Open Access

Establishment of a patient‑derived mucoepidermoid carcinoma cell line with the CRTC1‑MAML2 fusion gene

  • Authors:
    • Kazuma Noguchi
    • Shuji Kanda
    • Kazunari Yoshida
    • Yusuke Funaoka
    • Koji Yamanegi
    • Kyohei Yoshikawa
    • Kazuki Takaoka
    • Hiromitsu Kishimoto
    • Yoshiro Nakano
  • View Affiliations

  • Published online on: February 2, 2022     https://doi.org/10.3892/mco.2022.2508
  • Article Number: 75
  • Copyright: © Noguchi 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

Mucoepidermoid carcinoma (MEC) is the most common malignant tumor of the major and minor salivary glands. Surgical resection is the only curative treatment and there is no effective post‑operative therapy for MEC. The present study reports an Institutional Review Board‑approved case of a 45‑year‑old Japanese female diagnosed with low-grade MEC in the hard palate. Radical resection, supraomohyoid neck dissection and antero‑lateral thigh flap reconstruction was performed. A MEC cell line was then established from the resected tumor tissue. Short tandem repeat profiling confirmed the origin and authenticity of the cell line, that harbors a CRTC1‑MAML2 translocation, which is frequently observed in MEC. Amphiregulin (AREG), identified as one of the targets of the CRTC1‑MAML2 fusion gene, was expressed in the cell line. The AREG receptor, epidermal growth factor receptor (EGFR) was also highly phosphorylated. The results predicted that AREG‑EGFR signaling, which is required for tumor growth and survival, might be activated in the cell line in a cell‑autonomous manner. As AREG expression is associated with EGFR‑targeted drug resistance, this cell line might assist with the identification of novel strategies for MEC treatment.

Introduction

Mucoepidermoid carcinoma (MEC), representing 5% of all salivary gland tumors and 26% of malignant salivary gland tumors registered for the last 39 years in Hiroshima, Japan, is the most common malignant tumor of the major and minor salivary glands (1,2). MEC is characterized by its cellular heterogeneity and consists of mucin-producing, epidermoid and intermediate cells. Clinical and pathological parameters (age, tumor size, presence of cervical lymphadenopathy, distant spread, perineural invasion and histological grade) of MEC have been associated with tumor biological behavior and patient management (3). Pathological classification of MEC is graded as low-, intermediate- or high-grade based on adverse features, such as perineural invasion, angiolymphatic invasion, coagulative necrosis, infiltrative growth, high mitotic rate, anaplasia and cystic components of <20% (4).

An important genetic abnormality in MEC is the translocation between chromosomes 11q and 19p, which has been hypothesized to be an early event in the pathogenesis of MEC (5,6), and has been reported in >50% of MEC tumors (7). Low-grade tumors have a higher incidence rate of this fusion compared with that in high-grade tumors (8) and patients with fusion-positive cancer tend to have improved survival time, with significantly lower risks of recurrence, metastases or cancer-related mortality (9). The majority of fusion genes in MEC are associated with a specific chromosomal t(11;19) (q14-21;p12-13) translocation that joins exon 1 of the cAMP response element-binding (CREB) protein-binding domain of CREB-regulated transcription coactivator 1 (CRTC1) gene to exons 2-5 of the Notch coactivator mastermind-like gene 2 (MAML2) gene, resulting in the expression of a new CRTC1-MAML2 fusion gene (10). This translocation generates a fusion protein comprised of CRTC1 (also called MECT1, TORC1 or WAMP1) at 19q21 and the C-terminal transcriptional activation domain of MAML2 at 11q21 (11-14). Previous analysis suggested that another member of the CRTC family, at 15q26, CRTC3, also fused with MAML2 (15). Okabe et al (16) and Nakayama et al (17) showed that CRTC1-MAML2 or CRTC3-MAML2 fusions occurred in 40-80% of primary salivary gland MECs, and was associated with a distinct tumor subset that had favorable clinicopathological features and an indolent clinical course.

Previously, amphiregulin (AREG), a member of the epidermal growth factor (EGF) family, was identified as a target of the CRTC1-MAML2 fusion gene and secreted AREG was shown to activate EGF receptor (EGFR) signaling in an autocrine manner (18). Furthermore, mutations in EGFR itself are rare in salivary gland carcinomas (19), while copy number alternations in EGFR are frequently found in high-grade MEC, regardless of fusion gene positivity (20). The molecular pathology and oncology of MEC are still poorly understood. Established authentic cell lines are essential to determine the biological characteristics of MEC, and a number of cell cultures and models have emerged; however, the cell line usability is limited (21). The present study reports the establishment of a MEC cell line (HCM-MEC010) carrying the CRTC1-MAML2 fusion gene and activated EGFR. The potential uses for this cell line will also be discussed to understand the biological characteristics of MEC.

Materials and methods

Cell line generation and cell culture

A patient with MEC provided consent in accordance with Hyogo College of Medicine (Hyogo, Japan) institutional policies. Tumor samples were obtained according to an approved Institutional Review Board protocol of Hyogo College of Medicine (approval no. 276; Hyogo, Japan). The present study was also conducted in accordance with the Declaration of Helsinki. Clinical and pathological data were collected from the medical records of the patient. Tumor tissues were minced into 1-2-mm pieces with a disposable scalpel and placed in primary culture. To separate the stromal cells from the mass culture, a magnetic-activated cell sorting (MACS) system was used. Briefly, MACS buffer, containing 1X PBS, 0.5% BSA, 2 mM EDTA (pH 7.2) (cat. no. 130-042-901; Miltenyi Biotec Inc.), was pre-cooled to 4˚C. To remove the fibroblasts, the single cell suspension was centrifuged at 300 x g for 10 min at room temperature. and positive selection was performed using CD326 (EpCAM) MicroBeads and a MidiMACSTM Separator (Miltenyi Biotic GmbH), according to the manufacturer's instructions. The obtained primary human MEC cells were seeded in F-medium (22) with 10 µM Y-27632 (FUJIFILM Wako Pure Chemical Corporation). After 1 week, the culture medium was replaced with fresh medium, which was changed every 4 days thereafter. At the same time, the fibroblasts derived from the tumor tissue of the same patient, were obtained and grown in F-medium. Once cells reached confluence (80%), they were washed with PBS (Mg2+ and Ca2+ free) (23) and detached with 0.05% EDTA/trypsin for 5 min at 38˚C (24). After centrifugation at 167 x g for 5 min at 4˚C, the MEC cells were resuspended in F-medium, containing Y-27632 and seeded (0.3x106 cells) in 60 mm dishes. An epithelial cell line was successfully established from the sample of the patient and was termed HCM-MEC010. The morphology of the exponentially proliferating cells in a monolayer was reviewed and documented using inverted phase contrast microscopy. The cells were also tested for mycoplasma infection using the MycoAlert® Assay (Lonza Group, Ltd.) and the cell culture growth medium and with fluorescent microscopy using the Mycoplasma Hoechst Stain Assay (MP Biomedicals, LLC).

