Abstract
Glial cell line-derived neurotrophic factor (GDNF), a potent neurotrophic factor, has been shown to affect cancer cell metastasis and invasion. However, the molecular mechanisms underlying GDNF-induced colon cancer cell migration remain unclear. GDNF is found to be positively correlated with malignancy in human colon cancer patients. The migratory activities of two human colon cancer cell lines, HCT116 and SW480, were found to be enhanced in the presence of human GDNF. The expression of vascular endothelial growth factor (VEGF) was also increased in response to GDNF stimulation, along with VEGF mRNA expression and transcriptional activity. The enhancement of GDNF-induced cancer cell migration was antagonized by a VEGF-neutralizing antibody. Our results also showed that the expression of VEGF receptor 1 (VEGFR1) was increased in response to GDNF stimulation, whereas GDNF-induced cancer cell migration was reduced by a VEGFR inhibitor. The GDNF-induced VEGF expression was regulated by the p38 and PI3K/Akt signaling pathways. Treatment with GDNF increased nuclear hypoxia-inducible factor 1 α (HIF1α) accumulation and its transcriptional activity in a time-dependent manner. Moreover, GDNF increased hypoxia responsive element (HRE)-containing VEGF promoter transcriptional activity but not that of the HRE-deletion VEGF promoter construct. Inhibition of HIF1α by a pharmacological inhibitor or dominant-negative mutant reduced the GDNF-induced migratory activity in human colon cancer cells. These results indicate that GDNF enhances the migration of colon cancer cells by increasing VEGF–VEGFR interaction, which is mainly regulated by the p38, PI3K/Akt, and HIF1α signaling pathways.
Introduction
Vascular and neural neoplastic invasions have been found in patients with colon cancer (Bellis et al. 1993). As vascular and neural infiltrations are known to be ominous prognostic factors, their identification has great clinical relevance. Increasing evidence shows that perineural invasion or the neural cell adhesion molecule might be a prognostic factor for various cancers, such as bile duct cancer (Seki et al. 1993, Kayahara et al. 1994), gallbladder cancer (Seki et al. 1995), breast cancer (Lo et al. 1997), esophageal cancer (Tanaka et al. 1998), pancreatic cancer (Pour et al. 2003, Levy et al. 2006, Kayahara et al. 2007), prostate cancer (Li et al. 2003), and colorectal cancer (Gavert et al. 2005, Zhou et al. 2009, Li et al. 2011). Importantly, a recent report has shown that the severity of the neural invasion in rectal adenocarcinoma is an important factor in a scoring system to assess the prognostic value, showing high correlation with localization and severity (Ceyhan et al. 2010). Patients with a surgically resected pancreatic cancer showed a significantly higher frequency of positive glial cell line-derived neurotrophic factor (GDNF) immunostaining, and the intrapancreatic neural invasion by cancer cells was significantly related to the overexpression of GDNF (Zeng et al. 2008). Importantly, a strongly positive expression of GDNF was significantly more frequent than lesser grades of GDNF expression in patients with pancreatic cancer (Zeng et al. 2008).
GDNF, being a neurotrophic factor, promotes the survival and development of various neural cells in the CNS and peripheral nervous system. Previous reports have shown that GDNF also affects the survival of cancer cells and their proliferation in gliomas (Wiesenhofer et al. 2000b) and enhances the metastasis (Funahashi et al. 2005) and invasion potential (Kikuchi 2004, Ito et al. 2005, Qiao et al. 2009, Lu et al. 2010) of pancreatic cancer. GDNF and its receptor are expressed in human cancers such as gliomas (Wiesenhofer et al. 2000a). It has also been reported that GDNF is overexpressed at the transcript level in non-small-cell lung carcinomas (Garnis et al. 2005). Increased levels of GDNF in the urine could serve as a basis for the adjunct diagnosis, monitoring, and treatment of interstitial cystitis in patients with bladder cancer (Okragly et al. 1999). Recently, we reported that GDNF increases cell migration and metastasis in gliomas, chondrosarcomas, and oral squamous cell carcinomas (Su et al. 2009, Lu et al. 2010, Chuang et al. 2013). Moreover, it has also been reported that the GDNF expression in colon adenocarcinoma cells may play an important role in the pathogenesis of intestinal ganglioneuromatosis (Qiao et al. 2009), where an increased expression of GDNF is associated with diffuse ganglioneuromatosis (Furuta et al. 2007). Moreover, the enhancement of integrin expression by GDNF strongly influences its invasion of extracellular matrix proteins in colorectal cancer (Furuta et al. 2007). However, little is known about the detailed mechanism by which GDNF mediates colon cancer cell migration.
Vascular endothelial growth factor A (VEGF-A), also simply referred to as VEGF, is divided into different isoforms based upon their sequences of amino acids (Zachary & Gliki 2001). VEGF is a well-known angiogenic factor and is known to play an essential role in neovascularization (Ferrara et al. 1993). The VEGF variant is the predominant and the most critical regulator of the development of the vascular system (Zachary & Gliki 2001). The affinity of VEGF to its receptor 1 (VEGFR1) is higher than to VEGFR2 (Kaplan et al. 2005). Recent studies have shown that VEGFR1 is a critical mediator of physiological and developmental cell migration, as well as tumor-mediated metastasis (Ferrara 2004, Hicklin & Ellis 2005, Kaplan et al. 2005). However, the tyrosine kinase activity of VEGFR1 is much weaker than that of VEGFR2, and thus VEGFR2 has been considered to play the key role in angiogenesis (Ferrara 2004). Generally, increases in VEGF levels have been found to correlate with increases in the incidence of metastasis, decreases in overall survival, and poor prognosis (Takahashi et al. 1995, 1998). Importantly, VEGF is expressed in both primary and metastatic human colon cancers, and VEGF expression is higher in the metastatic cells (Hicklin & Ellis 2005). Moreover, serum levels of VEGF could be useful for screening and detecting the early stages of colon cancer (Ferrara et al. 1993). It has been demonstrated that VEGF is associated with the progression, invasion, and metastasis of colorectal cancer, and the overexpression of VEGF mRNA in the primary tumor is assumed to be closely correlated with a poor prognosis in patients with colorectal cancer (Ishigami et al. 1998). It has also been reported that treatment with monoclonal anti-VEGF antibody reduces metastases to the liver in patients with colorectal cancer (Zachary & Gliki 2001, Ferrara 2004). This study therefore sought to determine whether GDNF affects the expression of VEGF in human colon cancer. We also investigated the intracellular signaling pathways involved in the GDNF-induced upregulation of VEGF expression.
