Overexpression of CAP1 and its significance in tumor cell proliferation, migration and invasion in glioma

  • Authors:
    • Yue-Chao Fan
    • Chen-Chen Cui
    • Yi-Shuo Zhu
    • Lei Zhang
    • Meng Shi
    • Jin-Song Yu
    • Jin Bai
    • Jun-Nian Zheng
  • View Affiliations

  • Published online on: July 13, 2016     https://doi.org/10.3892/or.2016.4936
  • Pages: 1619-1625
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Adenylate cyclase-associated protein 1 (CAP1), a protein related to the regulation of actin filaments and the Ras/cAMP pathway, is associated with tumor progression. Nevertheless, the expression level and effects of CAP1 in regards to glioma have not been reported. In the present study, we examined the expression of CAP1 in glioma and tumor adjacent normal brain tissues by tissue microarray and immunohistochemistry. Our results showed that CAP1 was overexpressed in glioma tissues in comparison with that noted in the tumor adjacent normal brain tissues and increased staining of CAP1 was found to be correlated with WHO stage. In addition, we discovered that knockdown of CAP1 by specific RNA interference markedly inhibited cell growth and caused downregulation of the proliferation markers, PCNA and cyclin A. We further demonstrated that knockdown of CAP1 inhibited cell metastatic abilities by downregulating N-cadherin and vimentin and upregulating E-cadherin. These findings revealed that CAP1 expression is markedly increased in human glioma and that downregulation of CAP1 in tumors may serve as a treatment for glioma patients.

Introduction

Glioma is the most common primary intracranial tumor that arises from the neuroectoderm, and thereby is also known as neuroectodermal or neuroepithelial tumor, and accounts for 50% of primary intracranial tumors (1,2). As the most aggressive type of glioma, glioblastoma multiform (GBM) is a grade IV histological malignancy according to the WHO classification, with a median patient survival period of 12–14 months (3). The poor prognosis of glioblastomas is largely attributed to their rapid growth, high invasive and migratory abilities and high rate of recurrence (4). Therefore, a comprehensive understanding of the molecular mechanisms which expedite glioma tumor proliferation, migration and invasion is urgently needed in order to develop more effective therapies against glioma.

It is known in cell physiology that suppression of tumor cell metastasis includes regulation of cell-cell junctions, cytoskeletal structure and cell morphology. The actin cytoskeleton is pivotal in tumor cell motility and transition, as its remodeling regulates important cellular processes, such as cell adhesion, metastasis and morphological transformation (58). Cyclase-associated protein 1 (CAP1) was initially cloned from budding yeast and is located in the downstream of the ras gene (9). The N-terminal region of CAP binds to Cyr1 and is associated with Ras responsiveness in yeast (1012). The C-terminal region of this protein binds monomeric actin with high affinity, and is necessary for normal cellular morphology (13) Previous research has shown that CAP1 is overexpressed in hepatocellular carcinoma (14), breast (15) and lung cancer (16), and esophageal squamous cell carcinoma (17). Yet, there is sparse research on the influence of CAP1 in gliomas.

In the present study, to evaluate the relationship between the expression of CAP1 and clinicopathological features of glioma, we used a tissue microarray (TMA) of human glioma patient tissues and immunohistochemistry. We discovered that CAP1 expression was obviously upregulated in the glioma tissues when compared with that noted in the tumor adjacent normal brain tissues and this increased expression was correlated with WHO stage. We found that CAP1 was markedly upregulated in glioma tissues when compared to that in normal brain tissues. We then demonstrated that knockdown of CAP1 suppressed glioma cell proliferation, migratory and invasive abilities in glioma cells. We also investigated the molecular mechanisms of CAP1 in glioma cells.

Materials and methods

Patients and samples

We purchased a glioma TMA kit from Shanxi Alenabio Biotechnology (Xi'an, Shanxi, China) (no. GL2083a). Based on WHO criteria, the tumors were divided according to the pathological grade as follows: 134 cases with grades I–II and 58 cases with grades III–IV. Furthermore, it included 8 cases of normal brain tissue and 8 cases of tumor adjacent normal brain tissue. The array dot diameter was 1.0 mm, and each dot represented a tissue spot from one individual specimen that was selected and pathologically confirmed.

