Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2023, Vol. 17 Issue (2) : 263-274    https://doi.org/10.1007/s11684-022-0935-0
RESEARCH ARTICLE
NETO2 promotes melanoma progression via activation of the Ca2+/CaMKII signaling pathway
Susi Zhu1,2,3,4,5, Xu Zhang1,2,3,4, Yeye Guo1,2,3,4, Ling Tang1,2,3,4,5, Zhe Zhou1,2,3,4, Xiang Chen1,2,3,4(), Cong Peng1,2,3,4()
1. Department of Dermatology, Xiangya Hospital, Central South University, Changsha 41000, China
2. Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha 41000, China
3. Hunan Engineering Research Center of Skin Health and Disease, Changsha 41000, China
4. National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 41000, China
5. Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 41000, China
 Download: PDF(4221 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Melanoma is the most aggressive cutaneous tumor. Neuropilin and tolloid-like 2 (NETO2) is closely related to tumorigenesis. However, the functional significance of NETO2 in melanoma progression remains unclear. Herein, we found that NETO2 expression was augmented in melanoma clinical tissues and associated with poor prognosis in melanoma patients. Disrupting NETO2 expression markedly inhibited melanoma proliferation, malignant growth, migration, and invasion by downregulating the levels of calcium ions (Ca2+) and the expression of key genes involved in the calcium signaling pathway. By contrast, NETO2 overexpression had the opposite effects. Importantly, pharmacological inhibition of CaMKII/CREB activity with the CaMKII inhibitor KN93 suppressed NETO2-induced proliferation and melanoma metastasis. Overall, this study uncovered the crucial role of NETO2-mediated regulation in melanoma progression, indicating that targeting NETO2 may effectively improve melanoma treatment.

Keywords melanoma      neuropilin and tolloid-like 2      Ca2+/CaMKII signaling pathway     
Corresponding Author(s): Xiang Chen,Cong Peng   
Just Accepted Date: 01 December 2022   Online First Date: 07 February 2023    Issue Date: 26 May 2023
 Cite this article:   
Susi Zhu,Xu Zhang,Yeye Guo, et al. NETO2 promotes melanoma progression via activation of the Ca2+/CaMKII signaling pathway[J]. Front. Med., 2023, 17(2): 263-274.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-022-0935-0
https://academic.hep.com.cn/fmd/EN/Y2023/V17/I2/263
Fig.1  NETO2 overexpression in melanoma tissues correlated with melanoma prognosis. (A) Relative NETO2 mRNA expression in normal skin, nevi, and melanoma tissues; data were obtained from the GEO datasets GSE46517 and GSE3189. (B) Relative NETO2 mRNA expression in normal and melanoma tissues; data were obtained from GEPIA (TCGA). (C) IHC analyses of NETO2 protein levels in a human melanoma tissue microarray (left). Scale bar, 50 μm. The IHC score for NETO2 in benign and malignant and metastasic melanoma tissues (right). (D) The Kaplan–Meier overall and disease-free survival curves for melanoma patients correlated with NETO2 expression; data were obtained from GEPIA. The horizontal lines in all dot plots represent the means ± SEMs. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig.2  NETO2 promoted melanoma cell proliferation in vitro. (A) NETO2 expression in PIG1 cells and melanoma cell lines. GAPDH was used as a loading control. (B) SK-Mel-5 and SK-Mel-28 cells that had been infected with shMOCK or shNETO2 lentivirus. (C) Cell proliferation was assessed after NETO2 knockdown in SK-Mel-5 and SK-Mel-28 cells with CCK-8 assays. (D) Colony-formation capability was assessed in SK-Mel-5 and SK-Mel-28 cells with NETO2 knockdown; the number of clones of SK-Mel-5 and SK-Mel-28 cells was quantified. (E) WM35 cells were infected with control or NETO2-expressing lentivirus. (F) CCK-8 assays were conducted in NETO2-overexpressing WM35 cells. (G) Colony-formation capability was assessed in NETO2-overexpressing WM35 cells; the number of WM35 cell colonies was quantified. The data (means ± SEMs) were obtained from triplicate experiments. **P < 0.01, ***P < 0.001.
