Ovarian Tumor Cell Expression of Claudin-4 Reduces Apoptotic Response to Paclitaxel · Cell Fate...

11
Cell Fate Decisions Ovarian Tumor Cell Expression of Claudin-4 Reduces Apoptotic Response to Paclitaxel Christopher Breed 1,2 , Douglas A. Hicks 1 , Patricia G. Webb 1 , Carly E. Galimanis 1 , Benjamin G. Bitler 1 , Kian Behbakht 1,2 , and Heidi K. Baumgartner 1,2 Abstract A signicant factor contributing to poor survival rates for patients with ovarian cancer is the insensitivity of tumors to standard-of-care chemotherapy. In this study, we investigated the effect of claudin-4 expression on ovarian tumor cell apoptotic response to cisplatin and paclitaxel. We manipulat- ed claudin-4 gene expression by silencing expression [short hairpin RNA (shRNA)] in cells with endogenously expressed claudin-4 or overexpressing claudin-4 in cells that natively do not express claudin-4. In addition, we inhibited claudin-4 activity with a claudin mimic peptide (CMP). We monitored apoptotic response by caspase-3 and Annexin V binding. We examined proliferation rate by counting the cell number over time as well as measuring the number of mitotic cells. Prox- imity ligation assays, immunoprecipitation (IP), and immu- nouorescence were performed to examine interactions of claudin-4. Western blot analysis of tubulin in cell fractions was used to determine the changes in tubulin polymerization with changes in claudin-4 expression. Results show that clau- din-4 expression reduced epithelial ovarian cancer (EOC) cell apoptotic response to paclitaxel. EOCs without claudin-4 proliferated more slowly with enhanced mitotic arrest com- pared with the cells expressing claudin-4. Furthermore, our results indicate that claudin-4 interacts with tubulin, having a profound effect on the structure and polymerization of the microtubule network. In conclusion, we demonstrate that claudin-4 reduces the ovarian tumor cell response to micro- tubule-targeting paclitaxel and disrupting claudin-4 with CMP can restore apoptotic response. Implications: These results suggest that claudin-4 expression may provide a biomarker for paclitaxel response and can be a target for new therapeutic strategies to improve response. Introduction Because of the lack of adequate early detection screening, the majority (>85%) of the patients with epithelial ovarian cancer (EOC) are diagnosed at advanced stages of disease. Delayed detection means that ovarian tumor cells have disseminated beyond their site of origin (fallopian tube or ovary) into the peritoneal cavity. The standard of care for these patients is surgical debulking of the tumor, followed by adjuvant platinum- and taxane-based chemotherapies (cisplatin/carboplatin and paclitax- el/docetaxel; ref. 1). One third of patients with ovarian cancer have tumors that do not respond to initial chemotherapy and of the patients that do respond, approximately 75% of these patients will have disease recurrence (2). Patients with recurrent EOC succumb to the disease following the development of chemoresistance (3). There are several proposed mechanisms responsible for the reduced therapeutic response that include both de novo pathways that inherently make tumor cells resistant and the acquired path- ways that are upregulated in response to repeated chemothera- peutic insult. These pathways include, but are not limited to, drug transporters (i.e. P-glycoprotein) that either inhibit drug uptake or enhance drug efux (46), epigenetic changes (i.e. MLH1 and Tap73 methylation, miR-214, and miR-376c upregulation) that prevent expression of antitumorigenic genes (710), expression or mutations in proapoptotic proteins [i.e., p53, Bcl-2 family proteins, PTEN (1114)], and cytoskeletal organization that pre- vents binding of microtubule-targeting agents (15). Despite the increased understanding of chemoresistance mechanisms, it is unlikely that a single pathway can be targeted to restore tumor sensitivity to all cytotoxic drugs. However, a deeper understanding of patients' genetic and proteomic characteristics, which can predict response and inform optimal therapeutic strategies, is a signicant step forward. Claudin-4 is a member of a large family of transmembrane proteins, of which there are 27 different subtypes (16). Clau- dins have been most studied for their canonical role in tight junctions, where they mediate the specic paracellular barrier properties of an epithelium. Although claudin-4 has been shown to alter the permeability of epithelial monolayers in cell culture (1719), it is rarely localized to tight junctions in human and mouse tissue and is generally found along baso- lateral membranes and throughout the cytosol (2022). The ndings that claudin-4 knockout (KO) mice are phenotypically normal with only a subclinical increase in permeability to small molecules in lung epithelium, but have a reduced capacity for wound healing, further suggest a noncanonical role for claudin- 4 (23). Furthermore, it has been well documented that claudin- 4 is signicantly upregulated in multiple epithelia-derived 1 Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado. 2 Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado. Note: Supplementary data for this article are available at Molecular Cancer Research Online (http://mcr.aacrjournals.org/). Corresponding Author: Heidi K. Baumgartner, University of Colorado Denver, Mail Stop 8613, 12700 E. 19th Ave. Aurora, CO 80045, Phone: 303-724-3505: Fax: 303-724-3512; E-mail: [email protected] doi: 10.1158/1541-7786.MCR-18-0451 Ó2019 American Association for Cancer Research. Molecular Cancer Research www.aacrjournals.org 741 on June 2, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from Published OnlineFirst January 3, 2019; DOI: 10.1158/1541-7786.MCR-18-0451

Transcript of Ovarian Tumor Cell Expression of Claudin-4 Reduces Apoptotic Response to Paclitaxel · Cell Fate...

  • Cell Fate Decisions

    Ovarian Tumor Cell Expression of Claudin-4Reduces Apoptotic Response to PaclitaxelChristopher Breed1,2, Douglas A. Hicks1, Patricia G.Webb1, Carly E. Galimanis1,Benjamin G. Bitler1, Kian Behbakht1,2, and Heidi K. Baumgartner1,2

    Abstract

    A significant factor contributing to poor survival rates forpatients with ovarian cancer is the insensitivity of tumors tostandard-of-care chemotherapy. In this study, we investigatedthe effect of claudin-4 expression on ovarian tumor cellapoptotic response to cisplatin and paclitaxel. We manipulat-ed claudin-4 gene expression by silencing expression [shorthairpin RNA (shRNA)] in cells with endogenously expressedclaudin-4 or overexpressing claudin-4 in cells that natively donot express claudin-4. In addition, we inhibited claudin-4activity with a claudin mimic peptide (CMP). We monitoredapoptotic response by caspase-3 and Annexin V binding. Weexamined proliferation rate by counting the cell number overtime as well as measuring the number of mitotic cells. Prox-imity ligation assays, immunoprecipitation (IP), and immu-nofluorescence were performed to examine interactions ofclaudin-4. Western blot analysis of tubulin in cell fractions

    was used to determine the changes in tubulin polymerizationwith changes in claudin-4 expression. Results show that clau-din-4 expression reduced epithelial ovarian cancer (EOC) cellapoptotic response to paclitaxel. EOCs without claudin-4proliferated more slowly with enhanced mitotic arrest com-pared with the cells expressing claudin-4. Furthermore, ourresults indicate that claudin-4 interacts with tubulin, having aprofound effect on the structure and polymerization of themicrotubule network. In conclusion, we demonstrate thatclaudin-4 reduces the ovarian tumor cell response to micro-tubule-targeting paclitaxel and disrupting claudin-4 with CMPcan restore apoptotic response.