Subjects and methods
Materials
GDNF was purchased from PeproTech (Rocky Hill, NJ, USA). Fetal bovine serum (FBS), DMEM, OPTI-MEM, and Lipofectamine 2000 (LF2000) were purchased from Gibco BRL (Invitrogen Life Technologies). Primary antibodies against β-actin, ERK2, JNK, Akt, p-Akt, and p38 were purchased from Santa Cruz Biotechnology. The MAB specific for GDNF was purchased from R&D Systems (Minneapolis, MN, USA). Primary antibodies against p-p38 and p-JNK were purchased from Cell Signaling and Neuroscience (Danvers, MA, USA). The primary antibody against hypoxia-inducible factor 1α (HIF1α) was purchased from Novus Biologicals (Littleton, CO, USA). The luciferase assay kit was purchased from Promega. The dominant-negative (DN) mutant of HIF1α, HIF1α luciferase, and VEGF-luciferase 1.5 k and 1.2 k (deletion of hypoxia responsive element (HRE)) plasmids were gifts from Dr W-M Fu (National Taiwan University, Taipei, Taiwan). The DN mutant of p38 (DN-p38) was a gift from Dr C-H Tang (China Medical University, Taichung, Taiwan). The HIF1α inhibitor 3-(2-(4-adamantan-1-yl-phenoxy)-acetylamino)-4-hydroxybenzoic acid methyl ester and the VEGF receptor inhibitor N-(4-chlorophenyl)-2-[(pyridin-4-ylmethyl)amino]benzamide were purchased from Merck Co.
Immunohistochemistry
The protocol of immunohistochemistry was described in our previous reports (Lin et al. 2013, Lu et al. 2013). The human colon cancer tissue array was purchased from Biomax (Rockville, MD, USA) in the form of 5 μm sections of paraffin-embedded tissue on glass slides. Tissue specimens were rehydrated and quenched for endogenous peroxidases by soaking in 3% hydrogen peroxide in PBS. After trypsinization, sections were blocked by incubation with 5% BSA. The anti-GDNF primary antibody (1:50) was applied to the tissue specimens at 4 °C overnight. Then, after several washes with PBS, the samples were incubated with a biotin-conjugated secondary antibody (1:50). Bound antibodies were detected with an ABC reaction kit (Vector Laboratories, Burlington, CA, USA). The tissue specimens were developed with diaminobenzidine and counterstained with hematoxylin.
Cell cultures
The human colon carcinoma cell lines HCT116 and SW480 were obtained from the American Type Culture Collection (Manassas, VA, USA). The cell culture was performed according to a protocol described previously (Huang et al. 2011). Briefly, cells were grown in plastic cell culture dishes in DMEM, supplemented with 10% heat-inactivated FBS, 100 U/ml penicillin, and 100 mg/ml streptomycin, at 37 °C in a humidified incubator under an atmosphere of 5% CO2 and 95% air.
Migration assay
In vitro migration assay was performed using Costar Transwell inserts (Costar, New York, NY, USA; pore size, 8 μm) and as described in our previous reports (Chen et al. 2011, Lu et al. 2012). Before performing the migration assay, cells were pretreated for 30 min with different concentrations of inhibitors, or transfected with various DN mutants for 24 h. According to a cell viability assay, the various concentrations of inhibitors used did not affect colon cancer cell death (data not shown). Approximately 1×104 cells in 100 μl of serum-free medium were placed in the upper chamber, and 400 μl of the same medium containing GDNF was placed in the lower chamber. The plates were incubated for 24 h at 37 °C in 5% CO2, and then the cells were fixed in methanol for 15 min and stained with 0.05% crystal violet in PBS for 15 min. Cells on the upper sides of the filters were removed with cotton-tipped swabs, and the filters were washed with PBS. Cells on the undersides of the filters were examined and counted under a microscope.
Transfection
HCT116 cells were transiently transfected with 100 nM growth factor receptor α1 (GFR-α1), VEGF-A, or control siRNA by LF2000 for 24 h. Plasmid DNA and LF2000 were premixed in OPTI medium for 20 min and then applied to the cells. An equal volume of medium containing 20% FBS was added 5 h later. After transfection for 24 h, the cell cultures were washed, replenished with fresh serum-free medium, and treated with GDNF for another 24 h.
Reverse transcription PCR
Total RNA was extracted from cells using a TRIzol kit (MDBio, Inc., Taipei, Taiwan). The RT reaction was performed using 2 μg total RNA that was reverse transcribed into cDNA using the oligo(dT) primer and then amplified using the following oligonucleotide primers: VEGF, 5′-CTACCTCCACCATGCCAAGT-3′ and 5′-GCAGTAGCTGCGCTGATAGA-3′; GAPDH, 5′-ACCACAGTCCATGCCATCAC-3′ and 5′-TCCACCACCCTGTTGCTGTA-3′. Each PCR cycle was carried out for 30 s at 95 °C, 30 s at 55 °C, and 1 min at 68 °C. The PCR products were then separated electrophoretically on a 2% agarose gel and stained with ethidium bromide.
Western blot analysis
Cells were treated with GDNF for various times and then lysed briefly in homogenizing buffer on ice. The supernatants were collected by centrifugation. Equal amounts of the samples were separated by SDS–PAGE and transferred to polyvinylidene difluoride membranes (Millipore, Bedford, MA, USA). The membranes were blocked with non-fat milk in PBS and then probed with primary antibodies. After undergoing three PBS washes, the membranes were incubated with secondary antibodies. The blots were visualized by enhanced chemiluminescence using Kodak X-OMAT LS film (Eastman Kodak).
Preparation of nuclear extracts
Nuclear extracts was prepared as described previously (Lin et al. 2011, Chen et al. 2012). Cells were rinsed with PBS and suspended in hypotonic buffer A for 10 min on ice and vortexed for 10 s. The lysates were separated by centrifugation at 12 000 g for 10 min, after which the supernatants containing cytosolic proteins were collected. The pellets containing the nuclear fractions were resuspended in buffer C for 30 min on ice. The suspensions were then centrifuged at 13 000 g for 20 min, whereupon the supernatants containing nuclear proteins were collected and stored at −80 °C.