Fig.3  NETO2 promoted the growth of melanoma tumors in vivo. (A,B) Representative images of nude mice injected with differentially treated SK-Mel-28 cells (infected with shMOCK, shNETO2#2, or shNETO2#3) and their corresponding tumors. (C) Tumor size was analyzed quantitatively. (D,E) Immunohistochemical staining of Ki-67, P-CaMKII, and P-CREB in xenograft tumors; scale bar, 50 µm. The data (means ± SEMs) were obtained from triplicate experiments. **P < 0.01.
Fig.4  NETO2 promotes melanoma cell migration and invasion by inducing MMPs. (A) Wound healing assay in SK-Mel-5 and SK-Mel-28 cells expressing NETO2 and control shRNA, respectively; quantified data from wound healing assays at 0, 24, and 48 h are shown. (B,C) Transwell migration and invasion assays with NETO2 knockdown melanoma cells. (D) Western blot analysis of MMP2 and MMP9 levels in melanoma cells infected with shNETO2 lentivirus. (E) Wound healing assay in WM35 cells expressing NETO2 (pLVX-NETO2) or the control (pLVX-IRES). Quantitative analysis of the wounded areas of WM35 cells at 0, 12, and 24 h. (F) Migration (12 h) and invasion (24 h) assays in WM35 cells overexpressing NETO2. (G) Western blot analysis of MMP2 and MMP9 levels in melanoma cells infected with pLVX-NETO2 lentivirus; band intensities were quantified using ImageLab. The data (means ± SEMs) were obtained from triplicate experiments. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig.5  NETO2 activates the Ca2+/CaMKII pathways. (A,B) Flow cytometry detection of calcium ions in SK-Mel-5 and SK-Mel-28 cells expressing NETO2 or control shRNA, respectively. (C) Western blot analysis of CaMKII, P-CaMKII, CREB, P-CREB, and GAPDH in SK-Mel-5 and SK-Mel-28 cells infected with shNETO2 lentivirus; band intensities were quantified using ImageLab. (D,E) Flow cytometry detection of calcium ions in WM35 cells infected with NETO2 lentivirus. (F) Western blot analysis of CaMKII, P-CaMKII, CREB, P-CREB, and GAPDH in WM35 cells infected with NETO2-expressing lentivirus; band intensities were quantified using ImageLab. The data (mean ± SEM) were obtained from triplicate experiments. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig.6  Inhibition of CaMKII antagonized NETO2-mediated proliferation, migration, and invasion of melanoma cells. (A) Western blot analysis of NETO2, P-CaMKII, P-CREB, and MMP2 levels in NETO2-overexpressing melanoma cells with or without 10 μmol/L KN93 treatment; band intensities were quantified using ImageLab. (B) CCK-8 assays were conducted to analyze the effects of KN93 on melanoma cell proliferation. (C) Colony-formation capability was assessed in WM35 cells with or without 10μmol/L KN93 treatment. (D,E) Wound healing and Transwell assays were used to analyze melanoma cell migration and invasion, respectively. The data (mean ± SEM) were obtained from triplicate experiments. *P < 0.05, **P < 0.01, ***P < 0.001.
Fig.7  Schematic diagram depicting the NETO2-mediated induction of the CaMKII/CREB pathway.
