Open Access

CAP1 is overexpressed in human epithelial ovarian cancer and promotes cell proliferation

  • Authors:
    • Minhui Hua
    • Sujuan Yan
    • Yan Deng
    • Qinghua Xi
    • Rong Liu
    • Shuyun Yang
    • Jian Liu
    • Chunhui Tang
    • Yingying Wang
    • Jianxin Zhong
  • View Affiliations

  • Published online on: February 4, 2015     https://doi.org/10.3892/ijmm.2015.2089
  • Pages: 941-949
  • Copyright: © Hua et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 3.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Adenylate cyclase-associated protein 1 (CAP1) regulates both actin filaments and the Ras/cAMP pathway in yeast, and has been found play a role in cell motility and in the development of certain types of cancer. In the present study, we investigated CAP1 gene expression in human epithelial ovarian cancer (EOC). Western blot analysis and immunohistochemistry were performed using EOC tissue samples and the results revealed that CAP1 expression increased with the increasing grade of EOC. In the normal ovarian tissue samples however, CAP1 expression was barely detected. Using Pearson's χ2 test, it was demonstrated that CAP1 expression was associated with the histological grade and Ki-67 expression. Kaplan-Meier analysis revealed that a higher CAP1 expression in patients with EOC was associated with a poorer prognosis. In in vitro experiments using HO-8910 EOC cells, the expression of CAP1 was knocked down using siRNA. The proliferation of the HO-8910 cells was then determined by cell cycle analysis and cell proliferation assay using the cell counting kit-8 and flow cytometry. The results revealed that the loss of CAP1 expression inhibited cell cycle progression. These findings suggest that a high expression of CAP1 is involved in the pathogenesis of EOC, and that the downregulation of CAP1 in tumor cells may be a therapeutic target for the treatment of patients with EOC.

Introduction

Epithelial ovarian cancer (EOC) is one of the most common types of ovarian tumors and is the leading cause of mortality among gynecological malignancies (1,2). Despite the rapid development of surgical and chemotherapy treatments, the 5-year survival rate for patients with EOC remains at approximately 30–50% due to the lack of effective early diagnostic methods. Thus, there is an urgent need for the identification of biological factors with sensitivity and specificity in order to improve prognosis and reduce the threat to the lives and health of women.

It is well known that cell physiology, including cell-cell junctions, cytoskeletal structure and cell morphology regulate cancer cell migration and invasion. The actin cytoskeleton plays a key role in cancer cell motility and metastasis, as its remodeling regulates a number of important cellular processes, such as cell adhesion, migration and morphological changes (35). Cyclase-associated proteins (CAPs) which exist in organisms from mammals to apicomplexan parasites (6) are a family of evolutionarily conserved proteins which are key to regulating actin dynamics and participate in Ras-mediated adenylyl cyclase activity (7,8). In mammal, cells have 2 CAP genes encoding the related CAP1 and CAP2 (9), both of which localize in the cell membrane and cytoplasm, and contain a C-CAP/co-factor C-like domain (1013). The adenylate CAP1 gene, which encodes an actin monomer-binding protein is thought to facilitate processes, such as the establishment of cell polarity and mRNA localization (14). It also has the ability to control the cytoskeleton (1519). The reorganization of the actin filament is regulated by actin-binding proteins in some signaling pathways that are essential for cell migration and several other intracellular events (20,21). CAP1 also plays an important role in actin filament turnover by effectively recycling cofilin and actin (22). On both ends of the actin filament, CAP1 rapidly translocates to the mitochondria upon treatment with agents that induce apoptosis (23). Studies have demonstrated that CAP1 is overexpressed in hepatocelluar carcinoma (24), breast cancer (25), lung cancer (26) and esophageal squamous cell carcinoma (27). However, to the best of our knowledge, no more information is available to date regarding the role of CAP1 in ovarian tumorigenesis.

Therefore, in the present study, we aimed to investigate the expression and function of CAP1 in EOC. We analyzed the expression of CAP1 protein in 119 EOC tissue specimens by immunohistochemistry and western blot analysis. We also determined the correlation of CAP1 expression with clinicopathological characteristics and evaluated the prognostic value of CAP1 in EOC by survival analysis. In addition, we knocked down CAP1 expression in order to explore the potential involvement of CAP1 in the regulation of cell proliferation. Our results indicate that CAP1 is a newly identified biomarker for EOC and may be used as a therapeutic target for the treatment of EOC in the future.

