Open Access

Amine oxidase, copper containing 3 exerts anti‑mesenchymal transformation and enhances CD4+ T‑cell recruitment to prolong survival in lung cancer

  • Authors:
    • Chao-Yuan Chang
    • Kuan-Li Wu
    • Yung-Yun Chang
    • Pei-Hsun Tsai
    • Jen-Yu Hung
    • Wei-An Chang
    • Shu-Fang Jian
    • Yung-Chi Huang
    • Inn-Wen Chong
    • Ying-Ming Tsai
    • Ya-Ling Hsu
  • View Affiliations

  • Published online on: July 23, 2021     https://doi.org/10.3892/or.2021.8154
  • Article Number: 203
  • Copyright: © Chang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Lung cancer remains notorious for its poor prognosis. Despite the advent of tyrosine kinase inhibitors and immune checkpoint inhibitors, the probability of curing the disease in lung cancer patients remains low. Novel mechanisms and treatment strategies are needed to provide hope to patients. Advanced strategies of next generation sequencing (NGS) and bioinformatics were used to analyze normal and lung cancer tissues from lung cancer patients. Amine oxidases have been linked to leukocyte migration and tumorigenesis. However, the roles of amine oxidases in lung cancer are not well‑understood. Our results indicated that amine oxidase, copper containing 3 (AOC3) was significantly decreased in the tumor tissue compared with the normal tissue, at both the mRNA and protein level, in the included lung cancer patients and public databases. Lower expression of AOC3 conferred a poorer survival probability across the different cohorts. Epigenetic silencing of AOC3 via miR‑3691‑5p caused tumor promotion and progression by increasing migration and epithelial‑mesenchymal transition (EMT). Furthermore, knockdown of AOC3 caused less CD4+ T‑cell attachment onto lung cancer cells and reduced transendothelial migration in vitro, as well as reducing CD4+ T‑cell trafficking to the lung in vivo. In conclusion, the present study revealed that downregulation of AOC3 mediated lung cancer promotion and progression, as well as decrease of immune cell recruitment. This novel finding could expand our understanding of the dysregulation of the tumor immune microenvironment and could help to develop a novel strategy for the treatment of lung cancer.

Introduction

Lung cancer is the leading cause of cancer-related deaths worldwide with a high annual incidence and a 5-year survival rate of <20% regardless of its stage at diagnosis (1,2). However, when it metastasizes, the 5-year survival rate is less than 5% (3). Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, representing ~80% of all cases (4,5). The two most prevalent NSCLC subtypes are lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC), which constitute 35 and 25% of all cases, respectively (5). There have been notable advances in our understanding of lung cancer and its underlying mechanisms of action, which have been clearly elucidated and exemplified (6). There have also been marked improvements recently in the treatments available for lung cancer. The current treatment protocol consists of a platinum doublet, tyrosine kinase inhibitors for epithelial growth factor receptor (EGFR), echinoderm microtubule-associated protein-like 4-anaplastic lymphoma kinase (EML4-ALK) fusion protein and immune checkpoint inhibitors (3). However, the prognosis for lung cancer remains poor and a more detailed mechanism and treatment options are needed. In addition to the malignant cells, the surrounding microenvironment is also critical for tumorigenesis (7,8). Amine oxidases refer to a class of enzymes that catalyze the deamination of amine groups to produce aldehydes, ammonia and hydrogen peroxide (9). There are a variety of amine oxidases consisting of four classes of monoamine oxidases (MAOs), including MAO-A and MAO-B, polyamine oxidases, lysyl oxidases, and copper-containing amine oxidases (CAOs) (10). CAOs have been revealed to participate in the regulation of a variety of pathological and physiological processes, such as cell proliferation, differentiation, glucose uptake and immune regulation (11). Changes in CAO activity are correlated with a variety of human diseases, including diabetes mellitus, Alzheimer's disease, and inflammatory disorders (12,13). The four complete genes for CAOs are amine oxidase, copper containing (AOC)1-4. AOC1 consists of a homodimeric glycoprotein with an apparent molecular mass of 186 kDa; it is secreted as a diamine oxidase to generate hydrogen peroxide (14,15). AOC1 is strongly expressed in the kidneys, placenta, intestine and lungs (14). Little is known about the molecular mechanisms regulating AOC1 gene expression. The AOC2 gene encodes retina-specific amine oxidase (16), which was originally identified in ganglion cells. Its functions remain unclear but it may play a role in hereditary retinal diseases (16). The AOC4 gene encodes a soluble plasma amine oxidase in cows as bovine AOC4 (17) but not in humans, mice or rats. The AOC3 gene encodes vascular adhesion protein-1, which is primarily expressed on the endothelial cell surface but also in smooth muscle cells and adipocytes (18). In addition to its amine oxidase activity, AOC3 functions as a non-classical inflammation-inducible endothelial molecule which is linked to leukocyte-subtype specific rolling under physiological shear (18). It has been revealed that the enzymatic activity of AOC3 is functionally important, and leukocyte recruitment is impaired if its activity is abolished (10).

The surrounding microenvironment of cancer contributes to its promotion and progression (19). Due to its unique tumor microenvironment (TME), cancer promotes and strengthens its own progression as a result of its interactions. The cells inside the TME include cancer-associated fibroblasts, endothelial cells and immune cells (20), which form the tumor immune microenvironment (TIME). The functions and densities of different tumor-infiltrating immune cells in the TIME are closely associated with prognosis and prediction of the treatment response (7). Therefore, there is an urgent need for improved understanding of immune dysfunction inside the TIME and the mechanisms by which the tumor modifies its environment to remove the functional immunity of the body. The present study aimed to verify the role of AOC3 in lung cancer progression and the relevant anticancer immunity.

Materials and methods

Cell lines and reagents

Murine Lewis lung carcinoma (LLC) cell line and human umbilical vein endothelial cells (HUVECs) were purchased from American Type Culture Collection (ATCC). Human lung cancer CL1-5 cells were kindly provided by Dr Pan-Chyr Yang of National Taiwan University (Taipei City, Taiwan) and were cultured in RPMI-1640 medium (Lonza Group, Ltd.) supplemented with 10% fetal bovine serum (FBS), 100 U/ml penicillin and 100 µg/ml streptomycin (Thermo Fisher Scientific, Inc.; Waltham, MA, USA) at 37°C. Recombinant human and mouse AOC3 were obtained from R&D Systems, Inc. Knockdown of AOC3 in CL1-5 cells was performed using either pLKO_005 plasmid as a control or AOC3-shRNA plasmid (14 µg shRNA plasmid for 5×105 cells in a 6-well plate) obtained from the National RNAi Core Facility (Academia Sinica, Taipei, Taiwan). The plasmid was transfected into cells using Lipofectamine 2000™ Thermo Fisher Scientific, Inc.) for 2 days, and the stable clone of AOC3-knockdown cells were established by puromycin selection (5 µg/ml). All cells were authenticated by short tandem repeat (Promega Corporation) and examined for mycoplasma contamination using a MycoAlert™ mycoplasma detection kit (Lonza Group, Ltd.) according to the manufacturer's protocol every three months.