Short tandem repeat (STR) authentication of the MEC cell line

To verify the identity of the cell line, genomic DNA was extracted from the blood of the patient, whose tumor sample was used to generate the HCM-MEC010 cell line, as well as from the cell line using the QIAamp DNA Mini kit (Qiagen, Inc.) according to the manufacturer's protocol. DNA genotyping using STR profiling was performed using the GenePrint 10 System (Promega Corporation) and the Applied Biosystems 3130xl Analyzer (Applied Biosystems; Thermo Fisher Scientific, Inc.) and analyzed by BEX Co., Ltd. The evaluation value (EV) was determined using the following equation: EV=(number of coincidental peaks) x 2/total number of peaks in cell A and total number of peaks in cell B.

Reverse transcription (RT)-PCR of the CRTC1-MAML2 fusion oncogene

The HCM-MEC010 cell line was plated in 100-mm dishes and cultured to 90% confluence. RNA was extracted using TRIzol® (Invitrogen; Thermo Fisher Scientific, Inc.) and RT-PCR was performed using the PrimeScript RT-PCR kit (Takara Bio, Inc.) according to the manufacturer's instructions. The following primers were used: CRTC1 forward 1, 5'-TTCGAGGAGGTCATGAAGGA-3' and 2, 5'-ATGGCGACTTCGAACAATCCGCGGAA-3'; MAML2 reverse 1, 5'-TTGCTGTTGGCAGGAGATAG-3' and 2, 5'-GGGTCGCTTGCTGTTGGCAGGAG-3' (18), which amplified 101 and 194 bp fragments, respectively. Amplification of the GAPDH gene (forward, 5'-CAATGACCCCTTCATTGACC-3' and reverse, 5'-GACAAGCTTCCCGTTCTCAG-3') was performed as a control. Successfully amplified RT-PCR products of the CRTC1-MAML2 fusion gene were purified and sequenced (24) using BigDye Terminator v3.1 Cycle Sequencing kit (Thermo Fisher Scientific, Inc.) and 2% agarose gel electrophoresis.

Western blot analysis

The culture medium was removed and the cells were washed with PBS (Mg2+ and Ca2+ free). RIPA buffer was added (cat. no. sc-24948; Santa Cruz, Inc.) and the cells were incubated at 4˚C for 60 min, then centrifuged at 12,000 x g for 20 min 4˚C. The supernatant was the total cell lysate. Proteins were extracted from the HCM-MEC010 and human tongue squamous cell carcinoma (SAS; purchased from the Japanese Collection of Research Bioresources Cell Bank) cell lines as previously described (25). Protein concentration was measured using a Bradford assay (26) Western blot analysis was performed as previously described (25). The primary and secondary antibodies are listed in Table I. The protein expression ratio, compared with that in SAS cells, was measured using ImageJ v1.53e software (National Institutes of Health). The data are presented as the mean ± SD. The experiment was repeated three times.

Table I

Primary and secondary antibodies used for western blot analysis and immunofluorescence.

Table I

Primary and secondary antibodies used for western blot analysis and immunofluorescence.

A, Primary antibodies
 Dilution 
NameCat. no.Western blot ImmunofluorescenceSupplier
Rabbit monoclonal anti-EGFR42671/10001/60CST
Rabbit monoclonal anti-p-EGFR37771/1000 CST
Rabbit monoclonal anti-AKT46911/1000 CST
Rabbit monoclonal anti-p-AKT40601/1000 CST
Rabbit monoclonal anti-AREG16036-1-AP1/1000 ProteinTech Group, Inc.
Rabbit monoclonal anti-E-cadherin31951/10001/100CST
Rabbit monoclonal anti-N-cadherin131161/10001/100CST
Rabbit monoclonal anti-vimentin57411/10001/100CST
Rabbit monoclonal anti-tubulin21481/1000 CST
Mouse monoclonal anti-actin477781/1000  
B, Secondary antibodies
 Dilution 
NameCat. no.Western blot ImmunofluorescenceSupplier
Alexa Flur 488 goat anti-rabbit IgG (H+L)A-11008 1/400Molecular Probes; Thermo Fisher Scientific, Inc.
Anti-mouse IgG, HRP-linked70761/1000 CST
Anti-IgG (H+L chain) rabbit pAb-HRP4581/10000 Molecular and Biological Laboratories Co., Ltd.
Goat anti-mouse HRPab970231/1000 Abcam

[i] CST, Cell Signaling Technology, Inc.; p, phosphorylated.

Immunofluorescence staining

The cultured HCM-MEC010 and SAS cell lines were fixed in 3.7% formaldehyde for 20 min at room temperature. After permeabilization with 0.2% Triton-X/PBS for 5 min at room temperature, the cells were blocked with 2% (w/v) BSA (Nacalai Tesque, Inc.)/PBS, then washed with PBS (Mg2+ and Ca2+ free) and incubated with the primary antibodies overnight at 4˚C. The cells were washed with PBS (Mg2+ and Ca2+ free), then incubated with the secondary antibody and Rhodamine phalloidin (Cytoskeleton, Inc.) for 2 h at room temperature. The samples were mounted in Vecta shield containing DAPI (Vector Laboratories). Fluorescent images were captured using a confocal laser-scanning microscope (LSM780; Zeiss AG). The primary and secondary antibodies are listed in Table I.

RNA analysis

RNA-Sequencing (RNA-Seq) libraries were generated using RNA extracted from the HCM-MEC010 cell line, as previously described (27), with the TruSeq Stranded mRNA Library Prep kit for Illumina, Inc., following the manufacturer's instructions, then sequenced on a NovaSeq 6000 System (Illumina, Inc.). The analysis was performed by Takara Bio, Inc.