    Implications: These results suggest that claudin-4 expressionmay provide a biomarker for paclitaxel response and can be atarget for new therapeutic strategies to improve response.

    IntroductionBecause of the lack of adequate early detection screening, the

    majority (>85%) of the patients with epithelial ovarian cancer(EOC) are diagnosed at advanced stages of disease. Delayeddetection means that ovarian tumor cells have disseminatedbeyond their site of origin (fallopian tube or ovary) into theperitoneal cavity. The standard of care for these patients is surgicaldebulking of the tumor, followed by adjuvant platinum- andtaxane-based chemotherapies (cisplatin/carboplatin andpaclitax-el/docetaxel; ref. 1). One third of patients with ovarian cancerhave tumors that do not respond to initial chemotherapy and ofthe patients that do respond, approximately 75%of these patientswill have disease recurrence (2). Patients with recurrent EOCsuccumb to the disease following the development ofchemoresistance (3).

    There are several proposed mechanisms responsible for thereduced therapeutic response that include both de novo pathways

    that inherently make tumor cells resistant and the acquired path-ways that are upregulated in response to repeated chemothera-peutic insult. These pathways include, but are not limited to, drugtransporters (i.e. P-glycoprotein) that either inhibit druguptake orenhance drug efflux (4–6), epigenetic changes (i.e. MLH1 andTap73 methylation, miR-214, and miR-376c upregulation) thatprevent expression of antitumorigenic genes (7–10), expressionor mutations in proapoptotic proteins [i.e., p53, Bcl-2 familyproteins, PTEN (11–14)], and cytoskeletal organization that pre-vents binding of microtubule-targeting agents (15). Despite theincreased understanding of chemoresistance mechanisms, it isunlikely that a single pathway can be targeted to restore tumorsensitivity to all cytotoxic drugs.However, a deeper understandingof patients' genetic and proteomic characteristics, which canpredict response and inform optimal therapeutic strategies, is asignificant step forward.

    Claudin-4 is a member of a large family of transmembraneproteins, of which there are 27 different subtypes (16). Clau-dins have been most studied for their canonical role in tightjunctions, where they mediate the specific paracellular barrierproperties of an epithelium. Although claudin-4 has beenshown to alter the permeability of epithelial monolayers incell culture (17–19), it is rarely localized to tight junctions inhuman and mouse tissue and is generally found along baso-lateral membranes and throughout the cytosol (20–22). Thefindings that claudin-4 knockout (KO) mice are phenotypicallynormal with only a subclinical increase in permeability to smallmolecules in lung epithelium, but have a reduced capacity forwound healing, further suggest a noncanonical role for claudin-4 (23). Furthermore, it has been well documented that claudin-4 is significantly upregulated in multiple epithelia-derived

    1Department of Obstetrics and Gynecology, Division of Reproductive Sciences,Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado.2Department of Obstetrics and Gynecology, Division of Gynecologic Oncology,Anschutz Medical Campus, University of Colorado Denver, Aurora, Colorado.

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

    Corresponding Author: Heidi K. Baumgartner, University of Colorado Denver,Mail Stop 8613, 12700 E. 19th Ave. Aurora, CO 80045, Phone: 303-724-3505:Fax: 303-724-3512; E-mail: [email protected]

    doi: 10.1158/1541-7786.MCR-18-0451

    �2019 American Association for Cancer Research.

    MolecularCancerResearch

    www.aacrjournals.org 741

    on June 2, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst January 3, 2019; DOI: 10.1158/1541-7786.MCR-18-0451

    http://crossmark.crossref.org/dialog/?doi=10.1158/1541-7786.MCR-18-0451&domain=pdf&date_stamp=2019-2-7http://mcr.aacrjournals.org/

  • cancer cells that lack traditional tight junction structures.High claudin-4 expression has, in fact, been associated witha phenotypically aggressive ovarian cancer cell [chemoresistant,highly mobile, and stem-like (24–26)]. The mechanism bywhich claudin-4 exerts these effects is currently unknown.

    To investigate the noncanonical role of claudin-4 in bothnormal and tumor cell biology, we previously designed a smallclaudin mimic peptide (CMP) that interferes with the DFYNPsequence in the second extracellular loop of claudin-4 that isthought to be important in claudin–claudin interactions (27).Wehave recently shown that disrupting claudin-4 activity with CMPincreased tumor cell response to the potent apoptotic inducer,staurosporine, and inhibited cell migration (28). In addition,treatment ofmice bearing EOCcell line–derived xenograft tumorswith CMP significantly reduced the tumor burden. These observa-tions suggest that claudin-4 expression in ovarian cancer cellsplays a functional role in tumor progression.

    To determine whether claudin-4 plays a role in the apoptoticresponse of ovarian tumors to standard chemotherapeutics, weexamined whether the presence of the protein altered the apo-ptotic response of EOC cell lines to cisplatin and paclitaxel in thepresence and absence of CMP. We found that claudin-4 expres-sion led to reduced tumor cell sensitivity to paclitaxel-inducedapoptosis. Tumor cell sensitivity to paclitaxel was restored bysilencing claudin-4 expression or co-treatment withCMP.We alsoshow claudin-4 involvement in cell-cycle progression and aninteraction of claudin-4 with microtubules, providing a possiblemechanism by which claudin-4 drives resistance to paclitaxel.

    Materials and MethodsCell culture

    Human-derived OVCAR3, OVCAR4, OVCAR5, OVCAR8,PEO4, OV429, and DOV13 ovarian tumor cells were obtainedfrom the laboratory of Monique A. Spillman (University ofColorado Denver, Aurora, CO) and cultured in RPMI1640 medi-um (Gibco, Thermo Fisher Scientific) plus 10% heat-inactivatedFBS (Access Biologicals) and 1% penicillin/streptomycin (Gibco,Thermo Fisher Scientific) at 37�C and 5% CO2. Cells were trypsi-nized (0.25% trypsin, EDTA,Mediatech) and plated 1:3 every 3–4days. All cell lines were authenticated at the beginning ofthis study by short tandem repeat profiling, as described previ-ously (29). Cells were passaged approximately 10 times beforethawing out a fresh vial of authenticated cells. Authenticationwas then repeated at the end of the study. Cells were plated at 1�104 cells/well into type I collagen-coated 8-well chamber slides(Lab-Tek, NUNC) for treatment and analysis. Upon 80%–90%confluence, cells were treated with either 400 mmol/L controlpeptide [(NH2-GDGYNPG-OH, D-amino acid conformation,University of Colorado Protein and Peptide ChemistryCore (27)], 400 mmol/L claudin mimic peptide (CMP, NH2-GDFYNPG-OH, D-amino acid conformation, University of Col-orado Protein and Peptide Core), 10 nmol/L paclitaxel (Sigma-Aldrich), and/or 10 mmol/L cisplatin (Sigma-Aldrich) for 24hours. To examine the apoptotic mechanism, cells were alsotreated with 50 mmol/L etoposide (Sigma-Aldrich).

