A glycolytic mechanism regulating an angiogenic switch in prostate cancer

12
A Glycolytic Mechanism Regulating an Angiogenic Switch in Prostate Cancer Jianhua Wang, 1 Jincheng Wang, 1 Jinlu Dai, 4 Younghun Jung, 1 Chuen-Long Wei, 2 Yu Wang, 1 Aaron M. Havens, 1 Phillip J. Hogg, 6 Evan T. Keller, 4 Kenneth J. Pienta, 5 Jacques E. Nor, 2 Cun-Yu Wang, 3 and Russell S. Taichman 1 1 Department of Periodontics and Oral Medicine, 2 Department of Cariology, Restorative Sciences, and Endodontics, and 3 Division of Prosthodontics, Department of Biologic and Materials Sciences, University of Michigan School of Dentistry; 4 Department of Urology and 5 Division of Internal Medicine and Urology, University of Michigan School of Medicine, Ann Arbor, Michigan and 6 Center for Vascular Research, University of New South Wales, Sydney, New South Wales, Australia Abstract The generation of an ‘angiogenic switch’ is essential for tumor growth, yet its regulation is poorly understood. In this investigation, we explored the linkage between metastasis and angiogenesis through CXCL12/CXCR4 signaling. We found that CXCR4 regulates the expression and secretion of the glycolytic enzyme phosphoglycerate kinase 1 (PGK1). Over- expression of PGK1 reduced the secretion of vascular endothelial growth factor and interleukin-8 and increased the generation of angiostatin. At metastatic sites, however, high levels of CXCL12 signaling through CXCR4 reduced PGK1 expression, releasing the angiogenic response for metastastic growth. These data suggest that PGK1 is a critical downstream target of the chemokine axis and an important regulator of an ‘angiogenic switch’ that is essential for tumor and metastatic growth. [Cancer Res 2007;67(1):149–59] Introduction Hypoxia is a central feature of many solid tumors, including prostate cancer, making them extremely resistant to chemotherapy. Transcriptional activation of hypoxia-inducible genes are known to play a critical first step in the adaptation of tumors to hypoxia, including positive [vascular endothelial growth factor (VEGF), interleukin (IL)-8, and SDF-1/(CXCL12)] and negative regulation of the angiogenic cascade. Hypoxia-inducible genes also regulate many glycolytic enzymes to facilitate anaerobic metabolism. These molecules are required to provide for the high energetic demands of growing neoplasms and to generate macromolecules critical for growth (1). Consequently, the shift in energy production from oxidative phosphorylation to glycolysis, the so-called ‘‘Warburg effect,’’ is likely to be a fundamental property of cancer cells and is known to correlate with enhanced tumor progression (2–4). Phosphoglycerate kinase (PGK) 1 is an ATP-generating glycolytic enzyme that forms part of the glycolytic pathway. PGK1 is regulated by hypoxia-inducible factor-1a (HIF-1a; ref. 5) and it is often overexpressed in prostate cancer (6). PGK1 is located in a chromosomal region associated with familial prostate cancers, hypospadias, and androgen insensitivity (X chromosome; Xq11- Xq13; ref. 7). In the nucleus, PGK1 influences DNA replication and repair. Surprisingly, PGK1 is secreted extracellularly by tumors, where it acts as a disulfide reductase capable of cleaving plasmi- nogen to generate the vascular inhibitor angiostatin (8–13). Overexpression of PGK1 is therefore likely to restrict tumor growth by limiting angiogenesis. Thus, a delicate equilibrium may be established between the hypoxic response needed to generate proangiogenic factors and events essential for anaerobic metabo- lism (e.g., PGK1) for tumors to grow. The challenge is to regulate PGK1 secretion. Our previous work has focused on the role that chemokine axis of CXCL12 and its receptor CXCR4 plays in metastatic prostate cancer (14–16). We observed that CXCR4 expression is related to increasing tumor grade (14) and that CXCL12 signaling through CXCR4 triggers the adhesion of prostate cancers to bone marrow endothelial cells, possibly by activating a v h 3 integrins (17) and CD164 (18). Most critically, antibody to CXCR4 significantly reduces the number of prostate cancer bone metastasis in an in vivo model of metastasis (15). Further work has shown that CXCR4 signaling disrupts the delicate equilibrium between proangiogenic and antiangiogenic signals in prostate cancer disease (16). In the present investigation, we found that CXCR4 signaling regulates PGK1 expression, which regulates angiogenesis by generating angiostatin and by reducing the secretion of the proangiogenic cytokines VEGF and IL-8. At sites of high CXCL12 production, however (bone, lymph node, and liver), PGK1 secretion is inhibited by the CXCL12/CXCR4 axis. Thus, CXCL12 signaling through CXCR4 generates an ‘angiogenic switch’ necessary for metastatic growth. Together, these data further show that CXCL12/ CXCR4 chemokine axis and PGK1 represent at least one of the critical events necessary for metastasis of prostate cancer as well as a mechanism for a proangiogenic switch to promote tumor growth. These findings add a new dimension to our understanding of the relationship between chemokines and metabolism. Moreover, they point to potential therapeutic interventions to disrupt these pathways and control prostate cancer growth and its metastasis. Materials and Methods Cell cultures. The LNCaP, LNCaP metastatic subline C4-2B and PC3, and MCF-7 breast cancer cells were obtained from the American Type Culture Collection (Rockville, MD) and cultured in RPMI 1640 supplemented with 10% fetal bovine serum (FBS; Invitrogen Corp., Carlsbad, CA). The human prostate epithelial cell line (RWPE-1) was cultured in keratinocyte medium supplemented with 5 ng/mL human recombinant epithelial growth factor and 50 Ag/mL bovine pituitary extract (Invitrogen). Human dermal microvascular endothelial cells (HDMEC) were grown as described previously (15, 19, 20). Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Requests for reprints: Russell S. Taichman, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Avenue, Ann Arbor, MI 48109-1078. Phone: 734-764-9952; Fax: 734-763-5503; E-mail: rtaich@ umich.edu. I2007 American Association for Cancer Research. doi:10.1158/0008-5472.CAN-06-2971 www.aacrjournals.org 149 Cancer Res 2007; 67: (1). January 1, 2007 Research Article Research. on January 31, 2016. © 2007 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Transcript of A glycolytic mechanism regulating an angiogenic switch in prostate cancer

A Glycolytic Mechanism Regulating an Angiogenic

Switch in Prostate Cancer

Jianhua Wang,1Jincheng Wang,

1Jinlu Dai,

4Younghun Jung,

1Chuen-Long Wei,

2

Yu Wang,1Aaron M. Havens,

1Phillip J. Hogg,

6Evan T. Keller,

4Kenneth J. Pienta,

5

Jacques E. Nor,2Cun-Yu Wang,

3and Russell S. Taichman

1

1Department of Periodontics and Oral Medicine, 2Department of Cariology, Restorative Sciences, and Endodontics, and 3Division ofProsthodontics, Department of Biologic and Materials Sciences, University of Michigan School of Dentistry; 4Department ofUrology and 5Division of Internal Medicine and Urology, University of Michigan School of Medicine, Ann Arbor, Michiganand 6Center for Vascular Research, University of New South Wales, Sydney, New South Wales, Australia

