Skip to main content

EDN1 facilitates cisplatin resistance of non-small cell lung cancer cells by regulating the TNF signaling pathway

Abstract

Background

Cisplatin (DDP) is a commonly utilized chemotherapeutic agent. Nevertheless, the development of resistance to DDP significantly diminishes the effectiveness of DDP-based chemotherapy in patients with non-small cell lung cancer (NSCLC). In this study, we investigated the impact of endothelin 1 (EDN1) on the resistance to DDP in NSCLC.

Methods

The proliferation, invasion, and migration of NSCLC cells were detected by cell counting kit-8 and Transwell migration and invasion assays. ELISA was performed to analyze the inflammatory cytokines concentrations. The related protein levels of tumor necrosis factor (TNF) signaling pathway were analyzed by Western blot. Besides, a xenograft tumor mice model was established to explore the role of EDN1 in vivo.

Results

The results showed that DDP-resistance upregulated EDN1 expression, cell viability, invasion, migration, and inflammation in NSCLC cells, while the results were reversed after EDN1 inhibition. EDN1 affected DDP-resistance of NSCLC by regulating TNF signaling pathway. Overexpression of TNF receptor-1 (TNFR1) reversed the decreased cell viability, invasion, migration, and inflammation induced by silencing EDN1 in A549/DDP cells. Moreover, silencing EDN1 inhibited tumor growth and the protein levels of EDN1 and TNFR1.

Conclusion

EDN1 promoted DDP resistance in NSCLC cells through the modulation of the TNF signaling pathway, suggesting a potential therapeutic intervention strategy for NSCLC.

Introduction

Non-small cell lung cancer (NSCLC) is a term that encompasses a group of lung cancers that behave similarly and are characterized by the presence of large cells. It accounts for approximately 85% of all lung cancer diagnoses, making it the most prevalent type of lung cancer [1]. Long-term smoking, heredity, chronic obstructive pulmonary diseases, occupational causes (exposure to silica, asbestos, and inorganic arsenic), and obesity are common inductive factors for NSCLC [2, 3]. Cisplatin (DDP), also known as cis-diamminedichloroplatinum(II), is a platinum-based metal complex that plays a significant role in cancer treatment due to its unique mechanism of action. Upon administration, cisplatin enters the cells and undergoes hydrolysis, leading to the formation of reactive platinum-DNA adducts, which primarily induce cross-linking between DNA strands, disrupting the normal structure and function of the DNA helix [4]. Unfortunately, the emergence of DDP-resistance poses an obstacle to the therapeutic efficacy and reduces the survival rate of NSCLC patients [5]. Research has identified potential biomarkers for DDP resistance in NSCLC, including ubiquitin-specific protease 14 (USP14) and the long non-coding RNA small nucleolar RNA host gene 7 (SNHG7) [6, 7]. Additionally, several innovative treatment strategies for DDP-resistant NSCLC are under investigation, such as the combination of cordycepin with DDP, aptamer-guided nano-zinc carriers loaded with USP14 siRNA, and immune checkpoint inhibitors [7,8,9].

Yanagisawa and his co-workers first identified endothelin 1 (EDN1) in porcine aortic endothelial cells in 1988 [10]. Endothelin-1 (EDN1) is a powerful vasoconstrictor and mitogen, consisting of a chain of 21 amino acids. This peptide is essential in numerous physiological and pathological processes occurring in the body, such as blood supply, tissue metabolism, inflammation, homeostasis, and antioxidant stress, making it a significant focus of research in cardiovascular health and disease [11]. Besides, the role of EDN1 in the pathogenesis of different diseases has been illustrated, including obesity, infective diseases, periodontal diseases, and cancers [12,13,14,15]. In NSCLC, a previous study has found that EDN1 promotes drug resistance in NSCLC by promoting vasoconstriction and restricting blood and drug delivery [16]. In addition, another study suggests that EDN1 level in exhaled breath condensate in NSCLC patients is of diagnostic value for NSCLC [17]. While previous studies have highlighted the significance of EDN1 in NSCLC, its specific contributions to the mechanisms underlying drug resistance in NSCLC are still largely unknown.

Tumor necrosis factor (TNF), a pro-inflammatory cytokine, significantly influences tumor progression [18]. It influences a wide range of cellular functions and functions through complex signaling pathways, serving an essential role in the management of numerous diseases [19]. TNF receptor-1 (TNFR1) is a critical transducer of inflammatory pathways. TNF/TNFR1 pathway playing pleiotropic roles in the development of many cancers, including NSCLC [20, 21]. The impact of the TNF/TNFR1 signaling pathway on DDP-resistant NSCLC remains unclear.