Materials and methods

Patients and tissue samples

All investigations described in this study were carried out after obtaining informed consent and in accordance with an Institutional Review Board protocol approved by the Partners Human Research Committee at the Affiliated Hospital of Nantong University, Nantong, China. A total of 119 human EOC tissue specimens (45 papillary serous adenocarcinoma, 16 papillary mucinous carcinoma, 14 endometrioid adenocarcinoma and 14 clear cell carcinoma specimens, as well as 30 specimens classified as ‘other’) were provided by the Department of Pathology, the Affiliated Hospital of Nantong University from 2004 to 2009. None of the patients had received chemotherapy or radiotherapy prior to surgery. A total of 10 tissue samples were obtained at the time of surgery and were immediately stored in liquid nitrogen and maintained at −80°C until used in western blot analysis, including 1 normal tissue sample from a woman who underwent hysterectomy for benign disease. The clinicopathological characteristics of all the participants are shown in Table I. According to the WHO system, the histological classification of tumors was graded as follows: well differentiated [grade 1 (G1); n=15], moderately differentiated [grade 2 (G2); n=33] and poorly differentiated [grade 3 (G3); n=71].

Table I

Expression of CAP1 in the 119 human ovarian cancer specimens.

Table I

Expression of CAP1 in the 119 human ovarian cancer specimens.

Clinicopathological characteristicsNo. of casesCAP1 expression
P-valuea
LowHigh
Age (years)0.775
 ≤50351619
 >50843648
FIGO stage, n (%)0.477
 I381820
 II311021
 III331518
 IV1798
Histological grade, n (%)0.010b
 115123
 2331320
 3712744
Histological subtype, n (%)0.071
 Serous452025
 Mucinous16106
 Endometrioid1477
 Clear cell1486
 Others30723
Menopause0.395
 Absent432122
 Present763145
Lymph node status, n (%)0.209
 Negative873552
 Positive321715
Ascites, n (%)0.666
 Absent663036
 Present532231
Malignant tumor cells in peritoneal fluid, n (%)0.472
 Absent904149
 Present291118
Metastases to other organs, n (%)0.666
 Absent663036
 Present532231
Ki-67<0.001b
 Low expression503713
 High expression691554

a Statistical analyses were performed using the Pearson’s χ2 test.

b P<0.05 indicates statistical significance. CAP1, cyclase-associated protein 1.

Antibodies

The antibodies used for western blot analysis and immunohistochemistry were as follows: mouse anti-CAP1 monoclonal antibody (SC-376286), mouse anti-human Ki-67 monoclonal antibody (SC-101861), mouse anti-human proliferating cell nuclear antigen (PCNA) monoclonal antibody (sc-56), rabbit anti-human cyclin A polyclonal antibody (sc-751) and rabbit anti-human glyceraldehyde 3-phosphate dehydrogenase (GAPDH) polyclonal antibody (sc-25778; all from Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA).

Western blot analysis

Total protein extracts (100 mg) were subjected to 10% sodium dodecyl sulfate-polyacrylamide gels electrophoresis (SDS-PAGE) and then transferred onto polyvinylidene difluoride (PVDF) filter membranes (Millipore, Bedford, MA, USA). After the membranes were blocked in 5% non-fat milk in TBST (150 mM NaCl, 20 mM Tris, 0.05% Tween-20) for 2 h, they were incubated with the primary antibodies overnight at 4°C. After washing the membranes with TBST 3 times for 5 min each, horseradish peroxidase-linked IgG secondary antibodies (donkey anti-mouse IgG-HRP, sc-2314; Santa Cruz Biotechnology, Inc.) were added followed by incubation for 2 h at room temperature. The membranes were developed using the ECL detection system.

