siRNA-mediated inhibition of endogenous brain‑derived neurotrophic factor gene modulates the biological behavior of HeLa cells Corrigendum in /10.3892/or.2021.7922

Retraction in: /10.3892/or.2022.8438

siRNA-mediated inhibition of endogenous brain‑derived neurotrophic factor gene modulates the biological behavior of HeLa cells

Corrigendum in: /10.3892/or.2021.7922 Retraction in /10.3892/or.2022.8438

  • Authors:
    • Chun-Yan Sun
    • Zhang-Bo Chu
    • Jing Huang
    • Lei Chen
    • Jian Xu
    • Ao-Shuang Xu
    • Jun-Ying Li
    • Yu Hu
  • View Affiliations

  • Published online on: April 11, 2017     https://doi.org/10.3892/or.2017.5569
  • Pages: 2751-2760
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Brain-derived neurotrophic factor (BDNF) is expressed in a number of neural and non-neuronal tumors. The present study investigated the effect of endogenous BDNF on the biological behavior of cervix cancer cells using small interfering RNA (siRNA). HeLa, a cervix cancer cell line with high expression of BDNF, was used as a living model to screen out the effective sequences of short hairpin RNA of the BDNF gene, and the effects of RNA interference on proliferation, apoptosis, migration and invasion of these cells were evaluated. Among the 4 siRNAs examined, siRNA1 caused a 99% reduction in the relative BDNF mRNA level, while a 58% decrease in the relative BDNF protein level (p<0.01) was noted, and thus this siRNA was selected as the most efficient for use in the present study. In subsequent experiments, MTT assay revealed that BDNF silencing caused marked inhibition of HeLa cell proliferation while Hoechst 33258 staining assay demonstrated apoptosis-related changes in cell morphology. Downregulation of BDNF expression induced cell cycle arrest in the G1 phase as shown by flow cytometry. As indicated by Transwell migration and invasion assays, downregulation of BDNF expression suppressed the migratory and invasive capabilities of the HeLa cells. Together, our data revealed that BDNF modulates the proliferation, apoptosis, migratory and invasive capabilities of HeLa cells. BDNF siRNA may represent a novel therapy or drug target for preventing the tumorigenesis of cervical cancer.

Introduction

Brain-derived neurotrophic factor (BDNF), a member of the neurotrophin family, plays an essential role in promoting the growth, survival and differentiation of developing neurons in the central and peripheral nervous systems (1). In addition, BDNF is also involved in peripheral sensory and motor neuron regeneration at the site of nerve injury (2). Apart from its well-known effects on neurons, recent studies have demonstrated that BDNF and its high-affinity tyrosine kinase (Trk) receptor TrkB are constitutively expressed in a number of neural and non-neuronal tumors, including neuroblastoma (3), hepatocellular carcinoma (4), multiple meloma (5), prostatic carcinoma (6), pancreatic ductal adenocarcinoma (6), adenoid cystic carcinoma (7) and retinoblastoma (8). Moreover, it has been reported that BDNF and TrkB are preferentially expressed in more aggressive neuroblastoma with N-myc amplification (9). BDNF is expressed in 33 and 100% of typical (TC) and atypical (AC) pulmonary carcinoids, respectively, indicating the unfavorable prognosis of patients (10). In addition to malignant cells, BDNF can be produced by osteoblasts, and bone marrow (BM) endothelial cells in BM stroma (5), implying that the paracrine and autocrine functions of BDNF play critical roles in interaction of MM plasma cells with BM microenvironment as interleukin-6 (IL-6) (11). Exogenous BDNF participates in the promotion of the growth and survival of tumor cells, protection of tumor cells from chemotherapy-induced apoptosis (12), enhancing invasive and migratory capabilities of tumor cells in a dose-dependent manner (13), cooperation with TrkB in transforming rat intestinal epithelia cells to malignant cells and suppression of anoikis (apoptosis resulting from loss of cell-matrix interactions) (14), induction of the tube formation of human umbilical vein endothelial cells (HUVEC) in vitro and modulation of angiogenesis in tumors (15). These studies suggest the potential roles of BDNF in tumorigenicity and the progression of cancers; however, the mechanisms have not yet been completely clarified.

RNA interference (RNAi) is a process of post-transcriptional gene silencing in which double-stranded RNA (dsRNA) inhibits gene expression in a sequence-dependent manner via degradation of the corresponding mRNA (1618). RNAi is initiated by an event whereby dsRNA is recognized by Dicer (19). The Dicer enzyme cleaves dsRNA into 19–22 nucleotide short interfering RNA (siRNA). These siRNA duplexes are incorporated into a protein complex called the RNAi-induced silencing complex (RISC), where they are cleaved into 2 single strands (guide and passenger strand). The latter is degraded, while the former, antisense siRNA, pairs with the complementary sequence in mRNA to induce its cleavage (20). A previous study investigated the RNAi-mediated inhibition of BDNF in rat myoblasts. The reduction in the level of BDNF promoted myoblasts to exit the cell cycle and initiate the myogenic differentiation program (21). RNAi-mediated, sequence-specific gene silencing revealed that inhibition of BDNF expression enhanced cocaine cytotoxicity in neuroblastoma SK-N-AS cells and primary rat hippocampal neurons (22).