Hematoxylin and eosin-staining

A section of the hard palate was fixed in 10% formalin solution at room temperature for 24 h and embedded in paraffin. Sections (5-µm thick) were cut from the paraffin blocks and stained with hematoxylin (0.09%) for 5 min and eosin (0.13%) for 9 min at room temperature according to standard methods (28). The images were captured using a light microscope (BX51; Olympus Corporation).

Patient

A 45-year-old Japanese female noticed spontaneous dull pain and swelling in her hard palate for 1 month and was referred to Hyogo College of Medicine, Nishinomiya, Hyogo, Japan on January, 2019. On examination, diffuse swelling was observed in the right hard palate. There was no trismus. The surface of the mass was smooth and was soft on palpation (Fig. 1A). Bilateral cervical lymph nodes were palpable, but painless and mobile. Magnetic resonance imaging showed an irregular mass measuring 30x20x18 mm in the right hard palate, and resorption in the nasal septum and posterior wall of the maxillary sinus (Fig. 1B). The clinical diagnosis was a malignant tumor of the hard palate. A biopsy was performed intraorally and the lesion was pathologically diagnosed as low-grade MEC using Armed Forces Institute of Pathology (29).

Results

The patient was admitted to Hyogo College of Medicine, Nishinomiya, Hyogo, Japan and treated by partial resection of the hard palate, supraomohyoid neck dissection and reconstruction using an anterolateral thigh flap under general anesthesia. Hematoxylin and eosin-stained tumor tissue microscopically showed an overlying stratified squamous epithelium, mucous cells and squamous cells that were polygonal-to-ovoid in shape with eosinophilic cytoplasms (Fig. 1C). The mucous cells were cuboidal or goblet-like and tended to line the cysts. The squamous cells formed solid sheets. The tumor was diagnosed as mucoepidermoid carcinoma, low-grade type, pT4aN0M0 MEC of the hard palate. All dissected cervical lymph nodes showed no metastatic cells. At the 30-month follow up, the patient's prognosis was excellent and she had maintained a disease-free status.

Establishment of a MEC cell line from a patient tumor

A new MEC cell line, termed HCM-MEC010 was established, which maintained a cobblestone epithelial-like morphology for at least 30 passages (Fig. 2A and B). To confirm that the HCM-MEC010 cell line was derived from the tumor sample of the patient, STR profiling was performed using the DNA extracted from the high-passage HCM-MEC010 cell line and the blood from the patient. Genotypic analysis confirmed that the cell line was derived from the tumor and no contamination with other cell types was detected (EV, 1.0). (Table SI; Figs. S1 and S2).

RT-PCR analysis reveals that HCM-MEC010 cells express the CRTC1-MAML2 fusion gene

As the CRTC1-MAML2 gene fusion is common in MEC (9), the fusion event was analyzed in the HCM-MEC010 cell line using RT-PCR. Fig. 3A shows the translocation event between chromosomes 11 and 19, while Fig. 3B shows the RT-PCR amplified fragments (lane 1, 101 bp and lane 2, 196 bp) using primer sets 1 or 2, respectively. The fusion transcript of CRTC1 and MAML2 genes was confirmed using Sanger sequencing (Fig. 3C). This revealed the fusion products of CRTC1 exon 1 and MAML2 exon 2 with the predicted splicing event, indicating that a translocation event had occurred between the first introns of CRTC1 and MAML2.

Protein expression in the HCM-MEC010 cell line

Next, the protein expression of the epithelial and mesenchymal markers in the HCM-MEC010 cell line was confirmed using immunofluorescent staining. EGFR and E-cadherin were expressed on the cell membrane in the HCM-MEC010 cells, while N-cadherin expression was only faintly detected. Vimentin expression was also detected in HCM-MEC010 cells (Fig. 4).

HCM-MEC010 cells express AREG and show EGFR activation

As the AREG-EGFR signaling cascade has been identified as a CRTC1-MAML2 fusion gene target (18), AREG expression and the status of the EGFR cascade was analyzed in the HCM-MEC010 cell line. The human tongue SAS cell line was used as a comparison as the SAS cell line contains a mutation in the HER4 gene, which encodes one of the other types of human EGFR, and the authentic EGFR pathway is not involved in cell proliferation (30). EGFR was expressed in both cell types, but the AREG expression level was much higher in the HCM-MEC010 cell line compared with that in the SAS cell line (Fig. 5). Furthermore, EGFR was phosphorylated (p) in the HCM-MEC010 cell line compared with that in the SAS cell line, indicating the activation of the EGFR pathway. In addition, the expression level of AKT and p-AKT was lower in the HCM-MEC010 cell line compared with that in the SAS cell line. In the SAS cell line, AKT can be phosphorylated by both the AREG-EGFR and HER4 pathways (31,32), and high levels of AKT phosphorylation in the SAS cell line must represent an additive effect of HER4 pathway activation (33). E-cadherin was expressed at higher levels in the HCM-MEC010 cell line compared with that in the SAS cell line. Vimentin expression was detected in small amounts in both the HCM-MEC010 and SAS cell lines (Fig. 5).

RNA-seq analysis of the HCM-MEC010 cell line revealed epidermoid characteristics

To further characterize the HCM-MEC010 cell line, RNA-Seq analysis was performed. MEC is known to be composed of a mixture of mucous, epidermoid, and intermediate cells (34). RNA-Seq analysis revealed the high expression level of genes in the keratin family, including KRT5, KRT14, KRT6A, KRT17, and KRT7. Table II lists the top 200 expressed genes. However, expression of the mucous cell marker MUC was not detected. These results, together with the cell morphology results, suggest that the HCM-MEC010 cell line is considered to be of epidermoid, but not mucinous, origin.

Table II

RNA-Sequencing data for the MEC cell line.

Table II

RNA-Sequencing data for the MEC cell line.