Immunohistochemistry

Serial sections (5-μm-thick) were mounted on glass slides coated with 10% polylysine and were dewaxed in xylene and rehydrated in graded ethanol. Endogenous peroxidase activity was blocked by soaking in 0.3% hydrogen peroxide. The sections were then processed in 10 mmol/l citrate buffer (pH 6.0) and heated to 121°C in an autoclave for 20 min to retrieve the antigen. After rinsing in phosphate-buffered saline (PBS, pH 7.2), the sections were incubated with mouse anti-human CAP1 antibody (diluted 1:10,000) and mouse anti-human Ki-67 antibody (diluted 1:600) for 2 h at room temperature. Negative control slides were also processed in parallel using a non-specific immunoglobulin IgG (Santa Cruz Biotechnology, Inc.) at the same concentration as the primary antibody. All slides were processed using the peroxidase-anti-peroxidase method (Dako, Hamburg, Germany). Following rinsing with PBS, the peroxidase reaction was visualized by incubating the sections with the liquid mixture, DAB (0.1% phosphate buffer solution, 0.02% diaminobenzidine tetrahydrochloride and 3% H2O2). After rinsing in water, the sections were counterstained with hematoxylin, dehydrated and cover-slipped. All the immunostained sections were observed under a Leica fluorescence microscope (Leica Microsystems, Wetzlar, Germany).

For the assessment of CAP1 and Ki-67, at least 10 high-power fields in each specimen were randomly selected, and the percentage of cells with nuclear and cytoplasmic staining was examined with a total number of >500 cells counted to determine the labeling index in a single section. Tissues with no staining were rated as 0, with a faint staining or moderate to strong staining in ≤25% of cells as 1, with moderate staining or strong staining in 25–50% of cells as 2, and strong staining in ≥50% of cells as 3. For statistical analysis, a score of <2 was counted as low expression, while a score of ≥2 was counted as overexpression, as previously described (24). When evaluating Ki-67 protein immunoreaction, staining was scored in a semi-quantitative manner. A cut-off value of 50.7% or more positively stained nuclei in 5 high-power fields were used to identify Ki-67 staining as follows: the high expression group (≥50.7%) and low expression group (<50.7%). All the immunostained sections were evaluated in a blinded manner without knowledge of the clinicopathological variables of the patients. In half of the samples, staining was repeated twice to avoid technical errors, but similar results were obtained in these samples.

Cell culture

The human EOC cells, HO-8910, were purchased from the Shanghai Institute of Cell Biology and cultured in RPMI-1640 supplemented with 10% heat-inactivated fetal calf serum and 100 U/ml penicillin-streptomycin mixture (both from Gibco-BRL, Grand Island, NY, USA) at 37°C and 5% CO2.

Cell cycle analysis

Cells were harvested at the proper time and fixed in 70% ethanol overnight at 4°C and then incubated with 1 mg/ml RNase A for 30 min at 37°C. Subsequently, the cells were stained with propidium iodide (PI, 50 mg/ml; Becton-Dickinson, San Jose, CA, USA) in PBS, 0.5% Tween-20, and analyzed using a flow cytometer BD FACScan (Becton-Dickinson, San Jose, CA, USA) as well as CellQuest acquisition and analysis programs. As regards cell synchronization, serum deprivation was used for cell cycle G1-S phase arrest.

siRNA and transfection

Duplex siRNAs were synthesized as follows by Biomics Biotechnologies Co., Ltd. (Nantong, China): The sequences were as follows CAP1-specific, 5′-GAAAUGAA UGAUGCCAdTdT-3′ and control, 5′-UGGCGGCAUUCAUU UCdTdT-3′. The HO-8910 cells were seeded the day prior to transfection at a confluence of 50–70%. In addition, a Mock group with no transfection was also used as a control. The transient transfection of CAP1 and non-specific siRNA oligos was carried out using Lipofectamine 2000 (Invitrogen, St. Louis, MO, USA) in accordance with the manufacturer’s instructions. The cells were harvested 48 h after transfection and used for the experiment. The experiments were repeated at least 3 times.

Cell proliferation assay

The cell counting kit-8 (CCK-8) was used to measure cell proliferation according to the manufacturer’s instructions (Dojindo, Kumamoto, Japan). The cells were seeded into a 96-well cell culture cluster plates (Corning, Inc., Corning, NY, USA) at a concentration of 2×104 cells/well in a volume of 100 μl culture medium and grown overnight. CCK-8 reagents (Dojindo) were then added to each well followed by incubation for an additional 2 h at 37°C. The absorbency was measured at a test wavelength of 490 nm and a reference wavelength of 630 nm using a microplate reader (Bio-Rad, Hercules, CA, USA). The experiments were repeated at least 3 times.

