SPARC for talk

download SPARC for talk

of 17

Transcript of SPARC for talk

  • 8/7/2019 SPARC for talk

    1/17

    2007;5:1015-1030. Published online October 19, 2007.Mol Cancer ResNeveen Said, Matthew J. Socha, Jeffrey J. Olearczyk, et al.SPARCNormalization of the Ovarian Cancer Microenvironment by

    Updated Version10.1158/1541-7786.MCR-07-0001doi:

    Access the most recent version of this article at:

    Cited Articleshttp://mcr.aacrjournals.org/content/5/10/1015.full.html#ref-list-1

    This article cites 85 articles, 46 of which you can access for free at:

    Citing Articleshttp://mcr.aacrjournals.org/content/5/10/1015.full.html#related-urls

    This article has been cited by 1 HighWire-hosted articles. Access the articles at:

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

    SubscriptionsReprints and

    [email protected] Department atTo order reprints of this article or to subscribe to the journal, contact the AACR

    [email protected] at

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

    American Association for Cancer ResearchCopyright 2007on February 8, 2011mcr.aacrjournals.orgDownloaded from

    DOI:10.1158/1541-7786.MCR-07-0001

    http://mcr.aacrjournals.org/lookup/doi/10.1158/1541-7786.MCR-07-0001http://mcr.aacrjournals.org/lookup/doi/10.1158/1541-7786.MCR-07-0001http://mcr.aacrjournals.org/lookup/doi/10.1158/1541-7786.MCR-07-0001http://mcr.aacrjournals.org/content/5/10/1015.full.html#ref-list-1http://mcr.aacrjournals.org/content/5/10/1015.full.html#ref-list-1http://mcr.aacrjournals.org/content/5/10/1015.full.html#related-urlshttp://mcr.aacrjournals.org/content/5/10/1015.full.html#related-urlshttp://mcr.aacrjournals.org/cgi/alertshttp://mcr.aacrjournals.org/cgi/alertsmailto:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]:[email protected]://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://mcr.aacrjournals.org/mailto:[email protected]:[email protected]://mcr.aacrjournals.org/cgi/alertshttp://mcr.aacrjournals.org/content/5/10/1015.full.html#related-urlshttp://mcr.aacrjournals.org/content/5/10/1015.full.html#ref-list-1http://mcr.aacrjournals.org/lookup/doi/10.1158/1541-7786.MCR-07-0001
  • 8/7/2019 SPARC for talk

    2/17

    Normalization of the Ovarian Cancer Microenvironmentby SPARC

    Neveen Said,1 Matthew J. Socha,1 Jeffrey J. Olearczyk,1 Ahmed A. Elmarakby,1

    John D. Imig,1,2 and Kouros Motamed1,3

    1Vascular Biology Center and Departments of 2Physiology and3Pathology,

    Medical College of Georgia, Augusta, Georgia

    AbstractMalignant ascites is a major source of morbidity and

    mortality in ovarian cancer patients. It functions as a

    permissive reactive tumor-host microenvironment and

    provides sustenance for the floating tumor cells through

    a plethora of survival/metastasis-associated molecules.

    Using a syngeneic, immunocompetent model of

    peritoneal ovarian carcinomatosis in SP/ mice, we

    investigated the molecular mechanisms implicated in

    the interplay between host secreted protein acidic and

    rich in cysteine (SPARC) and ascitic fluid prosurvival/

    prometastasis factors that result in the significantly

    augmented levels of vascular endothelial growth factor

    (VEGF) and matrix metalloproteinases (MMP). Ascitic

    fluidenhanced ID8 invasiveness was mediated through

    VEGF via a positive feedback loop with MMP-2 and

    MMP-9 and through activation of Av and B1 integrins.

    Host SPARC down-regulated the VEGF-MMP axis at the

    transcriptional and posttranscriptional levels. In vitro,

    SPARC attenuated the basal as well as VEGF-induced

    integrin activation in tumor cells. SPARC inhibited theVEGF- and integrin-mediated ID8 proliferation in vitro

    and significantly suppressed their tumorigenicity

    in vivo. Relative to SP+/+, SP/ ascitic fluid contained

    significantly higher levels of bioactive lipids and exerted

    stronger chemotactic, proinvasive, and mitogenic

    effects on ID8 cells in vitro. SP/ ascites also

    contained high levels of interleukin-6, macrophage

    chemoattractant protein-1, and 8-isoprostane

    (prostaglandin F2A) that were positively correlated with

    extensive infiltration of SP/ ovarian tumors and

    ascites with macrophages. In summary, our findings

    strongly suggest that host SPARC normalizes the

    microenvironment of ovarian cancer malignant ascitesthrough down-regulation of the VEGF-integrin-MMP

    axis, decreases the levels and activity of bioactive lipids,

    and ameliorates downstream inflammation.

    (Mol Cancer Res 2007;5(10):101530)

    IntroductionOvarian cancer is the leading cause of death among

    gynecologic cancers in the United States. Metastatic

    dissemination to peritoneal and pleural effusions is consid-

    ered the major cause of patient morbidity and mortality in

    ovarian cancer. The ability of floating ovarian cancer cells

    in these malignant effusions to survive, proliferate, and

    disseminate in the absence of immediate proximity to a solid

    scaffold and vascular structures has been attributed to the

    unique, yet poorly defined, permissive microenvironment of

    these dense exudative fluids (1). The high autonomous

    proliferation rate of the metastatic ovarian cancer cells, their

    ability to develop alternative means of survival, as well as

    the contributions of inflammatory cells present in malignant

    effusions are believed to be responsible for the pluoripotent

    nature of these malignant effusions (2-5). Ovarian cancer

    cells and peritoneal mesothelial cells constitutively produce

    bioactive lipids and cytokines that further promote moto-

    genesis and mitogenesis of ovarian cancer cells in both

    autocrine and paracrine fashion (6-9). The autocrine secretion

    of cytokines, peptide growth factors, as well as proteolyticenzymes by malignant cells not only contributes to ascites

    accumulation but also enhances their survival and dissemi-

    nation (1, 10-12). Malignant ascites contains inflammatory

    cells such as macrophages that further contribute to the

    production of bioactive phospholipids, inflammatory cyto-

    kines, and peptide growth factors in the ascitic fluid (13-15).

    The interplay between different components of ascitic fluid is

    complex and reflects the tumor-host interactions that support

    the malignant process. Identifying and dissecting the factors

    involved in the crosstalk between the components of such

    microenvironments will not only improve our understanding

    of the disease but will ultimately enable us to provide better

    patient care.

    Secreted protein, acidic, and rich in cysteine (SPARC) is a

    matricellular protein involved in the modulation of cell

    adhesion, motility, and interactions with components of the

    extracellular matrix (16, 17). Recent reports highlight the role

    of this molecule as a positive or negative modulator in the

    pathogenesis of different malignancies (18-24). We and others

    have shown that SPARC functions as a tumor suppressor in

    ovarian cancer (25-28). As a tumor suppressor, effect of

    SPARC in different cancers was attributed not only to its

    counteradhesive, antiproliferative functions but also to its role

    in modulating angiogenesis as well as regulation of the

    production, assembly, and organization of the structural

    extracellular matrix proteins including type I collagen and

    fibronectin (23, 25, 29-32). Interestingly, the quality and

    Received 1/1/07; revised 5/28/07; accepted 6/7/07.

    Grant support: NIH grants K01-CA089689 (K. Motamed) and HL59699 (J.D.Imig).

    The costs of publication of this article were defrayed in part by the payment ofpage charges. This article must therefore be hereby markedadvertisement in

    accordance with 18 U.S.C. Section 1734 solely to indicate this fact.Requests for reprints: Kouros Motamed, Vascular Biology Center, CB-3306,

    Medical College of Georgia, 1459 Laney Walker Boulevard, Augusta, GA 30912.Phone: 706-721-9796; Fax: 706-721-9799. E-mail: [email protected] 2007 American Association for Cancer Research.

    doi:10.1158/1541-7786.MCR-07-0001

    Mol Cancer Res 2007;5(10). October 2007 1015

    American Association for Cancer ResearchCopyright 2007on February 8, 2011mcr.aacrjournals.orgDownloaded from

    DOI:10.1158/1541-7786.MCR-07-0001

    http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://mcr.aacrjournals.org/
  • 8/7/2019 SPARC for talk

    3/17

    quantity of mature collagen bundles reported in tumor stroma

    were positively correlated with levels of both tumor cell

    SPARC (32, 33) and host SPARC (19, 25, 34). Thus, it was

    hypothesized that SPARC-induced changes in the tumor

    microenvironment were responsible, at least in part, for its

    anticancer effects (32).

    Herein, to assess the role of host SPARC in modulating the

    ovarian cancer microenvironment, specifically ascitic fluid,

    we used a syngeneic model of ovarian carcinomatosis in

    immunocompetent SP/ mice by peritoneal implantation of

    ID8 mouse ovarian cancer cells. The results of our in vitro and

    in vivo studies have identified SPARC as a novel tumor

    suppressor that normalizes the ovarian cancer microenviron-

    ment, in part, through significant attenuation of the vascular

    endothelial growth factor (VEGF)-integrin-MMP signaling

    axis as well as the inflammatory processes that result from,

    augment, and/or contribute to this axis.

    ResultsSPARC Inhibits VEGF-Induced Proliferation and Survival

    of Ovarian Cancer Cells In vitro and In vivo

    VEGF is known to exert an autocrine as well as paracrine

    mitogenic effect on a variety of tumor cells including

    melanoma, prostate cancer, and ovarian cancer in vivo

    (22, 35-38). Our pilot studies indicated that VEGF stimulates

    the proliferation of ID8 cells in a concentration-dependent

    manner (data not shown). To investigate the autocrine effect

    of VEGF and to mimic the human disease, where VEGF is

    constitutively up-regulated in cancer cells, we used an ID8 cell

    line stably expressing VEGF164 (ID8-VEGF). This cell line has

    previously been shown to dramatically accelerate tumor cell

    growth and survival, tumor angiogenesis, and ascites formation

    in a syngeneic mouse model of ovarian carcinomatosis

    (39). ID8-VEGF exhibited higher proliferation rates than ID8

    cells (Fig. 1A). In accord with our earlier report (25), the

    FIGURE 1. SPARC inhibits VEGF-induced survival of ovarian cancer cells in vitro and in vivo. A. Proliferation of ID8 and ID8-VEGF cells was determinedby MTS assay. ID8-VEGF cells exhibited significantly higher rates of proliferation than ID8 cells, and both were inhibited by SPARC in a concentration-dependent manner. B. Animals injected intraperitoneally with ID8-VEGF cells exhibited decreased survival rates than those injected with ID8. SP/ miceexhibited decreased survival than SP+/+ counterparts. C. SP/ mice injected with ID8-VEGF developed larger volumes of ascitic fluid as compared withSP+/+. D. SP/ mice developed significantly larger ID8-VEGF subcutaneous tumors than SP+/+ counterparts. E. The mean vascular density (MVD) in ID8and ID8-VEGF tumors in SP/ and SP+/+ mice was determined by CD31 immunostaining. Columns, number of CD31-positive vessels counted in fivemicroscopic fields of at least four different tumor samples; bars, SE. #, P < 0.05, between ID8-VEGF tumors in SP+/+ and SP/ mice. {, P < 0.05, between

    ID8 and ID8-VEGF tumors in SP/ mice.

