Overexpression of RhoGDI2 correlates with the progression and prognosis of pancreatic carcinoma

  • Authors:
    • Bin Yi
    • Yi Zhang
    • Dongming Zhu
    • Lifeng Zhang
    • Shiduo Song
    • Songbing He
    • Bing Zhang
    • Dechun Li
    • Jian Zhou
  • View Affiliations

  • Published online on: January 7, 2015     https://doi.org/10.3892/or.2015.3707
  • Pages: 1201-1206
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Rho GDP dissociation inhibitor 2 (RhoGDI2) has been found to be a regulator of tumor metastasis. However, the expression of RhoGDI2 and its clinicopathological significance as well as the pathway of RhoGDI2 in tumor metastasis have yet to be investigated. To investigate the role of RhoGDI2 in the progression and prognosis of pancreatic carcinoma (PC), the expression of RhoGDI2 in human PC tissues was examined and compared with the clinicopathological characteristics and prognosis. Moreover, the relationship between RhoGDI2 and E-cadherin was examined. The results indicated that RhoGDI2 was overexpressed in PC tissues and associated with clinicopathological characteristics, including clinical stage and lymph node metastasis. Patients with a RhoGDI2‑negative expression had a significantly longer survival time than those with a RhoGDI2‑positive expression. Additionally, the expression of RhoGDI2 was negatively correlated with the expression of E-cadherin in PC tissues. Taken together, the findings suggest that RhoGDI2 is important in the progression and prognosis of PC, and may be used as a potential prognostic biomarker and a therapeutic target for PC.

Introduction

Human pancreatic carcinoma (PC) is a highly aggressive malignant cancer with a poor prognosis. Numerous treatment protocols have been applied to PC, however, the 5-year survival rate remains <5%, partly due to PC cells being resistant to chemotherapy and radiation (1,2). Vascular invasion and distant metastasis are the critical features in the aggressive phenotype of PC, and contribute to the principal causes of PC deaths. Thus, biomarkers associated with the invasion and metastasis and survival of PC are required to predict patient prognosis and to aid in the design of effective target therapy.

Rho GTPases, including Rac1, Cdc42 and RhoC, are involved in the regulation of cell migration, cell motility, cell cycle progression and cytoskeleton organization (3,4). Aberrant signaling of these proteins is commonly observed in many types of human cancer and is associated with aggressive phenotype. The biological activities of GTPases are regulated by guanine nucleotide exchange factors (GEFs), GTPase-activating proteins (GAPs) and Rho GDP dissociation inhibitors (RhoGDIs) (5). Rho GDP dissociation inhibitor 2 (RhoGDI2), which belongs to a family of RhoGDIs, is verified to be differentially expressed in human cancers (6,7). Accumulating evidence has shown that RhoGDI2 acts as a positive or negative regulator of cancer progression depending on the tumor type (8). In a previous study, we showed that RhoGDI2 promoted PC cell invasion and metastasis in vitro (9). However, the expression of RhoGDI2 and its correlation with poor prognosis in PC patients as well as the pathway of RhoGDI2 in tumor metastasis remain to be examined.

Epithelial to mesenchymal transition (EMT) is a critical morphologic conversion during tumor progression and results in the promotion of cell motility, invasion and metastasis (10). Increasing evidence suggests that EMT occurs in several types of cancer, including colorectal cancer (11), gastric cancer (12), and breast cancer (13). Loss of expression of the epithelial cell adhesion molecule E-cadherin is a prerequisite of EMT.

In this study, we examined the expression of RhoGDI2 in human PC tissues, and compared it with the clinicopathological characteristics and prognosis. Moreover, we investigated the association between RhoGDI2 and E-cadherin, a critical factor of EMT, and determined the possible pathway that RhoGDI2 may be involved in the aggressive phenotype of PC.

Materials and methods

Clinical samples

Tissue samples were collected from 77 PC patients during surgical resections performed at the First Affiliated Hospital of Soochow University between January, 2008 and December, 2010. Tumorous tissues and adjacent non-tumorous tissues (NT) were frozen immediately after surgical removal in liquid nitrogen and stored at −80°C. The patients had not received any preoperative chemo-, radio- or immunotherapy. Grades of differentiation and clinical stage were classified according to the World Health Organization. All the samples were obtained following patient consent and approval by the Ethics Committee of Soochow University.

