Mesothelin binding to CA125/MUC16 promotes pancreatic cancer cell motility and invasion via MMP-7 activation

Shih-Hsun Chen, Wei-Chien Hung, Pu Wang, Colin Paul, Konstantinos Konstantopoulos, Shih-Hsun Chen, Wei-Chien Hung, Pu Wang, Colin Paul, Konstantinos Konstantopoulos

Abstract

Mesothelin (MSLN) and cancer antigen125/mucin 16 (CA125/MUC16) are potential biomarkers for pancreatic cancer (PC) that are co-overexpressed at the invading edges of PC tissues, and their expression correlates with poor survival rates. However, the role of MSLN-MUC16 molecular interaction in PC cell motility and invasion has yet to be elucidated. Using sophisticated bioengineering and molecular biology tools, we report that the binding of MSLN to MUC16 markedly enhances PC cell motility and invasion via the selective induction of matrix metalloproteinase (MMP)-7. MSLN-mediated MMP-7 upregulation in MUC16-expressing PC cells occurs via a p38 MAPK-dependent pathway. Depletion of MMP-7 or inhibition of p38 activity abolishes MSLN-mediated PC motility and invasion. These findings provide a novel perspective on the enhanced invasive potential associated with MSLN and MUC16 co-overexpression, and the mechanism underlying MMP-7 activation in PC invasion and metastasis.