Immunohistochemistry of TMA

TMA slices were dewaxed at 62°C for 2 h, followed by two 30-min washes with xylene. The sections were blended by 5-min washes in 100, 95 and 80% ethanol and sequentially distilled water. Using a microwave the citrate buffer (pH 6.0) was heated to 95°C, and then the sections were placed in the buffer for 15 min. The citrate buffer was cooled to room temperature after the restoration. Phosphate-buffered saline (PBS) was washed out by three 5-min washings, and then hydrogen peroxide was added to the slices for 30 min in order to block endogenous peroxidase activity. After incubating the sections for 30 min with 10% goat serum, the sections were incubated with monoclonal rabbit anti-CAP1 antibody (1:500) (Abcam, Cambridge, MA, USA) at 4°C overnight. On the following day, the slices were removed at 4°C and rewarmed at room temperature for 30 min, and PBS was washed out by three washings for 5 min. Then, the sections were incubated with a biotinylated secondary antibody at room temperature for 30 min, followed by incubation with streptavidin-peroxidase (both from Zhongshan Biotech, Beijing, China) for an additional 30 min. After rinsing with PBS 3 times for 5 min, the sections were stained using DAB (Zhongshan Biotech), rinsed in distilled water and counterstained with hematoxylin. Dehydration was then performed in 80, 95, 95 and 100% ethanol and distilled water sequentially. Then, the sections were sealed with coverslips. Negative controls were stained with non-immune serum to replace primary antibodies.

Evaluation of immunohistochemical staining

The evaluation of CAP1 staining was blindly and independently examined by the intensity of staining and the proportion of tumor cells showing an unequivocal positive reaction. The CAP1 staining intensity was scored as 0–3 (0, negative; 1, weak; 2, moderate; and 3, strong). The proportion of CAP1-positive stained cells was also scored according to 4 categories: 1 (0–25%), 2 (26–50%), 3 (51–75%) and 4 (76–100%). For statistical analyses, the level of CAP1 staining was evaluated by the immunoreactive score (IRS), which was calculated by multiplying the score of the staining intensity and the proportion of positive cells. According to the IRS, the CAP1 staining pattern was categorized as negative (IRS, 0), weak (IRS, 1–3), moderate (IRS, 4–6) and strong (IRS, 8–12).

Cell culture and transfection

U251 and U87 human glioblastoma cell lines were originally obtained from the Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (Shanghai, China). The cells were cultivated in high glucose Dulbecco's modified Eagle's medium (DMEM) with 10% fetal bovine serum (FBS) (Invitrogen, Shanghai, China) in a 5% CO2 humidified atmosphere at 37°C. When cells grew to 50% confluency, they were then transfected. Non-specific control siRNA or CAP1 siRNA (both from Qiagen, Mississauga, ON, Canada) were transfected with siLentFect lipid reagent (Bio-Rad, Hercules, CA, USA) according to the manufacturer's instructions. The medium embodying the transfection reagents was replaced after 24 h. Then, the cells were irrigated twice with PBS and maintained in fresh medium. Forty-eight hours after transfection, the cells were lysed for western blot assay and were used for CCK-8 cell proliferation, cell migration and Matrigel invasion assays.

Western blot analysis

Treated and untreated cells were lysed in radioimmunoprecipitation assay buffer with a freshly added protease inhibitor cocktail (Roche Applied Science, Indianapolis, IN, USA). The protein density was determined using the bicinchoninic acid (BCA) assay (Pierce, Rockford, IL, USA). All protein samples were formulated and denatured. Protein was separated by SDS-PAGE on 8–12% gradient poly-acrylamide gel and transferred onto polyvinylidene difluoride membranes (Millipore, Billerica, MA, USA). The membranes were blocked in 5% non-fat milk for 3 h at room temperature, and then incubated with the primary antibody overnight at 4°C. The following antibodies were applied: rabbit anti-CAP1 (Abcam), rabbit anti-N-cadherin, rabbit anti-vimentin, rabbit anti-E-cadherin (all from Cell Signaling Technology, Beverly, MA, USA), rabbit anti-PCNA and rabbit anti-cyclin A (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) and rabbit anti-β-actin (Cell Signaling Technology). The membranes were then incubated with peroxidase-conjugated Affinipure goat anti-rabbit IgG secondary antibody (Zhongshan Biotech, Beijing, China) for 2 h at room temperature. Detection was performed using an enhanced chemiluminescence method (Pierce).

