Home CSF-1R promotes vasculogenic mimicry via epithelial-mesenchymal transition in nasopharyngeal carcinoma cells
Article Open Access

CSF-1R promotes vasculogenic mimicry via epithelial-mesenchymal transition in nasopharyngeal carcinoma cells

  • Huaqing Mo , Yanru Lv , Shan Gao , Zenan Chen , Zhiyong Xu , Jingyi Shen , Shu Zhou , Mengjie Yin , Yanyan Xie EMAIL logo and Yanrong Hao EMAIL logo
Published/Copyright: April 12, 2023

Abstract

Objectives

In nasopharyngeal carcinoma (NPC), the main factors for treatment failure are local recurrence and metastasis. Vasculogenic mimicry (VM), formation by invasive cancer cells mimicking the vasculogenic network, is strongly correlated with tumor therapy resistance and distant metastasis. CSF-1R was substantially expressed in NPC patients with a poor prognosis, according to an earlier study of ours. However, whether CSF-1R affects progression through vasculogenic mimicry deserves consideration.

Methods

By cultivating NPC cells that had CSF-1R overexpression in three-dimensional culture and labeling the NPC xenografts with CD34-PAS vasculogenic mimicry markers, the effect of CSF-1R on VM formation, migration, and invasion of NPC cells was evaluated. Finally, the underlying mechanisms were investigated by western blot.

Results

In vitro and in vivo, overexpressing CSF-1R in NPC cells causes the development of vessel-like structures. Meanwhile, NPC cells migrated and invaded more readily in the Transwell experiment when CSF-1R was highly expressed. Mechanistically, our research indicates that CSF-1R may control cell plasticity by activating the PI3K/AKT signaling pathway, promoting the formation of VM in these cells by facilitating the epithelial-mesenchymal transition.

Conclusions

CSF-1R in NPC progression by increasing VM production to increase nutrient supply to tumor cells and promote cancer cell invasion. Furthermore, these findings suggest that CSF-1R is a new promising therapeutic target aimed at treating VM in NPC.

Introduction

Nasopharyngeal carcinoma (NPC) is an epithelial tumor that develops in the nasopharynx and is extremely invasive and capable of metastasizing [1, 2]. Despite the use of radiotherapy and systemic therapies to treat NPC, distant metastases, and possible local recurrence resulting from resistance to radiotherapy, are the main reasons for treatment failure in NPC [3]. Therefore, the development of more active therapeutic approaches a good understanding of the mechanisms behind tumor spread and treatment resistance.

Tumor angiogenesis ensures that tumors receive a steady supply of nutrients, which is essential for tumor growth and metastasis [4]. Vasculogenic mimicry (VM), a tumor microcirculatory system that varies from vascular systems produced in traditional angiogenesis, was originally discovered in melanoma cells by Maniotis et al. [5] VM consists of a microvascular channel with epithelioid carcinoma cells that are CD34-negative and periodic acid-Schiff (PAS) positive basement membrane [6]. As the tumor cells are directly mosaiced in the lumen and are exposed to blood flow, the tumor cells can very easily enter the microcirculation via blood flow and thereby lead to distant metastasis [7], [8], [9]. Patients with NPCs that have VM are more likely to experience metastasis and have a poor clinical prognosis [10, 11]. The strong association between metastasis or worsening survival outcomes in cancer and tumor cells lining the VM structures is well documented [7], [8], [9]. More importantly, the presence of VM may clinically explain the inability of tumor-suppressive drugs to prevent metastasis [12]. As there are no effective clinical strategies for VM, new targets with VM-blocking effects are urgently needed to improve therapeutic outcomes for NPC patients.

The colony-stimulating factor-1 receptor (CSF-1R), a potential gene of interest, displayed significant differential expression in a comparative genomic study of biopsy specimens from patients with radiation-sensitive and radiation-resistant malignancies [13]. CSF-1R [14] is a member of the type III receptor tyrosine kinase family and is generated by the proto-oncogene c-fms. CSF-1R is a polypeptide that consists of 972 amino acids, which contain four sequences, and is considered a major immune system regulator [15]. CSF-1R–ligand binding is amplified via multiple cellular signaling pathways that induce the phosphorylation of tyrosine residues in the intracellular structural domain and consequent CSF-1R activation, which initiates and stimulates signal transduction pathways for target cell proliferation [16]. The high levels of CSF-1R [17] and its ligand leads to sustained tyrosine kinase activity that alters the biological effects of the functional protein, thereby enabling the continued proliferation of target cells to eventually develop cancer. The tissues of NPC patients have higher levels of CSF-1R expression, and this increased expression is linked to a higher risk of tumor recurrence, metastasis, and death [18, 19]. Thus, a potential regulatory role for CSF-1R in tumor progression has been indicated, although the effect of CSF-1R in VM in NPC has not been investigated.

We investigated the biological effects of CSF-1R on NPC cells by transfecting lentiviral or control vectors expressing the CSF-1R gene into NPC cell lines to illuminate the potential pathways of CSF-1R-induced effects in cellular and animal model experiments for monitoring VM development. Our goal was to clarify the role of CSF-1R and the related downstream signaling in NPC about VM, migration, and invasion. We intend to provide a rationale for developing therapeutic drug targets that precisely target VM in NPC patients.

Materials and methods

Cell lines and cultures

The non-metastatic human NPC 6–10B cell line (Sun Yat-sen University, Guangzhou, China) was used in this study. Fetal bovine serum (FBS; Gibco, USA) 10 % in RPMI 1640 medium (Gibco, USA) was used to cultivate cells in a humid incubator at 37 °C and 5 % CO2.

