ARTICLE

Role of Thymosin {beta}4 in Tumor Metastasis and Angiogenesis

Hee-Jae Cha, Moon-Jin Jeong, Hynda K. Kleinman

Affiliations of authors: Craniofacial Developmental Biology and Regeneration Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD (HJC, HKK); Department of Oral Histology, College of Dentistry, Chosun University, Gwanju, Korea (MJJ).

Correspondence to: Hynda K. Kleinman, PhD, Craniofacial Developmental Biology and Regeneration Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892 (e-mail: hkleinman{at}dir.nidcr.nih.gov)


    ABSTRACT
 Top
 Abstract
 Introduction
 Materials and Methods
 Results
 Discussion
 References
 
Background: Expression of the small peptide thymosin {beta}4 is associated with angiogenesis induction, accelerated wound healing, and the metastatic potential of tumor cells. However, little is known about the mechanism(s) by which thymosin {beta}4 promotes metastasis. Methods: Northern blot analysis and immunohistochemistry were used to examine thymosin {beta}4 expression in mouse melanoma B16 cell lines and in B16-F10 cells derived from metastatic mouse lung tumors, respectively. B16-F10 cells infected with adenoviruses containing a thymosin {beta}4 expression vector or an empty vector were injected subcutaneously and intravenously into C57BL/6 mice to evaluate tumor growth and metastatic potential, respectively. In vitro assays were used to study cell migration, invasion, matrix metalloproteinase activity, cell proliferation, and angiogenic activity of adenovirus-infected B16-F10 cells. Statistical significance of all results was analyzed by two-tailed Student’s t tests. Results: Thymosin {beta}4 mRNA was expressed in primary cultured B16-F10 cells derived from lung metastases and in B16-F10 cells that had formed lung tumors after being injected into mice but not in the B16-F1, B16-F10, or B16-BL6 cell lines. The mean tumor sizes in mice 20 days after injection with B16-F10 cells infected with thymosin {beta}4–expressing adenovirus and with control adenovirus were 21.7 mm (95% confidence interval [CI] = 17.7 to 25.7 mm) and 13.3 mm (95% CI = 11.1 to 15.3 mm), respectively (difference = 8.4 mm; P = .036). The mean numbers of metastatic lung nodules in mice (n = 20) 2 weeks after intravenous injection with thymosin {beta}4–expressing adenovirus and with control adenovirus were 46.7 (95% CI = 35.0 to 57.7) and 10.9 (95% CI = 6.2 to 15.6), respectively (difference = 35.8 metastatic lung nodules, P<.001). Thymosin {beta}4 overexpression was associated with a mean 2.3-fold increase (95% CI = 1.9- to 2.7-fold increase; P<.001) in B16-F10 cell migration and a mean 4.4-fold increase (95% CI = 3.3- to 5.5-fold increase; P<.001) in the number of blood vessels in solid tumors derived from injected B16-F10 cells but had no effect on cell invasion, proliferation, or matrix metalloproteinase activity. This induction of angiogenesis by thymosin {beta}4 was associated with induction of vascular endothelial growth factor expression. Conclusion: Thymosin {beta}4 may stimulate tumor metastasis by activating cell migration and angiogenesis.



    INTRODUCTION
 Top
 Abstract
 Introduction
 Materials and Methods
 Results
 Discussion
 References
 
Thymosin {beta}4 is a 43-amino-acid polypeptide that was originally described as a thymic maturation factor (1). Since then, several observations have suggested that thymosin {beta}4 is also involved in angiogenesis (3) and wound healing (4,5). For example, endothelial cells that form capillary-like structures on Matrigel have higher thymosin {beta}4 mRNA levels than endothelial cells grown on plastic (2). Transfection of endothelial cells with a thymosin {beta}4 expression vector was associated with an increase in the rates of cell attachment, cell spreading on plastic, and tube formation on Matrigel. Furthermore, antisense oligonucleotides specific for thymosin {beta}4 inhibited these effects. It has been proposed that thymosin {beta}4 might be involved early in the differentiation of endothelial cells and in vessel formation (3). Thymosin {beta}4 also acts as a chemoattractant for endothelial cells and keratinocytes (3,5).

Recently, the angiogenic effects of several members of the thymosin family of peptides were studied in the chick chorioallantoic membrane model (6). Thymosin {beta}4, prothymosin, and thymosin {alpha}1 were associated with enhancement of angiogenesis, whereas parathymosin, thymosin {beta}9, and thymosin {beta}10 were associated with inhibition of angiogenesis. However, the mechanisms responsible for the regulation of angiogenesis by thymosin {beta}4 have not been defined. Thymosin {beta}4 also promotes wound healing and decreases inflammation in corneas damaged by exposure to heptanol (7) or alkali (8). In the latter case, alkali-exposed mouse corneas topically treated with thymosin {beta}4 demonstrated accelerated re-epithelialization and decreased inflammation when compared with alkali-exposed corneas not treated with thymosin {beta}4 (8).

Thymosin {beta}4 also appears to be involved in tumor cell metastasis. Expression of thymosin {beta}4, thymosin {beta}10, and thymosin {beta}15 are associated with the metastatic potential of tumor cells (9-13). In addition, thymosin {beta}4 expression is elevated in metastatic melanoma compared with non-metastatic melanoma counterparts (10), and fibrosarcoma cells possessing greater metastatic potential express high levels of thymosin {beta}4, whereas fibrosarcoma cells of low metastatic potential express little or no thymosin {beta}4 (14). However, little is known about the mechanisms by which thymosin {beta}4 promotes metastasis. We used an adenovirus-based expression vector to examine the effects of overexpressing thymosin {beta}4 on various malignant activities of mouse melanoma B16-F10 cells in vitro and in vivo.