    Ovarian tumor samplesDe-identified patient tumor samples were collected from the

    University of Colorado Gynecologic Tissue and Fluid Bankunder an Institutional Review Board–approved protocol

    (COMIRB 05-1081). Each tumor sample was flash frozen inliquid nitrogen and stored at�80�C until processed for protein.Frozen tissue was placed in lysis buffer [30 mmol/L Tris HClpH7.4, 150 mmol/L NaCl, 1% Triton X-100, 10% glycerol, 2mmol/L EDTA, 0.57 mmol/L phenylmethylsulfonylfluoride(PMSF), 1� cOmplete Protease Inhibitor Cocktail] and homog-enized using a Polytron tissue homogenizer (BrinkmannInstruments, Thermo Fisher Scientific). Lysates were then spunat 13,000 rpm for 10 minutes and the supernatant collected forprotein analysis.

    Western blot analysisTo determine levels of claudin-4 protein expression, tumor

    cells were scraped from culture plates in the presence of lysisbuffer (30 mmol/L Tris HCl pH 7.4, 150 mmol/L NaCl, 1%Triton X-100, 10% glycerol, 2 mmol/L EDTA, 0.57 mmol/LPMSF, 1� cOmplete Protease Inhibitor Cocktail), placed on ashaker for 10 minutes and spun at 13,000 rpm for 10 minutes.Supernatant was collected and 20 mg of total protein wasdenatured, resolved on 10% SDS-PAGE, and transferred to apolyvinylidene difluoride membrane (Bio-Rad). Membraneswere blocked with 5% nonfat dry milk in Tris-buffered salinewith 0.1% Tween-20 (TBST) for 1 hour at room temperaturebefore treatment with either rabbit anti-human claudin-4(1:500, Invitrogen), mouse anti-human claudin-4 (1:500; Invi-trogen), or rabbit antihuman GAPDH (1:10,000; Sigma) over-night at 4�C. Membranes were then washed four times withTBST for 15 minutes before treatment with horseradish perox-idase (HRP)-conjugated goat anti-rabbit (1:10,000; GE Health-care) or goat anti-mouse (1:10,000; Jackson ImmunoResearchLaboratories) antibodies for 1 hour at room temperature.Membranes were washed with TBST as described above andthen visualized using an ECL Prime Western Blotting DetectionReagent (GE Healthcare) and X-ray film (CL-XPosure Film,Thermo Fisher Scientific).

    Caspase activation assayApoptosis was measured by determining the number of cells

    exhibiting activated caspase-3. After treatment, cells werewashed with PBS (Gibco, Thermo Fisher Scientific) and fixedwith 10% phosphate-buffered formalin (Thermo Fisher Scien-tific) at room temperature for 15 minutes. Cells were washedtwice with PBS before cell membrane permeablization with0.5% Triton X-100 (IBI Scientific) for 5 minutes and washedagain with PBS. Cells were treated with blocking buffer (2%BSA; Sigma-Aldrich) for 1 hour before application of primaryantibody directed to cleaved caspase-3 (1:400; rabbit anti-human cleaved caspase-3, Cell Signaling Technology) over-night at 4�C. Cells were then washed with PBS five times beforeapplication of secondary antibody conjugated to CY3 (1:100;donkey anti-rabbit, Jackson ImmunoResearch Laboratories)and 5 mg/mL 40,6-diamidino-2-phenylindole (DAPI; Sigma)for 45 minutes at room temperature followed by washing withPBS five times. After removal of PBS, o-phenylenediaminedihydrochloride (20 mg/mL; OPDA) in 1 mol/L Tris (pH 8.5)was added to the slides to preserve fluorescence and cover-slipped. The Olympus FV1000/RICS confocal microscope(University of Colorado AMC Light Microscopy Core) was usedto image fluorescence. The percent of cells positive for activecaspase was determined using SlideBook software (IntelligentImaging Innovations, Inc.).

    Breed et al.

    Mol Cancer Res; 17(3) March 2019 Molecular Cancer Research742

    on June 2, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst January 3, 2019; DOI: 10.1158/1541-7786.MCR-18-0451

    http://mcr.aacrjournals.org/

  • shRNA knockdownOVCAR3 and PEO4 cells were plated 3.2 � 104 in a 96-well

    plate and incubated at 37�C for 24hours.When cells reached 70%confluence, 10 mL of claudin-4 short hairpin RNA (shRNA; TRC#:TRCN0000116627, TRCN0000116628, or TRCN0000116631)or control shRNA (SHC001, pLK0.1-puro EmptyVector) lentiviralsuspension (Sigma-Aldrich MISSION shRNA, University ofColorado Functional Genomics Facility) was added to the cellsand incubated overnight at 37�C. Fresh medium was added toremove lentivirus and cells were allowed to recover for 24 hoursbefore being treated with 0.5 mg/mL puromycin for selection andexpansion of transduced cells. Western blot analysis (as describedabove) confirmed loss of claudin-4 expression.

    Overexpression of claudin-4GFP-tagged claudin-4 and aGFP-only control (pLenti-C-mGFP

    vector; OriGene) were transduced into OVCAR8 and OVCAR4cells via lentiviral particles in the presence of 10 mg/mL hexadi-methrine bromide. GFP-positive cells were flow-sorted andexpanded.

    Mitotic assaysTo examine the number of cells in mitosis, we examined the

    images of DAPI fluorescence in OVCAR3 cells cultured in 8-wellchamber slides. Cells that had clearly condensed chromatin thataligned along a single plane were counted as mitotic cells andcalculated as the percentage of total number of nuclei per field ofview (at least 12 fields of view were analyzed for each condition).In addition, a Mitotic Assay Kit (Active Motif) was used as permanufacturer's instructions. Briefly, cells were cultured in 96-wellplates, fixed, and treated with a phospho-histone H3 (Ser 28)monoclonal primary antibody followed by an HRP-conjugatedsecondary antibody. Absorbance at 450 nm was read after 20-minute incubation with the Developing Solution. Crystal violetwas used to normalize for total cell number.

    Proliferation assayA total of 10,000 cells were plated in each well of 12-well

    culture plates. Cells were trypsinized (0.25% trypsin, EDTA) and

    counted by aCountess AutomatedCell Counter (Invitrogen) after24, 48, and 72 hours in culture.

    Cell-cycle analysisCells were plated in 100-mm culture dishes at 5 � 105 cells

    per dish. After 24 hours in culture, media were changed toserum-free RPMI1640 for 48 hours. Cells were then exposed tomedia containing serum and harvested 6 hours later. Cells wereplaced in Krishan stain (0.224 g sodium citrate 2H2O, 9.22 mgpropidium iodide, 2.0 mL 1% NP40 in H2O, 2.0 mL 1 mg/mLRNase in 200 mL H2O) overnight at 4�C before being sent tothe University of Colorado Flow Cytometry Core Facility,Aurora, CO). Cells were analyzed by flow cytometry (BeckmanCoulter Gallios, University of Colorado Flow Cytometry CoreFacility, Aurora, CO), measuring fluorescence emission at >575nm (FL3). FlowJo software was used to determine number ofcells in G2–M phase.

