pai-1 leads to g -phase cell-cycle progression through cyclin … · pai-1 zymography of note, 30...

14
Cell Cycle and Senescence PAI-1 Leads to G 1 -Phase Cell-Cycle Progression through Cyclin D3/cdk4/6 Upregulation Evan Gomes Giacoia 1 , Makito Miyake 1 , Adrienne Lawton 3 , Steve Goodison 1,2,4 , and Charles J. Rosser 1,2 Abstract The canonical function of plasminogen activator inhibitor-1 (PAI-1/SERPINE1) is as an inhibitor of urokinase- type plasminogen activator for blood clot maintenance, but it is now also considered a pleiotropic factor that can exert diverse cellular and tumorigenic effects. However, the mechanism controlling its pleiotropic effects is far from being understood. To elucidate the tumorigenic role of PAI-1, we tested the effects of PAI-1 after manipulation of its expression or through the use of a small-molecule inhibitor, tiplaxtinin. Downregulation of PAI-1 signicantly reduced cellular proliferation through an inability to progress from the G 0 G 1 phase of the cell cycle. Accordingly, overexpression of PAI-1 augmented proliferation by encouraging S-phase entry. Biochemically, cell-cycle arrest was associated with the depletion of the G 1 -phase transition complexes, cyclin D3/cdk4/6 and cyclin E/cdk2, in parallel with the upregulation of the cell-cycle inhibitors p53, p21Cip1/Waf1, and p27Kip1. PAI-1 depletion signicantly decreased the tumor size of urothelial T24 and UM-UC-14 xenografts, and overexpression of PAI-1 substantially increased the tumor size of HeLa xenografts. Finally, immunohistochemical analysis of human bladder and cervical tumor tissue microarrays revealed increased expression of PAI-1 in cancerous tissue, specically in aggressive tumors, supporting the relevance of this molecule in human tumor biology. Implications: Targeting PAI-1 has benecial antitumoral effects and should be further investigated clinically. Mol Cancer Res; 12(3); 32234. Ó2014 AACR. Introduction PAI-1 is an endogenous inhibitor of urokinase-type plas- minogen activator (uPA). Specically, PAI-1 normally func- tions as part of the plasminogen activation system, which includes the serine protease uPA, its receptor uPA-R, tissue- type plasminogen activator (tPA), and inhibitors PAI-1 and PAI-2 (13). PAI-1 expression is regulated by many intrinsic factors (e.g., cytokines and growth factors) and extrinsic factors (e.g., cellular stress; ref. 4). Although the canonical function of PAI-1 has been known as an inhibitor of uPA to maintain clot formation, it is now regarded as a pleiotropic factor exerting diverse cellular effects, many potentially related to tumorigenesis, including cell migration, invasion, adhesion, and angiogenesis. Some reports suggest that PAI-1 inuences tumor pro- gression indirectly, through effects on the extracellular matrix that perturb migration, invasion, and angiogenesis (5, 6), but other reports claim that PAI-1 itself promotes tumor growth directly (7). For example, high levels of PAI-1 in human and murine cancer cells were associated with the retardation of tumor growth and invasion in a rodent model (8, 9). Thus, the role of PAI-1 in tumorigenesis and growth is complicated, varying with experimental design and its cel- lular origin, so more preclinical and mechanistic studies are required to elucidate the role of PAI-1 in human cancers. Dysregulation of cellular proliferation is a hallmark of cancer (1013). The cell cycle is a highly controlled process that involves tight regulation of key molecules that allow cells to progress between phases of the cell cycle. In an attempt to investigate the effect of PAI-1 on cell proliferation and cell- cycle progression, we created stable knockdown clones of PAI-1 in the urothelial T24 and UM-UC-14 cells and overexpressing PAI-1 clones in the cervical tumor cell HeLa. Next, we also inhibited PAI-1 activity with the small- molecule antagonist of PAI-1, tiplaxtinin (PAI-039; refs. 14, 15). Silencing of PAI-1 in T24 and UM-UC-14 cells via short hairpin RNA (shRNA) or tiplaxtinin treatment was associated with a marked inhibition of cellular proliferation causing a cell-cycle arrest in G 1 S phase. The silencing of PAI-1 by either genetic or pharmacologic techniques con- ferred depletion of the G 1 S transition molecules, cyclin D3/ cdk4/6 and cyclin E/cdk2, with a concomitant increase in the cell-cycle inhibitors p21 Cip1/Waf1 and p27 kip1 . In addi- tion, in vitro results were corroborated in a xenograft model Authors' Afliations: 1 Cancer Research Institute, MD Anderson Cancer Center Orlando; 2 Nonagen Bioscience Corp; 3 Department of Pathology, Orlando Health, Orlando, Florida; and 4 Department of Health Sciences Research, Mayo Clinic, Jacksonville, Florida Note: Supplementary data for this article are available at Molecular Cancer Research Online (http://mcr.aacrjournals.org/). Correspondence Author: Charles J. Rosser, Clinical and Translational Research, University of Hawaii Cancer Center, University of Hawaii School of Medicine, 701 Ilalo Street, Room 327, Honolulu, HI 96813. Phone: 808- 564-3812; Fax: 808-586-2984; E-mail: [email protected] doi: 10.1158/1541-7786.MCR-13-0543 Ó2014 American Association for Cancer Research. Molecular Cancer Research Mol Cancer Res; 12(3) March 2014 322 on December 16, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from Published OnlineFirst January 24, 2014; DOI: 10.1158/1541-7786.MCR-13-0543

Upload: others

Post on 25-Aug-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: PAI-1 Leads to G -Phase Cell-Cycle Progression through Cyclin … · PAI-1 zymography Of note, 30 mg of total cell lysate from PAI-1 clones (T24Scr, T24-PAI-1KD, UM-UC-14Scr, UM-UC-14-PAI-

Cell Cycle and Senescence

PAI-1 Leads to G1-Phase Cell-Cycle Progression throughCyclin D3/cdk4/6 Upregulation

Evan Gomes Giacoia1, Makito Miyake1, Adrienne Lawton3, Steve Goodison1,2,4, and Charles J. Rosser1,2

AbstractThe canonical function of plasminogen activator inhibitor-1 (PAI-1/SERPINE1) is as an inhibitor of urokinase-

type plasminogen activator for blood clot maintenance, but it is now also considered a pleiotropic factor that canexert diverse cellular and tumorigenic effects. However, the mechanism controlling its pleiotropic effects is far frombeing understood. To elucidate the tumorigenic role of PAI-1, we tested the effects of PAI-1 after manipulation ofits expression or through the use of a small-molecule inhibitor, tiplaxtinin. Downregulation of PAI-1 significantlyreduced cellular proliferation through an inability to progress from the G0–G1 phase of the cell cycle. Accordingly,overexpression of PAI-1 augmented proliferation by encouraging S-phase entry. Biochemically, cell-cycle arrest wasassociated with the depletion of theG1-phase transition complexes, cyclinD3/cdk4/6 and cyclin E/cdk2, in parallelwith the upregulation of the cell-cycle inhibitors p53, p21Cip1/Waf1, and p27Kip1. PAI-1 depletion significantlydecreased the tumor size of urothelial T24 and UM-UC-14 xenografts, and overexpression of PAI-1 substantiallyincreased the tumor size of HeLa xenografts. Finally, immunohistochemical analysis of human bladder and cervicaltumor tissue microarrays revealed increased expression of PAI-1 in cancerous tissue, specifically in aggressivetumors, supporting the relevance of this molecule in human tumor biology.

Implications: Targeting PAI-1 has beneficial antitumoral effects and should be further investigated clinically.MolCancer Res; 12(3); 322–34. �2014 AACR.

IntroductionPAI-1 is an endogenous inhibitor of urokinase-type plas-

minogen activator (uPA). Specifically, PAI-1 normally func-tions as part of the plasminogen activation system, whichincludes the serine protease uPA, its receptor uPA-R, tissue-type plasminogen activator (tPA), and inhibitors PAI-1 andPAI-2 (1–3). PAI-1 expression is regulated bymany intrinsicfactors (e.g., cytokines and growth factors) and extrinsicfactors (e.g., cellular stress; ref. 4). Although the canonicalfunction of PAI-1 has been known as an inhibitor of uPA tomaintain clot formation, it is now regarded as a pleiotropicfactor exerting diverse cellular effects, many potentiallyrelated to tumorigenesis, including cell migration, invasion,adhesion, and angiogenesis.Some reports suggest that PAI-1 influences tumor pro-

gression indirectly, through effects on the extracellular

matrix that perturb migration, invasion, and angiogenesis(5, 6), but other reports claim that PAI-1 itself promotestumor growth directly (7). For example, high levels of PAI-1in human and murine cancer cells were associated with theretardation of tumor growth and invasion in a rodent model(8, 9). Thus, the role of PAI-1 in tumorigenesis and growth iscomplicated, varying with experimental design and its cel-lular origin, so more preclinical and mechanistic studies arerequired to elucidate the role of PAI-1 in human cancers.Dysregulation of cellular proliferation is a hallmark of

cancer (10–13). The cell cycle is a highly controlled processthat involves tight regulation of keymolecules that allow cellsto progress between phases of the cell cycle. In an attempt toinvestigate the effect of PAI-1 on cell proliferation and cell-cycle progression, we created stable knockdown clones ofPAI-1 in the urothelial T24 and UM-UC-14 cells andoverexpressing PAI-1 clones in the cervical tumor cell HeLa.Next, we also inhibited PAI-1 activity with the small-molecule antagonist of PAI-1, tiplaxtinin (PAI-039; refs. 14,15). Silencing of PAI-1 in T24 and UM-UC-14 cells viashort hairpin RNA (shRNA) or tiplaxtinin treatment wasassociated with a marked inhibition of cellular proliferationcausing a cell-cycle arrest in G1–S phase. The silencing ofPAI-1 by either genetic or pharmacologic techniques con-ferred depletion of theG1–S transitionmolecules, cyclinD3/cdk4/6 and cyclin E/cdk2, with a concomitant increase inthe cell-cycle inhibitors p21Cip1/Waf1 and p27kip1. In addi-tion, in vitro results were corroborated in a xenograft model