    MATERIALS AND METHODS
 Top
 Abstract
 Introduction
 Materials and Methods
 Results
 Discussion
 References
 
Cell Culture and Treatment With Thymosin {beta}4 Peptide

Mouse melanoma B16-F1, B16-F10, and B16-BL6 cells were obtained from American Type Culture Collection (ATCC; Manassas, VA). Primary cultured B16-F10 cells derived from a lung tumor were also obtained from ATCC. All cells were grown in Dulbecco’s modified Eagle medium (DMEM) containing 10% fetal bovine serum (FBS) (Invitrogen, Carlsbad, CA). For some experiments, B16-F10 cells were cultured for 18 hours in serum-free DMEM containing a synthetic thymosin {beta}4 peptide (1 ng to 10 µg; RegeneRx Biopharmaceuticals, Bethesda, MD).

Recombinant Adenoviral Constructs and Infection

We used a high-fidelity polymerase chain reaction (PCR) system (Roche, Indianapolis, IN) to amplify a thymosin {beta}4 cDNA from total mouse embryo RNA (Stratagene, La Jolla, CA). Amplification conditions for thymosin {beta}4 were 0.5 min at 95 °C, 0.5 min at 60 °C, and 0.5 min at 72 °C for 30 cycles. These cycles were followed by a 10-minute elongation step at 72 °C. The forward and reverse primers used for this amplification contained HindIII and EcoRI restriction sites (underlined), respectively (forward primer, 5'-GTCAGTAAGCTTCTCCTTCCAGCAACCATGTC-3'; reverse primer, 5'-GTCAGTGAATTCAATGTACAGTGCATATTGGC-3'). The amplified product (0.2 kilobase [kb]) was digested with HindIII and EcoRI and cloned into the HindIII and EcoRI sites of the adenoviral plasmid shuttle vector pAC-EF1, where its expression was under control of the elongation factor 1 promoter (15). The sequence of the cloned mouse thymosin {beta}4 cDNA was confirmed by DNA sequence analysis (National Institute of Dental and Craniofacial Research [NIDCR] Core Sequence Facility, Bethesda, MD). The resulting adenoviral vector was transfected into human epithelial kidney A293 cells for adenovirus production, as previously described (15). The high-expressing virus was expanded by infecting A293 cells. Large batches of recombinant adenovirus were purified by centrifugation through two consecutive cesium chloride gradients (16). Adenovirus containing an empty shuttle vector was used as a control. For adenovirus infection, 106 B16-F10 cells were incubated in serum-free DMEM for 2 hours with different amounts of virus (200–2500 plaque-forming units [pfu]), after which the cells were washed and cultured in DMEM with 10% FBS.

Subcutaneous Tumor Growth and Experimental Metastasis Assays

We injected the dorsal neck region of C57BL/6 mice (The Jackson Laboratory, Bar Harbor, ME) subcutaneously with B16-F10 cells infected with either control adenovirus or adenovirus expressing mouse thymosin {beta}4 (2 x 105 cells/0.2 mL; 20 mice per group) and used calipers to measure the size of the resulting tumors several times per week for 20 days (17).

We also examined the metastatic potential of the adenovirus-infected cells by using a lung colonization assay, as described previously (17). In brief, we injected B16-F10 cells infected with either control adenovirus or adenovirus expressing mouse thymosin {beta}4 (1 x 105 cells/0.2 mL) into the tail veins of C57BL/6 mice (20 mice per group). Two weeks later, the mice were killed by asphyxiation with CO2 and their lungs were removed. The metastatic nodules on the surface of their lungs were counted. All animal work was performed under protocols approved by the NIDCR Animal Care and Use Committee.

Northern Blot Analysis

We used an RNA mini-prep kit (Stratagene) to purify total RNA from B16 cells. Ten micrograms of each RNA sample was separated by formaldehyde–agarose gel electrophoresis, transferred to a Nytran membrane (Schleicher and Schuell, Keene, NH), and hybridized to a [32P] dCTP-labeled thymosin {beta}4 cDNA probe, as previously described (18).