1 MJ Berridge, MD Bootman, P Lipp. Calcium—a life and death signal. Nature 1998; 395(6703): 645–648
https://doi.org/10.1038/27094 pmid: 9790183
2 MJ Berridge, MD Bootman, HL Roderick. Calcium signalling: dynamics, homeostasis and remodelling. Nat Rev Mol Cell Biol 2003; 4(7): 517–529
https://doi.org/10.1038/nrm1155 pmid: 12838335
3 TA Stewart, KT Yapa, GR Monteith. Altered calcium signaling in cancer cells. Biochim Biophys Acta 2015; 1848(10 Pt B): 2502–2511
https://doi.org/10.1016/j.bbamem.2014.08.016 pmid: 25150047
4 T Long, J Su, W Tang, Z Luo, S Liu, Z Liu, H Zhou, M Qi, W Zeng, J Zhang, X Chen. A novel interaction between calcium-modulating cyclophilin ligand and basigin regulates calcium signaling and matrix metalloproteinase activities in human melanoma cells. Cancer Lett 2013; 339(1): 93–101
https://doi.org/10.1016/j.canlet.2013.07.019 pmid: 23879967
5 Q Lin, K Balasubramanian, D Fan, SJ Kim, L Guo, H Wang, M Bar-Eli, KD Aldape, IJ Fidler. Reactive astrocytes protect melanoma cells from chemotherapy by sequestering intracellular calcium through gap junction communication channels. Neoplasia 2010; 12(9): 748–754
https://doi.org/10.1593/neo.10602 pmid: 20824051
6 AJ Miller, MC Jr Mihm. Melanoma. N Engl J Med 2006; 355(1): 51–65
https://doi.org/10.1056/NEJMra052166 pmid: 16822996
7 JJ Luke, KT Flaherty, A Ribas, GV Long. Targeted agents and immunotherapies: optimizing outcomes in melanoma. Nat Rev Clin Oncol 2017; 14(8): 463–482
https://doi.org/10.1038/nrclinonc.2017.43 pmid: 28374786
8 S Tomita, PE Castillo. Neto1 and Neto2: auxiliary subunits that determine key properties of native kainate receptors. J Physiol 2012; 590(10): 2217–2223
https://doi.org/10.1113/jphysiol.2011.221101 pmid: 22431337
9 BA Copits, GT Swanson. Dancing partners at the synapse: auxiliary subunits that shape kainate receptor function. Nat Rev Neurosci 2012; 13(10): 675–686
https://doi.org/10.1038/nrn3335 pmid: 22948074
10 W Zhang, F St-Gelais, CP Grabner, JC Trinidad, A Sumioka, M Morimoto-Tomita, KS Kim, C Straub, AL Burlingame, JR Howe, S Tomita. A transmembrane accessory subunit that modulates kainate-type glutamate receptors. Neuron 2009; 61(3): 385–396
https://doi.org/10.1016/j.neuron.2008.12.014 pmid: 19217376
11 M Mennesson, E Orav, A Gigliotta, N Kulesskaya, S Saarnio, A Kirjavainen, S Kesaf, F Winkel, Pou M Llach, J Umemori, V Voikar, V Risbrough, J Partanen, E Castrén, SE Lauri, I Hovatta. Kainate receptor auxiliary subunit NETO2-related cued fear conditioning impairments associate with defects in amygdala development and excitability. eNeuro 2020; 7(4): ENEURO.0541–19.2020
https://doi.org/10.1523/ENEURO.0541-19.2020 pmid: 32788298
12 D Sargin. Heightened fear in the absence of the kainate receptor auxiliary subunit NETO2: implications for PTSD. Neuropsychopharmacology 2019; 44(11): 1841–1842
https://doi.org/10.1038/s41386-019-0376-x pmid: 30976121
13 JC Xu, TY Chen, LT Liao, T Chen, QL Li, JX Xu, JW Hu, PH Zhou, YQ Zhang. NETO2 promotes esophageal cancer progression by inducing proliferation and metastasis via PI3K/AKT and ERK pathway. Int J Biol Sci 2021; 17(1): 259–270