Authors' Affiliations: 1Cancer Research Institute, MD Anderson CancerCenter Orlando; 2Nonagen Bioscience Corp; 3Department of Pathology,Orlando Health, Orlando, Florida; and 4Department of Health SciencesResearch, Mayo Clinic, Jacksonville, Florida

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

Correspondence Author: Charles J. Rosser, Clinical and TranslationalResearch, University of Hawaii Cancer Center, University of Hawaii Schoolof Medicine, 701 Ilalo Street, Room 327, Honolulu, HI 96813. Phone: 808-564-3812; Fax: 808-586-2984; E-mail: [email protected]

doi: 10.1158/1541-7786.MCR-13-0543

�2014 American Association for Cancer Research.

MolecularCancer

Research

Mol Cancer Res; 12(3) March 2014322

on December 16, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2014; DOI: 10.1158/1541-7786.MCR-13-0543

Page 2: PAI-1 Leads to G -Phase Cell-Cycle Progression through Cyclin … · PAI-1 zymography Of note, 30 mg of total cell lysate from PAI-1 clones (T24Scr, T24-PAI-1KD, UM-UC-14Scr, UM-UC-14-PAI-

in which silencing of PAI-1 led to a reduction in tumori-genicity with decreased levels of cyclin D3, and increasedexpression of p21Cip1/Waf1 and p27kip1, whereas overexpres-sion of PAI-1 in HeLa xenografts led to an enhancement intumor growth, which was associated with increased cyclinD3 expression along with a decrease expression ofp21Cip1/Waf1 and p27kip1. Finally, immunohistochemicalanalysis of bladder and cervical tissues revealed that PAI-1levels were increased in both human urothelial and cervicalcancer patient tissue samples, and higher expression wascorrelated with more aggressive tumors.

Materials and MethodsImmunohistochemical staining of human tumors forPAI-1After approval from the Institutional Review Board of

MD Anderson Cancer Center Orlando (Orlando, FL), 4-mm tissue sections from 163 bladder cancer patient speci-mens and commercially available tissue microarrays (TMA)BNC12011 (US Biomax, Inc.), consisting of 37 benignbladder controls, as well as CR805 (US Biomax, Inc.),consisting of 70 cervical cancers and 10 benign cervicaltissues, were examined by immunohistochemical staining.Details of the protocol and antibodies are available inSupplementary Material.

Cell culture and reagentsThe human urothelial cell lines T24, UM-UC-14, RT4

(American Type Culture Collection, ATCC), andUROtsa, derived from benign urothelial tissue (a generousgift from Dr. Donald Sens, University of North DakotaSchool of Medicine, Grand Forks, ND) as well as thehuman cervical cancer cell line, HeLa (ATCC), were usedfor these studies and maintained in media as previouslydescribed (16, 17). T24, UM-UC-14, and HeLa cells werecultured in RPMI-1640 (ATCC) medium supplementedwith 10% FBS, 50 U/mL penicillin and 50 mg/mL strep-tomycin. RT4 cells were cultured in the McCoy mediumsupplemented with 10% FBS, 50 U/mL penicillin, and50mg/mL streptomycin (InvitrogenCorp.). Because of thedifficulty in culturing RT4 cells, they were used in limitedexperimental assays. UROtsa cells were cultured in Dul-becco's Modified Eagle Medium with low glucose (Invi-trogen Corp.) supplemented with 1 mg/mL of glucose,10% FBS, 50 U/mL penicillin, and 50 mg/mL strepto-mycin. All cells were maintained in a standard humidifiedincubator at 37�C in 5%CO2. PAI-1 inhibitor, tiplaxtinin(PAI-039; Axon MedChem) was dissolved in dimethylsulfoxide at a stock concentration of 10 mmol/L and storedat �20�C.

Generation of PAI-1 knockdown/overexpression stablecell linesStable cells containing a functional null knockdown of

PAI-1 in T24 (T24-PAI-1KD) and UM-UC-14 (UM-UC-14-PAI-1KD) were generated using a plasmid with PAI-1shRNA cloned within a pGFP–V–RS vector (OriGene

Technologies) and transfected into T24 and UM-UC-14cells at 80% confluency in a 6-well plate using Fugene HDtransfection reagent (Roche Diagnostics). A plasmid with ascrambled (Scr) noneffective shRNA construct in pGFP–V–RS was concomitantly produced as a negative control(T24Scr and UM-UC-14Scr). The cells were selected inmedium containing 1 mg/mL of puromycin (Life Technol-ogies) for 14 days and subcloned by limiting dilution in 96-well plates. Pilot dose–response studies were performed todetermine the lowest dose of G418 or puromycin that cankill all the untransfected cells. PAI-1 shRNA sequences areavailable in SupplementaryMaterial. Next, HeLa cells stablyoverexpressing a functionally active PAI-1 (HeLa-PAI-1OE)were produced using a plasmid containing a sequenceverified human PAI-1 cDNA cloned into a pCMV6 entryvector and transfected intoHeLa cells at 80% confluency in a6-well plate using Fugene HD transfection reagent. Aplasmid with vector alone was transfected as an emptycontrol (HeLaEmpty). The cells were selected in mediumcontaining 1,200 mg/mL of G418 (Life Technologies) for14 days and subcloned by limiting dilution in 96-well plates.All generated stable cell lines were maintained in mediacontaining 0.25 mg/mL of puromycin for T24 clones (T24-PAI-1KD and T24Scr) and UM-UC-14 clones (UM-UC-14-PAI-1KD and UM-UC-14Scr) and in media containing 500mg/mL of G418 for HeLa clones (HeLe-PAI-1OE andHeLaEmpty).

ImmunoblottingCell lysate and immunoblotting were performed using

standard protocols as previously described (16). Antibodydetails are available in Supplementary Material.

Real-time reverse transcription PCRRNA was extracted from cells using the RNeasy Mini Kit

(Qiagen) as per the manufacturer's instructions. Conversionto cDNA was achieved through the High Capacity cDNAReverse Transcription Kit (Life Technologies). Quantitativereverse transcriptase (RT) PCR was carried out using ABI7300Real-Time PCRSystem (Life Technologies) in a 20mLreaction volume containing 1mL of the first-strand cDNA, 1mL of gene-specific TaqMan primer and probe mix. Primersets can be found in Supplementary Material. The foldchange in mRNA expression levels was calculated by the2�DDCt method (18).

Measurement of secreted PAI-1 by ELISAParental tumor cells (T24, UM-UC-14, and HeLa) and

PAI-1 clones (T24Scr, T24-PAI-1KD, UM-UC-14Scr,UM-UC-14-PAI-1KD, HeLaEmpty, and HeLa-PAI-1OE)were plated onto 6-well plate at a density of 2 � 105

cells in 2 mL of growth media per well. After incubationfor 48 hours, the cultured supernatant was collected andcentrifuged at 1,000 rpm for 5 minutes to remove anydead cells. The concentration of secreted PAI-1 in theculture supernatant was determined by using a plasmin-ogen activator inhibitor 1 (PAI-1) Human ELISA Kit(ab99970; Abcam).

PAI-1 Regulates Cyclin D3

www.aacrjournals.org Mol Cancer Res; 12(3) March 2014 323

on December 16, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2014; DOI: 10.1158/1541-7786.MCR-13-0543

Page 3: PAI-1 Leads to G -Phase Cell-Cycle Progression through Cyclin … · PAI-1 zymography Of note, 30 mg of total cell lysate from PAI-1 clones (T24Scr, T24-PAI-1KD, UM-UC-14Scr, UM-UC-14-PAI-

PAI-1 zymographyOf note, 30 mg of total cell lysate from PAI-1 clones

(T24Scr, T24-PAI-1KD, UM-UC-14Scr, UM-UC-14-PAI-1KD, HeLaEmpty, and HeLa-PAI-1OE) and parental tumorcells (T24, UM-UC-14, andHeLa treated in the presence orabsence of tiplaxtinin) were electrophoresed on 10% SDS–PAGE gel containing 1 mg/mL casein (Sigma), 10 mg/mLplasminogen (Sigma), and 0.5 mU/mL uPA under nonre-ducing conditions. After electrophoresis, SDS removal andPAI-1 renaturation were achieved by washing the gel for 1hour in buffer containing 2.5% Triton-X 100, 50 mmol/LTris pH 7.4, 5 mmol/L CaCl2, and 1 mmol/L ZnCl2. Thegel was then incubated in a reaction buffer containing 50mmol/L Tris pH 7.4, 5 mmol/L CaCl2, 1 mmol/L ZnCl2,and 0.02%NaN3 pH 8.0 for 18 hours at 37�C, followed bystaining with Coomassie blue. PAI-1 activity was indicatedas lytic zones of plasmin generation. Gel was photographedusing the KODAK Gel Logic 200 Imaging System withCarestream Molecular Imaging Software Standard Editionv5.0.7.24 (Carestream Health) when opaque bandsappeared on a clear background.