Measurement of cell viability
Cell viability was assessed by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Cells cultured in 24-well plates were treated with various concentrations of GDNF for 24 h. After incubation, MTT (0.5 mg/ml) was added for 60 min, after which the culture medium was removed and the cells were dissolved in dimethyl sulfoxide and shaken for 10 min. The absorbency (OD) values at 550 nm were immediately measured in a microplate reader. The absorbency values indicate the enzymatic activity of mitochondria and provide information on cell viability.
Statistical analysis
Statistical analysis was performed using the software GraphPad Prism 4.01 (GraphPad Software, Inc., San Diego, CA, USA). Values are the mean±s.e.m. Statistical analysis of the difference between two samples was performed using the Student's t-test. Statistical comparisons involving more than two groups were performed using one-way ANOVA with Dunnett's post-hoc test. In all cases, a value of P <0.05 was considered to be significant.
Results
GDNF increases the metastasis and migration of colon cancer cells
Higher GDNF expression was found to be associated with higher clinical pathological stages in tissue specimens of human colon cancer (Fig. 1A). The quantification results for GDNF intensity are shown in Fig. 1B. To elucidate a link between the GDNF expression and migration of colon cancer, we examined the migratory activity of human colon cancers by using transwell assays. In our results, GDNF directed the migration of both human colon cancer HCT116 cells (Fig. 2A) and SW480 cells (Fig. 2B) in a concentration-dependent manner. The pictures of the migrated cells are shown in Fig. 2D. Our results also showed that the GDNF-directed enhancement of colon cancer cell migration was abolished by treatment with a GDNF-neutralizing antibody (Fig. 2C). The receptor for GDNF is thought to be a complex of GFR-α1, which acts as a ligand-binding domain (Jing et al. 1996, Treanor et al. 1996). In order to investigate the role of the GDNF receptor in GDNF-mediated colon cancer cell migration, we transfected GFR-α1 siRNAs into HCT116 human colon cancer cells. The GDNF-induced colon cancer migration was antagonized by transfection with GFR-α1 siRNA in a concentration-dependent manner (Fig. 3A). GDNF has been identified as a ligand for Ret, which acts as the signal-transducing domain (Durbec et al. 1996, Trupp et al. 1996) and increases GDNF-induced pancreatic cancer cell invasion (Sawai et al. 2005). We therefore examined whether any interaction between GDNF and its receptor is involved in the signal transduction pathways leading to GDNF-induced colon cancer cell migration. The level of phosphorylation of the Ret receptor, after GDNF stimulation of HCT116 cells, was assessed by western blot analysis. As shown in Fig. 3B, GDNF induced Ret phosphorylation in a time-dependent manner. These data indicate that the GDNF-induced cancer cell migration may occur via activated Ret and GFR-α1 receptors in human colon cancers.
GDNF-directed colon cancer cell migration involves VEGF expression
Because angiogenesis has been found to be associated with increases in the growth and migratory potential of tumor cells, we studied the GDNF-induced expression of the angiogenic factor VEGF in HCT116 cells. As shown in Fig. 4A and B, GDNF increased the VEGF protein expression in concentration- and time-dependent manners. In addition, GDNF increased the VEGF mRNA expression in HCT116 cells (Fig. 4C). The VEGF-luciferase reporter gene assay was further used to examine whether the GDNF upregulated VEGF through a transcriptional pathway. As shown in Fig. 4D, GDNF also increased the VEGF transcriptional activity in HCT116 cells. Furthermore, the incubation of colon cancer cells with a VEGF-neutralizing antibody markedly inhibited the GDNF-induced cancer cell migration (Fig. 4E). VEGF exhibits high binding affinity to VEGFR1 (Kaplan et al. 2005). We therefore examined whether VEGFR1 is involved in the GDNF-induced migration of human colon cancer cells. After GDNF stimulation of the HCT116 cells, the expression of VEGFR1 was assessed by western blot analysis. As shown in Fig. 5A and B, GDNF induced VEGFR1 upregulation in concentration- and time-dependent manners. Moreover, pretreatment with a VEGFR pharmacological inhibitor, N-(4-chlorophenyl)-2-[(pyridin-4-ylmethyl)amino]benzamide, effectively reduced the enhancement of GDNF-induced colon cancer migration (Fig. 5C). These data indicate that GDNF-induced colon cancer migration may occur through the interaction of VEGF and VEGFR1.
MAP kinase signaling pathways are involved in GDNF-mediated VEGF upregulation and colon cancer cell migration
Previous studies and our recent report demonstrated that GDNF activates MAP kinase, and that Akt exerts biological and pathophysiological functions (Tremblay et al. 2006, Lai et al. 2010, Gu et al. 2011, Tomas et al. 2012). Thus, we examined whether MAP kinase and the Akt signaling pathway are involved in GDNF-induced VEGF expression and colon cancer migration. Our results showed that the stimulation of cells with GDNF increased the phosphorylation of ERK, p38, and JNK (Fig. 6A, B, and C respectively). On the other hand, GDNF also increased Akt phosphorylation in a time-dependent manner (Fig. 6D). Moreover, pretreatment with SB203580, LY294002, or Akt inhibitors, but not PD98059 or SP600125, reduced the GDNF-induced VEGF transcriptional activity (Fig. 6E) and VEGF expression (Fig. 6F). Transfection with p38-DN, p85-DN, and Akt-DN reduced the GDNF-enhanced migration of HCT116 cells. These results indicate that the GDNF-mediated VEGF upregulation and cell migration occur via the p38 and PI3K/Akt signaling pathways.
HIF1α is involved in GDNF-induced migration of human HCT116 colon cancer cells
As mentioned previously, intratumoral hypoxia correlates with poor prognosis and enhanced metastases formation. HIF1α is a key molecule in the hypoxic response and plays critical roles in cell detachment, invasion, energy metabolism, and induction of angiogenesis (Gort et al. 2008). Moreover, HIF1α also plays a role in this relationship by promoting expression by tumor cells of growth factors, such as VEGF (Ikeda et al. 1995, Forsythe et al. 1996). We sought to determine whether HIF1α activation is involved in GDNF-induced VEGF transcriptional activity. Our results showed that GDNF increased HIF1α accumulation in the nucleus (Fig. 7A). HIF1 binds to the HRE, which regulates the expression of a variety of genes that mediate the cellular response to hypoxic stress (Harris 2002). HCT116 cells were transfected with a VEGF promoter containing the HRE construct (1.5 k) for 24 h. As shown in Fig. 7B, the GDNF-stimulated VEGF transcriptional activity increased. However, in cells that were transfected with an HRE-deletion VEGF promoter construct (1.2 k), GDNF could not increase the VEGF transcriptional activity (Fig. 7B). Furthermore, pretreatment with an HIF1 inhibitor for 30 min or transfection with HIF1α-DN for 24 h antagonized the GDNF-enhanced migration of the HCT116 human colon cancer cells (Fig. 7C). These results indicate that the activation of HIF1α plays an important role in GDNF-induced VEGF expression and migration of human colon cancer cells.