Statistical analysis

All statistical analyses were performed using SPSS 17.0 statistical software. The association between CAP1 and Ki-67 expression and clinicopathological characteristics was analyzed using Pearson’s χ2 test. Multivariate analysis was performed using Cox’s proportional hazards model. CAP1 and Ki-67 expression was quantified using Pearson’s correlation co-efficient. Kaplan-Meier analysis was used to evaluate the survival curve, and the log-rank test was performed for analysis.

Results

CAP1 is overexpressed in EOC tissues

To confirm the role of CAP1 in EOC, western blot analysis for CAP1 was performed using surgical specimens. We found that CAP1 protein expression gradually increased in 9 EOC tissues from 3 tumors classified as G1 to G3 (3 tissues from each tumor) in comparison with 1 normal ovarian tissue sample in which CAP1 expression was barely detected (Fig. 1). In order to determine the role of CAP1 in the progression and development of EOC, we examined the intracellular expression of CAP1 and Ki-67 in 119 specimens of EOC by immunohistochemical analysis. We found that CAP1 was mainly located in the cytoplasm of the EOC cells (Fig. 2). The high expression of CAP1 was accompanied by the high expression of Ki-67 localized in the nucleus. In addition, CAP1 was highly expressed in the poorly differentiated tumor specimens compared to the well differentiated ones; similar results were obtained for Ki-67 expression.

Correlation of CAP1 expression with clinicopathologic characteristics in EOC

To clarify the clinicopathological significance of CAP1, we analyzed the correlation of CAP1 expression with the clinicopathologic characteristics of patients with EOC (Table I). We divided the tumor specimens into 2 groups according to the expression of CAP1. CAP1 expression was significantly associated with the histological grade (P=0.010) and Ki-67 expression (P<0.001), whereas there was no correlation observed with the FIGO stage (P=0.477), menopause (P=0.395), ascites (P=0.666), lymph node status (P=0.209), malignant tumor cells (P=0.472), metastases to other organs (P=0.666), age (P=0.775) and histologic subtype (P=0.071). In addition, Pearson’s correlation co-efficient revealed that there was a positive correlation between CAP1 expression and Ki-67 expression (r=0.722, P<0.001) (Fig. 3).

Expression of CAP1 and prognosis of EOC

Kaplan-Meier analysis revealed that out of the 119 clinical cases examined, the patients with a high expression of CAP1 had a poorer overall survival than those with a lower expression (P<0.001; Fig. 4). Pearson’s χ2 test indicated that the histological grade (P=0.047), CAP1 (P<0.001) and Ki-67 (P<0.001) were independent prognostic indicators of EOC (Table II). Moreover, multivariate analysis using Cox’s proportional hazards model showed that the histological grade (P<0.001), CAP1 (P= 0.029) and Ki-67 (P<0.001) were independent prognostic indicators for patient overall survival (Table III). The survival curve indicated that the median survival for patients with a high CAP1 expression was significantly shorter than the median survival for those patients with a low CAP1 expression (Fig. 4).

Table II

Survival status and clinicopathological characteristics in the 119 human ovarian cancer specimens.

Table II

Survival status and clinicopathological characteristics in the 119 human ovarian cancer specimens.

Clinicopathological characteristicsTotalSurvival status
P-valuea
AliveDeceased
Age (years)0.099
 ≤50351520
 >50842361
FIGO stage, n (%)0.655
 I381523
 II31823
 III331023
 IV17512
Histological grade, n (%)0.047b
 115510
 233528
 3712843
Histological subtype, n (%)0.229
 Serous451926
 Mucinous16313
 Endometrioid1459
 Clear cell1459
 Others30624
Menopause0.081
 Absent431825
 Present762056
Lymph node status, n (%)0.217
 Negative872562
 Positive321319
Ascites, n (%)0.051
 Absent662640
 Present531241
Malignant tumor cells in peritoneal fluid, n (%)0.135
 Absent903258
 Present29623
Metastases to other organs, n (%)0.051
 Absent662640
 Present531241
CAP1<0.001b
 Low expression522824
 High expression671057
Ki-67<0.001b
 Low expression503416
 High expression69465

a Statistical analyses were performed using the Pearson’s χ2 test.

b P<0.05 indicates statistical significance. CAP1, cyclase-associated protein 1.