Cell growth assay

Cell growth was analyzed using the Cell Counting Kit-8 (CCK-8; Beyotime, Nantong, China). U87 and U251 cells were cultured at a density of 5×103 cells/well and suspended in 100 μl high glucose DMEM containing 10% FBS in 96-well plates and incubated for 1, 2, 3 and 4 days, respectively. Then, 10 μl CCK-8 solution was added to each well and incubated at 37°C for 1 h. The optical density was measured at 450 nm on an ELX-800 spectrometer reader (Bio-Tek Instruments, Winooski, VT, USA).

Wound-healing assay

Cells were seeded into 6-well plates at a density of 5×105 cells/well in culture medium and transfected with siRNA. Twenty-four hours after transfection, the cells were scratched on the monolayer with a 10-μl pipette tip and washed twice with PBS. Then, the cells were maintained in high glucose DMEM for an additional 24 h. Images were captured using an inverted Leica phase-contrast microscope (Leica DFC300 FX) at 0 and 48 h time points. The percentage of wound healing of the cells was determined by the ratio of the healing width at each time point to the wound width at 0 h.

Migration assay

Cell migration was determined using a modified two-chamber plate with pore size of 8-μm. For the cell migration assay, 1×105 U251 and U87 cells were seeded in serum-free medium in the upper chamber. To stimulate migration, 10% of serum containing culture medium was added to the bottom wells. After the cell was incubated for 24 h at 37°C, cells on the top surface of the insert were carefully removed with a cotton swab and the cells that had traversed the membrane were fixed in methanol, stained with crystal violet to visualize nuclei and the number of migrating cells was counted under a magnification of ×200 in 5 fields (up, down, median, left and right), and the means for each chamber were determined.

Invasion assay

The invasion assay was performed using a modified two-chamber plate with a pore size of 8-μm. The Transwell filter inserts were coated with Matrigel, and 1×105 U251 and U87 cells were seeded in serum-free medium in the upper chamber. To stimulate invasion, 10% of serum containing culture medium was added to the bottom wells. After 24 h of incubation at 37°C, the cells in the upper chamber were carefully removed with a cotton swab and the cells that had traversed the membrane were fixed in methanol, stained with Giemsa and counted. For counting, the cells in 5 fields (up, down, median, left and right; magnification, ×200) per filter were counted under a microscope.

Statistical analysis

Statistical analysis was carried out using SPSS 19.0 software (SPSS, Inc., Chicago, IL, USA). Values are expressed as the means ± SD. The relationship between CAP1 staining and the clinicopathological parameters of the glioma patients, such as age, gender, WHO grade and histological type was assessed by χ2 test. For CCK-8 cell growth assays, the Student's t-test was used. For migration and invasion assays, the results are expressed as the means ± SD. Each experiment was performed at least 3 times. Differences were considered significant at a P-value <0.05.

Results

CAP1 expression is upregulated in glioma tissues

To investigate whether differential expression of CAP1 exists in glioma tissues, we applied a TMA to detect the CAP1 expression in normal brain and tumor adjacent normal brain tissues, benign tumors (grade I and II) and malignant tumors (grade III and IV). We found that CAP1 was mainly localized in the cytoplasm, yet it was occasionally observed in the nucleus. Representative images are presented in Fig. 1A–D. CAP1-positive staining was detected in 2 of 8 (25.00%) normal brain tissues, 3 of 8 (37.50%) tumor adjacent normal brain tissues and 147 of 192 (76.56%) glioma tissues (Fig. 1E). A significant difference in CAP1 staining was noted between the normal brain and glioma tissues (P<0.05; χ2 test) and between tumor adjacent normal brain and glioma tissues (P<0.05; χ2 test).