Discussion
Colon cancer ranks among the most frequent malignancies in Taiwan and is one of the leading causes of cancer-related deaths worldwide. The metastatic dissemination of primary tumors is linked directly to patient survival and accounts for about 90% of all colon cancer deaths. Current clinical predictions on whether colon cancer will metastasize are defined mainly by histopathological staging, describing the tumor spread within a surgical specimen. It should therefore be obvious that tools and methodologies that allow early cancer detection directly will affect the patient survival time. The molecular mechanisms controlling the invasion, dissemination to blood or lymphatic systems, and spread of tumor cells to distant organs are still poorly understood. Beyond a potential application to new therapeutics, the underlying novel hypotheses and mechanistic studies will have substantial potential to reveal the interlocking nature of growth factor pathways and the way they modulate pathophysiological states. Thus, perineural invasion and hypoxia would be related to GDNF and VEGF expression in colon cancer cells, and subsequently, the induction of cancer angiogenesis and cellular migration. Our study also elucidated that downstream signaling of GDNF-receptor, together with HIF1α activation and VEGF expression, is responsible for cancer cell migration. The successful implementation of these screening procedures will contribute to a reduction in the disease-associated mortality of colon carcinoma.
Although beneficial for patients with stage III colon cancer, the role of adjuvant chemotherapy is not yet established for stage II disease. Currently, patients with stage II colonic cancer are treated mostly by surgery alone. However, patients with high-risk stage II disease, with a prognosis similar to stage III patients, should be offered chemotherapy under the current guidelines (O'Connor et al. 2011). In spite of the potentially curative resection, about 15–30% of patients with stage II disease will eventually develop recurrent locoregional disease or distant metastases within 5 years (Koebrugge et al. 2011). Possibly, other high-risk factors can influence or predict the outcome in patients with stage II colon cancer, and further research seems warranted (Iddings et al. 2006, van Schaik et al. 2009, Bilchik et al. 2010). The elucidation of the molecular biology of cancer cells is currently being exploited to develop potential therapeutic targets. In this study, we found that GDNF enhances the migratory ability of human colon cancer cells. Interestingly, it has been reported that the exogenous administration of GDNF could increase VEGF expression in spinal cord injury (Kao et al. 2008). It has also been reported that GDNF induces the migration of lung (Garnis et al. 2005) and pancreatic cancer cells (Okada et al. 2003, Funahashi et al. 2005, Gil et al. 2010).
The combination of radiotherapy and surgery has significantly improved local control and overall survival in patients with colon cancer, but cell migration and metastasis still remain a significant problem (S.R.C. 1997). Metastasis formation is a major factor in cancer progression and in the majority of mortalities. Recent observations have indicated that the metastatic phenotype may already be present during the angiogenic switch of tumors. In order for solid tumors to grow, an increase in oxygen delivery to cells via angiogenesis and glycolysis activation have to occur (Seagroves et al. 2001). Intratumoral hypoxia correlates with poor prognosis and enhanced metastases formation, where HIFs play critical roles during tumor cell expansion. Increasing evidence implicates HIF function in metastatic cell characteristics such as cell detachment, invasion, energy metabolism, and induction of angiogenesis (Gort et al. 2008). HIF1 is a heterodimeric transcription factor that is activated by low oxygen tension and it subsequently binds to HRE, which regulates the expression of a variety of genes that mediate the cellular response to hypoxic stress (Harris 2002). HIF1 plays a role in this relationship by promoting tumor cells' expression of growth factors, such as VEGF (Ikeda et al. 1995, Forsythe et al. 1996). It has been shown that blocking VEGF results in an increase in the radiosensitivity of the tumor vasculature and increases the overall tumor radiation response (Geng et al. 2001, Toiyama et al. 2010). Importantly, xenograft studies have revealed that HIF1α-deficient human colon cancer cells displayed lower rates of proliferation and migration, effectively inhibiting tumor growth (Imamura et al. 2009).
Numerous studies have indicated that VEGF and VEGFR1 are implicated in colon cancer progression. Evidence has shown that expression of VEGF and VEGFR1 correlates with vascularity, metastasis, and proliferation in human colon cancer specimens (Takahashi et al. 1995, Ellis et al. 2000). Recent studies have also demonstrated that VEGF and VEGFR1 are expressed in human colon cancer tissue specimens, and that VEGF generated by colon cancer cells stimulates their growth directly through an autocrine mechanism that is independent of its primary function of angiogenesis induction (Ahluwalia et al. 2013). Moreover, in clinical studies that compared the efficacy of standard metastatic colorectal chemotherapy (irinotecan, 5-fluorouracil, and leucovorin) with and without bevacizumab, an anti-VEGF antibody, increased median survival with bevacizumab indicates a relationship between VEGF and metastatic colon cancer (Ferrara et al. 2004, Roskoski 2007). Several lines of evidence indicate that VEGFR2 mediates the stimulatory effects of VEGF on angiogenesis, whereas VEGFR1 actually attenuates the actions of VEGF by binding VEGF without activating angiogenesis (Zachary & Gliki 2001). Importantly, a previous study reported that colon cancer cells express VEGFR1, but not VEGFR2 (Fan et al. 2005). In addition, VEGFR1 has been reported to stimulate endothelial cell migration in response to VEGF-A (Kanno et al. 2000). A recent report has also shown that prostaglandin E2 regulates cell migration via the induction of VEGFR1 in HCA7 human colon cancer cells (Fujino et al. 2011). Similarly, in this study, GDNF induced VEGFR1 upregulation in concentration- and time-dependent manners. Moreover, pretreatment with a VEGFR pharmacological inhibitor effectively reduced the enhancement of GDNF-induced colon cancer migration. Conclusively, therefore, GDNF directs cell migration by the interaction of VEGF and VEGFR1.