Figures

Figure 1. MUC16 expression correlates with pancreatic…
Figure 1. MUC16 expression correlates with pancreatic cancer cell binding capacity to MSLN.
(A) hTERT-HPNE, HPDE, PANC-1, SW1990, and Pa03C cells were seeded in 96-well plates pre-coated with either mesothelin (MSLN) or BSA (control). After 30 min of incubation, the relative number of adherent cells to MSLN- versus BSA-coated wells was quantified by WST-1 assay. Data represent the mean ± S.E. of three independent experiments. *, p < 0.05 with respect to control. (B) Representative flow cytometric histograms of MUC16 surface expression on hTERT-HPNE, HPDE, PANC-1, SW1990, and Pa03C cells using indirect single-color immunofluorescence and an anti-MUC16 mAb or an isotype control. (C) MUC16-overexpressing SW1990 or Pa03C cells were pre-incubated with either a function-blocking anti-MUC16 mAb M11 (20 μg/ml) or an isotype control for 1 h at 37°C, and then used in the cell binding assay as described in (A). (D) Scramble control or MUC16-KD SW1990 and Pa03C cells were seeded in 96-well plates pre-coated with MSLN or BSA (control). After 30 min of incubation, adherent cells were quantified by WST-1 assay. Data represent the mean ± S.E. of three independent experiments. *, p < 0.05 with respect to MUC16-KD cells.
Figure 2. MSLN and MUC16 co-immunoprecipitate in…
Figure 2. MSLN and MUC16 co-immunoprecipitate in pancreatic cancer cells.
Whole lysates from scramble control (A) or MUC16-KD (B) SW1990 and Pa03C cells were immunoprecipitated with either an anti-MUC16 or anti-MSLN or an isotype control antibody, and analyzed for MSLN or MUC16 reactivity by immunoblotting. Lysates from SW1990 and Pa03C cells were also analyzed as input controls.
Figure 3. MUC16 expression correlates with MSLN-mediated…
Figure 3. MUC16 expression correlates with MSLN-mediated MMP-7 induction in pancreatic cancer cells.
hTERT-HPNE, HPDE, PANC-1, SW1990, and Pa03C cells were starved in serum-free medium and incubated in the presence or absence of MSLN (1 μg/ml) overnight. (A) MMP-7, (B) MMP-2, and (C) MMP-9 mRNA levels were then determined by qRT-PCR. GAPDH served as internal control. Data represent the mean ± S.E. of at least three independent experiments. *, p < 0.01 with respect to corresponding control. (D) MUC16, MSLN, and MMP-7 expression in pancreatic cancer tissue specimens was determined by immunohistochemical staining using an anti-MUC16, anti-MSLN, or anti-MMP-7 antibody, respectively. Strong immunostaining was detected in ∼ 90% of cancerous tissues, whereas no immunoreactivity was present in the ducts or acini of normal pancreas tissue.
Figure 4. MSLN-MUC16 interaction induces MMP-7 synthesis…
Figure 4. MSLN-MUC16 interaction induces MMP-7 synthesis in pancreatic cancer cells.
(A) SW1990 cells were starved in serum-free medium overnight and incubated with MSLN (1 μg/ml) for the indicated periods of time. The mRNA levels of MMP-7 were quantified by qRT-PCR. GAPDH served as internal control. Data represent the mean ± S.E. of at least three independent experiments. *, p < 0.05 with respect to cells treated with MSLN (1 μg/ml) for 0 h. MMP-7 protein expression from SW1990 cell lysates was analyzed using Western blotting. β-actin served as an internal control. (B) SW1990 cells were starved in serum-free medium overnight and incubated with different concentrations of MSLN (0–2.0 μg/ml) for 12 h. MMP-7 mRNA and protein expression were then analyzed by qRT-PCR and Western blotting, respectively. Data from qRT-PCR represent the mean ± S.E. of three independent experiments. *, p < 0.05 with respect to untreated control cells. (C, D) Scramble control or MUC16-KD SW1990 and Pa03C cells were serum-starved overnight and then incubated for 12 h in the presence or absence of MSLN (1 μg/ml). MMP-7 mRNA (C) and protein (D) levels were quantified by qRT-PCR and immunoblotting, respectively. Data represent the mean ± S.E. of at least three independent experiments. *, p < 0.01 and §, p < 0.05 with respect to the corresponding scramble control cells. (E) Conditioned media from scramble control or MUC16-KD pancreatic cancer cells pre-treated with MSLN (1 μg/ml) for 12 h were collected and analyzed for MMP-7 secretion. Data are reported as the fold of MMP-7 levels detected in the conditioned medium of each cell line compared to that in the medium of untreated scramble control cells. Data represent the mean ± S.E. of three independent experiments. *, p < 0.05 with respect to corresponding untreated scramble control cells.
Figure 5. MSLN-MUC16 interaction promotes pancreatic cancer…