    Said et al.

    Mol Cancer Res 2007;5(10). October 2007

    1016

    American Association for Cancer ResearchCopyright 2007on February 8, 2011mcr.aacrjournals.orgDownloaded from

    DOI:10.1158/1541-7786.MCR-07-0001

    http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://mcr.aacrjournals.org/
  • 8/7/2019 SPARC for talk

    4/17

  • 8/7/2019 SPARC for talk

    5/17

    growth in mice (43). A reciprocal stimulation of the expression

    and activity of MMP-2 by VEGF has also been reported in

    ovarian cancer cell lines (44). Consistent with our previous

    study (25), ascitic fluid of SP/ ovarian tumorbearing mice

    showed enhanced MMP-2 and MMP-9 proteolytic activity

    (data not shown). Up-regulated levels of MMP-2 and MMP-9

    mRNA in SP/ tumors were concomitant with the down-

    regulation of tissue inhibitor of metalloproteinase (TIMP)-1 and

    TIMP-2 levels. Conversely, up-regulated TIMP-1 and TIMP-2

    levels in tumors of SP+/+ mice were associated with down-

    regulation of MMPs (Fig. 2A). Immunostaining of tumor

    sections also confirmed the increased expression of MMP-2 and

    MMP-9 and the decreased expression of TIMP-1 and TIMP-2

    in SP/ tumors (Fig. 2B). In vitro, treatment of ID8 cell line

    with SPARC (20 Ag/mL) for 6 h resulted in down-regulation of

    MMP-2 and MMP-9 mRNA expression (Fig. 2C). However,

    SPARC treatment did not have a significant effect on regulation

    of TIMP-1 and TIMP-2 mRNA levels (Fig. 1C). These

    results indicate that the tumor suppressor effect of SPARC is

    mediated through both down-regulation of the expression and

    activity of MMPs and up-regulation of their tissue inhibitors

    in ID8 tumor cells in vivo and in vitro. The elevated levels

    and activity of MMPs in SP/ tumor-bearing mice can also be

    attributed to the significantly increased tumor-associated and

    FIGURE 3. Augmentation of ID8 cell adhesion and invasion by ascitic fluid is VEGF and MMP mediated. A. ID8 cells (1 106/mL) were preincubated withsFlt2-11 (50 Ag/mL), KDR2 inhibitor IV (KDR2 I; 50 Amol/L), VEGF neutralizing antibody (VEGF-Ab; 20 Ag/mL), or vehicle control (DMSO) in DMEM-0.4%bovine serum albumin for 30 min before incubation with SP/ and SP+/+ ascitic fluid (final dilution of ascitic fluid was 1:2 in DMEM). Cells were allowed toadhere to fibronectin-coated wells of 96-well plates for 4 h. Adherent cells were stained and quantified by measuring absorbance ( OD) at 590 nm. B. ID8 cellswere treated exactly the same as in (A), and in the presence of MMP-2/MMP-9 inhibitors (MMP-2/9 I; 50 Amol/L) or with the general MMP inhibitor GM 6001(50 Amol/L) for 30 min before being allowed to invade fibronectin-coated inserts for 5 h. Columns, mean of three independent experiments done in triplicates;bars, SE. *, P < 0.05, compared with vehicle control. #, P < 0.05, between SP/ and SP+/+. C. Basal adhesion of ID8 cells to fibronectin was measured afterpretreatment with SPARC (20 Ag/mL, 2 h), VEGF neutralizing antibody, Flt2-11, and KDR2 inhibitor IV as described above. D. Basal fibronectin invasion byID8 cells after pretreatment with SPARC, VEGF neutralizing antibody, Flt2-11, and KDR2 inhibitor IV was done exactly as described in ( A). ID8-VEGF celladhesion to (E) and invasion of (F) fibronectin were tested as described above. Adhesion or invasion of ID8-VEGF cells treated with vehicle alone (control)was assigned a value of 100%. Adhesion and invasion results are expressed as percent of control. Columns, mean of three independent experiments done in

    quadruplicates; bars, SE. *, P < 0.05.

    Said et al.

    Mol Cancer Res 2007;5(10). October 2007

    1018

    American Association for Cancer ResearchCopyright 2007on February 8, 2011mcr.aacrjournals.orgDownloaded from

    DOI:10.1158/1541-7786.MCR-07-0001

    http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://mcr.aacrjournals.org/
  • 8/7/2019 SPARC for talk

    6/17

    tumor-infiltrating macrophages. Interestingly, SP/ tumor

    tissues and ascites exhibited a significant increase in

    macrophage infiltration, as indicated by F4/80 staining relative

    to their SP+/+ counterparts (Fig. 2D). In accord with our

    previous study (25), these results were coincident with the up-

    regulation of the expression of VEGF and its receptors in

    tumor tissues and VEGF levels and activity in the ascitic fluid

    of SP/ mice.

    Augmentation of ID8 Cell Adhesion and Invasion by

    Ascitic Fluid Is VEGF and MMP Mediated

    The proinvasive properties of malignant ascites have been

    shown to be mediated through the interplay of VEGF and

    MMPs (2, 41, 45-47). Our previous findings that ascitic fluid

    from SP/ mice enhanced the invasiveness and, to a lesser

    extent, the adhesion of ID8 cells (25) led us to investigate the

    possible mechanisms by which SPARC modulates the interplay

    of VEGF and MMPs in the milieu of the ascitic fluid.

    Consistent with our earlier report, the difference between SP+/+

    and SP/ ascitic fluid induced adhesion was found to be

    insignificant but markedly inhibited by a VEGF neutralizing

    antibody, sFlt2-11, and a VEGFR2 (KDR) tyrosine kinase

    inhibitor, suggesting an autocrine effect of VEGF produced

    by ID8 cells themselves (Fig. 3A). On the other hand, SP/

    ascitic fluid significantly (>75%) enhanced the invasiveness

    of ID8 cells compared with SP+/+ ascitic fluid (Fig. 3B). This

    FIGURE 4. Modulation of integrin-mediated proliferation, adhesion, and invasion of ID8 cells by host SPARC. A. P4G11 stimulation (10 Ag/mL DMEM-2%fetal bovine serum) increased the proliferation of ID8 cells grown on uncoated (solid columns), fibronectin-coated (checkered columns), and collagenIcoated (open columns) wells by 1- to 1.2-fold over unstimulated controls. SPARC treatment significantly decreased P4G11-induced proliferation(*, P < 0.05). MnCl2 (Mn

    2+) increased ID8 proliferation by 1-fold (on uncoated wells) and f2-fold when grown on fibronectin and vitronectin (striped columns).SPARC significantly inhibited MnCl2-induced proliferation (**, P < 0.01). Columns, mean fold increase in cell proliferation, as compared with matched isotypecontrol antibody treated in DMEM-2% fetal bovine serum (assigned a value of 1); bars, SE. ID8 cells were preincubated with LM609, P1F6, and P4C10(15 Ag/mL each) or RGD peptide (10 Amol/L) for 30 min before stimulation with SP+/+ or SP/ ascitic fluid (final dilution is 1:2 in DMEM-0.4% bovine serumalbumin). Cells were allowed to adhere to fibronectin-coated wells for 4 h ( B) or invade fibronectin-coated inserts for 5 h ( C). *, P < 0.05, versus vehicle- ormatched isotype-treated SP+/+ controls. **, P < 0.05, versus vehicle- or matched isotype-treated SP/ controls. D. An ex vivo cell adhesion system showingsignificantly increased ID8-GFP adhesion to SP/ peritoneal explants compared with SP+/+. E. Adhesion of ID8-GFP cells to SP+/+ and SP/ murineperitoneal mesothelial cells is integrin mediated. ID8-GFP cells were pretreated exactly as described in ( B) and then were cocultured with primary SP+/+ andSP/ murine peritoneal mesothelial cells in 24-well plates for 5 h, as described in Materials and Methods. Columns, mean fluorescent intensity ( MFI) of sixfields per experimental condition from three independent experiments done in triplicates; bars, SE. *, P < 0.05, compared with SP+/+ vehicle- or isotype-treated controls. **, P < 0.05, compared with SP/ vehicle- or isotype-treated controls. F. Reverse transcription-PCR analysis of integrin expression in ID8peritoneal tumors grown in SP+/+ and SP/ mice. Columns, mean mRNA expression levels of integrin subunits from three different tumor samples in eachgroup following normalization to levels of rpS6 used as an internal control; bars, SE. Representative of three independent experiments.

    SPARC Normalizes Ovarian Cancer Microenvironment

    Mol Cancer Res 2007;5(10). October 2007

    1019

    American Association for Cancer ResearchCopyright 2007on February 8, 2011mcr.aacrjournals.orgDownloaded from

    DOI:10.1158/1541-7786.MCR-07-0001

    http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://mcr.aacrjournals.org/
  • 8/7/2019 SPARC for talk

    7/17

    invasion of fibronectin-coated inserts was not only inhibited

    significantly by VEGF neutralizing antibody and VEGFR

    inhibitors but also by MMP inhibitors. These results suggest

    that the proinvasive and, to a lesser extent, the proadhesive

    effects of ascitic fluid are mediated through the paracrine effects

    of VEGF and MMPs. In support of these findings, basal levels

    of ID8 cell adhesion to and invasion of fibronectin were shown

    to be significantly attenuated (f35% and 50%, respectively) in

    the presence of exogenous SPARC (Fig. 3C and D). Inhibitors

    of VEGF and VEGFR2, but not VEGFR1, showed significant

    suppression of ID8 cell adhesion (Fig. 3C), whereas inhibitors

    of VEGF, VEGFR, and MMP all resulted in significant

    diminutions in ID8 cell invasion (Fig. 3D). These findings

    suggest an autocrine effect exerted by VEGF that is produced

    by ID8 cells themselves.

    To further investigate the autocrine effect of VEGF, using

    ID8-VEGF cell line, we found that they exhibited a significant

    increase in adhesion to fibronectin (f20%) as compared with

    ID8 controls. This increase in adhesion was significantlyinhibited (up to 40%) by exogenous SPARC and inhibitors of

    VEGFRs but not MMPs (Fig. 3E). Furthermore, ID8-VEGF

    cells exhibited markedly increased (f37%) fibronectin inva-

    sion, relative to ID8 controls, which was significantly (30-70%)

    inhibited by exogenous SPARC as well as by inhibitors of

    VEGFRs and MMPs (Fig. 3F).

    Modulation of Integrin-Mediated Adhesion and Invasion

    of ID8 Cells by Host SPARC

    It has been shown that adhesion of ovarian cancer cells

    to the peritoneal mesothelial cells is integrin mediated (48-50).