Lentiviral vectors construction and transfection

The lentivirus overexpressing CSF-1R that carries the enhanced green fluorescent protein (EGFP) gene was purchased from GeneChem Co., Ltd. (Shanghai, China). The LV-CSF-1R vector was used to transfect the 6–10B cells (LV-CSF-1R group). Transfected LV-negative control vector (LV-Ctrl group) and untransfected vector (Mock group) were used as control groups. A lentiviral infection with a multiplicity of infection (MOI) of 15 was enabled for 18 h after the 6–10B cells were injected in 12 well plates. Stably infected cells were identified after 72 h of incubation by adding 3 g/mL puromycin (Solarbio, Beijing, China). After at least 10 consecutive generations, transfection efficiency was assessed based on EGFP protein expression by using inverted fluorescence microscopy (Olympus, Japan). Quantitative real-time PCR (RT-qPCR) and Western blotting were used to demonstrate the upregulation of CSF-1R expression.

Quantitative real-time-PCR

For total RNA extraction, the 6–10B cells were collected and put into TRIzol Reagent (Invitrogen, CA, USA). First, the reaction strand synthesis of cDNA was performed using the FastKing RT kit (GeneChem Co., Ltd., Beijing, China). Next, the SuperReal PreMix Plus SYBR Green PCR kit (Tiangen), CSF-1R primers (CSF-1R forward, 5′-TCT GGT CCT ATG GCA TCC TC-3′, reverse, 5′-GAT GCC AGG GTA GGG ATT C-3′), and β-actin primers (β-Actin forward, 5′-TGA CGT GGA CAT CCG CAA AG-3′, reverse, 5′-CTG GAA GGT GGACAG GGA-3′) in the ABI 7500 Real-Time PCR system (Thermo Fisher Scientific, USA) were used for quantitative PCR analysis. Lastly, the 2−ΔΔCq method was applied to quantify relative RNA levels, with β-actin serving as a normalizing factor [20].

Western blotting

Protease inhibitors were diluted 100:1 with RIPA lysis buffer (Beyotime, Shanghai, China) and added to the 6–10B cell culture dishes for protein extraction by lysis. Proteins were transferred to polyvinylidene difluoride membranes after electrophoresis in 8 % sodium dodecyl sulfate-polyacrylamide gels separated them (EMD Millipore, Bedford, MA, USA). The membranes were incubated with the primary antibody for 16 h at 4 °C after being blocked in 5 % nonfat milk for 1 h. Orders were placed with Cell Signaling Technology for all antibodies (Danvers, MA, USA). A nti-CSF-1R (1:1,000; #67455), anti-E-cadherin (1:1,000; #3195), anti-HIF-1α (1:1,000; #36169), anti-MMP-2 (1:1,000; #40994), anti-MMP9 (1:1,000; #13667), anti-phosphoinositide 3-kinase (PI3K; 1:1,000; #4255), anti-Akt (1:1,000; #9272), anti-p-Akt (1:1,000; #4060S), and anti-β-actin (1:1,000; #4970) were the primary antibodies. Protein bands were transferred to species-specific secondary antibodies (1:3,000; #7076S) conjugated to horseradish peroxidase polymer (HRP) the following day, and they were incubated for 2 h at 37 °C. Finally, bands were detected with enhanced chemiluminescence reagent (ECL; Meilun, Dalian, China) in an Odyssey FC system (LI-COR Biosciences, USA) and then quantified by ImageJ (NIH, Bethesda, MA, USA) using β-actin as an internal control.

Three-dimensional culture

Matrigel (354230, BD Biosciences, USA) and pipette tips were pre-chilled before performing the tube-formation assays [21]. Next, 10 µl Matrigel was added to the angiogenic slides (Ibidi, USA) for pretreatment. The Matrigel layer in each well was covered with a suspension of 6–10B cells in serum-free media (2 × 105 cells/mL, 50 μL), which was then incubated for 12 h at 37 °C. Five areas were randomly photographed with a microscope and the average number of tubes was calculated.

Transwell assay

Transwell chambers on 24-well plates that were either pre-coated with Matrigel or left uncoated (8-μm pore size, Corning, MA, USA) were used for migration and invasion tests. The 6–10B (8-μm pore size, Corning, MA, USA) were used for migration and invasion tests (8-μm pore size, Corning, MA, USA). The 6–10B cells were resuspended in serum-free media, and 500 μL of the medium containing 15 % FBS was introduced to the bottom chamber while 200 μL of the cell suspension (5 × 105 cells/ml) was pulled into the Transwell chamber. The cells were allowed to migrate or invade for 48 or 72 h before being entirely removed from the top of the Transwell chambers with cotton swabs. 4 % paraformaldehyde-fixed, and 0.1 % crystal violet (Shanghai Biyuntian Biotechnology Co., Ltd., China) stained the cells that had migrated or invaded for 30 min, respectively. The cells that had migrated or invaded were fixed in 4 % paraformaldehyde and stained for 30 min with 0.1 % crystal violet (Shanghai Biyuntian Biotechnology Co., Ltd., China). Finally, using a microscope, these cells were counted in five random regions of each well.

Animal models

The Ethics Committee of the People’s Hospital of the Guangxi Zhuang Autonomous Region authorized the use of animals in this study in compliance with institutional policies for the treatment of laboratory animals (KYGZR-2013-06, China). We bought 15 female BALB/c nude mice (4–5 weeks old; 15–21 g) from the Guangxi Medical University Laboratory Animal Center (SCXK(Gui)2014-0002, China). Mice were kept in a specialized pathogen-free (SPF) laminar flow chamber at a constant humidity level of 55 ± 15 % and temperature of 20–23 °C. The animals were divided into three groups of five at random. NPC models were established by subcutaneous injection of a 0.2-mL suspension of Mock, LV-Ctrl, and LV-CSF-1R overexpressing 6–10B cells (5 × 106 cells/mL) into the right dorsum of nude mice. Animals were monitored for disease progression and health status every 3 days, with care in noting any signs of distress or loss of health. The removed tumors were fixed in 10 % formalin and then implanted in paraffin after the mice were euthanized after three weeks.