Immunohistochemistry (IHC)

The samples were fixed with formalin, embedded in paraffin and sliced. Serial sections (4 μm) subjected to immunohistological staining were fixed with freshly prepared 3% H2O2 with 0.1% sodium azide to quench endogenous peroxidase and then treated with antigen retrieval solution for 15 min. After placing in blocking reagent for 15 min, the sections were incubated in primary anti-RhoGDI2 or anti-E-cadherin monoclonal antibody overnight at 4°C, followed by incubation with the secondary antibody and Extravidin-conjugated horseradish peroxidase. The staining intensity was scored as: 0, negative; 1, weak; 2, medium and 3, strong. The extent of staining was scored as: 0, 0%; 1, 1–25%; 2, 26–50%; 3, 51–75% and 4 >76%. The final score was obtained by the sum of the intensity score and the quantity score. A score of ≥3 was considered as positive expression, while a score of ≥6 was considered as strong-positive expression.

RT-PCR

Total RNA from samples was extracted by TRIzol (Invitrogen, Carlsbad, CA, USA). Total RNA (10 μg) was used to synthesize single-stranded cDNA for a PCR template by reacting with random primers and M-MLV reverse transcriptase (Promega, Madison, WI, USA). The relative expression of RhoGDI2 mRNA transcripts to that of the control (β-actin) was determined by RT-PCR. The primers used were: RhoGDI2 (606 bp) forward, 5′-ATGACTGAAAAAGCC CCA-3′ and reverse, 5′-TCATTCTGTCCACTCCTT-3′; β-actin (308 bp) forward, 5′-AGCGGGAAATCGTGCGTG-3′ and reverse, 5′-CAGGGTACATGGTGGTGCTGCC-3′. The PCR amplification was 40 cycles (95°C for 15 sec, 62°C for 45 sec and 72°C for 30 sec). The amplified segments were analyzed by 2.5% agarose gels.

Western blotting

Tissues were lysed in lysis buffer on ice. Total proteins were separated by 5–12% SDS-PAGE and transferred onto PVDF membrane. The membrane as placed in a TBST solution with 5% non-fat milk powder for 1 h at room temperature and incubated at 4°C overnight with primary antibodies: anti-RhoGDI2 antibody (1:200) ), anti-E-cadherin antibody (1:400; both from Abcam, Cambridge, UK) and anti-β-actin antibody (1:200), followed by incubation at room temperature for 1 h with a goat anti-mouse IgG (1:2,000, both from Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA), conjugated with horseradish peroxidase. Reactive bands were detected using ECL western blotting detection reagent.

Statistical analysis

SPSS version 17.0 was used for statistical analysis. Data were presented as mean ± SD. The t-test and Chi-square test were performed for inter-group comparison. The correlation between RhoGDI2 and E-cadherin expression was determined by the Pearson correlation analysis. Survival was assessed according to the Kaplan-Meier method and compared using the log-rank test. Multivariate analysis of prognostic markers was performed with the Cox proportional hazards regression model. P<0.05 was considered to indicate a statistically significant difference.

Results

Expression of RhoGDI2 in PC tissues

To investigate the expression pattern of RhoGDI2 in clinical fresh PC tissues, RT-PCR and western blotting were used in 20 paired PC tissues and adjacent non-tumorous tissues. The expression of RhoGDI2 in PC tissues was higher than that in non-tumorous tissues at the mRNA level (P<0.05) (Fig. 1). Similarly, western blotting results revealed that the expression of RhoGDI2 protein was upregulated in PC tissues compared with that in non-tumorous tissues (Fig. 2, P<0.05). These results indicated that RhoGDI2 was overexpressed in PC tissues.

