a CHEK1 inhibitor has a greater advantage thanCHEK1 expression alone because the high expression of CHEK1may not be functionally important. Indeed, p-CHEK1, instead ofCHEK1 expression levels, is a biomarker for CHEK1 inhibitors(45). We also reported that radioresistant breast cancer cellsthat carry high levels of oncogene and DDR proteins, includingATR/CHEK1, are more sensitive to CHEK1 inhibition (24),suggesting that expression of both oncogene and cell-cycle check-point proteins are features that could be targeted by CHEK1inhibitors. Thus, the role of RNF126 in promoting CHEK1 expres-sion, and perhaps also oncogene expression, determine the sen-sitivity of RNF126-positive breast cancer to CHEK1 inhibitors.

The current model for oncogene-induced replication stress isrelated to deregulated replication initiation, because an excess ofongoing replication forks will consume the limited dNTP pooland cause fork stalling (37). This will generate extensive ssDNAregions that are protected byRPA coating.With a limited supply ofRPA, uncoated ssDNA causes DSB. However, ATR/CHEK1 can beactivated during replication stress, which, in turn, suppressesoncogene-induced replication by targeting CDC25A for degrada-tion.Our studies provide evidence that further support the notionthat CHEK1 suppresses replication stress by inhibiting replicationinitiation, particularly in cells expressing RNF126 (Fig. 6). AsATR/CHEK1 can also promote the repair of DSBs by facilitatingHR, increased DSBs induced by CHEK1 inhibition may also berelated to the impaired HR repair of collapsed replication forks.Thus, multiple mechanisms are involved in CHEK1 inhibition–induced replication stress in cells expressing RNF126.

We have reported that RNF126 promotes the expression of HRprotein BRCA1 at the transcriptional level (12). The probability

that BRCA1 affects the efficacy of the CHEK1 inhibitor on breastcancer cells expressing relatively high levels of RNF126 is very low.Transient RNF126 overexpression increases mRNA expression ofthe BRCA1 (12). However, coexpression of these two proteinsmay not be seen in tumor tissues or cancer cell lines, as it is well-known that the BRCA1 promoter is frequently methylated, lead-ing to low expression (46, 47). Even if some high RNF126–expressing cell lines have high BRCA1 protein expression, itseffect may be to reduce, rather than increase, the sensitivityto CHEK1 inhibitors, as HR-defective cells are more sensitive toATR/CHEK1 inhibition (48).

Of note, despite the initial hypothesis that CHEK1 inhibitorscan increase efficacy in combination with IR and chemotherapydrugs, particularly in cells with TP53 deficiency, our studies showthat RNF126 promotes CHEK1 expression and affects sensitivityto CHEK1 inhibitors in cells, with or without wild-type TP53.Ourresults are consistent with previous publications showing thatATR/CHEK1 inhibition can target cancer cells as single agentsindependent of TP53 (24, 49). Thus, acting as single agents and incombinationwith other chemotherapy drugs/IR, themechanismsby which CHEK1 inhibitors lead to cell death may be distinct.

In summary, we identify that RNF126 is highly expressed ininvasive breast cancer and is an independent predictor of a pooroutcome for this disease. High RNF126 expressionmay be used asa potential biomarker for CHEK1 inhibitors. Our study providesproof of concept in preclinical models for a new paradigm fortreating breast cancer expressing high levels of RNF126 by CHEK1inhibitors (Fig. 6H). Identifying breast cancers with high levels ofRNF126 expression that can thenbe targeted byCHEK1 inhibitorswill significantly improve the efficacy of such agents. It will benecessary to validate our findings in breast cancer using largerandomized clinical trials. Thismay be done by assessing whethera simple IHC assay of RNF126 expression performed on routineparaffin-embedded tissue would be able to predict a patient'sresponse to CHEK1 inhibitors. We also need to evaluate whetherRNF126-positive breast tumors are more responsive to CHEK1 orATR inhibitors.

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

Authors' ContributionsConceptionanddesign:X. Yang, Y. Pan, Y. Zhang, S. Li, R.A.Keri, Z.Ma, J. ZhangDevelopment of methodology: X. Yang, Y. Pan, Z. Qiu, Z. Du, Y. Zhang, P. Fa,S. Ma, S. Li, J. ZhangAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): X. Yang, Z. Qiu, Y. Zhang, P. Fa, S. Gorityala, Y. Xu,Z. MaAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis):X. Yang, Z.Qiu, Y. Zhang, P. Fa, S.Gorityala,H.Wang,Y. Xu, C. Yan, Z. Ma, J. ZhangWriting, review, and/or revision of the manuscript: X. Yang, Y. Pan, H. Wang,Z. Ma, J. ZhangAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): Y. Pan, S. Ma, C. Chen, Y. Xu, R.A. KeriStudy supervision: Z. Ma, J. Zhang

AcknowledgmentsThe authors apologize to colleagues whose work was not cited because of

space limitations or ignorance. Our thanks for the service provided by BioMedProofreading LLC.

The work described was supported by a grant (R01CA154625) from theNational Cancer Institute and seed grants from the Case Comprehensive CancerCenter and VeloSano Bike to Cure Foundation (to J. Zhang); a National Natural

RNF126 Determines CHEK1 Inhibitor Use in Breast Cancer

www.aacrjournals.org Clin Cancer Res; 24(7) April 1, 2018 1641

Research. on September 25, 2020. © 2018 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 11, 2018; DOI: 10.1158/1078-0432.CCR-17-2242

Page 14: RNF126 as a Biomarker of a Poor Prognosis in Invasive Breast … · Cleveland State University, Cleveland, Ohio. 4Division of Oncology Breast Oncology Section, Washington University

Science Foundation of China grant (31571452 and 31271503) andGuangdongProvincial Natural Science Foundation of China grant (S2012010008368)and a startup fund from The First Affiliated Hospital of Sun Yat-sen University(to Z. Ma), and scholarships from the Chinese Scholarship Council (CSC). Thisresearch was also supported by the Radiation Resources Core Facility andCytometry & Imaging Microscopy Core Facility of the Case ComprehensiveCancer Center (P30 CA43703).

The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

Received August 1, 2017; revised November 10, 2017; accepted January 5,2018; published OnlineFirst January 11, 2018.

References1. Siegel RL, Miller KD, Jemal A. Cancer Statistics, 2017. CA Cancer J Clin

2017;67:7–30.2. BuchananCL,DornPL, Fey J, GironG,NaikA,Mendez J, et al. Locoregional

recurrence after mastectomy: incidence and outcomes. J Am Coll Surg2006;203:469–74.

3. Huston TL, SimmonsRM. Locally recurrent breast cancer after conservationtherapy. Am J Surg 2005;189:229–35.

4. Yard BD, Adams DJ, Chie EK, Tamayo P, Battaglia JS, Gopal P, et al.A genetic basis for the variation in the vulnerability of cancer to DNAdamage. Nat Commun 2016;7:11428.

5. Smith CJ, Berry DM, McGlade CJ. The E3 ubiquitin ligases RNF126 andRabring7 regulate endosomal sorting of the epidermal growth factorreceptor. J Cell Sci 2013;126(Pt 6):1366–80.

6. Smith CJ, McGlade CJ. The ubiquitin ligase RNF126 regulates the retro-grade sorting of the cation-independent mannose 6-phosphate receptor.Exp Cell Res 2014;320:219–32.

7. Rodrigo-Brenni MC, Gutierrez E, Hegde RS. Cytosolic quality control ofmislocalized proteins requires RNF126 recruitment to Bag6. Mol Cell2014;55:227–37.