CD34-PAS dual staining

Serially sectioned, dewaxed, and rehydrated mouse tissue samples were then used to perform thermally induced antigen repair in a pressure cooker with sodium citrate buffer. The tissues were treated with 3 % hydrogen peroxide to eliminate endogenous peroxidase. Sections were then sealed with 2 % BSA for 20 min at 37 °C, after which they were incubated for 16 h at 4 °C with diluted anti-CD34 antibody (1:200; #ab81289; Abcam, Cambridge, UK). Following that, the sections were treated for 30 min at 37 °C with an HRP-conjugated secondary antibody. The sections were then treated with Schiff’s solution for 20 min before being stained with 3-3′ diaminobenzidine (DAB), 0.5 % periodate solution (PAS), and then Schiff’s solution [21]. Lastly, the slices were re-stained with hematoxylin. The number of VM was calculated after five randomly selected locations from each section were photographed and examined under a microscope.

Statistical analysis

The data analyses were conducted using GraphPad Prism 9 (GraphPad Software, CA, USA) and SPSS 22.0 (IBM Corp., NY, USA). Data are expressed as mean ± standard deviation. One-way ANOVA was performed to compare the means of different groups, and the LSD t-test was used for pairwise comparisons between groups. The threshold for statistical significance was established at p<0.05 for all two-sided statistical tests.

Results

Stable CSF-1R overexpression in 6–10B cell line

To explore CSF-1R the impact on NPC cell lines. To investigate the effect of CSF-1R on NPC cell lines. In previous experiments [22], we evaluated CSF-1R expression in three NPC cell lines (5–8F, 6–10B, and CNE-2), and found that only 6–10B had relatively low CSF-1R expression. Therefore, in this study, 6–10B cells were selected to stably overexpress CSF-1R. Meanwhile, the sequence design for knocking down CSF-1R failed; therefore, knockdown experiments were not carried out.

We employed a lentivirus-mediated CSF-1R RNA (LV-CSR-1R group) transfection technique to upregulate CSF-1R expression levels and produce stably expressed cell lines to analyze the phenotypic changes (Figure 1A). As controls, control-RNA-infected (LV-Ctrl group) and uninfected (Mock group) cells were employed. By using RT-qPCR and western blotting, we examined the expression of CSF-1R in 6–10B cells. The results showed strong and stable expression of CSF-1R in CSF-1R RNA-transfected 6–10B cells as compared with the LV-Ctrl and Mock groups (p<0.001), and there was no statistical significance between the LV-Ctrl and Mock groups (Figure 1B and D).

Figure 1: 
CSF-1R expression is abnormally high in 6–10B cells. (A) Transfected 6–10B cells were observed by fluorescence microscopy (magnification, ×200). (B–D) RT-qPCR and western blotting were used to confirm CSF-1R expression in 6–10B cells. Mock and LV-Ctrl are the control samples for LV-CSF-1R. Data as mean ± SD (n=3). ***p<0.001.
Figure 1:

CSF-1R expression is abnormally high in 6–10B cells. (A) Transfected 6–10B cells were observed by fluorescence microscopy (magnification, ×200). (B–D) RT-qPCR and western blotting were used to confirm CSF-1R expression in 6–10B cells. Mock and LV-Ctrl are the control samples for LV-CSF-1R. Data as mean ± SD (n=3). ***p<0.001.

Alteration of CSF-1R expression affects tube formation in vitro

To observe whether CSF-1R overexpression affects the tube-formation ability, we next performed the tube formation assay. The results of 3D culture demonstrated the appearance of tube formation following CSF-1R high expression, which were tumor cells arranged in an arc (p<0.01), as illustrated in Figure 2A and B. LV-Ctrl and Mock groups of cells were oval and hardly formed complete tube structures. These results confirmed that CSF-1R induced tube formation in NPC cells in vitro.

Figure 2: 
CSF-1R overexpression increases VM formation in vitro. (A) VM channels in three-dimensional cultures of cells overexpressing CSF-1R (magnification, ×200). (B) The number of tubes that formed in each group was statistically analyzed. Data as mean ± SD (n=3). **p<0.01. ***p<0.001.
Figure 2:

CSF-1R overexpression increases VM formation in vitro. (A) VM channels in three-dimensional cultures of cells overexpressing CSF-1R (magnification, ×200). (B) The number of tubes that formed in each group was statistically analyzed. Data as mean ± SD (n=3). **p<0.01. ***p<0.001.

6–10B cell migration and invasion are promoted by high expression of CSF-1R

VM formation is crucial for the migration and invasion of tumor cells [23]. Figure 3A and B show that after high expression of CSF-1R, the number of cells migrating to the lower lumen was considerably higher than in the Mock and LV-Ctrl groups (p<0.01). Furthermore, the invasion assay indicated a more significant cell invasion potential in the LV-CSF-1R group (p<0.00). No significant differences were observed between the Mock and LV-Ctrl groups (Figure 3C and D). The findings imply that NPC cell invasion and migration are mediated by CSF-1R.

Figure 3: 
6–10B cells are driven to migrate and invade by CSF-1R. (A and C) Transwell assay (magnification, ×100) results show that upregulation of CSF-1R influences migration and invasion. (B and D) The bar chart shows the statistical outcomes. Data as mean ± SD (n=3).**p<0.01; ***p< 0.001.
Figure 3:

6–10B cells are driven to migrate and invade by CSF-1R. (A and C) Transwell assay (magnification, ×100) results show that upregulation of CSF-1R influences migration and invasion. (B and D) The bar chart shows the statistical outcomes. Data as mean ± SD (n=3).**p<0.01; ***p< 0.001.

CSF-1R induced VM formation in animal models

6–10B cells were injected subcutaneously into an immunocompromised mouse to create a xenograft cancer model to further support the effect of CSF-1R on VM formation in vivo. VM in the tumors was counted using CD34-PAS double staining. As shown in Figure 4A, LV-CSF-1R tumors generated significantly more VM channels than the LV-Ctrl and Mock groups (p<0.001). No significant differences were observed between the Mock and LV-Ctrl groups (Figure 4B). Our data suggest that CSF-1R overexpression in NPC cells promotes VM.

Figure 4: 
Overexpression of CSF-1R accelerated the VM effect in tumor tissues. (A) CD34-PAS double staining of each group in vitro. Red arrows show VM; Black arrows indicate endothelial vessels; top panel, magnification: 200×; Bottom panel, magnification: 40×. (B) Statistical analysis of VM formation in xenograft tumors. Data as mean ± SD (n=3). ***p< 0.001.
Figure 4:

Overexpression of CSF-1R accelerated the VM effect in tumor tissues. (A) CD34-PAS double staining of each group in vitro. Red arrows show VM; Black arrows indicate endothelial vessels; top panel, magnification: 200×; Bottom panel, magnification: 40×. (B) Statistical analysis of VM formation in xenograft tumors. Data as mean ± SD (n=3). ***p< 0.001.

CSF-1R promoted EMT by the PI3K/AKT pathway

The epithelial-mesenchymal transition (EMT) is critical for tumor development, and its stimulation causes tumor cell invasion and spread [24]. Research has confirmed that tumor cells can form VM through the EMT process [25]. Therefore, we examined whether the key gene CSF-1R affects the EMT process by western blotting. In the current research, overexpression of CSF-1R substantially reduced the levels of E-cadherin protein and raised the levels of HIF-1α, MMP-2, and MMP-9 proteins in comparison to the Mock and LV-Ctrl groups (Figure 5A). Taken together, the above data confirm the involvement of CSF-1R in the EMT process of NPC.

Figure 5: 
CSF-1R overexpression increases the protein levels involved in EMT, VM, and the PI3K/AKT signaling pathway in 6–10B cells. (A and B) Western blot and quantitative analysis revealed the expression of EMT and VM-related proteins (HIF-1α, MMP-2, MMP-9, and E-cadherin), as well as the PI3K/AKT signaling pathway (PI3K, AKT, p-AKT). Data as mean ± SD (n=3). ***p< 0.001. (C) Interaction network of CSF-1R with PIK3AP1, PIK3CA, PIK3CB, PIK3CD and PIK3CG proteins predicted by string website.
Figure 5:

CSF-1R overexpression increases the protein levels involved in EMT, VM, and the PI3K/AKT signaling pathway in 6–10B cells. (A and B) Western blot and quantitative analysis revealed the expression of EMT and VM-related proteins (HIF-1α, MMP-2, MMP-9, and E-cadherin), as well as the PI3K/AKT signaling pathway (PI3K, AKT, p-AKT). Data as mean ± SD (n=3). ***p< 0.001. (C) Interaction network of CSF-1R with PIK3AP1, PIK3CA, PIK3CB, PIK3CD and PIK3CG proteins predicted by string website.

We searched the string database and predicted the existence of protein interactions of CSF-1R with PIK3AP1, PIK3CA, PIK3CB, PIK3CD, and PIK3CG (Figure 5C). Moreover, the PI3K/Akt signaling axis regulates EMT, which is linked to tumor development [26]. Consequently, we explored changes in the PI3K/AKT signaling axis. Western blotting revealed that PI3K, AKT and p-AKT expressions were raised in the LV-CSF-1R group (Figure 5B). In conclusion, these findings imply that overexpression of CSF-1R can activate this signaling pathway in NPC cells, which mediates EMT to promote VM formation (Figure 6).

Figure 6: 
Schematic diagram showing that CSF-1R overexpression induces epithelial-mesenchymal transition promoted vasculogenic mimicry through the PI3K/AKT signaling pathways in nasopharyngeal carcinoma.
Figure 6:

Schematic diagram showing that CSF-1R overexpression induces epithelial-mesenchymal transition promoted vasculogenic mimicry through the PI3K/AKT signaling pathways in nasopharyngeal carcinoma.

Discussion

VM is a microvascular structure that is formed by cancer cells and is similar to a three-dimensional channel that provides the required nutrients for tumor growth [27]. Previous studies [10, 28] have confirmed the presence of VM structures in nasopharyngeal carcinoma, which correlates markedly with clinical stage, with shorter survival times in VM-positive patients. This suggests that VM is an indicator of poor prognosis in NPC. Thus, clarifying the key mechanisms underlying VM formation in NPC will help provide insight into the causes of tumor progression and treatment resistance, and to develop new clinical management strategies.

In the current research, we discovered that high CSF-1R expression not only significantly enhanced VM formation, invasion, and migration of NPC cell lines 6–10B in vitro, but also correlated with the VM density in vivo. Moreover, EMT is crucial in the formation of VM [29]. We examined the association of CSF-1R with EMT and VM marker proteins in NPC cells. CSF-1R had a positive correlation with the expression of HIF-1α, MMP2 and MMP9 and a negative correlation with E-cadherin. The above-described findings indicate that CSF-1R may promote VM formation in NPC through the induction of EMT.

Tyrosine phosphorylation of CSF-1R allows direct interaction with specific subunits of PI3K [30]. Then, PI3K initiates downstream signaling to control macrophage movement and promote cancer cell invasion [31]. String database predictions showed that CSF-1R mediates protein interactions with PIK3AP1, PIK3CA, PIK3CD, PIK3CB, and PIK3CG. We infer that CSF-1R may have a further function in NPC via PI3K [32]. In addition, it has been demonstrated in many experiments that the PI3K/AKT signaling axis can influence the EMT process [33], [34], [35], [36], [37], [38]. For instance, FAT4 controls the EMT in part through the PI3K-AKT signaling axis in colorectal cancer cells [39]. Our results showed this signaling pathway and supported the bioinformatics forecast. The above results illustrated that CSF-1R facilitates EMT formation through PI3K/AKT signaling pathway to regulate VM channels in NPC, which provides a new therapeutic target for NPC.

Recent evidence has denoted the crucial role of cancer cells’ mitochondria in cancer progression [40] and treatment resistance [41]. It has been demonstrated that mitochondrial reactive oxygen species (ROS) can promote vasculogenic mimicry through HIF-1α stabilization [42]. Besides, Chaturvedi et al. demonstrated that HIF-1α is an upstream regulator of CSF-1R to promote cancer progression [43]. The interaction between mitochondria, HIF-1α, and CSF-1R can provide new paradigms for future studies to enhance the targeted therapies against CSF-1R.

Researchers have confirmed that anti-angiogenic single agents are not sufficient for lasting clinical improvement [6, 44, 45] and VM formation may be the source of anti-VEGF resistance [46]. For example, treatment with sunitinib in mammary carcinoma-bearing mice demonstrated an increase in VM channels and increased the aggressivity of triple-negative breast cancer [47]. VM structure formation is driven by anti-angiogenic therapy (AAT) resistance in glioblastoma in vitro as well as in animal models [48]. Furthermore, sorafenib is a multikinase inhibitor that reduces angiogenesis and yet promotes disease progression in some patients with hepatocellular carcinoma [49]. Therefore, an effective strategy should inhibit both endothelium-dependent vascularity (EDV) and tumor cell-mediated VM [50]. Our study demonstrates that CSF-1R promotes VM formation in nasopharyngeal carcinoma and may be responsible for poor prognosis as this will contribute to the development of approaches to inhibit angiogenesis and improve prognosis.

CSF-1R/c-FMS is aberrantly overexpressed in various cancers and tumor-associated macrophages (TAM) and is a potential biomarker for poor prognosis in different types of malignancies [51]. Therefore, CSF-1R is used as a molecular target for malignancy treatment [52, 53]. Several CSF-1R/c-FMS inhibitors (e.g., ARRY-382, linifanib, PLX5622, PLX3397, DCC-3014, and Ki20227) are useful in cancer treatment after preclinical studies or clinical trials [54]. The study found that targeting recurrent human glioblastoma with CSF-1R inhibitors in combination with radiation therapy is a strategy for overcoming drug resistance [55]. The anti-CSF-1R antibody LY3022855’s tolerance and effectiveness in patients with solid tumors were defined in a phase 1 trial [56]. Multiple CSF-1/CSF-1R inhibitors are effective and safe in cancer patients, according to a significant body of research [57]. Based on our research CSF-1R directly binds to the PI3K/AKT axis and regulates VM in NPC. In the future, we performed computer-aided drug design to try to screen CSF-1R inhibitors, which are more accurate and effective in inhibiting the formation of VM.

There are limitations to this study. Only one human nasopharyngeal carcinoma cell line was used in this research. The role of CSF-1R pro-angiogenic mimicry should be validated in human nasopharyngeal carcinoma cell lines of different histological subtypes. In addition, we found VM in nude mouse transplanted tumors, but could not directly assess CSF-1R-induced VM formation in human nasopharyngeal carcinoma tissues. In the future, we will further confirm the VM-promoting role of CSF-1R in clinical specimens.

Conclusions

Cumulatively, the results of the current study showed that CSF-1R promoted cancer invasion, migration, and EMT to influence VM formation in NPC. Moreover, PI3K/AKT signaling was engaged in these processes. This study extends the therapeutic strategy for VM in NPC, thereby contributing to the further exploration of VM and adaptive drug resistance and also providing valuable targets for the treatment of NPC.


Corresponding author: Yanyan Xie and Yanrong Hao, Cancer Center, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, 530021 Guangxi, P.R. China, E-mail: ,
Huaqing Mo, Yanru Lv and Shan Gao contributed equally.

Award Identifier / Grant number: 81260348

Award Identifier / Grant number: GuiKe AB21196012

Acknowledgments

The authors thank Prof. Musheng Zeng from the Cancer Center of Sun Yat-sen University for providing the 6–10B cell line and Junde Ou (School of Foreign Languages, Hezhou University) for suggestions on language revisions to this paper.

  1. Research funding: This study was supported by a grant from the National Natural Science Foundation of China (grant no. 81260348) and the Key Research and Development Program of Guangxi (grant no. GuiKe AB21196012).

  2. Author contributions: The authors confirm their contribution to the paper as follows: study conception and design: HQM. Author, YYX. Author, YRH. Author; data collection: HQM. Author, YRL. Author, SG. Author; analysis and interpretation of results: ZNC. Author, ZYX. Author. JYS. Author; draft manuscript preparation: HQM. Author. YYX. Author, YRH. Author. All authors reviewed the results and approved the final version of the manuscript. All authors have accepted responsibility for the entire content of this manuscript and approved its submission.

  3. Competing interests: Authors state no conflict of interest.

  4. Informed consent: Not applicable.

  5. Ethical approval: All animal experiments followed the Guide for the Care and Use of Laboratory Animals Handling and were approved by the Ethics Committee of the People’s Hospital of Guangxi Zhuang Autonomous Region (approval no. KYGZR-2013-06, China).

  6. Availability of data and materials: The data used to support the findings of this study are included within the article.

References

1. Chen, YP, Chan, ATC, Le, QT, Blanchard, P, Sun, Y, Ma, J. Nasopharyngeal carcinoma. Lancet 2019;394:64–80. https://doi.org/10.1016/s0140-6736(19)30956-0.Search in Google Scholar

2. Guo, R, Mao, YP, Tang, LL, Chen, L, Sun, Y, Ma, J. The evolution of nasopharyngeal carcinoma staging. Br J Radiol 2019;92:20190244. https://doi.org/10.1259/bjr.20190244.Search in Google Scholar PubMed PubMed Central

3. Lee, AWM, Ng, WT, Chan, JYW, Corry, J, Mäkitie, A, Mendenhall, WM, et al.. Management of locally recurrent nasopharyngeal carcinoma. Cancer Treat Rev 2019;79:101890. https://doi.org/10.1016/j.ctrv.2019.101890.Search in Google Scholar PubMed

4. Chiang, AK, Mak, NK, Ng, WT. Translational research in nasopharyngeal carcinoma. Oral Oncol 2014;50:345–52. https://doi.org/10.1016/j.oraloncology.2013.11.005.Search in Google Scholar PubMed

5. Maniotis, AJ, Folberg, R, Hess, A, Seftor, EA, Gardner, LM, Pe’er, J, et al.. Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Am J Pathol 1999;155:739–52. https://doi.org/10.1016/s0002-9440(10)65173-5.Search in Google Scholar PubMed PubMed Central

6. Pezzella, F, Ribatti, D. Vascular co-option and vasculogenic mimicry mediate resistance to antiangiogenic strategies. Cancer Rep 2020;5:e1318. https://doi.org/10.1002/cnr2.1318.Search in Google Scholar PubMed PubMed Central

7. Cao, Z, Bao, M, Miele, L, Sarkar, FH, Wang, Z, Zhou, Q. Tumour vasculogenic mimicry is associated with poor prognosis of human cancer patients: a systemic review and meta-analysis. Eur J Cancer 2013;49:3914–23. https://doi.org/10.1016/j.ejca.2013.07.148.Search in Google Scholar PubMed

8. Salem, A, Salo, T. Vasculogenic mimicry in head and neck squamous cell carcinoma-time to take notice. Frontiers in oral health 2021;2:666895. https://doi.org/10.3389/froh.2021.666895.Search in Google Scholar PubMed PubMed Central

9. Yue, Y, Lou, Y, Liu, X, Peng, X. Vasculogenic mimicry in head and neck tumors: a narrative review. Transl Cancer Res 2021;10:3044–52. https://doi.org/10.21037/tcr-21-34.Search in Google Scholar PubMed PubMed Central

10. Luo, Y, Wang, J, Wang, F, Liu, X, Lu, J, Yu, X, et al.. Foxq1 promotes metastasis of nasopharyngeal carcinoma by inducing vasculogenic mimicry via the EGFR signaling pathway. Cell Death Dis 2021;12:411. https://doi.org/10.1038/s41419-021-03674-z.Search in Google Scholar PubMed PubMed Central

11. Xiang, T, Lin, YX, Ma, W, Zhang, HJ, Chen, KM, He, GP, et al.. Vasculogenic mimicry formation in EBV-associated epithelial malignancies. Nat Commun 2018;9:5009. https://doi.org/10.1038/s41467-018-07308-5.Search in Google Scholar PubMed PubMed Central

12. Wei, X, Chen, Y, Jiang, X, Peng, M, Liu, Y, Mo, Y, et al.. Mechanisms of vasculogenic mimicry in hypoxic tumor microenvironments. Mol Cancer 2021;20:7. https://doi.org/10.1186/s12943-020-01288-1.Search in Google Scholar PubMed PubMed Central

13. Yang, S, Chen, J, Guo, Y, Lin, H, Zhang, Z, Feng, G, et al.. Identification of prognostic biomarkers for response to radiotherapy by DNA microarray in nasopharyngeal carcinoma patients. Int J Oncol 2012;40:1590–600. https://doi.org/10.3892/ijo.2012.1341.Search in Google Scholar PubMed

14. Rettenmier, CW, Roussel, MF, Sherr, CJ. The colony-stimulating factor 1 (CSF-1) receptor (c-fms proto-oncogene product) and its ligand. J Cell Sci Suppl 1988;9:27–44. https://doi.org/10.1242/jcs.1988.supplement_9.2.Search in Google Scholar PubMed

15. Sherr, CJ. The colony-stimulating factor 1 receptor: pleiotropy of signal-response coupling. Lymphokine Res 1990;9:543–8.Search in Google Scholar

16. Stanley, ER, Berg, KL, Einstein, DB, Lee, PS, Yeung, YG. The biology and action of colony stimulating factor-1. Stem Cells 1994;12:15–24.Search in Google Scholar

17. Horiuchi, K, Toyama, Y. Posttranslational regulation of cell-surface colony-stimulating factor-1. Crit Rev Immunol 2008;28:215–27. https://doi.org/10.1615/critrevimmunol.v28.i3.30.Search in Google Scholar PubMed

18. Huang, L, Xu, X, Hao, Y. The possible mechanisms of tumor progression via CSF-1/CSF-1R pathway activation. Romanian J Morphol Embryol = Revue roumaine de morphologie et embryologie 2014;55:501–6.Search in Google Scholar

19. Huang, L, Xu, X, Hao, Y, Chen, J, Li, L, Cheng, J, et al.. Overexpression of CSF-1R in nasopharyngeal carcinoma. Rom J Morphol Embryol = Revue roumaine de morphologie et embryologie 2015;56:1279–83.Search in Google Scholar

20. Schmittgen, TD, Livak, KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc 2008;3:1101–8. https://doi.org/10.1038/nprot.2008.73.Search in Google Scholar PubMed

21. Folberg, R, Hendrix, MJ, Maniotis, AJ. Vasculogenic mimicry and tumor angiogenesis. Am J Pathol 2000;156:361–81. https://doi.org/10.1016/s0002-9440(10)64739-6.Search in Google Scholar

22. Chen, J, Hao, Y, Chen, J, Huang, L, Ao, W. Colony stimulating factor-1 receptor-mediated Bax/Bcl-2 expression inhibits apoptosis of human nasopharyngeal carcinoma 6-10B cells. Chin J Cancer Biother 2017;24:1386–90.Search in Google Scholar

23. Zhang, X, Zhang, J, Zhou, H, Fan, G, Li, Q. Molecular mechanisms and anticancer therapeutic strategies in vasculogenic mimicry. J Cancer 2019;10:6327–40. https://doi.org/10.7150/jca.34171.Search in Google Scholar PubMed PubMed Central

24. Liu, Q, Qiao, L, Liang, N, Xie, J, Zhang, J, Deng, G, et al.. The relationship between vasculogenic mimicry and epithelial-mesenchymal transitions. J Cell Mol Med 2016;20:1761–9. https://doi.org/10.1111/jcmm.12851.Search in Google Scholar PubMed PubMed Central

25. Fan, YL, Zheng, M, Tang, YL, Liang, XH. A new perspective of vasculogenic mimicry: EMT and cancer stem cells. Oncol Lett 2013;6:1174–80. https://doi.org/10.3892/ol.2013.1555.Search in Google Scholar PubMed PubMed Central

26. Xu, W, Yang, Z, Lu, N. A new role for the PI3K/Akt signaling pathway in the epithelial-mesenchymal transition. Cell Adhes Migr 2015;9:317–24. https://doi.org/10.1080/19336918.2015.1016686.Search in Google Scholar PubMed PubMed Central

27. Qiao, L, Liang, N, Zhang, J, Xie, J, Liu, F, Xu, D, et al.. Advanced research on vasculogenic mimicry in cancer. J Cell Mol Med 2015;19:315–26. https://doi.org/10.1111/jcmm.12496.Search in Google Scholar PubMed PubMed Central

28. Xu, S, Bai, J, Zhuan, Z, Li, B, Zhang, Z, Wu, X, et al.. EBV-LMP1 is involved in vasculogenic mimicry formation via VEGFA/VEGFR1 signaling in nasopharyngeal carcinoma. Oncol Rep 2018;40:377–84. https://doi.org/10.3892/or.2018.6414.Search in Google Scholar PubMed

29. Sun, B, Zhang, D, Zhao, N, Zhao, X. Epithelial-to-endothelial transition and cancer stem cells: two cornerstones of vasculogenic mimicry in malignant tumors. Oncotarget 2017;8:30502–10. https://doi.org/10.18632/oncotarget.8461.Search in Google Scholar PubMed PubMed Central

30. Sampaio, NG, Yu, W, Cox, D, Wyckoff, J, Condeelis, J, Stanley, ER, et al.. Phosphorylation of CSF-1R Y721 mediates its association with PI3K to regulate macrophage motility and enhancement of tumor cell invasion. J Cell Sci 2011;124:2021–31. https://doi.org/10.1242/jcs.075309.Search in Google Scholar PubMed PubMed Central

31. Mouchemore, KA, Sampaio, NG, Murrey, MW, Stanley, ER, Lannutti, BJ, Pixley, FJ. Specific inhibition of PI3K p110δ inhibits CSF-1-induced macrophage spreading and invasive capacity. FEBS J 2013;280:5228–36. https://doi.org/10.1111/febs.12316.Search in Google Scholar PubMed PubMed Central

32. Chen, J, Hao, Y, Chen, J, Huang, L, Ao, W, Yang, J, et al.. Colony stimulating factor-1 receptor promotes proliferation, migration and invasion in the human nasopharyngeal carcinoma 6-10B cell line via the phosphoinositide 3-kinase/Akt pathway. Oncol Lett 2018;16:1205–11. https://doi.org/10.3892/ol.2018.8750.Search in Google Scholar PubMed PubMed Central

33. Li, C, Hou, X, Yuan, S, Zhang, Y, Yuan, W, Liu, X, et al.. High expression of TREM2 promotes EMT via the PI3K/AKT pathway in gastric cancer: bioinformatics analysis and experimental verification. J Cancer 2021;12:3277–90. https://doi.org/10.7150/jca.55077.Search in Google Scholar PubMed PubMed Central

34. Ma, Z, Lou, S, Jiang, Z. PHLDA2 regulates EMT and autophagy in colorectal cancer via the PI3K/AKT signaling pathway. Aging 2020;12:7985–8000. https://doi.org/10.18632/aging.103117.Search in Google Scholar PubMed PubMed Central

35. Rumman, M, Jung, KH, Fang, Z, Yan, HH, Son, MK, Kim, SJ, et al.. HS-173, a novel PI3K inhibitor suppresses EMT and metastasis in pancreatic cancer. Oncotarget 2016;7:78029–47. https://doi.org/10.18632/oncotarget.12871.Search in Google Scholar PubMed PubMed Central

36. Wang, B, Wang, L, Lu, Y, Liang, W, Gao, Y, Xi, H, et al.. GRSF1 promotes tumorigenesis and EMT-mediated metastasis through PI3K/AKT pathway in gastric cancer. Biochem Biophys Res Commun 2021;555:61–6. https://doi.org/10.1016/j.bbrc.2021.03.121.Search in Google Scholar PubMed

37. Wang, H, Zhao, Y, Cao, L, Zhang, J, Wang, Y, Xu, M. Metastasis suppressor protein 1 regulated by PTEN suppresses invasion, migration, and EMT of gastric carcinoma by inactivating PI3K/AKT signaling. J Cell Biochem 2019;120:3447–54. https://doi.org/10.1002/jcb.27618.Search in Google Scholar PubMed

38. Xu, E, Xia, X, Jiang, C, Li, Z, Yang, Z, Zheng, C, et al.. GPER1 silencing suppresses the proliferation, migration, and invasion of gastric cancer cells by inhibiting PI3K/AKT-Mediated EMT. Front Cell Dev Biol 2020;8:591239. https://doi.org/10.3389/fcell.2020.591239.Search in Google Scholar PubMed PubMed Central

39. Wei, R, Xiao, Y, Song, Y, Yuan, H, Luo, J, Xu, W. FAT4 regulates the EMT and autophagy in colorectal cancer cells in part via the PI3K-AKT signaling axis. J Exp Clin Cancer Res CR 2019;38:112. https://doi.org/10.1186/s13046-019-1043-0.Search in Google Scholar PubMed PubMed Central

40. Taghizadeh-Hesary, F, Akbari, H, Bahadori, M, Behnam, B. Targeted anti-mitochondrial therapy: the future of oncology. Genes 2022;13:1728. https://doi.org/10.3390/genes13101728.Search in Google Scholar PubMed PubMed Central

41. Taghizadeh-Hesary, F, Houshyari, M, Farhadi, M. Mitochondrial metabolism: a predictive biomarker of radiotherapy efficacy and toxicity. J Cancer Res Clin Oncol 2023. https://doi.org/10.1007/s00432-023-04592-7.Search in Google Scholar PubMed

42. Comito, G, Calvani, M, Giannoni, E, Bianchini, F, Calorini, L, Torre, E, et al.. HIF-1α stabilization by mitochondrial ROS promotes Met-dependent invasive growth and vasculogenic mimicry in melanoma cells. Free Radic Biol Med 2011;51:893–904. https://doi.org/10.1016/j.freeradbiomed.2011.05.042.Search in Google Scholar PubMed

43. Chaturvedi, P, Gilkes, DM, Takano, N, Semenza, GL. Hypoxia-inducible factor-dependent signaling between triple-negative breast cancer cells and mesenchymal stem cells promotes macrophage recruitment. Proc Natl Acad Sci United States Am 2014;111:E2120–9. https://doi.org/10.1073/pnas.1406655111.Search in Google Scholar PubMed PubMed Central

44. Ribatti, D, Solimando, AG, Pezzella, F. The anti-VEGF(R) drug discovery legacy: improving attrition rates by breaking the vicious cycle of angiogenesis in cancer. Cancers 2021;13:3433. https://doi.org/10.3390/cancers13143433.Search in Google Scholar PubMed PubMed Central

45. Soda, Y, Myskiw, C, Rommel, A, Verma, IM. Mechanisms of neovascularization and resistance to anti-angiogenic therapies in glioblastoma multiforme. J Mol Med 2013;91:439–48. https://doi.org/10.1007/s00109-013-1019-z.Search in Google Scholar PubMed PubMed Central

46. Dey, N, De, P, Brian, LJ. Evading anti-angiogenic therapy: resistance to anti-angiogenic therapy in solid tumors. Am J Transl Res 2015;7:1675–98.10.1038/bjc.2014.439Search in Google Scholar

47. Sun, H, Zhang, D, Yao, Z, Lin, X, Liu, J, Gu, Q, et al.. Anti-angiogenic treatment promotes triple-negative breast cancer invasion via vasculogenic mimicry. Cancer Biol Ther 2017;18:205–13. https://doi.org/10.1080/15384047.2017.1294288.Search in Google Scholar PubMed PubMed Central

48. Angara, K, Borin, TF, Rashid, MH, Lebedyeva, I, Ara, R, Lin, PC, et al.. CXCR2-Expressing tumor cells drive vascular mimicry in antiangiogenic therapy-resistant glioblastoma. Neoplasia 2018;20:1070–82. https://doi.org/10.1016/j.neo.2018.08.011.Search in Google Scholar PubMed PubMed Central

49. Prieto-Domínguez, N, Ordóñez, R, Fernández, A, García-Palomo, A, Muntané, J, González-Gallego, J, et al.. Modulation of autophagy by Sorafenib: effects on treatment response. Front Pharmacol 2016;7:151. https://doi.org/10.3389/fphar.2016.00151.Search in Google Scholar PubMed PubMed Central

50. Fathi Maroufi, N, Taefehshokr, S, Rashidi, MR, Taefehshokr, N, Khoshakhlagh, M, Isazadeh, A, et al.. Vascular mimicry: changing the therapeutic paradigms in cancer. Mol Biol Rep 2020;47:4749–65. https://doi.org/10.1007/s11033-020-05515-2.Search in Google Scholar PubMed

51. Mo, H, Hao, Y, Lv, Y, Chen, Z, Shen, J, Zhou, S, et al.. Overexpression of macrophage-colony stimulating factor-1 receptor as a prognostic factor for survival in cancer: a systematic review and meta-analysis. Medicine 2021;100:e25218. https://doi.org/10.1097/md.0000000000025218.Search in Google Scholar

52. Peyraud, F, Cousin, S, Italiano, A. CSF-1R inhibitor development: current clinical status. Curr Oncol Rep 2017;19:70. https://doi.org/10.1007/s11912-017-0634-1.Search in Google Scholar PubMed

53. El-Gamal, MI, Al-Ameen, SK, Al-Koumi, DM, Hamad, MG, Jalal, NA, Oh, CH. Recent advances of colony-stimulating factor-1 receptor (CSF-1R) kinase and its inhibitors. J Med Chem 2018;61:5450–66. https://doi.org/10.1021/acs.jmedchem.7b00873.Search in Google Scholar PubMed

54. Kumari, A, Silakari, O, Singh, RK. Recent advances in colony stimulating factor-1 receptor/c-FMS as an emerging target for various therapeutic implications. Biomed Pharmacother = Biomedecine pharmacotherapie 2018;103:662–79. https://doi.org/10.1016/j.biopha.2018.04.046.Search in Google Scholar PubMed

55. Akkari, L, Bowman, RL, Tessier, J, Klemm, F, Handgraaf, SM, de Groot, M, et al.. Dynamic changes in glioma macrophage populations after radiotherapy reveal CSF-1R inhibition as a strategy to overcome resistance. Sci Transl Med 2020;12:eaaw7843. https://doi.org/10.1126/scitranslmed.aaw7843.Search in Google Scholar PubMed

56. Dowlati, A, Harvey, RD, Carvajal, RD, Hamid, O, Klempner, SJ, Kauh, JSW, et al.. LY3022855, an anti-colony stimulating factor-1 receptor (CSF-1R) monoclonal antibody, in patients with advanced solid tumors refractory to standard therapy: phase 1 dose-escalation trial. Invest New Drugs 2021;39:1057–71. https://doi.org/10.1007/s10637-021-01084-8.Search in Google Scholar PubMed

57. Ries, CH, Hoves, S, Cannarile, MA, Rüttinger, D. CSF-1/CSF-1R targeting agents in clinical development for cancer therapy. Curr Opin Pharmacol 2015;23:45–51. https://doi.org/10.1016/j.coph.2015.05.008.Search in Google Scholar PubMed

Received: 2022-12-22
Accepted: 2023-03-27
Published Online: 2023-04-12

© 2023 the author(s), published by De Gruyter, Berlin/Boston

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 25.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/oncologie-2022-1016/html
Scroll to top button