    We have previously shown that SPARC significantly inhibits

    human ovarian cancer cell adhesion to and invasion ofextracellular matrix proteins and human mesothelial cells

    (25), and that these functions were mediated through attenua-

    tions of tumor cell surface expression and/or clustering of

    av and h1 integrins (28). In the latter study, we found that

    SPARC inhibited integrin-mediated proliferation of human

    ovarian cancer cell lines (28). Herein, we show that activation

    ofav integrin with 2.5 mmol/L MnCl2 significantly stimulated

    proliferation of ID8 cells plated on fibronectin- and vitronectin-

    coated wells (f2.2-fold as compared with unstimulated cells,

    assigned a value of 1; Fig. 4A) as compared with uncoated

    wells. Exogenous SPARC (10 Ag/mL) attenuated MnCl2-

    induced proliferation by >1.5-fold. On the other hand,

    activation ofh1 integrin by P4G11 exerted a significant, albeit

    less pronounced, effect on ID8 proliferation whether they were

    seeded on uncoated, collagen Icoated, or fibronectin-coated

    plates. This effect was significantly inhibited by SPARC. These

    findings led us to investigate whether ascitic fluid induced ID8

    cell adhesion and invasion are also integrin mediated. We found

    that ascitic fluidinduced adhesion of ID8 cells to fibronectin

    was significantly suppressed by inhibitors of avh3 (LM609),

    avh5 (P1F6), and h1 (P4C10), as well as by RGD peptide

    (Fig. 4B). However, no significant difference was detected

    between the proadhesive effects of SP/ and SP

    +/+ ascitic

    fluids. Conversely, the proinvasive effect of SP/ ascitic fluid

    was significantly (>70%) higher than that of SP+/+ controls

    (Fig. 4C). This ascitic fluid induced invasion of fibronectin

    was also found to be inhibited by the aforementioned integrin

    inhibitors. Next, we tested whether SP/ peritoneal mesothe-

    lium supports adhesion of ID8 cells better than SP+/+ controls

    and whether this adhesion is integrin mediated. Using an

    ex vivo organ culture system, coincubation of freshly isolated

    peritoneal explants, from either diaphragmatic peritoneum or

    peritoneal lining of the anterior abdominal wall of SP/

    animals, resulted in a significant (f3-fold) increase in the

    adhesion of ID8-GFP cells relative to their SP+/+ counterparts

    (Fig. 4D). Therefore, we tested the effect of integrin activation

    and inhibition on the adhesion of ID8-GFP cells to isolated

    SP/ and SP+/+ primary murine peritoneal mesothelial cells

    in vitro. SP/ murine peritoneal mesothelial cells consistently

    showed a significantly increased (25%) basal rate of adhesion

    to ID8-GFP cells compared with SP+/+ controls (Fig. 4E).

    Activation of av and h1 integrins by Mn2+ and P4G11,

    respectively, resulted in a significant increase in adhesion of

    ID8-GFP cells over unstimulated controls and was markedly

    suppressed by function-blocking antibodies against avh3 and

    h1 (Fig. 4E). ID8-GFP adhesion rates to SP/ murine

    peritoneal mesothelial cells were consistently (15-25%) higher

    than SP+/+ control whether integrins were activated by Mn2+

    and P4G11 or inhibited by function-blocking antibodies and

    RGD peptide. These results indicated that although the

    adhesion of ID8 cells to murine peritoneal mesothelial cells

    was integrin mediated, the difference in the adhesion rates was

    correlated with the basal levels, which were mainly attributed

    to the absence of SPARC in SP/ murine peritoneal

    mesothelial cells. At the transcription level, absence of host

    SPARC had no significant effect on integrin mRNA levels

    (Fig. 4F) or protein levels (data not shown) in SP/ and SP

    +/+

    tumors. However, we cannot rule out differences in activation

    of tumor cell surface integrins due to unavailability ofantibodies against mouse-specific, activated integrin subunits.

    Taken together, these results indicated that the absence of host

    SPARC augments the adhesion of ID8 cells to extracellular

    matrix of the peritoneal mesothelium and that the adhesion is

    also integrin mediated. This integrin-mediated adhesion is

    independent of mesothelial SPARC but is hypothesized to be

    due to the effect of soluble SPARC itself on integrin activation

    and clustering and/or its effects on the soluble factors in ascitic

    fluid that activate integrins on the cell surface of ID8 cells.

    Interplay between VEGF and Integrins Influences Ligand

    Recognition and the Proinvasive Properties of Ovarian

    Cancer Cells

    VEGF has been reported to activate the key integrins in

    tumor cells as well as endothelial cells, thus influencing not

    only angiogenesis but also tumor growth and metastasis

    (22, 35-37, 51, 52). Ovarian cancer cells synthesize high levels

    of VEGF, which not only induces tumor angiogenesis but also

    functions as a permeability factor implicated in the production

    and maintenance of malignant ascitic fluid, which in turn

    supports tumor growth and metastasis (2, 47). Having shown

    that the proadhesive and proinvasive effects of SP/ and SP

    +/+

    ascitic fluid on ID8 cells are both VEGF and integrin mediated,

    we investigated whether SPARC itself inhibits integrin

    activation and/or inhibits integrin activation by VEGF in ID8

    cells. We found that ID8 cells exhibited high levels of basal

    adhesion to fibronectin that was significantly augmented by

    Said et al.

    Mol Cancer Res 2007;5(10). October 2007

    1020

    American Association for Cancer ResearchCopyright 2007on February 8, 2011mcr.aacrjournals.orgDownloaded from

    DOI:10.1158/1541-7786.MCR-07-0001

    http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://mcr.aacrjournals.org/
  • 8/7/2019 SPARC for talk

    8/17

    integrin activators P4G11 and Mn2+ (Fig. 5A). ID8-VEGF

    exhibited significantly (f40%) higher adhesion to fibronectin

    than ID8 cells, an increase that was comparable to that of

    integrin activation. SPARC inhibited both VEGF-mediated and

    integrin-mediated adhesion to fibronectin by 40% to 50%,

    respectively (Fig. 5A). To determine the integrins activated by

    VEGF, we used integrin inhibiting antibodies and RGD peptide.

    We found that the adhesion of ID8-VEGF cells to fibronectin

    was inhibited (up to 40%) by function-blocking antibodies to

    avh3 (LM609), avh5 (P1F6), h1 (P4C10), as well as RGD

    FIGURE 5. SPARC inhibits integrin-mediated and VEGF-enhanced adhesion of ID8 cells. A. ID8 cells were preincubated with SPARC (10 Ag/mL) for2 h before stimulation with P4G11 (10 Ag/mL) or MnCl2 (2.5 mmol/L) for 30 min. ID8-VEGF cells were also pretreated with SPARC as mentioned above. Cellsuspensions were allowed to bind to fibronectin-coated wells for 4 h. Adhesion of ID8 cells treated with vehicle alone was assigned a value of 100%.Adhesion is expressed as percent change from control untreated ID8 cells, which is assigned a value of 100%. *, P < 0.05, versus vehicle- or isotype-stimulated control. **, P < 0.05, versus vehicle- or isotype-stimulated ID8 or ID8-VEGF controls. B. ID8-VEGF cells were pretreated with SPARC or integrininhibitors as described earlier and were plated on fibronectin-coated wells. Adhesion is expressed as percent change from control untreated ID8-VEGF cells,which is assigned a value of 100%. *, P < 0.05, versus ID8-VEGF control (C) cells. C. ID8 cells were preincubated with SPARC as in ( A) before stimulationwith MnCl2 (2.5 mmol/L) for 30 min. ID8-VEGF cells were similarly pretreated with SPARC. Cell suspensions were allowed to bind to vitronectin-coated wellsfor 4 h. Adhesion is expressed as percent change from vehicle controltreated ID8 cells, which is assigned a value of 100%. *, P < 0.05, versus vehicle-treated ID8 control. **, P < 0.05, versus vehicle-treated ID8 or ID8-VEGF. D. ID8-VEGF cells were pretreated with SPARC or av integrin inhibitors, asdescribed earlier, and were plated on vitronectin-coated wells. Adhesion is expressed as percent change from vehicle- or isotype control treated ID8-VEGFcells, which is assigned a value of 100%. *, P < 0.05, versus ID8-VEGF cells. E. ID8 cells were preincubated with SPARC as in ( A) before stimulation withP4G11 for 30 min. ID8-VEGF cells were similarly pretreated with SPARC. Cell suspensions were allowed to bind to collagen I coated wells for 4 h. Adhesionis expressed as percent change from control untreated ID8 cells, which is assigned a value of 100%. *, P < 0.05, versus isotype control stimulated ID8control. **, P < 0.05, versus isotype-stimulated ID8 or ID8-VEGF. F. ID8-VEGF cells were pretreated with SPARC or h1 integrin inhibitor P4C10, as describedearlier, and were then added to collagen I coated wells. Adhesion is expressed as percent change from vehicle- or isotype control treated ID8-VEGF cells,

    which is assigned a value of 100%. *, P < 0.05, versus ID8-VEGF cells.

    SPARC Normalizes Ovarian Cancer Microenvironment

    Mol Cancer Res 2007;5(10). October 2007

    1021

    American Association for Cancer ResearchCopyright 2007on February 8, 2011mcr.aacrjournals.orgDownloaded from

    DOI:10.1158/1541-7786.MCR-07-0001

    http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://mcr.aacrjournals.org/
  • 8/7/2019 SPARC for talk

    9/17

    peptide (Fig. 5B). To further identify the VEGF-activated

    integrins, we first used vitronectin, the prototypic ligand of

    avh3, and found that ID8-VEGF exhibited a significantly

    increased (f50%) adhesion to vitronectin relative to ID8-GFP

    cells (Fig. 5C) and that the observed increased adhesion is

    significantly inhibited by LM609 and RGD but not by P1F6

    antibodies (Fig. 5D). Next, we used collagen I as a putative

    ligand for h1 integrins and found that the adhesion of ID8-

    VEGF was comparable to that of P4G11-activated ID8-GFP

    cells (Fig. 5E) and was significantly inhibited by P4C10

    (Fig. 5F). These data indicate that VEGF regulates adhesion

    and recognition of extracellular matrix molecules by ovarian

    cancer cells through activation ofav (avh3, avh5, and possibly

    avh1) and h1 integrin subunits.

    FIGURE 6. SPARC inhibits integrin-mediated and VEGF-enhanced ID8 invasion by ID8 cells. ID8 and ID8-VEGF cell lines were treated exactly asdescribed in Fig. 4. ID8 cells pretreated with SPARC or PBS, in the presence or absence of integrin activators, were added to the upper chamber offibronectin-coated inserts (A). After 5 h, fibronectin invasion was determined by counting the cells on the undersurface of the inserts. Invasion is expressed aspercent change from vehicle- or isotype control antibody treated ID8 cells, which is assigned a value of 100%. *, P < 0.05, versus ID8 controls. **, P < 0.05,versus matched stimulated ID8 or ID8-VEGF. B. ID8-VEGF cells were pretreated with SPARC, vehicle, or isotype control antibody (C), or integrin inhibitors,and allowed to invade fibronectin-coated inserts. Invasion is expressed as percent change from control ID8-VEGF cells, which is assigned a value of 100%.*, P < 0.05, versus ID8-VEGF cells. C. ID8 cells were preincubated with SPARC or PBS in the presence or absence of MnCl 2. ID8-VEGF cells werepretreated with SPARC or PBS and allowed to invade vitronectin-coated inserts. Invasion is expressed as percent change from control ID8 cells, which isassigned a value of 100. *, P < 0.05, versus untreated ID8 control. **, P < 0.05, versus matched stimulated ID8 or ID8-VEGF. D. ID8-VEGF cells werepretreated with SPARC, isotype control antibody, or av integrin inhibitors and allowed to invade vitronectin-coated inserts. Vitronectin invasion is expressedas percent change from control ID8-VEGF cells, which is assigned a value of 100%. *, P < 0.05, versus ID8-VEGF cells. E. ID8 cells were preincubated withSPARC in the presence of h1 integrin activator P4G11 or isotype control antibody. ID8-VEGF cells were pretreated with SPARC or PBS and allowed toinvade collagen I coated inserts. Collagen I invasion is expressed as percent change from control untreated ID8 cells, which is assigned a value of 100%.*, P < 0.05, versus isotype-treated ID8 control. **, P < 0.05, versus matched stimulated ID8 or ID8-VEGF. F. ID8-VEGF cells were pretreated with isotypecontrol antibody, SPARC, or h1 integrin inhibitor P4C10, as described earlier, and were added to the upper chambers of collagen I coated inserts. Invasion

    is expressed as percent change from control ID8-VEGF cells, which is assigned a value of 100. *, P < 0.05, versus ID8-VEGF cells.