    Proximity ligation assayClaudin-4 interaction with tubulin was examined using the

    DuoLink Proximity Ligation Assay Kit (Sigma), followingthe manufacturer's protocol. Briefly, cells were plated in 8-wellchamber slides and cultured to 70% confluence before beingfixed, permeablized, and blocked as described above forcapase-3 immunofluorescence. Cells were then treated with anti-bodies directed to claudin-4 (mouse anti-human claudin-4;1:200; Invitrogen) and a-tubulin (rabbit anti-human a-tubulin;1:100; Abcam) or b-tubulin (rabbit anti-human b-tubulin; 1:100;Abcam) overnight at 4�C. After washing with PBS, cells weretreated with PLA probes (1:5; anti-mouse PLUS, anti-rabbitMINUS) for 1 hour at 37�C in a preheated humidity chamber.After washing, Ligation Solution was added for 30 minutes at37�C followed by washing and addition of amplification poly-merase solution for 100 minutes at 37�C in a humidity chamber.Slides were washed and dried before adding mounting mediumand placing coverslip. Fluorescence was imaged using an Olym-pus FV1000/RICS confocal microscope (University of ColoradoAMC Light Microscopy Core).

    A

    HOSE HGSOC10

    100

    1,000

    10,000

    n = 10 n = 53

    ***

    CLDN4

    Expr

    essi

    on

    C

    Claudin-4

    GAPDH

    B

    Fallopiantuben = 4

    HGSOCn = 10

    OtherEOCn = 7

    0.5

    1.0

    1.5

    2.0

    2.5

    Den

    sito

    met

    ry (A

    .U.)

    Cla

    udin

    - 4 p

    rote

    in

    *

    (Primary tissue)

    Figure 1.

    Claudin-4 expression in HGSOCcells. A, CLDN4 gene expressionlevels in laser capturemicrodissected specimens ofhuman high-grade serous ovariantumors compared with isolatednormal human ovarian surfaceepithelial cells (36). Western blotanalysis of claudin-4 protein levelsin human patient samples of normalfallopian tube and ovarian cancer(B) as well as high-grade serousovarian tumor cell lines (C), usingGAPDH as a loading control. Mean� SEM, n¼ 10 HOSE, n¼ 10–53HGSOC tumor specimens, n¼ 7other EOC subtypes (� , P < 0.05;��� , P < 0.0001).

    Claudin-4 Reduces Response to Paclitaxel

    www.aacrjournals.org Mol Cancer Res; 17(3) March 2019 743

    on June 2, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst January 3, 2019; DOI: 10.1158/1541-7786.MCR-18-0451

    http://mcr.aacrjournals.org/

  • ImmunoprecipitationClaudin-4 (and interacting proteins) was pulled from cell

    lysates with an antibody directed to claudin-4 (mouse anti-human claudin-4, Invitrogen) bound to Protein A/G Dyna-beads Magnetic beads, using the Pierce Crosslink Immunopre-cipitation (IP) Kit (Thermo Fisher Scientific). Beads were incu-bated with lysate (�500 mg protein) for 1 hour at roomtemperature and then collected using a Magnetic SeparationRack (New England BioLabs). Proteins were eluted from beadsper manufacturer's protocol. Eluted proteins were run on 10%SDS-PAGE and Western blot analysis was performed asdescribed above, using antibodies directed to claudin-4 (rabbitanti-human claudin-4; 1:500) and tubulin (rabbit antihumana-tubulin or b-tubulin; 1:1,000).

    Visualization of claudin-4 and microtubulesImmunofluorescence was performed, as described above, for

    caspase-3 activation as well as to visualize claudin-4 and tubulinin tumor cells. Primary antibodies used in these assays were the

    same (including dilutions) as those described above for theproximity ligation assays.

    Tubulin polymerization assayThe Microtubules/Tubulin In Vivo Assay Kit (Cytoskeleton

    Inc.) was used to determine changes in tubulin polymerizationwith changes in claudin-4 expression. Cells were grown to 80%confluence before being washed with PBS and scraped from100-cm2 plates in tubulin stabilization buffer provided by themanufacturer of the kit and homogenized by pipetting. Cellsuspension was centrifuged at 1,000 � g (37�C) for 5 minutes.Cell pellets and supernatant were collected separately andresolved on 10% SDS-PAGE gel. Primary tubulin antibody andHRP-conjugated secondary antibody provided by the manu-facturer of the kit were used to determine polymerized (pelletfraction) and nonpolymerized (supernatant fraction) tubulinlevels. Densitometry, using ImageJ, was performed to quantifychanges in levels of polymerized to nonpolymerized tubulin ineach cell line.

    Figure 2.

    Disruption of claudin-4 activity with CMP enhances paclitaxel response. A, Representative images of nuclei (DAPI staining) and activated caspase-3 (fluorescentantibody directed to cleaved caspase-3) from OVCAR3 cells. Quantification of caspase-3 activation in OVCAR3 (B) and OVCAR8 (C) cells treated for 24 hourswith vehicle control (Veh cont), 400 mmol/L inactive control peptide (ContP), 400 mmol/L CMP, 10 mmol/L Cisplatin (Cis), CMPþ Cisplatin, 10 nmol/L paclitaxel(taxol), or CMPþ paclitaxel. Percent of cell population positive for caspase-3 activation was calculated using SlideBook software (3i). Mean� SEM; n¼ 3; NS, notsignificant; ��� , P < 0.001 versus control/drug only.

    Breed et al.

    Mol Cancer Res; 17(3) March 2019 Molecular Cancer Research744

    on June 2, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst January 3, 2019; DOI: 10.1158/1541-7786.MCR-18-0451

    http://mcr.aacrjournals.org/

  • Statistical analysisData are presented as mean � SEM. An unpaired Student t test

    was used for statistical comparison between control and treat-ment groups. A one-way ANOVA was used to determine varianceamong multiple groups, with a Bonferroni Multiple Comparisonposttest to determine significance between individual groups. A Pvalue of

  • paclitaxel with CMP cotreatment. As a secondary assay ofapoptotic response, we used flow cytometry to measure num-ber of OVCAR3 cells exhibiting Annexin V binding and con-firmed enhanced apoptosis (Annexin V binding) in response topaclitaxel when cotreated with CMP (Supplementary Fig. S3).

    Loss of claudin-4 expression improves tumor cell apoptoticresponse to paclitaxel

    Using a small hairpin RNA (shRNA) specific to claudin-4,we knocked down claudin-4 expression in OVCAR3 cells("shCLDN4_2" and shCLDN4_3; Fig. 3A). A control, emptyvector, shRNA, and shCLDN4_1 did not impact expression ofthe claudin-4 protein ("shCTRL" and "shCLDN4_1", respective-ly). Similar to parental OVCAR3 cells, shCTRL and shCLDN4_1cells showed increased apoptosis with CMP or paclitaxel aloneand a significantly enhanced apoptotic response to paclitaxel inthe presence of CMP (P < 0.0001; Fig. 3B). CMP did not induce anapoptotic response in OVCAR3 shCLDN4_2 and shCLDN4_3cells (cells that successfully lost claudin-4 expression) and CMPcotreatment did not enhance paclitaxel-induced apoptosis (P ¼0.8890 and 0.8851, respectively, for shCLDN4_2; P¼ 0.5024 and0.7734, respectively, for shCLDN4_3). However, the response topaclitaxel alone was significantly (P < 0.0001) higher in theshCLDN4_2 and shCLDN4_3 cells compared with shCTRL andshCLDN4_1 cells, indicating that disrupting claudin-4 augmentsthe apoptotic response to paclitaxel. Similar results were seen inthe claudin-4–expressing PEO4 cell line (Supplementary Figs. S4and S5).