Western Blot Analysis and Immunostaining

Western blot analysis was conducted as described (14). Briefly, 100 µg of cell lysate or conditioned media was separated by electrophoresis on a Novex 4-20% Tris–glycine gel (Invitrogen). Conditioned media were prepared from 106 cells cultured in DMEM without FBS for 18 hours and were concentrated with the use of Microcon YM-3 filters (Millipore, Bedford, MA). The protein concentrations of the lysates and conditioned media were determined by the bicinchoninic acid protein assay system (Pierce, Rockford, IL), and equal amounts of each sample were separated by electrophoresis on Novex 4-20% Tris–glycine gel. Equal protein loading was confirmed by Coomassie blue staining of duplicate gels after electrophoresis. The gels were incubated for 1 hour in phosphate-buffered saline (PBS) containing 10% glutaraldehyde (Sigma-Aldrich, St. Louis, MO), washed three times for 20 minutes in PBS, and further incubated in a blotting buffer containing 1x Novex Tris–glycine transfer buffer (Invitrogen) and 20% methanol for 30 minutes at room temperature. Proteins were transferred to a polyvinylidene difluoride membrane (Invitrogen) by electrotransfer. The membrane was pre-incubated for 2 hours in PBS containing 5% skim milk and 0.05% Tween 20 (PBS-T). The membrane was incubated for 1 hour at room temperature in PBS-T plus antibodies (rabbit polyclonal thymosin {beta}4, 1 : 1000 dilution; ALPCO Diagnostics, Windham, NH), goat polyclonal vascular endothelial growth factor (VEGF; 1 : 1000 dilution; R&D Systems, Minneapolis, MN), goat polyclonal fibroblast growth factor-1 (FGF-1; 1 : 100 dilution; Santa Cruz Biotechnology, Santa Cruz, CA), mouse monoclonal fibroblast growth factor (FGF-2; 1 : 1000 dilution; Upstate Biotechnology, Lake Placid, NY), mouse monoclonal insulin-like growth factor (IGF; 1 : 1000 dilution; R&D Systems), goat polyclonal platelet-derived growth factor (PDGF; 1 : 1000 dilution; Upstate Biotechnology), or rabbit polyclonal epidermal growth factor (EGF; 1 : 100 dilution; Upstate Biotechnology). The membranes were washed five times with PBS-T and then incubated with a species-appropriate horseradish peroxidase–conjugated secondary antibody (Santa Cruz Biotechnology) for 1 hour at room temperature. The membranes were washed five times with PBS-T, and bound antibody was detected with an enhanced chemiluminescence detection kit (Amersham Biosciences, Buckinghamshire, UK). Recombinant VEGF and FGF-1, used as positive controls in western blot, were purchased from R&D Systems and Santa Cruz Biotechnology, respectively.

For immunohistochemistry, mouse lungs containing tumors derived from B16-F10 cells were fixed with 4% formaldehyde (Sigma-Aldrich) and paraffinized. The sections were deparaffinized and immunostained with a rabbit polyclonal antibody to thymosin {beta}4 (1 : 100 dilution; a gift from Allan Goldstein, George Washington University, Washington, DC); antibody binding was detected with the use of an EnVision+ peroxidase system (DAKO, Carpinteria, CA).

For immunocytochemistry, B16-F10 cells infected with either thymosin {beta}4–expressing or control adenovirus were grown on cover slips and visualized by confocal microscopy, as previously described (19). Briefly, B16-F10 cells on glass cover slips were rinsed three times with PBS, fixed for 10 minutes at room temperature by incubation in 4% paraformaldehyde, and then permeabilized by incubation in PBS containing 0.1% Triton X-100 for 5 minutes. The cells were incubated with 1% bovine serum albumin in PBS for 1 hour to block nonspecific antibody binding and then for 1 hour at room temperature with an anti-thymosin {beta}4 antibody (ALPCO Diagnostics) diluted 1 : 100 in PBS. The cover slips were washed three times with PBS and incubated with affinity-isolated tetramethylrhodamine isothiocyanate (TRITC)-conjugated goat anti-rabbit immunoglobulin G (Molecular Probes, Eugene, OR). For localization of F-actin, the fixed cells were washed three times with PBS for 15 minutes and incubated with fluorescein isothiocyanate–labeled phalloidin (Sigma-Aldrich). All labeled cells were rinsed three times with PBS and once with distilled water and were then mounted on glass slides with the use of a fluorescent mounting medium (DAKO). The cells were observed for epifluorescence with the use of a confocal laser-scanning microscope.

Cell Proliferation Assay

A methylthiazoletetrazolium (MTT)-based cell growth determination kit (Sigma-Aldrich) was used to measure the proliferation of the adenovirus-infected B16-F10 cells. Proliferation was measured every 24 hours for 3 days. The experiments were repeated three times, and samples were tested in triplicate. The mean values and 95% confidence intervals (CIs) were determined.

Gelatin-Based Zymography

Conditioned media were prepared from 106 adenovirus-infected B16-F10 cells cultured with DMEM without FBS for 18 hours and concentrated by Microcon YM-3 filters. Concentrated conditioned media were separated by Novex Zymogram (gelatin) gels (Invitrogen). After electrophoresis, the gels were rinsed with Novex Zymogram Renaturing Buffer (Invitrogen) for 30 minutes, and gels were equilibrated with Novex Zymogram Developing Buffer (Invitrogen) for 30 minutes at room temperature. The gels were then rinsed with fresh Novex Zymogram Developing Buffer and incubated at 37 °C for 18 hours. Gelatinase-like matrix metalloproteinases MMP-2 or MMP-9 were identified following staining of the gel in 0.25% Coomassie blue R250 and destaining with 7% acetic acid. The digested area appeared clear on a blue background, indicating the expression and activity of gelatinases.