In light of this context, the objective of this study was to investigate the role of EDN1 in DDP resistance in NSCLC and to elucidate the underlying mechanisms. The results could provide a promising therapeutic approach for managing NSCLC.

Methods and materials

Clinical study

Samples were harvested from 80 NSCLC patients undergoing DDP therapy from the Qidong People’s Hospital, Qidong Liver Cancer Institute, Affiliated Qidong Hospital of Nantong University. NSCLC patient tumors and adjacent paracancerous tissues (> 3 cm from malignant tissues) were harvested during surgical biopsy and surgical procedures, including 80 DDP-sensitive tissue samples and 80 DDP-resistant tissue samples, which were immediately stored at -80℃ for further analysis. All patients who participated in this study signed an informed consent form, and this study was approved by the Ethics Committee of Qidong People’s Hospital, Qidong Liver Cancer Institute, Affiliated Qidong Hospital of Nantong University. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013).

Bioinformatics analysis

The datasets GSE108214 and GSE135720 were obtained from the GEO database (https://www.ncbi.nlm.nih.gov/geo/). Data analysis was performed using the R programming language, with differentially expressed genes (DEGs) identified based on the criteria of p < 0.05 and|log2(fold change)|>1. KEGG pathway enrichment analysis was carried out utilizing the DAVID database (https://david.ncifcrf.gov/) and visualized through the clusterProfiler package in R software.

Antibodies

For Western blot: GAPDH (1/2500, ab9485, Abcam), TNFR1 (1/1000, ab223352, Abcam, Cambridge, MA, USA); TNFAIP3 ((1/1000, ab92324, Abcam); MAP2K6 (1/2000, K108065P, Thermo Fisher Scientific, Waltham, MA, USA), goat anti-rabbit IgG (1/2000, ab6721, Abcam).

For immunohistochemistry (IHC): EDN1 (5 µg/mL, ab113697, Abcam); Ki67 (1 µg/mL, ab15580, Abcam); and TNFR1 (1/100, ab223352, Abcam).

Cell culture

A549 cells and their DDP resistant cells (A549/DDP) were sourced from Procell Life Technology Co., LTD, (Wuhan, China). A549 cells were maintained in Ham’s F-12 K medium (Procell) containing 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin mixture. A549/DDP cells were grown in Ham’s F-12 K medium containing 10% FBS, 1 µg/mL DDP (Yuanye Biotechnology Co., LTD, Shanghai, China) and 1% penicillin/streptomycin mixture.

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

Total RNA was extracted from cells using the commercial RNA Easy Fast Extraction kit (Tiangen Biochemical Technology Co., LTD, Beijing, China). HiScript III 1st Strand cDNA Synthesis Kit (Vazyme Biotechnology Co., LTD, Nanjing, China) was used to reverse transcribe RNA into cDNA and ChamQ Universal SYBR qPCR Master Mix (Vazyme) was used for qPCR amplification experiment. Gene primes were obtained from Genescript Biotechnology Technology Co., LTD, (Nanjing, China) and listed in Table 1 and 2. This experiment consisted of three biological replicates.

Table 1 Primer sequences used in qPCR
Table 2 Correlation analysis of EDN1 expression level and clinical features

Cell transfection

EDN1 short hairpin RNA (sh-EDN1) and its negative control (sh-NC) as well as overexpression vector (pcDNA3.1-TNFR1) and the empty vector (pcDNA3.1-NC) were sourced from Genescript. Transfection was conducted utilizing Lipofectamine 3000 (Vazyme) in accordance with the supplied guidelines when the cell confluence approached approximately 80%, for a duration of 48 h.

Cell counting kit-8 (CCK-8) assay

Cell viability was detected by a commercial CCK-8 kit (Beyotime). A549 and A549/DDP cells were seeded into 96-well plates (Beyotime) at the density of 1 × 103 cells/well and treated with DDP at the dose of 1, 2, 4, 8, 16, 32, or 64 µmol/L for 48 h at 37˚C, respectively. Then, 90 µL of Ham’s F-12 K medium and 10 µL of CCK-8 solution were added into each well of the plates to incubate for 2 h in the incubator after the medium with DDP were removed. The 50% inhibiting concentration (IC50) value was calculated via GraphPad Prism (v8.0.1). Additionally, A549 and A549/DDP cells were maintained in the incubator for 24 h at 37˚C. After that, 10 µL of CCK-8 solution was added to incubate with cells for 2 h at 37˚C. The absorbance was assessed at 450 nm using a microplate reader (Thermo Fisher). This experiment consisted of three biological replicates.