    Overexpression of claudin-4 reduces tumor cell response topaclitaxel

    We next evaluated the impact of overexpressing claudin-4 inOVCAR8 andOVCAR4 cells, which endogenously express no/lowclaudin-4 protein (Fig. 1).We transfected both cell lineswithGFP-tagged claudin-4 or a control GFP vector. Western blot analysisshowed strong expression of the claudin-4-GFP (Fig. 4A). GFPcontrol cells didnot respond toCMP treatment; apoptosiswas notinduced with CMP and paclitaxel apoptotic response was notenhanced with CMP cotreatment. However, cells with claudin-4-GFP overexpression had a decreased apoptotic response to pac-litaxel and were responsive to CMP, with induction of apoptosiswith CMP alone and enhanced apoptotic response to paclitaxelin the presence of CMP (Fig. 4B, representative images of nucleiand activated caspase-3 in claudin-4 nonexpressing control andclaudin-4 overexpressing tumor cells are shown in SupplementaryFig. S6). Similar responses were seen in OVCAR4 cells (Supple-mentary Fig. S7).

    Claudin-4 expression results in increased mitotic fidelityWe have shown in the experiments above that claudin-4

    diminishes the apoptotic response to paclitaxel. We nextwanted to determine a potential mechanism by which clau-din-4 could affect cell biology to influence paclitaxel response.Upon examination of nuclei in the OVCAR3 cell line, weobserved significantly more mitotic figures in shCLDN4_2 cellscompared with the claudin-4–expressing shCTRL cells (P <0.0001; Fig. 5A and B). To confirm this observation, we useda colorimetric mitotic assay kit that utilizes a phospho-histoneH3 (Ser28) mAb with HRP-conjugated secondary antibody todetect the level of mitosis. The loss of claudin-4 expressionsignificantly increased the number of cells in the M-phase of

    mitosis (Fig. 5C, P < 0.0001). To determine whether thisincreased mitotic index resulted in an increase in cell prolifer-ation, we measured cell number over time in the shCTRL andshCLDN4_2 cell lines. We found that the claudin-4 KO cellsproliferated significantly more slowly than the control cells(P ¼ 0.0453, Fig. 5D). Cell-cycle analysis, using flow cytometry(Fig. 5E and F), confirmed significantly (P < 0.0001) moretumor cells in the G2–M phase at 6 hours postreleasefrom serum-free conditions in cells that did not express clau-din-4 (shCLDN4_2) compared with the cells that did expressclaudin-4 (shCTRL). These findings suggest that the increase inmitotic figures in the cells lacking claudin-4 expression is notthe result of increased proliferation, but reflects an inhibition ofG2–M progression.

    contG

    FP C

    ontP

    contG

    FP C

    MP

    contG

    FP Ta

    xol

    contG

    FP Ta

    xol+C

    MP

    cld4G

    FP C

    ontP

    cld4G

    FP C

    MP

    cld4G

    FP Ta

    xol

    cld4G

    FP Ta

    xol+C

    MP0

    10

    20

    30

    40

    50

    % P

    opul

    atio

    n ca

    sp-3

    pos

    itive

    B

    A

    NS

    NS

    ***

    ***

    **

    Claudin-4-GFP

    Claudin-4

    GPDH

    Figure 4.

    Overexpression of claudin-4 reduces paclitaxel response. A,Western blotanalysis of claudin-4 protein in OVCAR8 cells transduced with GFP only orclaudin-4-GFP. GAPDHwas used as a loading control. B,Immunofluorescence analysis of fixed OVCAR8 cells that were treated with400 mmol/L inactive control peptide (ContP), 400 mmol/L CMP, 10 nmol/Lpaclitaxel (taxol), or CMPþ paclitaxel for 24 hours. Cells were treated withfluorescent antibody directed to cleaved caspase-3 and DAPI (nuclei) andpercent of cell population positive for caspase-3 was calculated usingSlideBook software (3i). Mean� SEM; n¼ 3 per treatment group; NS, notsignificant; �� , P < 0.01; ��� , P < 0.001 versus control/paclitaxel only.

    Breed et al.

    Mol Cancer Res; 17(3) March 2019 Molecular Cancer Research746

    on June 2, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst January 3, 2019; DOI: 10.1158/1541-7786.MCR-18-0451

    http://mcr.aacrjournals.org/

  • Microtubules are known to play an important role in bothpaclitaxel response and cell-cycle progression. Therefore, weexamined whether claudin-4 interacted with microtubules.Proximity ligation assays were performed on OVCAR3 cellswith antibodies directed to claudin-4 and either a-tubulin orb-tubulin. Claudin-4 was found to be within interacting dis-tance of both a-tubulin and b-tubulin (Fig. 6A). IP of claudin-4confirmed the interaction with a-tubulin (Fig. 6B), but wewere unable to confirm the interaction with b-tubulin by thismethod. However, immunofluorescence assays revealed thatclaudin-4 was colocalized with b-tubulin at the mitotic spindlein tumor cells undergoing mitosis (Fig. 6C). In addition,images of microtubules in claudin-4–expressing (shCTRL cells)and nonexpressing (shCLDN4_2) OVCAR3 cells revealed sig-nificant changes in microtubule structure when claudin-4expression was present (Fig. 6D).

    Claudin-4 expression alters polymerization of tubulinTo identify a potential role for claudin-4 in altering micro-

    tubule stability, we examined the levels of tubulin withinmicrotubules (polymerized tubulin) compared with free tubu-lin (nonpolymerized tubulin) in OVCAR3 cells expressing andnot expressing claudin-4. Figure 7A shows a shift betweenpolymerized (pellet: p) and nonpolymerized (supernatant: s)tubulin levels when claudin-4 expression is silenced. Densi-tometry analysis revealed that there was a significant (P ¼0.0054) decrease in the percentage of total tubulin foundwithin microtubules (polymerized) in the claudin-4 KO

    (shCLDN4_2) compared with claudin-4 expressing control(shCTRL) cells (Fig. 7B), suggesting that claudin-4 is enhancingthe polymerization of microtubules.

    DiscussionIn this study, we have shown that claudin-4 expression reduces

    the apoptotic response of ovarian tumor cells to paclitaxel.Disruption of claudin-4 expression (shRNA knockdown) or activ-ity (CMP treatment) can enhance or restore the paclitaxel apo-ptotic response through activation of apoptosis. In addition, wedemonstrated that loss of claudin-4 delayed progression throughthe cell cycle and reduced proliferation. The effect on bothapoptosis and the proliferative response in tumor cells may bedue the interaction of claudin-4 withmicrotubules and alterationof tubulin polymerization.

    Previously, high claudin-4 expression was shown to be asso-ciated with ovarian tumor cells that are resistant to chemother-apeutic agents (24, 25, 31). For example, CD44þ ovarian tumorcells were shown to have significantly higher expression levels ofclaudin-4 compared with CD44� populations isolated from thesame tumor. The CD44þ/high claudin-4–expressing cells weremore resistant to carboplatin and paclitaxel treatment comparedwith the low/no expressing cells (25). In addition, Gao andcolleagues have demonstrated that inhibiting claudin-3 and -4expression with a fragment of the Clostridium perfringens entero-toxin increased the ovarian tumor cell response to both paclitaxeland carboplatin (31). Although our data shows that cells

    B***

    shCTRL shCLDN4_2

    12345

    % P

    opul

    atio

    n m

    itotic

    A shCTRL shCLDN4_2

    20 μm

    F

    shCTRL shCLDN4_2

    10

    20

    30

    40

    50

    % P

    opul

    atio

    n in

    G2/

    M

    ***

    E

    Propidium iodide

    Cel

    l cou

    nt

    shCLDN4_2shCTRL

    500

    400

    300

    200

    100

    0104 105

    C

    OD

    450.