    Said et al.

    Mol Cancer Res 2007;5(10). October 2007

    1022

    American Association for Cancer ResearchCopyright 2007on February 8, 2011mcr.aacrjournals.orgDownloaded from

    DOI:10.1158/1541-7786.MCR-07-0001

    http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://mcr.aacrjournals.org/
  • 8/7/2019 SPARC for talk

    10/17

    Next, we tested whether VEGF influences the integrin-

    mediated invasiveness of ovarian cancer cells. Accordingly, the

    effect of overexpression of VEGF164 on ID8 cell invasion was

    assessed. In a transwell system, ID8-VEGF exerted a profound

    increase in invasion of fibronectin, vitronectin, and collagen I

    matrices. As shown in Fig. 6A, fibronectin invasion by ID8-

    VEGF was significantly (f60%) higher than that of ID8-GFP

    and was comparable to that induced by activating h1 integrins

    (using P4G11) and av integrins (using Mn2+). This invasion

    was inhibited substantially (up to 40%) by LM609, RGD

    peptide, P1F6, P4C10, as well as SPARC (Fig. 6B). To

    delineate the identity of the activated integrins, we used

    vitronectin and collagen I as substrates. We found that basal

    invasion of vitronectin by ID8 cells was significantly increased

    (>55%) by Mn2+ and inhibited (>60%) by SPARC (Fig. 6C).

    ID8-VEGF exhibited a significant (>40%) increase in vitro-

    nectin invasion as compared with ID8-GFP. Vitronectin

    invasion by ID8-VEGF was significantly inhibited (up to

    40%) by LM609 and RGD peptide but not by P1F6 (Fig. 6D),

    suggesting that avh3 is involved in VEGF-mediated invasion

    of ID8 cells. Similarly, invasion of collagen I by ID8 cells

    was markedly (f40%) augmented by h1 integrin activating

    antibody P4G11, comparable to that of levels found in ID8-

    VEGF (Fig. 6E). Collagen I invasion by ID8-VEGF was

    significantly (up to 35%) inhibited by P4C10 and SPARC

    (Fig. 6F). These results suggest that the constitutive activity of

    integrins on ID8 cells was induced by a VEGF-dependent

    autocrine loop through activation of av and h1 integrins.

    SPARC attenuated not only the constitutive activity of integrins

    in tumor cell lines but also their activated levels induced by

    specific integrin activators and VEGF.

    Modulation of the Activity of Bioactive Lipids in the Ascitic

    Fluid by Host SPARC

    We and others have recently shown that biologically active

    lipids, mainly lysophosphatidic acid (LPA), in the heat-

    insensitive fraction of mesothelial cell conditioned medium

    exert a chemotactic and proinvasive effect on human ovarian

    cancer cells in vitro (9, 27). In the latter study, we also reported

    that exogenous SPARC significantly inhibited LPA-mediated

    invasiveness and proliferation of human ovarian cancer cells.

    These findings prompted us to test whether the proinvasive and

    FIGURE 7. Modulation of the activity of bioactive lipids in the ascitic fluid by host SPARC. Pooled SP+/+ and SP/ ascitic fluids with equivalent proteinconcentration were heated at 95jC for 10 min to inactivate the biologically active peptides and proteins. The effect of heated and unheated samples onfibronectin invasion (A) or migration (B) of ID8 cells was measured as described earlier. The chemotactic effect of heated and unheated SP/ and SP+/+

    ascitic fluids was tested by adding 600 AL of ascitic fluid to the bottom chamber of fibronectin-coated transwell inserts. ID8 cells (1 105/100 AL of DMEM-0.4% bovine serum albumin) were added to the upper chamber. After 5 h, cells that migrated to the undersurface of the inserts were stained and counted.*, P < 0.05, compared with unheated SP+/+ ascitic fluid. **, P < 0.05, between heated and unheated ascitic fluid. The proliferative effect of SP/ and SP+/+

    ascitic fluids on ID8 cells was determined by MTS assay (C). *, P < 0.05, compared with unheated SP+/+ ascitic fluid. **, P < 0.05, between heated andunheated ascitic fluid. Cell proliferation of ID8 cells in response to indicated concentrations of LPA, in the presence and absence of SPARC (10 Ag/mL), wasassessed by MTS assay (D). Columns, mean of three independent experiments done in quadruplicates; bars, SE. *, P < 0.05, compared with vehicle control.**, P < 0.05, compared with corresponding LPA stimulation.

    SPARC Normalizes Ovarian Cancer Microenvironment

    Mol Cancer Res 2007;5(10). October 2007

    1023

    American Association for Cancer ResearchCopyright 2007on February 8, 2011mcr.aacrjournals.orgDownloaded from

    DOI:10.1158/1541-7786.MCR-07-0001

    http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://mcr.aacrjournals.org/
  • 8/7/2019 SPARC for talk

    11/17

    chemotactic effects of SP+/+ and SP

    / ascitic fluids are

    mediated by bioactive lipids, especially LPA. We found that the

    proinvasive activity of ascitic fluids from intraperitoneal

    tumorbearing SP+/+ and SP

    / mice was significantly

    decreased (f23% and 27%, respectively) after heating at

    95jC for 10 min (Fig. 7A). A significant (f2-fold) difference

    in the proinvasive activity was also observed between heated

    and unheated SP+/+ and SP

    / ascitic fluids. Similarly, we

    found that the ascitic fluid from SP/ mice exerted a

    significantly more potent chemotactic activity effect on ID8

    cells than that of SP+/+ mice (Fig. 7B). As anticipated, heating

    significantly decreased this chemotactic effect (>33%). Fur-

    thermore, the mitogenic effect of ascitic fluids on ID8 cells was

    also significantly (f35%) inhibited by heating and revealed a

    statistically significant difference between the proliferative

    capacities of SP+/+ and SP

    / ascitic fluids (Fig. 7C). These

    results indicate that bioactive lipids representf60% to 65% of

    the activity of the ascitic fluid. The major identified component

    of these bioactive lipids was LPA, which influences almostevery aspect of ovarian cancer progression (7, 9). In agreement

    with our recent report (27), LPA was also shown to induce

    proliferation of ID8 cells directly in a concentration-dependent

    manner, and addition of exogenous SPARC (10 Amol/L)

    significantly diminished the proliferation induced by low

    concentrations (5-10 Amol/L) of LPA but had no significant

    effect when higher concentrations (>20 Amol/L) of LPA were

    used (Fig. 7D). These data suggest that the significantly

    augmented proinvasive, chemotactic, and mitogenic effects of

    SP/ ascitic fluid on ID8 cells are mediated, at least in part, by

    the biologically active lipids in the ascitic fluid, of which LPA

    is well recognized. Moreover, these pronounced effects

    contribute to the increased tumor burden in SP/ mice, which

    in turn increases LPA production not only from tumor cells but

    also from mesothelial cells and other stromal components of the

    ascitic fluid.

    SPARC Ameliorates the Peritoneal Tumor Induced

    Inflammation

    Ovarian cancer cells have been reported to producecytokines that further promote tumor survival and invasiveness

    FIGURE 8. SPARC ameliorates peritoneal tumor induced inflammation. Determination of the level of MCP-1 (A), 8-isoprostane (B), and IL-6 (C) inSP/ and SP+/+ ascitic fluids by ELISA. Columns, mean of eight SP/ and five SP+/+ ascitic fluid samples; bars, SE. *, P < 0.01, compared with SP+/+. D.Chemotactic effect of ascitic fluid on ID8 cells was determined as described in legend of Fig. 7B. IL-6 (50 ng/mL) was added with SP+/+ ascitic fluid and IL-6neutralizing antibody (IL-6 Ab, 50 Ag/mL) was preincubated with SP/ ascitic fluid for 30 min before being added to the bottom chambers. *, P < 0.05,compared with vehicle control treated cells. **, P < 0.01, compared with SP+/+. E. The proinvasive activity of IL-6 in the ascitic fluid was tested in afibronectin invasion assay. ID8 cells (1 105/100 AL ascitic fluid) were added to the upper chamber of fibronectin-coated inserts in the presence and absenceof IL-6 and IL-6 neutralizing antibody as described earlier. The bottom chambers contained DMEM-5% fetal bovine serum. Columns, mean of threeindependent experiments done in triplicates; bars, SE. *, P < 0.05, compared with control untreated ascitic fluid. **, P < 0.01, compared with SP+/+. F. Cellproliferation of ID8 cells in response to indicated concentrations of IL-6, in the presence and absence of SPARC (10 Ag/mL), was assessed by MTS assay.Columns, mean of three independent experiments done in quadruplicates; bars, SE. *, P < 0.05, compared with control cells that are not stimulated (NS). **,

    P < 0.05, compared with the corresponding IL-6 stimulation.

    Said et al.

    Mol Cancer Res 2007;5(10). October 2007

    1024

    American Association for Cancer ResearchCopyright 2007on February 8, 2011mcr.aacrjournals.orgDownloaded from

    DOI:10.1158/1541-7786.MCR-07-0001

    http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://mcr.aacrjournals.org/
  • 8/7/2019 SPARC for talk

    12/17

    (53). High interleukin-6 (IL-6) levels in the ascitic fluid of

    ovarian cancer patients were correlated with poor prognosis

    (6). Other studies have highlighted the crucial role of IL-6 in

    the pathogenesis of ovarian cancer (54, 55). In the present

    study, we found significantly higher levels of macrophage

    chemoattractant protein-1 (MCP-1; >2.5-fold) in SP/ ascitic

    fluid (Fig. 8A), and these levels coincided with the extensive

    macrophage infiltration in SP/ tumors and ascites shown in

    Fig. 2D, highlighting a potent anti-inflammatory effect for

    SPARC. We also found that the levels of arachidonic acid

    inflammatory mediator, 8-isoprostane, were significantly

    higher (4.3-fold) in SP/ ascitic fluid compared with SP

    +/+

    (Fig. 8B), indicating the hypoxic stress in the ascitic fluid

    microenvironment, which correlates with LPA, tumor burden,

    as well as inflammation. Consistently, ascitic fluid from SP/

    mice contained significantly (f2-fold) higher levels of IL-6

    as compared with SP+/+ fluid (Fig. 8C). These data suggest

    that SPARC, through down-regulation of MCP-1, decreases

    tumor infiltration by macrophages, and hence decreases IL-6and 8-isoprostane proinflammatory mediators in the ascitic

    fluid. Furthermore, our in vitro data indicated that the

    augmented chemotactic (Fig. 8D) and proinvasive (Fig. 8E)

    effects of SP/ over SP+/+ ascitic fluid were, in part, due to

    the high IL-6 content, as confirmed by restoration of these

    activities in SP+/+

    ascitic fluid by exogenous IL-6 and

    neutralizing them by IL-6 neutralizing antibody in SP/

    ascites. Moreover, IL-6 exerted a concentration-dependent

    increase in ID8 proliferation, which was shown to be inhibited

    by exogenous SPARC (Fig. 8F).