Abstract

The generation of an ‘angiogenic switch’ is essential for tumorgrowth, yet its regulation is poorly understood. In thisinvestigation, we explored the linkage between metastasisand angiogenesis through CXCL12/CXCR4 signaling. We foundthat CXCR4 regulates the expression and secretion of theglycolytic enzyme phosphoglycerate kinase 1 (PGK1). Over-expression of PGK1 reduced the secretion of vascularendothelial growth factor and interleukin-8 and increasedthe generation of angiostatin. At metastatic sites, however,high levels of CXCL12 signaling through CXCR4 reduced PGK1expression, releasing the angiogenic response for metastasticgrowth. These data suggest that PGK1 is a critical downstreamtarget of the chemokine axis and an important regulator of an‘angiogenic switch’ that is essential for tumor and metastaticgrowth. [Cancer Res 2007;67(1):149–59]

Introduction

Hypoxia is a central feature of many solid tumors, includingprostate cancer, making them extremely resistant to chemotherapy.Transcriptional activation of hypoxia-inducible genes are knownto play a critical first step in the adaptation of tumors to hypoxia,including positive [vascular endothelial growth factor (VEGF),interleukin (IL)-8, and SDF-1/(CXCL12)] and negative regulation ofthe angiogenic cascade. Hypoxia-inducible genes also regulatemany glycolytic enzymes to facilitate anaerobic metabolism. Thesemolecules are required to provide for the high energetic demandsof growing neoplasms and to generate macromolecules critical forgrowth (1). Consequently, the shift in energy production fromoxidative phosphorylation to glycolysis, the so-called ‘‘Warburgeffect,’’ is likely to be a fundamental property of cancer cells and isknown to correlate with enhanced tumor progression (2–4).Phosphoglycerate kinase (PGK) 1 is an ATP-generating glycolytic

enzyme that forms part of the glycolytic pathway. PGK1 isregulated by hypoxia-inducible factor-1a (HIF-1a; ref. 5) and it isoften overexpressed in prostate cancer (6). PGK1 is located in achromosomal region associated with familial prostate cancers,hypospadias, and androgen insensitivity (X chromosome; Xq11-

Xq13; ref. 7). In the nucleus, PGK1 influences DNA replication andrepair. Surprisingly, PGK1 is secreted extracellularly by tumors,where it acts as a disulfide reductase capable of cleaving plasmi-nogen to generate the vascular inhibitor angiostatin (8–13).Overexpression of PGK1 is therefore likely to restrict tumor growthby limiting angiogenesis. Thus, a delicate equilibrium may beestablished between the hypoxic response needed to generateproangiogenic factors and events essential for anaerobic metabo-lism (e.g., PGK1) for tumors to grow. The challenge is to regulatePGK1 secretion.Our previous work has focused on the role that chemokine axis

of CXCL12 and its receptor CXCR4 plays in metastatic prostatecancer (14–16). We observed that CXCR4 expression is related toincreasing tumor grade (14) and that CXCL12 signaling throughCXCR4 triggers the adhesion of prostate cancers to bone marrowendothelial cells, possibly by activating avh3 integrins (17) andCD164 (18). Most critically, antibody to CXCR4 significantly reducesthe number of prostate cancer bone metastasis in an in vivo modelof metastasis (15). Further work has shown that CXCR4 signalingdisrupts the delicate equilibrium between proangiogenic andantiangiogenic signals in prostate cancer disease (16).In the present investigation, we found that CXCR4 signaling

regulates PGK1 expression, which regulates angiogenesis bygenerating angiostatin and by reducing the secretion of theproangiogenic cytokines VEGF and IL-8. At sites of high CXCL12production, however (bone, lymph node, and liver), PGK1 secretionis inhibited by the CXCL12/CXCR4 axis. Thus, CXCL12 signalingthrough CXCR4 generates an ‘angiogenic switch’ necessary formetastatic growth. Together, these data further show that CXCL12/CXCR4 chemokine axis and PGK1 represent at least one of thecritical events necessary for metastasis of prostate cancer as well asa mechanism for a proangiogenic switch to promote tumor growth.These findings add a new dimension to our understanding of therelationship between chemokines and metabolism. Moreover, theypoint to potential therapeutic interventions to disrupt thesepathways and control prostate cancer growth and its metastasis.

Materials and Methods

Cell cultures. The LNCaP, LNCaP metastatic subline C4-2B and PC3, andMCF-7 breast cancer cells were obtained from the American Type Culture

Collection (Rockville, MD) and cultured in RPMI 1640 supplementedwith 10% fetal bovine serum (FBS; Invitrogen Corp., Carlsbad, CA). The

human prostate epithelial cell line (RWPE-1) was cultured in keratinocyte

medium supplemented with 5 ng/mL human recombinant epithelial growthfactor and 50 Ag/mL bovine pituitary extract (Invitrogen). Human dermalmicrovascular endothelial cells (HDMEC) were grown as described

previously (15, 19, 20).

Note: Supplementary data for this article are available at Cancer Research Online(http://cancerres.aacrjournals.org/).

Requests for reprints: Russell S. Taichman, Department of Periodontics and OralMedicine, University of Michigan School of Dentistry, 1011 North University Avenue,Ann Arbor, MI 48109-1078. Phone: 734-764-9952; Fax: 734-763-5503; E-mail: [email protected].

I2007 American Association for Cancer Research.doi:10.1158/0008-5472.CAN-06-2971

www.aacrjournals.org 149 Cancer Res 2007; 67: (1). January 1, 2007

Research Article

Research. on January 31, 2016. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

PGK and CXCR4 expression constructs. A 1.33-kb human PGK (hPGK)cDNA was isolated by reverse transcription-PCR from total RNA extracted

from PC3 cells. The forward and reverse primers were 5¶-AGTACA-TATGTCGCTTTCTAACAAGCTG-3¶ (positions, 80-100) and 5¶-AGTAGGAT-CCCTAATGCCAAGTGGAGATGCA-3¶ (positions, 1,409-1,389), respectively.The hPGK cDNA containing the open reading frame of hPGK was cloned

to pcDNA3.1D/V5-His-TOPO vector. Cells were transfected with the PGK

cDNA in pcDNA3.1D/V5-His-TOPO, pcDNA3.1-CXCR4-Tagged 3� hemag-

glutinin (HA), or an empty vector by lipofection (16).Small interfering RNA knockdown of PGK1. Short hairpin small

interfering RNAs (siRNA) under the control of the polymerase -IIIH1-RNA

promoter were used for these studies (Oligoengine, Seattle, WA) as

described previously for CXCR4 (16). For siRNA knockdown of PGK1,

two groups of primers corresponding to nucleotide sequences in open

reading frame were synthesized [position, 186-203; Si1, 5¶-gatccccACA-ACCAGAGGATTAAGGCttcaagagaGCCTTAATCCTCTGGTTGTttttta-3¶( forward oligonucleotide) and 5¶-agcttaaaaaACAACCAGAGGATTAAGGCt-ctcttgaaGCCTTAATCCTCTGGTTGTggg-3¶ (reverse oligonucleotide); position,192-210; Si2, 5¶-gatccccAGAGGATTAAGGCTGCTGTttcaagagaACAGCAG-CCTTAATCCTCTttttta-3¶ ( forward oligonucleotide) and 5¶-agcttaaaaa-AGAGGATTAAGGCTGCTCTtctcttgaaACAGCAGCCTTAATCCTCTggg-3¶(reverse oligonucleotide)]. A group scrambled primers [5¶-gatccccAAAACC-GACGGCTATCTCTttcaagagaAGAGATAGCCGTCGGTTTTttttta-3¶ ( forwardoligonucleotide) and 5¶-agcttaaaaaAAAACCGACGGCTATCTCTtctcttgaa-AGAGATAGCCGTCGGTTTTggg-3¶ (reverse oligonucleotide)] were used.