    ***

    5

    10

    15

    shCTRL shCLDN4_2

    *

    20 40 60 80

    20,00040,00060,00080,000

    100,000120,000

    Time (hr)

    shCTRLshCLDN4_2

    D

    Cel

    l num

    ber

    Figure 5.

    Loss of claudin-4 leads to delayed mitotic progression. A, Representative images of DAPI (DNA) staining in fixed monolayers of control knockdown(shCTRL) and claudin-4 knockdown (shCLDN4_2) OVCAR3 cells. Yellow circles highlight mitotic figures. B, Visual counting of mitotic figures fromDAPI images. C, Colorimetric measurement of phosphorylated H2B (mitotic marker) to quantify population of mitotic cells in shCTRL (white bars) andshCLDN4_2 (black bars) OVCAR3 cells. D, Proliferation rates of shCTRL (circles/solid line) and shCLDN4_2 (triangles/dotted line) OVCAR3 cells.E, Representative histograms of propidium iodide staining 6 hours after release from serum starvation synchronization in shCTRL and shCLDN4_2cells. F, Quantification of the percent of the total cell population in the G2–M phase of the cell cycle using FlowJo software. Mean � SEM; n ¼ 3(� , P < 0.05; ��� , P < 0.001 versus control/paclitaxel only).

    Claudin-4 Reduces Response to Paclitaxel

    www.aacrjournals.org Mol Cancer Res; 17(3) March 2019 747

    on June 2, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst January 3, 2019; DOI: 10.1158/1541-7786.MCR-18-0451

    http://mcr.aacrjournals.org/

  • expressing high levels of claudin-4 (OVCAR3) are more resistantto cisplatin compared with the cells that do not express claudin-4(OVCAR8), CMP was unable to enhance the apoptotic responseto cisplatin in the claudin-4–expressing tumor cells. These resultssuggest the possibility that claudin-4 may regulate the apoptoticresponse to cisplatin through a different mechanism than pacli-taxel, potentially through an interaction of claudin-4 that doesnot involve the second extracellular loop (which is targeted byCMP).

    Previous studies investigating a role for claudin-4 in prolifer-ation have not been consistent. Agarwal and colleagues demon-strated that overexpressing claudin-4 in normal HOSE cellsenhanced survival, but did not alter proliferation (32). On theother hand,Ma and colleagues demonstrated that loss of claudin-4 expression inhibits proliferation in MCF7 breast cancer cellsin vitro and in vivo (33). The discrepancies could be due todifferences in the regulation (or loss of regulation) of claudin-4in normal versus tumor cells. In addition, our results demonstratethat tumor cells still proliferate in the absence of claudin-4. Thissuggests that claudin-4 is not required for proliferation but doesenhance proliferationwhen it is present. The observation that loss

    of claudin-4 leads to increased number of cells inmitosis indicatesthat claudin-4 likely enhances proliferation by facilitating pro-gression through mitosis.

    A proteomic analysis of proteins within interacting distanceof claudin-4 in MDCK cells was performed by Fredricksson andcolleagues and b-tubulin was identified as a potential interact-ing partner of claudin-4 (34). Our results confirm an interac-tion of claudin-4 with both a-tubulin and b-tubulin in ovariantumor cells and that this interaction changes during the cellcycle. We were able to capture an interaction of claudin-4 atsites at or near the plasma membrane as well as within themitotic spindle. Further analysis of this interaction will beneeded to fully understand how the interaction of claudin-4with microtubules may be regulating tumor cell survival andgrowth. Our results do suggest that interaction of claudin-4with tubulin may alter the polymerization of tubulin. McGrailand colleagues have shown that ovarian tumor cells that areresistant to paclitaxel-induced death have altered microtubulestability (35). Therefore, it is possible that claudin-4 altersmicrotubule dynamics in a way that makes tumor cells moreresistant to paclitaxel-induced death.

    Whole

    Claudin-4 / α-tubulin

    Claudin-4 / β-tubulin a-Tubulin

    Claudin-4

    A B

    co-localization

    Claudin-4β tubulinDAPI

    cld4 β tubulin DAPIC

    20 µm

    20 µm

    D shCTRL (+ claudin-4)Tubulin DAPI

    Tubulin

    10 μm

    shCLDN4_2 (- claudin-4)Tubulin DAPI

    Tubulin

    10 μm

    Figure 6.

    Claudin-4 interacts with tubulin. A, Proximity ligation assay using antibodies directed at claudin-4 and a-tubulin or b-tubulin. Red fluorescence indicatesprotein–protein interaction. Yellow outlined boxes are higher magnification of cell from image. Arrows point to sites of protein-protein interaction. B, IP ofclaudin-4 (cld-4 IP) and IgG (mouse IgG IP) from OVCAR3 lysates blotted for presence of a-tubulin and claudin-4. C, Immunofluorescence of claudin-4 (green)and b-tubulin (red), with DAPI (blue), in mitotic OVCAR3 cell. Yellow color indicates colocalization of claudin-4 and b-tubulin. D, Immunofluorescence ofmicrotubules (b-tubulin; white/red) and nuclei (DAPI/blue) in cells expressing (shCTRL) and not expressing (shCLDN4_2) claudin-4.

    Breed et al.

    Mol Cancer Res; 17(3) March 2019 Molecular Cancer Research748

    on June 2, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst January 3, 2019; DOI: 10.1158/1541-7786.MCR-18-0451

    http://mcr.aacrjournals.org/

  • In conclusion, we have demonstrated that claudin-4 plays animportant role in tumor cell resistance to paclitaxel. Therefore,expression of claudin-4 by tumor cells may be a biomarker forpatient response to paclitaxel. In addition, the development of aclaudin-4 therapeutic (e.g., CMP) could provide a novel way toimprove the patient response to paclitaxel.

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

    Authors' ContributionsConception and design:C.A. Breed, B.G. Bitler, K. Behbakht, H.K. BaumgartnerDevelopment ofmethodology:C.A. Breed, D.A. Hicks, B.G. Bitler, K. Behbakht,H.K. BaumgartnerAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): C.A. Breed, D.A. Hicks, C.E. Galimanis, B.G. Bitler,K. Behbakht, H.K. BaumgartnerAnalysis and interpretation of data (e.g., statistical analysis,biostatistics, computational analysis): C.A. Breed, D.A. Hicks, K. Behbakht,H.K. BaumgartnerWriting, review, and/or revision of the manuscript: C.A. Breed, B.G. Bitler,K. Behbakht, H.K. BaumgartnerAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): P.G. Webb, C.E. Galimanis, K. Behbakht,H.K. BaumgartnerStudy supervision: K. Behbakht, H.K. Baumgartner

    AcknowledgmentsThe authors wish to acknowledge Dziuleta Cepeniene of the Peptide and

    Protein Chemistry Core at the University of Colorado for synthesizing CMP.Wewould also like to acknowledge that imaging was made possible by theUniversity of Colorado Advanced Light Microscopy Core, supported in partby NIH/NCRR CCTSI grant UL1 RR025780. Flow cytometry was performed bythe University of Colorado Cancer Center Flow Cytometry Shared Resource(supported by NCI Cancer Center Support Grant P30CA046934). Funding wasalso provided by the Department of Obstetrics and Gynecology, University ofColorado Denver.