    DiscussionOf the numerous growth factors involved in the patho-

    genesis of ovarian cancer, the angiogenic growth factor VEGF

    plays a central role in the development of exudative asci-

    tic fluidthe hallmark of peritoneal ovarian carcinomatosis

    (41, 56). Cross talk between VEGF and integrin cell adhesion

    receptors is believed to play a critical role in modulation of

    tumor growth, survival, vascularization, invasion, and metas-

    tasis (51). The metastatic potential of tumor cells has been

    correlated with up-regulation of VEGF and VEGF-induced

    cell-surface expression and clustering of integrins including

    avh3, avh5, and h1 (35-37, 51, 57). Consistent with these

    reports, our findings in this study provide evidence that (a) the

    proadhesive and proinvasive effects of ascitic fluid are

    mediated, in part, through VEGF binding to its cognate

    receptors and activation of av and h1 integrins in ovarian

    cancer cells; (b) host SPARC negatively regulates MMP-2 and

    MMP-9 levels and activity in peritoneal tumors and ascitic

    fluid; (c) the enhanced adhesion of ovarian cancer cells to

    SP/ peritoneal mesothelial cells and their augmented

    invasion of fibronectin in response to SP/ ascitic fluid are

    mediated, in part, through the inhibitory effect of SPARC on

    integrin expression and activation of ovarian cancer cells,

    either directly or by modulation of levels and/or activity of

    soluble factors in the ascitic fluid; (d) SPARC inhibits VEGF-

    and integrin-mediated ovarian cancer cell proliferation in vitro,

    as well as VEGF-mediated mitogenic and tumorigenic effects

    in vivo; and (e) host SPARC ameliorates the peritoneal

    tumor induced inflammatory response that accompanies

    ovarian cancer through attenuation of levels and activity of

    bioactive lipids, IL-6, MCP-1, and 8-isoprostane proinflam-

    matory mediators (Fig. 9).

    VEGF has been shown to activate MMP-2 in ascitic fluid

    during peritoneal dissemination of ovarian cancer (58).Moreover, VEGF-regulated ovarian cancer invasion and

    migration was recently reported to involve expression and

    activation of MMP-2, MMP-7, and MMP-9 (59). Conversely,

    MMP-2 and MMP-9 have been implicated in formation of

    ascites through induction of the release of VEGF by ovarian

    carcinoma cells (41), suggesting the existence of a positive

    autocrine feedback loop between VEGF and MMPs. A link

    between integrins and the up-regulated MMPs in SP/ ascites

    may be suggested based on the earlier findings that avh3integrin binds MMP-2 on the surface of melanoma and

    endothelial cells and both colocalize in caveolae and promote

    tumor growth and angiogenesis in vivo (60). avh3 integrin

    also cooperates with MMP-9 by increasing migration of

    metastatic breast cancer cells, as avh3 integrin does not adhere

    to native, intact collagen but it does adhere to collagen that

    has undergone proteolytic degradation due to exposure of an

    RGD binding site (61). Furthermore, av and h1 integrins,

    through outside in and inside out integrin signaling, have

    been reported to induce the transcription of MMP genes in a

    variety of cancer cell lines, including ovarian cancer (refs. 62, 63

    and the references reviewed therein). This well-orchestrated

    interplay of VEGF-integrin-MMP axis in ascitic fluid favors the

    proinvasive and migratory phenotype of ovarian cancer cells

    after temporary ligand recognition and binding. It is therefore

    plausible to suggest that perturbation of this axis at the

    transcriptional and posttranscriptional levels by SPARC attenu-

    ates ovarian cancer cell motility, invasiveness, and metastasis.

    FIGURE 9. Hypothesized roles of SPARC in modulating signalingpathways involved in the pathogenesis of ovarian cancer. The pathwaysinvolved in peritoneal ovarian carcinomatosis are complex, interrelated,often synergistic, and mostly converge on stimulation of tumor cellsurvival, proliferation, and invasion. This schema summarizes the majorsignaling pathways involved in the pathogenesis of ovarian carcinomatosisand their proposed regulation by SPARC (SP). These various roles ofSPARC were hypothesized based on our findings in the present study aswell as our published studies using human cells and mouse tumor models.PGs, prostaglandins; ROS, reactive oxygen species; FN, fibronectin; VN,vitronectin.

    SPARC Normalizes Ovarian Cancer Microenvironment

    Mol Cancer Res 2007;5(10). October 2007

    1025

    American Association for Cancer ResearchCopyright 2007on February 8, 2011mcr.aacrjournals.orgDownloaded from

    DOI:10.1158/1541-7786.MCR-07-0001

    http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://mcr.aacrjournals.org/
  • 8/7/2019 SPARC for talk

    13/17

    LPA has been identified as an ovarian cancer promoting

    factor because high levels of this bioactive lipid have been

    reported in both plasma and ascites of ovarian cancer patients

    with stage I disease, suggesting that LPA promotes early events

    in ovarian carcinoma dissemination (64). Up-regulation of

    potent prosurvival, proangiogenic, as well as proinflammatory

    cytokines and chemokines has been reported downstream of

    LPA activity in vivo and in vitro (6, 7, 65, 66). LPA is

    constitutively produced by peritoneal mesothelial cells and has

    been shown to be crucial for the chemotactic activity of shed

    ovarian cancer cells (9, 27). Moreover, LPA enhances motility

    and invasiveness of ovarian cancer cells, in part, through up-

    regulation and activation of MMPs in ovarian cancer cells (67).

    Through binding to its cognate receptor, LPA has been shown

    to synergize with signals induced by h1 integrin and activation

    of EGF or platelet-derived growth factor receptors to modulate

    cellular migration (68). Accordingly, our results in the present

    study suggest that LPA- and VEGF-induced integrin activation

    may function via convergent signal transduction pathways topromote MMP expression and/or processing, resulting in

    significant augmentations in the migratory and invasive

    behavior of ovarian cancer cells. Moreover, the mitogenic and

    prosurvival effects of LPA have been attributed to its direct

    effect on cell cycle progression, up-regulation of VEGF,

    transactivation of receptor tyrosine kinases (e.g., EGFR and

    VEGFRs), and induction of an inflammatory response (66). Our

    recent report on significant attenuation of the LPA-mediated

    crosstalk between mesothelial cells and human ovarian cancer

    cell lines by exogenous addition or ectopic overexpression of

    SPARC (27), as well as significant diminutions in proliferation,

    migration, and invasion of bioactive lipids in ascites of ovarian

    tumorbearing SP+/+

    mice, highlights the important role ofSPARC in suppression of LPA-mediated events in ovarian

    carcinoma.

    In ovarian cancer, tumor cells are not the only source of

    MMPs and proinflammtory cytokines. Tumor-associated mono-

    cytes/macrophages are recruited from peripheral blood mono-

    cytes in response to chemokines produced by tumor cells,

    mainly through the action of MCP-1 (CCL2; refs. 69, 70). As a

    consequence, tumor-associated monocytes/macrophages under-

    go marked phenotypic changes with activation of hypoxia-

    inducible factor 1a, dramatically up-regulating the expression

    of a large number of genes encoding mitogenic, proangiogenic,

    and prometastatic cytokines, enzymes, and bioactive lipids

    (70, 71). Interestingly, in vivo and in vitro studies have shown

    that VEGF, MMPs, and IL-6 in the tumor microenvironment

    inhibit the differentiation and maturation of dendritic cells and

    switch their differentiation toward the macrophage lineage

    (72, 73). Accordingly, the infiltration of malignant ascitic fluid

    and solid peritoneal tumors with macrophages in our syngeneic

    ovarian cancer model was directly correlated with the levels of

    ascitic fluid VEGF, MMPs, MCP-1, and IL-6. Our results that

    SP/ tumor-bearing mice had higher ascitic fluid MCP-1 and

    IL-6 levels and more extensive macrophage infiltration than

    SP+/+ mice further validate a negative regulatory role for host

    SPARCin thedevelopmentof ovarian carcinomatosis. In support

    of these findings, we have recently shown that IL-6 and MCP-1

    production by human ovarian cancer cell lines is enhanced by

    LPA stimulation and down-regulated by SPARC (27).

    In this study, further evidence for the anti-inflammatory

    role of host SPARC is provided by the dramatic increase in

    8-isoprostane (prostaglandin F2a) levels in SP/ ascitic

    fluid, compared with SP+/+ counterparts. Isoprostanes,

    including 8-isoprostane, are produced by the action of

    cyclooxygenase enzymes (Cox-1 and Cox-2) on arachidonic

    acid via a free radicalcatalyzed mechanism of phospholipid

    peroxidation, and have been associated with oxidative stress

    and high levels of LPA (74, 75). LPA has recently been

    shown to up-regulate Cox-1 and Cox-2 expression in ovarian

    cancer, with subsequent up-regulation of MMPs and growth

    factors, enhancing ovarian cancer cell survival and invasive-

    ness (66). Furthermore, in a recent study by Rask et al. (76),

    the expression of Cox-1 and Cox-2 in ovarian cancer, as well

    as their downstream prostaglandin E2, was associated with

    the high grade of ovarian cancer, suggesting an important

    role in their malignant transformation and progression. The

    observation that Cox-1 and Cox-2, as well as prostaglandin

    E2, were expressed in the tumor cells as well as in stroma ledthe authors to suggest a paracrine signaling mechanism

    mediated by growth factors, cytokines, and possibly prosta-

    glandins due to the interactions between tumor cells and

    stromal cells (fibroblasts and immune cells). Moreover,

    cytosolic phospholipase A2 (cPLA2) has been implicated in

    LPA production in ascitic fluid and was associated with the

    invasive metastatic phenotype (77). Recently, it has been

    reported that under the hypoxic conditions encountered in

    ovarian cancer ascitic fluid, LPA up-regulates cytosolic

    phospholipase A2 and augments the metastatic phenotype

    through a hypoxia-inducible factor 1a dependent pathway

    (78). Taken together, we can speculate that the up-regulation

    of 8-isoprostane in SP/

    ascitic fluid is attributed to theactivation and up-regulation of cLPA and the downstream Cox

    enzymes by LPA in the hypoxic microenvironment created by

    the rapidly proliferating tumor cells and other infiltrating

    inflammatory cells. To the best of our knowledge, this is the

    first report implicating the involvement of 8-isoprostane

    (prostaglandin F2a) in the ascitic fluid of ovarian cancer and

    its down-regulation by host SPARC. The anti-inflammatory

    effects of SPARC have previously been implicated in SP/

    mice as evidenced by decreased leukocyte infiltration in

    bleomycin-induced peritonitis (79), attenuated 12-O-tetradeca-

    noylphorbol-13-acetate induced skin inflammation (24), as

    well as lung and pancreatic tumors, and have been attributed,

    at least in part, to abundance of less dense, immature collagen

    fibrils and a more malleable extracellular matrix (19, 31).