To generate stable clones, the cells were transfected by empty pSUPER

vector, scrambled vector, or PGK1-Si (Si1 and Si2) vector by lipofection (16).

The treated cells were selected in f500 Ag/mL G418 (Invitrogen), and theresulting clones were screened by Western blot. Further subcloning was

done by limiting dilution at a density of 0.2 cells per well after verifying that

only 1 cell per well had been plated.

Indirect immunofluorescence. PC3 cells were fixed and permeabilizedusing Fix and Perm kit according to the manufacturer’s protocol (Caltag

Laboratories, Burlingame, CA). Nonspecific staining was blocked with 10%FBS in permeabilization buffer and anti-PGK1 sheep IgG (1:200), a-tubulin(1:200; Sigma Chemical Co., St. Louis, MO), and sheep IgG (1:100; Upstate,

Lake Placid, NY) antibodies were used followed by anti-mouse fluorescein-conjugated antibody (1:200; Upstate) and anti-mouse rhodamine-conjugated

antibody (1:200; Upstate), each used for 1 h in sequence. The samples were

then fixed in Slow and Fade light anti-fade kit (Molecular Probes, Eugene,

OR). Images were acquired with a Zeiss LSM510 (Thorwood, NY).Western blot analysis. Prostate cancer cells were cultured to

confluence, washed, and then serum starved in RPMI 1640 with 0.1%

bovine serum albumin for 24 h (16). PGK1 detection was done using a

monoclonal IgM antibody at 1:5,000 (1 Ag/mL) dilution (11). Detection ofCXCR4 or angiostatin was similarly done using 5% milk with antibody to

CXCR4 (1:1,000; Abcam, Inc., Cambridge, MA) or a mouse anti-human

monoclonal antibody (1 Ag/mL) angiostatin (1:1,000; Sigma Chemical) withanti-species–specific horseradish peroxidase–conjugated antibodies (Sigma

Chemical).

The endothelial sprout network formation assay in Matrigel. Growthfactor-reduced Matrigel was placed into eight-chamber slides (125 AL/chamber; BD Biosciences, San Diego, CA) and 0.8 � 104 endothelial cells

were added on top. The chambers were then incubated at 37jC for 24 h.

Figure 1. Reciprocal regulation of PGK1 expression by CXCR4 and CXCL12 in prostate cancer cells. A, stable siRNA targeting of CXCR4 (or a scrambled control) inLNCaP, C4-2B, and PC3 cells. Western blotting of whole-cell lysates was done with an antibody to CXCR4 to show the effectiveness of the down-regulation bysiRNA targeting. The blot was stripped and reprobed with anti–h-actin antibody to confirm equal protein loading. Quantitative densitometry of independent investigationswere used for direct comparison of the treatment. *, P < 0.05, significant difference from the controls. Bars, SD. B, indirect immunofluorescence for PGK1 in the prostatecancer cell lines. Representative images of a-tubulin (red) and PGK1 (green ) in PC3 cells transfected with a control or siRNA targeting vector to CXCR4. Originalmagnification, �40. Bar , 100 Am.

Cancer Research

Cancer Res 2007; 67: (1). January 1, 2007 150 www.aacrjournals.org

Research. on January 31, 2016. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Figure 1 Continued. C, Western blot analysis of PGK1 expression in PC3-, LNCaP-, and C4-2B–expressing control or siRNA to CXCR4 vector (left ). In somecases, the cells were starved (48 h) and stimulated with 200 ng/mL CXCL12 for 0 or 24 h at 37jC (left). A time course of PGK1 expression in response to200 ng/mL CXCL12 (right ). D, Western blot analysis of CXCR4 expression in prostate cancer cells overexpressing PGK1 or where PGK1 was targeted by siRNA.The data show that signaling through or expression of CXCR4 negatively regulates PGK1 expression, whereas PGK1 expression regulates CXCR4 expression.Quantitative densitometry was used for direct comparison. *, P < 0.05, significant difference from the controls. Bars , SD.

PGK1 Regulates an Angiogenic Switch

www.aacrjournals.org 151 Cancer Res 2007; 67: (1). January 1, 2007

Research. on January 31, 2016. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

After incubation, the slides were fixed with methanol and stained with Diff-Quick solution II (Sigma Chemical). The slides were examined, and the

sprouts were counted from five random fields under a microscope (�200).For coculture assays, equal numbers of prostate cancers and endothelial

cells were added together, or 500 AL of the prostate cancer conditionedmedium or control were added daily.

Cytokine and PGK1 ELISA analysis. Antibody sandwich ELISAs wereused to evaluate IL-6, IL-8, and VEGF in the prostate cancer conditioned

medium (R&D Systems, Minneapolis, MN). PGK1 levels were determined bydirect ELISA using 100 AL of 5 Ag/mL murine anti-PGK monoclonal IgM

against a recombinant hPGK1 (rhPGK1) standard as described (8, 16).

PGK1, IL-6, IL-8, and VEGF levels were normalized to total protein in the

conditioned medium (Sigma Chemical).Intracardiac, s.c., and intratibial injections. Intracardiac studies, (s.c.)

and intratibial (i.t.) injections were done to determine the role that PGK1

plays in metastasis (15). After 4 weeks, bioluminescence was used to followthe prostate cancer-derived bone metastases as the primary outcome. The

mice were injected i.p. with luciferin (100 AL at 40 mg/mL in PBS) beforeXenogen IVIS (Alameda, CA) (15). Short-term homing capabilities of

PC3PGK1 Luc or PC3Control Luc cells were evaluated at 24 h by real-time PCRfor luciferase 2CP gene [luc2CP, CGGCTGGCAGAAGCTATGAA ( forward),

TCGCTGCACACCACGAT (reverse), and 5¶-FAM-CTATGGGCTGAATA-CAAACC (TaqMan probe; ABI, Foster City, CA)]. Data were normalized to

mouse tissue h-actin (mm00607939-s1).Cell implants and tibias were fixed in 10% formalin at 4jC. Tibias were

further decalcified in 10% EDTA (pH 7.4) for 10 days and embedded in

paraffin. Longitudinal sections of tissues were cut and stained with H&E forhistologic evaluation. Where indicated, histomorphometric analyses of the

samples were done using a computer-assisted bone histomorphometric

analyzing system (Image-Pro Plus version 4.0, Media Cybernetics, Silver

Spring, MD). The numbers of stained microvessels were blindly counted in10 random fields per implant at 100 and 200 magnifications. Four or five

implants were analyzed per condition following immunostaining for human

von Willebrand factor (28 mg/mL anti-rabbit IgG; DAKO North America,

Inc., Carpinteria, CA; ref. 16).Statistical analysis. Numerical data are expressed as mean F SD.