Signaling molecules, including ERK, p38, JNK, and Akt, have been demonstrated to play important roles in colon cancer cell invasion and migration (Tremblay et al. 2006, Lai et al. 2010, Gu et al. 2011, Tomas et al. 2012). The activation of Ret by GDNF results in the activation of the MAP kinase pathways (van Weering & Bos 1997). Specifically, Ret activates the ERK and JNK signaling pathways (Marek et al. 2004, Asai et al. 2006). Our results showed that GDNF induced ERK, p38, JNK, and Akt phosphorylation in human colon cancer cells. Furthermore, SB203580 and Ly294002, as well as an Akt inhibitor, effectively antagonized the GDNF-induced VEGF transcriptional activity and protein expression. Knockdown of p38 and Akt also reduced the GDNF-induced cell migration in human colon cancer. These results indicate that GDNF directs the migration of colon cancer cells through the activation of the p38 and Akt signaling pathways via Ret activation. Our results indicate that GDNF directs the migration of colon cancer through the activation of Ret and GFR-α1, as well as via p38 and Akt activation. Moreover, GDNF increased VEGF expression via HIF-1α activation, thereby contributing to tumor migration. Our findings, which reveal the molecular mechanisms by which GDNF promotes colon cancer cell migration, may lead to a better understanding of the malignant progression of human colon cancers.
Declaration of interest
The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.
Funding
This work was supported by grants from the National Science Council of Taiwan (NSC 102-2320-B-039-051-MY3 and NSC 101-2314-B-303-016-MY3), China Medical University (CMU 102 S-21), and Taichung Tzuchi Hospital, The Buddhist Tzuchi Medical Foundation (TTCRD-101-12).
Author contribution statement
S-M Huang, T-S Chen, C-M Chiu, and L-K Chang performed experiments, acquisition and analysis of data, and drafted the manuscript. K-F Liao, H-M Tan and W-L Yeh helped to perform experiments and prepare the manuscript. S-M Huang, M-Y Wang and D-Y Lu conceived the study and participated in its design and coordination, S-M Huang, M-Y Wang and D-Y Lu were involved in drafting the manuscript and revising it for intellectual content. All authors read and approved the final manuscript.
References
Ahluwalia A, Jones MK, Szabo S & Tarnawski AS 2013 Aberrant, ectopic expression of VEGF and VEGF receptors 1 and 2 in malignant colonic epithelial cells. Implications for these cells growth via an autocrine mechanism. Biochemical and Biophysical Research Communications 437 515–520. (doi:10.1016/j.bbrc.2013.06.096).
Asai N, Fukuda T, Wu Z, Enomoto A, Pachnis V, Takahashi M & Costantini F 2006 Targeted mutation of serine 697 in the Ret tyrosine kinase causes migration defect of enteric neural crest cells. Development 133 4507–4516. (doi:10.1242/dev.02616).
Bellis D, Marci V & Monga G 1993 Light microscopic and immunohistochemical evaluation of vascular and neural invasion in colorectal cancer. Pathology, Research and Practice 189 443–447. (doi:10.1016/S0344-0338(11)80333-X).
Bilchik A, Nissan A, Wainberg Z, Shen P, McCarter M, Protic M, Howard R, Elashoff D, Tyler J & Peoples GE et al. 2010 Surgical quality and nodal ultrastaging is associated with long-term disease-free survival in early colorectal cancer: an analysis of 2 international multicenter prospective trials. Annals of Surgery 252 467–474.discussion 474–476 (doi:10.1097/SLA.0b013e3181f19767).
Ceyhan GO, Liebl F, Maak M, Schuster T, Becker K, Langer R, Demir IE, Hartel M, Friess H & Rosenberg R 2010 The severity of neural invasion is a crucial prognostic factor in rectal cancer independent of neoadjuvant radiochemotherapy. Annals of Surgery 252 797–804. (doi:10.1097/SLA.0b013e3181fcab8d).
Chen JH, Huang SM, Chen CC, Tsai CF, Yeh WL, Chou SJ, Hsieh WT & Lu DY 2011 Ghrelin induces cell migration through GHS-R, CaMKII, AMPK, and NF-κB signaling pathway in glioma cells. Journal of Cellular Biochemistry 112 2931–2941. (doi:10.1002/jcb.23209).
Chen JH, Huang SM, Tan TW, Lin HY, Chen PY, Yeh WL, Chou SC, Tsai CF, Wei IH & Lu DY 2012 Berberine induces heme oxygenase-1 up-regulation through phosphatidylinositol 3-kinase/AKT and NF-E2-related factor-2 signaling pathway in astrocytes. International Immunopharmacology 12 94–100. (doi:10.1016/j.intimp.2011.10.019).
Chuang JY, Tsai CF, Chang SW, Chiang IP, Huang SM, Lin HY, Yeh WL & Lu DY 2013 Glial cell line-derived neurotrophic factor induces cell migration in human oral squamous cell carcinoma. Oral Oncology 49 1103–1112. (doi:10.1016/j.oraloncology.2013.08.009).
Durbec P, Marcos-Gutierrez CV, Kilkenny C, Grigoriou M, Wartiowaara K, Suvanto P, Smith D, Ponder B, Costantini F & Saarma M et al. 1996 GDNF signalling through the Ret receptor tyrosine kinase. Nature 381 789–793. (doi:10.1038/381789a0).
Ellis LM, Takahashi Y, Liu W & Shaheen RM 2000 Vascular endothelial growth factor in human colon cancer: biology and therapeutic implications. Oncologist 5 (Suppl 1) 11–15. (doi:10.1634/theoncologist.5-suppl_1-11).
Fan F, Wey JS, McCarty MF, Belcheva A, Liu W, Bauer TW, Somcio RJ, Wu Y, Hooper A & Hicklin DJ et al. 2005 Expression and function of vascular endothelial growth factor receptor-1 on human colorectal cancer cells. Oncogene 24 2647–2653. (doi:10.1038/sj.onc.1208246).
Ferrara N 2004 Vascular endothelial growth factor: basic science and clinical progress. Endocrine Reviews 25 581–611. (doi:10.1210/er.2003-0027).
Ferrara N, Winer J, Burton T, Rowland A, Siegel M, Phillips HS, Terrell T, Keller GA & Levinson AD 1993 Expression of vascular endothelial growth factor does not promote transformation but confers a growth advantage in vivo to Chinese hamster ovary cells. Journal of Clinical Investigation 91 160–170. (doi:10.1172/JCI116166).
Ferrara N, Hillan KJ, Gerber HP & Novotny W 2004 Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nature Reviews. Drug Discovery 3 391–400. (doi:10.1038/nrd1381).
