Caveolin-1 (Cav-1), a principal structural component of caveolar membrane domains, contributes to cancer development but its precise functional roles and regulation remain unclear. In this study, we determined the oncogenic function of Cav-1 in preclinical models of pancreatic cancer and in human tissue specimens. Cav-1 expression levels correlated with metastatic potential and epithelial–mesenchymal transition (EMT) in both mouse and human pancreatic cancer cells. Elevated levels in cells promoted EMT, migration, invasion, and metastasis in animal models, whereas RNA interference (RNAi)-mediated knockdown inhibited these processes. We determined that levels of Cav-1 and the Forkhead transcription factor FoxM1 correlated directly in pancreatic cancer cells and tumor tissues. Enforced expression of FoxM1 increased Cav-1 levels, whereas RNAi-mediated knockdown of FoxM1 had the opposite effect. FoxM1 directly bound to the promoter region of Cav-1 gene and positively transactivated its activity. Collectively, our findings defined Cav-1 as an important downstream oncogenic target of FoxM1, suggesting that dysregulated signaling of this novel FoxM1-Cav-1 pathway promotes pancreatic cancer development and progression. Cancer Res; 72(3); 655–65. ©2011 AACR.

Pancreatic cancer represents one of the leading causes of cancer deaths in industrialized countries with an estimated 43,140 new cases and 36,800 deaths occurring annually in the United States (1). At presentation, the disease has, in most cases, already spread locally and to distant organs that precludes a resection, and only about 5% to 25% of cases constitute candidates for surgical resection. Among patients undergoing a potentially curative resection, the long-term outcome remains unsatisfactory due to early recurrence and metastatic disease (2). Effective systemic therapy capable of reversing the aggressive biology of this disease is currently not available. Therefore, there is an urgent need for a better understanding of the molecular mechanisms underlying pancreatic cancer pathogenesis (3, 4).

Caveolin-1 (Cav-1) is the major structural protein in caveolae; 50- to 100-nm omega-shaped invaginations of the plasma membrane. Caveolae are involved in signal transduction, wherein Cav-1 acts as a scaffold to organize multiple molecular complexes regulating a variety of cellular events (5). Recent studies have suggested that Cav-1 may play important roles in cancer development and progression (6–10). Paradoxically, Cav-1 seems to impact both positively and negatively on cancer biology (6), although recent studies suggest a positive association between Cav-1 expression and pancreatic cancer poor prognosis (11–13). Specifically, tumor cells in pancreatic cancer show significantly higher Cav-1 expression relative to tumor stroma, and Cav-1 knockdown significantly induces cell apoptosis and enhances the radiosensitivity of pancreatic cancer cells (14, 15). Furthermore, Cav-1 expression could be detected in the fibroblasts of the desmoplastic pancreatic cancer stroma but not in stromal cells of the normal pancreas. Importantly, coexpression of Cav-1 and fatty acid synthase significantly correlates with clinical features and survival status of patients with pancreatic cancer showing that Cav-1 and other factors may functionally cooperate in the process of pancreatic tumorigenesis (16). All of this experimental and clinical evidence supports that Cav-1 is an oncogene in pancreatic cancer and may be good candidate prognostic markers and targets for therapeutic intervention. Interestingly, researchers have shown that Cav-1 expression can be upregulated during epithelial–mesenchymal transition (EMT), and furthermore, once expressed, Cav-1 can influence cancer cell motility (17). Moreover, Cav-1 overexpression increased motility and invasion of human hepatocellular carcinoma cells through downregulation of E-cadherin while upregulation of vimentin showing the important roles of Cav-1 in EMT and metastasis of human cancer (18). However, the precise function and underlying mechanisms of Cav-1 in pancreatic cancer EMT and metastasis remain unclear and how Cav-1 expression is regulated is unknown.

FoxM1, a transcription factor of the Forkhead box (Fox) protein superfamily, is a key regulator of both G1–S and G2–M phases of the cell cycle and mitotic spindle integrity (19). Recent studies have strongly suggested that FoxM1 is oncogenic and plays important roles in angiogenesis, invasion, and metastasis (20, 21). Interestingly, FoxM1 may regulate EMT phenotype of pancreatic cancer cells by activation of mesenchymal cell markers (22) whereas the underlying mechanisms are unknown. In this study, we sought to determine the role of Cav-1 in pancreatic cancer EMT and metastasis and regulatory function of FoxM1 in Cav-1 expression and function. We discovered that the novel FoxM1-Cav-1 signaling pathway critically regulates pancreatic cancer EMT, invasion, and metastasis.

Cell lines and culture conditions

The human pancreatic adenocarcinoma cell lines AsPC-1, CaPan-1, MiaPaca-2, and PANC-1 were purchased from the American Type Culture Collection; MDA Panc-28, and MDA Panc-48 were gifts from Dr. Paul J. Chiao (MD Anderson Cancer Center, Houston, TX); and COLO357 human pancreatic adenocarcinoma metastasis and its fast growing and liver metastatic variant L3.3 and L3.7 in nude mice and the murine ductal adenocarcinoma cell line Panc02 and its highly metastatic variant Panc02-H7 were described previously (23–25). All of these cell lines were maintained in plastic flasks as adherent monolayers in Eagle's minimal essential medium supplemented with 10% FBS, sodium pyruvate, nonessential amino acids, l-glutamine, and vitamin solution (Flow Laboratories). The immortalized normal human pancreatic ductal epithelial (HPDE) cell line (provided by Dr. Tsao, Ontario Cancer Institute, Toronto, ON, Canada) was maintained in keratinocyte serum-free medium supplemented with epidermal growth factor and bovine pituitary extract (Invitrogen).

Western blot analysis

Standard Western blotting was carried out using whole-cell protein lysates and primary antibodies against FoxM1 (Santa Cruz Biotechnology), Cav-1, E-cadherin and vimentin (BD Biosciences), β-catenin and N-cadherin (Cell Signaling Technology); and a secondary antibody (anti-rabbit IgG or anti-mouse IgG; Santa Cruz Biotechnology). Equal protein sample loading was monitored using an anti-GAPDH antibody (Santa Cruz Biotechnology).

Plasmids and siRNAs

The plasmids pcDNA3.1-FoxM1b, pcDNA3.1-caveolin-1, and control vector pcDNA3.1 were described previously (26–28). The siRNA sequences targeting FoxM1 and Cav-1 were as follows: FoxM1, CUCUUCUCCCUCAGAUAUAdTdT (29); Cav-1, AGACGAGCUGAGCGAGAAGCAtt (30).

Transient transfection

Transfection of plasmids and siRNAs into pancreatic cancer cells was carried out using Lipofectamine LTX and Lipofectamine 2000 CD (Invitrogen), respectively. For transient transfection, cells were transfected with plasmids or siRNA at different doses as indicated for 48 hours before functional assays were carried out. Pancreatic cancer cells treated with transfection reagent alone were included as mock controls.