Correlation between RhoGDI2 expression and clinicopathological parameters

To elucidate the role of RhoGDI2 in the progression of PC, we detected the expression of RhoGDI2 protein in PC tissues by IHC staining. The subcellular location of RhoGDI2 protein was observed mainly in the cytoplasm of cancer cells in PC tissues (Fig. 2). Among 77 PC tissues, 49 cases (63.6%) exhibited a positive expression of RhoGDI2, including 31 strong-positive cases (40.3%) in tumor tissues. Among the non-tumorous tissues, there were 66 RhoGDI2-negative expression (85.7%) and 11 weak-positive expression (14.3%) cases, showing a significant difference (χ2=39.428, P=0.001). The association between RhoGDI2 expression and clinicopathological parameters showed that RhoGDI2 expression was significantly correlated with clinical stage (χ2=19.983, P=0.008) and lymph-node metastasis (χ2=16.418, P=0.013), but did not show a statistically significant association with gender, age, tumor location, tumor size and differentiation (P>0.05, Table I) . These results indicated that the overexpression of RhoGDI2 may be correlated with the progression of PC.

Table I

Relationship between RhoGDI2 expression and clinicopathological characteristics in PC.

Table I

Relationship between RhoGDI2 expression and clinicopathological characteristics in PC.

RhoGDI2

VariablesCasesNegativePositiveχ2P-value
Gender0.5060.625
 Male481632
 Female291217
Age (years)0.4300.632
 ≤65451530
 >65321319
Tumor location0.0750.807
 Head511833
 Body and tail261016
Tumor size (cm)0.2290.641
 ≤2331320
 >2441529
Differentiation0.0030.999
 Well22814
 Moderate25916
 Poor301119
Clinical stage19.9830.008a
 I23176
 II541143
Lymph node metastasis16.4180.013a
 Yes40634
 No372215

a P<0.01.

Correlation between RhoGDI2 expression and PC patient prognosis

Among 77 PC patients, the follow-up success rate was 100%. After 3 years of follow up, only 8 of 77 (10.4%) patients were alive and 69 patients (89.6%) were deceased. The median survival time was 20 months (RhoGDI2-negative expression) and 11 months (RhoGDI2-positive expression), respectively. Kaplan-Meier curve assessment showed that the patients with RhoGDI2-negative expression had a significantly longer survival time than those with a RhoGDI2-positive expression (log-rank test, P=0.001, Fig. 3).

The univariate analysis results revealed that RhoGDI2 expression (P=0.003), clinical stage (P=0.007) and lymph-node metastasis (P=0.006) were closely correlated with patient survival time (Table II). RhoGDI2 expression was closely associated with clinical stage and lymph-node metastasis. Thus, we used RhoGDI2 expression as the grouping variable, while clinical stage and lymph-node metastasis were considered the subgrouping variables. Stratified analysis showed that survival time of the RhoGDI2-positive expression group was significantly shorter than that of the RhoGDI2-negative expression group in the different subgroup levels (P<0.01, Table III). The multivariate analysis results revealed that RhoGDI2 expression is one of the independent prognostic factors by Cox proportional hazards model (P=0.008, Table IV). The results indicated that the overexpression of RhoGDI2 was correlated with poor prognosis.

Table II

Univariate analysis of survival time of PC patients (Kaplan-Meier).

Table II

Univariate analysis of survival time of PC patients (Kaplan-Meier).

VariablesCasesAverage survival period (months)P-value
Gender0.887
 Male4814.4±1.2
 Female2914.9±1.5
Age (years)0.495
 ≤654514.2±1.1
 >653215.2±1.6
Tumor location0.379
 Head5115.1±1.2
 Body and tail2613.6±1.4
Tumor size (cm)0.131
 ≤23313.0±1.3
 >24415.8±1.3
Differentiation0.178
 Well2212.5±1.5
 Moderate2514.2±1.5
 Poor3016.4±1.7
Clinical stage0.007a
 I2320.7±1.9
 II5412.0±0.8
Lymph node metastasis0.006a
 Yes4011.1±1.0
 No3718.4±1.4
RhoGDI2 expression0.003a
 Positive4911.1±0.8
 Negative2820.7±1.6

a P<0.01.

Table III

Stratified analysis of related prognostic markers of PC patients.