In the present study, small interfering RNA (siRNA) was used as a tool for suppressing the expression of the endogenous BDNF gene in HeLa, a cervical carcinoma cell line with high expression of BDNF, in order to investigate the effects of RNA interference on the proliferation, apoptosis, cell cycle distribution and the invasive and migratory capabilities of HeLa cells.

Materials and methods

Construction of the recombinant eukaryotic human-BDNF siRNA expression vector

BDNF siRNA was designed according to the siRNA design guidelines of Ambion Inc. (Carlsbad, CA, USA). General design guidelines are as follows. The selected 19–22 base siRNA sequences were designed with 30–50% guanine cytosine content to avoid inverted repeats (23). Four siRNAs were chosen based on the sequence of the human BDNF gene. They covered different regions of the BDNF sequence and showed no homology with other human genes. A scrambled siRNA was used as a negative control. The target sequences and corresponding hairpin siRNA sequences for the 5 siRNAs, designated siRNA1, siRNA2, siRNA3, siRNA4 and scramble siRNA, are shown in Fig. 1. The structure of hairpin siRNA was BamHI + sense + loop + antisense + termination signal + SalI + HindIII. pGenesil-1 [enhanced green fluorescent protein (EGFP) purchased from Genesil (Wuhan, China)] was introduced for the construction of recombinant eukaryotic human BDNF siRNA expression vectors. Five pairs of hairpin siRNA sense and antisense sequences were synthesized, annealed and cloned into the BamHI/HindIII cloning site of pGenesil-1, respectively. The products were transformed into DH5α-competent cells. Kanamycin-resistant colonies were chosen, identified by restriction digestion, and further confirmed by DNA sequencing. The synthesis of all DNA chains and DNA sequencing was performed by Genesil.

Cell culture and transfection

The cervical carcinoma cell line HeLa was cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum (FBS), 100 U/ml penicillin, 100 U/ml streptomycin, and was routinely maintained at 37̊C in 5% CO2. Medium was changed every 3 days.

HeLa cells were seeded in 6-well plates (5×105/ml). Transient transfection was performed using Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA) according to the manufacturer's instructions. For stable transfection, HeLa cells were cultured in the standard culture medium for 48 h after transfection. G418 selection at 800 µg/ml was applied and continued for 4 weeks until single colonies were formed. In parallel, non-transfected cells were also placed in standard culture medium to ensure the potency and selectivity of G418. Positive clones were maintained in the culture medium with G418 at 300 µg/ml. To determine the transfection efficiencies and expression effects of BDNF siRNA in HeLa cells, EGFP expression was examined by microscopy (magnification, ×400) at 24 h after transfection and after the positive clones were established.

RNA extraction and reverse-transcriptase polymerase chain reaction (RT-PCR)

Total RNA from HeLa cells was isolated using TRIzol extraction according to the manufacturer's protocol (Invitrogen). Complementary DNA (cDNA) was subsequently synthesized from total RNA (5 µg) using RevertAid First-Strand cDNA Synthesis kit (Fermentas, Burlington, Ontario, Canada). Then, 1 µg of cDNA was subjected to PCR for the selected genes. Primers used were as follows: 5′-GCAGCCTTCTTTTGTGTAACC-3′ and 5′-AGAGTGATGACCATCCTTTTC-3′ for BDNF (594 bp); as well as 5′-GAAGGTGAAGGTCGGAGTC-3′ and 5′-GAAGATGGTGATGGGATTC-3′ for glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (226 bp) as control; 5′-GTTTCATAAGATCCCACTGGATGG-3′ and 5′-TGCTGCTTAGCTGCCTGAGAGTTA-3′ for TrkB (260 bp); as well as 5′-TGAGACCTTCAACACCCCAG-3′, and 5′-GCCATCTCTTGCTCGAAGTC-3′ for β-actin (312 bp) as control. PCR profiles consisted of denaturation at 94̊C for 1 min, annealing at 55̊C for 30 sec, and extension at 72̊C for 1 min. The samples were amplified for 32 cycles. PCR products were separated by electrophoresis on 1.5% agarose gels, stained with ethidium bromide and photographed.

Western blot analysis

Cells were washed twice with ice-cold phosphate-buffered saline (PBS), and then harvested on ice with NP40 lysis buffer, containing 50 mmol/l Tris-HCl, pH 7.4; 150 mmol/l NaCl; 1% NP40; 5 mmol/l EDTA; 5 mmol/l NaF; 2 mmol/l sodium vanadate; 1 mmol/l phenylmethylsulfonyl fluoride; 5 mg/ml leupeptin; and 5 mg/ml aprotinin. The protein content was quantitated by Bio-Rad protein assay (Bio-Rad, Hercules, CA, USA). The lysate was then boiled for 5 min for protein denaturation. Protein samples containing an equal amount (60 µg) of protein were separated by electrophoresis on 10% polyacrylamide-SDS gels and transferred onto nitrocellulose membranes. Non-specific binding of antibodies was blocked by incubation in Tris-buffered saline (TBS) containing 0.1% Tween-20 (TBS-T) and 5% non-fat milk for 1 h, followed by overnight incubation with rabbit anti-human BDNF (SC-546) (1:500) or rabbit anti-human TrkB (SC-12) (both from Santa Cruz Biotechnology, Santa Cruz, CA, USA) (1:500) or rabbit polyclonal IgG to GAPDH (1:1,000) at 4̊C. Rabbit anti-pAKT (1:500) and anti-AKT (1:1,000) were obtained from Cell Signaling Technology (Beverly, MA, USA). After washing, the membranes were incubated with horseradish peroxidase (HRP)-conjugated goat anti-rabbit secondary antibodies (1:5,000) at room temperature for 1 h. Immunoreactive bands were visualized using the ECL kit.