Promoter reporter and dual luciferase assay

The Cav-1 promoter luciferase reporter pA3Luc-caveolin-1 (pLuc-Cav) was described previously (28). Pancreatic cancer cells were transfected with the indicated pLuc-Cav, siRNAs, or specific gene expression plasmids. The Cav-1 promoter activity was normalized by cotransfection with a β-actin/Renilla luciferase reporter containing a full-length Renilla luciferase gene (31). Luciferase activity in the cells was quantified using a Dual Luciferase Assay System (Promega) 24 hours after transfection.

Human tissue samples and immunohistochemical analysis

Cav-1 and FoxM1 expression was analyzed using human pancreatic cancer and normal tissue microarray (US Biomax). The use of the tissue samples was approved by the Institutional Review Board of MD Anderson Cancer Center. Standard immunohistochemical procedures were carried out using anti-FoxM1 antibody (Santa Cruz Biotechnology) and anti-Cav-1 antibody (BD Biosciences). The staining results were scored by 2 investigators blinded to the clinical data as described previously (32). For negative controls, the primary antibody was omitted and replaced by negative IgG (Supplementary Fig. S1).

Statistics

The significance of the data from patient specimens was determined by the Pearson correlation coefficient. The significance of the in vitro data and in vivo data was determined by Student t test (2-tailed), Mann–Whitney test (2-tailed), or one-way ANOVA. P < 0.05 was considered significant.

Reverse transcription-PCR, chromatin immunoprecipitation assay, cell scratch wound assay, cell migration assay, cell invasion assay, cell immunofluorescence, animals, subcutaneous tumor growth, and experimental liver metastases

For experimental liver metastases, refer to the work of Marvin and colleagues (33). Details are described in the Supplementary Materials and Methods.

Cav-1 overexpression and its direct association with pancreatic cancer pathologic features

We first investigated the expression of Cav-1 protein in the 70 primary pancreatic tumor and 10 normal pancreatic tissue specimens in a tissue microarray. We observed Cav-1–positive staining in the membrane and cytoplasm of the tumor cells and tumor stroma with Cav-1–negative or weak Cav-1–positive staining in adjacent normal pancreatic cells and normal pancreatic cells (Fig. 1A and Supplementary Fig. S2). Increasing Cav-1 expression correlated with decreased level of tumor differentiation and significant difference between well (grade I) versus poorly differentiated (grade III) tumors (Fig. 1B and Supplementary Fig. S3). In addition, Cav-1 expression was positively correlated with disease stage showing that Cav-1 is upregulated in late-stage tumors. This association was significant between stage I versus stage IV tumors (Fig. 1C and Supplementary Fig. S3). Moreover, Cav-1 expression in lymph node metastasis or distant metastasis specimens was significantly higher than that in nonmetastasis specimens (Fig. 1D and Supplementary Fig. S3). These findings strongly indicated that Cav-1 expression plays a critical role in pancreatic cancer development and progression and is a valuable biomarker for this disease.

Figure 1.

Cav-1 expression in pancreatic cancer specimens and its association with pancreatic cancer pathologic features. A, tissue microarray (TMA) immunostaining using a specific antibody against Cav-1. Representative images of Cav-1 protein expression in normal pancreatic tissue, adjacent normal pancreatic tissue, and pancreatic cancer tissue are shown (200×). Note that the majority of the normal and adjacent normal pancreatic tissue cells are negative for Cav-1 expression, whereas pancreatic cancer tissue cells and stroma are strongly positive for Cav-1 expression. B, Cav-1 expression was positively correlated with tumor differentiation (*, P < 0.01 in a comparison of the grade III with the grade I or II) and representative images of grade I and III tumors are shown (200×), the numbers of samples of grade I, II, and III were 10, 41, and 19, respectively. C, Cav-1 expression was positively correlated with disease stage (*, P < 0.01 in a comparison of the stage III, IV with the stage I, II) and representative images of stage I and IV tumors are shown (200×), the numbers of samples of stage I, II, III, and IV are 35, 26, 3, and 6, respectively. D, Cav-1 expression was positively correlated with tumor metastasis (*, P < 0.01 in a comparison of the metastasis group with the non-metastasis group) and representative images of tumors from patients with or without distant metastasis are shown (200×).

Figure 1.

Cav-1 expression in pancreatic cancer specimens and its association with pancreatic cancer pathologic features. A, tissue microarray (TMA) immunostaining using a specific antibody against Cav-1. Representative images of Cav-1 protein expression in normal pancreatic tissue, adjacent normal pancreatic tissue, and pancreatic cancer tissue are shown (200×). Note that the majority of the normal and adjacent normal pancreatic tissue cells are negative for Cav-1 expression, whereas pancreatic cancer tissue cells and stroma are strongly positive for Cav-1 expression. B, Cav-1 expression was positively correlated with tumor differentiation (*, P < 0.01 in a comparison of the grade III with the grade I or II) and representative images of grade I and III tumors are shown (200×), the numbers of samples of grade I, II, and III were 10, 41, and 19, respectively. C, Cav-1 expression was positively correlated with disease stage (*, P < 0.01 in a comparison of the stage III, IV with the stage I, II) and representative images of stage I and IV tumors are shown (200×), the numbers of samples of stage I, II, III, and IV are 35, 26, 3, and 6, respectively. D, Cav-1 expression was positively correlated with tumor metastasis (*, P < 0.01 in a comparison of the metastasis group with the non-metastasis group) and representative images of tumors from patients with or without distant metastasis are shown (200×).

Close modal

Association of Cav-1 expression with metastatic potential and EMT phenotype

Cav-1 is markedly upregulated in most pancreatic cancer cell lines as compared with a transformed HPDE cell line (Fig. 2A). Then, we carried out cell immunofluorescence and Western blot analyses for Cav-1 in the poorly metastatic human and mouse pancreatic cancer cell lines (COLO357, Panc02) as well as highly metastatic human and mouse pancreatic cancer cell lines (L3.7, Panc02-H7). The levels of Cav-1 expression directly correlated with metastatic ability (Fig. 2B and D and Supplementary Fig. S4A). We further observed the cell morphology under invert phase-contrast microscope. The poorly metastatic cells (COLO357, Panc02) exhibited a typical epithelial morphology, whereas the highly metastatic cells (L3.7, Panc02-H7) exhibited a typical mesenchymal morphology (Fig. 2C). The unique morphology changes were consistent with the expression of EMT markers, that is, the expression of epithelial marker (E-cadherin, β-catenin) in the poorly metastatic cells (COLO357, Panc02) was higher than that in the highly metastatic cells (L3.7, Panc02-H7; Fig. 2D). In contrast, the expression of mesenchymal marker (vimentin, N-cadherin) in the poorly metastatic cells (COLO357, Panc02) was lower than that in the highly metastatic cells (L3.7, Panc02-H7; Fig. 2D). These results strongly indicated that Cav-1 expression directly correlated with EMT phenotype and metastatic potential of pancreatic cancer cells.

Figure 2.

