Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2022 Sep;10(9):e005061.
doi: 10.1136/jitc-2022-005061.

N-cadherin protects oral cancer cells from NK cell killing in the circulation by inducing NK cell functional exhaustion via the KLRG1 receptor

Affiliations

N-cadherin protects oral cancer cells from NK cell killing in the circulation by inducing NK cell functional exhaustion via the KLRG1 receptor

Chao Lou et al. J Immunother Cancer. 2022 Sep.

Abstract

Background: Circulating tumor cells (CTCs) can survive in the circulation and return to primary tumors through a self-seeding process. However, the mechanisms underlying CTCs escape from natural killer (NK) cell-mediated immune surveillance remain unclear.

Method: Self-seeded tumor cells were isolated and characterized using a modified contralateral seeding model. A comparison of transcriptional profiles was performed between the parental cells and self-seeded cells. The molecular mechanism of self-seeded tumor cells escaping from NK cell was demonstrated through in vitro experiments and verified in a CTC-mimicking in vivo model. Then, the expression level of key protein mediating CTCs immune escape was detected in 24 paired primary and recurrent tumor samples of patients with oral cancer by the immunohistochemical method.

Result: Self-seeded cells displayed resistance to NK cell-mediated lysis and a higher tumor seeding ability than their parental cells. Elevated expression levels of the CDH2 gene and its protein product, N-cadherin were found in self-seeded cells. NK cells secreted cytokines, and fluid shear stress facilitated N-cadherin release by promoting A disintegrin and metalloprotease 10 (ADAM10) translation or converting the precursor ADAM10 to the mature form. Soluble N-cadherin triggered NK cell functional exhaustion by interacting with the killer cell lectin-like receptor subfamily G member 1 (KLRG1) receptor and therefore protected tumor cells from NK cell killing in the circulation. In vivo experimental results showed that overexpression of N-cadherin promoted tumor self-seeding and facilitated the survival of CTCs. Compared with primary tumors, N-cadherin expression was significantly increased in matched recurrent tumor tissues.

Conclusion: Together, our findings illustrate an unknown mechanism by which CTCs evaded NK cell-mediated immune surveillance, and indicate that targeting N-cadherin is an effective strategy to prevent CTCs from homing to primary tumor.

Keywords: biomarkers, tumor; gene expression profiling; head and neck neoplasms; immune evation; killer cells, natural.

PubMed Disclaimer

Conflict of interest statement

Competing interests: None declared.