MTT assay

Cell proliferation was assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT; Sigma Chemical Co., St. Louis, MO, USA) assay. Cells were seeded in a 96-well plate at a density of 1.5×104 cells/well and allowed to adhere overnight. Transient transfection was performed using Lipofectamine 2000 (Invitrogen), according to the manufacturer's instructions. After 12, 24, 48, 72 and 120 h of transfection, 20 µl of 5 mg/µl MTT solution was added into each well of the plate and cells were incubated at 37̊C for an additional 4 h. The culture supernatant was removed, 150 µl of dimethyl sulfoxide (DMSO; Sigma) was added to dissolve the crystals. Spectrophotometric absorbance of the samples was measured by a microtiter plate reader at 570 nm. Considering that the initial cell concentrations of each group may be not identical, data at 12 h was set as the control. The cell proliferation rate was calculated as follows: Proliferation rate (%) = (sample absorbance/control absorbance) × 100. Each value represents 6 replicates, and each experiment was repeated 3 times.

Flow cytometry

HeLa cells were divided into non-transfected (Pnon), pGenesil-1-tranfected (P0) and positive experimental groups (PBDNF1). Cells in the Pnon, P0 and PBDNF1 groups were cultured in 6-well plates. After 48 h of incubation, a total of 1×106 cells were harvested, washed twice with PBS and fixed with cold 70% ethanol overnight at −20̊C. Fixed cells were centrifuged at 1,200 rpm and washed with PBS. Cells were stained in the dark with propidium iodide (PI; 50 µg/ml; Sigma) and 0.1% RNaseA (Invitrogen) at 4̊C for 1 h. Measurement of nuclear DNA was performed using flow cytometry (FCM). The results obtained reflected the percentage of cells in each phase of the cell cycle.

Hoechst 33258 staining

After 24 h of incubation in 6-well plates, the supernatant were removed. Then, the cells were fixed with cold methanol (0̊C) for 10 min and washed twice with PBS. Hoechst 33258 (1 mg/ml) was added and incubation for 30 min at 4̊C was carried out in a dark place. Cells were then washed with PBS and the apoptotic cells were observed using an Olympus BH-2 fluorescence microscope (magnification, ×200; Olympus, Tokyo, Japan). Cells stained bright blue were considered as apoptotic cells. Random cells (500) were counted and the apoptotic rate was calculated as follows: Hoechst 33258-stained cells/500 cells×100%.

Migration and invasion assays

Cell migration was quantified by the number of cells that directionally migrated through a 8-µm-pore polycarbonate filter (porosity, 8 µm; Costar, Appleton Woods, Birmingham, UK) in Boyden chambers. Briefly, the lower surface of the filter was coated with 10 mg of gelatin. HeLa cells were serum-starved overnight and resuspended in serum-free medium, and then 200 µl of the cell suspension was replaced onto the upper chamber of each well at a final concentration of 1×106 cells/ml. FBS (10%)-containing medium was added to the bottom chamber and cells were allowed to migrate for 6 h at 37̊C. Non-migrated cells on the upper membrane surface were removed with a cotton swab. The migratory cells attached to the lower membrane surfaces were fixed with 4% paraformaldehyde in PBS and stained with Wright staining. Cells were counted at a magnification of ×400 using standard microscopy, and the mean number of cells/field in 5 random fields was recorded. Triplicate filters were used and the experiments were repeated 3 times. The invasion assay was performed as above except that the upper surface of the filters were coated with 25 µg Matrigel, the cell concentration was adjusted to 2×105 cells/ml and the time was prolonged to 24 h.

Statistical analysis

SPSS for Windows (SPSS 13.0; SPSS, Inc., Chicago, IL, USA) was used to analyze the data. Data are expressed as the mean ± standard deviation (SD) from at least 3 separate experiments. Student's t-test was used to determine the significant difference between 2 groups. For comparison between 3 or more groups, one-way ANOVA test was used to determine statistical significance. The level of significance was set at p<0.05.

Results

High expression levels of BDNF and TrkB in HeLa cells

As shown in Fig. 2, RT-PCR and western blot analysis revealed that BDNF and TrkB were highly expressed in the HeLa cells at both the mRNA (Fig. 2A) and protein level (Fig. 2B). BDNF may provide autocrine support for TrkB-expressing HeLa cells as reported in the nervous system (24).