    Lastly, SPARC has also been shown to negatively regulate the

    availability and/or activity of proinflammatory/angiogenic

    cytokines including platelet-derived growth factor (80),

    fibroblast growth factor-2 (81), and VEGF (82). Collectively,

    these data strongly suggest a negative regulatory role for host

    SPARC in the inflammatory process that accompanies ovarian

    cancer.

    In summary, we have provided evidence that SPARC,

    through interference with the VEGF-integrin-MMP axis as

    well as that of the inflammatory mediators/intermediates in

    the malignant ascitic fluid, plays a crucial role in the norma-

    lization of the reactive microenvironment of peritoneal ovarian

    carcinomatosis. These findings, in combination with the

    Said et al.

    Mol Cancer Res 2007;5(10). October 2007

    1026

    American Association for Cancer ResearchCopyright 2007on February 8, 2011mcr.aacrjournals.orgDownloaded from

    DOI:10.1158/1541-7786.MCR-07-0001

    http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://mcr.aacrjournals.org/
  • 8/7/2019 SPARC for talk

    14/17

    established antiproliferative, proapoptotic, and antimetastatic

    properties of SPARC in ovarian cancer, underscore its

    therapeutic potential as a promising novel inhibitor of

    peritoneal ovarian carcinomatosis.

    Materials and MethodsCell Culture

    ID8, a cell line that was derived from spontaneous in vitro

    malignant transformation of C57BL/6 mouse ovarian surface

    epithelial cells (83) and was stably transfected with a retroviral

    vector expressing green fluorescent protein (GFP) or VEGF164plus GFP (ID8-VEGF; ref. 39), was kindly provided by

    Dr. George Coukos (Abramson Family Cancer Research

    Institute, University of Pennsylvania, Philadelphia, PA). Cells

    were maintained in DMEM, supplemented with 2 mmol/L

    L-glutamine (Sigma), 5% fetal bovine serum (Atlanta Bio-

    logicals), and 100 units/mL penicillin and 100 Ag/mL

    streptomycin (Sigma).

    Reagents and Antibodies

    SPARC from mouse parietal yolk sac was purchased from

    Sigma. Bovine osteonectin was purchased from Haematologic

    Technologies. Collagen I from rat tail, human plasma fibronec-

    tin, and vitronectin were from Sigma. Integrin-activating and

    integrin-blocking antibodies, rabbit anti-VEGF neutralizing

    antibody, rabbit anticytokeratin 5, anticytokeratin 18, and

    anti-vimentin antibodies were from Chemicon. Polyclonal

    goat antibodies against MMP-2, MMP-9, TIMP-1, TIMP-2,

    and integrins were from Santa Cruz Biotechnology, Inc. Rat

    anti-mouse macrophage marker F4/80 was from Serotec,

    Inc. Rat anti-mouse CD31 was from Chemicon. MMP inhibitor

    kit II and VEGF inhibitors were from Calbiochem. Lysopho-

    spahtidic acid (LPA) and RGD peptides were purchased from

    Sigma. Recombinant murine IL-6 and anti-mouse IL-6 anti-

    bodies were from RayBiotech. All other chemicals were of

    analytic grade and were purchased from Sigma and Fisher

    Scientific.

    Mice

    C57BL/6 129SvJ SPARC-null (SP/) and wild-type

    (SP+/+) mice (6-8 weeks old) were a kind gift of Dr. Helene

    Sage (Hope Heart Program, Benaroya Research Institute at

    Virginia Mason, Seattle, WA). Mice were backcrossed against

    wild-type C57BL/6 mice for at least six generations beforeuse in tumor studies. All experimental procedures were

    approved by the Laboratory Animal Services of the Medical

    College of Georgia and were done in specific pathogen-free

    facilities.

    In vivo Tumor Generation

    Intraperitoneal and subcutaneous tumors were generated by

    injection of ID8 and ID8-VEGF cells into SP+/+ and SP/

    mice (n = 10 per group) as previously described (25, 39).

    Animal survival and tumor development were monitored twice

    weekly. At the end of the experimental period, animals were

    sacrificed and ascitic fluid and tumor tissues from the animals

    were collected and processed as previously reported (25, 39).

    RNA Isolation and Reverse Transcription-PCR

    RNA was isolated from tissues and cells with Trizol reagent

    (Invitrogen) and was purified with RNeasy isolation kit

    (Qiagen). Total RNA (2 Ag) was reverse transcribed using

    Improm II RT enzyme kit (Promega) and cDNA was amplified

    using Jumpstart Taq polymerase (Sigma) as recommended by

    the manufacturers. Specific oligonucleotide primers were

    synthesized according to published sequences (Table 1). PCR

    products were resolved in 2% agarose gels and were visualized

    with ethidium bromide staining under UV light. Gel imaging,

    documentation, and analysis were done with a Kodak Gel Logic

    100 imaging system equipped with Kodak D 3.6 software

    (Eastman-Kodak).

    Immunohistochemistry

    The expression of MMP-2, MMP-9, TIMP-1, TIMP-2, and

    F4/80 was determined in deparaffinized tumor sections after

    antigen retrieval with AutoZyme (BioMeda Corp.). Sections

    were incubated with specific primary antibodies overnight at

    4jC, washed with PBS-0.2% Tween 20, and incubated for

    1 h with peroxidase-labeled appropriate secondary antibodies

    (Jackson ImmunoResearch laboratories, Inc.). Sections were

    then developed with Vectastain ABC Elite kit (Vector

    Laboratories) and stable 3,3-diaminobenzidene (ResGen),

    counterstained with hematoxylin, and mounted in Permount

    (Fisher Scientific). Images were acquired with Leica micro-

    scope (DM5000) equipped with a Q-Imaging digital camera.

    The mean vascular density in tumor sections was determined

    by immunostaining with anti-CD31 antibody and averaging the

    TABLE 1. PCR Primers Used in Semiquantitative ReverseTranscription-PCR

    Primer Sequence Cycles

    rpS6

    F: 5-AAGCTCCGCACCTTCTATGAGA-3 24R: 5-TGACTGGACTCAGACTTAGAAGTAGAAGC-3

    MMP-2F: 5-TTGGATATTTGCAATGCAGCC-3 35R: 5-AAGGTTGAAGGAAACGAGCGA-3

    MMP-9

    F: 5-CCATTTCGACGACGAGT-3 35R: 5-CCAAATTTGCCGTCCTTATCGTA-3

    TIMP-1

    F: 5-CTGTGGGAAATGCCGATA-3 30R: 5-TGAGCAGGGCTCAGAGTACGC-3

    TIMP-2F: 5-TCATCAGGGCCAAGTTCGTG-3 30

    R: 5-GGATCATGGGACAGCGAGTGA-3 h3 integrin

    F: 5-CTGGTAAAAACGCCGTGAAT-3 35R: 5-GAGTAGCAAGGCCAATGAGC-3

    h1 integrin

    F: 5-GCCAGGGCTGGTTATACAGA-3 35R: 5-TCACAATGGCACACAGGTTT-3

    h5 integrinF: 5-TAGCTAGGCACGCACGGATAAT-3 35

    R: 5-TTTGTACACACAGACTCTTCCGGATA-3 av integrin

    F: 5-AAACGTGTCCGACCACCTC-3 35

    R: 5-CTTTACAATCCTAATTGCCCTAACG-3a5 integrin

    5-TCCTATCCAGTGCACCACCATT-3 355-GCAACAAAGTCTTCTGTGTCATCAC-3

    SPARC Normalizes Ovarian Cancer Microenvironment

    Mol Cancer Res 2007;5(10). October 2007

    1027

    American Association for Cancer ResearchCopyright 2007on February 8, 2011mcr.aacrjournals.orgDownloaded from

    DOI:10.1158/1541-7786.MCR-07-0001

    http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://mcr.aacrjournals.org/
  • 8/7/2019 SPARC for talk

    15/17

    number of positively stained vessels in five microscopic

    fields per section of tumors from at least four different animals

    (n = 4).

    Adhesion AssaysAdhesion assays were done as previously described (25).

    Harvested cells were preincubated with and without MMP

    inhibitors (40 Amol/L), VEGF inhibitors (40 Ag/mL), integrin

    blocking antibodies (10 Ag/mL), SPARC (10 Ag/mL), or RGD

    peptides (20 Amol/L) followed by stimulation with VEGF164(40 ng/mL), integrin activating antibody (5-10 Ag/mL), MnCl2(1-5 mmol/L), IL-6 (5-50 ng/mL), LPA (5-50 Amol/L), or

    ascitic fluid. Cell suspensions were then added to ligand-coated

    plates. After a 4-h incubation at 37jC/5% CO2, nonadherent

    cells were washed out and adherent cells were stained with

    Hemacolor 3 stain (Fisher Scientific) as recommended by the

    manufacturer. Cell adhesion was determined by measuring the

    absorbance at 590 nm.

    Isolation of Murine Peritoneal Mesothelial Cells

    Murine peritoneal mesothelial cells were isolated from SP+/+

    and SP/ mice by peritoneal lavage and from collagenase/

    dispase-digested explants from the mesentery, as previously

    described (84). The digested tissue was placed into collagen

    Icoated plates and cultured for 2 days in DMEM containing

    12% fetal bovine serum and EGF (20 ng/mL). Mesothelial cells

    used for these studies (passages 1-2) were confirmed to be

    positive for the expression of cytokeratin 5, cytokeratin 18, and

    vimentin, as determined by immunocytochemical staining.

    Adhesion of ID8-GFP Cells to SP+/+ and SP/ Peritoneal

    Explants In vitro

    Fresh peritoneal tissues were excised and prepared as

    previously described (85). Briefly, 6 6-mm2 peritoneal pieces

    were dissected from the diaphragmatic peritoneum and the

    peritoneum lining the anterior abdominal wall and were placed

    in 24-well plates with the mesothelium facing upward. ID8-

    GFP cells (5 105/500 AL of DMEM-0.4% bovine serum

    albumin) were added onto peritoneal pieces in each well. Plates

    were then incubated for 72 h in a humidified incubator at 37jC/

    5% CO2. At the end of the incubation period, peritoneal pieces

    were rinsed thrice in PBS to remove nonadherent cells. Adherent

    cells were visualized under a fluorescent stereomicroscope,

    photographed, and the mean green fluorescence intensity in six

    fields per experimental condition was determined.

    Invasion Assays

    Invasion assays were done as previously described (25).

    Harvested cells were treated exactly as described in adhesion

    assays and cell suspensions were added to the upper chamber of

    polycarbonate inserts (8-Am pore size, Corning Costar) pre-

    coated with fibronectin, collagen I, or vitronectin. After a

    5-h incubation at 37jC/5% CO2, cells in the upper chamber

    were removed by Q-tips (Fisher Scientific). Cells attached to

    the underside of the inserts (the invasive cells) were stained

    with Hemacolor 3 stain and counted under an inverted

    microscope equipped with DFC 320 digital camera (Leica)

    under200 magnification.