Table III

Contribution of various potential prognostic factors to survival by Cox regression analysis of the 119 human ovarian cancer specimens.

Table III

Contribution of various potential prognostic factors to survival by Cox regression analysis of the 119 human ovarian cancer specimens.

Hazard ratioP-value95.0% CI
Histological grade0.552<0.001a0.396–0.770
CAP1 expression0.5480.029a0.319–0.941
Ki-67 expression6.161<0.001a3.256–11.655

a P<0.05 indicates statistical significance. CI, confidence interval; CAP1, cyclase-associated protein 1.

CAP1 expression and cell cycle progression

It has been reported that the knockdown of the expression of CAP1 affects the breast cancer cell cycle, inhibiting the growth of breast cancer cancer (25). In this study, we found that a high CAP1 expression associated with a poor prognosis of patients with EOC. Thus, we hypothesized that CAP1 participates in the cell cycle of HO-8910 cells. We subjected the cells to serum starvation and serum re-feeding and found that the expression of CAP1 gradually increased, as well as that of the proliferation markers, PCNA and cyclin A, during cell progression. Flow cytometric analysis revealed that the HO-8910 cells subjected to serum deprivation for 48 h were arrested in the G1 phase, and following serum re-feeding, the population of HO-8910 cells in the S phase increased from 18.08 to 61.12% (Fig. 5C). To further validate these results, we collected HO-8910 cellular protein at different time points, and western blot analysis was performed to determine whether CAP1 expression is cell cycle-dependent in HO-8910 cells. We found that CAP1 expression was significantly increased as early as 4 h following serum re-feeding in the HO-8910 cells. The expression of the cell proliferation markers, PCNA and cyclin A, showed a similar tendency (Fig. 5A and B).

Knockdown of CAP1 inhibits the proliferation of HO-8910 cells

To further determine the effect of CAP1 on cell proliferation, we knocked down endogenous CAP1 in HO-8910 cells using siRNA targeting CAP1 and the effects to the cells transfected with control siRNA and mock siRNA (Fig. 6A and B). The knockdown of CAP1 using siRNA inhibited the accumulation of cyclin A and PCNA compared with control- and mock-transfected cells (Fig. 6A and B). Additionally, we found that the proliferation rate of the HO-8910 cells transfected with siRNA exhibited a decrease compared with the cells transfected with the control siRNA and the mock-transfected cells (Fig. 6D). From these results, we ascertained that CAP1 plays a positive role in the regulation of cell proliferation.

Discussion

CAP1, a member of the CAP family in mammalian cells, was first identified as a component of the yeast adenylyl cyclase complex and conserved in all eukaryotic organisms. The CAP family contains 4 highly conserved protein domains, one of which is the N-terminal which interacts with adenylyl cyclase and induces the activity of RAS following exogenous signals, whereas the C-terminal half of CAPs is involved in the cell differentiation and depolymerization of the F-actin filamentactin (28). As a monomeric actin binding protein, CAP is involved in cell polarization, the distribution of actin filaments and mRNA in a Dictyostelium (29). The expression of CAP is associated with an abnormally large cell size, random budding pattern and an abnormal actin distribution in yeast (30,31).

A number of scholars have started to investigate the association between CAP1 and cancer. CAP1 has been to be commonly overexpressed in pancreatic cancers, and its level in clinical cases has been shown to be associated with neuronal invasion and lymph node metastasis. The knockdown CAP1 has been shown to reduce cell motility and migration (32). It has been reported that CAP1 is upregulated in breast cancer. After knocking down its expression, the proliferation and migration of MDA-MB-231 cells was shown to decrease, inducing changes in morphology, which were associated with the arrangement of F-actin (25). Western blot analysis, real-time PCR and immunohistochemical analysis have been used to prove that CAP1 is overexpressed in hepatocellular carcinoma compared with adjacent non-cancerous liver tissues, and that it is positively associated with HCC cell metastasis (24). CAP1 overexpression has been shown to be significantly associated with lymph node status in esophageal squamous cell carcinoma. The knockdown of CAP1 in TE1 cells has been shown to result in a decreased migration capability and the overexpression of CAP1 promotes TE1 cell migration (27). In this study, we found that CAP1 was overexpressed in EOC. Using immunohistochemistry, we found that CAP1 was highly expressed in poorly differentiated specimens compared to well differentiated ones, and similar results were obtained for Ki-67 expression. Kaplan-Meier analysis revealed that out of the 119 clinical cases, the patients with a high expression of CAP1 had a poorer overall survival than those with a lower CAP1 expression. In addition, the knockdown of CAP1 expression in an in vitro experiment revealed that the loss of CAP1 inhibit the proliferation of HO-8910 cells.