Cav-1 expression and EMT and metastatic potential of pancreatic cancer cells. A (i), Western blot analysis of Cav-1 protein expression in pancreatic cancer cell lines; (ii), Western blot quantitative results were obtained by densitometric analysis, standardized to GAPDH and expressed as fold of HPDE. B, double immunofluorescence staining for Cav-1 (red) and nuclei (DAPI; blue). Note that highly metastatic L3.7 and Panc02-H7 cells displayed strong positive Cav-1 staining, whereas poorly metastatic COLO357 and Panc02 cells displayed weak positive Cav-1 staining. C, phase-contrast photomicrographs. Note that highly metastatic L3.7 and Panc02-H7 cells exhibited a typical mesenchymal morphology, whereas poorly metastatic COLO357 and Panc02 cells exhibited a typical epithelial morphology. D, Western blot analysis of expression levels of E-cadherin, β-catenin, vimentin, and N-cadherin. Note that highly metastatic L3.7 and Panc02-H7 cells possessed high expression of mesenchymal marker (vimentin, N-cadherin) and low epithelial marker (E-cadherin, β-catenin), whereas poorly metastatic COLO357 and Panc02 cells possessed high epithelial marker (E-cadherin and β-catenin) and low expression of mesenchymal marker (vimentin and N-cadherin). DAPI, 4′,6-diamidino-2-phenylindole; GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

Figure 2.

Cav-1 expression and EMT and metastatic potential of pancreatic cancer cells. A (i), Western blot analysis of Cav-1 protein expression in pancreatic cancer cell lines; (ii), Western blot quantitative results were obtained by densitometric analysis, standardized to GAPDH and expressed as fold of HPDE. B, double immunofluorescence staining for Cav-1 (red) and nuclei (DAPI; blue). Note that highly metastatic L3.7 and Panc02-H7 cells displayed strong positive Cav-1 staining, whereas poorly metastatic COLO357 and Panc02 cells displayed weak positive Cav-1 staining. C, phase-contrast photomicrographs. Note that highly metastatic L3.7 and Panc02-H7 cells exhibited a typical mesenchymal morphology, whereas poorly metastatic COLO357 and Panc02 cells exhibited a typical epithelial morphology. D, Western blot analysis of expression levels of E-cadherin, β-catenin, vimentin, and N-cadherin. Note that highly metastatic L3.7 and Panc02-H7 cells possessed high expression of mesenchymal marker (vimentin, N-cadherin) and low epithelial marker (E-cadherin, β-catenin), whereas poorly metastatic COLO357 and Panc02 cells possessed high epithelial marker (E-cadherin and β-catenin) and low expression of mesenchymal marker (vimentin and N-cadherin). DAPI, 4′,6-diamidino-2-phenylindole; GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

Close modal

Altered Cav-1 expression directly affects epithelial or mesenchymal phenotype of pancreatic cancer cells

To determine the effect of altered Cav-1 expression on epithelial or mesenchymal phenotype of pancreatic cancer cells, we transfected the Cav-1 expression vector pcDNA3.1-Cav-1 or the control vector pcDNA3.1 into COLO357 cells, which typically express low levels of Cav-1 and have epithelial phenotype. We found that elevated expression of Cav-1 in COLO357 cells (Fig. 3A and C, left; and Supplementary Fig. S4B) significantly increased vimentin but decreased E-cadherin expression (Fig. 3C, left) and caused typical morphology changes of EMT (Fig. 3B, top). In contrast, we transfected Cav-1 siRNA or the control siRNA into L3.7 cells, which typically express high levels of Cav-1 and have mesenchymal phenotype. This transfection significantly inhibited Cav-1 protein expression in L3.7 cells (Fig. 3A and C, right; and Supplementary Fig. S4C) and Cav-1 knockdown led to typical morphology changes of MET of L3.7 cells (Fig. 3B, bottom) and a concomitant decrease in vimentin but increase in E-cadherin expression (Fig. 3C, right). These results clearly indicated that altered Cav-1 expression affected epithelial or mesenchymal phenotype of pancreatic cancer cells.

Figure 3.

Influence of altered Cav-1 expression on pancreatic cancer cell epithelial or mesenchymal phenotype. A, double immunofluorescence staining for Cav-1 (red) and nuclei (DAPI; blue). COLO357 cells were transfected with control pcDNA3.1 vector (“control”) or pcDNA3.1-Cav-1 expression vector (“pCav-1”) or just transfection reagent (“mock”), whereas L3.7 cells were transfected with control siRNA (“control”) or siRNA against Cav-1 (“siCav-1) or just transfection reagent (“mock”). B, phase-contrast photomicrographs of COLO357 cells with Cav-1 overexpression or L3.7 cells with Cav-1 knockdown (as described in A). C, Western blot analysis of EMT markers. Total protein lysates were prepared from COLO357 cells with Cav-1 overexpression or L3.7 cells with Cav-1 knockdown (as described in A). GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

Figure 3.

Influence of altered Cav-1 expression on pancreatic cancer cell epithelial or mesenchymal phenotype. A, double immunofluorescence staining for Cav-1 (red) and nuclei (DAPI; blue). COLO357 cells were transfected with control pcDNA3.1 vector (“control”) or pcDNA3.1-Cav-1 expression vector (“pCav-1”) or just transfection reagent (“mock”), whereas L3.7 cells were transfected with control siRNA (“control”) or siRNA against Cav-1 (“siCav-1) or just transfection reagent (“mock”). B, phase-contrast photomicrographs of COLO357 cells with Cav-1 overexpression or L3.7 cells with Cav-1 knockdown (as described in A). C, Western blot analysis of EMT markers. Total protein lysates were prepared from COLO357 cells with Cav-1 overexpression or L3.7 cells with Cav-1 knockdown (as described in A). GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

Close modal

Altered Cav-1 expression affected migration and invasion in vitro and growth and metastases in vivo of pancreatic cancer cells

To determine the effect of altered Cav-1 expression on migration of pancreatic cancer cells, COLO357 and L3.7 cells were transfected with pcDNA3.1-Cav-1 and Cav-1 siRNA for 48 hours, respectively. The transfected cells were wounded by scratching and maintained at 37°C for additional 12 hours. The overexpression of Cav-1 strongly promoted the flattening and spreading of COLO357 cells [Fig. 4A (i)], whereas knockdown of Cav-1 attenuated the flattening and spreading of L3.7 cells [Fig. 4B (i)]. The results of cell migration assay also indicated that overexpression of Cav-1 promoted the migration ability of COLO357 cells [Fig. 4A (ii) and Supplementary Fig. S5A], whereas knockdown of expression of Cav-1 attenuated the migration ability of L3.7 cells [Fig. 4B (ii) and Supplementary Fig. S5B]. Similarly, overexpression of Cav-1 promoted the invasiveness of COLO357 cells [Fig. 4A (iii) and Supplementary Fig. S6A], whereas knockdown of expression of Cav-1 attenuated the invasiveness of L3.7 cells [Fig. 4B (iii) and Supplementary Fig. S6B]. Consistent with the impact of altered Cav-1 expression on migration and invasion of pancreatic cancer cells in vitro, pcDNA3.1-Cav-1 transfection significantly promoted pancreatic tumor growth [Fig. 5A (i), (ii), and (v)] and increased liver metastases of COLO357 cells [Fig. 5A (iii), (iv), and (vi) and Supplementary Fig. S7A], whereas Cav-1 siRNA transfection significantly inhibited pancreatic tumor growth [Fig. 5B (i), (ii), and (v)] and abrogated liver metastases of L3.7 cells [Fig. 5B (iii), (iv), and (vi) and Supplementary Fig. S7B] in nude mice. Thus, our data clearly established that Cav-1 is oncogenic and promotes invasion and metastasis of pancreatic cancer.