In Vitro Migration and Invasion Assays

In vitro migration and invasion assays were performed as previously described (17,20). Briefly, we used HTS FluoroBlok 24-well chambers containing membranes with an 8-µm pore size (Fisher Scientific, Pittsburgh, PA) for both assays. For the migration assay, we placed conditioned medium obtained by culturing mouse NIH-3T3 cells for 18 hours in serum-free DMEM into the lower chambers of each well. We resuspended 105 adenovirus-infected B16-F10 cells in 100 µL of serum-free DMEM and placed them in the upper chambers of each well. The fluoro blocks were incubated for 16 hours at 37 °C, and the cells in the lower chambers were stained with 5 µg/mL calcein AM (Molecular Probes) for 30 minutes. The fluorescence of the cells in the lower part of the chamber was determined with a computer-based fluorescence reader (Wallac Victor 2; PerkinElmer, Wellesley, MA). The invasion assay was performed in a similar fashion, except that the upper surface of the fluoro block filter was coated with 20 µL of 0.5 mg/mL Matrigel (BD Biosciences, Bedford, MA) before the cells were added to the upper chambers. All experiments were repeated at least three times, and each data point was measured in triplicate. The mean values and 95% confidence intervals were determined.

Quantitation of Blood Vessels in Solid Tumors

We determined the number of blood vessels in eight solid tumors derived from subcutaneous injection of adenovirus-infected (control and thymosin {beta}4–expressing) B16-F10 cells by staining with a rat polyclonal anti-CD31/platelet–endothelial cell adhesion molecule-1 (PECAM-1) antibody that recognizes PECAM-1 on endothelial cells (1 : 100 dilution; Pharmingen, San Diego, CA). Antibody binding was detected with the use of an EnVision+ peroxidase system (DAKO), and the number of blood vessels was counted in six fields per three randomly chosen sections.

Statistical Analysis

The size of the solid tumors, the number of metastatic nodules, and the number of blood vessels were measured for calculation of mean values and 95% confidence intervals. We calculated mean values and 95% confidence intervals for cell proliferation, migration, and invasion from data collected in at least three independent experiments. Statistical significance of differences among the groups was determined using a two-tailed Student’s t test. P values less than .05 were considered statistically significant.


    RESULTS
 Top
 Abstract
 Introduction
 Materials and Methods
 Results
 Discussion
 References
 
Expression of Thymosin {beta}4 in a Highly Metastatic Lung Tumor and in Cultured Melanoma Cells

We analyzed thymosin {beta}4 mRNA levels in B16 melanoma cell lines and in primary cultured B16-F10 cells derived from lung tumors using northern blot analysis. Thymosin {beta}4 mRNA was detected in the primary cultured B16-F10 cells derived from lung tumors but not in the B16-F1, B16-F10, or B16-BL6 cell lines (Fig. 1, A). Immunohistochemical staining indicated that thymosin {beta}4 was more highly expressed in the lung tumor tissue that was derived from the B16-F10 cells (which have distinctive large nuclei) than in surrounding non-tumor tissue (Fig. 1, B). These data suggest that thymosin {beta}4 expression is elevated in metastatic tumor cells in vivo and in primary cultured tumor cells derived from metastatic lesions.



View larger version (72K):
[in this window]
[in a new window]
 
Fig. 1. Expression of thymosin {beta}4 in cultured B16 melanoma cell lines and in primary cultured B16-F10 cells derived from lung metastases. A) Northern blot analysis was conducted to analyze the expression of thymosin {beta}4 in B16-F1, B16-F10, and B16-BL6 cells and in B16-F10 cells from a lung tumor (top panel). The amount of total RNA in all lanes was almost the same and was confirmed with 28S and 18S RNA (bottom panel). B) Immunohistochemical analysis of thymosin {beta}4 expression in a lung tumor derived from metastasized B16-F10 cells.

 
Expression of Thymosin {beta}4 by Adenovirus-Infected Cultured Melanoma Cells

To elucidate the role of thymosin {beta}4 in tumor metastasis, we constructed a thymosin {beta}4 adenoviral expression vector and used it to infect B16-F10 cells. We first determined the expression level of thymosin {beta}4 in lysates made from infected cells and found that the level of thymosin {beta}4 protein increased with the amount of virus used to infect the cells and that thymosin {beta}4 was detectable for at least 6 days after infection, although it peaked at 2–3 days (Fig. 2). The amount of total protein in the cell lysates, analyzed by Coomassie blue staining of the protein gel, did not change after virus infection (data not shown). Thymosin {beta}4 was not detected in the culture medium of infected cells, suggesting that thymosin {beta}4 was not secreted by the infected cells. In all of the subsequent experiments, we infected cells with 200 pfu of adenovirus per cell, which has the minimum dose of virus necessary to induce detectable thymosin {beta}4 expression. This dose of virus had no effect on cell proliferation or viability (data not shown).



View larger version (46K):
[in this window]
[in a new window]
 
Fig. 2. Expression of thymosin {beta}4 in adenovirus-infected B16-F10 cells. Expression was analyzed by western blotting for several days after infection. Both lysates and conditioned media were analyzed. Controls (Cont) were cell lysates and conditioned media obtained 3 days after infection of B16-F10 cells with control adenovirus.