Statistical differences between the means for the different groups were

evaluated with Instat 4.0 (GraphPad Software, San Diego, CA) using one-wayANOVA, with the level of significance at P < 0.05.

Results

Identification of PGK1. We reported previously the generationof cell lines, which stably express siRNAs to reduce CXCR4expression (Fig. 1A ; ref. 16). To further examine the differentialexpression of proteins regulated by CXCR4 in prostate cancers,a proteomic approach was taken. High-resolution two-dimensionalPAGE analysis of proteins derived from scramble or vector controlor cell lines with the reduced CXCR4 expression were examined.More than 500 individual protein signatures with pIs between3.5 and 10 and relative molecular masses between 20 and 55 kDawere detected (Supplementary Fig. S1). A computer-assisted com-parative analysis of the silver-stained patterns of paired cell linesamples identified several proteins that were increased in theCXCR4 down-regulated cells compared with controls. Theseprotein spots were excised from the gels and analyzed byMALDITOF-mass spectrometry. Two of the proteins, in whichpeptides were identified from the tryptic gel digests, had thesequence VLNNMEIGTSLFDEEGAK. A Matrix Science MascotSearch and BLAST analysis identified the sequence belonging totwo isoforms of PGK1.PGK1 expression is regulated by CXCL12. To confirm that the

CXCL12/CXCR4 axis regulates PGK1 expression, indirect immuno-fluorescence and Western blot analysis were done. Strongmembrane and cytoplasmic staining of PGK1 was observed in

cells expressing SiRNA targeting CXCR4 (Fig. 1B). Western blots ofprostate cancer cell lysates also show significant elevations ofPGK1 expression in the SiRNA cells compared with the respectivecontrols (Fig. 1C, left). Stimulation of wild-type PC3 cell lines withCXCL12 decreased PGK1 expression by as early as 30 min for PC3cells (Fig. 1C, right). The C4-2B cells also decreased their expressionof PGK1 in response to CXCL12, but the response was not as robustor rapid as seen for PC3 cells (Fig. 1C, right).PGK1 and CXCR4 reciprocally regulate each other expres-

sion. As shown, down-regulation of CXCR4 expression or CXCL12stimulation of prostate cancer cells alters PGK1 expression. Todetermine if overexpression of CXCR4 directly regulates PGK1expression, a HA-tagged vector containing the full-length cDNAof CXCR4 was used for transfection of the prostate cancer celllines. Overexpression of CXCR4 resulted in down-regulation ofPGK1 in the prostate cancer cell lines (Supplementary Fig. S2A).To determine whether CXCR4 and PGK1 reciprocally regulateeach other, stable transfection of the prostate cancer cells withPGK1 was done. Overexpression of PGK1 increased theexpression of CXCR4 in all of the prostate cancer cell lines(Fig. 1D, left). To determine if inhibition of PGK1 regulatesCXCR4 expression, siRNA was used to generate stable clones withdecreased PGK1 expression. Following selection, PGK1 levels wereevaluated and individual clones were selected and pooled.Inhibition of PGK1 reduced the expression of CXCR4 at bothlevel of transcription (data not shown) and protein level (Fig. 1D,right).Because elevated PGK1 levels are a feature of the neoplastic

transformation, the effects of overexpression of PGK1 on CXCR4expression were also evaluated in the normal human prostateepithelial cell line, RWPE-1. Overexpression of PGK1 enhancedthe expression of CXCR4 in the RWPE-1 cell line (SupplementaryFig. S2B). Similar alterations in CXCR4 expression were observedby overexpressing PGK1 in the breast cancer cell line MCF-7(Supplementary Fig. S2B). Likewise, siRNA targeting of CXCR4increased PGK1 expression in both cell lines (SupplementaryFig. S2C). Together, these findings suggest that signaling throughor expression of CXCR4 inversely correlates with PGK1 expres-sion, whereas PGK1 expression directly correlates with CXCR4expression.Downstream targets of PGK1 were next evaluated using gene

expression arrays. Overexpression of PGK1 in prostate cancer celllines induced several molecules involved in adhesion (E-cadherinand h-catenin), angiogenesis (thrombospondin-1, KILLER/deathreceptor 5/tumor necrosis factor–related apoptosis-inducing ligandreceptor 2, epidermal growth factor receptor, and VEGF), apoptosis(APAF1, Bax, BIRC5, and telomerase), and transcriptional regula-tion [RASA1, InB kinase-a (IKK-a), and c-Src; SupplementaryTable S1]. Western blots confirmed that the expression ofE-cadherin and h-catenin were down-regulated in LNCaP-PGK1and C4-2B-PGK1 cell lines compared with their respective controls.These data suggest that overexpression of PGK1 reduces tumorcell-cell adhesiveness, suggesting that invasion and metastasisregulated by PGK1 may be a possibility (Supplementary Fig. S3A).Most germane to the present investigations were the findingsthat regulation of CXCR4 expression by PGK1 links with extra-cellular signal-regulated kinase (ERK) 1/2 signaling (SupplementaryFig. S3B) and activation of IKK-a (data not shown). These datasuggest that regulation of CXCR4 expression by PGK1 may involveboth mitogen-activated protein/ERK kinase/ERK signaling cascadeand nuclear factor-nB activation.

Cancer Research

Cancer Res 2007; 67: (1). January 1, 2007 152 www.aacrjournals.org

Research. on January 31, 2016. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Overexpression of PGK1 increases themetastatic rate in vivo.Previously, we showed that antibody to CXCR4 blocked prostatecancer cell adhesion to endothelial cells and invasion into extra-cellular matrices. Most importantly, antibody to CXCR4 blockedprostate cancer cell metastasis in an animal model (14, 21).Therefore, overexpression of PGK1 should increase the metastaticrate due to its effect on enhanced CXCR4 expression. To test thishypothesis, luciferase-labeled control (PC3Control) and PGK1-over-expressing cells (PC3PGK1) were injected into the left cardiac ven-tricle of nude mice (Fig. 2A). The short-term homing capabilities ofthe prostate cancer cells were assessed in a variety of tissues at24 h by real-time PCR. The ability to form metastatic lesions wasalso determined at 1 month by Xenogen imaging (Fig. 2A).At 24 h, fewer PC3PGK1 Luc cells were recovered from the

peripheral blood than the PC3Control Luc cells (Fig. 2B). At the sametime, more cells overexpressing PGK1 were found in the heart,spinal, and tibial marrow than control cells (Fig. 2B). Thesefindings roughly correlate to the levels of CXCL12 found in murinetissues (14). At 1 month, bioluminescence imaging showed that

most of the animals treated with either of the PC3 cells haddeveloped metastatic tumors. Quite unexpectedly, however, thetotal metastatic burden of the animals receiving PC3PGK1 Luc

cells was considerably less than those animals injected withPC3Control Luc cells (Fig. 2C). Together, these data suggest thatoverexpression of PGK1 enhanced the metastatic homing butinterfered with the ability of the metastastic cells to grow.Accordingly, proliferation assays were done, which showed thatPGK1 levels in culture did not correlate directly with proliferativeactivity in the various cell lines (data not shown).PGK1 regulates an angiogenic phenotype in vitro. The effects

of PGK1 on metastastic rates suggest that overexpression of PGK1may lead to a diminished ability to recruit a vasculature necessaryto support metastasis while increasing the expression of CXCR4necessary for metastatic homing (14). Previously, we and othershave reported that changes in CXCR4 levels alter the secretion ofIL-6, IL-8, tissue inhibitor of metalloproteinase-2, and VEGF byprostate cancer cells (16, 22–24). To further determine if PGK1regulates an angiogenic phenotype, conditioned medium derived