Entrez gene IDGene symbolDescriptionTPM
6280S100A9S100 calcium binding protein A925012.1582
-RNR2-19355.6582
6277S100A6S100 calcium binding protein A615991.2832
1915EEF1A1Eukaryotic translation elongation factor 1 α 113189.91406
9168TMSB10Thymosin β 1011790.02637
3852KRT5Keratin 510268.93652
6590SLPISecretory leukocyte peptidase inhibitor9873.083008
6222RPS18Ribosomal protein S189865.65332
3861KRT14Keratin 149010.899414
301ANXA1Annexin A18724.607422
302ANXA2Annexin A28183.348633
6130RPL7ARibosomal protein l7a7233.334961
3853KRT6AKeratin 6A7038.019531
6205RPS11Ribosomal protein S116782.036133
3872KRT17Keratin 176779
6282S100A11S100 calcium binding protein A116758.137695
57402S100A14S100 calcium binding protein A146577.536133
6136RPL12Ribosomal protein L126571.3125
6202RPS8Ribosomal protein S85322.165039
23521RPL13ARibosomal protein l13a5300.688477
6175RPLP0Ribosomal protein lateral stalk subunit P05200.693848
6144RPL21(2)Ribosomal protein L215167.824219
7114TMSB4XThymosin β 4 X-linked4858.939453
6201RPS7Ribosomal protein S74807.380371
6281S100A10S100 calcium binding protein A104705.583984
6206RPS12Ribosomal protein S124461.196777
6230RPS25Ribosomal protein S254362.669922
6122RPL3Ribosomal protein L34190.952148
2597GAPDH Glyceraldehyde-3-phosphate dehydrogenase4084.755371
4502MT2AMetallothionein 2A4024.866699
3855KRT7Keratin 73954.171631
6194RPS6Ribosomal protein S63893.395996
6152RPL24Ribosomal protein L243876.068848
6142RPL18ARibosomal protein l18a3788.035645
60ACTBActin β3734.336914
6156RPL30Ribosomal protein L303719.687988
6279S100A8S100 calcium binding protein A83664.906982
10399RACK1Receptor for activated C kinase 13650.605225
100133941CD24CD24 molecule3593.078369
6191RPS4XRibosomal protein S4 X-linked3478.193848
-RNR1-3421.098877
2950GSTP1Glutathione S-transferase pi 13338.100586
6187RPS2Ribosomal protein S23280.663086
6207RPS13Ribosomal protein S133170.464111
11224RPL35Ribosomal protein L353153.293701
1937EEF1GEukaryotic translation elongation factor 1 γ3140.122559
6125RPL5Ribosomal protein L53137.963623
6170RPL39Ribosomal protein L393100.071045
4637MYL6Myosin light chain 63067.146484
3868KRT16Keratin 163044.359619
4736RPL10ARibosomal protein l10a2961.195801
6141RPL18Ribosomal protein L182929.649658
1476CSTBCystatin B2910.897217
6124RPL4Ribosomal protein L42865.387207
4070TACSTD2Tumor associated calcium signal transducer 22787.421387
6147RPL23ARibosomal protein l23a2730.734131
71ACTG1Actin γ 12705.085693
220ALDH1A3Aldehyde dehydrogenase 1 family member A32588.035156
6135RPL11Ribosomal protein L112561.839844
3880KRT19Keratin 192536.187012
6132RPL8Ribosomal protein L82522.498047
6181RPLP2Ribosomal protein lateral stalk subunit P22490.632813
3866KRT15Keratin 152464.382324
6699SPRR1BSmall proline rich protein 1B2444.126465
6159RPL29Ribosomal protein L292439.016113
2512FTLFerritin light chain2432.441895
6193RPS5Ribosomal protein S52432.29126
6233RPS27ARibosomal protein s27a2403.434326
6129RPL7Ribosomal protein L72332.271973
6273S100A2S100 calcium binding protein A22289.59375
6133RPL9Ribosomal protein L92237.880371
1475CSTACystatin A2159.565186
6128RPL6Ribosomal protein L62119.131592
2495FTH1Ferritin heavy chain 12094.474121
3921RPSARibosomal protein SA2085.400391
5266PI3Peptidase inhibitor 32079.049805
2171FABP5Fatty acid binding protein 52073.613281
5052PRDX1Peroxiredoxin 12053.132568
3956LGALS1Galectin 12031.178833
6143RPL19Ribosomal protein L192021.314087
25818KLK5Kallikrein related peptidase 51822.719238
3939LDHALactate dehydrogenase A1803.661499
6176RPLP1Ribosomal protein lateral stalk subunit P11802.172241
51458RHCGRh family C glycoprotein1785.147339
6303SAT1Spermidine/spermine N1-acetyltransferase 11763.299316
9982FGFBP1Fibroblast growth factor binding protein 11742.557251
7178TPT1Tumor protein, translationally-controlled 11741.52832
6227RPS21Ribosomal protein S211726.189087
3934LCN2Lipocalin 21720.297241
3315HSPB1Heat shock protein family B (small) member 11668.157104
1973EIF4A1Eukaryotic translation initiation factor 4A11623.40625
1938EEF2Eukaryotic translation elongation factor 21612.361694
5055SERPINB2Serpin family B member 21610.25
2810SFNStratifin1591.703979
6703SPRR2DSmall proline rich protein 2D1568.223389
26986PABPC1Poly(A) binding protein cytoplasmic 11534.452637
6204RPS10Ribosomal protein S101532.445679
10410IFITM3Interferon induced transmembrane protein 31529.12146
6189RPS3ARibosomal protein S3A1509.361816
6154RPL26Ribosomal protein L261432.493286
3918LAMC2Laminin subunit γ 21429.380249
83442SH3BGRL3SH3 domain binding glutamate rich protein like 31395.721313
6139RPL17Ribosomal protein L171375.231934
1933EEF1B2Eukaryotic translation elongation factor 1 β 21351.424194
10974ADIRFAdipogenesis regulatory factor1348.772461
6134RPL10Ribosomal protein L101336.026611
5268SERPINB5Serpin family B member 51335.237183
6700SPRR2ASmall proline rich protein 2A1285.784912
10094ARPC3Actin related protein 2/3 complex subunit 31270.268311
2152F3Coagulation factor III, tissue factor1268.36792
2197FAUFAU ubiquitin like and ribosomal protein S30 fusion1255.56189
9124PDLIM1PDZ and LIM domain 11252.652954
64065PERPP53 apoptosis effector related to PMP221252.282227
4869NPM1Nucleophosmin 11247.643188
7295TXNThioredoxin1169.833984
3553IL1BInterleukin 1 β1166.45752
5054SERPINE1Serpin family E member 11154.025146
6171RPL41Ribosomal protein L411152.395996
25824PRDX5Peroxiredoxin 51133.30603
6173RPL36ARibosomal protein l36a1111.359619
5315PKMPyruvate kinase M1/21092.81897
1072CFL1Cofilin 11085.361328
6289SAA2Serum amyloid A21073.526978
4071TM4SF1Transmembrane 4 L six family member 11063.068237
506ATP5F1BATP synthase F1 subunit β1047.457275
5834PYGBGlycogen phosphorylase B1047.218994
928CD9CD9 molecule1021.081299
10628TXNIPThioredoxin interacting protein1021.076111
103910MYL12BMyosin light chain 12B1012.033325
3854KRT6BKeratin 6B1011.945374
3688ITGB1Integrin subunit β 11004.073792
3312HSPA8Heat shock protein family A (Hsp70) member 81000.302063
6288SAA1Serum amyloid A1999.111145
1382CRABP2Cellular retinoic acid binding protein 2986.4415283
6224RPS20Ribosomal protein S20975.680481
10109ARPC2Actin related protein 2/3 complex subunit 2966.9124146
1992SERPINB1Serpin family B member 1952.090332
306ANXA3Annexin A3951.5344238
4501MT1XMetallothionein 1X939.5453491
5660PSAPProsaposin936.7683105
6286S100PS100 calcium binding protein P924.9679565
567B2M β-2-microglobulin919.1690674
3914LAMB3Laminin subunit β 3918.9204102
1308COL17A1Collagen type XVII α 1 chain916.5231323
824CAPN2Calpain 2912.717041
2706GJB2Gap junction protein β 2904.8463745
3860KRT13Keratin 13894.9153442
3646EIF3EEukaryotic translation initiation factor 3 subunit E893.5683594
5479PPIBPeptidylprolyl isomerase B883.137207
7316UBCUbiquitin C875.6885986
3326HSP90AB1Heat shock protein 90 α family class B member 1871.9744263
642587MIR205HGMIR205 host gene864.2874146
468ATF4Activating transcription factor 4850.9224243
140576S100A16S100 calcium binding protein A16849.9338989
6155RPL27Ribosomal protein L27841.65802
6228RPS23Ribosomal protein S23837.4863281
25984KRT23Keratin 23837.0656738
54541DDIT4DNA damage inducible transcript 4831.8173218
112694756LOC112694756Uncharaterized LOC112694756831.1845093
9349RPL23Ribosomal protein L23826.6482544
7184HSP90B1Heat shock protein 90 β family member 1826.4506836
1337COX6A1Cytochrome c oxidase subunit 6A1820.6051025
1974EIF4A2Eukaryotic translation initiation factor 4A2800.7364502
6188RPS3Ribosomal protein S3796.1228638
6157RPL27ARibosomal protein l27a790.3303833
5757PTMAProthymosin α790.0863037
826CAPNS1Calpain small subunit 1783.6133423
5328PLAUPlasminogen activator, urokinase780.4100342
2023ENO1Enolase 1778.8522949
1509CTSDCathepsin D771.4251709
10476ATP5PDATP synthase peripheral stalk subunit d768.3088989
7534YWHAZTyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein ζ767.7701416
292SLC25A5Solute carrier family 25 member 5758.4469604
5216PFN1Profilin 1753.312439
1340COX6B1Cytochrome c oxidase subunit 6B1751.3442383
8407TAGLN2Transgelin 2741.7597046
689BTF3Basic transcription factor 3738.1211548
374AREGAmphiregulin735.1116333
10376TUBA1BTubulin α 1b732.8063965
6210RPS15ARibosomal protein s15a728.9209595
3909LAMA3Laminin subunit α 3723.6885986
7086TKTTransketolase713.4926147
5650KLK7Kallikrein related peptidase 7708.7366333
4323MMP14Matrix metallopeptidase 14702.4146118
4312MMP1Matrix metallopeptidase 1700.8983154
6229RPS24Ribosomal protein S24700.0944824
10653SPINT2Serine peptidase inhibitor, Kunitz type 2695.8338623
4831NME2NME/NM23 nucleoside diphosphate kinase 2694.8643799
10971YWHAQTyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein τ692.3873291
5478PPIAPeptidylprolyl isomerase A682.8765869
7980TFPI2Tissue factor pathway inhibitor 2679.0671997
6146RPL22Ribosomal protein L22678.4135132
3945LDHBLactate dehydrogenase B671.2799683
351APPAmyloid β precursor protein665.9901733
1508CTSBCathepsin B665.0159302
10209EIF1Eukaryotic translation initiation factor 1664.9918213
8673VAMP8Vesicle associated membrane protein 8659.6922607
7416VDAC1Voltage dependent anion channel 1659.1289063
4946OAZ1Ornithine decarboxylase antizyme 1656.2600098
6168RPL37ARibosomal protein l37a649.401123