It has been demonstrated that the morphologic changes of malignant tumor cells enhance the migration capacity of the cells and lead to invasion and metastasis (33). To a certain degree, the maintainance and changes in the structure and function of cells are achieved by regulating the structure and function of the actin cytoskeleton, which is the key to the reorganization of the actin cytoskeleton. When stimulated, CAP regulates the polymerization and disassembly of downstream actin protein, thus affecting the growth and differentiation of cells. CAP1 also takes part in accelerating the turnover of actin filaments by the recycling of cofilin and actin on both ends of the actin filament (34). Therefore, we hyopthesized that the molecular mechanisms of action of CAP1 in the pathogenesis of EOC may involve its downstream actin protein. However, further studies are required to identify the precise signaling pathways involved.

As CAP1 is overexpressed in EOC, it may thus serve as a prognostic marker for EOC. Using western blot analysis, we found that the expression of CAP1 was higher in the 9 EOC tissues compared to the 1 normal tissue, in which CAP1 expression was barely detected. Furthermore, immunohistochemistry of the 119 paraffin-embedded tissue sections of EOC also revealed that CAP1 immunostaining was located in the cytoplasm and was upregulated in the poorly differentiated tumor cells compared to the well differentiated ones. These results were similar to those obtained for Ki-67 expression, highlighting that CAP1 expression significantly correlated with the proliferation of EOC cells. The association between CAP1 and markers of the cell cycle was then detected by a starvation-release experiment.

In conclusion, this study demonstrates that CAP1 is an independent prognostic factor in EOC. Our data suggest that CAP1 plays an important role in cell proliferation in EOC and that CAP1 may be a potential therapeutic target in EOC chemotherapy.

Acknowledgments

This study was supported by a grant from the National Natural Science Foundation of China (no. 81302285).

References

1 

Menczer J, Chetrit A and Sadetzki S; National Israel Ovarian Cancer Group: Uterine metastases in ovarian carcinoma: frequency and survival in women who underwent hysterectomy. J Gynecol Oncol. 21:191–195. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Park B, Park S, Kim TJ, et al: Epidemiological characteristics of ovarian cancer in Korea. J Gynecol Oncol. 21:241–247. 2010. View Article : Google Scholar

3 

Chang E, Heo KS, Woo CH, et al: 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 :

4 

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

5 

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

6 

Makkonen M, Bertling E, Chebotareva NA, Baum J and Lappalainen P: Mammalian and malaria parasite cyclase-associated proteins catalyze nucleotide exchange on G-actin through a conserved mechanism. J Biol Chem. 288:984–994. 2013. View Article : Google Scholar :

7 

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

8 

Field J, Vojtek A, Ballester R, 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

9 

Peche V, Shekar S, Leichter M, et al: CAP2, cyclase-associated protein 2, is a dual compartment protein. Cell Mol Life Sci. 64:2702–2715. 2007. View Article : Google Scholar : PubMed/NCBI

10 

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.PubMed/NCBI

11 

Zelicof A, Gatica J and Gerst JE: Molecular cloning and characterization of a rat homolog of CAP, the adenylyl cyclase-associated protein from Saccharomyces cerevisiae. J Biol Chem. 268:13448–13453. 1993.PubMed/NCBI

12 

Yu G, Swiston J and Young D: Comparison of human CAP and CAP2, homologs of the yeast adenylyl cyclase-associated proteins. J Cell Sci. 107:1671–1678. 1994.PubMed/NCBI

13 

Swiston J, Hubberstey A, Yu G and Young D: Differential expression of CAP and CAP2 in adult rat tissues. Gene. 165:273–277. 1995. View Article : Google Scholar : PubMed/NCBI