Table III

Stratified analysis of related prognostic markers of PC patients.

Clinical stageLymph-node metastasis


GroupCasesIIINoYes
 RhoGDI2 positive4916.8±5.310.7±0.815.8±2.39.9±0.8
 RhoGDI2 negative2822.1±1.817.3±2.320.2±1.717.7±3.4
P-value0.6960.003a0.1640.005a

a P<0.01.

Table IV

Multivariate analysis of prognostic markers in PC patients.

Table IV

Multivariate analysis of prognostic markers in PC patients.

VariablesHR95% CIP-value
Gender0.9660.591–1.5810.892
Age0.9560.585–1.5620.858
Tumor location0.6770.404–1.1360.140
Tumor size2.1701.274–3.6970.004
Differentiation1.1740.680–2.0280.565
Clinical stage0.4910.254–0.9480.034a
Lymph-node metastasis1.8891.085–3.2880.025a
RhoGDI2 expression3.3441.718–6.5050.008a

a P<0.05.

RhoGDI2 expression correlated with E-cadherin expression in PC tissues

E-cadherin is involved in epithelial to mesenchymal transition (EMT), which is involved in invasion and metastasis in PC. To clarify the association between RhoGDI2 and E-cadherin, we firstly examined the expression of E-cadherin protein in 77 PC tissues by IHC (Fig 4). Data of the statistical analysis suggested that the expression of RhoGDI2 was negatively correlated with the expression of E-cadherin in PC tissues (P=0.002, Table V).

Table V

Correlation between RhoGDI2 and E-cadherin expression in PC.

Table V

Correlation between RhoGDI2 and E-cadherin expression in PC.

E-cadherin expressionRhoGDI2 expressionrP-value

Positive (n)Negative (n)
Positive (n)621
Negative (n)437−0.6330.002a

a P<0.01.

Discussion

In this study, we examined the expression of RhoGDI2 in 30 matched clinical fresh tissues and 77 cases of paraffin-embedded PC tissues. The results show that RhoGDI2 was overexpressed in PC tissues at mRNA and protein levels, and that RhoGDI2 expression was correlated with clinical stage, lymph-node metastasis and vascular invasion. Additionally, RhoGDI2 was one of the independent prognostic factors. We also found that the expression of RhoGDI2 was negatively correlated with the expression of E-cadherin in PC tissues. These findings suggest that the upregulation of RhoGDI2 is involved in the progression and prognosis of PC.

RhoGDI2, also known as D4-GDI or LyGDI, has been identified as a regulator of Rho GTPases, which play important roles in cell motility, invasion and metastasis (14, 15). RhoGDI2 was preferentially expressed in hematopoietic tissues, predominantly in B and T lymphocytes (16). However, accumulating evidence reveals that RhoGDI2 is also aberrantly expressed in human cancers. In the majority of studies, RhoGDI2 has been shown to promote tumor cell invasion, angiogenesis and metastasis, such as in lung and gastric cancer (17,18). However, it can function as a metastasis-suppressor gene in bladder cancer and Hodgkin’s lymphoma (19,20). Our results indicate that RhoGDI2 was overexpressed in PC and associated with clinicopathological characteristics of PC patients, including clinical stage and lymph-node metastasis. The conflicting role of RhoGDI2 may result from the dual roles of RhoGDI2 in the regulation of activities of Rho GTPases during cancer progression. RhoGDI2 binds the majority of Rho GTPases in the cytoplasm, maintaining Rho in an inactive form and inducing the disruption of Rho-dependent cell motility (21,22). On the other hand, RhoGDI2 acted as an escort protein directing Rho GTPases to the membrane and is associated with active forms of Rho, Rac and Cdc42, maintaining them in an active form (23,24). However, the exact mechanisms remain to be determined.

In this study, we have demonstrated that RhoGDI2 expression is one of the independent prognostic factors in PC, and overexpression of RhoGDI2 was correlated with poor prognosis. Stratified analysis of survival time showed that in lymph-node positive patients, the prognosis of PC with RhoGDI2-positive expression was worse than that of ones with RhoGDI2-negative expression. Similar results were obtained in stage II of PC patients with different RhoGDI2 expression. This finding indicated that, for PC patients with lymph-node metastasis and clinical stage II, we may draw up individualized gene therapy and evaluate prognosis by detecting RhoGDI2 expression.

