Tetramethylpyrazine downregulates transcription of the CXC receptor 4 (CXCR4) via nuclear respiratory factor‑1 (Nrf‑1) in WERI‑Rb1 retinoblastoma cells

  • Authors:
    • Nandan Wu
    • Ying Yang
    • Na Yu
    • Yihui Wu
    • Xiaokun Han
    • Shuilian Chen
    • Jing Zhang
    • Xi Chen
    • Chuangran Wu
    • Meng Yang
    • Jin Qiu
    • Jian Ge
    • Keming Yu
    • Jing Zhuang
  • View Affiliations

  • Published online on: July 15, 2019     https://doi.org/10.3892/or.2019.7233
  • Pages: 1214-1224
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tetramethylpyrazine (TMP; an extract of the Chinese herbal medicine, Chuanxiong) has been shown to exert remarkable antiretinoblastoma (RB) effects. Based on our previous study, the target gene was found to be C‑X‑C chemokine receptor type 4 (CXCR4). CXCR4 is a prognostic marker in various types of cancer, but the exact mechanisms underlying the regulation of CXCR4 expression by TMP in WERI‑Rb1 cells have yet to be fully elucidated. In the present study, it was revealed that TMP significantly downregulated CXCR4 expression and inhibited CXCR4 promoter activity in WERI‑Rb1 cells, indicating that TMP inhibits CXCR4 expression in WERI‑Rb1 cells through transcriptional regulatory mechanisms. Among the numerous transcription factors involved in CXCR4 function, including Yin Yang 1 (YY1), nuclear respiratory factor‑1 (Nrf‑1), Krüppel‑like Factor 2 (KLF2), specificity protein 1 (SP1), and nuclear factor‑κB subunit 1 (NF‑κB1), only TMP led to a significant downregulation of Nrf‑1 expression. Chromatin immunoprecipitation assays further indicated that Nrf‑1 directly binds to the promoter region of CXCR4, and silencing Nrf‑1 via siRNA transfection notably inhibited CXCR4 expression in WERI‑Rb1 cells. In addition, the expression levels of both Nrf‑1 and CXCR4 increased concomitantly with WERI‑Rb1 cell density. Furthermore, the downregulation of Nrf‑1 and CXCR4 expression in RB by TMP was confirmed in vivo. Taken together, the results of the present study have uncovered a novel mechanism in which CXCR4 expression is regulated by Nrf‑1 in WERI‑Rb1 cells, thereby identifying novel potential targets for the treatment of RB, and providing evidence for the clinical application of TMP in adjuvant retinoblastoma therapy.

Introduction

Retinoblastoma (RB) is a pediatric retinal tumor and the most common primary intraocular childhood malignancy (13), with an incidence of ~1 in 15,000 live births (4). The occurrence and development of tumors seriously damages the function of the retina. In addition, the current effective treatment methods for RB, including local chemotherapy and radiotherapy, can lead to further retinal damage (5). Recent progress in RB diagnosis and treatment has pushed the goal of RB management from life-saving to eye and vision preservation. Numerous types of neuron exist in the retina, including ganglion cells, bipolar cells, horizontal cells, and photoreceptor cells. Light is collected by these neurons and transformed to electric signals that are transmitted to the brain, leading to the phenomenon of vision. Thus, ideal therapeutic agents for the adjuvant therapy of RB should possess the following properties: Inhibition of tumor cells, proliferation and protection of neurons against damage from excitotoxicity induced by tumor cells, and radiotherapy.

Chuanxiong has been used in Chinese traditional medicine for more than 2,000 years. Its bioactive component, tetramethylpyrazine (TMP), was extracted from Chuanxiong in 1973 (6). According to data from the China Food and Drug Administration (CFDA), there are 132 pharmaceutical factories that produce TMP injections or tablets in China (7). TMP has been widely used in the clinic to treat ischemia, cerebral infarction, degenerative diseases, and so on, albeit there are mild side effects (811). Furthermore, a paper published by our research group identified that TMP exerts neuroprotective effects, and revealed a mechanism underpinning TMP-mediated treatment that involved the inhibition of C-X-C chemokine receptor type 4 (CXCR4) expression in cerebral neurocytes and glioma cells (12). Accumulating evidence has confirmed that TMP can significantly attenuate chemotherapeutic multidrug resistance, and is able to inhibit the proliferation and metastasis of various types of cancer cells, including melanoma cells, lung cancer and gastric carcinoma cells (1318). More importantly, in our previous study, it was shown that TMP can significantly inhibit RB cell growth, and that CXCR4 is the target gene (19).

The chemokine receptor CXCR4 belongs to a large superfamily of G protein-coupled receptors with a 7-transmembrane spanning structure. CXCR4 is widely expressed in numerous types of cancerous tissues, including lung, kidney, breast, and retinal tumors, and is considered to serve a pivotal role in a number of biological processes that promote cancer growth and spreading, including angiogenesis, invasion, locomotion, extravasation, directional migration, homing, and cell survival (2026). Therefore, CXCR4 has been shown to be a prognostic marker in various types of cancer, including leukemia, breast cancer, and prostate cancer (2729). In addition, certain eye diseases have been shown to be associated with abnormal activation of CXCR4, such as primary open-angle glaucoma, angiogenesis, and eye inflammation (30,31). In our previous study, it was shown that CXCR4 is overexpressed in RB cells, and is a target gene of TMP to inhibit the growth of RB cells (19). Nevertheless, the mechanism underlying the regulation of CXCR4 expression by TMP in RB cells is not well defined.

The regulation of transcription is a vital process in all living organisms, and has a strong impact on gene expression. CXCR4 transcription is controlled by various mechanisms, depending on the cell type. In oral cancer cells, Krüppel-like factor 2 (KLF2) was shown to reduce CXCR4 promoter activity as a negative regulator of CXCR4 expression (32). In breast cancer cells, nuclear factor-κB (NF-κB) could directly bind to the CXCR4 promoter region (from −66 to +7 bp) and positively regulate CXCR4 expression, thus promoting tumor migration and metastasis (33). Our previous study also demonstrated that NF-κB and Nrf-1 transcriptionally co-regulate CXCR4 in corneal neovascularization (34). In addition, the transcription factors c-myc, Yin Yang 1 (YY1), specificity protein 1 (SP1), and hypoxia-inducible factor-1α (HIF-1α) have also been reported to be involved in the transcriptional regulation of CXCR4 (3537). However, the transcription factors regulating CXCR4 expression by TMP in RB cells remain unidentified.