    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 May 3, 2018; revised October 11, 2018; accepted December 20,2018; published first January 3, 2019.

    References1. Colombo N, Peiretti M, Parma G, Lapresa M, Mancari R, Carinelli S, et al.

    Newly diagnosed and relapsed epithelial ovarian carcinoma: ESMOClinical Practice Guidelines for diagnosis, treatment and follow-up.Ann Oncol 2010;21:v23–30.

    2. Agarwal R, Kaye SB. Ovarian cancer: strategies for overcoming resistance tochemotherapy. Nat Rev Cancer 2003;3:502–16.

    3. Muggia FM, Jeffers S, Muderspach L, Roman L, Rosales R,Groshen S, et al. Phase I/II study of intraperitoneal floxuridineand platinums (cisplatin and/or carboplatin). Gynecol Oncol1997;66:290–4.

    4. Kavallaris M, Leary JA, Barrett JA, Friedlander ML. MDR1 and multidrugresistance-associated protein (MRP) gene expression in epithelial ovariantumors. Cancer Lett 1996;102:7–16.

    5. Zajchowski DA, Karlan BY, Shawver LK. Treatment-related protein bio-marker expression differs between primary and recurrent ovarian carcino-mas. Mol Cancer Ther 2012;11:492–502.

    6. Svoboda M, Wlcek K, Taferner B, Hering S, Stieger B, Tong D, et al.Expression of organic anion-transporting polypeptides 1B1 and1B3 in ovarian cancer cells: relevance for paclitaxel transport.Biomed Pharmacother 2011;65:417–26.

    7. Zeller C, Dai W, Steele NL, Siddiq A, Walley AJ, Wilhelm-Benartzi CS, et al.Candidate DNA methylation drivers of acquired cisplatin resistance inovarian cancer identified by methylome and expression profiling.Oncogene 2012;31:4567–76.

    8. Ibrahim N, He L, Leong CO, Xing D, Karlan BY, Swisher EM, et al.BRCA1-associated epigenetic regulation of p73 mediates an effector

    pathway for chemosensitivity in ovarian carcinoma. Cancer Res 2010;70:7155–65.

    9. Yang H, Kong W, He L, Zhao JJ, O'Donnell JD, Wang J, et al. MicroRNAexpression profiling in human ovarian cancer: miR-214 induces cellsurvival and cisplatin resistance by targeting PTEN. Cancer Res 2008;68:425–33.

    10. YeG, FuG,Cui S, Zhao S, Bernaudo S, Bai Y, et al.MicroRNA376c enhancesovarian cancer cell survival by targeting activin receptor-like kinase 7:implications for chemoresistance. J Cell Sci 2011;124:359–68.

    11. O'Connor PM, Jackman J, Bae I, Myers TG, Fan S, Mutoh M, et al.Characterization of the p53 tumor suppressor pathway in cell lines of theNational Cancer Institute anticancer drug screen and correlations with thegrowth-inhibitory potency of 123 anticancer agents. Cancer Res 1997;57:4285–300.

    12. WongKK, Izaguirre DI, Kwan SY, King ER,DeaversMT, SoodAK, et al. Poorsurvival with wild-type TP53 ovarian cancer? Gynecol Oncol 2013;130:565–9.

    13. Yan X, Fraser M, Qiu Q, Tsang BK. Over-expression of PTEN sensitizeshuman ovarian cancer cells to cisplatin-induced apoptosis in a p53-dependent manner. Gynecol Oncol 2006;102:348–55.

    14. Ying H, Qu D, Liu C, Ying T, Lv J, Jin S, et al. Chemoresistance is associatedwith Beclin-1 andPTENexpression in epithelial ovarian cancers.Oncol Lett2015;9:1759–63.

    15. Dumontet C, Sikic BI. Mechanisms of action of and resistance to anti-tubulin agents: microtubule dynamics, drug transport, and cell death.J Clin Oncol 1999;17:1061–70.

    Figure 7.

    Claudin-4 alters tubulin polymerization. A, RepresentativeWestern blotanalysis of polymerized (P, pellet fraction) and nonpolymerized (S,supernatant fraction) tubulin levels in OVCAR3 cells expressing claudin-4(shCTRL) and cells with silenced claudin-4 expression (shCLDN4_2). B,Quantification of the percent of the total tubulin that is polymerized. Mean�SEM; n¼ 4 (�� , P < 0.01).

    www.aacrjournals.org Mol Cancer Res; 17(3) March 2019 749

    Claudin-4 Reduces Response to Paclitaxel

    on June 2, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst January 3, 2019; DOI: 10.1158/1541-7786.MCR-18-0451

    http://mcr.aacrjournals.org/

  • 16. Mineta K, Yamamoto Y, Yamazaki Y, Tanaka H, Tada Y, Saito K, et al.Predicted expansion of the claudin multigene family. FEBS Lett 2011;585:606–12.

    17. Van Itallie C, Rahner C, Anderson JM. Regulated expression of claudin-4decreases paracellular conductance through a selective decrease in sodiumpermeability. J Clinical Invest 2001;107:1319–27.

    18. Hou J, Gomes AS, Paul DL, Goodenough DA. Study of claudin function byRNA interference. J Biol Chemistry 2006;281:36117–23.

    19. Hou J, Renigunta A, Yang J, Waldegger S. Claudin-4 forms paracellularchloride channel in the kidney and requires claudin-8 for tight junctionlocalization. Proc Natl Acad Sci U S A 2010;107:18010–15.

    20. Rahner C, Mitic LL, Anderson JM. Heterogeneity in expression and sub-cellular localization of claudins 2, 3, 4, and 5 in the rat liver, pancreas, andgut. Gastroenterology 2001;120:411–22.

    21. Jakab C, Halasz J, Szasz AM, Batmunkh E, Kiss A, Schaff Z, et al. Expressionand localisationof claudin-1, -2, -3, -4, -5, -7 and -10proteins in the normalcanine mammary gland. Acta Vet Hung 2008;56:341–52.

    22. Garcia-Godinez A, Contreras RG, Gonzalez-Del-Pliego M, Aguirre-BenitezE, Acuna-Macias I, de la Vega MT, et al. Anterior and intermediate pituitarytissues express claudin 4 in follicle stellate cells and claudins 2 and 5 inendothelial cells. Cell Tissue Res 2014;357:309–21.

    23. Kage H, Flodby P, GaoD, Kim YH,Marconett CN, DeMaio L, et al. Claudin4 knockout mice: normal physiological phenotype with increased suscep-tibility to lung injury. Am J Physiol Lung Cell Mol Physiol 2014;307:L524–36.

    24. Stewart JJ, White JT, Yan X, Collins S, Drescher CW, Urban ND, et al.Proteins associated with Cisplatin resistance in ovarian cancer cells iden-tified by quantitative proteomic technology and integrated with mRNAexpression levels. Mol Cell Proteomics 2006;5:433–43.

    25. Casagrande F,Cocco E, Bellone S, Richter CE, BelloneM, Todeschini P, et al.Eradicationof chemotherapy-resistant CD44þhumanovarian cancer stemcells in mice by intraperitoneal administration of clostridium perfringensenterotoxin. Cancer 2011;117:5519–28.

    26. Yin G, Alvero AB, Craveiro V, Holmberg JC, Fu HH, Montagna MK, et al.Constitutive proteasomal degradation of TWIST-1 in epithelial-ovariancancer stem cells impacts differentiation and metastatic potential.Oncogene 2013;32:39–49.

    27. Baumgartner HK, Beeman N, Hodges RS, Neville MC. A d-Peptideanalog of the second extracellular loop of claudin-3 and -4 leads tomisLocalized claudin and cellular apoptosis in mammary epithelialcells. Chem Biol Drug Des 2011;77:124–36.

    28. HicksDA,Galimanis CE,Webb PG, SpillmanMA, Behbakht K,NevilleMC,et al. Claudin-4 activity in ovarian tumor cell apoptosis resistance andmigration. BMC Cancer 2016;16:788.

    29. Korch C, Spillman MA, Jackson TA, Jacobsen BM, Murphy SK, Lessey BA,et al. DNA profiling analysis of endometrial and ovarian cell lines revealsmisidentification, redundancy and contamination. Gynecol Oncol 2012;127:241–8.

    30. Domcke S, Sinha R, LevineDA, Sander C, Schultz N. Evaluating cell lines astumourmodels by comparison of genomic profiles. Nat Commun 2013;4:2126.

    31. GaoZ, XuX,McClane B, ZengQ, Litkouhi B,WelchWR, et al. C Terminus ofClostridium perfringens enterotoxin downregulates CLDN4 and sensitizesovarian cancer cells to Taxol and Carboplatin. Clin Cancer Res 2011;17:1065–74.

    32. Agarwal R, D'Souza T, Morin PJ. Claudin-3 and claudin-4 expression inovarian epithelial cells enhances invasion and is associated withincreased matrix metalloproteinase-2 activity. Cancer Res 2005;65:7378–85.

    33. Ma X,MiaoH, Jing B, PanQ, ZhangH, Chen Y, et al. Claudin-4 controls theproliferation, apoptosis, migration and in vivo growth of MCF-7 breastcancer cells. Oncol Rep 2015;34:681–90.

    34. Fredriksson K, Van Itallie CM, Aponte A, Gucek M, Tietgens AJ, AndersonJM. Proteomic analysis of proteins surrounding occludin and claudin-4reveals their proximity to signaling and trafficking networks. PLoS One2015;10:e0117074.

    35. McGrail DJ, Khambhati NN, Qi MX, Patel KS, Ravikumar N, BrandenburgCP, et al. Alterations in ovarian cancer cell adhesion drive taxol resistanceby increasing microtubule dynamics in a FAK-dependent manner. Sci Rep2015;5:9529.

    36. Mok SC, Bonome T, Vathipadiekal V, Bell A, JohnsonME,WongKK, et al. Agene signature predictive for outcome in advanced ovarian cancer identifiesa survival factor: microfibril-associated glycoprotein 2. Cancer Cell 2009;16:521–32.

    Mol Cancer Res; 17(3) March 2019 Molecular Cancer Research750

    Breed et al.

    on June 2, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst January 3, 2019; DOI: 10.1158/1541-7786.MCR-18-0451

    http://mcr.aacrjournals.org/

  • 2019;17:741-750. Published OnlineFirst January 3, 2019.Mol Cancer Res Christopher Breed, Douglas A. Hicks, Patricia G. Webb, et al. Response to PaclitaxelOvarian Tumor Cell Expression of Claudin-4 Reduces Apoptotic

    Updated version

    10.1158/1541-7786.MCR-18-0451doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://mcr.aacrjournals.org/content/suppl/2019/01/03/1541-7786.MCR-18-0451.DC1

    Access the most recent supplemental material at:

    Cited articles

    http://mcr.aacrjournals.org/content/17/3/741.full#ref-list-1

    This article cites 36 articles, 11 of which you can access for free at:

    Citing articles

    http://mcr.aacrjournals.org/content/17/3/741.full#related-urls

    This article has been cited by 1 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://mcr.aacrjournals.org/content/17/3/741To request permission to re-use all or part of this article, use this link

    on June 2, 2021. © 2019 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

    Published OnlineFirst January 3, 2019; DOI: 10.1158/1541-7786.MCR-18-0451

    http://mcr.aacrjournals.org/lookup/doi/10.1158/1541-7786.MCR-18-0451http://mcr.aacrjournals.org/content/suppl/2019/01/03/1541-7786.MCR-18-0451.DC1http://mcr.aacrjournals.org/content/17/3/741.full#ref-list-1http://mcr.aacrjournals.org/content/17/3/741.full#related-urlshttp://mcr.aacrjournals.org/cgi/alertsmailto:[email protected]://mcr.aacrjournals.org/content/17/3/741http://mcr.aacrjournals.org/

    /ColorImageDict > /JPEG2000ColorACSImageDict > /JPEG2000ColorImageDict > /AntiAliasGrayImages false /CropGrayImages false /GrayImageMinResolution 200 /GrayImageMinResolutionPolicy /Warning /DownsampleGrayImages true /GrayImageDownsampleType /Bicubic /GrayImageResolution 300 /GrayImageDepth -1 /GrayImageMinDownsampleDepth 2 /GrayImageDownsampleThreshold 1.50000 /EncodeGrayImages true /GrayImageFilter /DCTEncode /AutoFilterGrayImages true /GrayImageAutoFilterStrategy /JPEG /GrayACSImageDict > /GrayImageDict > /JPEG2000GrayACSImageDict > /JPEG2000GrayImageDict > /AntiAliasMonoImages false /CropMonoImages false /MonoImageMinResolution 600 /MonoImageMinResolutionPolicy /Warning /DownsampleMonoImages true /MonoImageDownsampleType /Bicubic /MonoImageResolution 900 /MonoImageDepth -1 /MonoImageDownsampleThreshold 1.50000 /EncodeMonoImages true /MonoImageFilter /CCITTFaxEncode /MonoImageDict > /AllowPSXObjects false /CheckCompliance [ /None ] /PDFX1aCheck false /PDFX3Check false /PDFXCompliantPDFOnly false /PDFXNoTrimBoxError true /PDFXTrimBoxToMediaBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXSetBleedBoxToMediaBox true /PDFXBleedBoxToTrimBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXOutputIntentProfile (None) /PDFXOutputConditionIdentifier () /PDFXOutputCondition () /PDFXRegistryName () /PDFXTrapped /False

    /CreateJDFFile false /Description > /Namespace [ (Adobe) (Common) (1.0) ] /OtherNamespaces [ > /FormElements false /GenerateStructure false /IncludeBookmarks false /IncludeHyperlinks false /IncludeInteractive false /IncludeLayers false /IncludeProfiles false /MarksOffset 18 /MarksWeight 0.250000 /MultimediaHandling /UseObjectSettings /Namespace [ (Adobe) (CreativeSuite) (2.0) ] /PDFXOutputIntentProfileSelector /NA /PageMarksFile /RomanDefault /PreserveEditing true /UntaggedCMYKHandling /LeaveUntagged /UntaggedRGBHandling /LeaveUntagged /UseDocumentBleed false >> > ]>> setdistillerparams> setpagedevice