[i] TPM, transcript per million.

Discussion

The isolation of primary tumor cells from patient samples is the first step for several genetic, biochemical and pharmacological experiments relevant to personalized cancer treatment (35). However, such studies are limited due to cell availability. The establishment of a cancer cell line is a traditional, but still powerful and informative method of studying human cancer. The present study reports the establishment of a MEC cell line with a CRTC1-MAML2 fusion gene.

Several studies have shown that the presence of the CRTC1/3-MAML2 fusion gene confers an improved prognosis, with improved disease-free survival and fewer distant metastasis in MEC (36,37). There are rare exceptions to this rule, including fusion-positive high-grade MEC with multiple additional genetic variations, such as mutations in CDKN2A, that have been associated with a poor prognosis (38).

The function of the CRTC1-MAML2 fusion gene has been intensively studied. Its transformation ability was identified using the RK3E cell line (39) and its importance for tumor state maintenance has also been demonstrated. Initially, it was hypothesized to cause tumor growth by the constitutive activation of Notch signaling via the MAML2 gene portion. Furthermore, the N terminus CRTC1 domain-mediated aberrant activation of cAMP/CREB signaling has also been identified as a cause of tumor formation (14,40). The interaction between AP-1 and MYC oncoprotein with CRTC1–MAML2 fusion proteins has been reported (41), suggesting that the CRTC1-MAML2 fusion gene regulates several different signaling pathways. AREG is a known cAMP/CREB-regulated gene, whose expression positively correlates with that of CRTC1-MAML2 in MEC (42). As AREG-EGFR signaling was identified as one of the CRTC1-MAML2 fusion gene targets, EGFR signaling could represent the mechanism of action by which the fusion gene promotes carcinogenesis.