https://doi.org/10.7150/ijbs.53795 pmid: 33390848
14 X Wang, Z Bian, C Hou, M Li, W Jiang, L Zhu. Neuropilin and tolloid-like 2 regulates the progression of osteosarcoma. Gene 2021; 768: 145292
https://doi.org/10.1016/j.gene.2020.145292 pmid: 33157203
15 JY Liu, L Jiang, T He, JJ Liu, JY Fan, XH Xu, B Tang, Y Shi, YL Zhao, F Qian, Y Wang, YH Cui, PW Yu. NETO2 promotes invasion and metastasis of gastric cancer cells via activation of PI3K/Akt/NF-κB/Snail axis and predicts outcome of the patients. Cell Death Dis 2019; 10(3): 162
https://doi.org/10.1038/s41419-019-1388-5 pmid: 30770791
16 Y Li, Y Zhang, J Liu. NETO2 promotes pancreatic cancer cell proliferation, invasion and migration via activation of the STAT3 signaling pathway. Cancer Manag Res 2019; 11: 5147–5156
https://doi.org/10.2147/CMAR.S204260 pmid: 31239769
17 H Ma, RD Groth, SM Cohen, JF Emery, B Li, E Hoedt, G Zhang, TA Neubert, RW Tsien. γCaMKII shuttles Ca2+/CaM to the nucleus to trigger CREB phosphorylation and gene expression. Cell 2014; 159(2): 281–294
https://doi.org/10.1016/j.cell.2014.09.019 pmid: 25303525
18 C Xiao, BF Yang, JH Song, H Schulman, L Li, C Hao. Inhibition of CaMKII-mediated c-FLIP expression sensitizes malignant melanoma cells to TRAIL-induced apoptosis. Exp Cell Res 2005; 304(1): 244–255
https://doi.org/10.1016/j.yexcr.2004.11.002 pmid: 15707589
19 W Chen, Y Chen, J Su, J Kang, Y Ding, W Ai, J Zhang, H Luo, P An. CaMKII mediates TGFβ1-induced fibroblasts activation and its cross talk with colon cancer cells. Dig Dis Sci 2022; 67(1): 134–145
https://doi.org/10.1007/s10620-021-06847-0 pmid: 33528688
20 SP Chen, J Sun, YQ Zhou, F Cao, C Braun, F Luo, DW Ye, YK Tian. Sinomenine attenuates cancer-induced bone pain via suppressing microglial JAK2/STAT3 and neuronal CAMKII/CREB cascades in rat models. Mol Pain 2018; 14: 1744806918793232
https://doi.org/10.1177/1744806918793232 pmid: 30027795
21 WC Hsu, HN Le, YJ Lin, MC Chen, TF Wang, CC Li, WW Kuo, B Mahalakshmi, CH Singh, MC Chen, CY Huang. Calmodulin/CaMKII-γ mediates prosurvival capability in apicidin-persistent hepatocellular carcinoma cells via ERK1/2/CREB/c-fos signaling pathway. J Cell Biochem 2021; 122(6): 612–625
https://doi.org/10.1002/jcb.29892 pmid: 33459431
22 B Li, E Rex, H Wang, Y Qian, AM Ogden, D Bleakman, KW Johnson. Pharmacological modulation of GluK1 and GluK2 by NETO1, NETO2, and PSD95. Assay Drug Dev Technol 2016; 14(2): 131–143
https://doi.org/10.1089/adt.2015.689 pmid: 26991362
23 C Lian, S Cao, W Zeng, Y Li, J Su, J Li, S Zhao, L Wu, J Tao, J Zhou, X Chen, C Peng. RJT-101, a novel camptothecin derivative, is highly effective in the treatment of melanoma through DNA damage by targeting topoisomerase 1. Biochem Pharmacol 2020; 171: 113716
https://doi.org/10.1016/j.bcp.2019.113716 pmid: 31751535
24 X Zhang, Z Huang, Y Guo, T Xiao, L Tang, S Zhao, L Wu, J Su, W Zeng, H Huang, Z Li, J Tao, J Zhou, X Chen, C Peng. The phosphorylation of CD147 by Fyn plays a critical role for melanoma cells growth and metastasis. Oncogene 2020; 39(21): 4183–4197
https://doi.org/10.1038/s41388-020-1287-3 pmid: 32291412