 
Effect of Thymosin {beta}4 Expression on Tumor Growth and Metastasis

We examined the effects of thymosin {beta}4 expression on tumor growth and metastasis by injecting separate groups of mice (n = 20 per test group) subcutaneously and intravenously, respectively, with B16-F10 cells infected with adenovirus containing control or thymosin {beta}4 expression vector. At 20 days after subcutaneous injection, mean tumor size was 21.7 mm (95% CI = 17.7 to 25.7 mm) for mice injected with B16-F10 cells infected with thymosin {beta}4–expressing adenovirus and 13.3 mm (95% CI = 11.1 to 15.3 mm) for mice injected with B16-F10 cells infected with control adenovirus (difference = 8.4 mm; P = .036) (Fig. 3, A). Two weeks after mice were injected intravenously with adenovirus-infected cells, the mean number of metastatic lung nodules was 46.7 (95% CI = 35.0 to 57.7) for mice injected with B16-F10 cells infected with thymosin {beta}4–expressing adenovirus and 10.9 (95% CI = 6.2 to 15.6) for mice injected with B16-F10 cells infected with control adenovirus (difference = 35.8 metastatic lung nodules; P<.001) (Fig. 3, B). Mice in the two groups had metastatic lung nodules that were small and of similar size (data not shown), presumably because we dissected the mice only 2 weeks after the B16-F10 cell injections. These data suggest that overexpression of thymosin {beta}4 is associated with increases in primary tumor growth and in the number of lung metastases.



View larger version (12K):
[in this window]
[in a new window]
 
Fig. 3. Effect of thymosin {beta}4 on tumor growth and metastasis in vivo. A) Subcutaneous tumor growth assay. Thymosin {beta}4–expressing adenovirus- and control adenovirus–infected B16-F10 cells were injected subcutaneously into mice (20 mice per group), and the size of the resulting tumors was measured several times per week for 20 days. Symbols {diamondsuit} and {blacksquare} designate control and thymosin {beta}4 adenovirus–infected B16-F10 cells, respectively. *P = .036 compared with control adenovirus infected cells at day 20. B) Experimental metastasis assay. Cells infected with control adenovirus (Cont) or thymosin {beta}4–expressing adenovirus (Thy{beta}4) were injected into the tail veins of mice (20 mice per group), and the mice were killed 2 weeks after injection. The number of metastatic lung nodules was determined by direct counting of the nodules on the lung. *P<.001 compared with control adenovirus–infected cells. Statistical significance of the difference between the tumors formed from the control and those from the adenovirus-infected cells was determined by using a two-tailed Student’s t test, and error bars correspond to 95% confidence intervals.

 
Effect of Thymosin {beta}4 Expression on Cell Proliferation, Migration, and Invasion and MMP Expression and Activity In Vitro

We used adenovirus-infected B16-F10 cells to examine the functional role(s) of thymosin {beta}4 in tumor metastasis. Cells infected with thymosin {beta}4–expressing adenovirus displayed the same level of cell proliferation as cells infected with the control adenovirus (Fig. 4). To analyze the effect of thymosin {beta}4 on the expression or activity of MMPs, i.e., enzymes that degrade the extracellular matrix, we conducted gelatin-based zymography with the conditioned media prepared from adenovirus-infected B16-F10 cells. Thymosin {beta}4 overexpression was not associated with the expression or activity of MMP-2 or MMP-9 (data not shown).



View larger version (10K):
[in this window]
[in a new window]
 
Fig. 4. Effect of thymosin {beta}4 overexpression on B16-F10 cell proliferation, migration, and invasion in vitro, and angiogenesis in vivo. Cell proliferation, migration, invasion, and angiogenic activity (i.e., the mean number of vessels per microscopic field) were measured at various times after cells were infected with either control adenovirus (open bars) or thymosin {beta}4–expressing adenovirus (solid bars). *P = .00002 and **P = .00011. Statistical significance of the differences between the control and adenovirus-infected cells was determined by using a two-tailed Student’s t test, and error bars correspond to 95% confidence intervals.

 
Thymosin {beta}4 is thought to be involved in cell migration because it binds F-actin and regulates actin polymerization (21-23). We conducted in vitro assays to examine the effect of thymosin {beta}4 overexpression on the migration and invasion activities of adenovirus-infected B16-F10 cells. The effect of thymosin {beta}4 expression on cell migration and invasion was analyzed by measuring the fluorescence of migrating or invading cells that were stained with calcein AM. The fold change in fluorescence was based on comparisons with the fluorescence of migrating or invading control adenovirus–infected cells. Thymosin {beta}4 overexpression was associated with a 2.3-fold increase in cell migration (95% CI = 1.9- to 2.7-fold; P<.001) but had no effect on cell invasion (Fig. 4).

We next used confocal laser-scanning immunofluorescence microscopy to examine the subcellular distribution of thymosin {beta}4 and actin in adenovirus-infected cells. Cells were co-stained with an anti-thymosin {beta}4 antibody (red) and FITC-labeled phalloidin (green), which binds to F-actin. Thymosin {beta}4 was detected in the cytoplasm and in the nucleus of cells infected with thymosin {beta}4–expressing adenovirus (Fig. 5) but was not detectable in cells infected with control adenovirus (data not shown). F-actin was localized to the cell surface in cells infected with control adenovirus and in cells infected with thymosin {beta}4–expressing adenovirus (data not shown), in agreement with previously published results (24). When we merged the images for thymosin {beta}4 and actin staining, we found that thymosin {beta}4 co-localized with F-actin only at the leading edges of the cell (Fig. 5).



View larger version (37K):
[in this window]
[in a new window]
 
Fig. 5. Immunofluorescence staining of B16-F10 cells infected with thymosin {beta}4–expressing adenovirus for thymosin {beta}4 and F-actin. Cells were stained 2 days after adenovirus infection with anti-thymosin {beta}4 antibody (red) and with FITC-labeled phalloidin (green), which binds to F-actin, and were examined by confocal laser-scanning microscopy. Image merging of thymosin {beta}4 and F-actin is shown in the bottom panel.

 
Effect of Thymosin {beta}4 Expression on Angiogenesis

We previously reported that thymosin {beta}4 expression is associated with the induction of endothelial cell differentiation and the promotion of angiogenesis (18,25). To analyze the angiogenic activity of thymosin {beta}4, we counted the number of blood vessels in the solid tumors formed by injecting mice with B16-F10 cells infected with control or thymosin {beta}4–expressing adenovirus. The fold increase in the number of blood vessels was determined by comparison with the number of blood vessels formed by injecting mice with control adenovirus–infected B16-F10 cells. Thymosin {beta}4 overexpression was associated with a 4.4-fold mean increase (95% CI = 3.3- to 5.5-fold mean increase; P<.001) in the number of blood vessels in solid tumors derived from injected B16-F10 cells (Fig. 4).

Although these data suggest that thymosin {beta}4 stimulates angiogenesis, it is possible that this effect may be an indirect reflection of changes in the levels of growth factors that are known to regulate tumor angiogenesis. We therefore measured the levels of VEGF, FGF-1, FGF-2, IGF, PDGF, and EGF in B16-F10 cells at different times after infection with control or thymosin {beta}4–expressing adenovirus (26-30). We found that VEGF levels in cells infected with thymosin {beta}4–expressing adenovirus peaked at 2 days after infection. The increased VEGF expression pattern paralleled that of thymosin {beta}4 in cells infected with thymosin {beta}4–expressing adenovirus (Fig. 6). By contrast, the levels of FGF-1, PDGF, and EGF did not change after the cells were infected with thymosin {beta}4–expressing adenovirus (Fig. 6). Neither FGF-2 nor IGF was detectable by western blotting in cells infected with the control or the thymosin {beta}4–expressing adenovirus (data not shown).



View larger version (61K):
[in this window]
[in a new window]
 
Fig. 6. Effect of thymosin {beta}4 overexpression on the expression of VEGF, FGF-1, EGF, and PDGF by B16-F10 cells infected with thymosin {beta}4–expressing adenovirus. Western blot analysis was done with concentrated conditioned medium obtained from cells at the indicted times after infection. The control (Cont) was concentrated conditioned medium obtained from B16-F10 cells infected with control adenovirus 3 days after infection. We used 50 ng of recombinant VEGF (rmVEGF) and FGF-1 (rmFGF-1) as positive controls. The protein concentrations were determined by the bicinchoninic acid protein assay, and equal protein loading was confirmed by Coomassie blue staining of duplicate gels after electrophoresis.

 
Previously, it was found that addition of a synthetic thymosin {beta}4 peptide to the medium of cultured cells was associated with increases in their angiogenic and wound-healing activities (3-8). We therefore treated B16-F10 cells with various amounts of a synthetic thymosin {beta}4 peptide and analyzed the expression of angiogenic growth factors. We found that cells treated with as little as 1 ng of the thymosin {beta}4 peptide had higher levels of VEGF than untreated cells, but no changes were observed in the levels of the other growth factors (Fig. 7). At high doses of peptide, VEGF levels decreased (Fig. 7). These results suggest that thymosin {beta}4 may induce angiogenesis, in part, by increasing the levels of VEGF.



View larger version (58K):
[in this window]
[in a new window]
 
Fig. 7. Effect of exogenous synthetic thymosin {beta}4 peptide on VEGF, FGF-1, EGF, and PDGF expression in B16-F10 cells. Western blot analysis of VEGF, FGF-1, EGF, and PDGF expression was conducted with concentrated conditioned media obtained from B16-F10 cells incubated with various amounts of synthetic thymosin {beta}4 peptide for 18 hours. rmVEGF and rmFGF-1 designate recombinant VEGF (50 ng) and FGF-1 (50 ng), respectively, which served as positive controls. The protein concentrations were determined by the bicinchoninic acid protein assay, and equal protein loading was confirmed by Coomassie blue staining of the duplicated gels after electrophoresis.

 

    DISCUSSION
 Top
 Abstract
 Introduction
 Materials and Methods
 Results
 Discussion
 References
 
Tumor metastasis is a multistep process that involves tumor cell migration and invasion, angiogenesis, and metastatic tumor cell growth (31,32). Results from several studies suggest that thymosin {beta}4 expression may be important in tumor metastasis. For example, Clark and colleagues (10) injected B16 melanoma cells intravenously into mouse tail veins and selected metastasized B16 melanoma cells from resulting lung tumors. They found that the metastatic potential of B16 cells increased when they repeated this in vivo selection scheme, and that thymosin {beta}4 levels were elevated in the highly metastatic B16-F10 cells cultured from the lung tumors. In addition, thymosin {beta}4 levels were elevated in human melanoma metastatic lesions compared with the parental cells from which those lesions were derived (10). Increased levels of thymosin {beta}4 are also thought to regulate metastasis, invasion, and migration of fibrosarcoma cells (14). However, the mechanism by which thymosin {beta}4 functions in tumor metastasis is not understood. Our results identify several activities that contribute to metastasis and are associated with increased thymosin {beta}4 levels. To confirm the result of Clark and colleagues (10), we compared the expression level of thymosin {beta}4 in B16 cell lines and primary cultured B16-F10 cells derived from lung metastasis. The expression of thymosin {beta}4 was detected only in the B16-F10 cells cultured from lung metastases and not in the other B16 cell lines. Furthermore, B16-F10 cells that metastasized to the lung expressed high levels of thymosin {beta}4. These data suggest that thymosin {beta}4 is involved in tumor metastasis. In addition, B16-F10 cells infected with thymosin {beta}4–expressing adenovirus displayed more tumor growth and metastasis in vivo than that observed with B16-F10 cells infected with control adenovirus. We also found that thymosin {beta}4 overexpression was associated with an increase in cell migration, which may reflect the intracellular colocalization of thymosin {beta}4 with F-actin. Ballweber and colleagues (21) showed that thymosin {beta}4 binds to filamentous actin, and the results of other studies (23) demonstrate that thymosin {beta}4 regulates actin polymerization. These results suggest that thymosin {beta}4 may stimulate cell migration by regulating actin polymerization.

We previously reported that thymosin {beta}4 has strong angiogenic activity (25). To confirm the effect of thymosin {beta}4 on angiogenesis, we counted blood vessels in the solid tumors derived from B16-F10 cells infected with control or thymosin {beta}4–expressing adenovirus. Thymosin {beta}4 overexpression was associated with the stimulation of blood vessel formation, as shown in Fig. 4. We addressed the possible mechanism for the angiogenic effect of thymosin {beta}4 by showing that VEGF expression was increased upon infection of cells with a thymosin {beta}4–expressing adenovirus or by treatment of cells with synthetic thymosin {beta}4 peptide. Thymosin {beta}4 is not secreted, but we have found that exogenously added thymosin {beta}4 localizes in the cytoplasm and nucleus of cells (data not shown). However, it is not clear whether thymosin {beta}4 alone is angiogenic, whether all of the angiogenic potential associated with thymosin {beta}4 is actually mediated by VEGF, or whether both VEGF and thymosin {beta}4 are functionally important. Thymosin {beta}4 is also reported to have anti-inflammatory activity (8,33); thus, it may also act in promoting tumor growth by inhibiting immune surveillance.


    REFERENCES
 Top
 Abstract
 Introduction
 Materials and Methods
 Results
 Discussion
 References
 

1 Low TL, Hu SK, Goldstein AL. Complete amino acid sequence of bovine thymosin beta 4: a thymic hormone that induces terminal deoxynucleotidyl transferase activity in thymocyte populations. Proc Natl Acad Sci U S A 1981;78:1162–6.[Abstract]

2 Grant DS, Kinsella JL, Kibbey MC, LaFlamme S, Burbelo PD, Goldstein AL, et al. Matrigel induces thymosin beta 4 gene in differentiating endothelial cells. J Cell Sci 1995;108(Pt 12):3685–94.[Abstract/Free Full Text]

3 Malinda KM, Goldstein AL, Kleinman HK. Thymosin beta 4 stimulates directional migration of human umbilical vein endothelial cells. FASEB J 1997;11:474–81.[Abstract/Free Full Text]

4 Frohm M, Gunne H, Bergman AC, Agerberth B, Bergman T, Boman A, et al. Biochemical and antibacterial analysis of human wound and blister fluid. Eur J Biochem 1996;237:86–92.[Abstract]

5 Malinda KM, Sidhu GS, Mani H, Banaudha K, Maheshwari RK, Goldstein AL, et al. Thymosin beta 4 accelerates wound healing. J Invest Dermatol 1999;113:364–8.[Abstract/Free Full Text]

6 Koutrafouri V, Leondiadis L, Avgoustakis K, Livaniou E, Czarnecki J, Ithakissios DS, et al. Effect of thymosin peptides on the chick chorioallantoic membrane angiogenesis model. Biochim Biophys Acta 2001;1568:60–6.[ISI][Medline]

7 Sosne G, Chan CC, Thai K, Kennedy M, Szliter EA, Hazlett LD, et al. Thymosin beta 4 promotes corneal wound healing and modulates inflammatory mediators in vivo. Exp Eye Res 2001;72:605–8.[CrossRef][ISI][Medline]

8 Sosne G, Szliter EA, Barrett R, Kernacki KA, Kleinman H, Hazlett LD. Thymosin beta 4 promotes corneal wound healing and decreases inflammation in vivo following alkali injury. Exp Eye Res 2002;74:293–9.[CrossRef][ISI][Medline]

9 Otto AM, Müller CS, Huff T, Hannappel E. Chemotherapeutic drugs change actin skeleton organization and the expression of beta-thymosins in human breast cancer cells. J Cancer Res Clin Oncol 2002;128:247–56.[CrossRef][ISI][Medline]

10 Clark EA, Golub TR, Lander ES, Hynes RO. Genomic analysis of metastasis reveals an essential role for RhoC. Nature 2000;406:532–5.[CrossRef][ISI][Medline]

11 Bao L, Loda M, Zetter BR. Thymosin beta15 expression in tumor cell lines with varying metastatic potential. Clin Exp Metastasis 1998;16:227–33.[CrossRef][ISI][Medline]

12 Bao L, Loda M, Janmey PA, Stewart R, Anand-Apte B, Zetter BR. Thymosin beta 15: a novel regulator of tumor cell motility upregulated in metastatic prostate cancer. Nat Med 1996;2:1322–8.[ISI][Medline]