Figure 2. PGK1 regulates metastasis in vivo .Intracardiac injections of PC3 cells intononobese diabetic/severe combinedimmunodeficient (NOD/SCID) animals weredone using cells that overexpressed PGK1(PC3PGK1 Luc) or a control vector (PC3Control Luc)and tracked by luciferase activity.A, experimental scheme. B, the short-termhoming capabilities of the prostate cancer wereevaluated at 24 h. The level of engraftmentwas assessed by real-time PCR for luciferaseand data were normalized to total mousetissue h-actin. C, the long-term effects of PGK1on total prostate cancer metastasis weredetermined at 4 weeks using a Xenogen IVISSystem. *, P < 0.05 (significant difference fromcontrols); #, P < 0.01. The data show that PGK1regulates early homing of prostate cancer,possibly by up-regulating CXCR4 but negativelyaffecting metastatic development.

PGK1 Regulates an Angiogenic Switch

www.aacrjournals.org 153 Cancer Res 2007; 67: (1). January 1, 2007

Research. on January 31, 2016. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Figure 3. The effect of PGK1 on an angiogenic phenotype in vitro . The levels of IL-6, IL-8, VEGF, and PGK1 were determined at 48 h by ELISA in the conditionedmedium derived from (A) PC3 cells that overexpressed PGK1 or control vectors or for cells expressing a siRNA targeting PGK1 or a scramble vector control.Columns, mean of triplicate determinations and normalized against total protein; bars, SD. *, P < 0.05, significant difference from controls. The data show thatoverexpression of PGK1 reduces the secretion of proangiogenic factors by prostate cancer cell lines, whereas reducing PGK1 expression increases their secretion.B, secreted PGK1 levels were evaluated by ELISA for prostate cancer cells overexpressing CXCR4 or a siRNA targeting CXCR4 or a scramble vector control at 48 h.The data show that signaling through or the expression of CXCR4 reduces the secretion of PGK1. C, prostate cancer cells were cultured for 12 h in the presence orabsence of CXCL12 (100 ng/mL), with or without Co2+ (400 Amol/L; Sigma Chemical) as a mimic for hypoxic conditions. The data show that CXCL12 signalingdecreases PGK1 secretion even under hypoxic conditions. D, angiostatin levels were determined by Western blot for prostate cancer cells overexpressing PGK1,in prostate cancer cell lines with reduced PGK1 expression by siRNA targeting compared with a scrambled vector control (scramble ; top left) and in prostate cancer celllines overexpressing CXCR4 or control vectors (bottom left ). Angiostatin levels in normal prostate epithelium (RWPE) or MCF-7 breast cancer cells transientlyoverexpressing PGK1 levels (bottom right ). Quantitative densitometry of independent investigations was used for direct comparison of the treatment (top right andbottom right ). *, P < 0.05, significant difference from the controls. The data show that PGK1 levels correspond with increased angiostatin levels.

Cancer Research

Cancer Res 2007; 67: (1). January 1, 2007 154 www.aacrjournals.org

Research. on January 31, 2016. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

from cells overexpressing PGK1 was evaluated for alterations inIL-6, IL-8, and VEGF levels. PC3 and C4-2B cells that overexpressedPGK1 secreted less VEGF into their conditioned medium than didthe parental or control cells (Fig. 3A ; data not shown in C4-2Bcells). At the same time, it was observed that elevated PGK1 levelscorrelated with reductions of IL-6 and IL-8 levels by PC3 cells(Fig. 3A). Conversely, PC3 cells expressing a siRNA that targetedPGK1 secreted more IL-6, IL-8, and VEGF into their conditionedmedium compared with controls (Fig. 3A).Secreted PGK1 results in enhanced angiostatin generation.

Our previous work linked CXCR4 signaling with the generation ofangiostatin in vitro (16), and secreted PGK1 is known to function

extracellularly as a disulfide reductase–cleaving plasminogen togenerate angiostatin (8–11). To explore the possibility that theCXCL12/CXCR4 axis alters angiostatin levels through secretedPGK1, the levels of secreted PGK1 were evaluated in theconditioned medium of the prostate cancer cell lines. As shownin Fig. 3B , reducing CXCR4 expression resulted in enhancedsecretion of PGK1 into the conditioned medium. Overexpressionof CXCR4, however, resulted in decreased PGK1 levels detected inthe conditioned medium (Fig. 3B).As PGK1 is regulated by both HIF-1a and CXCL12, we next

evaluated whether the secretion of PGK1 is regulated by either ofthese factors. For these studies, prostate cancer cell lines were

Figure 4. PGK1 levels alter angiogenesisin vitro . HDMECs were plated in growthfactor–reduced Matrigel as a functional,in vitro assay of blood vessel formation.After 24 h, the cultures were fixedand stained. A, left, PC3 cells thatoverexpressed PGK1 or control vectors,or cancer cells expressing a siRNAtargeting PGK1 expression (or a scrambledcontrol) were co-plated with either theHDMECs or their conditioned medium;right, vessel sprout formation wasquantified by direct microscopic counting.B, the vascular response to PC3 cellsoverexpressing CXCR4 or stimulated with200 ng/mL CXCL12 was reversed byrhPGK1 (2,000 ng/mL; top ) and quantified(bottom ). Columns, mean of triplicatedeterminations; bars, SD. *, P < 0.05,significant difference from control. Originalmagnification, �10. Bar, 200 Am.