These observations suggest an overall role for EGFR in the pathogenesis of MEC and the EGFR pathway could be a possible therapeutic target. As several drugs target this pathway, AREG–EGFR signaling was analyzed in the HCM-MEC010 cell line in the present study. The HCM-MEC010 cell line was found to express AREG and phosphorylate EGFR. Immunofluorescence analysis localized EGFR expression to the HCM-MEC010 cell membrane. These data suggest that the EGFR ligand, AREG, activated EGFR in an autocrine manner; therefore, antibodies that block AREG-EGFR binding or drugs that interfere with EGFR activation could be used for CRTC1-MAML2 fusion-positive MEC treatment. However, further analysis is required to identify suitable therapies.

MECs are composed of mucin-producing, epidermoid, and intermediate cells; however, RNA-Seq analysis of the HCM-MEC010 cell line detected little expression of MUC genes in the mucous cell marker family, indicating that mucin-producing cells and intermediate cells may have been removed during culture. MECs develop in excretory duct cells (43) and the mixture of three different cell types in MECs predicts their common origin. Duct and acinar cell differentiation are typically lineage-restricted; however, after irradiation, both duct and acinar cells can differentiate into different cell types (44). It is conceivable that established epidermoid-like cells are competent to differentiate into acinar cells, which is a predicted characteristic of injured duct stem cells. Further analysis will assist in the clarification into the origin of MECs. Cancer stem cells have been hypothesized to be involved in tumor formation (43). The results of the present study potentially indicate these cells may be of the same origin.

In conclusion, a MEC cell line, HCM-MEC010, with a CRTC1-MAML2 gene fusion was established. This cell line showed typical MEC characteristics, including AREG expression and EGFR activation; therefore, it could be used to assist in the identification of EGFR-targeted drugs for the treatment of CRTC1-MAML2 fusion gene-harboring MEC.

Supplementary Material

Results of STR test in cells. STR, short tandem repeat.
Result of STR test in blood. STR, short tandem repeat.
Resultsfrom short tandem repeat analysis.

Acknowledgements

The authors would like to thank Ms. Shinobu Osawa (Department of Oral and Maxillofacial Surgery, Hyogo College of Medicine, Nishinomiya, Japan) for preparation of the experiments and Ms. Takako Nanba (Department of Oral and Maxillofacial Surgery, Hyogo College of Medicine, Nishinomiya, Japan) for the management of the grants. The authors would also like to thank Nikki March and Sarah Williams for editing a draft version of the manuscript.

Funding

Funding: This study was supported by JSPS Grants-in-Aid for Scientific Research (grant nos. 16H11737 and 19H 10277), a Grant-in-Aid for Graduate Students, and a Hyogo College of Medicine and Hyogo Health Foundation Cancer Research Award.

Availability of data and materials

The datasets generated and/or analyzed during the current study are not publicly available due to a pending patent application, but are available from the corresponding author on reasonable request.

Authors' contributions

KN, SK, KaY, KT, HK and YN conceived and designed the present study. KN, SK, KaY, YF, KyY and YN performed the experiments. KN, SK, KoY and YN analyzed the data. KN, SK and YN wrote, reviewed, and revised the manuscript. All authors read and approved the final manuscript. KN and SK confirm the authenticity of all the raw data.

Ethics approval and consent to participate

The current study was approved by the Institutional Review Board of Hyogo College of Medicine (Hyogo, Japan) and was conducted in accordance with the Declaration of Helsinki. The patient provided written informed consent to participate.

Patient consent for publication

The patient provided written informed consent for the publication of their case study.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sentani K, Ogawa I, Ozawa K, Sadakane A, Utada M, Tsuya T, Kajihara H, Yonehara S, Takeshima Y and Yasui W: Characteristics of 5015 salivary gland neoplasms registered in the Hiroshima tumor tissue registry over a period of 39 years. J Clin Med. 8(566)2019.PubMed/NCBI View Article : Google Scholar

2 

Behboudi A, Enlund F, Winnes M, Andrén Y, Nordkvist A, Leivo I, Flaberg E, Szekely L, Mäkitie A, Grenman R, et al: Molecular classification of mucoepidermoid carcinomas-prognostic significance of the MECT1-MAML2 fusion oncogene. Genes Chromosomes Cancer. 45:470–481. 2006.PubMed/NCBI View Article : Google Scholar

3 

Ettl T, Schwarz-Furlan S, Gosau M and Reichert TE: Salivary gland carcinomas. Oral Maxillofac Surg. 16:267–283. 2012.PubMed/NCBI View Article : Google Scholar

4 

Katabi N, Ghossein R, Ali S, Dogan S, Klimstra D and Ganly I: Prognostic features in mucoepidermoid carcinoma of major salivary glands with emphasis on tumour histologic grading. Histopathology. 65:793–804. 2014.PubMed/NCBI View Article : Google Scholar

5 

El-Naggar AK, Lovell M, Killary AM, Clayman GL and Batsakis JG: A mucoepidermoid carcinoma of minor salivary gland with t(11;19)(q21;p13.1) as the only karyotypic abnormality. Cancer Genet Cytogenet. 87:29–33. 1996.PubMed/NCBI View Article : Google Scholar

6 

Saade RE, Bell D, Garcia J, Roberts D and Weber R: Role of CRTC1/MAML2 translocation in the prognosis and clinical outcomes of mucoepidermoid carcinoma. JAMA Otolaryngol Head Neck Surg. 142:234–240. 2016.PubMed/NCBI View Article : Google Scholar

7 

Bell D and El-Naggar AK: Molecular heterogeneity in mucoepidermoid carcinoma: Conceptual and practical implications. Head Neck Pathol. 7:23–27. 2013.PubMed/NCBI View Article : Google Scholar

8 

Jee KJ, Persson M, Heikinheimo K, Passador-Santos F, Aro K, Knuutila S, Odell EW, Mäkitie A, Sundelin K, Stenman G and Leivo I: Genomic profiles and CRTC1-MAML2 fusion distinguish different subtypes of mucoepidermoid carcinoma. Mod Pathol. 26:213–222. 2013.PubMed/NCBI View Article : Google Scholar

9 

O'Neill ID: t(11;19) translocation and CRTC1-MAML2 fusion oncogene in mucoepidermoid carcinoma. Oral Oncol. 45:2–9. 2009.PubMed/NCBI View Article : Google Scholar