Proliferation and viability assayPAI-1 clones (T24Scr, T24-PAI-1KD, UM-UC-14Scr,

UM-UC-14-PAI-1KD,HeLaEmpty, andHeLa-PAI-1OE) andparental tumor cells (T24, UM-UC-14, and HeLa) wereplated in white-walled 96-well dishes in triplicate (103 cells/well) and treated with or without tiplaxtinin at the indicatedconcentrations. Cell proliferation was determined by Cell-Titer-Glo Luminescent cell viability/proliferation assay(Promega) according to the manufacturer's instructions at24, 48, and 72 hours after seeding. Samples were incubatedin an orbital shaker for 2 minutes to facilitate cell lysisfollowed by additional 10 minutes incubation at roomtemperature, and luminescence was measured using aFLUOstar Optima Reader (BMG LABTECH). IC50 oftiplaxtinin was determined in GraphPad Prism (GraphPadSoftware, Inc.) for each cell line. At least three independentexperiments consisting of each condition tested in triplicatewells were used to calculate mean � SD values.

Cell synchronization and cell-cycle distributionFor cell-cycle analysis, propidium iodide staining and

fluorescence-activated cell sorting were performed in PAI-1 clones (T24Scr, T24-PAI-1KD, UM-UC-14Scr, UM-UC-14-PAI-1KD, HeLaEmpty, and HeLa-PAI-1OE) and parentaltumor cell lines (T24, UM-UC-14, and HeLa) treated withor without tiplaxtinin. Briefly, cells were trypsinized, fixed inethanol for 1 hour at 4�C, stained with propidium iodidesolution (10 mg/mL) with RNase A (250 mg/mL), incubatedat 37�C for 30 minutes, and analyzed using the BD FACS-Caliber with Cell Quest Pro Software (BD Biosciences) andModFit LT (Verity Software House). Next to obtain popu-lations of cells in G1-phase, the above cells were arrested bydouble thymidine block (19). Briefly, cells were blocked for18 hours with 2.5 mmol/L thymidine, released for 8 hoursby washing out the thymidine, and then arrested again withanother 2.5 mmol/L thymidine for 18 hours. The cells were

then released from the thymidine block with two washes offreshmedium and allowed to progress throughG1-phase andinto S-phase, then the cell-cycle position of the cells wasdetermined at 0, 3, 6, and 12 hours by propidium iodidestaining.Subsequently, the progression into and through the S-

phase was assessed in the above cells using the Click-iT EdUAlexa Fluor 647 Flow Cytometry Assay Kit (Life Technol-ogies). EdU (5-ethynyl-20-deoxyuridine) is a thymidineanalog that becomes incorporated into DNA during activecellular DNA synthesis. Detection is determined via acopper-catalyzed covalent reaction between an azide (con-jugated to Alexa Fluor 647) and an alkyne. Briefly, PAI-1clones were seeded at 105 cells per well in a 6-well plate andsubsequently treated with 10 mmol/L of EdU for 24 hoursbefore harvesting. Cells were trypsinized, fixed in 4% form-aldehyde, and EdU incorporation was determined with theClick-iT EdU Alexa Fluor 647 flow Cytometry Assay Kit(Molecular Probes) according to the manufacturer's instruc-tions. Cell-cycle distribution and DNA synthesis of syn-chronized cells were also assessed by flow cytometry usingthe BD FACSCaliber with Cell Quest Pro Software (BDBiosciences) and ModFit LT (Verity Software House) orFlowJo (TreeStar Inc.).

In vivo tumorigenicityThe importance of PAI-1 expression in regard to tumor-

igenicity was assessed in an in vivo mouse model. AthymicBALB/c (nu/nu)mice, 6- to 8-week-old, were obtained fromHarlan Laboratories. Animal care was in compliance withthe recommendations of The Guide for Care and Use ofLaboratory Animals (National Research Council) andapproved by the Institutional Animal Care and Use Com-mittee. Tumor growth of the T24 PAI-1 knockdown clone,T24-PAI-1KD-19, was compared with the T24Scr control,theUM-UC-14 PAI-1 knockdown clone,UM-UC-14-PAI-1KD-4, was compared with the UM-UC-14Scr control, andfinally the HeLa PAI-1 overexpression clone, HeLa-PAI-1OE-12, was compared with the HeLaEmpty control. Cellswere harvested and resuspended in a 1:1 mixture ofserum-free RPMI and Matrigel (BD Biosciences). T24and UM-UC-14 cells (2 � 106) and HeLa cells (106) in100 mL of RPMI-1640 were mixed with 100 mL ofMatrigel and injected subcutaneously utilizing a 27-gauge needle into the rear flank of the mice as describedpreviously (20). Tumor volumes were measured andrecorded weekly with digital calipers and calculated byvolume ðmm3Þ¼ length� ðwidthÞ2 � 0:5236. Sta-tistical analysis of tumor volume was performed inGraphPad Prism (GraphPad Software, Inc.). After 4weeks, mice were sacrificed and tumors resected foranalysis.

Immunohistochemical analysis of xenograft tumorsImmunohistochemistry (IHC) was conducted as

described in ref. 20. Details and antibodies listed in Sup-plementary Material.

Giacoia et al.

Mol Cancer Res; 12(3) March 2014 Molecular Cancer Research324

on December 16, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2014; DOI: 10.1158/1541-7786.MCR-13-0543

Page 4: PAI-1 Leads to G -Phase Cell-Cycle Progression through Cyclin … · PAI-1 zymography Of note, 30 mg of total cell lysate from PAI-1 clones (T24Scr, T24-PAI-1KD, UM-UC-14Scr, UM-UC-14-PAI-

Statistical analysisExperimental data were expressed as mean with SD. All

statistical analyses were conducted using the Student t test,the Mann–Whitney nonparametric U test, or the one-wayANOVA and compared with the control(s). The compar-ison between PAI-1 expression in human tumors, based ondisease status, tumor grade, and tumor stage, was calculatedusing the Fisher exact test. A P value of <0.05was consideredsignificant. All statistical analyses and figures were carried outusing GraphPad Prism software 5.0 (GraphPad Software,Inc.).

ResultsPAI-1 expression is upregulated in aggressive humancancersIn biomarker discovery studies, our group previously

profiled the transcriptome of exfoliated urothelia obtainedfrom urine samples from a cohort of 92 patients withknown bladder disease status. We identified PAI-1 as avaluable biomarker for the detection of bladder cancer(21). The PAI-1 expression correlation was validated in anindependent cohort of 81 subjects using quantitative real-time PCR (22), and in two independent studies comprisedof 435 subjects using ELISA assays (23, 24). In the presentstudy, we assessed PAI-1 expression in human tissuespecimens by immunostaining. Immunohistochemicalstaining of 37 benign bladder tissues and 163 bladder

tumor tissues revealed significantly higher expression ofPAI-1 predominantly within the epithelial component ofbladder cancer tissues than in benign tissue (P <0.0001; Fig. 1A–E). However, some immunoreactivitywas noted in the stroma, mostly associated with vascula-ture. Furthermore, a correlation between PAI-1 expressionstatus and clinicopathologic features of bladder cancerrevealed a significant association of PAI-1 expression inmuscle invasive bladder cancer (MIBC) compared withnonmuscle invasive bladder cancer (NMIBC; P ¼0.0172; Fig. 1F). No association with tumor grade(P ¼ 0.6021) was identified (Fig. 1G).Next, we expanded our investigation to cancer of

another organ, the cervix. A TMA consisting of 10normal and 70 cervical cancers was analyzed for PAI-1expression. PAI-1 was absent in benign cervical tissue,but significant (P < 0.0001) expression was observed incancerous tissue (Supplementary Fig. S1). PAI-1 expres-sion in cervical cancer specimens was located in thetumor cells predominantly, but was also associated withthe vasculature in the tumor-adjacent stroma. Unlike inhuman bladder cancer, PAI-1 expression was not asso-ciated with higher stage cervical cancer (P ¼ 0.3147);however, the expression was correlated with high-gradecervical cancer (P ¼ 0.0303; Supplementary Fig. S1).The data provide evidence that increased PAI-1 levels areassociated with aggressive cancers in bladder and cervicallesions.

A B C D

E

% o

f To

tal c

ases

% o

f To

tal c

ases

% o

f To

tal c

ases

F G150 P < 0.0001

P = 0.0172 P = 0.6021

Normal NMIBC MIBC Low High

Stage GradeTumor

Low

Moderate

High

Low

Moderate

High

Low

Moderate

High100

50

0

150

100

50

0

150

100

50

0

Figure 1. PAI-1 overexpression in humanbladder cancer. PAI-1 levels weremeasured by immunohistochemical analysis of 37 benign bladder and 163 bladdertumor tissues. Representative PAI-1 expression levels are shown in benign (A) and bladder cancer tissue samples with absent (B), weak (C), and strong(D) staining. Scale bars, 100 mm. Arrows depict stromal tissue. Correlation was assessed between PAI-1 expression among benign versus cancer tissue (E),NMIBC versus MIBC (F), and high-grade versus low-grade (G).