Correlation of CAP1 expression with clinicopathological parameters

Investigation of the relationship between CAP1 expression and clinicopathological parameters of the glioma cases are summarized in Table I. WHO grade and histological type are known as vital prognostic markers for glioma patients. Samples with IRS 0–3 and IRS 4–12 were classified as having low and high expression of CAP1. We discovered CAP1-positive staining in 97 of 134 (72.39%) benign tumor tissues and in 50 of 58 (86.21%) malignant tumor tissues. Moreover CAP1 staining was significantly increased in malignant (grade III and IV) compared with benign tumors (grade I and II) (P<0.05; χ2 test; Fig. 1F). However, no obvious correlations were noted between CAP1 expression and other clinicopathological parameters, including patient age (P=0.683), gender (P=0.562) and histological type (P=0.802) (Table I).

Table I

CAP1 staining and clinicopathological characteristics of the 192 glioma patients.

Table I

CAP1 staining and clinicopathological characteristics of the 192 glioma patients.

CAP1 staining
TotalP-valuea
Negative No. (%)Positive No. (%)
Age (years)
 <4622 (24.7)67 (75.3)890.320
 ≥4623 (22.3)80 (77.7)103
Gender
 Male27 (23.1)90 (76.9)1170.883
 Female18 (24.0)57 (76.0)75
WHO grade
 Benign (I–II)37 (27.6)97 (72.4)1340.038
 Malignant (III–IV)8 (13.8)50 (86.2)58
Histological type
 Astrocytoma32 (24.2)100 (75.8)1320.951
 Glioblastoma6 (18.2)27 (71.8)33
 Oligoastrocytoma2 (28.6)5 (71.4)7
 Ependymoma3 (27.3)8 (72.7)11
 Oligodendroglioma2 (22.2)7 (77.8)9

a χ2 test. CAP1, cyclase-associated protein 1.

Knockdown of CAP1 inhibits glioma cell proliferation in vitro

Owing to the fact that CAP1 expression is significantly increased in glioma tissues compared with tumor adjacent normal brain tissues, CAP1 may play vital roles in tumor progression. U251 and U87 cells were transfected with siRNA targeting CAP1 to knock down CAP1 expression. The results of the CCK-8 cell proliferation assays revealed decreased growth rates in the U251 and U87 cells depleted of CAP1 (Fig. 2A and B). To determine whether the decreased proliferative ability after CAP1 knockdown was due to alterations in the proliferation markers, PCNA and cyclin A, we performed western blot analysis. Western blot results showed downregulated levels of PCNA and cyclin A in the glioma cells with silenced CAP1 (Fig. 2C).

Knockdown of CAP1 inhibits glioma cell migration in vitro

Since actin organization plays a significant role in cell motility and migration, we speculated that CAP1 may exert a crucial function in the regulation of cell motility. To examine this hypothesis, we knocked down the expression of CAP1 in U251 and U87 cell lines and analyzed the role of CAP1 on cell migration by wound healing and Transwell assays. We observed that wound recovery was significantly reduced by 26.58 and 38.75% after CAP1 knockdown (Fig. 3A and B), and the percentage of cells that transversed through the micropore membrane in the Transwell assay was significantly decreased by 44.44 and 36.21% (Fig. 3C and D) in the U251 and U87 cells lines as compared with the non-transfected cells. In summary, these results indicated that interference of CAP1 had an evident inhibitory effect on glioma cell motility.

Knockdown of CAP1 inhibits glioma cell invasion in vitro

For the cell invasion assay, downregulation of CAP1 inhibited the cell invasive ability of the U251 and U87 cells by 31.43 and 70.00%, respectively (Fig. 4A and B). To investigate the mechanisms of the regulation of metastasis by CAP1, we utilized western blotting to detect alterations in the protein levels. A decrease in CAP1 resulted in the upregulation of epithelial marker E-cadherin and downregulation of N-cadherin and vimentin (Fig. 4C). Our results indicated that CAP1 expression may expedite migration and invasion by increasing the expression of N-cadherin and vimentin and downregulation of E-cadherin.