Therefore, the aim of the present study was to investigate the possible transcriptional mechanism by which TMP mediates the downregulation of CXCR4 in RB cells in vitro and in vivo. The results obtained will lead to the identification of novel potential targets for the treatment of RB, and provide evidence for the clinical application of TMP in adjuvant therapy of RB.

Materials and methods

Ethics statement

Thirty-two female nude mice (4–5 weeks old), weighing between 16 and 20 g, were obtained from the Laboratory Animal Center, Sun Yat-sen University (Guangzhou, China). All experimental procedures were performed in accordance with the ARVO Statements for the Use of Animals in Ophthalmic and Vision Research, and were approved through the Institutional Animal Ethical Committee of Zhongshan Ophthalmic Center, Sun Yat-sen University (permit no. 2014-007). All animals were maintained in a room (temperature: 20–26°C, atmosphere: 40–70%) with a light schedule of alternating 12 h periods of light and dark, and received adequate amounts of food and water freely.

Cell culture

Cells of the human WERI-Rb1 RB cell line were purchased from the American Type Culture Collection (ATCC, Manassas, VA, USA) and maintained in Invitrogen® RPMI-1640 medium (Thermo Fisher Scientific, Inc, Waltham, MA, USA) supplemented with 10% FBS containing 100 U/ml penicillin and 100 µg/ml streptomycin in a humidified atmosphere of 5% CO2 at 37°C. Primary mouse retinal neurocytes were cultured as described previously (38). For co-culture assay, Transwell 12-well plates (0.4 µm pore size; BD Biosciences, Bedford, MA, USA) were used. Approximately 5×104 primary mouse retinal neurocytes were cultured for 6 days in the lower chamber filled with 1 ml medium, and subsequently 200 µl WERI-Rb1 cell suspension (7.5×105 cells/ml) was seeded in the upper chamber. Cells were maintained in RPMI-1640 and DMEM medium (1:1) supplemented with 10% FBS. For the cell density assay, the WERI-Rb1 cells were seeded in 6-well plates and maintained in 2 ml Complete™ RPMI-1640 medium at different densities (1×105, 2.5×105, 5.0×105, 7.5×105, and 106 cells/ml) for 24 h; subsequently, cell proteins were extracted for performing western blot assays. TMP was purchased from Sigma (now a brand of Merck KGaA; Darmstadt, Germany) and dissolved in component solvent (DMSO: Saline=1:1) to an appropriate concentration. The component solvent was applied as a control in all experiments.

Immunohistofluorescence assay

Cultured primary retinal neurocytes or WERI-Rb1 cells were fixed with ice-cold 4% paraformaldehyde for 15 min. For mono-staining (of Map-2 or CXCR4), the fixed cells were blocked with 10% normal goat serum for 30 min. For double-staining (CXCR4 and Nrf-1), the fixed cells were incubated with 0.1% Triton X-100 for 10 min, and then blocked with 10% normal goat serum for 30 min. Subsequently, cells were incubated overnight at 4°C with primary antibodies against Map-2 (1:100; cat. no. BM1243, Boster Biological Technology, Ltd., Wuhan, China), CXCR4 (1:100; cat. no. ab2074, Abcam, Cambridge, UK) and Nrf-1 (1:100; cat. no. sc-23624, Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA). Alexa Fluor 555 anti-rabbit lgG (1:500; cat. no. 4413, Cell Signaling Technology, Inc., Dallas, TX, USA) and Alexa Fluor 488 anti-goat lgG (1:500; cat. no. A-11055, Thermo Fisher Scientific, Inc.) were used as secondary antibodies, and nuclei were stained with DAPI. Images were captured using fluorescence microscopy (Leica Microsystems, Wetzlar, Germany; original magnification, ×100).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) assay

Total RNA from WERI-Rb1 cells following treatment with TMP was isolated with Invitrogen® TRIzol® reagent (Thermo Fisher Scientific, Inc.). The DNA contaminants in total RNA isolates were eliminated by treatment with DNAse I for 30 min at 37°C. One microgram of total RNA was subjected to reverse transcription using a PrimeScript™ RT Reagent kit (Takara Biotechnology Co., Ltd., Dalian, China) following the manufacturer's protocol. Expression levels of CXCR4 and Nrf-1 were measured by RT-qPCR with the Roche LightCycler® 480 System (Roche, Indianapolis, IN, USA). The PCR program was as follows: 94°C for 5 min, followed by 40 cycles of 94°C for 30 sec, 60°C for 30 sec, and 72°C for 30 sec. The following primer pairs were used: CXCR4, 5′-CTTATCCTGCCTGGTATTGTC-3′ (forward) and 5′-CAATGTAGTAAGGCAGCCAAC-3′ (reverse); Nrf-1, 5′-GGAATTCCCATGGAGGAACACGGAGTGAC-3′ (forward) and 5′-CGGGATCCCGTTATTTCCTTTTCAGTTGCTG-3′; (reverse); and β-actin, 5′-TCACCCACACTGTGCCCAT-3′ (forward) and 5′-TCTTTAATGTCACGCACGATT-3′ (reverse). Relative target gene expression levels (measured against β-actin) were calculated using the 2ΔΔCq method (39).

Western blot assay

Western blotting assays were performed according to a standard protocol. Briefly, whole proteins were extracted by using a radioimmunoprecipitation (RIPA) lysate kit (Beyotime Institute of Biotechnology, Jiangsu, China). The protein concentration was determined using the bicinchoninic acid (BCA) method. Equal amounts of protein (30 µg/well) were separated on an 8% sodium dodecyl sulfate (SDS)-polyacrylamide gel by electrophoresis, which were subsequently electrophoretically transferred to a polyvinylidene difluoride membrane. The membranes were blocked with 5% non-fat milk for 1 h at room temperature, then incubated with primary antibodies. The following primary antibodies were used: CXCR4 (1:500; cat. no. ab2074, Abcam), Nrf-1 (1:1,000; cat. no. ab175932, Abcam), YY1 (1:300; cat. no. sc-1703×, Santa Cruz Biotechnology, Inc.), KLF2 (1:300; cat. no. ab203591, Abcam), SP1 (1:300; cat. no. BA1402, Boster Biological Technology, Ltd.), NF-kB1 (1:500; cat. no. BA1297, Boster Biological Technology, Ltd.) and GAPDH (1:1,000; cat. no. 10494-1-AP, ProteinTech Group, Inc., Chicago, IL, USA). GAPDH served as the loading control. Protein bands were detected using an enhanced chemiluminescence detection system (EMD Millipore, Billerica, MA, USA).