Next generation sequencing (NGS) and bioinformatics analysis

All of the participants selected from January 2018 to December 2019, provided written informed consent prior to inclusion in the present study. The patients who agreed and received surgical intervention were enrolled in this study. The adjacent non-tumor lung and tumor tissues of ten patients (7 from LUAD and 3 from LUSC) were obtained from the Division of Thoracic Surgery and Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital (Kaohsiung, Taiwan). The protocol of the present study was reviewed and approved (approval no. KMUH-IRB-20130054 and KMUH-IRB-G(II)-20180021) by the Institutional Review Board of Kaohsiung Medical University Hospital. The deep RNA-seq was carried out at a biotechnology company (Welgene, Inc.) using the Solexa platform. RNA and small RNA library construction was carried out using a sample preparation kit (Illumina, Inc.) following the protocol of the TruSeq RNA or Small RNA Sample Preparation Guide.

The expression of AOCs in lung cancer and normal specimens (cancer vs. normal) were extracted from the Oncomine® database (http://www.oncomine.org; Compendia Biosciences) (21) and The Cancer Genome Atlas (TCGA) cohort of UALCAN (http://ualcan.path.uab.edu/analysis.html) (22). The 16 cohorts from Oncomine® database included Su et al (23), Okayama et al (24), Landi et al (25), Beer et al (26), Stearman et al (27), Selamat et al (28), Garber et al (adenocarcinoma and squamous) (29), Hou et al (adenocarcinoma, squamous and large cell) (30), Wachi et al (squamous) (31), Bhattacharjee et al (adenocarcinoma, squamous, carcinoid and small cell) (32). Criteria in the analysis were fold change >2 and P-value <10−4, which was calculated using the Oncomine® database through two-sided Student's t-test. The data of AOC mRNA, copy number and overall survival in TCGA database, and AOC protein were analyzed by UALCAN website (http://ualcan.path.uab.edu/analysis-prot.html) (22). The immunohistochemical staining for AOC3 in lung cancer and normal lung tissue samples were acquired from The Human Protein Atlas (33). The association between gene expression and clinical outcome of lung cancer patients was evaluated by publicly available data using Kaplan-Meier (K-M) plotter and log-rank testing (https://kmplot.com/analysis/) (34), UALCAN and PROGgeneV2 (35). The post-transcriptional regulation was predicted using miRWalk (version 3.0) (36), miRanda (37) and miRDB (38) with restriction of >95% confidence.

Measurement of AOC3

All of the participants provided written informed consent prior to inclusion in the present study. The sera of 40 healthy donors and 40 lung cancer patients (healthy donors: Age range, 40–80 years old; M/F 31%/69%; lung cancer donors: Age range 30–90 years old; M/F 46/54%) were collected from the Division of Thoracic Surgery and Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital (Kaohsiung, Taiwan) from January 2018 to December 2019. The patients who agreed with written informed consent were enrolled in this study before starting definite treatment. These samples were assessed using Quantikine Human VAP-1 Immunoassay (R&D Systems, Inc.). The protocol of the present study was reviewed and approved (approval no. KMUH-IRB-20130054) by the Institutional Review Board of Kaohsiung Medical University Hospital.

Reverse transcription-quantitative (RT-q) PCR

Total RNAs were extracted from CL 1–5 lung cancer cells with TRIzol reagent (Thermo Fisher Scientific, Inc.) and reverse transcribed into cDNA using an oligo (dT) primer and reverse transcriptase (PrimeScript RT Reagent Kit; Takara Bio, Inc.) following the manufacturer's protocols. The reaction conditions were as follows: Priming for 5 min at 25°C, reverse transcription for 20 min at 46°C, and final inactivation of reverse transcriptase for 1 min at 95°C (40). The expression levels of specific genes were determined by a StepOne-Plus PCR instrument (Applied Biosystems; Thermo Fisher Scientific, Inc.), using real-time analysis with SYBR-Green (Thermo Fisher Scientific, Inc.). The following primers were used: AOC1 forward, 5′-AOC1_H_F2GTGATGGAGGCCAAGATGCA-3′ and reverse, 5′-AOC1_H_R2TCTGCAGTGTCTGGAAGCTG-3′; AOC2 forward, 5′-AOC2_H_F2GCCTTCCACTTCAAGCTGGA-3′ and reverse, 5′-AOC2_H_R2GCTCTCAGGTCCTCCTTTCC-3′; AOC3 forward, 5′-AOC3_H_F2GTGGGGCCATAGAAATACGA-3′ and reverse, 5′-AOC3_H_R2CAGACCCAGTTCTCCAGTCC-3′; and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) forward, 5′-TTCACCACCATGGAGAAGGC-3′ and reverse, 5′-GGCATGGACTGTGGTCATGA-3′. The RT-qPCR was performed at 95°C for 20 sec, followed by 40 cycles at 95°C for 5 sec and 60°C for 35 sec (39). Relative expression levels of the cellular mRNA were normalized to GAPDH. The relative standard method (2−ΔΔCq) was used to calculate relative RNA expression (40).

Cell proliferation and 5-bromo-2-deoxyuridine (BrdU) incorporation

Cells (3×103 cells/well) were seeded in a 96 well plate, and then cultured for 48 or 72 h. Cell proliferation was determined by cell proliferation reagent WST-1 proliferation assay kit (Takara Bio, Inc.) after 2-h incubation and measured at a 450-nm wavelength according to the manufacturer's instructions. Cells were labelled with BrdU (10 µM) at day 2 after seeding followed by fixation. In the BrdU incorporation assay, cells were fixed at room temperature for 30 min with 200 µl/well of the Fixing Solution (included in the kit undermentioned) and incubated at room temperature for 30 min. Integrated BrdU was assessed by ELISA-based method according to the manufacturer's protocol (BrdU Cell Proliferation Assay Kit; cat. no. 2750; EMD Millipore).