The recombinant eukaryotic BDNF siRNA expression vectors were successfully constructed and transfected into HeLa cells stably with high efficiencies

As shown in Fig. 3A-C, 4 experimental groups of plasmids pBDNF1, pBDNF2, pBDNF3 and pBDNF4, and the negative control group plasmid pScr were successfully generated. The multiple clone sites of pGenesil-1 are: HindIII, shRNA, BamHI, U6 promotor, EcoRI, SalI and XbaI. A specific SalI site was designed and synthesized into the shRNA sequences. Recombinant vectors were digested to a 400 bp fragment by SalI with the correct insertion. Most of the HeLa cells were effectively transfected with the recombinant BDNF siRNA expression vectors. The transient transfection efficiencies were ~60–80%. EGFP expression was not significantly reduced after the positive clones were generated (Fig. 3D-F).

siRNA1 is the most efficient sequence among the siRNAs examined

HeLa cells were divided into non-transfected (Pnon), pGenesil-1-tranfected (P0), pScr (PScr) and positive experimental groups (PBDNF1, PBDNF2, PBDNF3 and PBDNF4, respectively). As shown in Fig. 4, semi-quantitative RT-PCR and western blot analysis were performed when the positive clones had been established. The results revealed that siRNA1 (p<0.01) and siRNA4 (p<0.05) both led to significant reductions in BDNF expression without marked changes in GAPDH (GAPDH, data not shown), while pGenesil-1, siRNA2, siRNA3 and scramble siRNA barely altered the expression of BDNF (p>0.05). Compared with the level of BDNF in the Pnon group, siRNA1 led to nearly 99% reduction in the relative mRNA level, while a 58% decrease in the relative protein level was noted. siRNA1 was selected as the most efficient siRNA for use in the next experiments. Stable HeLa cell clones in the Pnon, P0 and PBDNF1 groups were used as experimental objects.

Downregulation of BDNF expression suppresses the proliferation of HeLa cells and induced cell cycle arrest in the G1 phase

FCM was performed to evaluate the cell cycle profile in the BDNF-siRNA-transfected HeLa cells. As shown in Fig. 5A-D, the percentage of cells in the G1 phase in the PBDNF1 group (70.73±4.15%) was much higher than that observed in the Pnon (55.33±5.64%) (p<0.01) and P0 group (57.47±2.98%) (p<0.05). Meanwhile, the percentage of cells in the G2 phase correspondingly decreased in the PBDNF1 group (11.15±2.88%) compared with the Pnon (24.83±3.67%) (p<0.01) and P0 group (21.28±5.38%) (p<0.05). The percentage of cells in the S phase had no significant change in the 3 groups (p>0.05) (Fig. 5A-D). Together, the results showed that BDNF-siRNA induced cell cycle arrest at the G1 phase and decreased the distribution of cells in the G2 phase. The growth curves of cells in the 3 groups as determined by the MTT assay showed significant growth inhibition of the BDNF-siRNA-transfected HeLa cells (Fig. 5E).

Downregulation of BDNF expression induces the apoptosis of HeLa cells

To analyze the involvement of BDNF in cell apoptosis, Hoechst 33258 staining was performed to investigate the apoptosis-related changes in cell morphology and to further evaluate the apoptotic rates in the Pnon, P0 and PBDNF1 groups. Observation with fluorescence microscopy (magnification, ×200) revealed a significant increase in the number of cells in the PBDNF1 group showing nuclear condensation and fragmentation which was not observed in the Pnon and P0 groups (Fig. 6A-C). As shown in Fig. 6D, the percentage of apoptotic/necrosis cells in the PBDNF1 group (27.14±4.57%) was much higher than those observed in the Pnon (0.84±0.39%) and the P0 group (2.68±1.02%) (p<0.01).

Downregulation of BDNF expression suppresses the migratory and invasive capabilities of HeLa cells

To evaluate the role of BDNF in cell migration and invasion, Transwell assay was used as a tool to determine the migratory and invasive capabilities of HeLa cells in the Pnon, P0 and PBDNF1 groups. As shown in the Fig. 7A-D, the number of cells in the P0 group that had migrated to the underside of the filters was similar to that of the cells in the Pnon group, whereas cells in the PBDNF1 group showed a significantly reduced migratory capability compared with the other 2 groups (p<0.01). Migrated cells/field in the PBDNF1 group (37±17), were significantly less than those in the Pnon (105±31) and P0 group (92±28). Migratory capability was impaired in the BDNF-knockdown cells compared with that noted in the non-transfected cells within at least a 2-fold reduction (p<0.01). As shown in Fig. 7E-H, similar to the migration assay, the number of invaded cells/field in the PBDNF1 group (24±12), was significantly less than those in the Pnon (85±26) and P0 groups (75±20). Invasive capability was significantly impaired in the BDNF-knockdown cells compared with non-transfected cells with at least a 3-fold reduction (p<0.01). Using stable cell lines expressing shRNA against BDNF, these results indicate that endogenous BDNF is essential for the ability of HeLa cells to migrate and invade normally.

Downregulation of BDNF expression inhibits the activation states of PI3K signaling

We next sought to study the effect of BDNF on the activation states of PI3K, a member of the signaling pathways known to be involved in mediating tumor cell proliferation and migration. AKT is a downstream target of PI3K-generated signals and becomes activated after phosphorylation of Ser473. The results showed that the level of phosphorylated AKT was significantly impaired in the BDNF-knockdown HeLa cells compared with the level noted in the non-transfected cells (Fig. 8).