Cell Counting Kit-8 (CCK-8) cell viability assay

The effect of TMP on the cell viability of retinal neurocytes and WERI-Rb1 cells in a co-culture system was measured using a CCK-8 assay (Dojindo Molecular Technologies, Inc., Kumamoto, Japan). Following treatment with 200 µM TMP or vehicle control for the relevant time periods (24, 48 or 72 h), WERI-Rb1 cells were transferred to new wells. Subsequently, CCK-8 reagent was added to each well, followed by incubation at 37°C for an additional 1 h. The absorbance of the product was measured at 450 nm using a fluorescence plate reader (Power Wave XS; BioTek China, Beijing, China). Cell viability was calculated by determining the optical density ratio of a treated culture over an untreated control.

RNA interference

The siRNA sequences used for targeted silencing of Nrf-1 and control sequences were as follows: Human Nrf-1 siRNA: 5′-CGTTAGATGAATATACTAC-3′ and the control, GGUUUGGCUGGGGUGUUAUdTdT. The oligos were purchased from Guangzhou RiboBio (Guangzhou, China). WERI-Rb1 cells (2×106 cells in 60 mm dishes) in good condition underwent lipidmediated transfection using Invitrogen® Lipofectamine™ RNAiMAX (Thermo Fisher Scientific, Inc.), as recommended by the manufacturer. The mRNA and protein expression levels of Nrf1 and CXCR4 were measured by RT-qPCR or western blotting assays, respectively, at 24 or 48 h after transfection.

Reporter and plasmid construction

A fragment spanning from −1,981 to +80 bp that included the CXCR4 promoter sequence was produced by PCR with the forward primer, 5′-GGGGTACCCCACACAATTCTGAATCCTGCCT-3′, and the common reverse primer, 5′-CCGCTCGAGCGGTCCAGATGCGGTGGCTACTG-3′. This fragment was fused to the promoter-less firefly luciferase gene of pGL3-Basic vector (Promega Corporation, Madison, WI, USA) to generate the constructed plasmid, named ‘PGL3-hCXCR4-pro’. PGL3-hCXCR4-pro was derived from the pGL3-Basic vector (Promega Corporation), in which human CXCR4 cDNA was inserted into pGL3-Basic vector using the restrictive digestion sites, KpnI and XhoI.

CXCR4 promoter-reporter assay

WERI-Rb1 cells (2×106 cells in 60 mm dishes) were transfected using Invitrogen® Lipofectamine™ 2000 (Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. The transfected plasmids contained 2 µg expression plasmid PGL3-hCXCR4-pro or pGL3-Basic vectors, and 100 ng Renilla luciferase reporter plasmid, pCMV-RL (Promega Corporation). The pCMV-RL plasmid encoding Renilla luciferase was included in all the samples to monitor transfection efficiency. At 24 h post-transfection, the levels of firefly and Renilla luciferase activity were measured sequentially from a single sample using the Dual-Glo Luciferase Assay system (Promega Corporation). The levels of firefly luciferase activity were normalized against Renilla luciferase activity.

Chromatin immunoprecipitation (ChIP) assay

ChIP assay was performed using the ChIP assay kit (Upstate Cell Signaling Solutions, Lake Placid, NY, USA), according to the manufacturer's protocol. Approximately 5×105 cells/ml were used for each assay. WERI-Rb1 cells were cross-linked by the addition of 1% formaldehyde for 10 min at room temperature, and the reaction was terminated upon the addition of glycine (final concentration, 0.125 M). Subsequently, cells were collected and incubated in 600 µl SDS lysis buffer containing protease inhibitors (2 µg/ml leupeptin, 2 µg/ml aprotinin, and 2 mM PMSF) for 10 min on ice. The samples were sonicated to yield fragments of chromatin of ~0.5 kb in length on ice (the duration of the sonication process was 8 min (6 sec ON-12 sec OFF) and the power was set at 400 W). After sonication, the lysate was centrifuged at 16,800 g for 10 min at 4°C. The supernatant was diluted in ChIP dilution buffer that included the protease inhibitors. A total 5% of the supernatant was saved as input DNA, and primary rabbit antibody Nrf-1 (Santa Cruz Biotechnology, Inc.) or rabbit normal IgG (Merck KGaA) was added to the supernatant and incubated overnight at 4°C with rotation. Following incubation with protein A-agarose, the immune complexes were eluted with elution buffer. A part of the captured immune-complex was subjected to western blotting analysis to detect whether the captured chromatins contained Nrf-1. Cross-linking was reversed by heating at 65°C overnight. RNA was subsequently degraded with RNase A for 30 min, whereas protein was degraded with proteinase K for 2 h. DNA was purified using an Ez-ChIP™ polypropylene spin column (Merck Millipore), and subjected to PCR amplification using the following primers for H-CXCR4-CHIP: 5′-GACCACCCGCAAACAGCAGG-3 (sense) and 5′-GCAGCCAACAAACTGAAGTTTC-3′ (antisense).

Murine xenograft model of retinoblastoma

All experimental procedures were approved by the Ethical Committee of Zhongshan Ophthalmic Center, Sun Yat-sen University (permit no. 2014-007). Female athymic nude mice aged 4–5 weeks old were obtained from the Laboratory Animal Center, Sun Yat-sen University. They were maintained in a standard environment with filter tops. After the WERI-Rb1 cells had been resuspended in PBS, the mice were anesthetized with 3% isoflurane delivered by mask, and 1×105 cells were injected into the vitreous cavity of the right eye using a Hamilton needle. All surgical procedures were performed under sterile conditions with a dissecting microscope; the left eyes were used as untreated controls. The eyes were observed on a weekly basis for tumor development. At 2 weeks after the mice had been xenotransplanted intravitreally with WERI-Rb1 cells, fundus photographs (Phoenix MicronIV™; Phoenix Research Labs, Pleasanton, CA, USA) were taken for all animals. All animals were subsequently randomly divided into two groups (n=16 mice in each group). Mice were anesthetized with 3% isoflurane delivered by mask, and each treatment group then received vitreous injections with TMP at a final dose of 200 µM, whereas the control group received vitreous injections with balanced salt solution. At 48 h following vitreous injection, the mice were euthanized by carbon dioxide inhalation (flow rate: 2–3 l/min) in a 10 l sealed box, and death of the mice was verified through several indicators, including absence of a heartbeat, respiratory arrest, and rigor mortis. The globes (n=4 for each group) were enucleated and fixed in 4% paraformaldehyde for 12 h. The eyeballs were subsequently processed into paraffin-embedded sections, and sequential meridian sections (6 µm thick) were created and stained with hematoxylin and eosin (H&E). To detect the mRNA and protein levels of Nrf-1 and CXCR4 following TPM treatment, tumor tissue was collected and analyzed by RT-qPCR and western blotting, respectively, after vitreous injection with TPM for 48 h (n=6 for each group).