Discussion

Cyclase-associated protein 1 (CAP1) was initially cloned by budding yeast and is located in the downstream of the ras gene (9). CAP consists of 4 domains which are involved in actin binding, adenylyl cyclase association in yeast, SH3 binding and oligomerization, respectively (18). As a monomeric actin binding protein, CAP takes part in cell polarization, the allocation of actin filaments and mRNA in Dictyostelium (19). The expression of CAP has a relationship with an anomalous large cell size, random budding pattern and an anomalous actin allocation in yeast (10,20). The human homologue of CAP1 was initially detected in the early 1990's (21). Both mammal and yeast CAPs interplay with actin (13) and play a part in actin turnover (22). Given the vital role of actin filament restructuring in cell metastasis and the regulatory role of CAP1 in actin filament reorganization (18,23), we hypothesized that CAP1 may function in tumor metastasis. In addition, the functions of CAPs in mammals and other vertebrates are not well known. Although a previous study demonstrated that CAP1 is overexpressed in pancreatic cancers and revealed an involvement of CAP1 in the aggressive behavior of pancreatic cancer cells (24), the role and molecular mechanism of CAP1 in glioma remain virtually unknown. In the present study, our data demonstrated that CAP1 was obviously increased in glioma tissues compared to tumor adjacent normal brain tissues by immunohistochemical (Fig. 1A–D). Furthermore, CAP1 expression was upregulated in malignant (grade III and IV) compared with benign tumor tissues (grade I and II) (Fig. 1F). This demonstrated that CAP1 may play a vital role in glioma development and progression.

Cyclin A, a major member of the cyclin family, is a component of the machinery required for progression through S phase (25,26). Fundamental expression of cyclin A has a relationship with tumorigenesis in diverse studies (27,28). Our results identified a significant decrease in cyclin A expression after silencing of CAP1. However, we found an obvious decrease in the expression of proliferating cell nuclear antigen (PCNA) after silencing of CAP1, an associated protein of DNA polymerase δ. Cyclin A and PCNA were simultaneously downregulated after silencing of CAP1 in the glioma cells. While speculative, these results warrant further study.

Epithelial to mesenchymal transition (EMT) is a step that is conducive to an aggressive cell phenotype which is accompanied by the loss of intercellular adhesion in tumors, leading to the formation of migratory mesenchymal cells with invasive characteristics (29,30). Therefore, EMT is crucial in tumor progression and metastasis (31). In epithelial cells, adhesion molecule, E-cadherin, regulates the interactions between cell-cell and tissue framework of organization. The fundamental mechanism involved in the process of EMT is upregulation of the mesenchymal marker vimentin and downregulation of the epithelial marker E-cadherin (32). In the present study, we discovered that downregulation of CAP1 in glioma cells inhibited cell metastatic abilities, which were caused by the downregulation of N-cadherin and vimentin and the upregulation of E-cadherin. However, a more detailed molecular mechanism of how CAP1 regulates the progression of glioma requires further investigation.

In summary, our results demonstrated that CAP1 plays a vital role in human glioma pathogenesis. Overexpression of CAP1 influenced tumor progression by enhancing cell proliferation, migration and invasion. Our results imply that CAP1 may be an important marker and a therapeutic target for gliomas. Thus, the present study may provide a valuable therapeutic strategy to help control the progression of gliomas.

References

1 

Hamza MA and Gilbert M: Targeted therapy in gliomas. Curr Oncol Rep. 16:3792014. View Article : Google Scholar : PubMed/NCBI

2 

Kim YZ: Altered histone modifications in gliomas. Brain Tumor Res Treat. 2:7–21. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Wen PY and Kesari S: Malignant gliomas in adults. N Engl J Med. 359:492–507. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Giese A, Bjerkvig R, Berens ME and Westphal M: Cost of migration: Invasion of malignant gliomas and implications for treatment. J Clin Oncol. 21:1624–1636. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Chang E, Heo KS, Woo CH, Lee H, Le NT, Thomas TN, Fujiwara K and Abe J: MK2 SUMOylation regulates actin filament remodeling and subsequent migration in endothelial cells by inhibiting MK2 kinase and HSP27 phosphorylation. Blood. 117:2527–2537. 2011. View Article : Google Scholar :

6 

Kirfel G, Rigort A, Borm B and Herzog V: Cell migration: Mechanisms of rear detachment and the formation of migration tracks. Eur J Cell Biol. 83:717–724. 2004. View Article : Google Scholar