Lactic dehydrogenase (LDH) release

A549 and A549/DDP cells (1 × 105 cells/mL) were seeded on 96-well plates, followed by centrifugation to collect the supernatants. LDH assay kit (ab65393, Abcam) was utilized to assess the release of LDH in the supernatant of the cell culture medium, following the guidelines provided by the manufacturer. Finally, the colorimetric change of cells at the absorbance of 492 nm was determined using a microplate reader.

Transwell migration and invasion assays

A549 and A549/DDP cells were resuspended in a serum-free medium. For the migration assay, a cell suspension of 5 × 10^4 cells in 100 µL was placed in the chamber without Matrigel. In the case of the invasion assay, the same number of cells was seeded in a chamber containing 100 µL of Matrigel. The lower chamber was filled with Ham’s F-12 K medium enriched with 10% FBS to serve as a chemoattractant. Following a 24-h incubation period, the cells on the outer surface were rinsed with PBS, fixed using 4% paraformaldehyde (Beyotime) for 20 min, and subsequently stained with a dye solution of 0.1% crystal violet (Beyotime) for visualization purposes. This experiment consisted of three biological replicates.

Western blot

Total protein was extracted by a commercial kit (Solarbio, Beijing China) and the BCA method (Vazyme) was used for protein concentration detection. Then, 50 µg of protein was subjected to separation via 10% SDS-PAGE (Beyotime) and subsequently transferred to a PVDF membrane (Vazyme). Following this, the membrane was blocked with 5% skim milk for one hour. The membrane was then incubated overnight at 4 °C with primary antibodies obtained from a commercial source. After three washes with TBST (Thermo Fisher), the membrane was treated with the secondary antibody for one hour at room temperature. Finally, protein signal detection was carried out using an enhanced chemiluminescence solution (Vazyme). This experiment consisted of three biological replicates.

Enzyme-linked immunosorbent assay (ELISA)

Commercial ELISA kits (Beyotime) were used for TNF-α, IL-6, and IL-1β concentration detection in accordance with the manufacturer’s guidelines. Absorbance readings were obtained utilizing a microplate reader. This experiment consisted of three biological replicates.

Xenograft mouse model establishment

Forty male BALB/c nude mice, aged 6 to 8 weeks and weighing approximately 25 g, were acquired from Charles River (Beijing, China). Following a one-week acclimatization period, the mice were randomly assigned to four groups (n = 10). Subcutaneous injections were administered in the left flank, using A549 or A549/DDP cells that had been stably transfected with either sh-NC or sh-EDN1 plasmids, at a concentration of 1 × 10^6 cells in 100 µL. After a duration of 28 days, the mice were euthanized with pentobarbital sodium at a dosage of 40 mg/kg (Sigma), and the tumors were subsequently excised, weighed, and preserved at -80℃.

Hematoxylin-eosin (H&E) staining

The tumor tissues in each group (n = 10) were preserved in 4% paraformaldehyde for a duration of 24 h, followed by dehydration using ethanol (Beyotime) and embedding in paraffin. Subsequently, the paraffin-embedded tissues were sectioned into slices of 4 μm thickness. The sections were then stained with hematoxylin (Beyotime) for 5 min and eosin (Beyotime) for 3 min. In conclusion, the sections were dehydrated and sealed, and images were captured using a biopathology microscope (Olympus, Japan).

IHC

Paraffin-embedded tumor tissues in each group (n = 10) were sectioned, dewaxed, and rehydrated before being treated with 3% hydrogen peroxide. Subsequently, the sections were subjected to treatment with 10 mM citrate buffer (pH 6.0, Beyotime), heated to 95 °C, and blocked at room temperature. Following this, the sections were incubated with primary antibodies overnight at 4 °C, and then were treated with goat anti-rabbit IgG at 37 °C for 1 h. The sections were then counterstained with hematoxylin (Beyotime) and blued using 1% ammonia water after the addition of DAB solution (Vazyme). Finally, the sections were dehydrated, sealed, and examined under a light microscope (Leica, Wetzlar, Germany).

Statistical analysis

The analysis of data was conducted using SPSS version 21.0. The results are presented as mean ± standard deviation (SD). For comparisons between two groups, Student’s t-test was employed, while ANOVA was utilized for comparisons among three groups. Statistical evaluations were carried out using GraphPad Prism (version 8.0.1). A p-value of less than 0.05 was considered indicative of a statistically significant difference.