Figure 4.

Influence of Cav-1 expression on pancreatic cancer cell migration and invasion. COLO357 (A) and L3.7 cells (B) were transfected with pcDNA3.1-Cav-1 and Cav-1 siRNA for 48 hours, respectively. For cell scratch wound assay, the cultures were wounded by scratching and maintained at 37°C for additional 12 hours. Cell cultures were photographed and cell migration was assessed by measuring gap sizes [inserted number represented percentage area of gap ± SD; A (i) and B (i)]. For cell migration assay, the transfected cells were maintained at 37°C for additional 24 hours. Representative tumor cell migrated through a membrane were photographed, whereas the numbers of cells that migrated through the membrane without Matrigel were counted in 5 random fields identified within the lower surface of the membrane and expressed as percentage of mock control (inserted numbers). Data represent mean ± SD of triplicates [A (ii) and B (ii)]. For cell invasion assay, the transfected cells were maintained at 37°C for additional 48 hours. Representative tumor cell invaded through Matrigel were photographed, whereas the numbers of invasive cells that penetrated through Matrigel-coated filter were counted in 5 random fields identified within the lower surface of the filters and expressed as percentage of mock control (inserted numbers). Data represent mean ± SD of triplicates [A (iii) and B (iii)]. *, P < 0.01 in a comparison of the pCav-1- or si-Cav-1–treated group with the mock or control groups.

Figure 4.

Influence of Cav-1 expression on pancreatic cancer cell migration and invasion. COLO357 (A) and L3.7 cells (B) were transfected with pcDNA3.1-Cav-1 and Cav-1 siRNA for 48 hours, respectively. For cell scratch wound assay, the cultures were wounded by scratching and maintained at 37°C for additional 12 hours. Cell cultures were photographed and cell migration was assessed by measuring gap sizes [inserted number represented percentage area of gap ± SD; A (i) and B (i)]. For cell migration assay, the transfected cells were maintained at 37°C for additional 24 hours. Representative tumor cell migrated through a membrane were photographed, whereas the numbers of cells that migrated through the membrane without Matrigel were counted in 5 random fields identified within the lower surface of the membrane and expressed as percentage of mock control (inserted numbers). Data represent mean ± SD of triplicates [A (ii) and B (ii)]. For cell invasion assay, the transfected cells were maintained at 37°C for additional 48 hours. Representative tumor cell invaded through Matrigel were photographed, whereas the numbers of invasive cells that penetrated through Matrigel-coated filter were counted in 5 random fields identified within the lower surface of the filters and expressed as percentage of mock control (inserted numbers). Data represent mean ± SD of triplicates [A (iii) and B (iii)]. *, P < 0.01 in a comparison of the pCav-1- or si-Cav-1–treated group with the mock or control groups.

Close modal
Figure 5.

Influence of Cav-1 expression on pancreatic cancer growth and metastasis. COLO357 cells with Cav-1 overexpression (A) or L3.7 cells with Cav-1 knockdown (B) were injected subcutaneously (1 × 105 (per mouse) into the right scapular region of nude mice, or injected intravenously (COLO357, 1 × 106 per mouse; L3.7, 1 × 105 per mouse) into the ileocolic vein of nude mice (n = 5). The tumor-bearing mice were sacrificed when they became moribund or on day 21 (intravenous injection) or day 35 (subcutaneous injection). Shown are gross tumors in the mice [A (i) and B (i)], the tumors removed from the mice [A (ii) and B (ii)], gross liver metastatic tumors in the liver [A (iii) and B (iii)] and hematoxylin and eosinH&E-stained sections of liver [A (iv) and B (iv)], tumors weights [A (v) and B (v)], and the numbers of liver surface metastases [A (vi) and B (vi)]. *, P < 0.01 in a comparison of the pCav-1- or si-Cav-1–treated group with the Mmock or Ccontrol groups.

Figure 5.

Influence of Cav-1 expression on pancreatic cancer growth and metastasis. COLO357 cells with Cav-1 overexpression (A) or L3.7 cells with Cav-1 knockdown (B) were injected subcutaneously (1 × 105 (per mouse) into the right scapular region of nude mice, or injected intravenously (COLO357, 1 × 106 per mouse; L3.7, 1 × 105 per mouse) into the ileocolic vein of nude mice (n = 5). The tumor-bearing mice were sacrificed when they became moribund or on day 21 (intravenous injection) or day 35 (subcutaneous injection). Shown are gross tumors in the mice [A (i) and B (i)], the tumors removed from the mice [A (ii) and B (ii)], gross liver metastatic tumors in the liver [A (iii) and B (iii)] and hematoxylin and eosinH&E-stained sections of liver [A (iv) and B (iv)], tumors weights [A (v) and B (v)], and the numbers of liver surface metastases [A (vi) and B (vi)]. *, P < 0.01 in a comparison of the pCav-1- or si-Cav-1–treated group with the Mmock or Ccontrol groups.

Close modal

Close relationship between altered expression of FoxM1 and Cav-1 in pancreatic cancer

To explore the mechanisms underlying Cav-1 overexpression, we initially analyzed both FoxM1 and Cav-1 expression in pancreatic cancer tissues and cell lines. Pancreatic cancer tissues expressed both FoxM1 and Cav-1 (Fig. 6A) and their direct correlation was found statistically significant (r = 0.574; P < 0.001; Fig. 6B). Consistently, the expression of Cav-1 directly correlated with the expression of FoxM1 in pancreatic cancer cell lines (Fig. 6C).

Figure 6.

Coexpression of FoxM1 and Cav-1 expression in pancreatic cancer. A, immunohistochemical staining with specific anti-FoxM1 and anti-Cav-1 antibodies were done on pancreatic cancer specimens. Shown were representative images of positive (left 2 panels) and negative (right 2 panels) FoxM1 and Cav-1 staining in pancreatic cancer sections (200×, 400× for the inserts). B, direct correlation between FoxM1 expression and Cav-1 expression in pancreatic cancer samples (n = 70; Pearson's correlation test, r = 0.574; P < 0.001). Note that some of the dots on the graphs represented more than 1 specimen (overlapped scores). C, Western blot analysis of FoxM1 and Cav-1 protein expression in pancreatic cancer cell lines. D, COLO357 and AsPC-1 cells were transfected with pcDNA3.1-FoxM1 or pcDNA3.1 (i) or L3.7 and PA-TU-8902 cells were transfected with FoxM1-siRNA or control siRNA (ii), and the cultures were incubated for 48 hours. Total RNA and protein lysates were harvested for determination of the levels of FoxM1 and Cav-1 expression using RT-PCR (left) and Western blot analysis (right). E, effects of FoxM1 expression on Cav-1 promoter activity. COLO357 and AsPC-1 cells were cotransfected with 0.8 μg of the Cav-1 promoter-luciferase construct pLuc-Cav and 0 to 0.8 μg of pcDNA3.1-FoxM1 or pcDNA3.1 (i), whereas L3.7 and PA-TU-8902 cells were cotransfected with 0.8 μg pLuc-Cav and 50 nmol/L FoxM1-siRNA or control siRNA (ii). Promoter activities were determined using Dual-Luciferase Assay Kit. GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

Figure 6.