Statistical analyses

All experiments were performed at least three times. Data are expressed as the mean ± SD. Differences between mean values were evaluated using two-tailed Student's t-test (for 2 groups) or analysis of variance (ANOVA; >2 groups). SPSS 21.0 software (IBM Corp., Armonk, NY, USA) was used for all statistical analyses. P<0.05 was considered to indicate a statistically significant value.

Results

TMP significantly promotes retinal neurocyte survival and suppresses WERI-Rb1 cell growth in co-culture systems

To assay the bioactivity of TMP on primary retinal neurocytes and WERI-Rb1 cells, a co-culture system was used to mimic RB physiological conditions. First, the primary rat retinal neurocytes were cultured 1 day after birth, and Map-2 staining was performed to verify the identity of these cells. As denoted by the green coloration in Fig. 1A, the majority of the cells were Map-2-positive cells. Subsequently, the primary retinal neurocytes were co-cultured with WERI-Rb1 cells, and treated with TMP or vehicle. At different time points following treatment, the cell viability of neurocytes and tumor cells were measured by CCK-8 assay. Our data showed that the viability of WERI-Rb1 cells was significantly inhibited following TMP treatment compared with the controls (at 24 h: Control, 1; and TMP, 0.82±0.04; at 48 h: Control, 1; TMP, 0.77±0.04; and at 72 h: Control, 1; TMP, 0.70±0.08) (P<0.05; Fig. 1B), results which were consistent with our previous study (19). Simultaneously, the viability of retinal neurocytes was significantly enhanced by TMP compared with controls (at 24 h: Control, 1; TMP, 1.10±0.04; at 48 h: Control, 1; TMP, 1.23±0.10; and at 72 h: Control, 1; TMP, 1.35±0.04) (P<0.05; Fig. 1C). These results indicated that TMP may serve as an ideal therapeutic agent for the adjuvant treatment of RB.

TMP downregulates CXCR4 expression in WERIRb1 cells

CXCR4 fulfills a critical role in fundamental aspects of cancer, including proliferation, migration, invasion, and metastases (2026). CXCR4 was found to be strongly expressed on the nuclear membrane of WERI-Rb1 cells (Fig. 2A). At 24 h after treatment with 200 µM TMP, total RNA and whole-cell lysates were extracted for RT-qPCR and western blot analyses, respectively. As shown in Fig. 2B, the level of CXCR4 mRNA in WERI-Rb1 cells following TMP treatment was significantly decreased compared with the control (control, 1; TMP, 0.63±0.14; P<0.05). The western blot assay also revealed that TMP treatment led to a notable attenuation of the CXCR4 protein level in WERI-Rb1 cells compared with the control (Fig. 2C). The relative quantification of CXCR4 expression is shown as a histogram in Fig. 2D (control, 1; TMP, 0.64±0.06; P<0.05).

To examine whether TMP regulated CXCR4 expression in WERI-Rb1 cells at the transcriptional level, a firefly luciferase reporter was constructed using the predicted CXCR4 promoter region (−1,981 to +80 bp) (Fig. 2E). Luciferase assay revealed that TMP significantly inhibited CXCR4 reporter activity (Control, 1; CXCR4Pro, 62.90±7.79; and CXCR4-Pro + TMP, 49.67±11.96) (P<0.05; P<0.001; Fig. 2F). These results indicated that TMP downregulates CXCR4 expression in WERI-Rb1 cells via inhibition of its transcription.

The transcription factor Nrf-1 is downregulated by TMP in WERI-Rb1 cells

To identify the transcription factors regulated by TMP in WERI-Rb1 cells, several reported transcription factors (i.e., KLF2, SP1, Nrf-1, YY1, and NF-κB1) that bind to the CXCR4 promoter were analyzed. After treatment with TMP for 24 or 48 h, whole-cell lysates of WERI-Rb1 were analyzed by western blotting. As shown in Fig. 3A, Nrf-1 expression was markedly downregulated in accordance with the expression of CXCR4 in WERI-Rb1 cells; however, the expression levels of KLF2, SP1, YY1, and NF-κB1 in WERI-Rb1 cells were not significantly altered following TMP treatment. The relative expression levels were then quantified by densitometry and normalized against GAPDH levels. As shown in Fig. 3B, Nrf-1 expression was significantly decreased by 27.62±7.62 and 33.65±9.52%, respectively, at 24 and 48 h following TMP treatment. Similarly, CXCR4 expression was decreased by 35.87±8.89 and 45.40±11.75%, respectively. For the expression of the other transcription factors, no significant differences were observed between the control group and the TMP-treated group (P>0.05). These results indicated that Nrf-1 may be involved in the transcriptional regulation of CXCR4 by TMP in WERI-Rb1 cells.

Nrf-1 directly binds to the CXCR4 promoter and positively regulates the expression of CXCR4 in WERI-Rb1 cells

To confirm whether Nrf-1 directly binds the CXCR4 promoter in WERI-Rb1 cells, ChIP assays were performed. Cells were sonicated to break up the chromatin molecules into ~0.5 kb fragments (Fig. 4A), followed by incubation with a rabbit Nrf-1 antibody or normal rabbit IgG. A portion of each immunoprecipitation reaction was subjected to western blot assay. Nrf-1 was shown to be readily detectable in the samples incubated with Nrf-1 antibody, but not normal rabbit IgG (Fig. 4B). In addition, the precipitated DNA was subjected to PCR amplification using primers designed to amplify a 200 bp fragment of the CXCR4 promoter region flanking the ATF site. As shown in Fig. 4C, a 200 bp band was only detected in DNA incubated with anti-Nrf-1 and the input DNA. These results indicated that Nrf-1 directly binds to the promoter region of CXCR4.