Wound healing analysis

CL 1–5 cells were seeded into a 12 well-pate at 90% confluence and cultured in 1% of FBS-containing medium for exogenous AOC3 (control, 10, 20 and 50 ng/ml) and 10% of FBS-containing medium (since cell proliferation was not affected by AOC3 knockdown, in order to mimic the physiologic conditions, 10% of FBS-containing medium was used and cells were not serum starved) for AOC3-shRNA knockdown at 37°C as previously described by Shao et al (41), and the cell migration was evaluated by measuring the migration of cells into the acellular region formed by a sterile yellow tip. The wound closure was observed after 8 h. The wound healing assay was closely observed via a Nikon inverted microscope (Nikon Corporation).

CD4+ T-cell isolation

Peripheral blood mononuclear cells (PBMCs) of healthy donors (eight healthy donors: Age range, 35–45 years old; male only) were obtained from the Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital (Kaohsiung, Taiwan) from January 2020 to December 2020. The protocol of the present study was reviewed and approved (approval no. KMUH-IRB-20130054) by the Institutional Review Board of Kaohsiung Medical University Hospital and the donors provided written informed consent. PBMCs were isolated using 7.5 ml Ficoll-Hypaque gradient reagent (EMD Millipore) in 1 ml blood mixing with 5 ml PBS, and human CD4+ T cells were isolated form PBMC using CD4+ T-cell Isolation Kit (MACS MicroBeads; Miltenyi Biotec GmbH) according to the manufacturer's instructions.

Cell adhesion and transendothelial migration

For transendothelial migration, HUVECs (5×104) were seeded onto inserts with polyester membranes of 3-µm pore size (EMD Millipore) and cultured at 37°C for 48 h to form a 100% confluent monolayer. CL1-5 (1×105) or AOC3-knockdown CL1-5 (1×105) cells were seeded in the bottom of a 24-well plate containing RPMI-1640 culture medium. PKH26-labeled (EMD Millipore) CD4+ T-cells were seeded onto HUVEC-coated inserts, which were placed in the wells of the 24-well plate and then incubated for 24 h at 37°C. The migratory cells were visualized in four randomly selected fields using a Nikon fluorescence microscope (Nikon Corporation).

Western blot analysis

Total proteins from primary tissues and cell lines were extracted using RIPA lysis buffer (Thermo Fisher Scientific, Inc.). An equal amount of total protein (2 µg) was quantitated by bicinchoninic acid (BCA) analysis and separated by SDS-PAGE (6-8%). After transferring, the PVDF membranes containing bound proteins were blocked at room temperature for 2 h using 5% milk containing TBST buffer (0.02% Tween-20) and then incubated overnight at 4°C with primary antibodies against a specific target protein. After incubation with HRP-coupled secondary antibodies (1:5,000; anti-mouse, 7076; anti-rabbit, 7074; Cell Signaling Technology) at room temperature for 1 h, the protein bands were visualized using ECL (EMD Millipore) and detected using a FluorChem HD2 System (ProteinSimple). The following primary antibodies were used: E-cadherin (1:500; cat. no. 610182) N-cadherin (1:500; cat. no. 610921) and vimentin (1:500; cat. no. 550513; all from BD Biosciences), Slug (1:500; product no. 9585S; Cell Signaling Technology, Inc.), and GAPDH (1:5,000; cat. no. MAB374; EMD Millipore). The quantitation of the results of the western blotting was performed using AlphaImager software (Version 6.0.0; ProteinSimple).

miRNA mimics transfection

CL1-5 cells were transfected with microRNA (miR)-3691-5p (AGUGGAUGAUGGAGACUCGGUAC; at a concentration of 100 nM; GE Healthcare Dharmacon, Inc.) or scrambled control (negative control 1; UCACAACCUCCUAGAAAGAGUAGA; at a concentration of 100 nM; GE Healthcare Dharmacon, Inc.) by using Dharmafect reagent 4 (GE Healthcare Dharmacon, Inc.) according to the manufacturer's instructions. The transfection efficacy was monitored by transfecting siGLO fluorescent oligonucleotides (catalog ID: D-001630-02-05; GE Healthcare Dharmacon, Inc.) concurrently after 24 h of transfection at 37°C according to the manufacturer's protocol. The expression of AOC1-3, cell migration and CD4+ T-cell migration as well as adhesion were assayed after a 48-h transfection.

Mouse studies

All mice procedures were approved by and conducted in accordance with the Institutional Animal Care and Use Committee at Kaohsiung Medical University (IACUC Approval No. 107104; Kaohsiung, Taiwan). C57BL/6 mice (12 males in total; weight, 18±2 g; 5 weeks old) were obtained from the Taiwan National Laboratory Animal Center (Taipei City, Taiwan). The mice were housed in a specific pathogen-free environment with the room temperature being maintained at ~20°C, the humidity at ~45% and a 12-h light/dark cycle. Each mouse had free access to food and water. The mice were subjected to implantation of LLC cells (1×106 cells) via tail vein and tumor growth in the lungs was allowed for 7 days. Mice were treated with PBS or recombinant mouse (rm) AOC3 twice (10 µg/mouse; on days 7 and 14) by intra-tracheal route. At the end of the experiment, the mice were euthanized by CO2 asphyxiation during which the CO2 gas flow rate displaced 10 to 30% of the cage volume per minute. CD4+ T cells of the lungs of mice were isolated by mouse CD4+ T cell isolation kit (MACS MicroBeads; Miltenyi Biotec GmbH) according to the manufacturer's instructions and counted after 21 days of LLC implantation. Lung tissue was collected and minced and incubated in RPMI-1640 medium with collagenase type 1 (400 U/ml; Worthington Biochemical Corporation) at 37°C for 1 h. The digested tissues were filtered through a 70-µm cell strainer and washed with RPMI-1640 medium. CD4+ T cells of the lung filtered solution were isolated by mouse CD4 isolation kit and counted after 21 days of LLC implantation.