Figure 5. MSLN-MUC16 interaction promotes pancreatic cancer cell invasion via MMP-7 activation.
Scramble control or MUC16-KD SW1990 (A) or Pa03C (B) cells were incubated with MSLN (1 μg/ml) in the presence of either a neutralizing MMP-7 antibody (2 μg/ml) or an isotype control and then used in transwell invasion assays. Data are reported as normalized cell number that invaded through the transwell membrane relative to untreated scramble control (100%), and represent the mean ± S.E. of three independent experiments. **, p < 0.01; ***, p < 0.001 (one-way ANOVA followed by Tukey test). (C) SW1990 and Pa03C pancreatic cancer cells were incubated with either human recombinant active MMP-7 (2 μg/ml) or vehicle control, and then used in transwell invasion assays. Data are reported as normalized cell number that invaded through the membrane relative to vehicle control-treated cells (100%), and represent the mean ± S.E. of three independent experiments. *, p < 0.05 with respect to vehicle control cells (Student's t-test).
Figure 6. MSLN binding to MUC16 promotes…
Figure 6. MSLN binding to MUC16 promotes pancreatic cancer cell motility via MMP-7 activation.
Representative time-lapse images of scramble control and MUC16-KD SW1990 (A) and Pa03C (C) pancreatic cancer cells migrating through channels of prescribed dimensions (WxHxL = 6 × 10 × 200 μm) using 10% FBS as chemoattractant. In all experiments, cells were incubated with or without MSLN (1 μg/ml) for 6 h before being seeded to the microchannel device. The average migration speed of individual scramble control and MUC16-KD SW1990 (B) and Pa03C (D) cells was determined over a 10 h period for > 30 cells from three independent experiments. *, p < 0.05 with respect to scramble control and MSLN-treated MUC16-KD cells. (E) The average migration speed of MSLN-stimulated SW1990 and Pa03C cells in the presence of either an MMP-7 neutralizing antibody (2 μg/ml) or an isotype control was quantified for > 30 cells from three independent experiments. *, p < 0.05 with respect to the control treatment. (F) The average migration speed of SW1990 and Pa03C cells in the presence or absence of human recombinant active MMP-7 (2 μg/ml) was quantified for > 30 cells from three independent experiments. *, p < 0.05 with respect to untreated control.
Figure 7. MSLN-MUC16 interaction promotes pancreatic cancer…
Figure 7. MSLN-MUC16 interaction promotes pancreatic cancer cell motility and invasion via MMP-7 induced by a p38 MAPK-dependent pathway.
(A) Scramble control and MUC16-KD SW1990 cells were serum-starved overnight, and then treated with MSLN (1 μg/ml) for 12 h. The levels of phospho-ERK1/2 (Thr202/Tyr204) and phospho-p38 MAPK (Thr180/Tyr182) from whole cell lysates were analyzed by immunoblotting using specific Abs. Equal loading in each lane was ensured by the similar intensities of total ERK1/2, p38 MAPK, and β-actin. (B) SW1990 cells were incubated with MSLN (1 μg/ml) for 12 h in the presence or absence of the p38 MAPK inhibitor SB203580 (20 μM). The levels of phospho-p38 MAPK (Thr180/Tyr182) and MMP-7 expression from cell lysates were analyzed by Western blotting using specific Abs. Equal loading in each lane is ensured by the similar intensities of total p38 MAPK and β-actin. These Western blots are representative of three independent experiments, all revealing similar results. The intensity of bands was quantified using the NIH ImageJ software and then normalized with respect to the value obtained for the untreated control. (C, D) SW1990 cells were stimulated with MSLN (1 μg/ml) in the presence or absence of SB203580 (20 μM) or an MMP-7 antisense oligonucleotide (50 nM), and then subjected to either transwell invasion or microchannel motility assays. (C) Data are reported as percentage of untreated control cells that invaded through the transwell membrane, and represent the mean ± S.E. of three independent experiments. (D) The average migration speed of individual cells was determined over a 10 h period for > 30 cells from three independent experiments for each of the indicated conditions. *, p < 0.01.
Figure 8. Proposed molecular pathway induced by…
Figure 8. Proposed molecular pathway induced by MSLN-MUC16 binding, which results in MMP-7 induction in pancreatic cancer cells.
The binding of MSLN to MUC16 present on the pancreatic cancer cell surface activates a p38 MAPK-dependent pathway, which in turn upregulates MMP-7 synthesis, resulting in increased invasive and migratory potentials. In the absence of MUC16 expression by pancreatic cancer cells, MSLN is capable of upregulating MMP-7 expression via activation of an ERK-dependent pathway.