Forsythe JA, Jiang BH, Iyer NV, Agani F, Leung SW, Koos RD & Semenza GL 1996 Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Molecular and Cellular Biology 16 4604–4613.
Fujino H, Toyomura K, Chen XB, Regan JW & Murayama T 2011 Prostaglandin E2 regulates cellular migration via induction of vascular endothelial growth factor receptor-1 in HCA-7 human colon cancer cells. Biochemical Pharmacology 81 379–387. (doi:10.1016/j.bcp.2010.11.001).
Funahashi H, Okada Y, Sawai H, Takahashi H, Matsuo Y, Takeyama H & Manabe T 2005 The role of glial cell line-derived neurotrophic factor (GDNF) and integrins for invasion and metastasis in human pancreatic cancer cells. Journal of Surgical Oncology 91 77–83. (doi:10.1002/jso.20277).
Furuta A, Funahashi H, Sawai H, Sato M, Okada Y, Takeyama H & Manabe T 2007 The relationship between GDNF and integrins in human colorectal cancer cell activity. Hepatogastroenterology 54 1398–1402.
Garnis C, Davies JJ, Buys TP, Tsao MS, MacAulay C, Lam S & Lam WL 2005 Chromosome 5p aberrations are early events in lung cancer: implication of glial cell line-derived neurotrophic factor in disease progression. Oncogene 24 4806–4812. (doi:10.1038/sj.onc.1208643).
Gavert N, Conacci-Sorrell M, Gast D, Schneider A, Altevogt P, Brabletz T & Ben-Ze'ev A 2005 L1, a novel target of β-catenin signaling, transforms cells and is expressed at the invasive front of colon cancers. Journal of Cell Biology 168 633–642. (doi:10.1083/jcb.200408051).
Geng L, Donnelly E, McMahon G, Lin PC, Sierra-Rivera E, Oshinka H & Hallahan DE 2001 Inhibition of vascular endothelial growth factor receptor signaling leads to reversal of tumor resistance to radiotherapy. Cancer Research 61 2413–2419.
Gil Z, Cavel O, Kelly K, Brader P, Rein A, Gao SP, Carlson DL, Shah JP, Fong Y & Wong RJ 2010 Paracrine regulation of pancreatic cancer cell invasion by peripheral nerves. Journal of the National Cancer Institute 102 107–118. (doi:10.1093/jnci/djp456).
Gort EH, Groot AJ, van der Wall E, van Diest PJ & Vooijs MA 2008 Hypoxic regulation of metastasis via hypoxia-inducible factors. Current Molecular Medicine 8 60–67. (doi:10.2174/156652408783565568).
Gu S, Papadopoulou N, Nasir O, Foller M, Alevizopoulos K, Lang F & Stournaras C 2011 Activation of membrane androgen receptors in colon cancer inhibits the prosurvival signals Akt/bad in vitro and in vivo and blocks migration via vinculin/actin signaling. Molecular Medicine 17 48–58. (doi:10.2119/molmed.2010.00120).
Harris AL 2002 Hypoxia – a key regulatory factor in tumour growth. Nature Reviews. Cancer 2 38–47. (doi:10.1038/nrc704).
Hicklin DJ & Ellis LM 2005 Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. Journal of Clinical Oncology 23 1011–1027. (doi:10.1200/JCO.2005.06.081).
Huang SM, Cheung CW, Chang CS, Tang CH, Liu JF, Lin YH, Chen JH, Ko SH, Wong KL & Lu DY 2011 Phloroglucinol derivative MCPP induces cell apoptosis in human colon cancer. Journal of Cellular Biochemistry 112 643–652. (doi:10.1002/jcb.22966).
Iddings D, Ahmad A, Elashoff D & Bilchik A 2006 The prognostic effect of micrometastases in previously staged lymph node negative (N0) colorectal carcinoma: a meta-analysis. Annals of Surgical Oncology 13 1386–1392. (doi:10.1245/s10434-006-9120-y).
Ikeda E, Achen MG, Breier G & Risau W 1995 Hypoxia-induced transcriptional activation and increased mRNA stability of vascular endothelial growth factor in C6 glioma cells. Journal of Biological Chemistry 270 19761–19766. (doi:10.1074/jbc.270.44.26460).
Imamura T, Kikuchi H, Herraiz MT, Park DY, Mizukami Y, Mino-Kenduson M, Lynch MP, Rueda BR, Benita Y & Xavier RJ et al. 2009 HIF-1α and HIF-2α have divergent roles in colon cancer. International Journal of Cancer 124 763–771. (doi:10.1002/ijc.24032).
Ishigami SI, Arii S, Furutani M, Niwano M, Harada T, Mizumoto M, Mori A, Onodera H & Imamura M 1998 Predictive value of vascular endothelial growth factor (VEGF) in metastasis and prognosis of human colorectal cancer. British Journal of Cancer 78 1379–1384. (doi:10.1038/bjc.1998.688).
Ito Y, Okada Y, Sato M, Sawai H, Funahashi H, Murase T, Hayakawa T & Manabe T 2005 Expression of glial cell line-derived neurotrophic factor family members and their receptors in pancreatic cancers. Surgery 138 788–794. (doi:10.1016/j.surg.2005.07.007).
Jing S, Wen D, Yu Y, Holst PL, Luo Y, Fang M, Tamir R, Antonio L, Hu Z & Cupples R et al. 1996 GDNF-induced activation of the ret protein tyrosine kinase is mediated by GDNFR-α, a novel receptor for GDNF. Cell 85 1113–1124. (doi:10.1016/S0092-8674(00)81311-2).
Kanno S, Oda N, Abe M, Terai Y, Ito M, Shitara K, Tabayashi K, Shibuya M & Sato Y 2000 Roles of two VEGF receptors, Flt-1 and KDR, in the signal transduction of VEGF effects in human vascular endothelial cells. Oncogene 19 2138–2146. (doi:10.1038/sj.onc.1203533).
Kao CH, Chen SH, Chio CC, Chang CK & Lin MT 2008 Exogenous administration of glial cell line-derived neurotrophic factor improves recovery after spinal cord injury. Resuscitation 77 395–400. (doi:10.1016/j.resuscitation.2008.01.023).
Kaplan RN, Riba RD, Zacharoulis S, Bramley AH, Vincent L, Costa C, MacDonald DD, Jin DK, Shido K & Kerns SA et al. 2005 VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 438 820–827. (doi:10.1038/nature04186).