Statistical analyses

Each experiment was repeated at least three times. Data are expressed as the mean ± standard deviation (SD) using GraphPad Prism version 7.04 (GraphPad Software, Inc.). Two treatment groups were compared by unpaired Student's t-test. Multiple group comparisons were performed by two-way analysis of variance with Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

AOC3 mRNA expression is reduced in lung cancer

The controversial roles of AOCs have been reported in various cancer types (4144), therefore their effect in lung cancer was investigated. Tumor tissue and adjacent normal tissue specimens from 10 lung cancer patients (7 LUAD and 3 LUSC) were analyzed via NGS (Table I). The expression of AOC2 (7 out of 10) and AOC3 (8 out of 10) was lower in most of the tumor tissue of patients compared with their normal tissue, however lower AOC1 in tumor tissue was observed in only 2 out of 10 patients with lung cancer (Fig. 1A-C). Using Oncomine® datasets, it revealed that AOC3, but not AOC1 or AOC2, was expressed at lower levels in tumor tissue compared with normal tissue in 16 lung cancer cohorts (Fig. 1D). Further analysis of these 16 cohorts revealed that the expression of AOC3 mRNA was lower in the tumor tissue for both the LUAD and non-adenocarcinoma patients (Fig. 1E). The expression of AOC2 and AOC3 in LUAD and LUSC was also retrieved from TCGA cohorts. Overall AOC2 (Fig. 1F) and AOC3 (Fig. 1G) expression was significantly lower in the tumor tissue compared with the adjacent normal tissue, even though this trend was not observed for all stages. Moreover, the expression of AOC3 was significantly lower in the N1 group (with lymph node metastasis) compared with the N0 group (without lymph node metastasis), implying that AOC3 may contribute to cancer metastasis (Fig. 1F and G).

Table I.

Characteristics of patients.

Table I.

Characteristics of patients.

GroupNumberSexAgePathological diagnosisStageTNM
I01M70Adenocarcinoma grade 32B300
02M66Adenocarcinoma grade 34B2a01c
03F51Adenocarcinoma grade 31B2a00
04M53Adenocarcinoma grade 33A320
05F60Adenocarcinoma grade 21A1b00
06M67Adenocarcinoma grade 11A1a00
07M60Adenocarcinoma grade 34A411a
II08M84Squamous cell carcinoma grade 22B300
09F65Squamous cell carcinoma grade 23A40b
10M69Squamous cell carcinoma grade 22B2b10
AOC3 protein expression is inversely associated with lung cancer grade

AOC protein expression was extracted from the National Cancer Institute Clinical Proteomic Tumor Analysis Consortium (CPTAC). AOC1 protein expression did not vary between the tumor and normal tissue for the different types of LUAD, grades or stages (Fig. 2A-D). However, AOC3 protein expression was lower in the tumor tissue compared with the normal tissue in every cell type (Fig. 2E and F). Moreover, AOC3 expression was negatively associated with the grades and stages (early and late) of LUAD (Fig. 2G and H). The soluble form of AOC3 has been detected in other cancer types such as colorectal cancer (41). To evaluate the role of soluble AOC3 in lung cancer, serum from lung cancer patients was collected. Soluble AOC3 in the sera from lung cancer patients was lower than in healthy donors (Fig. 2I). In addition, the public datasets for the expression of AOC3 in lung cancer were utilized. Compared with normal tissue, both LUAD and LUSC expressed lower levels of AOC3 from The Human Protein Atlas (Fig. 2J). The combination of these results and the mRNA expression results indicated that AOC3 could be a promising tumor suppressor in lung cancer.

Lower expression of AOC3 confers a poorer survival time

Since the tumor tissue of lung cancers expressed lower levels of AOC3, its prognostic vaule in patients was evaluated by survival analysis. There are several public websites that evaluate survival analysis, including the K-M plotter, UALCAN and PROGgeneV2. According to the K-M plotter, low AOC1 expression did not confer a poorer survival time, and it actually conferred a longer survival time in LUAD but not in LUSC patients (Fig. 3A; upper panel). AOC2 revealed the same pattern in both types of lung cancer (Fig. 3A; middle panel). However, analysis of AOC3 expression revealed that the lower the AOC3 expression was, the shorter the survival time was in LUAD patients but not in LUSC patients (Fig. 3A, lower panel). Moreover, the clinical implication of AOC3 expression as detemined by survival rates was validated by cohorts extracted from the UALCAN and PROGgeneV2 websites; low expression of AOC3 conferred a shorter surivival time but this was not observed for AOC1 (ABP1) or AOC2 (Fig. 3B and C). These results confirmed that AOC3 was strongly associated with clinical outcomes in lung cancer patients (Fig. 3A and B, lower panel; Fig. 3C).

Mechanism regulating the expression of AOC3

Since AOC3 was revealed to be critical in the prognosis of lung cancer patients, the dysregulation of AOC3 required investigation. Genetic modifications, as DNA copy number variation, DNA methylation, and post-transcriptional regulation by miRNAs were utilized. As determined by the TCGA cohort, variation in the DNA copy number of AOC3 was not correlated with the expression of AOC3 mRNA (R=−0.121; Fig. 4A). In addition, DNA methylation of AOC3 was not significantly associated with the expression of AOC3 mRNA (R=−0.034; Fig. 4B). Concerning post-transcriptional regulation, the miRNAs that epigenetically regulate AOC3 mRNA were predicted using miRWalk version 3.0 with miRanda and miRDB restrictions of >95% confidence. There was a total of 27 miRNAs listed as potential regulators of the AOC3 (Fig. 4C). This list of miRNAs was validated using the TCGA cohort and the ones with the highest probability were miR-3190 and miR-3691 since both of them had significantly increased expression in the tumor tissue compared with the normal tissue (Fig. 4D and E). Both of the predicted and highly probable miRNAs were verified in 10 lung cancer patients. The most likely miRNA to contribute to the regulation of AOC3 mRNA was miR-3691-5p because there was an undetectable read number in most specimens for miR-3190 in our samples (Fig. 4F and G).

Low AOC3 expression mediates epithelial-mesenchymal transition (EMT) in lung cancer

Low levels of AOC3 expression conferred poor clinical outcomes in lung cancer patients. Therefore, the present study set out to verify the mechanisms by which AOC3 mediated lung cancer progression. AOC3 expression was knocked down in the LUAD cell line (CL1-5) via the shRNA method with >50% efficiency (Fig. 5A). The cells were then studied to evaluate the effect of AOC3 knockdown on proliferation. Neither the WST-1 nor the BrdU assay indicated that AOC3 affected cell proliferation in lung cancer (Fig. 5B and C, respectively). Cell migration as evaluated via wound healing analysis, revealed enhanced healing (increased migration ability) after AOC3 knockdown (Fig. 5D). In addition, AOC3 knockdown enhanced the mesenchymal characteristics as N-cadherin, vimentin and Slug were increased (Fig. 5E). The rmAOC3 protein (rhAOC3) was added to confirm the observed changes in proliferation and migration. The proliferation did not change even at a high dose (50 ng/ml) of rhAOC3 as evaluated by either WST-1 or BrdU assays (Fig. 5F and G, respectively). Cell migration was reduced after the addition of rhAOC3 in a dose-dependent manner as revealed in the wound-healing assay (Fig. 5H). The mesenchymal characteristics transitioned to epithelial features as E-cadherin was increased and N-cadherin, vimentin and Slug were decreased in a dose-dependent manner (Fig. 5I). The aforementioned results indicated that reduced AOC3 expression played a role in lung cancer progression by increasing cell migration and EMT but not proliferation.