References

    1. Chang K. & Pastan I. Molecular cloning of mesothelin, a differentiation antigen present on mesothelium, mesotheliomas, and ovarian cancers. Proc Natl Acad Sci U S A 93, 136–40 (1996).
    1. Argani P. et al. Mesothelin is overexpressed in the vast majority of ductal adenocarcinomas of the pancreas: identification of a new pancreatic cancer marker by serial analysis of gene expression (SAGE). Clin Cancer Res 7, 3862–8 (2001).
    1. Hassan R. et al. Mesothelin is overexpressed in pancreaticobiliary adenocarcinomas but not in normal pancreas and chronic pancreatitis. Am J Clin Pathol 124, 838–45 (2005).
    1. Huang C. Y. et al. Serum mesothelin in epithelial ovarian carcinoma: a new screening marker and prognostic factor. Anticancer Res 26, 4721–8 (2006).
    1. Johnston F. M. et al. Circulating mesothelin protein and cellular antimesothelin immunity in patients with pancreatic cancer. Clin Cancer Res 15, 6511–8 (2009).
    1. Li M. et al. Mesothelin is a malignant factor and therapeutic vaccine target for pancreatic cancer. Mol Cancer Ther 7, 286–96 (2008).
    1. Chang M. C. et al. Mesothelin enhances invasion of ovarian cancer by inducing MMP-7 through MAPK/ERK and JNK pathways. Biochem J 442, 293–302 (2012).
    1. Servais E. L. et al. Mesothelin overexpression promotes mesothelioma cell invasion and MMP-9 secretion in an orthotopic mouse model and in epithelioid pleural mesothelioma patients. Clin Cancer Res 18, 2478–89 (2012).
    1. Wang K. et al. Inhibition of mesothelin as a novel strategy for targeting cancer cells. PLoS One 7, e33214 (2012).
    1. Rump A. et al. Binding of ovarian cancer antigen CA125/MUC16 to mesothelin mediates cell adhesion. J Biol Chem 279, 9190–8 (2004).
    1. Gubbels J. A. et al. Mesothelin-MUC16 binding is a high affinity, N-glycan dependent interaction that facilitates peritoneal metastasis of ovarian tumors. Mol Cancer 5, 50 (2006).
    1. Kaneko O. et al. A binding domain on mesothelin for CA125/MUC16. J Biol Chem 284, 3739–49 (2009).
    1. O'Brien T. J. et al. The CA 125 gene: an extracellular superstructure dominated by repeat sequences. Tumour Biol 22, 348–66 (2001).
    1. Kui Wong N. et al. Characterization of the oligosaccharides associated with the human ovarian tumor marker CA125. J Biol Chem 278, 28619–34 (2003).
    1. Streppel M. M. et al. Mucin 16 (cancer antigen 125) expression in human tissues and cell lines and correlation with clinical outcome in adenocarcinomas of the pancreas, esophagus, stomach, and colon. Hum Pathol 43, 1755–63 (2012).
    1. Haridas D. et al. Pathobiological implications of MUC16 expression in pancreatic cancer. PLoS One 6, e26839 (2011).
    1. Markman M., Federico M., Liu P. Y., Hannigan E. & Alberts D. Significance of early changes in the serum CA-125 antigen level on overall survival in advanced ovarian cancer. Gynecol Oncol 103, 195–8 (2006).
    1. Bast R. C. Jr et al. CA 125: the past and the future. Int J Biol Markers 13, 179–87 (1998).
    1. Shimizu A. et al. Coexpression of MUC16 and mesothelin is related to the invasion process in pancreatic ductal adenocarcinoma. Cancer Sci 103, 739–46 (2012).
    1. Einama T. et al. Co-expression of mesothelin and CA125 correlates with unfavorable patient outcome in pancreatic ductal adenocarcinoma. Pancreas 40, 1276–82 (2011).
    1. Yamamoto H. et al. Expression of matrix metalloproteinases and tissue inhibitors of metalloproteinases in human pancreatic adenocarcinomas: clinicopathologic and prognostic significance of matrilysin expression. J Clin Oncol 19, 1118–27 (2001).
    1. Bloomston M., Zervos E. E. & Rosemurgy A. S. 2nd Matrix metalloproteinases and their role in pancreatic cancer: a review of preclinical studies and clinical trials. Ann Surg Oncol 9, 668–74 (2002).
    1. Gress T. M. et al. Expression and in-situ localization of genes coding for extracellular matrix proteins and extracellular matrix degrading proteases in pancreatic cancer. Int J Cancer 62, 407–13 (1995).
    1. Crawford H. C., Scoggins C. R., Washington M. K., Matrisian L. M. & Leach S. D. Matrix metalloproteinase-7 is expressed by pancreatic cancer precursors and regulates acinar-to-ductal metaplasia in exocrine pancreas. J Clin Invest 109, 1437–44 (2002).
    1. Fukushima H. et al. Association of matrilysin mRNA expression with K-ras mutations and progression in pancreatic ductal adenocarcinomas. Carcinogenesis 22, 1049–52 (2001).
    1. Jones L. E., Humphreys M. J., Campbell F., Neoptolemos J. P. & Boyd M. T. Comprehensive analysis of matrix metalloproteinase and tissue inhibitor expression in pancreatic cancer: increased expression of matrix metalloproteinase-7 predicts poor survival. Clin Cancer Res 10, 2832–45 (2004).
    1. Chen S. H., Dallas M. R., Balzer E. M. & Konstantopoulos K. Mucin 16 is a functional selectin ligand on pancreatic cancer cells. FASEB J 26, 1349–59 (2012).
    1. Balzer E. M. et al. Physical confinement alters tumor cell adhesion and migration phenotypes. FASEB J 26, 4045–56 (2012).
    1. Tong Z. et al. Chemotaxis of cell populations through confined spaces at single-cell resolution. PLoS One 7, e29211 (2012).
    1. Veit C. et al. Activation of phosphatidylinositol 3-kinase and extracellular signal-regulated kinase is required for glial cell line-derived neurotrophic factor-induced migration and invasion of pancreatic carcinoma cells. Cancer Res 64, 5291–300 (2004).
    1. Singh A. K. et al. p38beta MAP kinase as a therapeutic target for pancreatic cancer. Chem Biol Drug Des 80, 266–73 (2012).
    1. Cwik G., Wallner G., Skoczylas T., Ciechanski A. & Zinkiewicz K. Cancer antigens 19-9 and 125 in the differential diagnosis of pancreatic mass lesions. Arch Surg 141, 968–73; discussion 974 (2006).
    1. Sato N. et al. Frequent hypomethylation of multiple genes overexpressed in pancreatic ductal adenocarcinoma. Cancer Res 63, 4158–66 (2003).
    1. Hucl T. et al. High cancer-specific expression of mesothelin (MSLN) is attributable to an upstream enhancer containing a transcription enhancer factor dependent MCAT motif. Cancer Res 67, 9055–65 (2007).
    1. Sternlicht M. D. & Werb Z. How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol 17, 463–516 (2001).
    1. Friedl P. & Alexander S. Cancer invasion and the microenvironment: plasticity and reciprocity. Cell 147, 992–1009 (2011).
    1. Remy L., Trespeuch C., Bachy S., Scoazec J. Y. & Rousselle P. Matrilysin 1 influences colon carcinoma cell migration by cleavage of the laminin-5 beta3 chain. Cancer Res 66, 11228–37 (2006).
    1. Wroblewski L. E. et al. Stimulation of MMP-7 (matrilysin) by Helicobacter pylori in human gastric epithelial cells: role in epithelial cell migration. J Cell Sci 116, 3017–26 (2003).
    1. Tan X. et al. Involvement of matrix metalloproteinase-7 in invasion-metastasis through induction of cell dissociation in pancreatic cancer. Int J Oncol 26, 1283–9 (2005).
    1. Jones S. et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 321, 1801–6 (2008).
    1. Dallas M. R. et al. Sialofucosylated podocalyxin is a functional E- and L-selectin ligand expressed by metastatic pancreatic cancer cells. Am J Physiol Cell Physiol 303, C616–24 (2012).
    1. Thomas S. N., Zhu F., Schnaar R. L., Alves C. S. & Konstantopoulos K. Carcinoembryonic antigen and CD44 variant isoforms cooperate to mediate colon carcinoma cell adhesion to E- and L-selectin in shear flow. J Biol Chem 283, 15647–55 (2008).
    1. Burdick M. M. & Konstantopoulos K. Platelet-induced enhancement of LS174T colon carcinoma and THP-1 monocytoid cell adhesion to vascular endothelium under flow. Am J Physiol Cell Physiol 287, C539–47 (2004).
    1. McCarty O. J., Jadhav S., Burdick M. M., Bell W. R. & Konstantopoulos K. Fluid shear regulates the kinetics and molecular mechanisms of activation-dependent platelet binding to colon carcinoma cells. Biophys J 83, 836–48 (2002).
    1. Wang P., Zhu F., Tong Z. & Konstantopoulos K. Response of chondrocytes to shear stress: antagonistic effects of the binding partners Toll-like receptor 4 and caveolin-1. Faseb J 25, 3401–15 (2011).
    1. Zhu F., Wang P., Kontrogianni-Konstantopoulos A. & Konstantopoulos K. Prostaglandin (PG)D(2) and 15-deoxy-Delta(12,14)-PGJ(2), but not PGE(2), mediate shear-induced chondrocyte apoptosis via protein kinase A-dependent regulation of polo-like kinases. Cell Death Differ 17, 1325–34 (2010).
    1. Thomas S. N., Schnaar R. L. & Konstantopoulos K. Podocalyxin-like protein is an E-/L-selectin ligand on colon carcinoma cells: comparative biochemical properties of selectin ligands in host and tumor cells. Am J Physiol Cell Physiol 296, C505–13 (2009).
    1. Hanley W. D. et al. Variant isoforms of CD44 are P- and L-selectin ligands on colon carcinoma cells. FASEB J 20, 337–9 (2006).
    1. Napier S. L., Healy Z. R., Schnaar R. L. & Konstantopoulos K. Selectin ligand expression regulates the initial vascular interactions of colon carcinoma cells: the roles of CD44v and alternative sialofucosylated selectin ligands. J Biol Chem 282, 3433–41 (2007).
    1. Wang P., Zhu F. & Konstantopoulos K. Interleukin-6 synthesis in human chondrocytes is regulated via the antagonistic actions of prostaglandin (PG)E2 and 15-deoxy-Delta(12,14)-PGJ2. PLoS One 6, e27630 (2011).

Source: PubMed

3
Suscribir