Kayahara M, Nagakawa T, Tsukioka Y, Ohta T, Ueno K & Miyazaki I 1994 Neural invasion and nodal involvement in distal bile duct cancer. Hepatogastroenterology 41 190–194.
Kayahara M, Nakagawara H, Kitagawa H & Ohta T 2007 The nature of neural invasion by pancreatic cancer. Pancreas 35 218–223. (doi:10.1097/mpa.0b013e3180619677).
Kikuchi K 2004 Expression of GDNF (glial cell line-derived neurotrophic factor) and Ret in normal human and cancerous pancreatic tissues. Hokkaido Igaku Zasshi 79 585–595.
Koebrugge B, Vogelaar FJ, Lips DJ, Pruijt JF, van der Linden JC, Ernst MF & Bosscha K 2011 The number of high-risk factors is related to outcome in stage II colonic cancer patients. European Journal of Surgical Oncology 37 964–970. (doi:10.1016/j.ejso.2011.08.135).
Lai KC, Huang AC, Hsu SC, Kuo CL, Yang JS, Wu SH & Chung JG 2010 Benzyl isothiocyanate (BITC) inhibits migration and invasion of human colon cancer HT29 cells by inhibiting matrix metalloproteinase-2/-9 and urokinase plasminogen (uPA) through PKC and MAPK signaling pathway. Journal of Agricultural and Food Chemistry 58 2935–2942. (doi:10.1021/jf9036694).
Levy MJ, Topazian M, Keeney G, Clain JE, Gleeson F, Rajan E, Wang KK, Wiersema MJ, Farnell M & Chari S 2006 Preoperative diagnosis of extrapancreatic neural invasion in pancreatic cancer. Clinical Gastroenterology and Hepatology 4 1479–1482. (doi:10.1016/j.cgh.2006.08.012).
Li R, Wheeler T, Dai H & Ayala G 2003 Neural cell adhesion molecule is upregulated in nerves with prostate cancer invasion. Human Pathology 34 457–461. (doi:10.1016/S0046-8177(03)00084-4).
Li D, Peng X, Yan D, Tang H, Huang F, Yang Y & Peng Z 2011 Msi-1 is a predictor of survival and a novel therapeutic target in colon cancer. Annals of Surgical Oncology 18 2074–2083. (doi:10.1245/s10434-011-1567-9).
Lin HY, Tang CH, Chen JH, Chuang JY, Huang SM, Tan TW, Lai CH & Lu DY 2011 Peptidoglycan induces interleukin-6 expression through the TLR2 receptor, JNK, c-Jun, and AP-1 pathways in microglia. Journal of Cellular Physiology 226 1573–1582. (doi:10.1002/jcp.22489).
Lin HY, Huang BR, Yeh WL, Lee CH, Huang SS, Lai CH, Lin H & Lu DY 2013 Antineuroinflammatory effects of lycopene via activation of adenosine monophosphate-activated protein kinase-α1/heme oxygenase-1 pathways. Neurobiology of Aging 35 191–202. (doi:10.1016/j.neurobiolaging.2013.06.020).
Lo JY, Baker JA, Kornguth PJ, Iglehart JD & Floyd CE Jr 1997 Predicting breast cancer invasion with artificial neural networks on the basis of mammographic features. Radiology 203 159–163.
Lu DY, Leung YM, Cheung CW, Chen YR & Wong KL 2010 Glial cell line-derived neurotrophic factor induces cell migration and matrix metalloproteinase-13 expression in glioma cells. Biochemical Pharmacology 80 1201–1209. (doi:10.1016/j.bcp.2010.06.046).
Lu DY, Yeh WL, Huang SM, Tang CH, Lin HY & Chou SJ 2012 Osteopontin increases heme oxygenase-1 expression and subsequently induces cell migration and invasion in glioma cells. Neuro-Oncology 14 1367–1378. (doi:10.1093/neuonc/nos262).
Lu DY, Huang BR, Yeh WL, Lin HY, Huang SS, Liu YS & Kuo YH 2013 Anti-neuroinflammatory effect of a novel caffeamide derivative, KS370G, in microglial cells. Molecular Neurobiology 48 863–874. (doi:10.1007/s12035-013-8474-y).
Marek L, Levresse V, Amura C, Zentrich E, Van Putten V, Nemenoff RA & Heasley LE 2004 Multiple signaling conduits regulate global differentiation-specific gene expression in PC12 cells. Journal of Cellular Physiology 201 459–469. (doi:10.1002/jcp.20087).
O'Connor ES, Greenblatt DY, LoConte NK, Gangnon RE, Liou JI, Heise CP & Smith MA 2011 Adjuvant chemotherapy for stage II colon cancer with poor prognostic features. Journal of Clinical Oncology 29 3381–3388. (doi:10.1200/JCO.2010.34.3426).
Okada Y, Eibl G, Duffy JP, Reber HA & Hines OJ 2003 Glial cell-derived neurotrophic factor upregulates the expression and activation of matrix metalloproteinase-9 in human pancreatic cancer. Surgery 134 293–299. (doi:10.1067/msy.2003.239).
Okragly AJ, Niles AL, Saban R, Schmidt D, Hoffman RL, Warner TF, Moon TD, Uehling DT & Haak-Frendscho M 1999 Elevated tryptase, nerve growth factor, neurotrophin-3 and glial cell line-derived neurotrophic factor levels in the urine of interstitial cystitis and bladder cancer patients. Journal of Urology 161 438–441.discussion 441–442 (doi:10.1016/S0022-5347(01)61915-3).
Pour PM, Bell RH & Batra SK 2003 Neural invasion in the staging of pancreatic cancer. Pancreas 26 322–325. (doi:10.1097/00006676-200305000-00002).
Qiao S, Iwashita T, Ichihara M, Murakumo Y, Yamaguchi A, Isogai M, Sakata K & Takahashi M 2009 Increased expression of glial cell line-derived neurotrophic factor and neurturin in a case of colon adenocarcinoma associated with diffuse ganglioneuromatosis. Clinical Neuropathology 28 105–112. (doi:10.5414/NPP28105).
Roskoski R Jr 2007 Vascular endothelial growth factor (VEGF) signaling in tumor progression. Critical Review in Oncology and Hematology 62 179–213. (doi:10.1016/j.critrevonc.2007.01.006).