To further verify that Nrf-1 exerts a crucial role in the regulation of CXCR4 in WERI-Rb1 cells, siRNA transfection was performed to silence Nrf-1 expression. As shown in Fig. 4D, the mRNA expression of CXCR4 was significantly decreased after Nrf-1 silencing. Western blot analysis also showed that silencing Nrf-1 notably inhibited CXCR4 protein expression in WERI-Rb1 cells (Fig. 4E). The relative quantification of Nrf-1 and CXCR4 protein expression is presented in histogram form in Fig. 4F. The protein expression of Nrf-1 was decreased by 42.19±11.46% after siRNA silencing. Accordingly, CXCR4 was also decreased by 35.42±11.98% (P<0.05). These data further suggested that Nrf-1 is involved in the positive transcriptional regulation of CXCR4 in WERI-Rb1 cells.

Nrf-1 and CXCR4 expression are increased in accordance with cell density

Our previous study demonstrated that CXCR4 downregulation by TMP is associated with cell density in WERI-Rb1 cells (19). Therefore, in the present study, the protein levels of Nrf-1 and CXCR4 following culture (24 h) of WERI-Rb1 cells plated at different densities (1×105, 2.5×105, 5.0×105, 7.5×105, and 106 cells/ml) were measured. To verify the expression location of Nrf-1 and CXCR4, and that Nrf-1 and CXCR4 are co-expressed in WERI-Rb1 cells, double immunohistofluorescence staining was performed. The results demonstrated that Nrf-1 was expressed in the nucleus, and that CXCR4 was expressed predominantly on the nuclear membrane, with moderate staining evident in the cytoplasm (Fig. 5A). Western blot analysis demonstrated that Nrf-1 and CXCR4 protein expression was increased in accordance with cell density (Fig. 5B). The relative expression of Nrf-1 and CXCR4 in WERI-Rb1 cells was also quantified by densitometry. The average ratio of Nrf-1 or CXCR4 to GAPDH in WERI-Rb1 cells at the lowest cell density investigated (1×105 cells/ml) was defined as 1. As shown in Fig. 5C, relative quantification revealed that Nrf-1 in WERI-Rb1 cells was significantly upregulated with increasing cell density, compared with cells at the lowest density (1×105 cells/ml, 1; 2.5×105 cells/ml, 1.30±0.05; 5×105 cells/ml, 1.58±0.14; 7.5×105 cells/ml, 1.82±0.10; and 106 cells/ml, 2.04±0.10) (P<0.05). The expression of CXCR4 was also significantly increased in cells, in essentially a stepwise manner, with increasing cell density (1×105 cells/ml, 1; 2.5×105 cells/ml, 1.26±0.10; 5×105 cells/ml, 1.47±0.21; 7.5×105 cells/ml, 1.83±0.09; and 106 cells/ml, 2.09±0.05) (P<0.05; Fig. 5D). These results demonstrated that the expression pattern of Nrf-1 is similar to that of CXCR4, further indicating that the expression of CXCR4 in WERI-Rb1 cells is regulated by the transcription factor Nrf-1.

Nrf-1 and CXCR4 are downregulated by TMP in vivo

Based on the in vitro results, an RB orthotopic xenotransplantation model was established to validate the TMP-mediated inhibition of CXCR4 by the transcription factor Nrf-1. Fundus photographs were taken for all animals aged over 2 weeks. Fig. 6A shows fundus photographs of normal mice eyes (left panel) and mice eyes with RB (center panel; the white arrowheads indicate RB tumors); H&E staining revealed that the cell mass of the RB is located in the vitreous cavity (right panel; the white arrowheads indicate RB tumors). Subsequently, the mice were divided into two groups receiving either vitreous injection of TMP, or vehicle. At 48 h following treatment, total RNA and protein lysates of RB were obtained for RT-qPCR and western blot analyses. As shown in Fig. 6B, the mRNA expression levels of both Nfr-1 and CXCR4 were significantly decreased in retinoblastoma treated with TMP, compared with controls (for Nrf-1: Control, 1; TMP-treated, 0.23±0.06; for CXCR4: Control, 1; TMP-treated, 0.47±0.22) (P<0.05). Similarly, the altered Nfr-1 and CXCR4 protein expression levels were consistent with the changes in mRNA levels (Fig. 6C). The relative quantification of Nrf-1 and CXCR4 protein expression is presented in histogram form in Fig. 6D (for Nrf-1: Control, 1; TMP-treated, 0.66±0.08; for CXCR4: Control, 1; TMP-treated, 0.40±0.04) (P<0.05). These data further demonstrated that CXCR4 is downregulated by TMP through Nrf-1 in vivo.

Discussion

TMP is commonly used in the clinic to treat vascular diseases and neurodegenerative diseases with mild side effects (811). According to our previous study, TMP possesses a remarkable anti-RB effect (19); however, its underlying molecular mechanism has not been fully elucidated. Currently, TMP is only used in certain Oriental countries, including China and Korea. Therefore, clarifying the anti-RB mechanism of TMP will extend its clinical therapeutic application in medical practice.

In the present study, RB cells and a retinal neurocyte co-culture system were used to mimic the RB ocular physiological environment, and the data obtained revealed that TMP is able to significantly inhibit the viability of WERIRb1 cells and to promote retinal neurocyte survival (Fig. 1). Furthermore, accumulating evidence has confirmed that combining TMP with other treatments could significantly attenuate the multidrug resistance of chemotherapy (1517). Therefore, TMP may be an ideal chemical for the adjuvant treatment of RB. CXCR4 is the target gene of TMP, which acts as an inhibitor of RB cell growth. In addition to the downregulation of mRNA and protein expression of CXCR4 by TMP, the present study also disclosed that the activity of the CXCR4 promoter was significantly reduced following TMP treatment in WERI-Rb1 cells, suggesting that TMP inhibits CXCR4 expression in WERI-Rb1 cells via transcriptional regulatory mechanisms.

The regulation of transcription is a vital process in all living organisms, which exerts a strong impact on gene expression. Previous studies in various cell lines have reported that the CXCR4 promoter is regulated by several transcription factors, including KLF2, SP1, Nrf-1, YY1, NF-κB1, and so on (3236). In the present study, however, TMP was only found to mediate the downregulation of Nrf-1. In addition, ChIP assays confirmed that Nrf-1 directly bound to the CXCR4 promoter region, and silencing Nrf-1 notably decreased CXCR4 expression. Furthermore, Nrf-1 and CXCR4 expression were downregulated by TMP in an RB model. These results strongly suggest that TMP downregulates the transcription of CXCR4 via Nrf-1 in WERI-Rb1 cells.

A previously published study by our research group demonstrated that NF-κB and Nrf-1 transcriptionally co-regulate CXCR4 expression in human umbilical vein endothelial cells and the alkali-burn cornea (34). However, in the present study, TMP was found not to affect NF-κB1 expression in WERI-Rb1 cells. Therefore, these results suggested that CXCR4 is regulated by different transcriptional factors in different cells. In addition, a previous study also demonstrated that Nrf-1 expression correlates with that of CXCR4 during retinal development (40). CXCR4 and Nrf-1 are expressed in the postnatal rat retina, and are silenced together in the adult rat retina. Furthermore, in the oxygen-induced retinopathy rat model, retinal hypoxia was able to concurrently induce the upregulation of CXCR4 and Nrf-1 expression. Therefore, the transcriptional mechanism of CXCR4 in WERI-Rb1 cells may be similar to that found in normal retinal tissue. RB is a malignant intraocular tumor derived from the retina; therefore, it is possible to speculate that transcription factors of CXCR4 are specific to host tissue.

In our previous study, it was shown that TMP downregulation of CXCR4 in WERI-Rb1 cells is sensitive to cell density (19). Interestingly, Nrf-1 and CXCR4 expression were found to be significantly increased in parallel with cell density in the present study. Nrf-1, a member of the basic leucine-zipper family of proteins (41,42), was first identified as a transcription factor that regulates mitochondria-associated genes, including p43, CREB, p53, and Stat3 (43,44). A further study also showed that Nrf-1 is able to mediate reactive oxygen species (ROS) and play a vital role in regulating cell metabolism and respiration (45). Therefore, these studies suggest that increasing the cell density induces the upregulation of Nrf-1, which subsequently promotes the transcription of CXCR4 in retinoblastoma cells.

In addition, previous studies demonstrated that Nrf-1 is involved in neural ischemic diseases. For example, the mRNA content of Nrf-1 rapidly increased 3–6 h following an ischemic-tolerant state (46). Nrf-1 was significantly upregulated in the retina of an ischemia-reperfusion surgery model, and decreased after treatment with lithium, a neuroprotective reagent (38). TMP has been used in patients with neural diseases, and our research group previously verified that TMP is able to downregulate CXCR4 expression in cerebral neurocytes, which inhibits increases in somatic Ca2+ and decreases glutamate release to protect neurons (12). Therefore, it is possible to speculate that TMP exerts a neuroprotective action by inhibiting Nrf-1 expression, with the subsequent decrease in CXCR4 expression.

In addition, Nrf-1 is known to regulate the expression of other genes, and our previous study demonstrated that Nrf-1 binds the promoter of ligase IV, which regulates DNA repair in retinal neurocytes (38). Therefore, in WERI-Rb1 cells, the Nrf-1-CXCR4 pathway might be one of several complicated signaling pathways that respond to TMP. Further investigation of this subject is required in the future.

In conclusion, the results obtained in the present study have helped to further elucidate the anti-RB molecular mechanism of TMP. The transcription factor Nrf-1 directly binds to the promoter sequence of CXCR4, and TMP downregulates CXCR4 expression at the transcriptional level via Nrf-1, thereby inhibiting the growth of WERI-Rb1 cells. Our study has identified novel potential targets for the treatment of RB, and provides evidence for the clinical application of TMP in adjuvant therapy of retinoblastoma.

Acknowledgements

Not applicable.

Funding

This study was supported by grants from the National Natural Science Foundation of China (project nos. 81470626 and 81670848).

Availability of data and materials

The datasets used and/or analyzed during the current study would be available from the corresponding author upon reasonable request.

Authors' contributions

NW and YY designed the study, performed the experiments, collected and analyzed the data, and wrote the manuscript. NY, YW, XH, and JQ performed experiments and analyzed the data. SC, JZ, XC, CW, and MY assisted in collecting data and assembling the figures. JG assisted in designing the study, and provided professional suggestions. KY and JZ designed the study, analyzed data, and wrote the manuscript. NW and JZ revised the manuscript in terms of its important intellectual content. All authors read and approved the manuscript, and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

All experimental procedures were performed in accordance with the ARVO statements for the Use of Animals in Ophthalmic and Vision Research, and were approved through the Institutional Animal Ethical Committee of Zhongshan Ophthalmic Center, Ethical Committee of Zhongshan Ophthalmic Center, Sun Yat-Sen University (permit no. 2014-007).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Chintagumpala M, ChevezBarrios P, Paysse EA, Plon SE and Hurwitz R: Retinoblastoma: Review of current management. Oncologist. 12:1237–1246. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Houston SK, Murray TG, Wolfe SQ and Fernandes CE: Current update on retinoblastoma. Int Ophthalmol Clin. 51:77–91. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Devesa SS: The incidence of retinoblastoma. Am J Ophthalmol. 80:263–265. 1975. View Article : Google Scholar : PubMed/NCBI

4 

Tamboli A, Podgor MJ and Horm JW: The incidence of retinoblastoma in the United States: 1974 through 1985. Arch Ophthalmol. 108:128–132. 1990. View Article : Google Scholar : PubMed/NCBI

5 

Francis JH, Brodie SE, Marr B, Zabor EC, MondesireCrump I and Abramson DH: Efficacy and toxicity of intravitreous chemotherapy for retinoblastoma: Fouryear experience. Ophthalmology. 124:488–495. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Beijing Pharmaceutical Industry Research Institute, . Active ingredient research of Chuanxiong: I. Extraction, isolation and structural identification of ligustrazine. Natl Med J Chin. 7:420–421. 1977.(In Chinese).

7 

China Food and Drug Administration. http://app1.sfda.gov.cn/datasearch/face3/base.jsp?tableId=25&tableName=TABLE25&title=%B9%FA%B2%FA%D2%A9%C6%B7&bcId=124356560303886909015737447882June 22–2019

8 

You JM, Zhang ZG, Lin C and Ji Y: Ischemic stroke and the regulation of syndrome of traditional Chinese medicine compound efficacy TMP combined. Chin Archives of Tradit Chin Med. 12:2666–2668. 2010.(In Chinese).

9 

Tang Z, Wang S and Lin Y: Progress in protective effects of tetramethylpyrazine on diabetes complications in nervous system and possible mechanisms. Chin J Pharmacol Toxicol. 25:114–118. 2011.(In Chinese).

10 

Chen Y and Liu M: Systemic evaluation of Security of ligustrazine for treatment of cerebral infarction. Chin J Clin Rehab. 07:1299–1301. 2004.(In Chinese).

11 

Yang XG and Jiang C: Ligustrazine as a salvage agent for patients with relapsed or refractory non-Hodgkin's lymphoma. Chin Med J (Engl). 123:3206–3211. 2010.(In Chinese). PubMed/NCBI

12 

Chen Z, Pan X, Georgakilas AG, Chen P, Hu H, Yang Y, Tian S, Xia L, Zhang J, Cai X, et al: Tetramethylpyrazine (TMP) protects cerebral neurocytes and inhibits glioma by down regulating chemokine receptor CXCR4 expression. Cancer Lett. 336:281–289. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Hu J and Li Q: Effect of tetramethypyrazine on the proliferation and apoptosis of lung cancer cell line of a549 and its mechanism. Med J Wuhan Univ. 3:50–51. 2010.(In Chinese).

14 

Lu X, Wang Z and Chen J: Tetramethylpyrazine induced expression of iNOS and NO to inhibit lung metastasis of melanoma bearing mice. Chin J Mod Med. 16:3208–3210. 2006.(In Chinese).

15 

Hua Z and Zhou Y: TMP inhibits the proliferation of gastric carcinoma cells and endothelial cells. J Changchun Univ Tradit Chin Med. 25:20–22. 2009.(In Chinese).

16 

Wang S, Lei T and Zhang M: The reversal effect and its mechanisms of Tetramethylpyrazine on multidrug resistance in human bladder cancer. PLoS One. 11:e01577592016. View Article : Google Scholar : PubMed/NCBI

17 

Zhang Y, Liu X, Zuo T, Liu Y and Zhang JH: Tetramethylpyrazine reverses multidrug resistance in breast cancer cells through regulating the expression and function of P-glycoprotein. Med Oncol. 29:534–538. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Wang XB, Wang SS, Zhang QF, Liu M, Li HL, Liu Y, Wang JN, Zheng F, Guo LY and Xiang JZ: Inhibition of tetramethylpyrazine on P-gp, MRP2, MRP3 and MRP5 in multidrug resistant human hepatocellular carcinoma cells. Oncol Rep. 23:211–215. 2010.PubMed/NCBI

19 

Wu N, Xu L, Yang Y, Yu N, Zhang Z, Chen P, Zhang J, Tang M, Yuan M, Ge J, et al: Tetramethylpyrazinemediated regulation of CXCR4 in retinoblastoma is sensitive to cell density. Mol Med Rep. 15:2481–2488. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Su L, Zhang J, Xu H, Wang Y, Chu Y, Liu R and Xiong S: Differential expression of CXCR4 is associated with the metastatic potential of human non-small cell lung cancer cells. Clin Cancer Res. 11:8273–8280. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Smith MC, Luker KE, Garbow JR, Prior JL, Jackson E, PiwnicaWorms D and Luker GD: CXCR4 regulates growth of both primary and metastatic breast cancer. Cancer Res. 64:8604–8612. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Schrader AJ, Lechner O, Templin M, Dittmar KE, Machtens S, Mengel M, ProbstKepper M, Franzke A, Wollensak T, Gatzlaff P, et al: CXCR4/CXCL12 expression and signalling in kidney cancer. Br J Cancer. 86:1250–1256. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Liang Z, Brooks J, Willard M, Liang K, Yoon Y, Kang S and Shim H: CXCR4/CXCL12 axis promotes VEGFmediated tumor angiogenesis through Akt signaling pathway. Biochem Biophys Res Commun. 359:716–722. 2007. View Article : Google Scholar : PubMed/NCBI

24 

Zhou Y, Larsen PH, Hao C and Yong VW: CXCR4 is a major chemokine receptor on glioma cells and mediates their survival. J Biol Chem. 277:49481–49487. 2002. View Article : Google Scholar : PubMed/NCBI

25 

Müller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME, McClanahan T, Murphy E, Yuan W, Wagner SN, et al: Involvement of chemokine receptors in breast cancer metastasis. Nature. 410:50–56. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Yin X, Liu Z, Zhu P, Wang Y, Ren Q, Chen H and Xu J: CXCL12/CXCR4 promotes proliferation, migration, and invasion of adamantinomatous craniopharyngiomas via PI3K/AKT signal pathway. J Cell Biochem. 120:9724–9736. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Jung SJ, Kim CI, Park CH, Chang HS, Kim BH, Choi MS and Jung HR: Correlation between chemokine receptor CXCR4 expression and prognostic factors in patients with prostate cancer. Korean J Urol. 52:607–611. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Spoo AC, Lübbert M, Wierda WG and Burger JA: CXCR4 is a prognostic marker in acute myelogenous leukemia. Blood. 109:786–791. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Zhang Z, Ni C, Chen W, Wu P, Wang Z, Yin J, Huang J and Qiu F: Expression of CXCR4 and breast cancer prognosis: A systematic review and meta-analysis. BMC Cancer. 14:492014. View Article : Google Scholar : PubMed/NCBI

30 

Yu N, Zhang Z, Chen P, Zhong Y, Cai X, Hu H, Yang Y, Zhang J, Li K, Ge J, et al: Tetramethylpyrazine (TMP), an active ingredient of chinese herb medicine Chuanxiong, attenuates the degeneration of trabecular meshwork through SDF1/CXCR4 axis. PLoS One. 10:e01330552015. View Article : Google Scholar : PubMed/NCBI

31 

Cai X, Chen Z, Pan X, Xia L, Chen P, Yang Y, Hu H, Zhang J, Li K, Ge J, et al: Inhibition of angiogenesis, fibrosis and thrombosis by tetramethylpyrazine: Mechanisms contributing to the SDF-1/CXCR4 axis. PLoS One. 9:e881762014. View Article : Google Scholar : PubMed/NCBI

32 

Uchida D, Onoue T, Begum NM, Kuribayashi N, Tomizuka Y, Tamatani T, Nagai H and Miyamoto Y: Vesnarinone downregulates CXCR4 expression via upregulation of Kruppel-like factor 2 in oral cancer cells. Mol Cancer. 8:622009. View Article : Google Scholar : PubMed/NCBI