13 Yamamoto T, Gotoh M, Kitajima M, Hirohashi S. Thymosin beta-4 expression is correlated with metastatic capacity of colorectal carcinomas. Biochem Biophys Res Commun 1993;193:706–10.[CrossRef][ISI][Medline]

14 Kobayashi T, Okada F, Fujii N, Tomita N, Ito S, Tazawa H, et al. Thymosin-beta4 regulates motility and metastasis of malignant mouse fibrosarcoma cells. Am J Pathol 2002;160:869–82.[Abstract/Free Full Text]

15 Becker TC, Noel RJ, Coats WS, Gomez-Foix AM, Alam T, Gerard RD, et al. Use of recombinant adenovirus for metabolic engineering of mammalian cell. Methods Cell Biol 1994;43 Pt A:161–89.[ISI][Medline]

16 Bett AJ, Haddara W, Prevec L, Graham FL. An efficient and flexible system for construction of adenovirus vectors with insertions or deletions in early regions 1 and 3. Proc Natl Acad Sci U S A 1994;91:8802–6.[Abstract]

17 Song SY, Nomizu M, Yamada Y, Kleinman HK. Liver metastasis formation by laminin-1 peptide (LQVQLSIR)-adhesion selected B16-F10 melanoma cells. Int J Cancer 1997;71:436–41.[CrossRef][ISI][Medline]

18 Grant DS, Kinsella JL, Kibbey MC, LaFlamme S, Burbelo PD, Goldstein AL, et al. Matrigel induces thymosin beta 4 gene in differentiating endothelial cells. J Cell Sci 1995;108(Pt 12):3685–94.[Abstract/Free Full Text]

19 Jeong MJ, Yoo J, Lee SS, Lee KI, Cho A, Kwon BM, et al. Increased GTP-binding to dynamin II does not stimulate receptor-mediated endocytosis. Biochem Biophys Res Commun 2001;283:136–42.[CrossRef][ISI][Medline]

20 Sunder-Plassmann G, Hofbauer R, Sengoelge G, Horl WH. Quantification of leukocyte migration: improvement of a method. Immunol Invest 1996;25:49–63.[ISI][Medline]

21 Ballweber E, Hannappel E, Huff T, Stephan H, Haener M, Taschner N, et al. Polymerisation of chemically cross-linked actin:thymosin beta(4) complex to filamentous actin: alteration in helical parameters and visualisation of thymosin beta(4) binding on F-actin. J Mol Biol 2002;315:613–25.[CrossRef][ISI][Medline]

22 Ballweber E, Galla M, Aktories K, Yeoh S, Weeds AG, Mannherz HG. Interaction of ADP-ribosylated actin with actin binding proteins. FEBS Lett 2001;508:131–5.[CrossRef][ISI][Medline]

23 Yarmola EG, Parikh S, Bubb MR. Formation and implications of a ternary complex of profilin, thymosin beta 4, and actin. J Biol Chem 2001;276:45555–63.[Abstract/Free Full Text]

24 Moroianu J, Fett JW, Riordan JF, Vallee BL. Actin is a surface component of calf pulmonary artery endothelial cells in culture. Proc Natl Acad Sci U S A 1993;90:3815–9.[Abstract]

25 Grant DS, Rose W, Yaen C, Goldstein A. Thymosin beta 4 enhances endothelial cell differentiation and angiogenesis. Angiogenesis 1999;3:125–35.[CrossRef][Medline]

26 Liekens S, De Clercq E, Neyts J. Angiogenesis: regulators and clinical applications. Biochem Pharmacol 2001;61:253–70.[CrossRef][ISI][Medline]

27 Veikkola T, Alitalo K. VEGFs, receptors and angiogenesis. Semin Cancer Biol 1999;9:211–20.[CrossRef][ISI][Medline]

28 Ferrara N. Vascular endothelial growth factor: molecular and biological aspects. Curr Top Microbiol Immunol 1999;237:1–30.[ISI][Medline]

29 Bussolino F, Albini A, Camussi G, Presta M, Viglietto G, Ziche M, et al. Role of soluble mediators in angiogenesis. Eur J Cancer 1996;32A:2401–12.[CrossRef]

30 Sato Y, Okamura K, Morimoto A, Hamanaka R, Hamaguchi K, Shimada T, et al. Indispensable role of tissue-type plasminogen activator in growth factor-dependent tube formation of human microvascular endothelial cells in vitro. Exp Cell Res 1993;204:223–9.[CrossRef][ISI][Medline]

31 Woodhouse EC, Chuaqui RF, Liotta LA. General mechanisms of metastasis. Cancer 1997;80:1529–37.[CrossRef][ISI][Medline]

32 Liotta LA, Steeg PS, Stetler-Stevenson WG. Cancer metastasis and angiogenesis: an imbalance of positive and negative regulation. Cell 1991;64:327–36.[ISI][Medline]

33 Young JD, Lawrence AJ, MacLean AG, Leung BP, McInnes IB, Canas B, et al. Thymosin beta 4 sulfoxide is an anti-inflammatory agent generated by monocytes in the presence of glucocorticoids. Nat Med 1999;5:1424–7.[CrossRef][ISI][Medline]

Manuscript received April 10, 2003; revised September 8, 2003; accepted September 15, 2003.


This article has been cited by other articles in HighWire Press-hosted journals:


Editorial about this Article

             
Copyright © 2003 Oxford University Press (unless otherwise stated)
Oxford University Press Privacy Policy and Legal Statement