Sawai H, Okada Y, Kazanjian K, Kim J, Hasan S, Hines OJ, Reber HA, Hoon DS & Eibl G 2005 The G691S RET polymorphism increases glial cell line-derived neurotrophic factor-induced pancreatic cancer cell invasion by amplifying mitogen-activated protein kinase signaling. Cancer Research 65 11536–11544. (doi:10.1158/0008-5472.CAN-05-2843).
van Schaik PM, Hermans E, van der Linden JC, Pruijt JR, Ernst MF & Bosscha K 2009 Micro-metastases in stages I and II colon cancer are a predictor of the development of distant metastases and worse disease-free survival. European Journal of Surgical Oncology 35 492–496. (doi:10.1016/j.ejso.2008.07.011).
Seagroves TN, Ryan HE, Lu H, Wouters BG, Knapp M, Thibault P, Laderoute K & Johnson RS 2001 Transcription factor HIF-1 is a necessary mediator of the pasteur effect in mammalian cells. Molecular and Cellular Biology 21 3436–3444. (doi:10.1128/MCB.21.10.3436-3444.2001).
Seki H, Tanaka J, Sato Y, Kato Y, Umezawa A & Koyama K 1993 Neural cell adhesion molecule (NCAM) and perineural invasion in bile duct cancer. Journal of Surgical Oncology 53 78–83. (doi:10.1002/jso.2930530205).
Seki H, Koyama K, Tanaka J, Sato Y & Umezawa A 1995 Neural cell adhesion molecule and perineural invasion in gallbladder cancer. Journal of Surgical Oncology 58 97–100. (doi:10.1002/jso.2930580205).
S.R.C. Improved survival with preoperative radiotherapy in resectable rectal cancer. Swedish Rectal Cancer Trial New England Journal of Medicine 336 1997 980–987. (doi:10.1056/NEJM199704033361402).
Su CM, Lu DY, Hsu CJ, Chen HT, Huang CY, Yang WH, Su YC, Yang SN, Fong YC & Tseng WP et al. 2009 Glial cell-derived neurotrophic factor increases migration of human chondrosarcoma cells via ERK and NF-κB pathways. Journal of Cellular Physiology 220 499–507. (doi:10.1002/jcp.21802).
Takahashi Y, Kitadai Y, Bucana CD, Cleary KR & Ellis LM 1995 Expression of vascular endothelial growth factor and its receptor, KDR, correlates with vascularity, metastasis, and proliferation of human colon cancer. Cancer Research 55 3964–3968.
Takahashi Y, Bucana CD, Cleary KR & Ellis LM 1998 p53, vessel count, and vascular endothelial growth factor expression in human colon cancer. International Journal of Cancer 79 34–38. (doi:10.1002/(SICI)1097-0215(19980220)79:1<34::AID-IJC7>3.0.CO;2-X).
Tanaka A, Matsumura E, Yosikawa H, Uchida T, Machidera N, Kubo R, Okuno K, Koh K, Watatani M & Yasutomi M 1998 An evaluation of neural invasion in esophageal cancer. Surgery Today 28 873–878. (doi:10.1007/s005950050245).
Toiyama Y, Inoue Y, Saigusa S, Okugawa Y, Yokoe T, Tanaka K, Miki C & Kusunoki M 2010 Gene expression profiles of epidermal growth factor receptor, vascular endothelial growth factor and hypoxia-inducible factor-1 with special reference to local responsiveness to neoadjuvant chemoradiotherapy and disease recurrence after rectal cancer surgery. Clinical Oncology 22 272–280. (doi:10.1016/j.clon.2010.01.001).
Tomas NM, Masur K, Piecha JC, Niggemann B & Zanker KS 2012 Akt and phospholipase Cγ are involved in the regulation of growth and migration of MDA-MB-468 breast cancer and SW480 colon cancer cells when cultured with diabetogenic levels of glucose and insulin. BMC Research Notes 5 214. (doi:10.1186/1756-0500-5-214).
Treanor JJ, Goodman L, de Sauvage F, Stone DM, Poulsen KT, Beck CD, Gray C, Armanini MP, Pollock RA & Hefti F et al. 1996 Characterization of a multicomponent receptor for GDNF. Nature 382 80–83. (doi:10.1038/382080a0).
Tremblay PL, Auger FA & Huot J 2006 Regulation of transendothelial migration of colon cancer cells by E-selectin-mediated activation of p38 and ERK MAP kinases. Oncogene 25 6563–6573. (doi:10.1038/sj.onc.1209664).
Trupp M, Arenas E, Fainzilber M, Nilsson AS, Sieber BA, Grigoriou M, Kilkenny C, Salazar-Grueso E, Pachnis V & Arumae U 1996 Functional receptor for GDNF encoded by the c-ret proto-oncogene. Nature 381 785–789. (doi:10.1038/381785a0).
van Weering DH & Bos JL 1997 Glial cell line-derived neurotrophic factor induces Ret-mediated lamellipodia formation. Journal of Biological Chemistry 272 249–254. (doi:10.1074/jbc.272.1.249).
Wiesenhofer B, Stockhammer G, Kostron H, Maier H, Hinterhuber H & Humpel C 2000a Glial cell line-derived neurotrophic factor (GDNF) and its receptor (GFR-α1) are strongly expressed in human gliomas. Acta Neuropathologica 99 131–137. (doi:10.1007/PL00007416).
Wiesenhofer B, Weis C & Humpel C 2000b Glial cell line-derived neurotrophic factor (GDNF) is a proliferation factor for rat C6 glioma cells: evidence from antisense experiments. Antisense & Nucleic Acid Drug Development 10 311–321. (doi:10.1089/oli.1.2000.10.311).
Zachary I & Gliki G 2001 Signaling transduction mechanisms mediating biological actions of the vascular endothelial growth factor family. Cardiovascular Research 49 568–581. (doi:10.1016/S0008-6363(00)00268-6).
Zeng Q, Cheng Y, Zhu Q, Yu Z, Wu X, Huang K, Zhou M, Han S & Zhang Q 2008 The relationship between overexpression of glial cell-derived neurotrophic factor and its RET receptor with progression and prognosis of human pancreatic cancer. Journal of International Medical Research 36 656–664. (doi:10.1177/147323000803600406).
Zhou J, Li J, Chen J, Liu Y, Gao W & Ding Y 2009 Over-expression of CDH22 is associated with tumor progression in colorectal cancer. Tumour Biology 30 130–140. (doi:10.1159/000225242).