Results

EDN1 was an upregulated gene in DDP-resistant NSCLC

The GSE108214 and GSE135720 datasets in GEO database were used to obtain DEGs in DDP-resistant NSCLC. The findings indicated that EDN1 was upregulated in DDP-resistant NSCLC (Fig. 1A and B). No previous studies have been conducted on EDN1 in NSCLC; consequently, this research seeks to explore the role of EDN1 in NSCLC that is resistant to DDP. The RT-qPCR analysis revealed that resistant NSCLC patients exhibited markedly higher expression level of EDN1 in NSCLC tissues compared to their sensitive counterparts (Fig. 1C). Furthermore, as detailed in Table 1, a cohort of 80 NSCLC patients was stratified into two groups, High (n = 40) and Low (n = 40), based on the expression levels of EDN1. Notably, there were no significant differences between these groups concerning age, sex, and the number of tumors. However, a greater proportion of patients in the High group presented with tumor sizes of ≥ 50 mm, advanced III/IV tumor stages, and evidence of distant metastasis when compared to those in the Low group.

Fig. 1
figure 1

EDN1 was an upregulated gene in DDP-resistant NSCLC. The A, GSE108214 and B, GSE135720 datasets in GEO database were used to obtain differentially expressed genes in DDP-resistant NSCLC; C, RT-qPCR was performed to detect the mRNA level of EDN1 in the sensitive (n = 80) and resistant (n = 80) groups. GEO, Gene Expression Omnibus; DDP, cisplatin; NSCLC, non-small cell lung cancer; RT-qPCR, reverse transcription-quantitative polymerase chain reaction; EDN1, endothelin 1

A549/DDP cell lines showed upregulated EDN1 expression, cell viability, invasion, migration, and inflammation

To assess the DDP resistance of the acquired A549/DDP cell lines, CCK-8 assay was conducted to evaluate cell viability. The findings revealed that A549/DDP cell viability was significantly lower compared to that of A549 cells. Additionally, the IC50 values for A549/DDP cells exceeded those of A549 cells (Fig. 2A), indicating that the A549/DDP cell lines exhibit resistance to DDP. Besides, A549/DDP cells showed an upregulated EDN1 mRNA level compared with A549 cells (Fig. 2B). Transwell migration and invasion assays results indicated that DDP-resistance A549 cells exhibited increases in invasion and migration in comparison with A549 cells (Fig. 2C-E). Furthermore, A549/DDP cells exhibited elevated levels of TNF-α, IL-6, and IL-1β in comparison to A549 cells (Fig. 2F-H), indicating that DDP resistance heightened the inflammatory response in A549 cells.

Fig. 2
figure 2

A549/DDP cell lines showed upregulated EDN1 expression, cell viability, invasion, migration, and inflammation. A, CCK-8 assay was performed to assess the cell viability of A549 and A549/DDP cells; B, Quantification analysis of EDN1 in A549 and A549/DDP cells; C, Transwell invasion assay was performed to detect the cell invasion in A549 and A549/DDP cells; D, The number of invaded cells in A549 and A549/DDP cells; E, Transwell migration assay was performed to detect the cell migration in A549 and A549/DDP cells; ELISA was used to analyze the concentrations of F, TNF-α, G, IL-6, and H, IL-1β in A549 and A549/DDP cells. (n = 3; **p < 0.01; ***p < 0.001).DDP, cisplatin; CCK-8, cell counting kit-8; TNF-α, tumor necrosis factor α; IL, interleukin; ELISA, enzyme-linked immunosorbent assay; EDN1, endothelin 1

Silencing EDN1 decreased the cell viability, invasion, migration, and inflammation and increased cytotoxicity in A549/DDP cells

To investigate the impact of EDN1 on the development of DDP resistance, we introduced sh-EDN1 into A549/DDP cells, resulting in the suppression of EDN1 expression (Fig. 3A). Furthermore, the suppression of EDN1 resulted in a reduction of cell viability and an increased of LDH release when compared to the sh-NC group in A549/DDP cells, suggesting that loss of EDN1 increased cytotoxicity in A549/DDP cells. Besides, EDN1 inhibition downregulated the cell invasion, migration, and levels of inflammatory cytokines (TNF-α, IL-6, and IL-1β) in A549/DDP cells compared with the sh-NC group (Fig. 3B-G).

Fig. 3
figure 3

Silencing EDN1 decreased the cell viability, invasion, migration, and inflammation and increased cytotoxicity in A549/DDP cells. A, RT-qPCR was used to detect the expression of EDN1 in A549/DDP cells after EDN1 silenced; B, The cell viability was assessed by CCK-8 assay; C, LDH release in each group was analyzed by a commercial kit; Detection and quantification of cell D, invasion and E, migration; The levels of F, TNF-α, G, IL-6, and H, IL-1β in A549 and A549/DDP cells were analyzed by ELISA. (n = 3; ***p < 0.001 vs. sh-NC group or A549 group; ##p < 0.01 and ###p < 0.001 vs. A549/DDP + sh-NC group).DDP, cisplatin; RT-qPCR, reverse transcription-quantitative polymerase chain reaction; TNF-α, tumor necrosis factor α; IL, interleukin; ELISA, enzyme-linked immunosorbent assay; CCK-8, cell counting kit-8; LDH, lactate dehydrogenase; EDN1, endothelin 1; shRNA, short hairpin RNA