PGK1 Regulates an Angiogenic Switch

www.aacrjournals.org 155 Cancer Res 2007; 67: (1). January 1, 2007

Research. on January 31, 2016. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

cultured in the presence or absence of CXCL12 with or withoutCo2+ (400 Amol/L) at 12 h to mimic hypoxic conditions. In eachcase, hypoxic conditions enhanced the secretion of PGK1 into theconditioned medium (Fig. 3C). Under these conditions, CXCL12reduced the secretion of PGK1 (Fig. 3C).Next, it was determined whether the expression of PGK1

corresponds with the generation of the vascular inhibitor angios-tatin. As predicted, the overexpression of PGK1 resulted in elevatedangiostatin levels (Fig. 3D, top left). Decreasing PGK1 expression bysiRNA resulted in reduced levels of angiostatin (Fig. 3D, top right).Similarly, overexpression of CXCR4 (Supplementary Fig. S2A)resulted in less angiostatin generated (Fig. 3D, bottom left). Theeffect of overexpression of PGK1 on angiostatin levels was alsoevaluated in RWPE-1 under normoxic conditions. As shown inFig. 3D (bottom right), overexpression of PGK1 enhanced the expres-sion of angiostatin by the RWPE-1 cell line and the MCF-7 cell line.PGK1 levels alter angiogenesis in vitro . To examine whether

the effect of secreted PGK1 has a biologically relevant effect on

endothelial cells, human microvascular endothelial cell migrationthrough Matrigel membranes was examined as a functional,in vitro assay measuring proangiogenic capacity (25). As shownin Fig. 4A , little or no endothelial sprout formation took place inthe absence of external stimuli. Coculture of the endothelial cellswith either conditioned medium of the PC3Control cells or thecells themselves stimulated robust sprout formation. Over-expression of PGK1 in PC3 cells (PC3PGK1) dramatically reversedthe activity on sprout formation of the parental cells. Down-regulation of PGK1 by siRNA had a stimulatory effect on bloodvessel tube formation (Fig. 4A). In parallel, overexpression ofCXCR4 or stimulation of PC3 cells with CXCL12 enhancedendothelial vessel formation that was reversed by the addition ofrhPGK1 (Fig. 4B).CXCL12/CXCR4 signaling regulates an angiogenic switch

through PGK1. The previous results suggest that overexpression ofPGK1 may promote metastasis by virtue of its effect on CXCR4expression. However, the effect of PGK1 secretion may be to limit

Figure 5. PGK1 expression regulates blood vesselformation stimulated by prostate cancers grown inlow- and high-CXCL12 environments. Histologic (A ) andquantitative (B ) analysis of microvessel formation resultingfrom NOD/SCID mice implanted with PC3 cells thatoverexpressed PGK1, CXCR4, or siRNA targetingPGK1 and implanted in either a low-CXCL12 (s.c.) orhigh-CXCL12 (i.t.) environments. Microvessel formationwas determined by immunohistochemistry for humanfactor VIII (arrows ) and quantified by direct microscopiccounting of 10 random high-power fields.

Cancer Research

Cancer Res 2007; 67: (1). January 1, 2007 156 www.aacrjournals.org

Research. on January 31, 2016. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

the size of the metastatic lesions based on its effects onangiogenesis. To directly test the role of PGK1 on angiogenesisin vivo , mice were implanted s.c. with cells engineered to expressaltered levels of PGK1 in an environment that expresses low levelsof CXC12. At 1 month, the tissues were recovered, weighed, andstained for factor VIII to identify blood vessels. Overexpression ofPGK1 in PC3 cells significantly reduced the number of bloodvessels formed in the tissue from 14.3 F 2.1 to 6.8 F 0.8 per high-power field relative to controls (Fig. 5A and B). Cancer cells withreduced expression of PGK1 or overexpression of CXCR4 generatedsignificantly larger and more abundant blood vessels than controls(Fig. 5A and B). To further test the role of PGK1 on angiogenesis,mice were implanted s.c. with scaffolds containing a mixture ofhuman dermal endothelial cells with PC3Control or PC3PGK1 cells.Overexpression of PGK1 resulted the generation of smaller tumor-derived tissues and significantly reduced the number of humanblood vessels formed in the tissue from 11.0 F 2 to 1.8 F 0.8 perhigh-power field (Supplementary Fig. S4).To directly determine if CXCL12 regulates an angiogenic switch

through PGK1 activity, PC3CXCR4, PC3PGK1, or PC3PGK1 SiRNA cellswere injected i.t. into an environment rich in CXCL12 (15). After4 weeks, the skeletal lesions were identified. Tumors generated in

high CXCL12 environment produced larger and more abundantblood vessels. Overexpression of PGK1 still reduced the number ofblood vessels, although these effects were reversed by the highlevels of CXCL12 (Fig. 5A and B). Cancer cells with enhancedexpression of CXCR4 or reduced expression of PGK1 generatedlarger and more abundant blood vessels than those produced bytumors expressing scrambled siRNA controls (Fig. 5A and B). Aradiographic and histologic analyses of the resulting tumors dis-played extensive bone destruction in the control groups (Fig. 5Cand D). In comparison, animals receiving PC3PGK1 cells had smallerlesions with less osteolytic damage (Fig. 5C and D). Quantitativehistomorphometry confirmed that the overall tumor area and bonearea associated with tumor were more extensive in the PC3Control

animals versus the PC3PGK1-injected animals (21.51 F 1.3 versus7.64 F 1.6 mm; 12.08 F 1.1 versus 2.21 F 0.8 mm; Fig. 5D). Astriking feature of the resulting tissues was the formation ofextracortical woven bone in the animals injected with the PC3PGK1

cells (Fig. 5C, dashed line). Here, the number of osteoblasts permillimeter of bone surface was higher in tissues resulting fromthe PC3PGK1 versus PC3Control cells (4.86 F 0.8/mm versus 3.01 F0.4/mm). However, there were no differences in the number ofosteoclasts (2.95 F 0.3/mm versus 2.37 F 0.6/mm; Fig. 5C).

Figure 5 Continued. C, radiographic (top )and histologic [middle (Mason’s tetracromestain) and bottom (TRAP stain)] analysis oflesions resulting from i.t. injection ofPC3PGK1 versus PC3Control cells. Blackarrows, osteoclast; dashed arrows,osteoblast; dashed line, cortical boneborder. D, quantitative histomorphometryfor tumor area, bone area associated withtumor, and osteoblast and osteoclastnumber per millimeter of bone surfaceusing a computerized image analysissystem (VisioLab 2000, Biocom, Paris,France). Columns, mean of triplicatedeterminations; bars, SD. *, P < 0.05,significant difference from control. Originalmagnification, �20. Bar , 100 Am (A).

PGK1 Regulates an Angiogenic Switch

www.aacrjournals.org 157 Cancer Res 2007; 67: (1). January 1, 2007

Research. on January 31, 2016. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

Together, these in vivo and in vitro data suggest that PGK1 is acritical downstream target of CXCL12/CXCR4 chemokine axis andlikely to play a critical role in the regulation of an ‘angiogenicswitch’ essential for tumor growth and metastasis.