8. Benini M, Fortuni S, Condo I, Alfedi G, Malisan F, Toschi N, et al. E3 ligaseRNF126 directly ubiquitinates frataxin, promoting its degradation: iden-tification of a potential therapeutic target for friedreich ataxia. Cell Rep2017;18:2007–17.

9. Delker RK, Zhou Y, Strikoudis A, Stebbins CE, Papavasiliou FN. Solubility-based genetic screen identifies RING finger protein 126 as an E3 ligase foractivation-induced cytidine deaminase. Proc Natl Acad Sci U S A 2013;110:1029–34.

10. Zhi X, Zhao D,Wang Z, Zhou Z, Wang C, ChenW, et al. E3 ubiquitin ligaseRNF126 promotes cancer cell proliferation by targeting the tumor sup-pressor p21 for ubiquitin-mediated degradation. Cancer Res 2013;73:385–94.

11. Ishida N, Nakagawa T, Iemura SI, Yasui A, Shima H, Katoh Y, et al.Ubiquitylation of Ku80 by RNF126 promotes completion of nonhomol-ogous end joining-mediated DNA repair. Mol Cell Biol 2017;37:e00347–16.

12. Wang Y, Deng O, Feng Z, Du Z, Xiong X, Lai J, et al. RNF126 promoteshomologous recombination via regulation of E2F1-mediated BRCA1expression. Oncogene 2015;35:1363–72.

13. Burger A, Amemiya Y, Kitching R, Seth AK.Novel RINGE3ubiquitin ligasesin breast cancer. Neoplasia 2006;8:689–95.

14. Burger AM, Gao Y, Amemiya Y, Kahn HJ, Kitching R, Yang Y, et al. A novelRING-type ubiquitin ligase breast cancer-associated gene 2 correlates withoutcome in invasive breast cancer. Cancer Res 2005;65:10401–12.

15. Wymant JM, Hiscox S, Westwell AD, Urbe S, Clague MJ, Jones AT. The roleof BCA2 in the endocytic trafficking of EGFR and significance as a prog-nostic biomarker in cancer. J Cancer 2016;7:2388–407.

16. Syljuasen RG, Sorensen CS, Hansen LT, Fugger K, Lundin C, Johansson F,et al. Inhibition of human Chk1 causes increased initiation of DNAreplication, phosphorylation of ATR targets, and DNA breakage. Mol CellBiol 2005;25:3553–62.

17. Lopez-Contreras AJ, Gutierrez-Martinez P, Specks J, Rodrigo-Perez S,Fernandez-Capetillo O. An extra allele of Chk1 limits oncogene-inducedreplicative stress and promotes transformation. J Exp Med 2012;209:455–61.

18. GiladO,Nabet BY, RaglandRL, SchoppyDW, Smith KD,DurhamAC, et al.Combining ATR suppression with oncogenic Ras synergistically increasesgenomic instability, causing synthetic lethality or tumorigenesis in adosage-dependent manner. Cancer Res 2010;70:9693–702.

19. Schrader KA, Hurlburt J, Kalloger SE, Hansford S, Young S, Huntsman DG,et al. Germline BRCA1 and BRCA2mutations in ovarian cancer: utility of a

histology-based referral strategy. Obstet Gynecol 2012;120(2 Pt 1):235–40.

20. Ma CX, Janetka JW, Piwnica-Worms H. Death by releasing the breaks:CHK1 inhibitors as cancer therapeutics. Trends Mol Med 2011;17:88–96.

21. Vance S, Liu E, Zhao L, Parsels JD, Parsels LA, Brown JL, et al. Selectiveradiosensitization of p53 mutant pancreatic cancer cells by combinedinhibition of Chk1 and PARP1. Cell Cycle 2011;10:4321–9.

22. Murga M, Campaner S, Lopez-Contreras AJ, Toledo LI, Soria R, MontanaMF, et al. Exploiting oncogene-induced replicative stress for the selectivekilling of Myc-driven tumors. Nat Struct Mol Biol 2011;18:1331–5.