Discussion

The present study was designed to investigate the biological role of BDNF in cervical carcinoma cell line HeLa. We showed that both BDNF and also its high-affinity receptor TrkB are expressed by HeLa cells. It has been documented that an autocrine loop exists between BDNF and TrkB in malignant tumors such as neuroblastoma (25), multiple myeloma (5) and ovarian cancer (26). In the autocrine loop, high expression of endogenous BDNF induced expression of TrkB. Hence, we postulated that BDNF may provide autocrine support for TrkB-expressing HeLa cells. Cervical carcinoma is the second leading cause of cancer morbidity and mortality among women worldwide, particularly in developing countries. It is well known that infection with high-risk human papilloma virus (HPV) is the predominant risk factor for cervical carcinoma. Although estrogens are a human carcinogen for a variety of cancers (2729), its effect on cervical carcinoma has not received much attention. However, it has been revealed that estrogens (E) and the estrogen receptor (ER) are overexpressed in cervical carcinoma tissue samples. The E/ER signaling pathway is essential for stimulating the expression of HPV E6 and E7 mRNA and cervical carcinoma cell proliferation, anchorage-independent growth and resistance to drug-induced apoptosis (30). The BDNF gene contains a sequence with close homology to the estrogen response element (ERE) and estrogen-ligand complexes are capable of binding this sequence and protecting it from DNase degradation (31). Ovariectomy in female rats was found to reduce BDNF expression and exogenous estrogen replacement restores it (32). Gonadectomized male rats show an increase in BDNF mRNA after estrogen treatment (33). Recent studies have also found that estrogen regulates BDNF expression via non-receptor-dependent mechanisms, involving disinhibition of GABA-ergic neurons (34). It remains unknown whether BDNF/TrkB is expressed in cervical carcinoma tissue samples, whether the expression level is correlated with the clinical stage, and whether BDNF-estrogen interaction is involved in the pathological mechanism of cervical carcinoma. We are currently conducting studies to address these issues.

In the present study, recombinant eukaryotic BDNF siRNA expression vectors were successfully constructed and transfected into HeLa cells. siRNA1 was selected as the most efficient siRNA used in the present study. siRNA-induced silencing of endogenous BDNF expression suppressed the proliferation of HeLa cells and induced cell cycle arrest in the G1 phase. These investigations are consistent with previous findings which demonstrated that BDNF promoted multiple myeloma (35), pancreatic cancer (36) and hepatocellular carcinoma cell (4) proliferation in a dose-dependent manner. Administration of BDNF to hepatocellular carcinoma cell lines induced significantly increased expression of cyclin D1 (4). Cyclin D1 is a key modulator in the G1 phase which controls the cell cycle switch to S phase. Upregulation of cyclin D1 by exogenous BDNF accelerated the G1 phase process and promoted tumor cell proliferation. Hence, we postulated that interference of endogenous BDNF expression in HeLa cells may alter the cell cycle profile in the G1 phase resulting in downregulation of the proliferation rate. However, BDNF/TrkB signal transduction in neuroblastoma cell lines SMS-KCN and SH-SY5Y, and retinoblastoma cell line RBL-15 are distinct from those observed in the present study. BDNF was barely able to alter cell proliferation or change cell cycle distribution (3,25). The data indicate that the role of BDNF in promoting proliferation is still controversial; much research must be carried out to fully elucidate the issue.

Hoechst 33258 staining assay revealed that interference of BDNF expression also increased cell apoptosis. Kurokawa et al (37) constructed a rat retinal ischemia-reperfusion injury model and found that exogenous BDNF intravitreally injected immediately after reperfusion decreased the number of caspase-2-positive cells in the retinal ganglion cell layer. Administration of K252α (a type of TrkB inhibitor) was able to activate caspase-3 and furthermore induce apoptosis in lung adenocarcinoma cell line A549 (38). BDNF may reduce neuron apoptosis by increasing the expression of the Bcl-2 anti-apoptosis protein and by inhibiting intracellular calcium overload (39). Anoikis is defined as apoptosis caused by lack of cell-matrix interactions (40), which has been suggested to act as a barrier to metastasis. Transgenic co-expression of BDNF/TrkB in rat intestinal epithelial cells resulted in complete transformation of the cells from normal cells to malignant cells. Transformed cells showed the capability of anti-apoptosis in systemic circulation and seeded to a distant place forming secondary tumors (14). A similar phenomenon was observed in ovarian cancer (41), BDNF and TrkB were found to be overexpressed in epithelial ovarian cancers and the BDNF/TrkB/PI3K-AKT signaling pathway may mediate anoikis suppression. Suppression of anoikis by BDNF may increase the survival of grafted Schwann cells in the case of therapy for spinal cord injury (42). In the present study, it was found that interference of BDNF expression significantly enhanced cell apoptosis and PI3K/AKT was involved in BDNF signal transduction in the cervical carcinoma cells. This evidence suggests that BDNF/TrkB functions as an anti-apoptosis signal to modulate tumor cell survival.

Metastasis is a major factor in the malignancy of cancers, and is often responsible for the failure of cancer treatment. Yu et al (41) clarified that the overexpression of BDNF/TrkB was significantly higher in greater omentum metastatic lesions and multicellular spheroids in ascites than in the corresponding primary lesions. In the present study, the migratory capability was impaired in the BDNF-knockdown cells compared with the non-transfected cells with at least a 2-fold reduction while the invasive capability was significantly attenuated with at least a 3-fold reduction. BDNF is a novel cytokine which induces the metastasis of HeLa cells as well as in multiple myeloma (35), and lung adenocarcinoma (43). Invasion of tumor cells into neighboring tissues requires degradation of the extracellular matrix by proteases. A recent study demonstrated that at least 2 discrete domains within the tissue-type plasminogen activator (tPA) gene promoter contribute to the BDNF response (44). Enhanced TrkB expression in neuroblastoma cells was associated with a significant increase in the secretion of subsets of matrix metalloproteinases (MMPs) (MMP-1, MMP-2 and MMP-9) and the urokinase-type plasminogen activator (uPA) and tPA, which resulted in an increase in their invasive capability via increased activity of proteolytic networks (45). These findings explain why BDNF/TrkB expression contributes to the migration and invasion of tumor cells.

In conclusion, we report the expression of BDNF/TrkB in human cervical carcinoma cell line HeLa. siRNA targeting the BDNF gene was used to prove that BDNF promotes HeLa cell proliferation and suppresses apoptosis, migration and invasion. In addition to the biological roles observed in the present study, BDNF was also found to act as a pro-angiogenic factor essential for the formation of tumor blood vessels (46). A new hypothesis suggests that tumors initiate their own innervations by the release of neurotrophic factors including nerve growth factor (NGF) and BDNF. By this process, which is termed neoneurogenesis, tumor cells come into close contact to nerve cells, forming a neuro-neoplastic synapse. Through these synapses, neurotransmitters are directly supplied to the tumor, which has impact on tumor growth and metastasis formation (47,48). Such findings provide hints to the possible mechanisms of the BDNF/TrkB signaling pathway in tumorigenesis which warrants further investigation for the possibility of alternative therapeutic targets.

Acknowledgements

The present study was supported by grants from the National Natural Sciences Foundation of China (no. 81272625 for C.-Y.S.; no. 81302042 to Z.-B.C.) and the Important New Drug Discovery (no. 2011ZX09302-002 to Y.H.).

Glossary

Abbreviations

Abbreviations:

BDNF

brain-derived neurotrophic factor

siRNA

small interfering RNA

Trk

tyrosine kinase

IL-6

interleukin-6

RNAi

RNA interference

dsRNA

double-stranded RNA

RISC

RNAi-induced silencing complex

FBS

fetal bovine serum

cDNA

complementary DNA

PBS

phosphate-buffered saline

TBS

Tris-buffered saline

MTT

3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide

DMSO

dimethyl sulfoxide

RT-PCR

reverse-transcriptase polymerase chain reaction

EGFP

enhanced green fluorescent protein

HPV

human papilloma virus

ER

estrogen receptor

MMPs

matrix metalloproteinases

uPA

urokinase-type plasminogen activator

tPA

tissue-type plasminogen activator

NGF

nerve growth factor

References

1 

Lewin GR and Barde YA: Physiology of the neurotrophins. Annu Rev Neurosci. 19:289–317. 1996. View Article : Google Scholar : PubMed/NCBI

2 

Novikov L, Novikova L and Kellerth JO: Brain-derived neurotrophic factor promotes axonal regeneration and long-term survival of adult rat spinal motoneurons in vivo. Neuroscience. 79:765–774. 1997. View Article : Google Scholar : PubMed/NCBI

3 

Lattanzio F, Carboni L, Carretta D, Candeletti S and Romualdi P: Treatment with the neurotoxic Aβ (25–35) peptide modulates the expression of neuroprotective factors Pin1, Sirtuin 1, and brain-derived neurotrophic factor in SH-SY5Y human neuroblastoma cells. Exp Toxicol Pathol. 68:271–276. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Guo D, Hou X, Zhang H, Sun W, Zhu L, Liang J and Jiang X: More expressions of BDNF and TrkB in multiple hepatocellular carcinoma and anti-BDNF or K252a induced apoptosis, supressed invasion of HepG2 and HCCLM3 cells. J Exp Clin Cancer Res. 30:972011. View Article : Google Scholar : PubMed/NCBI

5 

Pearse RN, Swendeman SL, Li Y, Rafii D and Hempstead BL: A neurotrophin axis in myeloma: TrkB and BDNF promote tumor-cell survival. Blood. 105:4429–4436. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Miknyoczki SJ, Wan W, Chang H, Dobrzanski P, Ruggeri BA, Dionne CA and Buchkovich K: The neurotrophin-trk receptor axes are critical for the growth and progression of human prostatic carcinoma and pancreatic ductal adenocarcinoma xenografts in nude mice. Clin Cancer Res. 8:1924–1931. 2002.PubMed/NCBI

7 

Jia S, Wang W, Hu Z, Shan C, Wang L, Wu B, Yang Z, Yang X and Lei D: BDNF mediated TrkB activation contributes to the EMT progression and the poor prognosis in human salivary adenoid cystic carcinoma. Oral Oncol. 51:64–70. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Stephan H, Zakrzewski JL, Bölöni R, Grasemann C, Lohmann DR and Eggert A: Neurotrophin receptor expression in human primary retinoblastomas and retinoblastoma cell lines. Pediatr Blood Cancer. 50:218–222. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Thiele CJ, Li Z and McKee AE: On Trk - the TrkB signal transduction pathway is an increasingly important target in cancer biology. Clin Cancer Res. 15:5962–5967. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Ricci A, Graziano P, Mariotta S, Cardillo G, Sposato B, Terzano C and Bronzetti E: Neurotrophin system expression in human pulmonary carcinoid tumors. Growth Factors. 23:303–312. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Rosean TR, Tompkins VS, Tricot G, Holman CJ, Olivier AK, Zhan F and Janz S: Preclinical validation of interleukin 6 as a therapeutic target in multiple myeloma. Immunol Res. 59:188–202. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Li Z, Zhang Y, Tong Y, Tong J and Thiele CJ: Trk inhibitor attenuates the BDNF/TrkB-induced protection of neuroblastoma cells from etoposide in vitro and in vivo. Cancer Biol Ther. 16:477–483. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Kupferman ME, Jiffar T, El-Naggar A, Yilmaz T, Zhou G, Xie T, Feng L, Wang J, Holsinger FC, Yu D, et al: TrkB induces EMT and has a key role in invasion of head and neck squamous cell carcinoma. Oncogene. 29:2047–2059. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Douma S, Van Laar T, Zevenhoven J, Meuwissen R, Van Garderen E and Peeper DS: Suppression of anoikis and induction of metastasis by the neurotrophic receptor TrkB. Nature. 430:1034–1039. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Sun CY, Hu Y, Wang HF, He WJ, Wang YD and Wu T: Brain-derived neurotrophic factor inducing angiogenesis through modulation of matrix-degrading proteases. Chin Med J. 119:589–595. 2006.PubMed/NCBI

16 

Sharp PA: RNA interference - 2001. Genes Dev. 15:485–490. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Hutvágner G and Zamore PD: RNAi: Nature abhors a double-strand. Curr Opin Genet Dev. 12:225–232. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Hannon GJ and Rossi JJ: Unlocking the potential of the human genome with RNA interference. Nature. 431:371–378. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Bernstein E, Caudy AA, Hammond SM and Hannon GJ: Role for a bidentate ribonuclease in the initiation step of RNA interference. Nature. 409:363–366. 2001. View Article : Google Scholar : PubMed/NCBI

20 

Hammond SM, Boettcher S, Caudy AA, Kobayashi R and Hannon GJ: Argonaute2, a link between genetic and biochemical analyses of RNAi. Science. 293:1146–1150. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Mousavi K and Jasmin BJ: BDNF is expressed in skeletal muscle satellite cells and inhibits myogenic differentiation. J Neurosci. 26:5739–5749. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Yan QS, Feng MJ and Yan SE: RNA interference-mediated inhibition of brain-derived neurotrophic factor expression increases cocaine's cytotoxicity in cultured cells. Neurosci Lett. 414:165–169. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Reynolds A, Leake D, Boese Q, Scaringe S, Marshall WS and Khvorova A: Rational siRNA design for RNA interference. Nat Biotechnol. 22:326–330. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Eaton MJ and Whittemore SR: Autocrine BDNF secretion enhances the survival and serotonergic differentiation of raphe neuronal precursor cells grafted into the adult rat CNS. Exp Neurol. 140:105–114. 1996. View Article : Google Scholar : PubMed/NCBI

25 

Nakamura Y, Suganami A, Fukuda M, Hasan MK, Yokochi T, Takatori A, Satoh S, Hoshino T, Tamura Y and Nakagawara A: Identification of novel candidate compounds targeting TrkB to induce apoptosis in neuroblastoma. Cancer Med. 3:25–35. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Streiter S, Fisch B, Sabbah B, Ao A and Abir R: The importance of neuronal growth factors in the ovary. Mol Hum Reprod. 22:3–17. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Clemons M and Goss P: Estrogen and the risk of breast cancer. N Engl J Med. 344:276–285. 2001. View Article : Google Scholar : PubMed/NCBI

28 

Burry K and Cain JM: Estrogen replacement therapy and risk of ovarian cancer in postmenopausal women. JAMA. 288:2538author reply 2539. 2002. View Article : Google Scholar : PubMed/NCBI

29 

Chen GG, Zeng Q and Tse GM: Estrogen and its receptors in cancer. Med Res Rev. 28:954–974. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Nair HB, Luthra R, Kirma N, Liu YG, Flowers L, Evans D and Tekmal RR: Induction of aromatase expression in cervical carcinomas: Effects of endogenous estrogen on cervical cancer cell proliferation. Cancer Res. 65:11164–11173. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Sohrabji F, Miranda RC and Toran-Allerand CD: Identification of a putative estrogen response element in the gene encoding brain-derived neurotrophic factor. Proc Natl Acad Sci USA. 92:11110–11114. 1995. View Article : Google Scholar : PubMed/NCBI

32 

Singh M, Meyer EM and Simpkins JW: The effect of ovariectomy and estradiol replacement on brain-derived neurotrophic factor messenger ribonucleic acid expression in cortical and hippocampal brain regions of female Sprague-Dawley rats. Endocrinology. 136:2320–2324. 1995. View Article : Google Scholar : PubMed/NCBI

33 

Solum DT and Handa RJ: Estrogen regulates the development of brain-derived neurotrophic factor mRNA and protein in the rat hippocampus. J Neurosci. 22:2650–2659. 2002.PubMed/NCBI

34 

Blurton-Jones M, Kuan PN and Tuszynski MH: Anatomical evidence for transsynaptic influences of estrogen on brain-derived neurotrophic factor expression. J Comp Neurol. 468:347–360. 2004. View Article : Google Scholar : PubMed/NCBI