    ELISA

    Mouse IL-6 ELISA kit (RayBiotech), MCP-1 ELISA kit

    (BD Biosciences), and 8-isoprostane EIA kits (Cayman

    Chemical) were used to determine IL-6, MCP-1, and 8-

    isoprostane levels in ascitic fluid samples, respectively,

    according to the manufacturers instructions.

    Immunoblotting

    Tumor tissues from SP+/+ and SP

    / animals (n = 4) were

    homogenized in lysis buffer [20 mmol/L Tris (pH 7.4),

    150 mmol/L NaCl, 1 mmol/L EDTA, 50 mmol/L NaF, 0.5%

    sodium deoxycholate, 1% NP40, 1 mmol/L Na3VO4, and

    1 protease inhibitor cocktail mixture] as previously described

    (25). One hundred micrograms of cell lysates were resolved

    by 10% SDS-PAGE under nonreducing conditions and

    transferred onto polyvinylidene difluoride membranes (Bio-

    Rad). The membranes were then incubated overnight at 4jC

    with antimouse specific integrin antibodies (Santa Cruz Bio-

    technology) and monoclonal antiglyceraldehyde-3-phosphate

    dehydrogenase (Ambion) as loading control. Protein detection

    was carried out with horseradish peroxidase conjugated

    secondary antibodies and a SuperSignal West Dura Chemilu-

    minescence kit (Pierce).

    Nonradioactive Cell Proliferation Assay

    CellTiter96 kit (Promega) was used according to the

    manufacturers instructions. The number of proliferating cells

    was determined colorimetrically by measuring the absorbance

    at 590 nm (A590) of the dissolved formazan product after the

    addition of 3-(4,5-dimethyl-thiazol-2yl)-5-(3-carboxymethoxy-

    phenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) for 3 h.

    Statistical Analysis

    Statistical analysis was done using GraphPad Prism version

    3.1 for Windows (GraphPad Software). Survival curves were

    generated using the Kaplan-Meier method followed by log-rank

    test for comparison of curves. The concentration-dependent

    studies were done by one-way ANOVA followed by Newman-

    Keuls multiple comparison test. All other statistical analyses

    were determined by Students t test. Differences were

    considered significant at P < 0.05.

    AcknowledgmentsWe thank Dr. George Coukos for providing ID8-GFP and ID8-VEGF-GFP cells

    and Dr. E. Helene Sage for providing SP/ and SP+/+ mice.

    References1. Kohn EC, Travers LA, Kassis J, Broome U, Klominek J. Malignant effusionsare sources of fibronectin and other promigratory and proinvasive components.

    Diagn Cytopathol 2005;33:300 8.

    2. Kassis J, Klominek J, Kohn EC. Tumor microenvironment: what can effusions

    teach us? Diagn Cytopathol 2005;33:316 9.

    3. Mukherjee TM, Swift JG, Henderson DW. Freeze-fracture study of

    intercellular junctions in benign and malignant mesothelial cells in effusionsand a comparison with those seen in pleural mesotheliomas (solid tumour).J Submicrosc Cytol Pathol 1988;20:195 208.

    4. Fiegl M, Massoner A, Haun M, et al. Sensitive detection of tumour cells ineffusions by combining cytology and fluorescence in situ hybridisation (FISH).

    Br J Cancer 2004;91:558 63.

    5. Fiegl M, Haun M, Massoner A, et al. Combination of cytology, fluorescence

    in situ hybridization for aneuploidy, and reverse-transcriptase polymerase chain

    Said et al.

    Mol Cancer Res 2007;5(10). October 2007

    1028

    American Association for Cancer ResearchCopyright 2007on February 8, 2011mcr.aacrjournals.orgDownloaded from

    DOI:10.1158/1541-7786.MCR-07-0001

    http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://mcr.aacrjournals.org/
  • 8/7/2019 SPARC for talk

    16/17

    reaction for human mammaglobin/mammaglobin B expression improves

    diagnosis of malignant effusions. J Clin Oncol 2004;22:474 83.

    6. Chou CH, Wei LH, Kuo ML, et al. Up-regulation of interleukin-6 in human

    ovarian cancer cell via a Gi/PI3K-Akt/NF-nB pathway by lysophosphatidic acid,an ovarian cancer-activating factor. Carcinogenesis 2005;26:45 52.

    7. Fang X, Yu S, Bast RC, et al. Mechanisms for lysophosphatidic acid-induced

    cytokine production in ovarian cancer cells. J Biol Chem 2004;279:9653 61.

    8. Bian D, Mahanivong C, Yu J, et al. The G12/13-RhoA signaling pathway

    contributes to efficient lysophosphatidic acid-stimulated cell migration. Oncogene2006;25:2234 44.

    9. Ren J, Xiao YJ, Singh LS, et al. Lysophosphatidic acid is constitutively

    produced by human peritoneal mesothelial cells and enhances adhesion,migration, and invasion of ovarian cancer cells. Cancer Res 2006;66:3006 14.

    10. Yamada T, Sato K, Komachi M, et al. Lysophosphatidic acid (LPA) inmalignant ascites stimulates motility of human pancreatic cancer cells throughLPA1. J Biol Chem 2004;279:6595 605.

    11. Kobayashi H. Suppression of urokinase expression and tumor metastasis bybikunin overexpression [mini-review]. Hum Cell 2001;14:233 6.

    12. Fishman DA, Bafetti LM, Stack MS. Membrane-type matrix metalloprotei-nase expression and matrix metalloproteinase-2 activation in primary humanovarian epithelial carcinoma cells. Invasion Metastasis 1996;16:150 9.

    13. Liotta LA, Kohn EC. Stromal therapy: the next step in ovarian cancertreatment. J Natl Cancer Inst 2002;94:1113 4.

    14. Chu CS, Woo EY, Toll AJ, et al. Tumor-associated macrophages as a sourceof functional dendritic cells in ovarian cancer patients. Clin Immunol 2002;102:

    291301.

    15. Zavadova E, Loercher A, Verstovsek S, Verschraegen CF, Micksche M,

    Freedman RS. The role of macrophages in antitumor defense of patients withovarian cancer. Hematol Oncol Clin North Am 1999;13:135 44.

    16. Brekken RA, Sage EH. SPARC, a matricellular protein: at the crossroads ofcell-matrix communication. Matrix Biol 2001;19:816 27.

    17. Framson PE, Sage EH. SPARC and tumor growth: where the seed meets thesoil? J Cell Biochem 2004;92:679 90.

    18. Brekken RA, Sage EH. SPARC, a matricellular protein: at the crossroads ofcell-matrix. Matrix Biol 2000;19:569 80.

    19. Brekken RA, Puolakkainen P, Graves DC, Workman G, Lubkin SR, SageEH. Enhanced growth of tumors in SPARC null mice is associated with changesin the ECM. J Clin Invest 2003;111:487 95.

    20. Chlenski A, Liu S, Crawford SE, et al. SPARC is a key Schwannian-derivedinhibitor controlling neuroblastoma tumor angiogenesis. Cancer Res 2002;62:

    735763.

    21. Ledda F, Bravo AI, Adris S, Bover L, Mordoh J, Podhajcer OL. Theexpression of the secreted protein acidic and rich in cysteine (SPARC) is

    associated with the neoplastic progression of human melanoma. J Invest Dermatol

    1997;108:210 214.

    22. De S, Chen J, Narizhneva NV, et al. Molecular pathway for cancer metastasis

    to bone. J Biol Chem 2003;278:39044 50.

    23. Chlenski A, Liu S, Guerrero LJ, et al. SPARC expression is associated withimpaired tumor growth, inhibited angiogenesis and changes in the extracellular

    matrix. Int J Cancer 2006;118:310 6.

    24. Sangaletti S, Stoppacciaro A, Guiducci C, Torrisi MR, Colombo MP.

    Leukocyte, rather than tumor-produced SPARC, determines stroma and collagentype IV deposition in mammary carcinoma. J Exp Med 2003;198:1475 85.

    25. Said N, Motamed K. Absence of host-secreted protein acidic and rich incysteine (SPARC) augments peritoneal ovarian carcinomatosis. Am J Pathol

    2005;167:1739 52.

    26. Yiu GK, Chan WY, Ng SW, et al. SPARC (secreted protein acidic and rich in

    cysteine) inducesapoptosis in ovariancancercells. Am J Pathol2001;159:609 22.

    27. Said NA, Najwer I, Socha MJ, et al. SPARC inhibits LPA-mediated

    mesothelial-ovarian cancer cell crosstalk. Neoplasia 2007;9:23 35.

    28. Said N, Najwer I, Motamed K. Secreted protein acidic and rich in cysteine

    (SPARC) inhibits integrin-mediated adhesion and growth factor-dependentsurvival signaling in ovarian cancer. Am J Pathol 2007;170:1054 63.

    29. Gilles C, Bassuk JA, Pulyaeva H, Sage EH, Foidart JM, Thompson EW.SPARC/osteonectin induces matrix metalloproteinase 2 activation in humanbreast cancer cell lines. Cancer Res 1998;58:5529 36.

    30. Koblinski JE, Kaplan-Singer BR, VanOsdol SJ, et al. Endogenousosteonectin/SPARC/BM-40 expression inhibits MDA-MB-231 breast cancer cell

    metastasis. Cancer Res 2005;65:7370 7.

    31. Puolakkainen PA, Brekken RA, Muneer S, Sage EH. Enhanced growth of

    pancreatic tumors in SPARC-null mice is associated with decreased deposition of

    extracellular matrix and reduced tumor cell apoptosis. Mol Cancer Res 2004;2:

    21524.

    32. Chlenski A, Guerrero LJ, Yang Q, et al. SPARC enhances tumor stromaformation and prevents fibroblast activation. Oncogene 2007;26:4513 22.

    33. Chlenski A, Liu S, Baker LJ, et al. Neuroblastoma angiogenesis is inhibitedwith a folded synthetic molecule corresponding to the epidermal growth factor-

    like module of the follistatin domain of SPARC. Cancer Res 2004;64:7420 5.

    34. Koukourakis MI, Giatromanolaki A, Brekken RA, et al. Enhanced expression

    of SPARC/osteonectin in the tumor-associated stroma of non-small cell lungcancer is correlated with markers of hypoxia/acidity and with poor prognosis ofpatients. Cancer Res 2003;63:5376 80.

    35. Chen J, De S, Brainard J, Byzova TV. Metastatic properties of prostate cancercells are controlled by VEGF. Cell Commun Adhes 2004;11:1 11.

    36. De S, Razorenova O, McCabe NP, OToole T, Qin J, Byzova TV. VEGF-integrin interplay controls tumor growth and vascularization. Proc Natl Acad Sci

    U S A 2005;102:7589 94.

    37. Byzova TV, Goldman CK, Pampori N, et al. A mechanism for modula-

    tion of cellular responses to VEGF: activation of the integrins. Mol Cell 2000;6:85160.

    38. Zhang L, Yang N, Park JW, et al. Tumor-derived vascular endothelial growthfactor up-regulates angiopoietin-2 in host endothelium and destabilizes host

    vasculature, supporting angiogenesis in ovarian cancer. Cancer Res 2003;63:

    340312.39. Zhang L, Yang N, Garcia JR, et al. Generation of a syngeneic mouse model

    to study the effects of vascular endothelial growth factor in ovarian carcinoma.