Lung cancers with low levels of AOC3 fail to recruit CD4+ T cells to the tumor in vitro and in vivo

CD4+ T-cell infiltration is a critical factor for determining the TIME against cancer (26). The role of AOC3 in the recruitment of CD4+ T cells remains unclear in lung cancer. To validate the role of AOC3 in mediating the TIME in lung cancer, in vitro and in vivo studies were performed. CD4+ T-cell migration and attachment to lung cancer cells were evaluated. As determined by a cell adhesion assay, CD4+ T-cell attachment to lung cancer cells (CL1-5) was decreased after AOC3 knockdown (Fig. 6A). Before CD4+ T cells arrive at tumor sites, they must traverse the endothelia. A transendothelial migration assay was utilized to reveal the transit of CD4+ T cells through the vascular endothelia. When AOC3 was silenced in cancer cells, CD4+ T-cell migration was reduced, (Fig. 6B) indicating that the lower the AOC3 expression, the fewer CD4+ T cells were recruited. Conversely, when rhAOC3 was added, more CD4+ T cells attached to the lung cancer cells (Fig. 6C). The addition of rhAOC3 increased CD4+ T-cell migration through the vascular endothelial cells (Fig. 6D). These results indicated that AOC3 increased the recruitment of CD4+ T cells to lung cancer sites. AOC3 facilitation of CD4+ T-cell recruitment was validated using an animal model. The in vivo study investigated the number of CD4+ T cells in the lungs of mice with tumors and revealed that the number of CD4+ T cells was increased after rmAOC3 was instilled two times (10 µg/mouse) (Fig. 6E). These results indicated that AOC3 promoted CD4+ T-cell recruitment into the TIME.

miR-3691-5 regulates EMT and cancer migration via epigenetic downregulation of AOC3

To further verify the possible regulatory role of miR-3691-5p in AOC3 expression, miR-3691 mimics were transfected to CL1-5 cells and then their biological functions were assessed. The transfection efficacy of miRNA mimics was monitored by Dharmacon™ siGLO™ transfection indicators and the results revealed that >90% efficacy was achieved (Fig. 7A). miR-3691-5p downregulated expression of AOC3 but not of AOC1 and AOC2 (Fig. 7B). The transfected lung cancer cells were then adopted for wound healing analysis and revealed enhanced healing process in a dose-dependent manner (increased migration ability) (Fig. 7C). Moreover, with miR-3691-5p transfection, AOC3 protein expression was decreased whereas the expression of the mesenchymal markers N-cadherin, vimentin and Slug were increased (Fig. 7D). These results indicated that miR-3691-5p affected cell migration and EMT through AOC3. Furthermore, to evaluate the miR-3691-5p-AOC3 axis in mediating the TIME in lung cancer, in vitro studies were performed. CD4+ T-cell migration and adhesion to lung cancer cells were evaluated. As determined by a cell adhesion assay, CD4+ T-cell attachment to lung cancer cells (CL1-5) was decreased after miR-3691-5p transfection in a dose-dependent manner (Fig. 7E). A transendothelial migration assay revealed that migration of CD4+ T cells through endothelia was reduced in a dose-dependent manner with miR-3691-5p transfection (Fig. 7F). These results indicated that miR-3691-5p attenuated the recruitment of CD4+ T cells. Collectively, it was indicated that miR-3691-5p regulated AOC3 expression to perform its biological functions.

Discussion

The present study attempted to identify a novel factor affecting the different aspects of lung cancer pathogenesis. Through analysis of lung cancer patients via a high-throughput NGS tool, and the utilization of genomic data from different cohorts, the present study determined that AOC3 contributed to lung cancer pathogenesis. Different findings at both transcriptional and translational levels revealed that low levels of AOC3 were a critical factor contributing to cancer development. Low-level AOC3 facilitated mesenchymal transformation and decreased CD4+ T-cell recruitment to lung cancer tumors. It was also revealed that AOC3 expression was under miR3691-5p epigenetic regulation. The strong negative association between AOC3 and the survival rate in patients indicated that it is a key factor involved in lung cancer, and that AOC3 could be a valuable target for drug development (Fig. 8). AOCs have different effects in different types of cancer (14,4244,45,46). High levels of AOCs can act as oncogenes and confer worse clinical outcomes, such as AOC1 in gastric cancer (14) and AOC3 in human glioma (45). On the other hand, low levels of AOCs are associated with worse clinical outcomes, such as AOC3 in colorectal (42,47) and gastric cancer (46). Moreover, decreased AOC3 levels are correlated with lymph node and hepatic metastasis in colorectal cancer (47). The present study is the first one, to the best of our knowledge, which revealed that low-level AOC3 is the critical amine oxidase in lung cancer pathogenesis but not AOC1 or AOC2. Furthermore, miR-3691-5p regulation on AOC3 and its biological functions were defined. miR-3691-5p has been demonstrated to enhance migration and invasion in hepatocellular carcinoma (48). Low expression of AOC3 conferred poor clinical outcomes and lymph node metastasis, supporting the theory that AOC3 acts as a tumor suppressor in lung cancer. Biological function analyses revealed that AOC3 did not affect cell proliferation but that it did influence cell migration. The process of EMT is essential for the enhancement of cell migration (49). AOC3 knockdown enhanced mesenchymal characteristics as revealed by an increase in N-cadherin, vimentin and Slug and attenuated epithelial characteristics as revealed by a decrease in E-cadherin. Exogenous rhAOC3 reversed these mesenchymal patterns. This finding revealed that AOC3 is involved in the maintenance of epithelial characteristics to decrease the metastasis ability of lung cancer. For the first time, the present study has revealed the pathogenic roles of AOC3 in malignant cells and AOC3 per se providing a useful biomarker and prognostic factor in lung cancer patients for clinical diagnosis and treatment.

AOC3 contributes to both innate and acquired immunity (50). Endothelial AOC3 mediates the adhesion of tumor infiltration lymphocytes, lymphokine-activated killer cells and natural killer cells (51) in inflammatory tissue (52) and tumor tissue. An absence of AOC3 leads to a marked reduction in antigen-specific CD4+ recruitment into the airway bronchial lymph nodes (50). In the present study, knockdown of AOC3 in lung cancer cells caused a reduction in CD4+ T-cell extravasation through the endothelial layer and attachment to cancer cells. On the other hand, exogenous rhAOC3 increased transendothelial migration and enhanced CD4+ T-cell attachment onto lung cancer cells. Furthermore, rmAOC3 facilitated CD4+ T-cell recruitment to preexisting lung tumor in a mouse model. These results indicated that AOC3 could modulate the TIME in lung cancer cells, and it may be possible to potentiate its effectiveness by immunotherapy. However, before reaching a definite conclusion, there are some limitations in the present study. Firstly, CD4+ T cells were utilized as a recruiting immune cell to the lung. However, there are more immune cells such as dendritic cells and macrophages and consequently, further investigation may be necessary. Secondly, data from immunohistochemical staining for membrane-bound AOC3 in tumor tissues, which would limit the role of AOC3 in lung cancer, are lacking.

Collectively, the results of the present study confirmed the axis of miR-3691-5p-AOC3 as having a critical role in lung cancer via inhibiting EMT and migration and a determining factor for the recruitment of CD4+ T cells to restore anticancer immunity in the TME. AOC3 expression in lung cancer specimens may provide valuable information for patient prognosis and could have valuable applications when determining a therapeutic strategy in immunotherapy/chemotherapy.

Acknowledgements

The authors would like to thank the CPTAC of UALCAN and the Human Protein Atlas who generated the data used in this publication. The authors would also like to thank the Center for Research Resources and Development in Kaohsiung Medical University for the assistance in Bioinformatics.

Funding

The present study was supported by the Ministry of Science and Technology (grant nos. 110-2314-B-037-124-MY3, 109-2314-B-037-091 and 108-2320-B-037-024-MY3), the Kaohsiung Medical University (grant no. KMU-DK108008), the Kaohsiung Medical University Hospital (grant nos. KMUH108-8R15, KMUH108-8R16 and MUH106-6T06) and the Kaohsiung Municipal Ta-Tung Hospital (grant nos. KMTTH-103-019 and KMTTH-105-051).

Availability of data and materials

The datasets generated and/or analyzed during the present study are not publicly available due to ongoing study in our laboratory but are available from the corresponding author on reasonable request.

Authors' contributions

CYC, YMT and YLH conceptualized the present study. SFJ, PHT and YCH provided the technical support, performed the experiments and acquired the data. CYC provided the software management and analyzed the data. YYC, JYH, WAC and IWC validated the results. KLW, YMT and YLH performed the formal analysis. YLH pursued the investigation and provided the resources. YMT, KLW and YLH performed data curation and interpreted the data. YMT and KLW wrote original draft. YYC and YLH wrote, reviewed and edited the final manuscript. IWC performed visualization of the imaging data. YLH supervised the study, was the project administrator and acquired the funding. YMT and YLH critically revised the manuscript for important intellectual content. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

The protocol of the present study was approved (approval no. KMUH-IRB-20130054 and and KMUH-IRB-G(II)-20180021) by the Institutional Review Board of Kaohsiung Medical University Hospital (Kaohsiung, Taiwan) and written informed consents were acquired from all enrolled patients. All mice procedures were approved by the Institutional Animal Care and Use Committee at Kaohsiung Medical University (Kaohsiung, Taiwan).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

EML4-ALK

echinoderm microtubule-associated protein-like 4-anaplastic lymphoma kinase

AOC3

amine oxidase, copper containing 3

CAO

copper-containing amine oxidase

EGFR

epithelial growth factor receptor

EMT

epithelial-mesenchymal transition

MAO

monoamine oxidase

NGS

next generation sequencings

TIME

tumor immune microenvironment

TME

tumor microenvironment

References

1 

Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Piñeros M, Znaor A and Bray F: Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer. 144:1941–1953. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2019. CA Cancer J Clin. 69:7–34. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Arbour KC and Riely GJ: Systemic therapy for locally advanced and metastatic non-small cell lung cancer: A review. JAMA. 322:764–774. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Miller KD, Nogueira L, Mariotto AB, Rowland JH, Yabroff KR, Alfano CM, Jemal A, Kramer JL and Siegel RL: Cancer treatment and survivorship statistics, 2019. CA Cancer J Clin. 69:363–385. 2019. View Article : Google Scholar : PubMed/NCBI

5 

National Lung Screening Trial Research Team, . Lung Cancer Incidence and mortality with extended follow-up in the National lung screening trial. J Thorac Oncol. 14:1732–1742. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Elmore LW, Greer SF, Daniels EC, Saxe CC, Melner MH, Krawiec GM, Cance WG and Phelps WC: Blueprint for cancer research: Critical gaps and opportunities. CA Cancer J Clin. 71:107–139. 2021. View Article : Google Scholar : PubMed/NCBI

7 

Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, Coussens LM, Gabrilovich DI, Ostrand-Rosenberg S, Hedrick CC, et al: Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 24:541–550. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Duan Q, Zhang H, Zheng J and Zhang L: Turning cold into hot: Firing up the tumor microenvironment. Trends Cancer. 6:605–618. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Vakal S, Jalkanen S, Dahlstrom KM and Salminen TA: Human copper-containing amine oxidases in drug design and development. Molecules. 25:12932020. View Article : Google Scholar : PubMed/NCBI

10 

Mondovì B and Finazzi Agrò A: Structure and function of amine oxidase. Adv Exp Med Biol. 148:141–153. 1982. View Article : Google Scholar : PubMed/NCBI

11 

Buffoni F and Ignesti G: The Copper-containing amine oxidases: Biochemical aspects and functional role. Mol Genet Metab. 71:559–564. 2000. View Article : Google Scholar : PubMed/NCBI

12 

Boomsma F, Bhaggoe UM, van der Houwen AM and van den Meiracker AH: Plasma semicarbazide-sensitive amine oxidase in human (patho)physiology. Biochim Biophys Acta. 1647:48–54. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Salmi M and Jalkanen S: Vascular adhesion protein-1: A cell surface amine oxidase in translation. Antioxid Redox Signal. 30:314–332. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Xu F, Xu Y, Xiong JH, Zhang JH, Wu J, Luo J and Xiong JP: AOC1 Contributes to tumor progression by promoting the AKT and EMT pathways in gastric cancer. Cancer Manag Res. 12:1789–1798. 2020. View Article : Google Scholar : PubMed/NCBI