35 

Hu Y, Sun CY, Wang HF, Guo T, Wei WN, Wang YD, He WJ, Wu T, Tan H and Wu TC: Brain-derived neurotrophic factor promotes growth and migration of multiple myeloma cells. Cancer Genet Cytogenet. 169:12–20. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Ketterer K, Rao S, Friess H, Weiss J, Büchler MW and Korc M: Reverse transcription-PCR analysis of laser-captured cells points to potential paracrine and autocrine actions of neurotrophins in pancreatic cancer. Clin Cancer Res. 9:5127–5136. 2003.PubMed/NCBI

37 

Kurokawa T, Katai N, Shibuki H, Kuroiwa S, Kurimoto Y, Nakayama C and Yoshimura N: BDNF diminishes caspase-2 but not c-Jun immunoreactivity of neurons in retinal ganglion cell layer after transient ischemia. Invest Ophthalmol Vis Sci. 40:3006–3011. 1999.PubMed/NCBI

38 

Perez-Pinera P, Hernandez T, García-Suárez O, de Carlos F, Germana A, Del Valle M, Astudillo A and Vega JA: The Trk tyrosine kinase inhibitor K252a regulates growth of lung adenocarcinomas. Mol Cell Biochem. 295:19–26. 2007. View Article : Google Scholar : PubMed/NCBI

39 

Liu Z, Ma D, Feng G, Ma Y and Hu H: Recombinant AAV-mediated expression of human BDNF protects neurons against cell apoptosis in Abeta-induced neuronal damage model. J Huazhong Univ Sci Technolog Med Sci. 27:233–236. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Frisch SM and Screaton RA: Anoikis mechanisms. Curr Opin Cell Biol. 13:555–562. 2001. View Article : Google Scholar : PubMed/NCBI

41 

Yu X, Liu L, Cai B, He Y and Wan X: Suppression of anoikis by the neurotrophic receptor TrkB in human ovarian cancer. Cancer Sci. 99:543–552. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Koda M, Someya Y, Nishio Y, Kadota R, Mannoji C, Miyashita T, Okawa A, Murata A and Yamazaki M: Brain-derived neurotrophic factor suppresses anoikis-induced death of Schwann cells. Neurosci Lett. 444:143–147. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Sinkevicius KW, Kriegel C, Bellaria KJ, Lee J, Lau AN, Leeman KT, Zhou P, Beede AM, Fillmore CM, Caswell D, et al: Neurotrophin receptor TrkB promotes lung adenocarcinoma metastasis. Proc Natl Acad Sci USA. 111:10299–10304. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Daniel PB, Lux W, Samson AL, Schleuning WD, Niego B, Weiss TW, Tjärnlund-Wolf A and Medcalf RL: Two conserved regions within the tissue-type plasminogen activator gene promoter mediate regulation by brain-derived neurotrophic factor. FEBS J. 274:2411–2423. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Hecht M, Schulte JH, Eggert A, Wilting J and Schweigerer L: The neurotrophin receptor TrkB cooperates with c-Met in enhancing neuroblastoma invasiveness. Carcinogenesis. 26:2105–2115. 2005. View Article : Google Scholar : PubMed/NCBI

46 

Usui T, Naruo A, Okada M, Hayabe Y and Yamawaki H: Brain-derived neurotrophic factor promotes angiogenic tube formation through generation of oxidative stress in human vascular endothelial cells. Acta Physiol. 211:385–394. 2014. View Article : Google Scholar

47 

Mancino M, Ametller E, Gascón P and Almendro V: The neuronal influence on tumor progression. Biochim Biophys Acta. 1816:105–118. 2011.PubMed/NCBI

48 

Entschladen F, Palm D, Niggemann B and Zaenker KS: The cancer's nervous tooth: Considering the neuronal crosstalk within tumors. Semin Cancer Biol. 18:171–175. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2017
Volume 37 Issue 5

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
Sun C, Chu Z, Huang J, Chen L, Xu J, Xu A, Li J and Hu Y: siRNA-mediated inhibition of endogenous brain‑derived neurotrophic factor gene modulates the biological behavior of HeLa cells Corrigendum in /10.3892/or.2021.7922 Retraction in /10.3892/or.2022.8438. Oncol Rep 37: 2751-2760, 2017
APA
Sun, C., Chu, Z., Huang, J., Chen, L., Xu, J., Xu, A. ... Hu, Y. (2017). siRNA-mediated inhibition of endogenous brain‑derived neurotrophic factor gene modulates the biological behavior of HeLa cells Corrigendum in /10.3892/or.2021.7922 Retraction in /10.3892/or.2022.8438. Oncology Reports, 37, 2751-2760. https://doi.org/10.3892/or.2017.5569
MLA
Sun, C., Chu, Z., Huang, J., Chen, L., Xu, J., Xu, A., Li, J., Hu, Y."siRNA-mediated inhibition of endogenous brain‑derived neurotrophic factor gene modulates the biological behavior of HeLa cells Corrigendum in /10.3892/or.2021.7922 Retraction in /10.3892/or.2022.8438". Oncology Reports 37.5 (2017): 2751-2760.
Chicago
Sun, C., Chu, Z., Huang, J., Chen, L., Xu, J., Xu, A., Li, J., Hu, Y."siRNA-mediated inhibition of endogenous brain‑derived neurotrophic factor gene modulates the biological behavior of HeLa cells Corrigendum in /10.3892/or.2021.7922 Retraction in /10.3892/or.2022.8438". Oncology Reports 37, no. 5 (2017): 2751-2760. https://doi.org/10.3892/or.2017.5569