    Am J Pathol 2002;161:2295 309.

    40. Taverna S, Ghersi G, Ginestra A, et al. Shedding of membrane vesicles

    mediates fibroblast growth factor-2 release from cells. J Biol Chem 2003;278:51911 9.

    41. Belotti D, Paganoni P, Manenti L, et al. Matrix metalloproteinases (MMP9

    and MMP2) induce the release of vascular endothelial growth factor (VEGF) byovarian carcinoma cells: implications for ascites formation. Cancer Res 2003;63:

    52249.

    42. Graves LE, Ariztia EV, Navari JR, Matzel HJ, Stack MS, Fishman DA.

    Proinvasive properties of ovarian cancer ascites-derived membrane vesicles.Cancer Res 2004;64:7045 9.

    43. Huang S, Van Arsdall M, Tedjarati S, et al. Contributions of stromal

    metalloproteinase-9 to angiogenesis and growth of human ovarian carcinoma inmice. J Natl Cancer Inst 2002;94:1134 42.

    44. Janat-Amsbury MM, Yockman JW, Anderson ML, Kieback DG, Kim SW.Comparison of ID8 MOSE and VEGF-modified ID8 cell l ines in an

    immunocompetent animal model for human ovarian cancer. Anticancer Res2006;26:2785 9.

    45. Davidson B, Goldberg I, Gotlieb WH, et al. Coordinated expression of

    integrin subunits, matrix metalloproteinases (MMP), angiogenic genes and Ets

    transcription factors in advanced-stage ovarian carcinoma: a possible activationpathway? Cancer Metastasis Rev 2003;22:103 15.

    46. Davidson B, Goldberg I, Gotlieb WH, et al. The prognostic value of

    metalloproteinases and angiogenic factors in ovarian carcinoma. Mol CellEndocrinol 2002;187:39 45.

    47. Boyce EA, Kohn EC. Ovarian cancer in the proteomics era: diagnosis,prognosis, and therapeutics targets. Int J Gynecol Cancer 2005;15 Suppl 3:

    26673.

    48. Lessan K, Aguiar DJ, Oegema T, Siebenson L, Skubitz AP. CD44 andh1integrin mediate ovarian carcinoma cell adhesion to peritoneal mesothelial cells.

    Am J Pathol 1999;154:1525 37.

    49. Cannistra SA, Kansas GS, Niloff J, DeFranzo B, Kim Y, Ottensmeier C.Binding of ovarian cancer cells to peritoneal mesotheliumin vitro is partly

    mediated by CD44H. Cancer Res 1993;53:3830 8.

    50. Strobel T, Cannistra SA. h1-integrins partly mediate binding of ovarian

    cancer cells to peritoneal mesotheliumin vitro. Gynecol Oncol 1999;73:362 7.

    51. Byzova TV, Kim W, Midura RJ, Plow EF. Activation of integrina(V)h(3)regulates cell adhesion and migration to bone sialoprotein. Exp Cell Res 2000;

    254:299 308.

    52. Byzova T. Integrins in bone recognition and metastasis. J Musculoskelet

    Neuronal Interact 2004;4:374.

    53. Costanzo ES, Lutgendorf SK, Sood AK, Anderson B, Sorosky J, LubaroffDM. Psychosocial factors and interleukin-6 among women with advanced ovarian

    cancer. Cancer 2005;104:305 13.

    54. Kryczek I, Grybos M, Karabon L, Klimczak A, Lange A. IL-6 production in

    ovarian carcinoma is associated with histiotype and biological characteristics of

    the tumour and influences local immunity. Br J Cancer 2000;82:621 8.

    SPARC Normalizes Ovarian Cancer Microenvironment

    Mol Cancer Res 2007;5(10). October 2007

    1029

    American Association for Cancer ResearchCopyright 2007on February 8, 2011mcr.aacrjournals.orgDownloaded from

    DOI:10.1158/1541-7786.MCR-07-0001

    http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://www.aacr.org/http://mcr.aacrjournals.org/
  • 8/7/2019 SPARC for talk

    17/17

    55. Yao V, Platell C, Hall JC. Peritoneal mesothelial cells produce inflammatory

    related cytokines. ANZ J Surg 2004;74:997 1002.

    56. Laird AD, Vajkoczy P, Shawver LK, et al. SU6668 is a potent antiangiogenic

    and antitumor agent that induces regression of established tumors. Cancer Res2000;60:4152 60.

    57. Byzova TV, Plow EF. Activation of avh3 on vascular cells controls

    recognition of prothrombin. J Cell Biol 1998;143:2081 92.

    58. Yabushita H, Shimazu M, Noguchi M, et al. Vascular endothelial growthfactor activating matrix metalloproteinase in ascitic fluid during peritonealdissemination of ovarian cancer. Oncol Rep 2003;10:89 95.

    59. Wang FQ, So J, Reierstad S, Fishman DA. Vascular endothelial growthfactor-regulated ovarian cancer invasion and migration involves expression and

    activation of matrix metalloproteinases. Int J Cancer 2006;118:879 88.

    60. Stefanidakis M, Koivunen E. Cell-surface association between matrix

    metalloproteinases and integrins: role of the complexes in leukocyte migrationand cancer progression. Blood 2006;108:1441 50.

    61. Ahmed N, Riley C, Oliva K, Stutt E, Rice GE, Quinn MA. Integrin-linkedkinase expression increases with ovarian tumour grade and is sustained by

    peritoneal tumour fluid. J Pathol 2003;201:229 37.

    62. Ahmed N, Pansino F, Clyde R, et al. Overexpression ofa(v)h6 integrin inserous epithelial ovarian cancer regulates extracellular matrix degradation via the

    plasminogen activation cascade. Carcinogenesis 2002;23:237 44.

    63. Curran S, Murray GI. Matrix metalloproteinases: molecular aspects of theirroles in tumour invasion and metastasis. Eur J Cancer 2000;36:1621 30.

    64. Xu Y, Gaudette DC, Boynton JD, et al. Characterization of an ovarian cancer

    activating factor in ascites from ovarian cancer patients. Clin Cancer Res 1995;1:122332.

    65. Huang MC, Lee HY, Yeh CC, Kong Y, Zaloudek CJ, Goetzl EJ. Induction ofprotein growth factor systems in the ovaries of transgenic mice overexpressing

    human type 2 lysophosphatidic acid G protein-coupled receptor (LPA2).Oncogene 2004;23:122 9.

    66. Symowicz J, Adley BP, Woo MMM, Auersperg N, Hudson LG, Stack MS.Cyclooxygenase-2 functions as a downstream mediator of lysophosphatidic acidto promote aggressive behavior in ovarian carcinoma cells. Cancer Res 2005;65:

    223442.

    67. Fishman DA, Liu Y, Ellerbroek SM, Stack MS. Lysophosphatidic acid

    promotes matrix metalloproteinase (MMP) activation and MMP-dependentinvasion in ovarian cancer cells. Cancer Res 2001;61:3194 9.

    68. Sakai T, de la Pena JM, Mosher DF. Synergism among lysophosphatidic acid,h1A integrins, and epidermal growth factor or platelet-derived growth factor inmediation of cell migration. J Biol Chem 1999;274:15480 6.

    69. Milliken D, Scotton C, Raju S, Balkwill F, Wilson J. Analysis of chemokinesand chemokine receptor expression in ovarian cancer ascites. Clin Cancer Res

    2002;8:1108 14.

    70. Murdoch C, Lewis CE. Macrophage migration and gene expression in

    response to tumor hypoxia. Int J Cancer 2005;117:701 8.

    71. Duyndam MC, Hilhorst MC, Schluper HM, et al. Vascular endothelial

    growth factor-165 overexpression stimulates angiogenesis and induces cyst

    formation and macrophage infiltration in human ovarian cancer xenografts. Am JPathol 2002;160:537 48.

    72. Gabrilovich D, Ishida T, Oyama T, et al. Vascular endothelial growth factorinhibits the development of dendritic cells and dramatically affects thedifferentiation of multiple hematopoietic lineages in vivo . Blood 1998;92:

    415066.

    73. Menetrier-Caux C, Montmain G, Dieu MC, et al. Inhibition of thedifferentiation of dendritic cells from CD34(+) progenitors by tumor cells: role

    of interleukin-6 and macrophage colony-stimulating factor. Blood 1998;92:

    477891.

    74. Brault S, Gobeil F, Jr., Fortier A, et al. Lysophosphatidic acid inducesendothelial cell death by modulating the redox environment. Am J Physiol Regul

    Integr Comp Physiol 2007;292:R1174 83.

    75. Siess W. Platelet interaction with bioactive lipids formed by mild oxidation oflow-density lipoprotein. Pathophysiol Haemost Thromb 2006;35:292 304.

    76. Rask K, Zhu Y, Wang W, Hedin L, Sundfeldt K. Ovarian epithelial cancer: arole for PGE2 synthesis and signalling in malignant transformation and

    progression. Mol Cancer 2006;5:62.

    77. Sengupta S, Xiao YJ, Xu Y. A novel laminin-induced LPA autocrine loop inthe migration of ovarian cancer cells. FASEB J 2003;17:1570 2.

    78. Kim KS, Sengupta S, Berk M, et al. Hypoxia enhances lysophosphatidic acid

    responsiveness in ovarian cancer cells and lysophosphatidic acid induces ovariantumor metastasis in vivo. Cancer Res 2006;66:7983 90.

    79. Savani RC, Zhou Z, Arguiri E, et al. Bleomycin-induced pulmonary injuryin mice deficient in SPARC. Am J Physiol Lung Cell Mol Physiol 2000;279:

    L743 750.

    80. Raines EW, Lane TF, Iruela-Arispe ML, Ross R, Sage EH. The extracellular

    glycoprotein SPARC interacts with platelet-derived growth factor (PDGF)-ABand -BB and inhibits the binding of PDGF to its receptors. Proc Natl Acad Sci

    U S A 1992;89:1281 5.

    81. Motamed K, Blake DJ, Angello JC, et al. Fibroblast growth factor receptor-1

    mediates the inhibition of endothelial cell proliferation and the promotion ofskeletal myoblast differentiation by SPARC: a role for protein kinase A. J CellBiochem 2003;90:408 23.

    82. Kupprion C, Motamed K, Sage EH. SPARC (BM-40, osteonectin) inhibitsthe mitogenic effect of vascular endothelial growth factor on microvascular

    endothelial cells. J Biol Chem 1998;273:29635 40.

    83. Roby KF, Taylor CC, Sweetwood JP, et al. Development of a syngeneicmouse model for events related to ovarian cancer. Carcinogenesis 2000;21:

    58591.

    84. Muller J, Yoshida T. Interaction of murine peritoneal leukocytes and

    mesothelial cells: in vitro model system to survey cellular events on serosalmembranes during inflammation. Clin Immunol Immunopathol 1995;75:231 8.

    85. Groothuis PG, Koks CAM, de Goeij AFPM, Dunselman GAJ, Arends JW,Evers JLH. Adhesion of human endometrial fragments to peritoneum in vitro.Fertil Steril 1999;71:1119 24.

    Said et al.1030

    DOI:10.1158/1541-7786.MCR-07-0001