EMT is an essential cell mechanism during tumor progression, which induces tumor cell migration, invasion and metastasis (25). In all EMT processes, cells lose the expression of a cell-to-cell adhesion molecule known as E-cadherin, which functions as a molecular glue that attaches cells to one another (26). To clarify the underlying mechanism of RhoGDI2 in the progression of tumor invasion and metastasis, we also investigated the relationship between and in PC tissues. Our results (data not shown) indicated E-cadherin was down-regulated in PC tissues and was negatively correlated with the expression of RhoGDI2. In our previous study, RhoGDI2 was known to promote PC cell invasion and migration in vitro, but to the best of our knowledge, this is the first study showing that RhoGDI2 expression was correlated with E-cadherin expression in PC tissues.

In conclusion, our study has demonstrated that the overexpression of RhoGDI2 was associated with PC progression and played an important role in predicting the prognosis of PC patients. Moreover, upregulation of RhoGDI2 was associated with reversal of E-cadherin expression in PC tissues. These findings indicate that targeting RhoGDI2 may be a useful strategy for inhibiting the invasion and metastasis of PC.

Acknowledgements

This study was supported by the Project of Nature Science Foundation of China (81201905), Nature Science Research Grants at the University of Jiangsu Province of P.R. China (14KJB320019) as well as the Project of Medical Research of Jiangsu Province (Q201402).

References

1 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2013. CA Cancer J Clin. 63:11–30. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Vincent A, Herman J, Schulick R, Hruban RH and Goggins M: Pancreatic cancer. Lancet. 378:607–620. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Vega FM and Ridley AJ: Rho GTPases in cancer cell biology. FEBS Lett. 582:2093–2101. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Reymond N, Riou P and Ridley AJ: Rho GTPases and cancer cell transendothelial migration. Methods Mol Biol. 827:123–142. 2012. View Article : Google Scholar

5 

Garcia-Mata R, Boulter E and Burridge K: The ‘invisible hand’: regulation of RHO GTPases by RHOGDIs. Nat Rev Mol Cell Biol. 12:493–504. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Cho HJ, Baek KE and Yoo J: RhoGDI2 as a therapeutic target in cancer. Expert Opin Ther Targets. 14:67–75. 2010. View Article : Google Scholar

7 

Agarwal NK, Chen CH, Cho H, Boulbes DR, Spooner E and Sarbassov DD: Rictor regulates cell migration by suppressing RhoGDI2. Oncogene. 32:2521–2526. 2013. View Article : Google Scholar

8 

Griner EM and Theodorescu D: The faces and friends of RhoGDI2. Cancer Metastasis Rev. 31:519–528. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Yi B, Hu Y, Qin G, et al: Depletion of RhoGDI2 expression inhibits the ability of invasion and migration in pancreatic carcinoma. Int J Mol Med. 34:205–212. 2014.PubMed/NCBI

10 

Rhim AD, Mirek ET, Aiello NM, et al: EMT and dissemination precede pancreatic tumor formation. Cell. 148:349–361. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Kevans D, Wang LM, Sheahan K, et al: Epithelial-mesenchymal transition (EMT) protein expression in a cohort of stage II colorectal cancer patients with characterized tumor budding and mismatch repair protein status. Int J Surg Pathol. 19:751–760. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Matsuoka J, Yashiro M, Doi Y, et al: Hypoxia stimulates the EMT of gastric cancer cells through autocrine TGFβ signaling. PLoS One. 8:e623102013. View Article : Google Scholar

13 

Burgess DJ: Breast cancer: Circulating and dynamic EMT. Nat Rev Cancer. 13:1482013. View Article : Google Scholar : PubMed/NCBI