15 

Lopes de Carvalho L, Bligt-Linden E, Ramaiah A, Johnson MS and Salminen TA: Evolution and functional classification of mammalian copper amine oxidases. Mol Phylogenet Evol. 139:1065712019. View Article : Google Scholar : PubMed/NCBI

16 

Imamura Y, Kubota R, Wang Y, Asakawa S, Kudoh J, Mashima Y, Oguchi Y and Shimizu N: Human retina-specific amine oxidase (RAO): cDNA cloning, tissue expression, and chromosomal mapping. Genomics. 40:277–283. 1997. View Article : Google Scholar : PubMed/NCBI

17 

Bonaiuto E, Lunelli M, Scarpa M, Vettor R, Milan G and Di Paolo ML: A structure-activity study to identify novel and efficient substrates of the human semicarbazide-sensitive amine oxidase/VAP-1 enzyme. Biochimie. 92:858–868. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Salmi M and Jalkanen S: A 90-kilodalton endothelial cell molecule mediating lymphocyte binding in humans. Science. 257:1407–1409. 1992. View Article : Google Scholar : PubMed/NCBI

19 

Stankovic B, Bjørhovde HAK, Skarshaug R, Aamodt H, Frafjord A, Müller E, Hammarström C, Beraki K, Bækkevold ES, Woldbæk PR, et al: Immune cell composition in human non-small cell lung cancer. Front Immunol. 9:31012019. View Article : Google Scholar : PubMed/NCBI

20 

Oja AE, Piet B, van der Zwan D, Blaauwgeers H, Mensink M, de Kivit S, Borst J, Nolte MA, van Lier RAW, Stark R and Hombrink P: Functional heterogeneity of CD4+ tumor-infiltrating lymphocytes with a resident memory phenotype in NSCLC. Front Immunol. 9:26542018. View Article : Google Scholar : PubMed/NCBI

21 

Rhodes DR, Yu J, Shanker K, Deshpande N, Varambally R, Ghosh D, Barrette T, Pandey A and Chinnaiyan AM: ONCOMINE: A cancer microarray database and integrated data-mining platform. Neoplasia. 6:1–6. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Chandrashekar DS, Bashel B, Balasubramanya SAH, Creighton CJ, Ponce-Rodriguez I, Chakravarthi BVSK and Varambally S: UALCAN: A portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia. 19:649–658. 2017. View Article : Google Scholar : PubMed/NCBI

23 

Su LJ, Chang CW, Wu YC, Chen KC, Lin CJ, Liang SC, Lin CH, Whang-Peng J, Hsu SL, Chen CH and Huang CY: Selection of DDX5 as a novel internal control for Q-RT-PCR from microarray data using a block bootstrap re-sampling scheme. BMC Genomics. 8:1402007. View Article : Google Scholar : PubMed/NCBI

24 

Okayama H, Kohno T, Ishii Y, Shimada Y, Shiraishi K, Iwakawa R, Furuta K, Tsuta K, Shibata T, Yamamoto S, et al: Identification of genes upregulated in ALK-positive and EGFR/KRAS/ALK-negative lung adenocarcinomas. Cancer Res. 72:100–111. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Landi MT, Dracheva T, Rotunno M, Figueroa JD, Liu H, Dasgupta A, Mann FE, Fukuoka J, Hames M, Bergen AW, et al: Gene expression signature of cigarette smoking and its role in lung adenocarcinoma development and survival. PLoS One. 3:e16512008. View Article : Google Scholar : PubMed/NCBI

26 

Beer DG, Kardia SL, Huang CC, Giordano TJ, Levin AM, Misek DE, Lin L, Chen G, Gharib TG, Thomas DG, et al: Gene-expression profiles predict survival of patients with lung adenocarcinoma. Nat Med. 8:816–824. 2002. View Article : Google Scholar : PubMed/NCBI

27 

Stearman RS, Dwyer-Nield L, Zerbe L, Blaine SA, Chan Z, Bunn PA Jr, Johnson GL, Hirsch FR, Merrick DT, Franklin WA, et al: Analysis of orthologous gene expression between human pulmonary adenocarcinoma and a carcinogen-induced murine model. Am J Pathol. 167:1763–1775. 2005. View Article : Google Scholar : PubMed/NCBI

28 

Selamat SA, Chung BS, Girard L, Zhang W, Zhang Y, Campan M, Siegmund KD, Koss MN, Hagen JA, Lam WL, et al: Genome-scale analysis of DNA methylation in lung adenocarcinoma and integration with mRNA expression. Genome Res. 22:1197–1211. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Garber ME, Troyanskaya OG, Schluens K, Petersen S, Thaesler Z, Pacyna-Gengelbach M, van de Rijn M, Rosen GD, Perou CM, Whyte RI, et al: Diversity of gene expression in adenocarcinoma of the lung. Proc Natl Acad Sci USA. 98:13784–13789. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Hou J, Aerts J, den Hamer B, van Ijcken W, den Bakker M, Riegman P, van der Leest C, van der Spek P, Foekens JA, Hoogsteden HC, et al: Gene expression-based classification of non-small cell lung carcinomas and survival prediction. PLoS One. 5:e103122010. View Article : Google Scholar : PubMed/NCBI

31 

Wachi S, Yoneda K and Wu R: Interactome-transcriptome analysis reveals the high centrality of genes differentially expressed in lung cancer tissues. Bioinformatics. 21:4205–4208. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Bhattacharjee A, Richards WG, Staunton J, Li C, Monti S, Vasa P, Ladd C, Beheshti J, Bueno R, Gillette M, et al: Classification of human lung carcinomas by mRNA expression profiling reveals distinct adenocarcinoma subclasses. Proc Natl Acad Sci USA. 98:13790–13795. 2001. View Article : Google Scholar : PubMed/NCBI

33 

Uhlen M, Zhang C, Lee S, Sjöstedt E, Fagerberg L, Bidkhori G, Benfeitas R, Arif M, Liu Z, Edfors F, et al: A pathology atlas of the human cancer transcriptome. Science. 357:eaan25072017. View Article : Google Scholar : PubMed/NCBI

34 

Gyorffy B, Surowiak P, Budczies J and Lanczky A: Online survival analysis software to assess the prognostic value of biomarkers using transcriptomic data in non-small-cell lung cancer. PLoS One. 8:e822412013. View Article : Google Scholar : PubMed/NCBI

35 

Mende DR, Letunic I, Maistrenko OM, Schmidt TS, Milanese A, Paoli L, Hernández-Plaza A, Orakov AN, Forslund SK, Sunagawa S, et al: proGenomes2: An improved database for accurate and consistent habitat, taxonomic and functional annotations of prokaryotic genomes. Nucleic Acids Res. 48:D621–D625. 2020.PubMed/NCBI