Coexpression of FoxM1 and Cav-1 expression in pancreatic cancer. A, immunohistochemical staining with specific anti-FoxM1 and anti-Cav-1 antibodies were done on pancreatic cancer specimens. Shown were representative images of positive (left 2 panels) and negative (right 2 panels) FoxM1 and Cav-1 staining in pancreatic cancer sections (200×, 400× for the inserts). B, direct correlation between FoxM1 expression and Cav-1 expression in pancreatic cancer samples (n = 70; Pearson's correlation test, r = 0.574; P < 0.001). Note that some of the dots on the graphs represented more than 1 specimen (overlapped scores). C, Western blot analysis of FoxM1 and Cav-1 protein expression in pancreatic cancer cell lines. D, COLO357 and AsPC-1 cells were transfected with pcDNA3.1-FoxM1 or pcDNA3.1 (i) or L3.7 and PA-TU-8902 cells were transfected with FoxM1-siRNA or control siRNA (ii), and the cultures were incubated for 48 hours. Total RNA and protein lysates were harvested for determination of the levels of FoxM1 and Cav-1 expression using RT-PCR (left) and Western blot analysis (right). E, effects of FoxM1 expression on Cav-1 promoter activity. COLO357 and AsPC-1 cells were cotransfected with 0.8 μg of the Cav-1 promoter-luciferase construct pLuc-Cav and 0 to 0.8 μg of pcDNA3.1-FoxM1 or pcDNA3.1 (i), whereas L3.7 and PA-TU-8902 cells were cotransfected with 0.8 μg pLuc-Cav and 50 nmol/L FoxM1-siRNA or control siRNA (ii). Promoter activities were determined using Dual-Luciferase Assay Kit. GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

Close modal

To provide casual evidence for the direct correlation between the expression FoxM1 and Cav-1, we determined the impacts of altered FoxM1 expression on Cav-1 expression in human pancreatic cancer cell lines that have either low (COLO357 and AsPC-1) or high (L3.7 and PA-TU-8902) levels of FoxM1 expression. We found that increased expression of FoxM1 in COLO357 and AsPC-1 cells [Fig. 6D (i)] led to significantly increased Cav-1 mRNA and protein [Fig. 6D (i)]. Conversely, knockdown of FoxM1 expression by transfection of FoxM1-siRNA into L3.7 and PA-TU-8902 cells [Fig. 6D (ii)] led to significantly decreased Cav-1 mRNA and protein in the cells [Fig. 6D (ii)].

To investigate the regulatory role of FoxM1 in Cav-1 transcription, we cotransfected the Cav-1 promoter-luciferase construct pLuc-Cav into COLO357 and AsPC-1 cells with pcDNA3.1-FoxM1b or the control vector pcDNA 3.1. FoxM1b activated Cav-1 promoter in both cell lines [Fig. 6E (i)]. Conversely, knockdown of FoxM1 expression by cotransfecting FoxM1-siRNA (50 nmol/L) and pLuc-Cav-1 into L3.7 and PA-TU-8902 cells suppressed Cav-1 promoter activities [Fig. 6E (ii)]. Collectively, our findings strongly indicated that FoxM1 regulates Cav-1 expression most likely at the transcriptional level.

Direct interaction of FoxM1 with Cav-1 promoter

Finally, we analyzed the sequence of the Cav-1 promoter for the presence of potential FoxM1-binding elements by using the FoxM1 consensus sequence 5′-A(C/T)AAA(C/T)AA-3′ (34), 5′-AGATTGAGTA-3′ (35), and 5′-TAATCA-3′ (31). We identified 3 putative FoxM1-binding elements (referred to as DNA sequences #1, #2, and #3) in the Cav-1 promoter region (Fig. 7A). To provide direct proof that FoxM1 is recruited to the endogenous Cav-1 promoter during transcription in vivo, we conducted chromatin immunoprecipitation assays using chromatins prepared from COLO357, AsPC-1, L3.7, and PA-TU-8902 cells and 3 primer sets franking the 345-bp (−1,771 to −1,427; #1 site), 108-bp (−1,260 to −1,153; #2 site), and 181-bp (−1,174 to −994; #3 site) regions of the Cav-1 promoter (Fig. 7A). The 181-bp DNA fragment was amplified from the precipitates by anti-FoxM1 antibodies but not by control IgG in COLO357, AsPC-1, L3.7, and PA-TU-8902 cells (Fig. 7B, right), suggesting that endogenous FoxM1 bound to the region between −1,174 and −994 bp of the Cav-1 promoter in pancreatic cancer cells. However, we did not observe any detectable binding of FoxM1 to the site #1 and #2 (Fig. 7B, left and middle). These results were further confirmed using COLO357 and AsPC-1 cells with engineered overexpression of FoxM1, which led to increased FoxM1 recruitment to Cav-1 promoter (Fig. 7C), and using L3.7 and PA-TU-8902 with FoxM1 knockdown, which led to decreased FoxM1 recruitment to Cav-1 promoter (Fig. 7D). The altered FoxM1 recruitments were consistent with changes of Cav-1 promoter activities (Fig. 7E and F). Collectively, these findings showed that FoxM1 bound primarily at position −1,055 to −1,050 bp of Cav-1 promoter and positively regulates Cav-1 transcription.

Figure 7.

Direct binding of FoxM1 to Cav-1 promoter. A, sequences and positions of putative FoxM1-binding elements on the Cav-1 promoter (#1, #2, and #3). B, chromatin immunoprecipitation (ChIP) assay. Chromatins were isolated from COLO357, AsPC-1, L3.7, and PA-TU-8902 cells and binding of FoxM1 to the Cav-1 promoter was determined using a specific anti-FoxM1 antibody and oligonucleotides flanking the Cav-1 promoter regions containing putative FoxM1-binding sites as described in Materials and Methods. C and D, similar ChIP assays were conducted using chromatins isolated from COLO357, COLO357-FoxM1, AsPC-1, and AsPC-1-FoxM1 cells (C) or L3.7, L3.7-siFoxM1, PA-TU-8902, and PA-TU-8902-siFoxM1 cells (D). Normal IgG was used as a control, and 1% of the total cell lysates was also subjected to PCR before immunoprecipitation (input control). E and F, COLO357 and AsPC-1 cells were cotransfected with 0.8

Figure 7.