23. Ferrao PT, Bukczynska EP, Johnstone RW, McArthur GA. Efficacy of CHKinhibitors as single agents in MYC-driven lymphoma cells. Oncogene2012;31:1661–72.

24. Zhang Y, Lai J, Du Z, Gao J, Yang S, Gorityala S, et al. Targeting radio-resistant breast cancer cells by single agent CHK1 inhibitor via enhancingreplication stress. Oncotarget 2016;7:34688–702.

25. BrooksK,Oakes V, Edwards B, RanallM, LeoP, Pavey S, et al. A potent Chk1inhibitor is selectively cytotoxic in melanomas with high levels of repli-cative stress. Oncogene 2013;32:788–96.

26. Stevens C, La Thangue NB. The emerging role of E2F-1 in the DNA damageresponse and checkpoint control. DNA Repair 2004;3:1071–9.

27. Degregori J. A new role for E2F1 inDNA repair: all for the greater good. CellCycle 2011;10:1716.

28. Johnson DG, Degregori J. Putting the oncogenic and tumor suppressiveactivities of E2F into context. Curr Mol Med 2006;6:731–8.

29. Bracken AP, Ciro M, Cocito A, Helin K. E2F target genes: unraveling thebiology. Trends Biochem Sci 2004;29:409–17.

30. Lu J, Pan Y, Xia X, GuY, Lei Y. Prognostic significance ofmTORandPTEN inpatients with esophageal squamous cell carcinoma. BioMed Res Int2015;2015:417210.

31. Friedrichs K, Gluba S, Eidtmann H, Jonat W. Overexpression of p53 andprognosis in breast cancer. Cancer 1993;72:3641–7.

32. Xu X, Stern DF. NFBD1/KIAA0170 is a chromatin-associated proteininvolved in DNA damage signaling pathways. J Biol Chem 2003;278:8795–803.

33. Zhuang J, Zhang J, Willers H, Wang H, Chung JH, van Gent DC, et al.Checkpoint kinase 2-mediated phosphorylation of BRCA1 regulates thefidelity of nonhomologous end-joining. Cancer Res 2006;66:1401–8.

34. Seiler JA, Conti C, Syed A, Aladjem MI, Pommier Y. The intra-S-phasecheckpoint affects both DNA replication initiation and elongation: single-cell and -DNA fiber analyses. Mol Cell Biol 2007;27:5806–18.

35. Wang A, Schneider-Broussard R, Kumar AP, MacLeod MC, Johnson DG.Regulation of BRCA1 expression by the Rb-E2F pathway. J Biol Chem2000;275:4532–6.

36. Mooney LM, Al-Sakkaf KA, Brown BL, Dobson PR. Apoptotic mechanismsinT47DandMCF-7humanbreast cancer cells. Br JCancer 2002;87:909–17.

37. Jones RM, Mortusewicz O, Afzal I, Lorvellec M, Garcia P, Helleday T, et al.Increased replication initiation and conflicts with transcription underlieCyclin E-induced replication stress. Oncogene 2013;32:3744–53.

38. GuoC, Kumagai A, Schlacher K, ShevchenkoA,DunphyWG. Interaction ofChk1with Treslin negatively regulates the initiation of chromosomal DNAreplication. Mol Cell 2015;57:492–505.

39. Wang ZJ, Churchman M, Campbell IG, Xu WH, Yan ZY, McCluggage WG,et al. Allele loss and mutation screen at the Peutz-Jeghers (LKB1) locus(19p13.3) in sporadic ovarian tumours. Br J Cancer 1999;80:70–2.

40. Yanaihara N, Okamoto A, Matsufuji S. A commonly deleted region inovarian cancer on chromosome 19p13.3, not including theOAZ1 gene. IntJ Oncol 2003;23:567–75.

41. Amfo K, Neyns B, Teugels E, Lissens W, Bourgain C, De Sutter P, et al.Frequent deletion of chromosome 19 and a rare rearrangement of 19p13.3

Yang et al.