36 

Sticht C, De La Torre C, Parveen A and Gretz N: miRWalk: An online resource for prediction of microRNA binding sites. PLoS One. 13:e02062392018. View Article : Google Scholar : PubMed/NCBI

37 

Enright AJ, John B, Gaul U, Tuschl T, Sander C and Marks DS: MicroRNA targets in Drosophila. Genome Biol. 5:R12003. View Article : Google Scholar : PubMed/NCBI

38 

Chen Y and Wang X: miRDB: An online database for prediction of functional microRNA targets. Nucleic Acids Res. 48:D127–D131. 2020. View Article : Google Scholar : PubMed/NCBI

39 

Ke HL, Li WM, Lin HH, Hsu WC, Hsu YL, Chang LL, Huang CN, Li CC, Chang HP, Yeh HC, et al: Hypoxia-regulated MicroRNA-210 overexpression is associated with tumor development and progression in upper tract urothelial carcinoma. Int J Med Sci. 14:578–584. 2017. View Article : Google Scholar : PubMed/NCBI

40 

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

41 

Shao L, Li H, Chen J, Song H, Zhang Y, Wu F, Wang W, Zhang W, Wang F, Li H and Tang D: Irisin suppresses the migration, proliferation, and invasion of lung cancer cells via inhibition of epithelial-to-mesenchymal transition. Biochem Biophys Res Commun. 485:598–605. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Ward ST, Weston CJ, Shepherd EL, Hejmadi R, Ismail T and Adams DH: Evaluation of serum and tissue levels of VAP-1 in colorectal cancer. BMC Cancer. 16:1542016. View Article : Google Scholar : PubMed/NCBI

43 

Sun WY, Choi J, Cha YJ and Koo JS: Evaluation of the expression of amine oxidase proteins in breast cancer. Int J Mol Sci. 18:27752017. View Article : Google Scholar : PubMed/NCBI

44 

Kostoro J, Chang SJ, Clark Lai YC, Wu CC, Chai CY and Kwan AL: Overexpression of vascular adhesion protein-1 is associated with poor prognosis of astrocytomas. APMIS. 124:462–468. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Chang SJ, Tu HP, Lai YCC, Luo CW, Nejo T, Tanaka S, Chai CY and Kwan AL: Increased vascular adhesion protein 1 (VAP-1) levels are associated with alternative M2 macrophage activation and poor prognosis for human gliomas. Diagnostics (Basel). 10:2562020. View Article : Google Scholar : PubMed/NCBI

46 

Yasuda H, Toiyama Y, Ohi M, Mohri Y, Miki C and Kusunoki M: Serum soluble vascular adhesion protein-1 is a valuable prognostic marker in gastric cancer. J Surg Oncol. 103:695–699. 2011. View Article : Google Scholar : PubMed/NCBI

47 

Toiyama Y, Miki C, Inoue Y, Kawamoto A and Kusunoki M: Circulating form of human vascular adhesion protein-1 (VAP-1): Decreased serum levels in progression of colorectal cancer and predictive marker of lymphatic and hepatic metastasis. J Surg Oncol. 99:368–372. 2009. View Article : Google Scholar : PubMed/NCBI

48 

Du W, Zhang X and Wan Z: miR-3691-5p promotes hepatocellular carcinoma cell migration and invasion through activating PI3K/Akt signaling by targeting PTEN. Onco Targets Ther. 12:4897–4906. 2019. View Article : Google Scholar : PubMed/NCBI

49 

Leggett SE, Hruska AM, Guo M and Wong IY: The epithelial-mesenchymal transition and the cytoskeleton in bioengineered systems. Cell Commun Signal. 19:322021. View Article : Google Scholar : PubMed/NCBI

50 

Dunkel J, Aguilar-Pimentel JA, Ollert M, Fuchs H, Gailus-Durner V, de Angelis MH, Jalkanen S, Salmi M and Veres TZ: Endothelial amine oxidase AOC3 transiently contributes to adaptive immune responses in the airways. Eur J Immunol. 44:3232–3239. 2014. View Article : Google Scholar : PubMed/NCBI

51 

Irjala H, Salmi M, Alanen K, Grénman R and Jalkanen S: Vascular adhesion protein 1 mediates binding of immunotherapeutic effector cells to tumor endothelium. J Immunol. 166:6937–6943. 2001. View Article : Google Scholar : PubMed/NCBI

52 

Stolen CM, Marttila-Ichihara F, Koskinen K, Yegutkin GG, Turja R, Bono P, Skurnik M, Hänninen A, Jalkanen S and Salmi M: Absence of the endothelial oxidase AOC3 leads to abnormal leukocyte traffic in vivo. Immunity. 22:105–115. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2021
Volume 46 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chang C, Wu K, Chang Y, Tsai P, Hung J, Chang W, Jian S, Huang Y, Chong I, Tsai Y, Tsai Y, et al: Amine oxidase, copper containing 3 exerts anti‑mesenchymal transformation and enhances CD4<sup>+</sup> T‑cell recruitment to prolong survival in lung cancer. Oncol Rep 46: 203, 2021
APA
Chang, C., Wu, K., Chang, Y., Tsai, P., Hung, J., Chang, W. ... Hsu, Y. (2021). Amine oxidase, copper containing 3 exerts anti‑mesenchymal transformation and enhances CD4<sup>+</sup> T‑cell recruitment to prolong survival in lung cancer. Oncology Reports, 46, 203. https://doi.org/10.3892/or.2021.8154
MLA
Chang, C., Wu, K., Chang, Y., Tsai, P., Hung, J., Chang, W., Jian, S., Huang, Y., Chong, I., Tsai, Y., Hsu, Y."Amine oxidase, copper containing 3 exerts anti‑mesenchymal transformation and enhances CD4<sup>+</sup> T‑cell recruitment to prolong survival in lung cancer". Oncology Reports 46.3 (2021): 203.
Chicago
Chang, C., Wu, K., Chang, Y., Tsai, P., Hung, J., Chang, W., Jian, S., Huang, Y., Chong, I., Tsai, Y., Hsu, Y."Amine oxidase, copper containing 3 exerts anti‑mesenchymal transformation and enhances CD4<sup>+</sup> T‑cell recruitment to prolong survival in lung cancer". Oncology Reports 46, no. 3 (2021): 203. https://doi.org/10.3892/or.2021.8154