25 E Guney, AP Arruda, G Parlakgul, E Cagampan, N Min, GY Lee, L Greene, E Tsaousidou, K Inouye, MS Han, RJ Davis, GS Hotamisligil. Aberrant Ca2+ signaling by IP3Rs in adipocytes links inflammation to metabolic dysregulation in obesity. Sci Signal 2021; 14(713): eabf2059
https://doi.org/10.1126/scisignal.abf2059 pmid: 34905386
26 F Gu, A Krüger, HG Roggenkamp, R Alpers, D Lodygin, V Jaquet, F Möckl, C LC Hernandez, K Winterberg, A Bauche, A Rosche, H Grasberger, JY Kao, D Schetelig, R Werner, K Schröder, M Carty, AG Bowie, S Huber, C Meier, HW Mittrücker, J Heeren, KH Krause, A Flügel, BP Diercks, AH Guse. Dual NADPH oxidases DUOX1 and DUOX2 synthesize NAADP and are necessary for Ca2+ signaling during T cell activation. Sci Signal 2021; 14(709): eabe3800
https://doi.org/10.1126/scisignal.abe3800 pmid: 34784249
27 Y Tan, D Mui, S Toan, P Zhu, R Li, H Zhou. SERCA overexpression improves mitochondrial quality control and attenuates cardiac microvascular ischemia-reperfusion injury. Mol Ther Nucleic Acids 2020; 22: 696–707
https://doi.org/10.1016/j.omtn.2020.09.013 pmid: 33230467
28 GR Monteith, FM Davis, SJ Roberts-Thomson. Calcium channels and pumps in cancer: changes and consequences. J Biol Chem 2012; 287(38): 31666–31673
https://doi.org/10.1074/jbc.R112.343061 pmid: 22822055
29 J Mookerjee-Basu, R Hooper, S Gross, B Schultz, CK Go, E Samakai, J Ladner, E Nicolas, Y Tian, B Zhou, MR Zaidi, W Tourtellotte, S He, Y Zhang, DJ Kappes, J Soboloff. Suppression of Ca2+ signals by EGR4 controls Th1 differentiation and anti-cancer immunity in vivo. EMBO Rep 2020; 21(5): e48904
https://doi.org/10.15252/embr.201948904 pmid: 32212315
30 Q Jia, S Hu, D Jiao, X Li, S Qi, R Fan. Synaptotagmin-4 promotes dendrite extension and melanogenesis in alpaca melanocytes by regulating Ca2+ influx via TRPM1 channels. Cell Biochem Funct 2020; 38(3): 275–282
https://doi.org/10.1002/cbf.3465 pmid: 31743468
31 A Schwab, T Loeck, K Najder-Nalepa. STIM2: redox-sensor and effector of the (tumor) microenvironment. Cell Calcium 2021; 94: 102335
https://doi.org/10.1016/j.ceca.2020.102335 pmid: 33387846
32 A Kondratskyi, M Yassine, K Kondratska, R Skryma, C Slomianny, N Prevarskaya. Calcium-permeable ion channels in control of autophagy and cancer. Front Physiol 2013; 4: 272
https://doi.org/10.3389/fphys.2013.00272 pmid: 24106480
33 YJ Li, GF Duan, JH Sun, D Wu, C Ye, YY Zang, GQ Chen, YY Shi, J Wang, W Zhang, YS Shi. Neto proteins regulate gating of the kainate-type glutamate receptor GluK2 through two binding sites. J Biol Chem 2019; 294(47): 17889–17902
https://doi.org/10.1074/jbc.RA119.008631 pmid: 31628192
34 R Falcón-Moya, P Losada-Ruiz, TS Sihra, A Rodríguez-Moreno. Cerebellar kainate receptor-mediated facilitation of glutamate release requires Ca2+-calmodulin and PKA. Front Mol Neurosci 2018; 11: 195
https://doi.org/10.3389/fnmol.2018.00195 pmid: 29928192
35 MR Hansen, J Bok, AK Devaiah, XM Zha, SH Green. Ca2+/calmodulin-dependent protein kinases II and IV both promote survival but differ in their effects on axon growth in spiral ganglion neurons. J Neurosci Res 2003; 72(2): 169–184