PAI-1 Regulates Cyclin D3

www.aacrjournals.org Mol Cancer Res; 12(3) March 2014 325

on December 16, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2014; DOI: 10.1158/1541-7786.MCR-13-0543

Page 5: PAI-1 Leads to G -Phase Cell-Cycle Progression through Cyclin … · PAI-1 zymography Of note, 30 mg of total cell lysate from PAI-1 clones (T24Scr, T24-PAI-1KD, UM-UC-14Scr, UM-UC-14-PAI-

Downregulation of PAI-1 leads to inhibition of cellularproliferationThe baseline expression of PAI-1 was explored in a panel

of cell lines; UROtsa, RT4, T24, UM-UC-14, and HeLa(Supplementary Fig. S2).Western blot analysis revealed thatPAI-1 is differentially expressed in the panel, with increasedexpression apparent in T24 and UM-UC-14 cells. Theseresults were confirmed at the mRNA level and by evaluationof secreted PAI-1 protein by ELISA. Given the progressiveupregulation of PAI-1 in human bladder and cervical can-cers, we investigated how PAI-1 influences key cell processesby genetically manipulating the expression of PAI-1 in T24,UM-UC-14, and HeLa cells. Briefly, we selected two stablytransfected PAI-1 knockdown clones of urothelial T24(T24-PAI-1KD-19 and T24-PAI-1KD-22) and UM-UC-14 cells (UM-UC-14-PAI-1KD-4 and UM-UC-14-PAI-1KD-17) using shRNA, and two stably transfected HeLacervical cancer cells (HeLa-PAI-1OE-12 andHeLa-PAI-1OE-18) that overexpressed a functionally active human PAI-1.Efficiency of shRNA-mediated depletion and exogenousPAI-1 overexpression was documented by Western blotanalysis, RT-PCR, and ELISA assay of the conditionedmedia (Supplementary Fig. S2). In addition to shRNAtechnology, inhibition of PAI-1 was achieved through treat-ment of parental T24, UM-UC-14, and HeLa cells with thesmall-molecule inhibitor, tiplaxtinin, a novel antagonistcapable of silencing PAI-1 activity. PAI-1 activity wasabrogated in urothelial parental cells and HeLa parentalcells treated with tiplaxtinin, as demonstrated by the appear-ance of lytic zones of plasmin generation in zymography gelscontaining plasminogen and uPA under nonreducing con-ditions. The expected activity of PAI-1 in urothelial cellknockdowns and overexpressingHeLa clones was confirmedby zymography (Supplementary Fig. S2).In an in vitro proliferation assay, proliferation of T24 cells

(Fig. 2A), UM-UC-14 cells (Fig. 2B), and HeLa cells (Fig.2C) correlated with PAI-1 expression, i.e., reduced PAI-1expression in T24 and UM-UC-14 clones was associatedwith a reduction in cellular proliferation. Treatment of allparental cells with tiplaxtinin resulted in a significant reduc-tion in cellular proliferation (Fig. 2A–C). IC50 values fortiplaxtinin were determined for UROtsa cells (70.3 � 0.1mmol/L), T24 cells (43.7 � 6.3 mmol/L), UM-UC-14 cells(52.8 � 1.6 mmol/L), and HeLa cells (29.9 � 3.1 mmol/L).Our results show that reduction of PAI-1 activity in T24,UM-UC-14, or HeLa cells via genetic manipulation ortiplaxtinin treatment causes significant cell growthinhibition.

Loss of PAI-1 causes cell-cycle arrest by differentiallyregulating key cell-cycle proteinsNext, we investigated the effects of silencing PAI-1 expres-

sion on cell-cycle progression. The percentage of cells inG0–G1, S, and G2–Mphases of the cell cycle were monitoredin cells with functional inhibition of PAI-1 (T24-PAI-1KD-19and T24-PAI-1KD-22; UM-UC-14-PAI-1KD-4 and UM-UC-14-PAI-1KD-17), and in overexpressors of PAI-1(HeLa-PAI-1OE-12 and HeLa-PAI-1OE-18) using flow

cytometry (Fig. 3A). Inhibition of PAI-1 in T24 clonesresulted in the accumulation of cells in G0–G1 phase from65% in T24Scr control cells to 74% in T24-PAI-1KD-19 and75% in T24-PAI-1KD-22, along with a reduction in S-phasecells from 29% in T24Scr control to 19% in both T24-PAI-1KD-19 and -22 clones. In UM-UC-14 clones, there was anaccumulation of cells inG0–G1 phase from 55% inUM-UC-14Scr to 82% in UM-UC-14-PAI-1KD-4 and 73% in UM-UC-14-PAI-1KD-17, along with a reduction in S-phasecells from 28% in UM-UC-14Scr to 9% in UM-UC-14-PAI-1KD-4 and 3% in UMUC14-PAI-1KD-17.Similar to PAI-1 knockdown, inhibition of PAI-1 with

tiplaxtinin in parental cells caused a G0–G1 phase cell-cyclearrest (Fig. 3B). In T24 cells, tixplaxtinin treatment wasassociated with an increase in the percentage of cells inG0–G1 phase from 50% to 74% and a correspondingdecrease in S-phase cells from 34% in T24 control cells to8% in T24 cells treated with 50 mmol/L tiplaxtinin. UM-UC-14 cells exhibited an accumulation in G0–G1 phasefrom 59% in control cells to 76% in cells treated with 50mmol/L tiplaxtinin, and depletion in cells in S-phase from28% in control cells to 13% in tiplaxtinin-treated cells. InHeLa cells, G0–G1 phase, accumulation was observed from57% in control cells to 75% in HeLa cells treated with 50mmol/L tiplaxtinin, and a concomitant decrease in S-phasecells from 27% in control to 11% was observed.To elucidate the molecular changes associated with the

observed G0–G1 phase cell-cycle arrest in PAI-1–depletedcells, we assessed the expression of a panel of key regulatoryproteins of the cell-cycle machinery byWestern blot analysis(Fig. 3C and D). Inhibition of PAI-1 in T24-PAI-1KD orUM-UC-14KD clones and in parental T24, UM-UC-14,and HeLa cells treated with tiplaxtinin resulted in thedepletion of the G1–S transition molecules cyclin D3,cdk4/6, cyclin E, and cdk2. A concomitant increase wasobserved in the levels of the cell-cycle inhibitors p53,phospho-p53, p21Cip1/Waf1, and p27kip1, which all playsignificant roles in G0–G1 to S-phase progression. Thus,our data suggest that the loss of PAI-1 prevents cell-cycle progression of urothelial and cervical cancer cellsthrough G1 into S-phase through the regulation of keycell-cycle factors.

PAI-1 knockdown interferes with progression throughthe G1-phase of the cell cycleTo further validate that abrogation of PAI-1 can lead to

growth phase arrest of human cells, T24 clones (T24-PAI-1KD-19 and T24-PAI-1KD-22) and UM-UC-14 clones(UM-UC-14-PAI-1KD-4 andUMUC14-PAI-1KD-17) weresynchronized in G0–G1 phase using a double thymidineblock, then cell-cycle progression was monitored by flowcytometry following thymidine release. Although controlsT24Scr and UM-UC-14Scr were able to progress out ofG1-phase, T24 and UM-UC-14 clones depleted of PAI-1were primarily arrested in G1-phase with little to no pro-gression to S-phase (Fig. 4A). To confirm this finding, thepercentage of cells entering S-phase was alsomonitored usingthe Click-iT EdU Alexa Fluor 647 Flow Cytometry Assay

Giacoia et al.

Mol Cancer Res; 12(3) March 2014 Molecular Cancer Research326

on December 16, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2014; DOI: 10.1158/1541-7786.MCR-13-0543

Page 6: PAI-1 Leads to G -Phase Cell-Cycle Progression through Cyclin … · PAI-1 zymography Of note, 30 mg of total cell lysate from PAI-1 clones (T24Scr, T24-PAI-1KD, UM-UC-14Scr, UM-UC-14-PAI-

Kit. Specifically, the incorporation of the thymidine analog,EdU was examined in cells at 0, 3, 6, and 12 hours afterrelease from thymidine block (Fig. 4B). Depletion of PAI-1did significantly delay the exit of cells from G1-phase, withonly 11% and 15% of the T24-PAI-1KD-19 and T24-PAI-1KD-22 clone cells in S-phase, respectively, within 3 hours,whereas 43% of the T24Scr control cells progressed into S-phase within 3 hours (P < 0.01; Fig. 4B). In UM-UC-14clones, only 10% of UM-UC-14-PAI-1KD-4 and 8% ofUMUC14-PAI-1KD-17 cells were able to progress intoS-phase, whereas 30% of UM-UC-14Scr control cells were

in S-phase (P< 0.01; Fig. 4B).Our results suggest that PAI-1expression is necessary to allow transition of cells fromG1-phase to S-phase of the cell cycle. This is the first reportthat has linked PAI-1 to cell-cycle changes, and specificallyassessed cell promotion through G0–G1 phase.

Inhibition of PAI-1 prevents xenograft tumor growthSubcutaneous xenograft tumor growth of T24, UM-UC-

14, and HeLa cells was assessed in athymic nude mice. Invivo experiments showed amarked decrease in tumor volumein the T24-PAI-1KD-19 and UM-UC-14-PAI-1KD-4

150

100

50

0

150

100

50

0Cel

l pro

lifer

atio

n %

co

ntr

ol

150

100

50

0Cel

l pro

lifer

atio

n %

co

ntr

ol

200

150

100

50

0

Cel

l pro

lifer

atio

n %

co

ntr

ol

150

100

50

0Cel

l pro

lifer

atio

n %

co

ntr

ol

150

100

50

0Cel

l pro

lifer

atio

n %

co

ntr

ol

Cel

l pro

lifer

atio

n %

co

ntr

ol

Scr

Scr

Em

pty

Co

ntr

ol

OE

cl.

12

OE

cl.

18

Em

pty

OE

cl.

12

OE

cl.

18

Em

pty

OE

cl.

12

OE

cl.

18

Scr

Co

ntr

ol

Co

ntr

ol

Co

ntr

ol

Co

ntr

ol

Tip

lax 3

0 μ

mo

l/L

Tip

lax 3

0 μ

mo

l/L

Tip

lax 3

0 μ

mo

l/L

Tip

lax 5

0 μ

mo

l/L

Co

ntr

ol

Tip

lax 3

0 μ

mo

l/L

Tip

lax 5

0 μ

mo

l/L

Co

ntr

ol

Tip

lax 3

0 μ

mo

l/L

Tip

lax 5

0 μ

mo

l/L

Tip

lax 5

0 μ

mo

l/L

Co

ntr

ol

Tip

lax 3

0 μ

mo

l/L

Tip

lax 5

0 μ

mo

l/L

Co

ntr

ol

Tip

lax 3

0 μ

mo

l/L

Tip

lax 5

0 μ

mo

l/L

Tip

lax 5

0 μ

mo

l/L

Tip

lax 3

0 μ

mo

l/L

Tip

lax 3

0 μ

mo

l/L

Tip

lax 5

0 μ

mo

l/L

Tip

lax 5

0 μ

mo

l/L

Scr

Scr

KD

cl.

19

KD

cl.

4

KD

cl.

4

KD

cl.

17

KD

cl.

17

Scr

KD

cl.

4

KD

cl.

17

KD

cl.

19

KD

cl.

19

KD

cl.

22

KD

cl.

22

KD

cl.

22

24 h

24 h

48 h 72 h

T24-PAI-1KD

UM-UC-14-PAI-1KD

HeLa-PAI-1OE HeLa

UM-UC-14

T24

24 h 48 h 72 h

48 h 72 h

A

B

C

24 h 48 h 72 h

24 h

24 h 48 h 72 h

48 h 72 h

Figure 2. Depletion of PAI-1 inhuman tumor cell lines inhibits cellproliferation. Proliferation rateswere measured in cancer cellclones T24-PAI-1KD (A), UM-UC-14-PAI-1KD (B), and HeLa-PAI-1OE

(C) over a 72-hour period. Similarly,proliferation rates were measuredin parental T24 (A), UM-UC-14 (B),andHeLa (C) cells without andwithtiplaxtinin at 30 and 50 mmol/L overa 72-hour period. Data,mean�SDrelative to untreated cells, whichare set to 100%. Threeindependent experiments wereperformed in triplicate.Significance compared withcontrol cells is denoted by�, P < 0.05.

PAI-1 Regulates Cyclin D3

www.aacrjournals.org Mol Cancer Res; 12(3) March 2014 327

on December 16, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2014; DOI: 10.1158/1541-7786.MCR-13-0543

Page 7: PAI-1 Leads to G -Phase Cell-Cycle Progression through Cyclin … · PAI-1 zymography Of note, 30 mg of total cell lysate from PAI-1 clones (T24Scr, T24-PAI-1KD, UM-UC-14Scr, UM-UC-14-PAI-

T24-Scr

T24 Control

UMUC-14 Control

HeLa Control HeLa Tiplax 30 μmol/L HeLa Tiplax 50 μmol/L

UMUC-14 Tiplax 30 μmol/L UMUC-14 Tiplax 50 μmol/L

T24 Tiplax 30 μmol/L T24 Tiplax 50 μmol/L

UMUC-14-Scr UMUC-14-PAI-1KD-4 UMUC-14-PAI-1KD-17

HeLa-empty HeLa-PAI-1OE-12 HeLa-PAI-1OE-18

T24-PAI-1KD-19 T24-PAI-1KD-22

Cyclin D3

T24

Contr

ol

Scr

KD

cl.

19

KD

cl.

22

Scr

Em

pty

OE

cl.

12

OE

cl.

18

KD

cl.

4

KD

cl.

17

Tip

lax 3

0 μ

mol/L

Tip

lax 5

0 μ

mol/L

Contr

ol

Tip

lax 3

0 μ

mol/L

Tip

lax 5

0 μ

mol/L

Contr

ol

Tip

lax 3

0 μ

mol/L

Tip

lax 5

0 μ

mol/L

CDK4

CDK6

Cyclin E

CDK 2

p53

Phospho-p53

p21

p27

β-Actin

Cyclin D3

CDK4

CDK6

Cyclin E

CDK 2

p53

Phospho-p53

p21

p27

β-Actin

T24-PAI-1KD UM-UC-14-PAI-1KD HeLa-PAI-1OE

UM-UC-14 HeLa

A C

B D

Figure 3. Inhibition of PAI-1 promotes cell-cycle arrest by depleting cell-cycle regulatory proteins associated with G1–S transition. A, cell-cycle progressionwas examined by flow cytometry in generated T24 clones (T24Scr, T24-PAI-1KD-19, and T24-PAI-1KD-22), UM-UC-14 clones (UM-UC-14Scr, UM-UC-14-PAI-1KD-4, and UMUC14-PAI-1KD-17) both depleted of PAI-1, in addition to HeLa clones (HeLaEmpty, HeLa-PAI-1OE-12, and HeLa-PAI-1OE-18) overexpressingPAI-1. B, furthermore, cell-cycle progression was noted in parental cells (T24, UM-UC-14, and HeLa) treated with tiplaxtinin. All experiments wererepeated at least three times and a representative of one experiment is depicted. The percentage of cells in G0–G1, S, and G2–M phase of the cell cycle wasobservedby flowcytometry after propidium iodide staining of cellular DNAand analyzed by theModFit software. Expression levels of several knownG0–G1, S,and G2–M phase cell-cycle regulatory factors were analyzed by immunoblotting of clones in C and parental cells treated with tiplaxtinin in D.

Giacoia et al.

Mol Cancer Res; 12(3) March 2014 Molecular Cancer Research328

on December 16, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2014; DOI: 10.1158/1541-7786.MCR-13-0543

Page 8: PAI-1 Leads to G -Phase Cell-Cycle Progression through Cyclin … · PAI-1 zymography Of note, 30 mg of total cell lysate from PAI-1 clones (T24Scr, T24-PAI-1KD, UM-UC-14Scr, UM-UC-14-PAI-

knockdown clones when compared with their respectivecontrols, T24Scr and UM-UC-14Scr (Fig. 5). Upon com-pletion of the study at 4 weeks, T24Scr control xenograftswere noted to have an average volume of 516mm3 comparedwith 168 mm3 for T24-PAI-1KD-19 xenografts (P ¼0.0005). UM-UC-14Scr xenografts had a volume of 1,550mm3 compared with 633 mm3 for UM-UC-14-PAI-1KD-4

xenografts (P¼ 0.0039). Conversely, overexpression of PAI-1 in HeLa-PAI-1OE-12 enhanced tumor growth consider-ably relative to HeLaEmpty control cells. HeLaEmpty xeno-grafts had a tumor volume of 933 mm3 compared withHeLa-PAI-1OE-12, which exhibited a dramatic increase intumor volume to 1,858 mm3 (P ¼ 0.0195; Fig. 5A). Weconfirmed the expected differences in tissue PAI-1 levels in

A

B

Figure 4. Depletion of PAI-1 prevents passage of tumor cells out of G1 phase of cell cycle. A, T24 clones (T24Scr, T24-PAI-1KD-19, and T24-PAI-1KD-22) andUM-UC-14 clones (UM-UC-14Scr, UM-UC-14-PAI-1KD-4, and UMUC14-PAI-1KD-17) were synchronized in G0–G1 via a double thymidine block andcell-cycle progression was monitored following thymidine release by propidium iodide staining by flow cytometry analysis at 0 (orange), 3 (green), 6 (blue),and 12 hours (red). All experiments were repeated at least three times and a representative of one experiment is depicted. B, the percentage of cells enteringS-phase was quantified using the Click-iT EdU Alexa Fluor 647 Flow Cytometry Assay Kit. Incorporation of EdU was examined in cells at 0, 3, 6, and12 hours after release of thymidine block. All experiments were repeated at least three times. Data, mean þ SD. �, P < 0.05, relative to control.

PAI-1 Regulates Cyclin D3

www.aacrjournals.org Mol Cancer Res; 12(3) March 2014 329

on December 16, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2014; DOI: 10.1158/1541-7786.MCR-13-0543

Page 9: PAI-1 Leads to G -Phase Cell-Cycle Progression through Cyclin … · PAI-1 zymography Of note, 30 mg of total cell lysate from PAI-1 clones (T24Scr, T24-PAI-1KD, UM-UC-14Scr, UM-UC-14-PAI-

800

600

400

200

0

2,000

1,500

1,000

500

0

4,000

3,000

2,000

1,000

0

Tu

mo

r vo

lum

e (

mm

3)

Pro

lifera

tion Index

Pro

lifera

tion Index

Pro

lifera

tion Index

Tu

mo

r vo

lum

e (

mm

3)

T24Scr

T24Scr

T24

Scr

H&E

PAI-1

Cyclin D3

p27 Cip1/Waf1

p21kip1

T24-PAI-1KD-19

T24-PAI-1KD-19

T24-P

AI-

1K

D-1

9

UM-UC-14Scr

UM-UC-14Scr

UM-UC-14-PAI-1KD-4

UM-UC-14-PAI-1KD-4

UM

-UC

-14

Scr

UM

-UC

-14-P

AI-

1K

D-4

HeLaEmpty

HeLaEmpty

HeLa-PAI-1OE-12

HeLa-PAI-1OE

HeLa

Em

pty

HeLa-P

AI-

1O

E-1

2

1 2

Weeks after inoculation

3 4 1 2

Weeks after inoculation

3 4 1 2

Weeks after inoculation

3 4

30

20

10

0

25

20

15

10

5

0

50

40

30

20

10

0

A

B

C

*

*

*

*

**

Giacoia et al.

Mol Cancer Res; 12(3) March 2014 Molecular Cancer Research330

on December 16, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2014; DOI: 10.1158/1541-7786.MCR-13-0543

Page 10: PAI-1 Leads to G -Phase Cell-Cycle Progression through Cyclin … · PAI-1 zymography Of note, 30 mg of total cell lysate from PAI-1 clones (T24Scr, T24-PAI-1KD, UM-UC-14Scr, UM-UC-14-PAI-

T24 and UM-UC-14 knockdown xenografts and HeLaoverexpressor xenografts (Fig. 5B).The expression levels of the proliferative marker (Ki-67)

were also monitored in the excised xenograft tissues by IHC,and a proliferation index was quantified (Fig. 5C). Weobserved a significant reduction in proliferative capacity inT24-PAI-1KD-19 and UM-UC-14-PAI-1KD-4 tumorsdepleted of PAI-1, and an enhanced proliferative capacityin HeLa-PAI-1OE-12. Specifically, T24-PAI-1KD-19showed a reduction in proliferative index of 91% comparedwith T24Scr (P ¼ 0.0007) and UM-UC-14-PAI-1KD-4exhibited a reduction of 72% compared with UM-UC-14Scr

(P < 0.0001). The overexpression of PAI-1 in HeLa–PAI-1OE-12 led to a prominent increase in proliferation of 120%compared withHeLaEmpty control (P¼ 0.0002; Fig. 5C). Inaddition, immunohistochemical analysis confirmed our invitro cell-cycle data. Reduction of a functional PAI-1 in T24-PAI-1KD-19 and UM-UC-14-PAI-1KD-4 was accompaniedby a dramatic reduction in the expression of cyclinD3 and anincrease in the cell-cycle inhibitors, p21Cip1/Waf1 and p27kip1

, alluding to a cell-cycle arrest in the G1-phase. Staining forHeLa-PAI-1OE-12 xenografts is noted in Fig. 5B. Reductionof a functional PAI-1 in T24-PAI-1KD-19 and UM-UC-14-PAI-1KD-4 was not consistently accompanied by a reductionin the expression of cyclin E, cdk2 or p53 (data not shown).Collectively, the xenograft tumor data show that the down-regulation of PAI-1 is associated with a diminution in tumorgrowth through the attenuation of proliferation coupledwith the inhibition of key cyclin/CDK complexes and aninduction of cell-cycle inhibitors involved in G1–S cell-cyclephase progression.

DiscussionPAI-1 is the primary inhibitor of tPA and uPA, and acts to

suppress tissue and plasma fibrinolysis via plasmin conver-sion (25). PAI-1 is known to play a major role in benigndisorders such as deep vein thrombosis, myocardial infarc-tion, atherosclerosis, and stroke (26), and moreover, hasbeen linked to some cancers (27, 28). Although PAI-1activity has been associated with some tumor cell activities(7–9, 29–32), no data have linked PAI-1 to specific changeswith cellular proliferation and the cell cycle to date.Given our compelling data linking PAI-1 expression to cell

proliferation, we focused our studies in further studying therole of PAI-1 in cell-cycle progression. In this study, weexamined the tumorigenic and proliferative effects of PAI-1on human urothelial and cervical cell lines by geneticallyaltering the expression of PAI-1 and by interfering with

PAI-1 activity with the small-molecule inhibitor tiplaxtinin.In both in vitro and in vivo experiments, we found that thedownregulation of PAI-1 led to (i) inhibition in cell prolif-eration and (ii) potent arrest in the G1–S phase of the cellcycle, a phenomena associated with reduction in cyclin D3/cdk4/6 and cyclin E/cdk2 and an increase in cell-cycleinhibitors, p53, p21Cip1/Waf1, and p27kip1.Cell proliferation relies on the activation of cyclins, which

in turn bind to cyclin-dependent kinases (CDK), formingcomplexes that drive progression through the differentphases of the cell cycle, and ultimately leads to cell growth(12, 13). This is coupled to regulatory mechanisms by CDKinhibitors that allow or disallow cells to progress through thecell cycle. Because uncontrolled cyclin/CDK activity is awell-established contributing factor for human cancer pro-gression and tumorigenesis (13), we evaluated whether PAI-1 knockdown in tumor cells would affect cell-cycle devel-opment. Until now, there has been no evidence availableshowing the effects of functional PAI-1 as it relates to cell-cycle progression. Cyclin Dmembers (D1, D2, and D3) areprotooncogenic regulators of theG1–S phase checkpoint andhave been linked with the pathogenesis of several tumortypes (13). In addition, cyclin E and its binding partner cdk2lead to the phosphorylation of downstream targets forgrowth progression (33–35). Our data suggest that PAI-1may confer its protumor properties by causing cell progres-sion through G1-phase via the upregulation of cyclin D3/cdk4/6 and cyclin E/cdk2 complexes.In this study, the impairment of progression through G1

into S-phase in thePAI-1–depleted cells was also accompaniedby prominent increases in the levels of the master cell-cycleregulator p53, and activation of specific downstream targets,including the CDK inhibitors, p21Cip1/Waf1 and p27kip1,which both bind to and inhibit cyclin–CDK complexes toregulate cell-cycle progression through G1-phase. Followingantimitogenic signals, p21Cip1/Waf1 and p27kip1 typically bindto cyclin–CDK complexes to inhibit their catalytic activityand induce cell-cycle arrest (36, 37).Cancer cells are known totightly regulate such cell-cycle inhibitors to prevent arrest andthereby facilitate cell proliferation (38–40). Furthermore, thecyclinE/cdk2complex is known to regulate p27kip1 activity byflagging it for ubiquitination and subsequent degradation,which allows cyclin A expression and progression to S-phase(41). This perhaps explains the observed activation of p27kip1

promoting G1-phase arrest of T24 and UM-UC-14 tumorcells in which PAI-1 was silenced.TGF-b is known to arrest epithelial cells inG1-phase (42),

whereas it is also known as a strong inducer of PAI-1expression (43). Our data clearly illustrate that PAI-1 acts

Figure 5. PAI-1 knockdown results in reduction of xenograft tumor growth. A, tumor growth of human xenografts was assessed in vivo in athymic nudemice inthe PAI-1 T24 cells (T24Scr and T24-PAI-1KD-19) and UM-UC-14 cells (UM-UC-14Scr and UM-UC-14-PAI-1KD-4). In addition, PAI-1–overexpressing cells(HeLa-PAI-1OE-12 and HeLaEmpty) were evaluated (n¼ 10/group). Tumor volumes were measured weekly for 4 weeks and plotted as mean� SD. �, P < 0.05,relative to control. B, tumors were resected, fixed in 10% buffered formalin, and embedded in paraffin. Hematoxylin and eosin (H&E) images are included toidentify and define tumor histology. Immunohistochemical analysis of xenograft tumors (T24Scr, T24-PAI-1KD-19, UM-UC-14Scr, UM-UC-14-PAI-1KD-4,HeLaEmpty, and HeLa-PAI-1OE-12) for PAI-1, cyclin D3, p21Cip1/Waf1, and p27kip1 was conducted. Representative images from the four groups along areillustrated. These in vivo results confirmed our in vitro results; PAI-1 correlates with cyclin D3 expression and inversely correlates with p21Cip1/Waf1 and p27kip1

expression. C, proliferative index was quantified on the basis of Ki-67 staining in each group. Data, mean þ SD. �, P < 0.05, relative to control.

PAI-1 Regulates Cyclin D3

www.aacrjournals.org Mol Cancer Res; 12(3) March 2014 331

on December 16, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2014; DOI: 10.1158/1541-7786.MCR-13-0543

Page 11: PAI-1 Leads to G -Phase Cell-Cycle Progression through Cyclin … · PAI-1 zymography Of note, 30 mg of total cell lysate from PAI-1 clones (T24Scr, T24-PAI-1KD, UM-UC-14Scr, UM-UC-14-PAI-

as a G1-phase cell-cycle promoter and thus, generates someconflict with respect to the effect of TGF-b on cell-cycleprogression. We are in the process of delving into thismechanism, but believe that PAI-1 and TGF-1 interactwith each other via a feedback loop (e.g., elevated TGF-bleads to elevated PAI-1, which then shuts down TGF-b).Furthermore, negative regulators in these pathways havebeen shown to be important, adding complexity and versa-tility to PAI-1 gene regulation (44).Aberrant expression of PAI-1 has been observed in some

tumor types (e.g., breast and pancreatic) and has beenassociated with poor prognosis (45, 46). Little attention hasbeen given to PAI-1 in human bladder or cervical tumors.For example, only two groups have reported on PAI-1 levelsin patients with human bladder cancer. Goodison andcolleagues noted a significant increase in urothelial cellPAI-1 levels in patients bearing bladder tumors comparedwith non–tumor-bearing patients (22), and the same groupreported increased urinary PAI-1 protein levels (23) inpatients with bladder cancer in an independent study.Becker and colleagues reported significantly higher PAI-1levels in tissue and plasma samples, but not in urine, frompatients with bladder cancer compared with controls (47).In cervical cancer, the literature on PAI-1 is even morescant. Kobayashi and colleagues demonstrated a significantlyhigher lymph node–positive rate in patients that had cervicaltumors with strong urokinase plasminogen activator and/orPAI-1 tissue staining, than in those with tumors with weakexpression (48). In another study, Tee and colleagues noted aspecific PAI-1 allelic polymorphism in women with cervicalcancer compared with women without cervical cancer (49).Thus, further exploration of PAI-1 in human tumors iswarranted. Interestingly, PAI-1 expression was noted to beincreased in MIBC compared with NMIBC (Fig. 1F), butno association was noted with tumor grade (Fig. 1G).Furthermore, in cervical cancer, PAI-1 expression was notassociated with higher stage cervical cancer, but was associ-ated with high-grade cervical cancer (Supplementary Fig.S1). This stage- and grade-specific discrepancy could be dueto inherited differences between bladder and cervical tumors,or if larger cohorts were studied, the discrepancies may bereduced or eliminated.Recently, we reported that the reduction or inhibition of

PAI-1 by tiplaxtinin resulted in the reduction of cellularproliferation, cell adhesion, and colony formation, and theinduction of apoptosis and anoikis in vitro. Furthermore,treatment of T24 xenografts with tiplaxtinin resulted ininhibition of angiogenesis and induction of apoptosis, lead-ing to a significant reduction in tumor growth. Similarresults were obtained through evaluation of the humancervical cancer HeLa cell line, showing that PAI-1–mediatedeffects are not restricted to tumor cells of bladder origin (50).

Of note, in our in vitro studies when PAI-1 was silenced, theability of cell to progress through the cell cycle was severelyhampered (Fig. 4). However, we must point out that littleexpression of PAI-1 was evident in xenograft tumors inwhich PAI-1 was silenced. Despite genetically silencing PAI-1, xenograft tumors demonstrated a minimal growth poten-tial over time, not a reduction in growth (Fig. 5). Thus, webelieve that within xenograft tumors there are other factorsthat can help the tumors escape from the effects of silencingPAI-1, because these tumors still show some, albeit reduceproliferative potential. We are currently planning follow-upstudies to closely look into this aspect of the project.Collectively, these data along with the present data showthat targeting PAI-1 may be beneficial and support thenotion that novel drugs such as tiplaxtinin could be inves-tigated as anticancer agents. In this setting, care must beexercised when titrating tiplaxtinin to reduce any unsus-pected bleeding issues.Taken together, the results presented here show the

importance of PAI-1 as it relates to tumor cell proliferation.We are the first to show that cell-cycle arrest associated withthe downregulation of PAI-1 was accompanied by thedepletion of G1 cell-cycle transition complexes in parallelwith the upregulation of key cell-cycle inhibitors. Althoughits function is complex, further investigation into PAI-1 iswarranted in the hope that it can provide insight into specificaspects of tumor biology and the identification of tumor cellvulnerabilities for therapeutic exploitation.

Disclosure of Potential Conflicts of InterestS. Goodison is a CEO and has patent co-ownership in Nonagen Bioscience Corp.

C.J. Rosser is a president and has ownership interest (including patents) in NonagenBioscience Corp—patents on bladder signature. No potential conflicts of interest weredisclosed by the other authors.

Authors' ContributionsConception and design: E.G. Giacoia, S. Goodison, C.J. RosserDevelopment of methodology: E.G. Giacoia, M. MiyakeAcquisition of data (provided animals, acquired and managed patients, providedfacilities, etc.): E.G. Giacoia, A. LawtonAnalysis and interpretation of data (e.g., statistical analysis, biostatistics, compu-tational analysis): E.G. Giacoia, A. Lawton, C.J. RosserWriting, review, and/or revision of the manuscript: E.G. Giacoia, S. Goodison,C.J. RosserAdministrative, technical, or material support (i.e., reporting or organizing data,constructing databases): E.G. Giacoia, M. Miyake, C.J. RosserStudy supervision: S. Goodison, C.J. Rosser

Grant SupportThis work is funded in part by the James and Esther King Biomedical Team Science

Project, 1KT-01 (to C.J. Rosser) and NIH/NCI RO1 CA108597 (to S. Goodison).The costs of publication of this article were defrayed in part by the payment of page

charges. This article must therefore be herebymarked advertisement in accordance with18 U.S.C. Section 1734 solely to indicate this fact.

Received October 11, 2013; revised December 4, 2013; accepted December 27,2013; published OnlineFirst January 24, 2014.

References1. Lee CC, Huang TS. Plasminogen activator inhibitor-1: the expression,

biological functions, and effects on tumorigenesis and tumor celladhesion and migration. J Cancer Mol 2005;1:25–36.

2. Wilkins-Port CE, Freytag J, Higgins SP, Higgins PJ. PAI-1: a multi-functional SERPIN with complex roles in cell signaling and migration.Cell Commun Insights 2010;3:1–10.

Giacoia et al.

Mol Cancer Res; 12(3) March 2014 Molecular Cancer Research332

on December 16, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2014; DOI: 10.1158/1541-7786.MCR-13-0543

Page 12: PAI-1 Leads to G -Phase Cell-Cycle Progression through Cyclin … · PAI-1 zymography Of note, 30 mg of total cell lysate from PAI-1 clones (T24Scr, T24-PAI-1KD, UM-UC-14Scr, UM-UC-14-PAI-

3. Lijnen HR. Pleiotropic functions of plasminogen activator inhibitor-1.Thromb Haemost 2005;3:35–45.

4. Jankun J, Keck RW, Skrzypczak-Jankun E, Swiercz R. Inhibitors ofurokinase reduce size of prostate cancer xenografts in severe com-bined immunodeficient mice. Cancer Res 1997;57:559–63.

5. McMahonGA,PetitclercE, StefanssonS,Smith E,WongMK,WestrickRJ, et al. Plasminogen activator inhibitor-1 regulates tumor growth andangiogenesis. J Biol Chem 2001;276:33964–8.

6. Binder BR, Mihaly J. The plasminogen activator inhibitor "paradox" incancer. Immunol Lett 2008;118:116–24.

7. Soff GA, Sanderowitz J, Gately S, Verrusio E, Weiss I, Brem S, et al.Expression of plasminogen activator inhibitor type 1 by human pros-tate carcinoma cells inhibits primary tumor growth, tumor-associatedangiogenesis, and metastasis to lung and liver in an athymic mousemodel. J Clin Invest 1995;96:2593–600.

8. Ma D, Gerard RD, Li XY, Alizadeh H, Niederkorn JY. Inhibition ofmetastasis of intraocular melanomas by adenovirus-mediated genetransfer of plasminogen activator inhibitor type 1 (PAI-1) in an athymicmouse model. Blood 1997;90:2738–46.

9. Czekay RP, Wilkins-Port CE, Higgins SP, Freytag J, Overstreet JM,Klein RM, et al. PAI-1: an integrator of cell signaling andmigration. Int JCell Biol 2011;2011:562481.

10. Hudson MA, McReynolds LM. Urokinase and the urokinase receptor:association with in vitro invasiveness of human bladder cancer celllines. J Natl Cancer Inst 1997;89:709–17.

11. Vermeulen K, Van Bockstaele DR, Berneman ZN. The cell cycle: areview of regulation, deregulation, and therapeutic targets in cancer.Cell Prolif 2003;36:131–49.

12. Malumbres M, Hunt SL, Sotillo R, Martín J, Odajima J, Martín A, et al.Driving the cell cycle to cancer. Adv Exp Med Biol 2003;532:1–11.

13. Elokdah H, Abou-Gharbia M, Hennan JK, McFarlane G, Mugford CP,KrishnamurthyG, et al. Tiplaxtinin, a novel, orally efficacious inhibitor ofplasminogen activator inhibitor-1: design, synthesis, and preclinicalcharacterization. J Med Chem 2004;47:3491–4.

14. Leik CE, Su EJ, Nambi P, Crandall DL, Lawrence DA. Effect ofpharmacologic plasminogen activator inhibitor-1 inhibition on cellmotility and tumor angiogenesis. J Thromb Haemost 2006;4:2710–5.

15. Miyake M, Goodison S, Giacoia EG, Rizwani W, Ross S, Rosser CJ.Influencing factors on theNMP-22urine assay: anexperimentalmodel.BMC Urol 2012;12:23–7.

16. Cao W, Yacoub S, Shiverick KT, Namiki K, Sakai Y, Porvasnik S, et al.Dichloroacetate (DCA) sensitizes both wild-type and overexpressingBcl-2 prostate cancer cells in vitro to radiation. Prostate 2008;68:1223–31.

17. PfafflMW.A newmathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res 2001;29:e45.

18. Whitfield ML, Sherlock G, Saldanha AJ, Murray JI, Ball CA, AlexanderKE, et al. Identification of genes periodically expressed in the humancell cycle and their expression in tumors. Mol Biol Cell 2002;13:1977–2000.

19. Anai S, Goodison S, Shiverick K, Hirao Y, Brown BD, Rosser CJ.Knock-down of Bcl-2 by antisense oligodeoxynucleotides inducesradiosensitization and inhibition of angiogenesis in human PC-3 pros-tate tumor xenografts. Mol Cancer Ther 2007;6:101–11.

20. Rosser CJ, Liu L, Sun Y, Villicana P, McCullers M, Porvasnik S, et al.Bladder cancer-associated gene expression signatures identified byprofiling of exfoliated urothelia. Cancer Epidemiol Biomarkers Prev2009;18:444–53.

21. Urquidi V,GoodisonS, Cai Y, SunY,RosserCJ. A candidatemolecularbiomarker panel for the detection of bladder cancer. Cancer EpidemiolBiomarkers Prev 2012;21:2149–58.

22. Goodison S, Chang M, Dai Y, Urquidi V, Rosser CJ. A multi-analyteassay for the non-invasive detection of bladder cancer. PLoS ONE2012;7:e47469.

23. Rosser CJ, Ross S, Chang M, Dai Y, Mengual L, Zhang G, et al.Multiplex protein signature for the detection of bladder cancer invoided urine samples. J Urol 2013;5347:4581–3.

24. Wiman B, Collen D. Purification and characterization of human anti-plasmin, the fast-acting plasmin inhibitor in plasma. Eur J Biochem1977;78:19–26.

25. Stefansson S, Haudenschild CC, Lawrence DA. Beyond fibrino-lysis: the role of plasminogen activator inhibitor-1 and vitronectinin vascular wound healing. Trends Cardiovasc Med 1998;8:175–80.

26. Fang H, Placencio VR, DeClerck YA. Protumorigenic activity of plas-minogen activator inhibitor-1 through an antiapoptotic function. J NatlCancer Inst 2012;104:1470–84.

27. Wilkins-Port CE, Ye Q, Mazurkiewicz JE, Higgins PJ. TGF-beta1 þEGF-initiated invasive potential in transformed human keratinocytes iscoupled to a plasmin/MMP-10/MMP-1-dependent collagen remodel-ing axis: role for PAI-1. Cancer Res 2009;69:4081–91.

28. Almholt K, Lund LR, Rygaard J, Nielsen BS, Danø K, Rømer J, et al.Reduced metastasis of transgenic mammary cancer in urokinase-deficient mice. Int J Cancer 2005;113:525–32.

29. Shapiro RL, Duquette JG, Roses DF, Nunes I, Harris MN, Kamino H,et al. Induction of primary cutaneous melanocytic neoplasms inurokinase-type plasminogen activator (uPA)-deficient and wild-type mice: cellular blue nevi invade but do not progress to malignantmelanoma in uPA-deficient animals. Cancer Res 1996;56:3597–604.

30. Sabapathy KT, Pepper MS, Kiefer F, M€ohle-Steinlein U, Tacchini-Cottier F, Fetka I, et al. Polyoma middle T-induced vascular tumorformation: the role of the plasminogen activator/plasmin system. JCellBiol 1997;137:953–63.

31. Praus M, Collen D, Gerard RD. Both u-PA inhibition and vitro-nectin binding by plasminogen activator inhibitor 1 regulateHT1080 fibrosarcoma cell metastasis. Int J Cancer 2002;102:584–91.

32. Stamatakos M, Palla V, Karaiskos I, Xiromeritis K, Alexiou I, Pateras I,et al. Cell cyclins: triggering elements of cancer or not? World J SurgOncol 2010;8:111–4.

33. Donnellan R, Chetty R. Cyclin E in human cancers. FASEB J1999;13:773–80.

34. Malumbres M, Barbacid M. Cell cycle, CDKs, and cancer: a changingparadigm. Nat Rev Cancer 2009;9:153–66.

35. Slingerland J, Pagano M. Regulation of the cdk inhibitor p27 and itsderegulation in cancer. J Cell Physiol 2000;183:10–7.

36. Viglietto G, Motti ML, Fusco A. Understanding p27(kip1) deregulationin cancer: downregulation or mislocalization. Cell Cycle 2002;1:394–400.

37. Nho RS, Sheaff RJ. p27kip1 contributions to cancer. Prog Cell CycleRes 2003;5:249–59.

38. Coqueret O. New roles for p21 and p27 cell-cycle inhibitors: afunction for each cell compartment? Trends Cell Biol 2003;13:65–70.

39. Romanov VS, Pospelov VA, Pospelova TV. Cyclin-dependent kinaseinhibitor p21(Waf1): contemporary view on its role in senescence andoncogenesis. Biochemistry 2012;77:575–84.

40. Sheaff RJ, Groudine M, Gordon M, Roberts JM, Clurman BE.Cyclin E-CDK2 is a regulator of p27Kip1. Genes Dev 1997;11:1464–78.

41. Duffy MJ. Urokinase plasminogen activator and its inhibitor, PAI-1, asprognostic markers in breast cancer: from pilot to level 1 evidencestudies. Clin Chem 2002;48:1194–7.

42. Ewen ME. p53-dependent repression of cdk4 synthesis in transform-ing growth factor-beta-induced G1 cell-cycle arrest. J Lab Clin Med1996;128:355–60.

43. Samarakoon R, Higgins PJ. Integration of non-SMAD and SMADsignaling in TGF-beta1-induced plasminogen activator inhibitortype-1 gene expression in vascular smooth muscle cells. ThrombHaemost 2008;100:976–83.

44. Nagamine Y. Transcriptional regulation of the plasminogen activatorinhibitor type 1–with an emphasis on negative regulation. ThrombHaemost 2008;100:1007–13.

45. Eljuga D, Razumovic JJ, Bulic K, Petrovecki M, Draca N, Bulic SO.Prognostic importance of PAI-1 in node negative breast cancerpatients–results after 10 years of follow-up. Pathol Res Pract2011;207:290–4.

46. Smith R, Xue A, Gill A, Scarlett C, Saxby A, Clarkson A, et al. Highexpression of plasminogen activator inhibitor-2 (PAI-2) is a predictor of

PAI-1 Regulates Cyclin D3

www.aacrjournals.org Mol Cancer Res; 12(3) March 2014 333

on December 16, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2014; DOI: 10.1158/1541-7786.MCR-13-0543

Page 13: PAI-1 Leads to G -Phase Cell-Cycle Progression through Cyclin … · PAI-1 zymography Of note, 30 mg of total cell lysate from PAI-1 clones (T24Scr, T24-PAI-1KD, UM-UC-14Scr, UM-UC-14-PAI-

improved survival inpatientswithpancreatic adenocarcinoma.World JSurg 2007;31:493–502.

47. Becker M, Szarvas T, Wittschier M, vom Dorp F, T€otsch M, SchmidKW, et al. Prognostic impact of plasminogen activator inhibitortype 1 expression in bladder cancer. Cancer 2010;116:4502–12.

48. Kobayashi H, Fujishiro S, Terao T. Impact of urokinase-type plasmin-ogen activator and its inhibitor type 1 onprognosis in cervical cancer ofthe uterus. Cancer Res 1994;54:6539–48.

49. Tee YT, Wang PH, Tsai HT, Lin LY, Lin HT, Yang SF, et al. Geneticpolymorphism of urokinase-type plasminogen activator is interactingwith plasminogen activator inhibitor-1 to raise risk of cervical neopla-sia. J Surg Oncol 2012;106:204–8.

50. Gomes-Giacoia E, Miyake M, Goodison S, Rosser CJ. Targetingplasminogen activator inhibitor-1 inhibits angiogenesis and tumorgrowth in a human cancer xenograft model. Mol Can Ther 2013;12:2697–708.

Giacoia et al.

Mol Cancer Res; 12(3) March 2014 Molecular Cancer Research334

on December 16, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2014; DOI: 10.1158/1541-7786.MCR-13-0543

Page 14: PAI-1 Leads to G -Phase Cell-Cycle Progression through Cyclin … · PAI-1 zymography Of note, 30 mg of total cell lysate from PAI-1 clones (T24Scr, T24-PAI-1KD, UM-UC-14Scr, UM-UC-14-PAI-

2014;12:322-334. Published OnlineFirst January 24, 2014.Mol Cancer Res   Evan Gomes Giacoia, Makito Miyake, Adrienne Lawton, et al.   D3/cdk4/6 Upregulation

-Phase Cell-Cycle Progression through Cyclin1PAI-1 Leads to G

  Updated version

  10.1158/1541-7786.MCR-13-0543doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mcr.aacrjournals.org/content/suppl/2014/01/24/1541-7786.MCR-13-0543.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://mcr.aacrjournals.org/content/12/3/322.full#ref-list-1

This article cites 50 articles, 12 of which you can access for free at:

   

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

  Subscriptions

Reprints and

  [email protected]

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

  Permissions

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

.http://mcr.aacrjournals.org/content/12/3/322To request permission to re-use all or part of this article, use this link

on December 16, 2020. © 2014 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst January 24, 2014; DOI: 10.1158/1541-7786.MCR-13-0543