EDN1 affected DDP-resistant NSCLC by regulating TNF signaling pathway

Through the construction of the Venn diagram, we obtain the differentially expressed genes that were co-expressed in the two datasets (GSE108214 and GSE 135720) (Fig. 4A). KEGG enrichment analysis was conducted to assess gene functional enrichment. The findings revealed that the 15 differentially expressed genes were predominantly associated with TNF signaling pathways (Fig. 4B). Thus, we analyzed the related protein levels of TNF signaling pathways using Western blot, and the results found EDN1 inhibition decreased the protein levels of TNFR1, TNFAIP3, and MAP2K6 (Fig. 4C), illustrating that EDN1 affected DDP-resistant NSCLC by regulating TNF signaling pathway.

Fig. 4
figure 4

EDN1 affected DDP-resistant NSCLC by regulating TNF signaling pathway. A, The Venn diagram was used to screen genes that were upregulated in both GSE108214 and GSE135720 datasets; B, The KEGG pathway enrichment analysis. The color indicates the p-value (from the lowest in green to the highest in red), and the bubble size indicates the number of genes; C, The related protein levels of TNF signaling pathway were assayed by Western blot (n = 3). KEGG, Kyoto Encyclopedia of Genes and Genomes; TNF, tumor necrosis factor; EDN1, endothelin 1

Overexpressing of TNFR1 increased cell viability, invasion, migration, and inflammation in A549/DDP cells

To investigate the role of TNFR1 in the progression of DDP-resistant NSCLC, we introduced pcDNA-3.1-TNFR1 into A549/DDP cells, resulting in an elevated expression of TNFR1 (Fig. 5A). Furthermore, the overexpression of TNFR1 counteracted the reduction in cell viability, invasion, migration, and levels of inflammatory cytokines (TNF-α, IL-6, and IL-1β) that were caused by the silencing of EDN1 in A549/DDP cells (Fig. 5B-G).

Fig. 5
figure 5

Overexpressing of TNFR1 reversed the decreased cell viability, invasion, migration, and inflammation induced by silencing EDN1 in A549/DDP cells. A, RT-qPCR was performed to detect the mRNA level of TNFR1 in A549/DDP cells after TNFR1 overexpression; B, The cell viability in each group was assessed by CCK-8 assay; Detection and quantification of cell C, invasion and D, migration in each group; The levels of E, TNF-α, F, IL-6, and G, IL-1β in A549 and A549/DDP cells were assessed using ELISA. (n = 3; **p < 0.01 and ***p < 0.001 vs. pcDNA3.1-NC group or sh-NC group; #p < 0.05, ##p < 0.01, and ###p < 0.001 vs. sh-EDN1 + pcDNA3.1-NC group). RT-qPCR, reverse transcription-quantitative polymerase chain reaction; TNF-α, tumor necrosis factor α; IL, interleukin; ELISA, enzyme-linked immunosorbent assay; CCK-8, cell counting kit-8; EDN1, endothelin 1; DDP, cisplatin; shRNA, short hairpin RNA; TNFR1, tumor necrosis factor receptor-1

Silencing EDN1 inhibited tumor growth of mice

Finally, the in vivo findings revealed that the silencing of EDN1 resulted in a reduction of both tumor size and weight in A549 and A549/DDP cells (Fig. 6A and B). H&E results indicated that EDN1 inhibition alleviated the outcomes of cytoplasmic reduction, cytoboundary ambiguity, and cell necrosis. In addition, IHC assay showed that silencing EDN1 downregulated the protein levels of EDN1, Ki67, and TNFR1 (Fig. 6C).

Fig. 6
figure 6

Silencing EDN1 inhibited tumor growth of mice. A, The tumor size in each group; B, Tumor weight in each group; C, H&E and IHC assays were performed to assess the pathological status of tumor tissue and the protein levels of EDN1, Ki67, and TNFR1 in each group. (n = 10; ***p < 0.001 vs. A549 + sh-NC group; ###p < 0.001 vs. A549/DDP + sh-NC group). H&E, hematoxylin-eosin; sh-RNA, short hairpin RNA; IHC, immunohistochemistry; TNFR1, tumor necrosis factor receptor-1; DDP, cisplatin

Discussion

The emergence of DDP resistance in patients with NSCLC represents a significant challenge in the treatment of this disease. As cancer cells develop resistance to DDP, their sensitivity to the drug diminishes, leading to a situation where higher doses may be required to achieve the same therapeutic effect. This reduced sensitivity can occur through various mechanisms, including alterations in drug uptake and efflux, changes in DNA repair pathways, and modifications in cellular signaling that promote survival despite the presence of the drug [22, 23].

Research indicates that the upregulation of EDN1 could contribute to various cellular processes that promote tumor survival and proliferation despite the presence of chemotherapeutic agents. EDN1 is recognized for its role in pathways that govern cell proliferation, angiogenesis, and apoptosis, all of which are essential elements in the advancement of cancer and the response to treatment [24, 25]. In our study, GEO databases showed that EDN1 was an upregulated gene in DDP-resistant NSCLC. Similarly, Pulido et al. [16] indicate that secretion of EDN1 mediates drug resistance in epidermal growth factor receptor mutant NSCLC by promoting vasoconstriction, limiting blood and drug delivery. Besides, we found that DDP-resistance upregulated EDN1 expression, cell viability, invasion, migration, and inflammation in A549 cells, suggesting that DDP-resistance promoted the malignant progression of NSCLC. Similarly, other DDP-resistant tumor cells also show greater migration and invasion abilities and increased inflammation levels [26, 27]. EDN1 is a crucial peptide in cancer progression and is upregulated in different cancers [13, 28]. In the current investigation, the inhibition of EDN1 resulted in a reduction of cell viability, invasion, migration, and inflammation in A549/DDP cells, aligning with findings from earlier research [29]. In addition, among other lung diseases, EDN1 has been shown to be associated with pulmonary airway dysfunction [30], idiopathic pulmonary fibrosis [31], and pulmonary inflammation [32].

To further explore the mechanisms of EDN1, we used the KEGG database for enrichment function analysis and found that differentially expressed genes were mainly involved in TNF signaling pathways. The role of TNF signaling pathways in pathophysiology in different cancers has been intensively investigated [33]. We found that related protein levels of TNF signaling pathways were decreased after EDN1 silenced and overexpression of TNFR1 reversed the decreased cell viability, invasion, migration, and inflammation induced by silencing EDN1 in A549/DDP cells, implying that EDN1 facilitated DDP resistance of NSCLC by regulating the TNF signaling pathway. TNF-α can bind to two different receptors, TNFR1 and TNFR2 [34]. TNFR1, a member of TNFR superfamily, is a 455 amino acid polypeptide that contains a death domain in its C-terminal region [35]. Similar with our results, a recent review has indicated that TNFR1 overexpression promotes cell dedifferentiation and metastasis in lung squamous cell carcinoma [35]. In addition, Yang et al. demonstrates that upregulation of squalene synthase promotes lung cancer metastasis by increasing TNFR1 expression [36]. Interestingly, inflammation has been described as a favorable environment for tumor development in breast cancer, and TNF-α alone or in combination with chemotherapy and radiotherapy can be used as adjuvant treatment for breast cancer [37], implying that TNF signaling pathway plays different roles in different tumors. Moreover, the results of in vivo study demonstrated that silencing EDN1 inhibited tumor growth of mice and the protein levels of EDN1 and TNFR1, which was consistent with our in vitro study.

In summary, this research indicated that EDN1 enhances DDP resistance in NSCLC through the modulation of the TNF signaling pathway, potentially offering a novel reference for the treatment of NSCLC. Nevertheless, it is necessary to note that the findings of this study were validated solely through cellular and animal experiments, lacking clinical verification. This limitation will be addressed in future comprehensive investigations.

Data availability

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

References

  1. Celik B, Bulut T, Khoor A. Subtyping of non-small cell lung cancer by cytology specimens: a proposal for resource-poor hospitals. Cytojournal. 2019;16:8.

    Article  PubMed  PubMed Central  Google Scholar 

  2. You D, Wang D, Wu Y, Chen X, Shao F, Wei Y, et al. Associations of genetic risk, BMI trajectories, and the risk of non-small cell lung cancer: a population-based cohort study. BMC Med. 2022;20:203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Chen P, Liu Y, Wen Y, Zhou C. Non-small cell lung cancer in China. Cancer Commun (Lond). 2022;42:937–70.

    Article  PubMed  Google Scholar 

  4. Makovec T. Cisplatin and beyond: molecular mechanisms of action and drug resistance development in cancer chemotherapy. Radiol Oncol. 2019;53:148–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Chen TY, Zhou J, Li PC, Tang CH, Xu K, Li T, et al. SOX2 knockdown with siRNA reverses cisplatin resistance in NSCLC by regulating APE1 signaling. Med Oncol. 2022;39:36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. She K, He S, Lu X, Yu S, Li M, Xiong W, et al. LncRNA SNHG7 promotes non-small cell lung cancer progression and cisplatin resistance by inducing autophagic activity. J Thorac Dis. 2023;15:155–67.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Zhao X, Wu X, Wang H, Lai S, Wang J. Targeted therapy for cisplatin-resistant lung cancer via aptamer-guided nano-zinc carriers containing USP14 siRNA. MedComm (2020) 2023; 4:e237.

  8. Liao XZ, Gao Y, Zhao HW, Zhou M, Chen DL, Tao LT, et al. Cordycepin reverses Cisplatin Resistance in Non-small Cell Lung Cancer by activating AMPK and Inhibiting AKT Signaling Pathway. Front Cell Dev Biol. 2020;8:609285.

    Article  PubMed  Google Scholar 

  9. Jiang Z, Yang Y, Yang Y, Zhang Y, Yue Z, Pan Z, et al. Ginsenoside Rg3 attenuates cisplatin resistance in lung cancer by downregulating PD-L1 and resuming immune. Biomed Pharmacother. 2017;96:378–83.

    Article  CAS  PubMed  Google Scholar 

  10. Yanagisawa M, Kurihara H, Kimura S, Tomobe Y, Kobayashi M, Mitsui Y, et al. A novel potent vasoconstrictor peptide produced by vascular endothelial cells. Nature. 1988;332:411–5.

    Article  CAS  PubMed  Google Scholar 

  11. Banecki K, Dora KA. Endothelin-1 in Health and Disease. Int J Mol Sci 2023; 24.

  12. Jenkins HN, Rivera-Gonzalez O, Gibert Y, Speed JS. Endothelin-1 in the pathophysiology of obesity and insulin resistance. Obes Rev. 2020;21:e13086.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Tocci P, Blandino G, Bagnato A. YAP and endothelin-1 signaling: an emerging alliance in cancer. J Exp Clin Cancer Res. 2021;40:27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Freeman BD, Machado FS, Tanowitz HB, Desruisseaux MS. Endothelin-1 and its role in the pathogenesis of infectious diseases. Life Sci. 2014;118:110–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Khalid W, Vargheese SS, Lakshmanan R, Sankari M, Jayakumar ND. Role of endothelin-1 in periodontal diseases: a structured review. Indian J Dent Res. 2016;27:323–33.

    Article  PubMed  Google Scholar 

  16. Pulido I, Ollosi S, Aparisi S, Becker JH, Aliena-Valero A, Benet M, et al. Endothelin-1-Mediated Drug Resistance in EGFR-Mutant Non-small Cell Lung Carcinoma. Cancer Res. 2020;80:4224–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Chen JL, Lv XD, Ma H, Chen JR, Huang JA. Detection of cancer embryo antigen and endothelin-1 in exhaled breath condensate: a novel approach to investigate non-small cell lung cancer. Mol Clin Oncol. 2016;5:124–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Salomon BL, Leclerc M, Tosello J, Ronin E, Piaggio E, Cohen JL. Tumor Necrosis Factor Alpha and Regulatory T Cells in Oncoimmunology. Front Immunol. 2018;9:444.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Holbrook J, Lara-Reyna S, Jarosz-Griffiths H, McDermott M. Tumour necrosis factor signalling in health and disease. F1000Res 2019; 8.

  20. Niiya M, Niiya K, Kiguchi T, Shibakura M, Asaumi N, Shinagawa K, et al. Induction of TNF-alpha, uPA, IL-8 and MCP-1 by doxorubicin in human lung carcinoma cells. Cancer Chemother Pharmacol. 2003;52:391–8.

    Article  PubMed  Google Scholar 

  21. Xiao Z, Shi G, Xi S, Singh AK, Willette-Brown J, Li X, et al. A TNFR1-UBCH10 axis drives lung squamous cell carcinoma dedifferentiation and metastasis through a cell-autonomous signaling loop. Cell Death Dis. 2022;13:885.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Du J, Chen W, Yang L, Dai J, Guo J, Wu Y, et al. Disruption of SHH signaling cascade by SBE attenuates lung cancer progression and sensitizes DDP treatment. Sci Rep. 2017;7:1899.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Wang S, Liu F, Zhu J, Chen P, Liu H, Liu Q, et al. DNA repair genes ERCC1 and BRCA1 expression in Non-small Cell Lung Cancer Chemotherapy Drug Resistance. Med Sci Monit. 2016;22:1999–2005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lee YJ, Jung E, Choi J, Hwang JS, Jeong EJ, Roh Y et al. The EDN1/EDNRA/beta–arrestin axis promotes colorectal cancer progression by regulating STAT3 phosphorylation. Int J Oncol 2023; 62.

  25. Zhu Y, Liu W, Wang Z, Wang Y, Tan C, Pan Z, et al. ARHGEF2/EDN1 pathway participates in ER stress-related drug resistance of hepatocellular carcinoma by promoting angiogenesis and malignant proliferation. Cell Death Dis. 2022;13:652.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Liao J, Yang Z, Carter-Cooper B, Chang ET, Choi EY, Kallakury B, et al. Suppression of migration, invasion, and metastasis of cisplatin-resistant head and neck squamous cell carcinoma through IKKbeta inhibition. Clin Exp Metastasis. 2020;37:283–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Liu Y, Han S, Li Y, Liu Y, Zhang D, Li Y, et al. MicroRNA-20a contributes to cisplatin-resistance and migration of OVCAR3 ovarian cancer cell line. Oncol Lett. 2017;14:1780–6.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Kaplan M, Kilinc A, Ozturk S, Ilhan N, Gurocak S, Gonen M. Effect of radiation on brain tissue endothelin-1 level and tumor development. Asian J Neurosurg. 2017;12:185–8.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Zhang ZY, Chen LL, Xu W, Sigdel K, Jiang XT. Effects of silencing endothelin-1 on invasion and vascular formation in lung cancer. Oncol Lett. 2017;13:4390–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Low A, George S, Howard L, Bell N, Millar A, Tulloh R. Lung function, inflammation, and Endothelin-1 in congenital heart Disease-Associated Pulmonary arterial hypertension. J Am Heart Assoc 2018; 7.

  31. Pulito-Cueto V, Genre F, Lopez-Mejias R, Mora-Cuesta VM, Iturbe-Fernandez D, Portilla V et al. Endothelin-1 as a biomarker of idiopathic pulmonary fibrosis and interstitial lung Disease Associated with Autoimmune diseases. Int J Mol Sci 2023; 24.

  32. Manitsopoulos N, Nikitopoulou I, Maniatis NA, Magkou C, Kotanidou A, Orfanos SE. Highly selective Endothelin-1 receptor a inhibition prevents Bleomycin-Induced Pulmonary inflammation and fibrosis in mice. Respiration. 2018;95:122–36.

    Article  CAS  PubMed  Google Scholar 

  33. Manohar SM. At the crossroads of TNF alpha signaling and Cancer. Curr Mol Pharmacol. 2024;17:e060923220758.

    PubMed  Google Scholar 

  34. Benoot T, Piccioni E, De Ridder K, Goyvaerts C. TNFalpha and Immune Checkpoint Inhibition: friend or foe for Lung Cancer? Int J Mol Sci 2021; 22.

  35. Shi G, Hu Y. TNFR1 and TNFR2, which link NF-kappaB activation, Drive Lung Cancer Progression, Cell Dedifferentiation, and Metastasis. Cancers (Basel) 2023; 15.

  36. Yang YF, Jan YH, Liu YP, Yang CJ, Su CY, Chang YC, et al. Squalene synthase induces tumor necrosis factor receptor 1 enrichment in lipid rafts to promote lung cancer metastasis. Am J Respir Crit Care Med. 2014;190:675–87.

    Article  PubMed  Google Scholar 

  37. Martinez-Reza I, Diaz L, Garcia-Becerra R. Preclinical and clinical aspects of TNF-alpha and its receptors TNFR1 and TNFR2 in breast cancer. J Biomed Sci. 2017;24:90.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

The work was supported by Nantong City Science and Technology Bureau Project under grant number MSZ2023214.

Author information

Authors and Affiliations

Authors

Contributions

All authors participated in the design, interpretation of the studies and analysis of the data and review of the manuscript. J Z drafted the work and revised it critically for important intellectual content; L S, L J, S Z and Y C were responsible for the acquisition, analysis, or interpretation of data for the work; Z L made substantial contributions to the conception or design of the work. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Zhenwei Li.

Ethics declarations

Ethics approval and consent to participate

This study protocol was approved by the Ethics Committee of Qidong People’s Hospital, Qidong Liver Cancer Institute, Affiliated Qidong Hospital of Nantong University. The study was conducted in accordance with the Declaration of Helsinki (as revised in 2013). All animal experiments should comply with the ARRIVE guidelines. All methods were carried out in accordance with relevant guidelines and regulations.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhen, J., Sun, L., Ji, L. et al. EDN1 facilitates cisplatin resistance of non-small cell lung cancer cells by regulating the TNF signaling pathway. World J Surg Onc 23, 71 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12957-025-03692-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12957-025-03692-7

Keywords