14 

Pollard TD and Cooper JA: Actin, a central player in cell shape and movement. Science. 326:1208–1212. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Baum B, Li W and Perrimon N: A cyclase-associated protein regulates actin and cell polarity during Drosophila oogenesis and in yeast. Curr Biol. 10:964–973. 2000. View Article : Google Scholar : PubMed/NCBI

16 

Baum B and Perrimon N: Spatial control of the actin cytoskeleton in Drosophila epithelial cells. Nat Cell Biol. 3:883–890. 2001. View Article : Google Scholar : PubMed/NCBI

17 

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

18 

Kang CM and Jiang YW: Genome-wide survey of non-essential genes required for slowed DNA synthesis-induced filamentous growth in yeast. Yeast. 22:79–90. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Yusof AM, Hu NJ, Wlodawer A and Hofmann A: Structural evidence for variable oligomerization of the N-terminal domain of cyclase-associated protein (CAP). Proteins. 58:255–262. 2005. View Article : Google Scholar

20 

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

21 

Pollard TD, Blanchoin L and Mullins RD: Molecular mechanisms controlling actin filament dynamics in nonmuscle cells. Annu Rev Biophys Biomol Struct. 29:545–576. 2000. View Article : Google Scholar : PubMed/NCBI

22 

Bertling E, Hotulainen P, Mattila PK, Matilainen T, Salminen M and Lappalainen P: Cyclase-associated protein 1 (CAP1) promotes cofilin-induced actin dynamics in mammalian nonmuscle cells. Mol Biol Cell. 15:2324–2334. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Wang C, Zhou GL, Vedantam S, Li P and Field J: Mitochondrial shuttling of CAP1 promotes actin- and cofilin-dependent apoptosis. J Cell Sci. 121:2913–2920. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Liu Y, Cui X, Hu B, 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

25 

Yu XF, Ni QC, Chen JP, et al: 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

26 

Tan M, Song X, Zhang G, et al: Overexpression of adenylate cyclase-associated protein 1 is associated with metastasis of lung cancer. Oncol Rep. 30:1639–1644. 2013.PubMed/NCBI

27 

Li M, Yang X, Shi H, et al: 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

28 

Zelicof A, Protopopov V, David D, Lin XY, Lustgarten V and Gerst JE: Two separate functions are encoded by the carboxyl-terminal domains of the yeast cyclase-associated protein and its mammalian homologs. Dimerization and actin binding. J Biol Chem. 271:18243–18252. 1996. View Article : Google Scholar : PubMed/NCBI

29 

Noegel AA, Blau-Wasser R, Sultana H, et al: 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 :

30 

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.PubMed/NCBI

31 

Vojtek A, Haarer B, Field J, et al: 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

32 

Yamazaki K, Takamura M, Masugi Y, 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

33 

Friedl P and Wolf K: Tumour-cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer. 3:362–374. 2003. View Article : Google Scholar : PubMed/NCBI

34 

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

Related Articles

Journal Cover

April-2015
Volume 35 Issue 4

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Hua M, Yan S, Deng Y, Xi Q, Liu R, Yang S, Liu J, Tang C, Wang Y, Zhong J, Zhong J, et al: CAP1 is overexpressed in human epithelial ovarian cancer and promotes cell proliferation. Int J Mol Med 35: 941-949, 2015
APA
Hua, M., Yan, S., Deng, Y., Xi, Q., Liu, R., Yang, S. ... Zhong, J. (2015). CAP1 is overexpressed in human epithelial ovarian cancer and promotes cell proliferation. International Journal of Molecular Medicine, 35, 941-949. https://doi.org/10.3892/ijmm.2015.2089
MLA
Hua, M., Yan, S., Deng, Y., Xi, Q., Liu, R., Yang, S., Liu, J., Tang, C., Wang, Y., Zhong, J."CAP1 is overexpressed in human epithelial ovarian cancer and promotes cell proliferation". International Journal of Molecular Medicine 35.4 (2015): 941-949.
Chicago
Hua, M., Yan, S., Deng, Y., Xi, Q., Liu, R., Yang, S., Liu, J., Tang, C., Wang, Y., Zhong, J."CAP1 is overexpressed in human epithelial ovarian cancer and promotes cell proliferation". International Journal of Molecular Medicine 35, no. 4 (2015): 941-949. https://doi.org/10.3892/ijmm.2015.2089