Discussion

In this investigation, the connection between angiogenesis,anaerobic metabolism, and chemokine signaling was examined.Using siRNA targeting of CXCR4 in prostate cancer cell lines, weidentified that the expression of PGK1 was enhanced followingdown-regulation of the chemokine receptor. Western blot analysisshowed that CXCL12 signaling through CXCR4, or overexpressionof CXCR4, inhibited the expression of PGK1. Similarly, when PGK1was overexpressed, CXCR4 expression was enhanced. In vivostudies corroborated these findings to show that more tumor cellsexited the blood and found their way to the spinal and tibialmarrow of recipient animals when the cells overexpressed PGK1.However, the size of the resulting tumors generated by PC3PGK1

cells were smaller than those made by PC3Control cells. Similarly,tumors generated by the PC3PGK1 cells following s.c. or i.t.injections were also smaller. These results are most likely due tothe fact that cells secreting PGK1 have a diminished ability torecruit a vasculature necessary to support metastasis. This mayoccur for two reasons; elevated PGK1 levels in prostate cancer celllines reduced the secretion of proangiogenic factors, includingVEGF, IL-6, and IL-8. Elevated intracellular levels of PGK1 alsoresulted in enhanced secretion of PGK1 that generated theangiogenesis inhibitor angiostatin (8–13). Alternatively, thereduced metastatic burden at 1 month might suggest a decreasein survival of the early metastatic colonies due to the apoptoticderegulation (Supplementary Table S1). Thus, our data suggest

that the role of PGK1 in cancer is a double-edged sword. PGK1regulates CXCR4 expression resulting in changes in the metastaticrate. Yet, the enhanced expression of PGK1 also leads to decreasedsecretion of proangiogenic factors and secretion of PGK1 itselfthat inhibits angiogenesis through the generation of angiostatin.These results lead us to conclude that there is a reciprocalrelationship between CXCL12/CXCR4 signaling and PGK1 expres-sion (model; Fig. 6).Prostate tumors as well as those arising in many other tissues

display a remarkable propensity to invade and survive in bone.Although the reasons for this are not entirely clear, severalmechanisms seem to facilitate this complex phenomenon. Ourprevious work has shown that prostate cancers use CXCR4 as a keyelement in their migration to tissues rich in CXCL12, includingthe lymph nodes, liver, adrenal glands, and bone (15, 16, 21). Onemechanism to account for these observations is that CXCL12transiently regulates adhesion molecules, including CD164 and thenumber and affinity of the avh3 integrin expressed by prostatecancer cells to enhance their binding to marrow endothelial cellsand components of the bone marrow extracellular matrix (17, 18).Furthermore, recent data indicate that CD44 expression mayregulate the adhesion of prostate and breast cancer cells tohaluronic acid expressed by human bone marrow endothelial cells(26). The participation of CD44 in PGK1-mediated events is alsopossible as PGK1-overexpressing prostate cancer cells significantlyoverexpressed the protein (Supplementary Table S1).Recent studies have also shown that E-cadherin/h-catenin/Wnt

signaling regulates cell-cell adhesion, migration, and tumorigen-esis (27). Likewise, PGK1 was shown to regulate E-cadherin/h-catenin, suggesting that overexpression of PGK1 promotesdecreased tumor cell-cell adhesion that potentially linked to Wntsignaling and up-regulation of cell migration (27). A second

Figure 6. Model of PGK1 action inprostate cancer. CXCL12 signalingthrough the CXCR4 receptor leads to an‘angiogenic switch’ that is mediated byextracellular PGK1 levels in prostatecancer. At site of low CXCL12, CXCR4signaling regulates PGK1 expression,which reduces angiogenesis by generatingangiostatin and by reducing the secretionof the proangiogenic cytokines VEGFand IL-8. At sites of high CXCL12production (e.g., bone, lymph node,and liver), however, PGK1 secretion isinhibited by CXCL12/CXCR4 axis favoringangiogenesis. Thus, CXCL12 signalingthrough CXCR4 generates an ‘angiogenicswitch’ necessary for metastatic growth.

Cancer Research

Cancer Res 2007; 67: (1). January 1, 2007 158 www.aacrjournals.org

Research. on January 31, 2016. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

mechanism is that CXCL12 may be critical for tumor growth. Suchthat antibody to CXCL12 decreased PC3 proliferation in vitro (14)and anti-CXCR4 antibody or a anti-CXCR4 peptide decreased thesize of the skeletal lesions generated in mice following i.t.injection (15). A third and novel mechanism is supported by thePGK1 data; when prostate cancers metastasize to bone, they enteran environment that is high in CXCL12. PGK1 in this newenvironment, which may also be achieved if metastatic cells wereto secrete CXCL12 themselves, undergo an ‘angiogenic switch,’ inwhich vascular inhibition by PGK1 is reduced and angiogenicgrowth of the tumor is promoted (Fig. 6). Critically, these dataalso suggest a mechanism to explain metastatic localizationpatterns first observed by Paget (28). Only tissues rich in CXCL12are able to overcome the vascular inhibition produced by PGK1 tofacilitate tumor growth.One of the most striking findings was the effect of PGK1

expression by prostate cancers on bone. Overexpression of PGK1lead to the generation of extracortical bone with enhanced levelsof osteoblasts and decreased numbers of osteoclasts present inthe bone. Previous work has shown that PGK1 can be secretedby tumors, where it cleaves plasminogen generating the vascularinhibitor angiostatin (8–11). Angiostatin limits tumor size bylimiting angiogenesis and by directly suppressing osteoclastdevelopment (3, 4). This is thought to be critical as activeosteoclast-mediated bone resorption may be required for bothintraosseous growth and metastatic localization (14, 29). Otherwork by our colleagues has shown that VEGF produced byprostate cancers induce initial differentiation of osteoblasts butrequire other factors, present in C4-2B but not LNCaP cells, to

induce mineralization. Therefore, how the PGK1 effects onsecretion of VEGF and other proangiogenic factors by prostatecancers result in extracortical bone formation is unclear.In summary, the results presented herein show that high levels

of PGK1 are essential for tumor growth but limit angiogenesiswhen secreted extracellularly. However, at sites of high CXCL12production, such as bone, lymph node, and liver; however,PGK1 secretion is likely to be inhibited. Thus, CXCL12 signalingthrough CXCR4 generates an ‘angiogenic switch’ that may benecessary for metastatic growth (Fig. 6). Together, these datafurther show that CXCL12/CXCR4 chemokine axis and PGK1represent at least one of the critical determinants for metastasisof prostate cancer as well as a mechanism for a proangiogenicswitch that promotes tumor growth. These findings add a newdimension to our understanding of relationship between chemo-kine and glucose metabolism enzyme and point to potentialtherapeutic interventions.

Acknowledgments

Received 8/10/2006; revised 9/25/2006; accepted 10/24/2006.Grant support: CA93900 (E.T. Keller, K.J. Pienta, and R.S. Taichman) awards from

the NIH, DE13701 (R.S. Taichman) and the Department of Defense DAMD17-02-1-0100and PC060857 (R.S. Taichman).The costs of publication of this article were defrayed in part by the payment of page

charges. This article must therefore be hereby marked advertisement in accordancewith 18 U.S.C. Section 1734 solely to indicate this fact.We thank Mary Caponite Hurley (Michigan Proteome Consortium, University of

Michigan, Ann Arbor, MI) for her technical assistance doing the two-dimensional gelsand their analysis, Dr. Hyunsuk Shim (Emory University School of Medicine, Atlanta,GA) for cDNA clones and Drs. L.K. McCouley (University of Michigan) and N.S.Taichman (University of Pennsylvania) for helpful discussions.

References1. Warburg O. Origin of cancer cells. Science 1956;123:309–14.

2. Detterbeck FC, Falen S, Rivera MP, Halle JS, SocinskiMA. Seeking a home for a PET, part 2: defining theappropriate place for positron emission tomographyimaging in the staging of patients with suspected lungcancer. Chest 2004;125:2300–8.3. Peyruchaud O, Serre CM, NicAmhlaoibh R, Fournier P,Clezardin P. Angiostatin inhibits bone metastasisformation in nude mice through a direct anti-osteoclas-tic activity. J Biol Chem 2003;278:45826–32.4. Peyruchaud O, Serre CM, NicAmhlaoibh R, SveigaardC, Clezardin P. Does tumor angiogenesis play a role inbone metastatic process? Rev Med Suisse Romande2004;124:83–4.5. Staller P, Sulitkova J, Lisztwan J, Moch H, Oakeley EJ,Krek W. Chemokine receptor CXCR4 downregulated byvon Hippel-Lindau tumour suppressor pVHL [seecomment]. Nature 2003;425:307–11.6. LaTulippe E, Satagopan J, Smith A, et al. Comprehensivegene expression analysis of prostate cancer revealsdistinct transcriptional programs associated with meta-static disease. Cancer Res 2002;62:4499–506.7. Riley DE, Krieger JN. Short tandem repeat polymor-phism linkage to the androgen receptor gene in prostatecarcinoma. Cancer 2001;92:2603–8.8. Chen G, Gharib TG, Wang H, et al. Protein profilesassociated with survival in lung adenocarcinoma. ProcNatl Acad Sci U S A 2003;100:13537–42.9. Migita T, Oda Y, Naito S, Morikawa W, Kuwano M,Tsuneyoshi M. The accumulation of angiostatin-likefragments in human prostate carcinoma. Clin CancerRes 2001;7:2750–6.10. Daly EB, Wind T, Jiang XM, Sun L, Hogg PJ. Secretionof phosphoglycerate kinase from tumour cells iscontrolled by oxygen-sensing hydroxylases. BiochimBiophys Acta 2004;1691:17–22.

11. Lay AJ, Jiang XM, Kisker O, et al. Phosphoglyceratekinase acts in tumour angiogenesis as a disulphidereductase. Nature 2000;408:869–73.12. Zhang D, Tai LK, Wong LL, Chiu LL, Sethi SK, KoayES. Proteomic study reveals that proteins involved inmetabolic and detoxification pathways are highlyexpressed in HER-2/neu -positive breast cancer. MolCell Proteomics 2005;4:1686–96.13. Hwang TL, Liang Y, Chien KY, Yu JS. Overexpressionand elevated serum levels of phosphoglycerate kinase 1in pancreatic ductal adenocarcinoma. Proteomics 2006;6:2259–72.14. Sun Y-X, Wang J, Shelburne CE, et al. The expressionof CXCR4 and CXCL12 (SDF-1) in human prostatecancers (PCa) in vivo . J Cell Biochem 2003;89:462–73.15. Sun YX, Schneider A, Jung Y, et al. Skeletallocalization and neutralization of the SDF-1(CXCL12)/CXCR4 axis blocks prostate cancer metastasis andgrowth in osseous sites in vivo . J Bone Miner Res 2005;20:318–29.16. Wang JH, Wang J, Sun Y-X, et al. Diverse signalingpathways through the SDF-1/CXCR4 chemokine axis inprostate cancer cell lines leads to altered patterns ofcytokine secretion and angiogenesis. Cell Signal 2005;17:1578–92.17. Sun Y-X, Fang M, Wang JH, Cooper CR, Pienta KJ,Taichman RS. Expression and activation of avh3integrins by SDF-1/CXCL12 increases the aggressivenessof prostate cancer cells. Prostate. In press 2006.18. Havens AM, Jung Y, Sun YX, Taichman RS. The role ofsialomucin CD164 (MGC-24v or endolyn) in prostatecancer metastasis. BMC Cancer 2006;6:195.19. Nor JE, Peters MC, Christensen JB, et al. Engineer-ing and characterization of functional human micro-vessels in immunodeficient mice. Lab Invest 2001;81:453–63.20. Lehr JE, Pienta KJ. Preferential adhesion of prostatecancer cells to a human bone marrow endothelial cellline. J Natl Cancer Inst 1998;90:118–23.

21. Taichman RS, Cooper C, Keller ET, Pienta KJ,Taichman N, McCauley LK. Use of the stromal cell-derived factor-1/CXCR4 pathway in prostate cancermetastasis to bone. Cancer Res 2002;62:1832–7.22. Lee LF, Louie MC, Desai SJ, et al. Interleukin-8 confersandrogen-independent growth and migration of LNCaP:differential effects of tyrosine kinases Src and FAK.Oncogene 2004;23:2197–205.23. Steeve KT, Marc P, Sandrine T, Dominique H, YannickF. IL-6, RANKL, TNF-a/IL-1: interrelations in boneresorption pathophysiology. Cytokine Growth FactorRev 2004;15:49–60.24. Lu KV, Jong KA, Rajasekaran AK, Cloughesy TF,Mischel PS. Upregulation of tissue inhibitor ofmetalloproteinases (TIMP)-2 promotes matrix metal-loproteinase (MMP)-2 activation and cell invasion ina human glioblastoma cell line. Lab Invest 2004;84:8–20.25. Zeng Q, Li S, Chepeha DB, et al. Crosstalk betweentumor and endothelial cells promotes tumor angiogen-esis by MAPK activation of Notch signaling. Cancer Cell2005;8:13–23.26. Draffin JE, McFarlane S, Hill A, Johnston PG, WaughDJ. CD44 potentiates the adherence of metastaticprostate and breast cancer cells to bone marrowendothelial cells. Cancer Res 2004;64:5702–11.27. Yang F, Li X, Sharma M, et al. Linking h-catenin toandrogen-signaling pathway. J Biol Chem 2002;277:11336–44.28. Paget S. The distribution of secondary growths incancer of the breast. Lancet 1889;1:571–3.29. Schneider A, Kalikin LM, Mattos AC, et al. Boneturnover mediates preferential localization of pros-tate cancer in the skeleton. Endocrinology 2005;146:1727–36.30. Kitagawa Y, Dai JL, Zhang J, et al. Vascularendothelial growth factor contributes to prostatecancer-mediated osteoblastic activity. Cancer Res 2005;65:10921–9.

PGK1 Regulates an Angiogenic Switch

www.aacrjournals.org 159 Cancer Res 2007; 67: (1). January 1, 2007

Research. on January 31, 2016. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from

2007;67:149-159. Cancer Res   Jianhua Wang, Jincheng Wang, Jinlu Dai, et al.   Prostate CancerA Glycolytic Mechanism Regulating an Angiogenic Switch in

  Updated version

  http://cancerres.aacrjournals.org/content/67/1/149

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2006/12/27/67.1.149.DC1.html

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/67/1/149.full.html#ref-list-1

This article cites 28 articles, 11 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/67/1/149.full.html#related-urls

This article has been cited by 11 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] at

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

  Permissions

  [email protected] at

To request permission to re-use all or part of this article, contact the AACR Publications

Research. on January 31, 2016. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from