Clin Cancer Res; 24(7) April 1, 2018 Clinical Cancer Research1642

Research. on September 25, 2020. © 2018 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 11, 2018; DOI: 10.1158/1078-0432.CCR-17-2242

Page 15: RNF126 as a Biomarker of a Poor Prognosis in Invasive Breast … · Cleveland State University, Cleveland, Ohio. 4Division of Oncology Breast Oncology Section, Washington University

involving the insulin receptor gene in human ovarian cancer. Oncogene1995;11:351–8.

42. Shen CY, Yu JC, Lo YL, Kuo CH, Yue CT, Jou YS, et al. Genome-wide searchfor loss of heterozygosity using laser capturemicrodissected tissue of breastcarcinoma: an implication for mutator phenotype and breast cancerpathogenesis. Cancer Res 2000;60:3884–92.

43. Benada J, Macurek L. Targeting the checkpoint to kill cancer cells. Biomo-lecules 2015;5:1912–37.

44. Puigvert JC, Sanjiv K, Helleday T. Targeting DNA repair, DNA metabolismand replication stress as anti-cancer strategies. FEBS J 2016;283:232–45.

45. Bryant C, Rawlinson R, Massey AJ. Chk1 inhibition as a novel therapeuticstrategy for treating triple-negative breast and ovarian cancers. BMCCancer2014;14:570.

46. Esteller M, Silva JM, Dominguez G, Bonilla F, Matias-Guiu X, LermaE, et al. Promoter hypermethylation and BRCA1 inactivation insporadic breast and ovarian tumors. J Natl Cancer Inst 2000;92:564–9.

47. Matros E,WangZC, LodeiroG,MironA, Iglehart JD, RichardsonAL. BRCA1promoter methylation in sporadic breast tumors: relationship to geneexpression profiles. Breast Cancer Res Treat 2005;91:179–86.

48. Krajewska M, Fehrmann RS, Schoonen PM, Labib S, de Vries EG, Franke L,et al. ATR inhibition preferentially targets homologous recombination-deficient tumor cells. Oncogene 2015;34:3474–81.

49. Schoppy DW, Ragland RL, Gilad O, Shastri N, Peters AA, Murga M, et al.Oncogenic stress sensitizesmurine cancers to hypomorphic suppression ofATR. J Clin Invest 2012;122:241–52.

www.aacrjournals.org Clin Cancer Res; 24(7) April 1, 2018 1643

RNF126 Determines CHEK1 Inhibitor Use in Breast Cancer

Research. on September 25, 2020. © 2018 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 11, 2018; DOI: 10.1158/1078-0432.CCR-17-2242

Page 16: RNF126 as a Biomarker of a Poor Prognosis in Invasive Breast … · Cleveland State University, Cleveland, Ohio. 4Division of Oncology Breast Oncology Section, Washington University

2018;24:1629-1643. Published OnlineFirst January 11, 2018.Clin Cancer Res   Xiaosong Yang, You Pan, Zhaojun Qiu, et al.   Cancer and CHEK1 Inhibitor Efficacy in Breast Cancer CellsRNF126 as a Biomarker of a Poor Prognosis in Invasive Breast

  Updated version

  10.1158/1078-0432.CCR-17-2242doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://clincancerres.aacrjournals.org/content/suppl/2018/01/10/1078-0432.CCR-17-2242.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://clincancerres.aacrjournals.org/content/24/7/1629.full#ref-list-1

This article cites 49 articles, 12 of which you can access for free at:

  Citing articles

  http://clincancerres.aacrjournals.org/content/24/7/1629.full#related-urls

This article has been cited by 2 HighWire-hosted articles. Access the articles 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/24/7/1629To request permission to re-use all or part of this article, use this link

Research. on September 25, 2020. © 2018 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 11, 2018; DOI: 10.1158/1078-0432.CCR-17-2242