https://doi.org/10.1002/jnr.10551 pmid: 12671991
36 K Pan, Y Xie. LncRNA FOXC2-AS1 enhances FOXC2 mRNA stability to promote colorectal cancer progression via activation of Ca2+-FAK signal pathway. Cell Death Dis 2020; 11(6): 434
https://doi.org/10.1038/s41419-020-2633-7 pmid: 32513911
37 C Shi, Y Cai, Y Li, Y Li, N Hu, S Ma, S Hu, P Zhu, W Wang, H Zhou. Yap promotes hepatocellular carcinoma metastasis and mobilization via governing cofilin/F-actin/lamellipodium axis by regulation of JNK/Bnip3/SERCA/CaMKII pathways. Redox Biol 2018; 14: 59–71
https://doi.org/10.1016/j.redox.2017.08.013 pmid: 28869833
38 RM Lomash, N Sheng, Y Li, RA Nicoll, KW Roche. Phosphorylation of the kainate receptor (KAR) auxiliary subunit Neto2 at serine 409 regulates synaptic targeting of the KAR subunit GluK1. J Biol Chem 2017; 292(37): 15369–15377
https://doi.org/10.1074/jbc.M117.787903 pmid: 28717010
39 AY Wen, KM Sakamoto, LS Miller. The role of the transcription factor CREB in immune function. J Immunol 2010; 185(11): 6413–6419
https://doi.org/10.4049/jimmunol.1001829 pmid: 21084670
40 YQ Zhou, DQ Liu, SP Chen, J Sun, XR Zhou, F Luo, YK Tian, DW Ye. Cellular and molecular mechanisms of calcium/calmodulin-dependent protein kinase II in chronic pain. J Pharmacol Exp Ther 2017; 363(2): 176–183
https://doi.org/10.1124/jpet.117.243048 pmid: 28855373
41 Z Jie, Z Xie, X Zhao, X Sun, H Yu, X Pan, S Shen, A Qin, X Fang, S Fan. Glabridin inhibits osteosarcoma migration and invasion via blocking the p38- and JNK-mediated CREB-AP1 complexes formation. J Cell Physiol 2019; 234(4): 4167–4178
https://doi.org/10.1002/jcp.27171 pmid: 30146723
42 H Li, Z Qiu, F Li, C Wang. The relationship between MMP-2 and MMP-9 expression levels with breast cancer incidence and prognosis. Oncol Lett 2017; 14(5): 5865–5870
https://doi.org/10.3892/ol.2017.6924 pmid: 29113219
43 T Brabletz, R Kalluri, MA Nieto, RA Weinberg. EMT in cancer. Nat Rev Cancer 2018; 18(2): 128–134
https://doi.org/10.1038/nrc.2017.118 pmid: 29326430
[1] FMD-22012-OF-PC_suppl_1 Download
[1] Feifei Bao, Mengjie Liu, Wenhua Gai, Yuwei Hua, Jing Li, Chao Han, Ziyu Zai, Jiahuang Li, Zichun Hua. Bacteria-mediated tumor-targeted delivery of tumstatin (54-132) significantly suppresses tumor growth in mouse model by inhibiting angiogenesis and promoting apoptosis[J]. Front. Med., 2022, 16(6): 873-882.
[2] Yan Zhang, Baoyuan Zhang, Yongyun Li, Yuting Dai, Jiaoyang Li, Donghe Li, Zhizhou Xia, Jianming Zhang, Ping Liu, Ming Chen, Bo Jiao, Ruibao Ren. Palmitoylation of GNAQ/11 is critical for tumor cell proliferation and survival in GNAQ/11-mutant uveal melanoma[J]. Front. Med., 2022, 16(5): 784-798.
[3] Jianwen REN, Zhenhui PENG, Birong GUO, Min PAN. Celecoxib in combination with retinoid CD437 inhibits melanoma A375 cell in vitro[J]. Front Med Chin, 2009, 3(1): 108-112.
[4] HUANG Hongying, LIU Guangchao, QI Yijun, DU Yaowu, CHEN Jugao, MA Yuanfang. Inhibitory activity of Bifidobacterium adolescent combined with cisplatin on melanoma in mice and its mechanism[J]. Front. Med., 2008, 2(2): 186-190.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed