Immune Netw. 2023 Aug;23(4):e34. English.
Published online Aug 11, 2023.
Copyright © 2023. The Korean Association of Immunologists
Original Article

Tumor Promoting Function of DUSP10 in Non-Small Cell Lung Cancer Is Associated With Tumor-Promoting Cytokines

Xing Wei,1, Chin Wen Png,2,3, Madhushanee Weerasooriya,2,3 Heng Li,2,3 Chenchen Zhu,1 Guiping Chen,1 Chuan Xu,4 Yongliang Zhang,2,3 and Xiaohong Xu1
    • 1Breast Surgery Department, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China.
    • 2Department of Microbiology and Immunology, and NUSMED Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore.
    • 3Immunology Programme, Institute of Life Sciences, National University of Singapore, Singapore 117545, Singapore.
    • 4Department of Oncology & Cancer Institute, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 610056, China.
Received April 30, 2023; Revised July 09, 2023; Accepted July 31, 2023.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Lung cancer, particularly non-small cell lung cancer (NSCLC) which contributes more than 80% to totally lung cancer cases, remains the leading cause of cancer death and the 5-year survival is less than 20%. Continuous understanding on the mechanisms underlying the pathogenesis of this disease and identification of biomarkers for therapeutic application and response to treatment will help to improve patient survival. Here we found that a molecule known as DUSP10 (also known as MAPK phosphatase 5) is oncogenic in NSCLC. Overexpression of DUSP10 in NSCLC cells resulted in reduced activation of ERK and JNK, but increased activation of p38, which was associated with increased cellular growth and migration. When inoculated in immunodeficient mice, the DUSP10-overexpression NSCLC cells formed larger tumors compared to control cells. The increased growth of DUSP10-overexpression NSCLC cells was associated with increased expression of tumor-promoting cytokines including IL-6 and TGFβ. Importantly, higher DUSP10 expression was associated with poorer prognosis of NSCLC patients. Therefore, DUSP10 could severe as a biomarker for NSCLC prognosis and could be a target for development of therapeutic method for lung cancer treatment.

Keywords
Dual specificity phosphatase; Non-small cell lung cancer; Mitogen-activated protein kinases; Cytokine

INTRODUCTION

Lung cancer remains the leading cause of cancer death globally, and is the most frequently diagnosed cancer and cause of cancer death in men and women combined (1). For instance, in 2018, 2.09 million new cases of lung cancer were diagnosed and the number of lung cancer death was 1.76 million which contributed 18.4% to total cancer death. The two most common types of lung cancer are small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). The NSCLC which can be histologically divided into adenocarcinoma, large-cell carcinoma and squamous cell carcinoma (2), makes up about 80% of lung cancer cases. Despite advances in diagnosis and treatment, the overall survival of lung cancer patients remains poor. Early diagnosis, suitable and effective treatment would help to improve lung cancer patient survival.

Like various other types of cancer, the development of lung cancer is accompanied by accumulation of genetic mutations in both oncogenes and tumor suppressor genes (3). Among them, mutations in the epidermal growth factor receptor (EGFR) were found in about 10%–15% of Caucasian NSCLC patients and in about 30%–50% of Asian patients (4). EGFR belongs to the HER/erbB family of receptor tyrosine kinases, which includes HER1, HER2, HER3 and HER4 (5). Binding of their ligands results in dimerization and autophosphorylation of the cytoplasmic domain of the receptor, which coupled the receptor to downstream signaling pathways that ultimately leads to increased proliferation, angiogenesis, metastasis, and decreased apoptosis. Dysregulation of EGFR tyrosine kinase activity could be due to EGFR gene mutation, increased gene copy number, and EGFR protein overexpression. All of these could result in constitutive and oncogenic activation of the receptor, contributing to tumor growth and progression. The RAS/RAF/MEK/ERK is one of the main signaling pathways downstream of EGFR. Activation of the RAS GTPase downstream of EGFR results in a signaling cascade through phosphorylation, leading to the activation of RAF, MEK, and eventually the ERK1/2 phosphor-mitogen-activated protein (MAP) kinases. In addition to EGFR, RAS and RAF are also among the most commonly mutated genes in human cancer. For instance, mutations in RAS were identified in about 35% of lung cancer patients (6), whereas BRAF mutations were identified in 3-5% of NSCLC (7). Together, these mutations confer constitutive activation of MEK1/2-ERK1/2 to promote oncogenic transformation.

ERK1/2 are known as central regulators of an array of cellular processes including cell proliferation, growth, survival, and migration (8). The biological outcome of ERK1/2 activation reflects a balance between the activity of upstream pathway components and various negative regulatory mechanisms (9). A group of proteins known as dual-specificity phosphatases (DUSPs), also known as MAPK phosphatases (MKPs) were identified as the main negative regulators of MAPKs including ERK1/2. These DUSPs/MKPs were also found to play important roles in not only cancer development, but also cancer resistance to treatment, thereby influencing patient prognosis (10, 11, 12). For example, DUSP16/MKP7 was found to regulate cancer cell resistance to chemodrugs through regulation of BAX (12). DUSP10/MKP5, on the other hand, suppresses colorectal cancer development though inhibition of ERK1/2 in gut epithelial cells (11). However, the role of DUSP10 in lung cancer is unclear. Here, we examined the function of this molecule in lung cancer development through analysis of human lung cancer patient samples, lung cancer cell lines with DUSP10 overexpression and xenograft animal models. Our results showed that DUSP10 is oncogenic in lung cancer.

MATERIALS AND METHODS

Clinical specimens and survival analysis

A retrospective analysis of 195 cases from the First Affiliated Hospital of Zhejiang Chinese Medical University and the Sichuan Provincial People’s Hospital with NSCLC who were histologically diagnosed and treated with standard regiments from January 2014 to January 2022 in hospital were included. The tumor specimens consist of 113 adenocarcinoma, 57 squamous cell carcinoma, 20 adenosquamous carcinoma and 5 large cell lung carcinoma. Informed consent was obtained from patients before standard treatment for specimens to be used for this research. The study was approved by Institutional Review Board of the First Affiliated Hospital of Zhejiang Chines Medical University (2023-KL-031-02). All samples were re-evaluated by experienced pathologist according to the World Health Organization grading system and the General Rules for Clinical Lung Cancer.

Formalin-fixed, paraffin-embedded tissue sections of 4-μm thick obtained from 182 primary lung tumor tissues were immunostained using the Gene Tech EnVision™ Detection Kit (GK500710, Gene tech, Shanghai, China). After deparaffinization and dehydration through graded alcohols and xylene, endogenous peroxidase activity and non-specific Ag were blocked with 3% hydrogen peroxide. The sections were heated for 5 min twice at 100°C with Tris EDTA (pH 9.0) in a microwave oven for Ag retrieval. All sections were incubated with anti-DUSP10 Ab (Abcam #ab228987) at 37°C for 1 h. After washing, the sections were incubated with the ChemMate™ Envision™/HRP, Rabbit (ENV) reagent at 37°C for 30 min. Rinsed gently with PBS, the sections were visualized by the ChemMate TM DAB+ Chromogen, counterstained with hematoxylin, mounted in neutral gum, and analyzed using a bright field microscope.

The immunohistochemically stained tissue sections were reviewed and scored by two independent experienced pathologists blinded to the clinicopathological variables. For each section, 5 high power field were random selected and the staining was evaluated by the extent (the percentage of positive tumor cells or normal epithelial cells in relation to the whole tissue area: negative, 0; ≤10%,1; 10%–50%,2; ≥51%, 3) and the intensity (absent 0; weak 1; moderate 2; strong 3). The combined score (extensity×intensity) of 0–4 and 5–9 were defined as low and high expression levels of DUSP10 respectively.

Descriptive statistics comparing DUSP10 expression with the clinicopathological characteristics were analyzed by the chi-square test. Survival curves were calculated by the Kaplan–Meier method, and the differences were assessed by the log-rank test. Statistical analysis software (IBM SPSS Statistic 19.0; IBM Corp., Armonk, NY, USA) was used to perform the analyses, and a p-value of less than 0.05 (p<0.05) was considered statistically significant.

Animal experiment

Animal experiments were approved by the Institutional Animal Care and Use Committee of National University of Singapore. NSGS mice were obtained from Jackson Laboratory and were inbreed for all xenograft experiments in this study. For establishment of NSCLC cell line xenograft model, 5×106 of H460 and HCC827 cells stably overexpressing DUSP10 were subcutaneously injected into the flanks of the animals. Tumor xenografts were allowed to develop over 3 wk before harvest.

Cell culture

The human NSCLC cell lines, H460 and HCC827 cells (American Type Culture Collection, Manassas, VA, USA), were maintained in complete Roswell Park Memorial Institute media, RPMI (Biowest, Nuaillé, France) containing 10% FBS and 1% penicillin/streptomycin (Invitrogen, Carlsbad, CA, USA), at 37°C with 5% CO2. Cell harvest was done by incubating cells with 0.25% Trypsin-EDTA (Gibco, Grand Island, NY, USA) at 37°C for 5 min, till the adherent cells are detached from the surface. Detached cells were subsequently pelleted by centrifugation at 400 g for 3 min. Total live Quantification of live cell count was carried out using trypan blue (HyClone, Cramlington, UK) staining.

Generation of H460 and HCC827 stably overexpressing DUSP10 cell lines

Empty pcDNA plasmid vector (control) or pcDNA plasmid containing the full-length of human DUSP10 cDNA were transfected into both H460 and HCC827 cells using Lipofectamine® LTX and Plus™ Reagent (Invitrogen) according to the manufacturer’s instructions.

H460 and HCC827 cells that were positive for either the control vector or vector containing DUSP10 were identified based on geneticin (G418) (1.5 mg/ml) selection. The successfully transfected clones were selected and screened for DUSP10 overexpression by using quantitative real-time PCR (qPCR) to quantify DUSP10 mRNA level. Clones that stably overexpressed DUSP10 were maintained in complete RPMI medium with 1.5 mg/ml geneticin (G418).

Cell proliferation assay

H460- and HCC827-pcDNA control and H460- and HCC827-DUSP10 overexpression cells were seeded at a density of 0.08×106 cells/well in 6-well plates and incubated overnight at 37°C with 5% CO2. Subsequently, cells were incubated in serum free RPMI for 20 h at the same condition. After serum starvation, serum free RPMI was replaced with a complete RPMI for assessment of basal proliferation over a period of 5 days. At each time point, RPMI was removed and 500 μl crystal violet stain (0.5% crystal violet in 20% methanol) was added into the wells and was shaken for 5 min. Crytal violet stain was then removed, cells were washed with RO water and the plates were left to air-dry for 15 min. After drying, the stain was solubilized in 500 μl solubilisation buffer (0.1M sodium citrate in 50% absolute ethanol). Analysis of relative proliferation rate was done by measuring absorbance at 545nm using BioTek Synergy™ H1 Hybrid Multi-Mode Microplate Reader (BioTek Instruments, Winooski, VT, USA).

Spheroid formation assay

H460- and HCC827-pcDNA control and H460- and HCC827-DUSP10 overexpression cells were seeded at a density of 10,000 cells/well in 0.35% (w/v) sterile low melting agarose, in 48-well plates, which were pre-coated with 0.5% (w/v) sterile low melting agarose. Complete RPMI medium was added to each well immediately after the agar were solidified. Fresh culture medium was replaced every three days during culture. Spheroids were left to develop over 2 wk before microscopic assessment to determine spheroid sizes. Olympus IX80 inverted microscope (Olympus, Tokyo, Japan) was used to capture images of the spheroids, which were measured using ImageJ software.

Wound healing assay

H460- and HCC827-pcDNA control and H460- and HCC827-DUSP10 cells were harvested and the cell suspensions were adjusted to 0.8×106 cells/ml. A volume of 70 µL cell suspension was seeded into Ibidi culture insert (Ibidi, Fitchburg, WI, USA) forming a monolayer of cells. On the day of experiment, culture insert was removed before cells were treated with 10 μM of mitomycin-c (Sigma, St. Louis, MO, USA) for 2 h before imaging to determine the “wound” size at Time 0. Imaging was done after 24 h to determine percentage of “wound” closure for quantification of cell migration using Image J software (NIH, Bethesda, MD, USA).

Quantitative real-time polymerase chain reaction

Total RNA was extracted from cultured cells using TRizol (Invitrogen) and was used for cDNA synthesis using ImProm-II™ Reverse Transcription System (Promega, Madison, WI, USA). qPCR was performed with an Applied system 7900 Detection System using Fast SYBR Master Mix (Applied Biosystems, B.V., Singapore).

Western blot

Whole cell lysates were separated by 10% SDS-PAGE, and Western blotting was performed using antibodies against total and MAP kinases (Cell Signaling, Danvers, MA, USA), EGFR, c-Myc, COX2 (Santa Cruz, Santa cruz, CA, USA), and actin (Cell signaling). Immunoblots were developed with ECL donkey anti-rabbit IgG horseradish peroxidase-conjugated secondary antibodies (GE Healthcare, Chicago, IL, USA) and SuperSignal West Dura Chemiluminescent Substrate (Thermo Scientific, Waltham, MA, USA). The blots were exposed to Amersham® Hyperfilm® ECL™ and MP Autoradiography Films (GE Healthcare).

Statistical analysis

Data were expressed as the mean±SE. Statistical analysis was performed using two tailed Student’s unpaired t-test. A p-value <0.05 was considered to be statistically significant.

RESULTS

Overexpression of DUSP10 in lung cancer cells resulted in increased cell growth and migration

To understand the function of DUSP10 in lung cancer, particularly NSCLC, we generated DUSP10 overexpression HCC827 cells. Six clones were selected from vector- and DUSP10-overexpression cells respectively to examine DUSP10 expression using qPCR. The results showed increased expression of DUSP10 in the DUSP10-overexpression clones compared to that in vector-expression ones (Fig. 1A), showing the successful generation of DUSP10-overexpression HCC827 cells. One vector clone and two DUSP10-overexpression clones (clone 4 and 5) were selected to examine the effect of DUSP10 in cell growth using MTT assay. It was found that the two DUSP10-overexpression clones had significantly increased proliferation compared to that of vector clone (Fig. 1B). Next, to confirm the effect of DUSP10 overexpression on HCC827 growth, we examined the expression of DUSP10 protein in the vector clone and the HCC827 DUSP10-oveexpression clone 5 using western blot analysis. It was found that DUSP10 protein was not expressed in the vector-expression cells and obvious DUSP10 protein was detected in the overexpression clone, confirming the successful generation of the DUSP10-overexpression cells (Fig. 1C). Next, vector- and DUSP10-overexpression cells were cultured in the absence or presence of epidermal growth factor (EGF) over a period of three days followed by examination of cell growth by crystal violet staining. The results showed that DUSP10-overexpression cells had significantly increased growth compared to vector cells with or without EGF (Fig. 1D). Together, these results suggested that DUSP10 may promote cell growth in lung cancer.

Figure 1
Establishment of DUSP10-overexpression HCC827 stable cell clones. (A) NSCLC cell line HCC827 was transfected with pcDNA3 vector or pcDNA3 containing the full-length of human DUSP10 cDNA followed by drug selection for stably transfected clones. Six clones each from pcDNA3- and pcDNA3-DUSP10 transfected cells were selected for examination of DUSP10 expression by qPCR. (B) One vector clone and two DUSP10-overexpression clones (clone #4 & 5) were selected to examine the impact of DUSP10 on cell proliferation by MTT assay. (C) Western blot analysis of DUSP10 protein expression in vector and DUSP10-overexpression HCC827 cells (clone #5). (D) Vector- and DUSP10-overexpression HCC827 cells were cultured with or without hEGF (100 ng/ml) for various days indicated to assess cell proliferation by crystal violet assay. Data are representative of three independent experiments with similar results.
*p<0.05, **p<0.01, ***p<0.001.

To further exam the role of DUSP10 in lung cancer cells, we performed spheroid formation and wound healing assays. The results showed that overexpression of DUSP10 resulted increased sizes of spheroids (Fig. 2A). In addition, DUSP10-overexpression cells had greatly increased wound healing ability compared to that of vector cells (Fig. 2B), suggesting that overexpression of DUSP10 resulted in increased migration of lung cancer cells.

Figure 2
Overexpression of DUSP10 in NSCLC cells resulted in increased spheroid formation and wound healing. (A) Representative micrographs of multicellular spheroid aggregates, derived from vector- and DUSP10-overexpression HCC827 cells. Bar chart shows mean of spheroid size (n=30). (B) Representative micrographs showing wound closure in vector- and DUSP10-overexpression cells. Bar chart shows mean of percentage of wound closure (n=12). Results representative of three independent experiments.
***p<0.001.

To verify the function of DUSP10 in regulation of NSCLC cell growth and migration, we generated vector- and DUSP10-overexpression H460 cells (Supplementary Fig. 1A). The growth of vector- and DUSP10-overexpression cells with or without EGF over a period of 3 days was examined by crystal violet staining assay. In consistent with the role of DUSP10 in HCC827 cells, DUSP10-oveexpression H460 cells had increased growth compared to vector transfected cells with or without EGF treatment (Supplementary Fig. 1B). Next, spheroid formation and would healing assays were carried out. The results showed that overexpression of DUSP10 in H460 cells resulted in increased spheroid formation and wound healing (Supplementary Fig. 2A and B), which is consistent with the function of this molecule in HCC827 cells (Fig. 2). Together, these results demonstrated that DUSP10 promotes cell growth and migration in lung cancer.

DUSP10 regulates the activation of MAPKs and the expression of growth-related molecules

To understand the mechanism underlying the regulation of DUSP10 in lung cancer cells, we first examined the phosphorylation of ERK, JNK and p38 in both vector- and DUSP10-overexpression HCC827 cells with or without EGF stimulation. It was found that ERK was constitutively phosphorylated in vector-cells and its phosphorylation was slightly increased at 0.5 h, followed by reduction at 3 and 6 h upon EGF treatment (Fig. 3A and Supplementary Fig. 3). Slightly reduced phosphorylation of ERK was observed in DUSP10-overexpression cells without EGF stimulation. Upon EGF stimulation, ERK phosphorylation was greatly reduced at 0.5, 3 and 6 h in DUSP10-overexpression cells compared to that in vector-cells. JNK was not phosphorylated in both vector- and DUSP10-overexpression cells without stimulation (Fig. 3A and Supplementary Fig. 3). EGF stimulation induced JNK phosphorylation in both vector- and DUSP10-overexression cells at 0.5 h, but the level of phosphorylation was much lower in DUSP10-overexpression cells compared to that in vector cells. Phosphorylation of JNK was weaned off in both vector- and DUSP10-overexpression cells at 3 and 6 h. Interestingly, phosphorylation of p38 was found to be increased in DUSP10-overexpression cells compared to that in vector cells (Fig. 3A and Supplementary Fig. 3). Weakly phosphorylated p38 was detected in vector cells without EGF stimulation, increased phosphorylation was observed at 0.5 h upon EGF stimulation and the level of phosphorylation was further increased at 3 h followed by a reduction at 6 h upon EGF stimulation. In contrast, higher level of p38 phosphorylation was detected in DUSP10-overexpression cells compared to that in vector-cells without EGF stimulation. EGF stimulation resulted in a similar increase in p38 phosphorylation as that in vector cells, but the levels of phosphorylation were higher than that in vector-cells at each time point upon EGF stimulation. Together, these results suggest that DUSP10 negatively regulates ERK and JNK phosphorylation, but positively regulates p38 phosphorylation.

Figure 3
DUSP10 increases p38 activation and inhibits the activation of ERK and JNK as well as ERK-regulated growth promoting molecules. Vector- and DUSP10-overexpression HCC827 cells were stimulated with 100 ng/ml of EGF. Cells were harvested at the indicated time points to prepare protein lysates. (A) The levels of pERK, pJNK and pP38 as well as tERK, tJNK and tP38 were determined by western blot analysis. The expression of c-Myc, COX2 and actin was analyzed. (B) The proliferation of vector- and DUSP10-overexpression HCC827 cells with or without p38-specific inhibitor SB203580 was determined by MTT or crystal violet assays. (C) Representative micrographs showing wound closure in vector- and DUSP10-overexpression cells with or without SB203580 treatment. Bar chart shows mean of percentage of wound closure (n=12). Results representative of three independent experiments.
pERK, phospho-ERK; tERK, total-ERK.

***p<0.001.

Next, we examined the expression of cellular growth-related molecules including c-Myc and COX2. In consistent with reduced phosphorylation of ERK, the expression of both c-Myc and COX2 which are targets of ERK was reduced in DUSP10-overexpression cells compared to vector cells (Fig. 3A and Supplementary Fig. 3). These results demonstrated that DUSP10 inhibits ERK and ERK- regulated molecules which is consistent with our previous findings (11). However, the increased cell growth and migration in DUSP10-overexpression cells suggested that this molecule regulates lung cancer though other mechanisms, possibly through p38.

To test if p38 contributes to the increased growth and migration of lung cancer cells, we treated vector- and DUSP10-overexpression cells with p38 inhibitor SB203580 followed by examination of cell growth and migration. Western blot analysis showed the successful inhibition of p38 activation by the inhibitor (Supplementary Fig. 3B). Without p38 inhibition, DUSP10-overexpression HC827 cells had increased cell growth determined by both MTT and crystal violet assays (Fig. 3B), which was consistent with previous findings (Fig. 1). Upon inhibition, both the vector- and DUSP10-overexpression cells had reduced proliferation and the levels of proliferation became comparable between them. Similar results were obtained in vector- and DUSP10-overexpression H460 cells where overexpression increased cell growth and migration and p38 inhibition abrogated the differences in growth and migration between vector- and DUSP10-overexpression cells (Supplementary Fig. 4A and B). These results suggest that p38 plays an important role in NSCLC growth and development.

DUSP10 promotes lung cancer cell growth in vivo

To further understand the function of DUSP10 in lung cancer, vector- and DUSP10-overexpression cells were inoculated into NSG mice to access their growth. As shown in Fig. 4, DUSP10-overexpression cells formed significantly large tumors compared to these formed by vector-cells (Fig. 4A), demonstrating that DUSP10 promotes lung cancer cell growth in vivo.

Figure 4
DUSP10 promotes NSCLC growth in vivo. Vector- and DUSP10-overexpression HCC827 cells (A), or vector- and DUSP10-overexpression H460 cells (B) were inoculated into NSGS mice (n=5 for each cell type) respectively. After three weeks of growth, mice were sacrificed to assess tumor formation. Representative photographs of vector- and DUSP10-overexpression xenograft tumours and bar chart show the weights of the xenograft tumours. (C, D). Immunohistochemical analysis for p38 activation of vector- and DUSP10-overexpression HCC827 (C) and H460 (D) tumor sections. Representative images of phosphor-p38 and quantification of phosphor-p38 staining were shown (n=5). Scale bar = 100 µM. Results representative of two independent experiments with similar results.
*p<0.05, **p<0.01, ***p<0.001.

To confirm that the tumor promoting function of DUSP10 in NSCLC in vivo, we inoculated vector- and DUSP10-overexpression H46 cells inoculated into NSG mice. The results showed that larger tumors were formed by DUSP10-oeverexpression H460 cells compared to those formed by vector cells (Fig. 4B), which was consistent with the results from vector- and DUSP10-overexpression HCC827 cells (Fig. 4A). These results further support the oncogenic function of DUSP10 in NSCLC.

Next, we performed immunohistochemical staining to evaluate the activation of p38 and ERK. Consistent with western blot results (Fig. 3A), increased p38 staining was observed in DUSP10-overexpression xenografts compared to that in vector-transfected xenografts (Fig. 4C and D), whereas ERK staining was found to be reduced in xenografts with DUSP10-overexpression compared to that in vector-transfected ones (Supplementary Fig. 5). These results suggested that the increased growth of DUSP10-overexpression NSCLC was associated with increased p38 activation in vivo.

Higher DUSP10 expression in lung cancer patients is associated with poorer prognosis

Next, we examined the protein expression of DUSP10 in tumor tissues from 195 NSCLC cancer patients using immunohistochemistry staining. Based the signal, the samples were assigned a score from 0 to 9. Score 0 to 4 indicate low DUSP10 protein expression and sore 5–9 indicate high DUSP10 expression (Fig. 5A). Kaplan-Meier analysis was carried out to compare the survival of patients between DUSP10 low and DUSP10 high groups. It was found that patients with high DUSP10 protein expression had significant lower survival compared with patients with low DUSP10 (Fig. 5B). This result demonstrates that DUSP10 promotes lung cancer development and influences the clinical outcome of NSCLC.

Figure 5
Higher DUSP10 levels in tumors were associated with poorer prognosis of NSCLC patients. (A) Formalin-fixed, paraffin-embedded tissue sections from NSCLC patients were immunostained with Ab against human DUSP10. DUSP10 signal was viewed and scored from 0 to 3, with no DUSP10 staining as score 0, weak staining as 1, moderate staining as 2 and strong staining as 3. Representative photographs of score 0–3 of DUSP10 staining were shown. (B) Kaplan-Meier survival curve and log-rank (Mantel-Cox) test showing the survival of 195 NSCLC patients stratified by low or high DUSP10 expression in tumours cells.

Increased expression of IL-6 and TGFβ in DUSP10-overexpression lung cancer cells

Lung cancer cells are known to produce various cytokines which regulate tumor growth (13). In addition, p38 is known to be an important regulator of various cytokines. We therefore examine the regulation of cytokines by DUSP10 in lung cancer cells. Analysis of gene expression by qPCR confirmed that the expression of DUSP10 was significantly higher in DUSP10-overexpression HCC827 cells compared to that in vector cells (Fig. 6A). In addition, reduced expression of TNFα was observed in DUSP10-overexpression cells compared to that in vector cells, which is consistent with previous findings on the role of DUSP10 in the expression of these cytokines (14). Interestingly, the expression of TGFβ and IL-6 was found to be increased in DUSP10-overexpression cells compared to that in vector cells (Fig. 6A and B). To validate the role of p38 in regulation of these cytokines in NSCLC, SB203580 was used to treat vector- and DUSP10 overexpression cells followed by examination of TGFβ and IL-6 expression. The results confirmed the increased expression of these two cytokines in DUSP10-overexpression cells without p38 inhibition (Fig. 6C). Upon p38 inhibition, the expression of both cytokines was reduced in vector- and DUSP10-overexpression cells and the level of their expression become comparable between vector- and DUSP10-overexpression cells. These cytokines have been shown to play important roles in NSCLC progress (15, 16, 17, 18). Therefore, the tumor promoting function of DUSP10 in NCSLC is associated with increased p38 activation and the expression of p38-regulated tumor-promoting cytokines.

Figure 6
DUSP10 regulates tumor-promoting cytokines in NSCLC cells likely through p38. (A). Vector- and DUSP10-overexpression HCC827 cells were stimulated with 100 ng/ml EGF for 24 h before harvest to extract total RNA to prepare cDNA. The expression of the indicated cytokines was determined by qPCR. (B). Protein expression of TGFβ and IL-6 were assessed by ELISA. (C). Vector- and DUSP10-overexpression HCC827 cells were stimulated with 100 ng/ml EGF with or without SB20380 for 24 h before analysis of TGFβ and IL-6 by qPCR. Results representative of three independent experiments.
*p<0.05, **p<0.01.

DISCUSSION

Lung cancer, particularly NSCLC which contributes more than 80% to total lung cancer cases, is a leading cause of death world-wide. Over half of the patients diagnosed with lung cancer die within one year of diagnosis and the 5-year survival remains less than 20% (1, 19). Continuous expanding our knowledge of the genetics and molecular mechanisms underlying the pathogenesis of lung cancer will help to development new effective therapies to improve the treatment and management of this disease, hence improving patient survival. Here we showed that DUSP10 is oncogenic in NSCLC. Using gene overexpression approach, we first showed that this molecule suppressed growth of both HCC827 and H460, two NSCLC cell lines in both in vitro and in vivo. Interestingly, the activation of ERK was found to be reduced, but p38 activation was increased upon DUSP10 overexpression (Fig. 3A). Indeed, both c-Myc and COX2, two ERK-targeted molecules that promote cell growth were found to be reduced by DUSP10 overexpression. Meanwhile, p38 activation was found to be increased upon DUSP10 overexpression (Fig. 3A). Importantly, NSCLC patients with low DUSP10 expression showed better survival compared to those with high DUSP10 (Fig. 5). Together, these findings demonstrated that DUSP10 promotes NSCLC development and can be served as a marker for patient prognosis.

Previously, DUSP10 has been shown to suppress colorectal cancer development through ERK (11). This DUSP mainly dephosphorylates ERK in epithelial cells in the gut to suppress cell growth and wound healing, thereby inhibiting tumor development. It has also been shown to target JNK in immune cells to inhibit the expression of proinflammatory cytokines and regulate various immune effector cell function (14). In this study, it was found that the phosphorylation of both ERK and JNK was reduced upon DUSP10 overexpression, but p38 phosphorylation was increased (Fig. 3A). The increase of p38 phosphorylation could be due to crosstalk between ERK/JNK and p38. It is also possible that overexpression of DUSP10 led to reduced expression and function of p38-specific phosphatases, hence increasing p38 phosphorylation. Nevertheless, cell- and tissue-specific substrate preference is a common phenomenon for DUSPs (10).

Although DUSP10 inhibited ERK activation and the expression of ERK-target molecules including c-Myc and COX2, it promoted cell growth and migration in NSCLC, associated with p38 activation. The roles of p38 in cancer remains controversial. There are studies showing tumor suppressive function of p38. For instance, mice with liver-specific deletion of p38α had enhanced hepatocyte proliferation and tumorigenesis which was correlated with sustained activation of JNK-c-Jun pathway in response to chemical-induced liver cancer development (20). In addition, p38α was showed to be able to detect oxidative stress production early in the process of oncogenic H-Ras-induced transformation, thereby triggering apoptosis to negatively regulate malignant transformation (21). On the other hand, there are studies showing tumor promoting function of p38. For example, it has been shown that breast cancer epithelial cells rely on p38α to maintain their genome integrity and support tumor growth (22). As such, inhibition of p38α resulted in increased chromosome instability, increased DNA damage and impaired DNA replication, promoting cancer cell death and tumor regression. In NSCLC, increased activation of p38 has been detected in tumor tissues compared to that in normal tissues (23). In this study, using p38-specific inhibitor SB203580, we should that inhibition of p38 resulted in reduced proliferation and migration of both HCC827 and H460 cells (Fig. 3B and C, Supplementary Fig. 3). Importantly, the differences in both cell proliferation and migration between vector- and DUSP10-overexpression cells were abrogated upon p38 inhibition, demonstrating that DUSP10 promoting NSCLC through p38.

In addition, we found that DUSP10 promotes the expression of IL-6 and TGFβ in NSCLC cells through p38 (Fig. 6). It has been shown that IL-6 is a predicator of severe lung cancer and increased IL-6 levels are correlated with worse patient survival (15). It also promotes resistance to immunotherapy in NSCLC patients (16). For TGFβ, a multifunctional cytokine, its tumor-promoting function has been well documented. For instance, it has been shown that TGFβ1 promoted NSCLC cell proliferation, anchorage-independent cell growth and migration (24). In addition, it induces NSCLC epithelial-to-mesenchymal-transition (EMT) and metastasis (17). Furthermore, the invasion and metastasis of NSCLC cells was positively corelated with serum levels of TGFβ1 (25), and the expression of TGFβ1 can significantly predict the worse prognosis of NSCLC patients (18). Overall, it can induce tumor cell migration and stimulates EMT, promotes tumorigenesis indirectly by action on the tumor microenvironment, as well as contributes to tumor resistance to treatment. Moreover, TGFβ1 activates JNK and p38 through the non-canonical pathway and the activation of p38 is an obligatory requirement for TGFβ-induced EMT (26). It is possible that in NSCLC cells, p38 is not targeted by DUSP10, therefore, increased TGFβ results in increased p38 activation which in turn contributes to the increased growth and migration. It is also possible that the increased p38 activation contributes to increased TGFβ expression. Further investigation is need to clarify how DUSP10 regulates p38 activation which in turn contributes to the tumor-promoting function of DUSP10.

In summary, findings from this study demonstrated that DUSP10 promotes NSCLC cell growth and migration which is correlated with increased p38 activation and increased tumor-promoting cytokine expression such as the expression of TGFβ. High DUSP10 is associated with poorer prognosis in NSCLC patients. DUSP10 therefore could severe as a biomarker for NSCLC prognosis and could be targeted to develop therapeutic intervention for lung cancer treatment. Future study may focus on the detailed mechanism underlying the oncogenic function of this molecule in NSCLC.

SUPPLEMENTARY MATERIALS

Supplementary Figure 1

Establishment of DUSP10-overexpression H460 stable cell clones. (A) NSCLC cell line H460 was transfected with pcDNA3 vector or pcDNA3 containing the full-length of human DUSP10 cDNA followed by drug selection for stably transfected clones. One clone each from pcDNA3- and pcDNA3-DUSP10 transfected cells were selected for examination of DUSP10 expression by quantitative real-time PCR (qPCR). (B) Cell growth of vector- and DUSP10-overexpression over a period of 3 days with or without hEGF was analyzed by crystal violet staining.

Click here to view.(754K, ppt)

Supplementary Figure 2

Overexpression of DUSP10 in NSCLC cells resulted in increased spheroid formation and wound healing. (A) Representative micrographs of multicellular spheroid aggregates, derived from vector- and DUSP10-overexpression HCC827 cells. Bar chart shows mean of spheroid size (n=30). (B) Representative micrographs showing wound closure in vector- and DUSP10-overexpression cells. Bar chart shows mean of percentage of wound closure (n=12). Results representative of three independent experiments.

Click here to view.(2M, ppt)

Supplementary Figure 3

Quantification of MAPK activation, cMyc and COX2 protein expression in vector- and DUSP10-overexpression HCC827 cells with or without EGF stimulation. (A) Quantification of phospho-ERK, pP38 and pJNK against their respective total protein expression. Quantification of cMyc and COX2 protein expression was based on expression of actin. The quantification of the blots was performed using imageJ software. (B) The results presented were the average of three independent experiments.

Click here to view.(1M, ppt)

Supplementary Figure 4

Important function of p38 in regulation of NSCL growth and migration. (A) Representative micrographs of multicellular spheroid aggregates, derived from vector- and DUSP10-overexpression HCC827 cells. Bar chart shows mean of spheroid size. (B) Representative micrographs showing wound closure in vector- and DUSP10-overexpression cells. Bar chart shows mean of percentage of wound closure (n=12). Results representative of three independent experiments.

Click here to view.(2M, ppt)

Supplementary Figure 5

Decreased ERK activation in DUSP10-overexpression xenografts. IHC analysis ERK activation in vector- and DUSP10-overexpression H640 xenografts. Representative images of pERK staining were shown.

Click here to view.(2M, ppt)

Notes

Conflicts of Interest:The authors declare no potential conflicts of interest.

Author Contributions:

  • Conceptualization: Xu X, Zhang Y, Png CW, Wei X.

  • Data curation: Wei X, Png CW, Weerasooriya M, Li H, Zhu C, Chen G, Xu C.

  • Formal analysis: Xu X, Zhang Y, Png CW, Wei X.

  • Funding acquisition: Xu X, Zhang Y, Png CW, Chen G.

  • Supervision: Xu X, Zhang Y.

  • Writing - original draft: Xu X, Zhang Y, Png CW, Wei X.

  • Writing - review & editing: Xu X, Zhang Y .

Abbreviations

DUSP dual-specificity phosphatase
EGF epidermal growth factor
EGFR epidermal growth factor receptor
EMT epithelial-to-mesenchymal-transition
MAP mitogen-activated protein
MKP MAPK phosphatase
NSCLC non-small cell lung cancer
SCLC small cell lung cancer
qPCR Quantitative real-time PCR

ACKNOWLEDGEMENTS

This work was supported by grants from the TCM Science and Technology Program of Zhejiang Province, China (2020ZA054/2020 to G.C.), and the Science and Technology Project of Zunyi, Guizhou province, China (Zunyi City Kehe Support NS (2020) No. 18/2020 to X.X.), the Singapore National Medical Research Council (NMRC/OFIRG/0059/2017 to Y. Z.) and the National University Health System (NUHSRO/2021/110/T1/Seed-Sep/03 to C.W. P.).

References

    1. Bade BC, Dela Cruz CS. Lung cancer 2020: epidemiology, etiology, and prevention. Clin Chest Med 2020;41:1–24.
    1. Nooreldeen R, Bach H. Current and future development in lung cancer diagnosis. Int J Mol Sci 2021;22:8661.
    1. Kozaki K, Miyaishi O, Tsukamoto T, Tatematsu Y, Hida T, Takahashi T, Takahashi T. Establishment and characterization of a human lung cancer cell line NCI-H460-LNM35 with consistent lymphogenous metastasis via both subcutaneous and orthotopic propagation. Cancer Res 2000;60:2535–2540.
    1. Takahashi M, Okamoto Y, Kato Y, Shirahama H, Tsukahara S, Sugimoto Y, Tomida A. Activating mutations in EGFR and PI3K promote ATF4 induction for NSCLC cell survival during amino acid deprivation. Heliyon (Lond) 2023;9:e14799
    1. da Cunha Santos G, Shepherd FA, Tsao MS. EGFR mutations and lung cancer. Annu Rev Pathol 2011;6:49–69.
    1. Prior IA, Lewis PD, Mattos C. A comprehensive survey of Ras mutations in cancer. Cancer Res 2012;72:2457–2467.
    1. Dankner M, Rose AA, Rajkumar S, Siegel PM, Watson IR. Classifying BRAF alterations in cancer: new rational therapeutic strategies for actionable mutations. Oncogene 2018;37:3183–3199.
    1. Lavoie H, Gagnon J, Therrien M. ERK signalling: a master regulator of cell behaviour, life and fate. Nat Rev Mol Cell Biol 2020;21:607–632.
    1. Caunt CJ, Keyse SM. Dual-specificity MAP kinase phosphatases (MKPs): shaping the outcome of MAP kinase signalling. FEBS J 2013;280:489–504.
    1. Low HB, Zhang Y. Regulatory roles of MAPK phosphatases in cancer. Immune Netw 2016;16:85–98.
    1. Png CW, Weerasooriya M, Guo J, James SJ, Poh HM, Osato M, Flavell RA, Dong C, Yang H, Zhang Y. DUSP10 regulates intestinal epithelial cell growth and colorectal tumorigenesis. Oncogene 2016;35:206–217.
    1. Low HB, Wong ZL, Wu B, Kong LR, Png CW, Cho YL, Li CW, Xiao F, Xin X, Yang H, et al. DUSP16 promotes cancer chemoresistance through regulation of mitochondria-mediated cell death. Nat Commun 2021;12:2284.
    1. Fukuyama T, Ichiki Y, Yamada S, Shigematsu Y, Baba T, Nagata Y, Mizukami M, Sugaya M, Takenoyama M, Hanagiri T, et al. Cytokine production of lung cancer cell lines: correlation between their production and the inflammatory/immunological responses both in vivo and in vitro. Cancer Sci 2007;98:1048–1054.
    1. Zhang Y, Blattman JN, Kennedy NJ, Duong J, Nguyen T, Wang Y, Davis RJ, Greenberg PD, Flavell RA, Dong C. Regulation of innate and adaptive immune responses by MAP kinase phosphatase 5. Nature 2004;430:793–797.
    1. An J, Gu Q, Cao L, Yang H, Deng P, Hu C, Li M. Serum IL-6 as a vital predictor of severe lung cancer. Ann Palliat Med 2021;10:202–209.
    1. Liu C, Yang L, Xu H, Zheng S, Wang Z, Wang S, Yang Y, Zhang S, Feng X, Sun N, et al. Systematic analysis of IL-6 as a predictive biomarker and desensitizer of immunotherapy responses in patients with non-small cell lung cancer. BMC Med 2022;20:187.
    1. Tong X, Wang S, Lei Z, Li C, Zhang C, Su Z, Liu X, Zhao J, Zhang HT. MYOCD and SMAD3/SMAD4 form a positive feedback loop and drive TGF-β-induced epithelial-mesenchymal transition in non-small cell lung cancer. Oncogene 2020;39:2890–2904.
    1. Li J, Shen C, Wang X, Lai Y, Zhou K, Li P, Liu L, Che G. Prognostic value of TGF-β in lung cancer: systematic review and meta-analysis. BMC Cancer 2019;19:691.
    1. Zappa C, Mousa SA. Non-small cell lung cancer: current treatment and future advances. Transl Lung Cancer Res 2016;5:288–300.
    1. Komnenovic V, Scheuch H, Beug H, Wagner EF. p38α suppresses normal and cancer cell proliferation by antagonizing the JNK–c-Jun pathway. Nat Genet 2007;39:741–749.
    1. Dolado I, Swat A, Ajenjo N, De Vita G, Cuadrado A, Nebreda AR. p38α MAP kinase as a sensor of reactive oxygen species in tumorigenesis. Cancer Cell 2007;11:191–205.
    1. Cánovas B, Igea A, Sartori AA, Gomis RR, Paull TT, Isoda M, Pérez-Montoyo H, Serra V, González-Suárez E, Stracker TH, et al. Targeting p38α increases DNA damage, chromosome instability, and the anti-tumoral response to taxanes in breast cancer cells. Cancer Cell 2018;33:1094–1110.e8.
    1. Greenberg AK, Basu S, Hu J, Yie TA, Tchou-Wong KM, Rom WN, Lee TC. Selective p38 activation in human non-small cell lung cancer. Am J Respir Cell Mol Biol 2002;26:558–564.
    1. Liu H, Zhang M, Xu S, Zhang J, Zou J, Yang C, Zhang Y, Gong C, Kai Y, Li Y. HOXC8 promotes proliferation and migration through transcriptional up-regulation of TGFβ1 in non-small cell lung cancer. Oncogenesis 2018;7:1.
    1. Liu SG, Yuan SH, Wu HY, Liu J, Huang CS. The clinical research of serum VEGF, TGF-β1, and endostatin in non-small cell lung cancer. Cell Biochem Biophys 2015;72:165–169.
    1. Zhang YE. Non-Smad pathways in TGF-beta signaling. Cell Res 2009;19:128–139.

Metrics
Share
Figures

1 / 6

PERMALINK