7 

Hall A: The cytoskeleton and cancer. Cancer Metastasis Rev. 28:5–14. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Fedor-Chaiken M, Deschenes RJ and Broach JR: SRV2, a gene required for RAS activation of adenylate cyclase in yeast. Cell. 61:329–340. 1990. View Article : Google Scholar : PubMed/NCBI

9 

Field J, Vojtek A, Ballester R, Bolger G, Colicelli J, Ferguson K, Gerst J, Kataoka T, Michaeli T, Powers S, et al: Cloning and characterization of CAP, the S. cerevisiae gene encoding the 70 kd adenylyl cyclase-associated protein. Cell. 61:319–327. 1990. View Article : Google Scholar : PubMed/NCBI

10 

Gerst JE, Ferguson K, Vojtek A, Wigler M and Field J: CAP is a bifunctional component of the Saccharomyces cerevisiae adenylyl cyclase complex. Mol Cell Biol. 11:1248–1257. 1991. View Article : Google Scholar : PubMed/NCBI

11 

Mintzer KA and Field J: Interactions between adenylyl cyclase, CAP and RAS from Saccharomyces cerevisiae. Cell Signal. 6:681–694. 1994. View Article : Google Scholar : PubMed/NCBI

12 

Nishida Y, Shima F, Sen H, Tanaka Y, Yanagihara C, Yamawaki-Kataoka Y, Kariya K and Kataoka T: Coiled-coil interaction of N-terminal 36 residues of cyclase-associated protein with adenylyl cyclase is sufficient for its function in Saccharomyces cerevisiae ras pathway. J Biol Chem. 273:28019–28024. 1998. View Article : Google Scholar : PubMed/NCBI

13 

Freeman NL, Chen Z, Horenstein J, Weber A and Field J: An actin monomer binding activity localizes to the carboxyl-terminal half of the Saccharomyces cerevisiae cyclase-associated protein. J Biol Chem. 270:5680–5685. 1995. View Article : Google Scholar : PubMed/NCBI

14 

Liu Y, Cui X, Hu B, Lu C, Huang X, Cai J, He S, Lv L, Cong X, Liu G, et al: Upregulated expression of CAP1 is associated with tumor migration and metastasis in hepatocellular carcinoma. Pathol Res Pract. 210:169–175. 2014. View Article : Google Scholar

15 

Yu XF, Ni QC, Chen JP, Xu JF, Jiang Y, Yang SY, Ma J, Gu XL, Wang H and Wang YY: Knocking down the expression of adenylate cyclase-associated protein 1 inhibits the proliferation and migration of breast cancer cells. Exp Mol Pathol. 96:188–194. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Tan M, Song X, Zhang G, Peng A, Li X, Li M, Liu Y and Wang C: Overexpression of adenylate cyclase-associated protein 1 is associated with metastasis of lung cancer. Oncol Rep. 30:1639–1644. 2013.PubMed/NCBI

17 

Li M, Yang X, Shi H, Ren H, Chen X, Zhang S, Zhu J and Zhang J: Downregulated expression of the cyclase-associated protein 1 (CAP1) reduces migration in esophageal squamous cell carcinoma. Jpn J Clin Oncol. 43:856–864. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Hubberstey AV and Mottillo EP: Cyclase-associated proteins: CAPacity for linking signal transduction and actin polymerization. FASEB J. 16:487–499. 2002. View Article : Google Scholar : PubMed/NCBI

19 

Noegel AA, Blau-Wasser R, Sultana H, Müller R, Israel L, Schleicher M, Patel H and Weijer CJ: The cyclase-associated protein CAP as regulator of cell polarity and cAMP signaling in Dictyostelium. Mol Biol Cell. 15:934–945. 2004. View Article : Google Scholar :

20 

Vojtek A, Haarer B, Field J, Gerst J, Pollard TD, Brown S and Wigler M: Evidence for a functional link between profilin and CAP in the yeast S. cerevisiae. Cell. 66:497–505. 1991. View Article : Google Scholar : PubMed/NCBI

21 

Matviw H, Yu G and Young D: Identification of a human cDNA encoding a protein that is structurally and functionally related to the yeast adenylyl cyclase-associated CAP proteins. Mol Cell Biol. 12:5033–5040. 1992. View Article : Google Scholar : PubMed/NCBI

22 

Moriyama K and Yahara I: Human CAP1 is a key factor in the recycling of cofilin and actin for rapid actin turnover. J Cell Sci. 115:1591–1601. 2002.PubMed/NCBI

23 

Loisel TP, Boujemaa R, Pantaloni D and Carlier MF: Reconstitution of actin-based motility of Listeria and Shigella using pure proteins. Nature. 401:613–616. 1999. View Article : Google Scholar : PubMed/NCBI

24 

Yamazaki K, Takamura M, Masugi Y, Mori T, Du W, Hibi T, Hiraoka N, Ohta T, Ohki M, Hirohashi S, et al: Adenylate cyclase-associated protein 1 overexpressed in pancreatic cancers is involved in cancer cell motility. Lab Invest. 89:425–432. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Krek W, Ewen ME, Shirodkar S, Arany Z, Kaelin WG Jr and Livingston DM: Negative regulation of the growth-promoting transcription factor E2F-1 by a stably bound cyclin A-dependent protein kinase. Cell. 78:161–172. 1994. View Article : Google Scholar : PubMed/NCBI

26 

Lees E, Faha B, Dulic V, Reed SI and Harlow E: Cyclin E/cdk2 and cyclin A/cdk2 kinases associate with p107 and E2F in a temporally distinct manner. Genes Dev. 6:1874–1885. 1992. View Article : Google Scholar : PubMed/NCBI

27 

Wang J, Chenivesse X, Henglein B and Bréchot C: Hepatitis B virus integration in a cyclin A gene in a hepatocellular carcinoma. Nature. 343:555–557. 1990. View Article : Google Scholar : PubMed/NCBI

28 

Barlat I, Fesquet D, Bréchot C, Henglein B, Dupuy d'Angeac A, Vié A and Blanchard JM: Loss of the G1-S control of cyclin A expression during tumoral progression of Chinese hamster lung fibroblasts. Cell Growth Differ. 4:105–113. 1993.PubMed/NCBI

29 

Singh A and Settleman J: EMT, cancer stem cells and drug resistance: An emerging axis of evil in the war on cancer. Oncogene. 29:4741–4751. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Thiery JP: Epithelial-mesenchymal transitions in development and pathologies. Curr Opin Cell Biol. 15:740–746. 2003. View Article : Google Scholar : PubMed/NCBI

31 

Song Y, Washington MK and Crawford HC: Loss of FOXA1/2 is essential for the epithelial-to-mesenchymal transition in pancreatic cancer. Cancer Res. 70:2115–2125. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Huber MA, Kraut N and Beug H: Molecular requirements for epithelial-mesenchymal transition during tumor progression. Curr Opin Cell Biol. 17:548–558. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2016
Volume 36 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Fan Y, Cui C, Zhu Y, Zhang L, Shi M, Yu J, Bai J and Zheng J: Overexpression of CAP1 and its significance in tumor cell proliferation, migration and invasion in glioma. Oncol Rep 36: 1619-1625, 2016
APA
Fan, Y., Cui, C., Zhu, Y., Zhang, L., Shi, M., Yu, J. ... Zheng, J. (2016). Overexpression of CAP1 and its significance in tumor cell proliferation, migration and invasion in glioma. Oncology Reports, 36, 1619-1625. https://doi.org/10.3892/or.2016.4936
MLA
Fan, Y., Cui, C., Zhu, Y., Zhang, L., Shi, M., Yu, J., Bai, J., Zheng, J."Overexpression of CAP1 and its significance in tumor cell proliferation, migration and invasion in glioma". Oncology Reports 36.3 (2016): 1619-1625.
Chicago
Fan, Y., Cui, C., Zhu, Y., Zhang, L., Shi, M., Yu, J., Bai, J., Zheng, J."Overexpression of CAP1 and its significance in tumor cell proliferation, migration and invasion in glioma". Oncology Reports 36, no. 3 (2016): 1619-1625. https://doi.org/10.3892/or.2016.4936