Direct binding of FoxM1 to Cav-1 promoter. A, sequences and positions of putative FoxM1-binding elements on the Cav-1 promoter (#1, #2, and #3). B, chromatin immunoprecipitation (ChIP) assay. Chromatins were isolated from COLO357, AsPC-1, L3.7, and PA-TU-8902 cells and binding of FoxM1 to the Cav-1 promoter was determined using a specific anti-FoxM1 antibody and oligonucleotides flanking the Cav-1 promoter regions containing putative FoxM1-binding sites as described in Materials and Methods. C and D, similar ChIP assays were conducted using chromatins isolated from COLO357, COLO357-FoxM1, AsPC-1, and AsPC-1-FoxM1 cells (C) or L3.7, L3.7-siFoxM1, PA-TU-8902, and PA-TU-8902-siFoxM1 cells (D). Normal IgG was used as a control, and 1% of the total cell lysates was also subjected to PCR before immunoprecipitation (input control). E and F, COLO357 and AsPC-1 cells were cotransfected with 0.8

Close modal

In this study, we determined the critical roles of Cav-1 and FoxM1 in pancreatic cancer pathogenesis and their underlying mechanisms. We found FoxM1 transcriptionally activated Cav-1 gene, constituting a novel signaling pathway that directly impact EMT, invasion, and metastasis of pancreatic cancer cells and its alterations inform the clinicopathologic behaviors of pancreatic cancer. Collectively, our novel clinical and mechanistic evidence strongly suggested that dysregulated FoxM1 expression causes abnormal Cav-1 expression and critically contributes to pancreatic cancer pathogenesis and aggressive biology.

Cav-1 is an essential constituent of caveolae and interacts with a variety of cellular proteins and regulates cell signaling events. However, the potential roles of Cav-1 in cancer development and progression are highly inconsistent, functioning from as a tumor suppressor to as an oncogene (6, 36). First possible reason could be due to tumor types. For example, Cav-1 seems to be tumor suppressor in breast cancer and ovarian carcinoma (37, 38), whereas Cav-1 acts more as an oncogene in gastrointestinal cancer, including pancreatic cancer (11–13, 39). Second possible reason is the tumor stage. For example, loss of Cav-1 is necessary and sufficient to promote fibroblasts cell transformation in the early stages of cancer progression, supporting the notion that it functions as a tumor suppressor (40). However, increased expression of Cav-1 correlates with advanced pathologic stage, the presence of metastasis, and poor cancer prognosis of non–small cell lung cancer (41) and esophageal carcinoma (9). Third possible reason could be due to research approaches. For example, most studies using human pancreatic tumor specimens have found a Cav-1 overexpression and its direct correlation with a poor prognosis (11–13). However, biology studies suggested otherwise (42). In our current study, we provided many lines of evidence to show Cav-1 as an oncogene. First of all, elevated Cav-1 expression directly correlated with poorer tumor differentiation and advanced disease stage, particularly with lymph node and distant metastasis. Thus, we provide the first clinical evidence that Cav-1 promotes pancreatic cancer metastasis.

Our notion is further supported by our novel findings using our unique cell line systems, that is, mouse pancreatic cancer cell lines Panc02 and Panc02-H7 and human pancreatic cancer cell lines COLO357 and L3.7, which derive from biologically isogenic cell lines but have distinct metastatic properties. Specifically, Cav-1 protein levels positively correlated with metastatic potential of pancreatic cancer cells. Elevated expression of Cav-1 promoted metastasis of poorly metastatic pancreatic cancer cells, whereas knockdown of Cav-1 attenuated migration and invasion abilities of highly metastatic pancreatic cancer cells. Our findings were consistent with recent studies, which have suggested a regulatory role for Cav-1 in metastasis of hepatocellular carcinoma and lung adenocarcinoma cells (10, 43).

Cancer metastasis is a complex process and its cellular and molecular basis remains to be determined (44, 45). Emerging evidences suggest that EMT plays a key role in metastasis (46, 47). In this study, we presented 2 cell systems with cell morphology, molecular biomarkers, and biology behaviors being consistent with EMT or MET. We used these systems to show the critical role of Cav-1 in pancreatic cancer EMT and progression. Specifically, the poorly metastatic human and mouse pancreatic cancer cell lines (COLO357 and Panc02) exhibit a typical epithelial morphology and have high expression of epithelial markers (E-cadherin and β-catenin), whereas the highly metastatic human and mouse pancreatic cancer cell lines (L3.7 and Panc02-H7) exhibit a typical mesenchymal morphology and have high expression of mesenchymal marker (vimentin and N-cadherin). Moreover, we observed that elevated Cav-1 expression could contribute to the acquisition of EMT phenotype by upregulation of mesenchymal cell marker vimentin and downregulation of epithelial cell marker E-cadherin, whereas decreased Cav-1 expression could contribute to the acquisition of MET phenotype by downregulation of mesenchymal cell marker vimentin and upregulation of epithelial cell marker E-cadherin. Our new findings were consistent with recent studies, which have shown one correlation between Cav-1 overexpression and downregulation of E-cadherin but upregulation of vimentin in human hepatocellular carcinoma cells (18) and another correlation between cancer cell EMT and Cav-1 upregulation (17). We showed for the first the critical role of Cav-1 in pancreatic cancer EMT.

Given the important role of Cav-1 in pancreatic cancer EMT and metastasis, we further determined the underlying mechanisms responsible for Cav-1 overexpression in malignances, which are currently unclear (48). Our current study showed that Cav-1 was a novel downstream target of FoxM1, an important transcription factor that has been implicated in the development and progression of many types of cancer (19–21). Our specific evidences include (i) the expression levels of Cav-1 directly correlated with the expression levels of FoxM1 both in pancreatic cancer cell lines and in pancreatic cancer tissues; (ii) overexpression of FoxM1 led to increased expression of Cav-1 whereas knockdown of FoxM1 did the opposite; and (iii) FoxM1 directly bound to the promoter regions of Cav-1 gene and transactivated Cav-1 gene transcription. Therefore, our findings provided clinical and mechanistic evidence supporting the existence of a novel FoxM1-Cav-1 signaling pathway and its critical contribution to pancreatic cancer growth and metastasis. In addition, there are reports indicating the regulatory roles of Sp1 and Stat3 in Cav-1 expression (49, 50). It would be interesting and significant to further determine the cross-talks between those transcription factors and FoxM1 in regulating Cav-1 expression and function, how these cross-talks are altered during pancreatic carcinogenesis and contribute to pancreatic cancer progression and metastasis, and future potential of designing effective therapies that target FoxM1-Cav-1 signaling.

In summary, this study provided critical insight into the role of the Cav-1 in pancreatic cancer progression and metastasis; the critical role of upstream FoxM1 transcription factor in Cav-1 expression; and critical role of this novel FoxM1-Cav-1 signaling in EMT and MET phenotypes and malignancy of pancreatic cancer. Collectively, our study not only provides a novel molecular mechanism for pancreatic cancer progression and metastasis but also undercovers this aberrant FoxM1-Cav-1 signaling as a promising new molecular target for designing novel therapeutic modalities to control pancreatic cancer progression and metastasis.

No potential conflicts of interest were disclosed.

The authors thank Don Norwood for editorial comments.