10 

Seethala RR, Dacic S, Cieply K, Kelly LM and Nikiforova MN: A reappraisal of the MECT1/MAML2 translocation in salivary mucoepidermoid carcinomas. Am J Surg Pathol. 34:1106–1121. 2010.PubMed/NCBI View Article : Google Scholar

11 

Tonon G, Modi S, Wu L, Kubo A, Coxon AB, Komiya T, O'Neil K, Stover K, El-Naggar A, Griffin JD, et al: t(11;19)(q21;p13) translocation in mucoepidermoid carcinoma creates a novel fusion product that disrupts a Notch signaling pathway. Nat Genet. 33:208–213. 2003.PubMed/NCBI View Article : Google Scholar

12 

Enlund F, Behboudi A, Andrén Y, Oberg C, Lendahl U, Mark J and Stenman G: Altered Notch signaling resulting from expression of a WAMTP1-MAML2 gene fusion in mucoepidermoid carcinomas and benign Warthin's tumors. Exp Cell Res. 292:21–28. 2004.PubMed/NCBI View Article : Google Scholar

13 

Wu L, Liu J, Gao P, Nakamura M, Cao Y, Shen H and Griffin JD: Transforming activity of MECT1-MAML2 fusion oncoprotein is mediated by constitutive CREB activation. EMBO J. 24:2391–2402. 2005.PubMed/NCBI View Article : Google Scholar

14 

Coxon A, Rozenblum E, Park YS, Joshi N, Tsurutani J, Dennis PA, Kisch IR and Kaye FJ: Mect1-Maml2 fusion oncogene linked to the aberrant activation of cyclic AMP/CREB regulated genes. Cancer Res. 65:7137–7144. 2005.PubMed/NCBI View Article : Google Scholar

15 

Fehr A, Röser K, Heidorn K, Hallas C, Löning T and Bullerdiek J: A new type of MAML2 fusion in mucoepidermoid carcinoma. Genes Chromosomes Cancer. 47:203–206. 2008.PubMed/NCBI View Article : Google Scholar

16 

Okabe M, Miyabe S, Nagatsuka H, Terada A, Hanai N, Yokoi M, Shimozato K, Eimoto T, Nakamura S, Nagai N, et al: MECT1-MAML2 fusion transcript defines a favorable subset of mucoepidermoid carcinoma. Clin Cancer Res. 12:3902–3907. 2006.PubMed/NCBI View Article : Google Scholar

17 

Nakayama T, Miyabe S, Okabe M, Sakuma H, Ijichi K, Hasegawa Y, Nagatsuka H, Shimozato K and Inagaki H: Clinicopathological significance of the CRTC3-MAML2 fusion transcript in mucoepidermoid carcinoma. Mod Pathol. 22:1575–1581. 2009.PubMed/NCBI View Article : Google Scholar

18 

Chen Z, Chen J, Gu Y, Hu C, Li JL, Lin S, Shen H, Cao C, Gao R, Li J, et al: Aberrantly activated AREG-EGFR signaling is required for the growth and survival of CRTC1-MAML2 fusion-positive mucoepidermoid carcinoma cells. Oncogene. 33:3869–3877. 2014.PubMed/NCBI View Article : Google Scholar

19 

Dahse R, Driemel O, Schwartz S, Dahse J, Kromeyer-Hauschild K, Berndt A and Kosmehl H: Epidermal growth factor receptor kinase domain mutations are rare in salivary gland carcinomas. Br J Cancer. 100:623–625. 2009.PubMed/NCBI View Article : Google Scholar

20 

Nakano T, Yamamoto H, Hashimoto K, Tamiya S, Shiratsuchi H, Nakashima T, Nishiyama K, Higaki Y, Komune S and Oda Y: HER2 and EGFR gene copy number alterations are predominant in high-grade salivary mucoepidermoid carcinoma irrespective of MAML2 fusion status. Histopathology. 63:378–392. 2013.PubMed/NCBI View Article : Google Scholar

21 

Warner KA, Adams A, Bernardi L, Nor C, Finkel KA, Zhang Z, McLean SA, Helman J, Wolf GT, Divi V, et al: Characterization of tumorigenic cell lines from the recurrence and lymph node metastasis of a human salivary mucoepidermoid carcinoma. Oral Onco. 49:1059–1066. 2013.PubMed/NCBI View Article : Google Scholar

22 

Liu X, Ory V, Chapman S, Yuan H, Albanese C, Kallakury B, Timofeeva OA, Nealon C, Dakic A, Simic V, et al: ROCK inhibitor and feeder cells induce the conditional reprogramming of epithelial cells. Am J Pathol. 180:599–607. 2012.PubMed/NCBI View Article : Google Scholar

23 

Dulbecco R and Vogt M: Plaque formation and isolation of pure lines with poliomyelitis viruses. J Exp Med. 99:167–182. 1954.PubMed/NCBI View Article : Google Scholar

24 

Noguchi K, Wakai K, Kiyono T, Kawabe M, Yoshikawa K, Hashimoto-Tamaoki T, Kishimoto H and Nakano Y: Molecular analysis of keratocystic odontogenic tumor cell lines derived from sporadic and basal cell nevus syndrome patients. Int J Oncol. 51:1731–1738. 2017.PubMed/NCBI View Article : Google Scholar

25 

Hiromoto T, Noguchi K, Yamamura M, Zushi Y, Segawa E, Takaoka K, Moridera K, Kishimoto H and Urade M: Up-regulation of neutrophil gelatinase-associated lipocalin in oral squamous cell carcinoma: Relation to cell differentiation. Oncol Rep. 26:1415–1421. 2011.PubMed/NCBI View Article : Google Scholar

26 

Gupta R, Kalita P, Patil O and Mohanty S: An investigation of folic acid-protein association sites and the effect of this association on folic acid self-assembly. J Mol Model. 21(308)2015.PubMed/NCBI View Article : Google Scholar

27 

Rio DC, Ares M Jr, Hannon GJ and Nilsen TW: Purification of RNA using TRIzol (TRI reagent). Cold Spring Harb Protoc. 2010(pdb.prot5439)2010.PubMed/NCBI View Article : Google Scholar

28 

Prophet EB, Mills B, Arrington JB and Sobin LH: Laboratory methods in histotechnology (Armed Forces Institute of Phatology). American Registry of Pathology, Washington, DC, 1992.