33 

Helbig G, Christopherson KW II, Bhat-Nakshatri P, Kumar S, Kishimoto H, Miller KD, Broxmeyer HE and Nakshatri H: NFkappaB promotes breast cancer cell migration and metastasis by inducing the expression of the chemokine receptor CXCR4. J Biol Chem. 278:21631–21638. 2003. View Article : Google Scholar : PubMed/NCBI

34 

Tang MJ, Yang Y, Yu JZ, Qiu J, Chen P, Wu YH, Wang QY, Xu ZJ, Ge J, Yu K and Zhuang J: Tetramethylpyrazine in a murine alkaliburn model blocks NFκB/NRF1/CXCR4-signaling-induced corneal neovascularization. Invest Ophth Vis Sci. 59:2133–2141. 2018. View Article : Google Scholar

35 

Moriuchi M, Moriuchi H, Margolis DM and Fauci AS: USF/c-Myc enhances, while Yin-Yang 1 suppresses, the promoter activity of CXCR4, a coreceptor for HIV-1 entry. J Immunol. 162:5986–5992. 1999.PubMed/NCBI

36 

Wegner SA, Ehrenberg PK, Chang G, Dayhoff DE, Sleeker AL and Michael NL: Genomic organization and functional characterization of the chemokine receptor CXCR4, a major entry co-receptor for human immunodeficiency virus type 1. J Biol Chem. 273:4754–4760. 1998. View Article : Google Scholar : PubMed/NCBI

37 

Schioppa T, Uranchimeg B, Saccani A, Biswas SK, Doni A, Rapisarda A, Bernasconi S, Saccani S, Nebuloni M, Vago L, et al: Regulation of the chemokine receptor CXCR4 by hypoxia. J Exp Med. 198:1391–1402. 2003. View Article : Google Scholar : PubMed/NCBI

38 

Yang Y, Wu N, Tian S, Li F, Hu H, Chen P, Cai X, Xu L, Zhang J, Chen Z, et al: Lithium promotes DNA stability and survival of ischemic retinal neurocytes by upregulating DNA ligase IV. Cell Death Dis. 7:e24732016. View Article : Google Scholar : PubMed/NCBI

39 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using realtime quantitative PCR and the 2(Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

40 

Chen P, Cai XX, Yang Y, Chen Z, Qiu J, Yu N, Tang MJ, Wang QY, Ge J, Yu K and Zhuang J: Nuclear respiratory factor-1 (NRF-1) regulates transcription of the CXC receptor 4 (CXCR4) in the rat retina. Invest Ophth Vis Sci. 58:4662–4669. 2017. View Article : Google Scholar

41 

Chan JY, Kwong M, Lu R, Chang J, Wang B, Yen TS and Kan YW: Targeted disruption of the ubiquitous CNC-bZIP transcription factor, Nrf-1, results in anemia and embryonic lethality in mice. EMBO J. 17:1779–1787. 1998. View Article : Google Scholar : PubMed/NCBI

42 

Chan JY, Cheung MC, Moi P, Chan K and Kan YW: Chromosomal localization of the human NF-E2 family of bZIP transcription factors by fluorescence in situ hybridization. Hum Genet. 95:265–269. 1995. View Article : Google Scholar : PubMed/NCBI

43 

LeighBrown S, Enriquez JA and Odom DT: Nuclear transcription factors in mammalian mitochondria. Genome Biol. 11:2152010. View Article : Google Scholar : PubMed/NCBI

44 

Huo L and Scarpulla RC: Mitochondrial DNA instability and peri-implantation lethality associated with targeted disruption of nuclear respiratory factor 1 in mice. Mol Cell Biol. 21:644–654. 2001. View Article : Google Scholar : PubMed/NCBI

45 

Zhang J, Gu JY, Chen ZS, Xing KC and Sun B: Astragalus polysaccharide suppresses palmitate-induced apoptosis in human cardiac myocytes: The role of Nrf1 and antioxidant response. Int J Clin Exp Pathol. 8:2515–2524. 2015.PubMed/NCBI

46 

Stetler RA, Leak RK, Yin W, Zhang LL, Wang SP, Gao YQ and Chen J: Mitochondrial biogenesis contributes to ischemic neuroprotection afforded by LPS pre-conditioning. J Neurochem. 123 (Suppl):S125–S137. 2012. View Article : Google Scholar

Related Articles

Journal Cover

September-2019
Volume 42 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
Wu N, Yang Y, Yu N, Wu Y, Han X, Chen S, Zhang J, Chen X, Wu C, Yang M, Yang M, et al: Tetramethylpyrazine downregulates transcription of the CXC receptor 4 (CXCR4) via nuclear respiratory factor‑1 (Nrf‑1) in WERI‑Rb1 retinoblastoma cells. Oncol Rep 42: 1214-1224, 2019
APA
Wu, N., Yang, Y., Yu, N., Wu, Y., Han, X., Chen, S. ... Zhuang, J. (2019). Tetramethylpyrazine downregulates transcription of the CXC receptor 4 (CXCR4) via nuclear respiratory factor‑1 (Nrf‑1) in WERI‑Rb1 retinoblastoma cells. Oncology Reports, 42, 1214-1224. https://doi.org/10.3892/or.2019.7233
MLA
Wu, N., Yang, Y., Yu, N., Wu, Y., Han, X., Chen, S., Zhang, J., Chen, X., Wu, C., Yang, M., Qiu, J., Ge, J., Yu, K., Zhuang, J."Tetramethylpyrazine downregulates transcription of the CXC receptor 4 (CXCR4) via nuclear respiratory factor‑1 (Nrf‑1) in WERI‑Rb1 retinoblastoma cells". Oncology Reports 42.3 (2019): 1214-1224.
Chicago
Wu, N., Yang, Y., Yu, N., Wu, Y., Han, X., Chen, S., Zhang, J., Chen, X., Wu, C., Yang, M., Qiu, J., Ge, J., Yu, K., Zhuang, J."Tetramethylpyrazine downregulates transcription of the CXC receptor 4 (CXCR4) via nuclear respiratory factor‑1 (Nrf‑1) in WERI‑Rb1 retinoblastoma cells". Oncology Reports 42, no. 3 (2019): 1214-1224. https://doi.org/10.3892/or.2019.7233