14 

Nitz MD, Harding MA and Theodorescu D: Invasion and metastasis models for studying RhoGDI2 in bladder cancer. Methods Enzymol. 439:219–233. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Li X, Wang J, Zhang X, Zeng Y, Liang L and Ding Y: Overexpression of RhoGDI2 correlates with tumor progression and poor prognosis in colorectal carcinoma. Ann Surg Oncol. 19:145–153. 2012. View Article : Google Scholar

16 

Scherle P, Behrens T and Staudt LM: Ly-GDI, a GDP-dissociation inhibitor of the RhoA GTP-binding protein, is expressed preferentially in lymphocytes. Proc Natl Acad Sci USA. 90:7568–7572. 1993. View Article : Google Scholar : PubMed/NCBI

17 

Niu H, Li H, Xu C and He P: Expression profile of RhoGDI2 in lung cancers and role of RhoGDI2 in lung cancer metastasis. Oncol Rep. 24:465–471. 2010.PubMed/NCBI

18 

Cho HJ, Baek KE, Kim IK, et al: Proteomics-based strategy to delineate the molecular mechanisms of RhoGDI2-induced metastasis and drug resistance in gastric cancer. J Proteome Res. 11:2355–2364. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Theodorescu D, Sapinoso LM, Conaway MR, Oxford G, Hampton GM and Frierson HF Jr: Reduced expression of metastasis suppressor RhoGDI2 is associated with decreased survival for patients with bladder cancer. Clin Cancer Res. 10:3800–3806. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Ma L, Xu G, Sotnikova A, et al: Loss of expression of LyGDI (ARHGDIB), a rho GDP-dissociation inhibitor, in Hodgkin lymphoma. Br J Haematol. 139:217–223. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Dovas A and Couchman JR: RhoGDI: multiple functions in the regulation of Rho family GTPase activities. Biochem J. 390:1–9. 2005. View Article : Google Scholar : PubMed/NCBI

22 

DerMardirossian C and Bokoch GM: GDIs: central regulatory molecules in Rho GTPase activation. Trends Cell Biol. 15:356–363. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Hart MJ, Maru Y, Leonard D, Witte ON, Evans T and Cerione RA: A GDP dissociation inhibitor that serves as a GTPase inhibitor for the Ras-like protein CDC42Hs. Science. 258:812–815. 1992. View Article : Google Scholar : PubMed/NCBI

24 

Chuang TH, Xu X, Knaus UG, Hart MJ and Bokoch GM: GDP dissociation inhibitor prevents intrinsic and GTPase activating protein-stimulated GTP hydrolysis by the Rac GTP-binding protein. J Biol Chem. 268:775–778. 1993.PubMed/NCBI

25 

Yilmaz M and Christofori G: EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev. 28:15–33. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Theys J, Jutten B, Habets R, et al: E-Cadherin loss associated with EMT promotes radioresistance in human tumor cells. Radiother Oncol. 99:392–397. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2015
Volume 33 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
Yi B, Zhang Y, Zhu D, Zhang L, Song S, He S, Zhang B, Li D and Zhou J: Overexpression of RhoGDI2 correlates with the progression and prognosis of pancreatic carcinoma. Oncol Rep 33: 1201-1206, 2015
APA
Yi, B., Zhang, Y., Zhu, D., Zhang, L., Song, S., He, S. ... Zhou, J. (2015). Overexpression of RhoGDI2 correlates with the progression and prognosis of pancreatic carcinoma. Oncology Reports, 33, 1201-1206. https://doi.org/10.3892/or.2015.3707
MLA
Yi, B., Zhang, Y., Zhu, D., Zhang, L., Song, S., He, S., Zhang, B., Li, D., Zhou, J."Overexpression of RhoGDI2 correlates with the progression and prognosis of pancreatic carcinoma". Oncology Reports 33.3 (2015): 1201-1206.
Chicago
Yi, B., Zhang, Y., Zhu, D., Zhang, L., Song, S., He, S., Zhang, B., Li, D., Zhou, J."Overexpression of RhoGDI2 correlates with the progression and prognosis of pancreatic carcinoma". Oncology Reports 33, no. 3 (2015): 1201-1206. https://doi.org/10.3892/or.2015.3707