29 

Seethala RR: An update on grading of salivary gland carcinomas. Head Neck Pathol. 3:69–77. 2009.PubMed/NCBI View Article : Google Scholar

30 

Ohnishi Y, Minamino Y, Kakudo K and Nozaki M: Resistance of oral squamous cell carcinoma cells to cetuximab is associated with EGFR insensitivity and enhanced stem cell-like potency. Oncol Rep. 32:780–786. 2014.PubMed/NCBI View Article : Google Scholar

31 

Meng C, Wang S and Wang X, Lv J, Zeng W, Chang R, Li Q and Wang X: Amphiregulin inhibits TNF-α-induced alveolar epithelial cell death through EGFR signaling pathway. Biomed Pharmacother. 125(109995)2020.PubMed/NCBI View Article : Google Scholar

32 

Telesco SE, Vadigepalli R and Radhakrishnan R: Molecular modeling of ErbB4/HER4 kinase in the context of the HER4 signaling network helps rationalize the effects of clinically identified HER4 somatic mutations on the cell phenotype. Biotechnol J. 8:1452–1464. 2013.PubMed/NCBI View Article : Google Scholar

33 

Li X, Huang Q, Wang S, Huang Z, Yu F and Lin J: HER4 promotes the growth and metastasis of osteosarcoma via the PI3K/AKT pathway. Acta Biochim Biophys Sin (Shanghai). 52:345–362. 2020.PubMed/NCBI View Article : Google Scholar

34 

Luna MA: Salivary mucoepidermoid carcinoma: Revisited. Adv Anat Pathol. 13:293–307. 2006.PubMed/NCBI View Article : Google Scholar

35 

Mitra A, Mishra L and Li S: Technologies for deriving primary tumor cells for use in personalized cancer therapy. Trends Biotechnol. 31:347–354. 2013.PubMed/NCBI View Article : Google Scholar

36 

Okumura Y, Miyabe S, Nakayama T, Fujiyoshi Y, Hattori H, Shimozato K and Inagaki H: Impact of CRTC1/3-MAML2 fusions on histological classification and prognosis of mucoepidermoid carcinoma. Histopathology. 59:90–97. 2011.PubMed/NCBI View Article : Google Scholar

37 

Tirado Y, Williams MD, Hanna EY, Kaye FJ, Batsakis JG and El-Naggar AK: CRTC1/MAML2 fusion transcript in high grade mucoepidermoid carcinomas of salivary and thyroid glands and Warthin's tumors: Implications for histogenesis and biologic behavior. Genes Chromosomes Cancer. 46:708–715. 2007.PubMed/NCBI View Article : Google Scholar

38 

Anzick SL, Chen WD, Park Y, Meltzer P, Bell D, El-Naggar AK and Kaye FJ: Unfavorable prognosis of CRTC1-MAML2 positive mucoepidermoid tumors with CDKN2A deletions. Genes Chromosomes Cancer. 49:59–69. 2010.PubMed/NCBI View Article : Google Scholar

39 

Komiya T, Park Y, Modi S, Coxon AB, Oh H and Kaye FJ: Sustained expression of Mect1-Maml2 is essential for tumor cell growth in salivary gland cancers carrying the t(11;19) translocation. Oncogene. 25:6128–6132. 2006.PubMed/NCBI View Article : Google Scholar

40 

Wu J, Wang N, Yang Y, Jiang G, Zhan H and Li F: LINC01152 upregulates MAML2 expression to modulate the progression of glioblastoma multiforme via Notch signaling pathway. Cell Death Dis. 12(115)2021.PubMed/NCBI View Article : Google Scholar

41 

Amelio AL, Fallahi M, Schaub FX, Zhang M, Lawani MB, Alperstein AS, Southern MR, Young BM, Wu L, Zajac-Kaye M, et al: CRTC1/MAML2 gain-of-function interactions with MYC create a gene signature predictive of cancers with CREB-MYC involvement. Proc Natl Acad Sci USA. 111:3260–3268. 2014.PubMed/NCBI View Article : Google Scholar

42 

Shinomiya H, Ito Y, Kubo M, Yonezawa K, Otsuki N, Iwae S, Inagaki H and Nibu KI: Expression of amphiregulin in mucoepidermoid carcinoma of the major salivary glands: A molecular and clinicopathological study. Hum Pathol. 57:37–44. 2016.PubMed/NCBI View Article : Google Scholar

43 

Porcheri C and Mitsiadis TA: Physiology, pathology and regeneration of salivary glands. Cells. 8(976)2019.PubMed/NCBI View Article : Google Scholar

44 

Weng PL, Aure MH, Maruyama T and Ovitt CE: Limited regeneration of adult salivary glands after severe injury involves cellular plasticity. Cell Rep. 24:1464–1470.e3. 2018.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

March-2022
Volume 16 Issue 3

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Noguchi K, Kanda S, Yoshida K, Funaoka Y, Yamanegi K, Yoshikawa K, Takaoka K, Kishimoto H and Nakano Y: Establishment of a patient‑derived mucoepidermoid carcinoma cell line with the <em>CRTC1‑MAML2</em> fusion gene. Mol Clin Oncol 16: 75, 2022
APA
Noguchi, K., Kanda, S., Yoshida, K., Funaoka, Y., Yamanegi, K., Yoshikawa, K. ... Nakano, Y. (2022). Establishment of a patient‑derived mucoepidermoid carcinoma cell line with the <em>CRTC1‑MAML2</em> fusion gene. Molecular and Clinical Oncology, 16, 75. https://doi.org/10.3892/mco.2022.2508
MLA
Noguchi, K., Kanda, S., Yoshida, K., Funaoka, Y., Yamanegi, K., Yoshikawa, K., Takaoka, K., Kishimoto, H., Nakano, Y."Establishment of a patient‑derived mucoepidermoid carcinoma cell line with the <em>CRTC1‑MAML2</em> fusion gene". Molecular and Clinical Oncology 16.3 (2022): 75.
Chicago
Noguchi, K., Kanda, S., Yoshida, K., Funaoka, Y., Yamanegi, K., Yoshikawa, K., Takaoka, K., Kishimoto, H., Nakano, Y."Establishment of a patient‑derived mucoepidermoid carcinoma cell line with the <em>CRTC1‑MAML2</em> fusion gene". Molecular and Clinical Oncology 16, no. 3 (2022): 75. https://doi.org/10.3892/mco.2022.2508