The work was supported in part by grants R01-CA129956, R01-CA148954, and R01CA152309 (to K. Xie), and R01-CA116528 and R01-CA157933 (to S. Huang) from the National Cancer Institute, NIH; and grant No.81101844 (to C. Huang) from the National Natural Science Foundation of China.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Jemal
A
,
Siegel
R
,
Xu
J
,
Ward
E
. 
Cancer statistics, 2010
.
CA Cancer J Clin
2010
;
60
:
277
300
.
2.
Nieto
J
,
Grossbard
ML
,
Kozuch
P
. 
Metastatic pancreatic cancer 2008: is the glass less empty
.
Oncologist
2008
;
13
:
562
76
.
3.
Pliarchopoulou
K
,
Pectasides
D
. 
Pancreatic cancer: current and future treatment strategies
.
Cancer Treat Rev
2009
;
35
:
431
6
.
4.
Stathis
A
,
Moore
MJ
. 
Advanced pancreatic carcinoma: current treatment and future challenges
.
Nat Rev Clin Oncol
2010
;
7
:
163
72
.
5.
Couet
J
,
Belanger
MM
,
Roussel
E
,
Drolet
MC
. 
Cell biology of caveolae and caveolin
.
Adv Drug Deliv Rev
2001
;
49
:
223
35
.
6.
Goetz
JG
,
Lajoie
P
,
Wiseman
SM
,
Nabi
IR
. 
Caveolin-1 in tumor progression: the good, the bad and the ugly
.
Cancer Metastasis Rev
2008
;
27
:
715
35
.
7.
Thomas
S
,
Overdevest
JB
,
Nitz
MD
,
Williams
PD
,
Owens
CR
,
Sanchez-Carbayo
M
, et al
Src and caveolin-1 reciprocally regulate metastasis via a common downstream signaling pathway in bladder cancer
.
Cancer Res
2011
;
71
:
832
41
.
8.
Tahir
SA
,
Yang
G
,
Ebara
S
,
Timme
TL
,
Satoh
T
,
Li
L
, et al
Secreted caveolin-1 stimulates cell survival/clonal growth and contributes to metastasis in androgen-insensitive prostate cancer
.
Cancer Res
2001
;
61
:
3882
5
.
9.
Kato
K
,
Hida
Y
,
Miyamoto
M
,
Hashida
H
,
Shinohara
T
,
Itoh
T
, et al
Overexpression of caveolin-1 in esophageal squamous cell carcinoma correlates with lymph node metastasis and pathologic stage
.
Cancer
2002
;
94
:
929
33
.
10.
Ho
CC
,
Huang
PH
,
Huang
HY
,
Chen
YH
,
Yang
PC
,
Hsu
SM
. 
Up-regulated caveolin-1 accentuates the metastasis capability of lung adenocarcinoma by inducing filopodia formation
.
Am J Pathol
2002
;
161
:
1647
56
.
11.
Suzuoki
M
,
Miyamoto
M
,
Kato
K
,
Hiraoka
K
,
Oshikiri
T
,
Nakakubo
Y
, et al
Impact of caveolin-1 expression on prognosis of pancreatic ductal adenocarcinoma
.
Br J Cancer
2002
;
87
:
1140
4
.
12.
Tanase
CP
. 
Caveolin-1: a marker for pancreatic cancer diagnosis
.
Expert Rev Mol Diagn
2008
;
8
:
395
404
.
13.
Tanase
CP
,
Dima
S
,
Mihai
M
,
Raducan
E
,
Nicolescu
MI
,
Albulescu
L
, et al
Caveolin-1 overexpression correlates with tumour progression markers in pancreatic ductal adenocarcinoma
.
J Mol Histol
2009
;
40
:
23
9
.
14.
Hehlgans
S
,
Eke
I
,
Storch
K
,
Haase
M
,
Baretton
GB
,
Cordes
N
. 
Caveolin-1 mediated radioresistance of 3D grown pancreatic cancer cells
.
Radiother Oncol
2009
;
92
:
362
70
.
15.
Cordes
N
,
Frick
S
,
Brunner
TB
,
Pilarsky
C
,
Grutzmann
R
,
Sipos
B
, et al
Human pancreatic tumor cells are sensitized to ionizing radiation by knockdown of caveolin-1
.
Oncogene
2007
;
26
:
6851
62
.
16.
Witkiewicz
AK
,
Nguyen
KH
,
Dasgupta
A
,
Kennedy
EP
,
Yeo
CJ
,
Lisanti
MP
, et al
Co-expression of fatty acid synthase and caveolin-1 in pancreatic ductal adenocarcinoma: implications for tumor progression and clinical outcome
.
Cell Cycle
2008
;
7
:
3021
5
.
17.
Bailey
KM
,
Liu
J
. 
Caveolin-1 up-regulation during epithelial to mesenchymal transition is mediated by focal adhesion kinase
.
J Biol Chem
2008
;
283
:
13714
24
.
18.
Cokakli
M
,
Erdal
E
,
Nart
D
,
Yilmaz
F
,
Sagol
O
,
Kilic
M
, et al
Differential expression of Caveolin-1 in hepatocellular carcinoma: correlation with differentiation state, motility and invasion
.
BMC Cancer
2009
;
9
:
65
.
19.
Wang
IC
,
Chen
YJ
,
Hughes
D
,
Petrovic
V
,
Major
ML
,
Park
HJ
, et al
Forkhead box M1 regulates the transcriptional network of genes essential for mitotic progression and genes encoding the SCF (Skp2-Cks1) ubiquitin ligase
.
Mol Cell Biol
2005
;
25
:
10875
94
.
20.
Gartel
AL
. 
FoxM1 inhibitors as potential anticancer drugs
.
Expert Opin Ther Targets
2008
;
12
:
663
5
.
21.
Park
HJ
,
Gusarova
G
,
Wang
Z
,
Carr
JR
,
Li
J
,
Kim
KH
, et al
Deregulation of FoxM1b leads to tumour metastasis
.
EMBO Mol Med
2011
;
3
:
21
34
.
22.
Bao
B
,
Wang
Z
,
Ali
S
,
Kong
D
,
Banerjee
S
,
Ahmad
A
, et al
Over-expression of FoxM1 leads to epithelial-mesenchymal transition and cancer stem cell phenotype in pancreatic cancer cells
.
J Cell Biochem
2011
;
112
:
2296
306
.
23.
Morgan
RT
,
Woods
LK
,
Moore
GE
,
Quinn
LA
,
McGavran
L
,
Gordon
SG
. 
Human cell line (COLO 357) of metastatic pancreatic adenocarcinoma
.
Int J Cancer
1980
;
25
:
591
8
.
24.
Vezeridis
MP
,
Meitner
PA
,
Tibbetts
LM
,
Doremus
CM
,
Tzanakakis
G
,
Calabresi
P
. 
Heterogeneity of potential for hematogenous metastasis in a human pancreatic carcinoma
.
J Surg Res
1990
;
48
:
51
5
.
25.
Wang
B
,
Wei
D
,
Crum
VE
,
Richardson
EL
,
Xiong
HH
,
Luo
Y
, et al
A novel model system for studying the double-edged roles of nitric oxide production in pancreatic cancer growth and metastasis
.
Oncogene
2003
;
22
:
1771
82
.
26.
Liu
M
,
Dai
B
,
Kang
SH
,
Ban
K
,
Huang
FJ
,
Lang
FF
, et al
FoxM1B is overexpressed in human glioblastomas and critically regulates the tumorigenicity of glioma cells
.
Cancer Res
2006
;
66
:
3593
602
.
27.
Fujimoto
T
,
Kogo
H
,
Nomura
R
,
Une
T
. 
Isoforms of caveolin-1 and caveolar structure
.
J Cell Sci
2000
;
113
:
3509
17
.
28.
Engelman
JA
,
Zhang
XL
,
Razani
B
,
Pestell
RG
,
Lisanti
MP
. 
p42/44 MAP kinase-dependent and -independent signaling pathways regulate caveolin-1 gene expression. Activation of Ras-MAP kinase and protein kinase A signaling cascades transcriptionally down-regulates caveolin-1 promoter activity
.
J Biol Chem
1999
;
274
:
32333
41
.
29.
Li
Q
,
Zhang
N
,
Jia
Z
,
Le
X
,
Dai
B
,
Wei
D
, et al
Critical role and regulation of transcription factor FoxM1 in human gastric cancer angiogenesis and progression
.
Cancer Res
2009
;
69
:
3501
9
.
30.
Sunaga
N
,
Miyajima
K
,
Suzuki
M
,
Sato
M
,
White
MA
,
Ramirez
RD
, et al
Different roles for caveolin-1 in the development of non-small cell lung cancer versus small cell lung cancer
.
Cancer Res
2004
;
64
:
4277
85
.
31.
Zhang
Y
,
Zhang
N
,
Dai
B
,
Liu
M
,
Sawaya
R
,
Xie
K
, et al
FoxM1B transcriptionally regulates vascular endothelial growth factor expression and promotes the angiogenesis and growth of glioma cells
.
Cancer Res
2008
;
68
:
8733
42
.
32.
Wang
L
,
Wei
D
,
Huang
S
,
Peng
Z
,
Le
X
,
Wu
TT
, et al
Transcription factor Sp1 expression is a significant predictor of survival in human gastric cancer
.
Clin Cancer Res
2003
;
9
:
6371
80
.
33.
Marvin
MR
,
Southall
JC
,
Trokhan
S
,
DeRosa
C
,
Chabot
J
. 
Liver metastases are enhanced in homozygous deletionally mutant ICAM-1 or LFA-1 mice
.
J Surg Res
1998
;
80
:
143
8
.
34.
Wierstra
I
,
Alves
J
. 
Despite its strong transactivation domain, transcription factor FOXM1c is kept almost inactive by two different inhibitory domains
.
Biol Chem
2006
;
387
:
963
76
.
35.
Dai
B
,
Kang
SH
,
Gong
W
,
Liu
M
,
Aldape
KD
,
Sawaya
R
, et al
Aberrant FoxM1B expression increases matrix metalloproteinase-2 transcription and enhances the invasion of glioma cells
.
Oncogene
2007
;
26
:
6212
9
.
36.
Quest
AF
,
Gutierrez-Pajares
JL
,
Torres
VA
. 
Caveolin-1: an ambiguous partner in cell signalling and cancer
.
J Cell Mol Med
2008
;
12
:
1130
50
.
37.
Fiucci
G
,
Ravid
D
,
Reich
R
,
Liscovitch
M
. 
Caveolin-1 inhibits anchorage-independent growth, anoikis and invasiveness in MCF-7 human breast cancer cells
.
Oncogene
2002
;
21
:
2365
75
.
38.
Wiechen
K
,
Diatchenko
L
,
Agoulnik
A
,
Scharff
KM
,
Schober
H
,
Arlt
K
, et al
Caveolin-1 is downregulated in human ovarian carcinoma and acts as a candidate tumor suppressor gene
.
Am J Pathol
2001
;
159
:
1635
43
.
39.
Henkhaus
RS
,
Roy
UK
,
Cavallo-Medved
D
,
Sloane
BF
,
Gerner
EW
,
Ignatenko
NA
. 
Caveolin-1-mediated expression and secretion of kallikrein 6 in colon cancer cells
.
Neoplasia
2008
;
10
:
140
8
.
40.
Koleske
AJ
,
Baltimore
D
,
Lisanti
MP
. 
Reduction of caveolin and caveolae in oncogenically transformed cells
.
Proc Natl Acad Sci U S A
1995
;
92
:
1381
5
.
41.
Ho
CC
,
Kuo
SH
,
Huang
PH
,
Huang
HY
,
Yang
CH
,
Yang
PC
. 
Caveolin-1 expression is significantly associated with drug resistance and poor prognosis in advanced non-small cell lung cancer patients treated with gemcitabine-based chemotherapy
.
Lung Cancer
2008
;
59
:
105
10
.
42.
Han
F
,
Zhu
HG
. 
Caveolin-1 regulating the invasion and expression of matrix metalloproteinase (MMPs) in pancreatic carcinoma cells
.
J Surg Res
2010
;
159
:
443
50
.
43.
Zhou
H
,
Jia
L
,
Wang
S
,
Wang
H
,
Chu
H
,
Hu
Y
, et al
Divergent expression and roles for caveolin-1 in mouse hepatocarcinoma cell lines with varying invasive ability
.
Biochem Biophys Res Commun
2006
;
345
:
486
94
.
44.
Steeg
PS
. 
Tumor metastasis: mechanistic insights and clinical challenges
.
Nat Med
2006
;
12
:
895
904
.
45.
Chaffer
CL
,
Weinberg
RA
. 
A perspective on cancer cell metastasis
.
Science
2011
;
331
:
1559
64
.
46.
Hugo
H
,
Ackland
ML
,
Blick
T
,
Lawrence
MG
,
Clements
JA
,
Williams
ED
, et al
Epithelial–mesenchymal and mesenchymal–epithelial transitions in carcinoma progression
.
J Cell Physiol
2007
;
213
:
374
83
.
47.
Huber
MA
,
Kraut
N
,
Beug
H
. 
Molecular requirements for epithelial-mesenchymal transition during tumor progression
.
Curr Opin Cell Biol
2005
;
17
:
548
58
.
48.
Dasari
A
,
Bartholomew
JN
,
Volonte
D
,
Galbiati
F
. 
Oxidative stress induces premature senescence by stimulating caveolin-1 gene transcription through p38 mitogen-activated protein kinase/Sp1-mediated activation of two GC-rich promoter elements
.
Cancer Res
2006
;
66
:
10805
14
.
49.
Chen
W
,
Chen
Y
,
Qin
L
,
Li
A
,
Zhao
X
,
Wang
X
, et al
Transcription factor Sp1 is essential for the regulation of the porcine caveolin-1 gene
.
DNA Cell Biol
2011
;
30
:
491
7
.
50.
Chiu
WT
,
Lee
HT
,
Huang
FJ
,
Aldape
KD
,
Yao
J
,
Steeg
PS
, et al
Caveolin-1 upregulation mediates suppression of primary breast tumor growth and brain metastases by Stat3 inhibition
.
Cancer Res
2011
;
71
:
4932
43
.

Supplementary data