Figures

Figure 1
Figure 1
Isolation and characterization of self-seeded cells. (A, B) Seeded tumors were visualized by fluorescence microscopy at 60 days (A) or 30 days (B) after inoculation using contralateral seeding model (left) or NK cell-depleted contralateral seeding model (middle), and fluorescence-based quantification was measured (Right) (n=6 per group). RFP-labeled SCC9 cells were defined as donor cells and GFP-labeled SCC9 cells were defined as recipient cells. Bars: 500 µm. (C) Isolation of SCC9-seeded cells by FACS. (D) Bright field and fluorescence images of isolated SCC9-seeded cells. Bars: 50 µm. (E–G) Comparison of wound healing (E), colony formation (F) and proliferation ability (G) between SCC9-parental and SCC9-seeded cells. (H) The expression of EMT markers in SCC9-parental and SCC9-seeded cells. (I) Cleaved-PARP expression in monolayer cultured or suspension cultured SCC9-seeded and SCC9-parental cells. (J) Lysis of SCC9-seeded and SCC9-parental cells by fresh NK cells at E/T ratios of 5:1, 10:1, and 20:1. (K, L) Seeded tumors were visualized by fluorescence microscopy at 60 days (K) or 30 days (L) after inoculation using contralateral seeding model (Left, middle), and fluorescence-based quantification was measured (Right) (n=6 per group). SCC9-seeded cells were inoculated as donor cells. Bar: 500 µm. Data represent three independent experiments done in triplicate (in vitro model) or with 6 mice per group (in vivo model) (mean±SD). *p<0.05, **p<0.01, ***p<0.001; ns, no significance; unpaired Student’s t-test (A, B, E, F, K and L), two-way ANOVA analysis (G, J). ANOVA, analysis of variance; E/T, effector/target; FACS, fluorescence activated cell sorting; GFP, green fluorescent protein; NK, natural killer; PARP, poly ADP-ribose polymerase; RFP, red fluorescent protein; shN-cad, short hairpin N-cadherin.
Figure 2
Figure 2
High expression of N-cadherin in self-seeded cells. (A) A comparison of transcriptional profiles was performed between SCC9-seeded and SCC9-parental cells. (B) Venn diagram analysis indicated three overlapping genes (CDH2, FN1 and HLA-E) between set 1 and set 2. Set 1 includes differentially expressed genes between SCC9-seeded and SCC9-parental cells; and set 2 includes membrane molecules related to NK cell function based on published literature. (C) The mRNA expression of CDH2, FN1 and HLA-E in SCC9-seeded and SCC9-parental cells was assessed using qRT-PCR analysis. (D) Homology of CDH1, CDH2 and CDH4 between humans and mice. (E) The mRNA expression of CDH1, CDH2 and CDH4 in SCC9-seeded and SCC9-parental cells was assessed using qRT-PCR analysis. (F) Protein levels of N-cadherin in SCC9-seeded and SCC9-parental cells. (G) Immunofluorescence staining of N-cadherin in SCC9-seeded and SCC9-parental cells. Bars: 25 µm. (H) Detection of N-cadherin in CTCs harvested from patients with oral cancer. From left to right: CK staining; CD45 staining; DAPI staining; N-cadherin staining; merged image. Bars: 50 µm. (I) Comparison of KLRG1 expression status on NK cells in healthy individuals and patients with oral cancer. Data represent three independent experiments done in triplicate (in vitro model) or with 10 patients with oral cancer (H) or 20 individuals (I, 10 patients with oral cancer and 10 healthy control) (mean±SD). **p<0.01, ****p<0.0001; ns, no significance; unpaired Student’s t-test (C, E and I). CK, cytokeratin; CTCs, circulating tumor cells; DAPI, 4’,6-diamidino-2-phenylindole; mRNA, message RNA; NK, natural killer; qRT-PCR, quantitative real time-PCR.
Figure 3
Figure 3
Self-seeded cell-derived soluble N-cadherin impaired cytotoxicity by interacting with the KLRG1 receptor. (A) Lysis of the indicated tumor cell lines by total NK populations (left), KLRG1 (+) subsets (Middle) and KLRG1 (−) subsets (Right) at E/T ratios of 5:1, 10:1 and 20:1. (B) Comparison of total NK populations (Left), KLRG1 (+) subsets (Middle) and KLRG1 (−) subsets (right) cytotoxicity against K562 cells after exposure to various tumor cell culture supernatants at E/T ratios of 5:1, 10:1 and 20:1. (C) Soluble N-cadherin levels in supernatants of the indicated cells. sN-cad: soluble N-cadherin. (D) Comparison of total NK populations (left), KLRG1 (+) subsets (Middle) and KLRG1 (−) subsets (Right) cytotoxicity against K562 cells with the indicated treatments at E/T ratios of 5:1, 10:1 and 20:1. (E) Cytokine production of total NK populations (left), KLRG1 (+) subsets (Middle) and KLRG1 (−) subsets (right) after the indicated treatments. Data represent three independent experiments done in triplicate (mean±SD). *p<0.05, **p<0.01, ***p<0.001; ns, no significance; two-way ANOVA analysis (A, B, D), one-way ANOVA and Tukey-Kramer multiple comparison tests (C, E) ANOVA, analysis of variance; E/T, effector/ target; IFN, interferon; NK, natural killer; N-cad Ab, N-cadherin blocking antibody; rhN-cad, recombinant human soluble N-cadherin; shNC, short hairpin negative control; shN-cad, short hairpin N-cadherin; TNF, tumor necrosis factor.
Figure 4
Figure 4
Soluble N-cadherin induces NK cell functional exhaustion in a KLRG1 receptor-dependent manner. Freshly isolated total NK populations were exposed to the indicated tumor cell culture supernatants or the recombinant human soluble N-cadherin with or without an N-cadherin blocking antibody. KLRG1 (+) subsets and KLRG1 (−) subsets were administered recombinant human soluble N-cadherin with or without an N-cadherin blocking antibody. The NK populations were then phenotyped. (A) Statistics for FACS data of cytolytic effector molecules expression (left), NKARs expression middle) and NKIRs expression (right) in NK-cell populations following indicated treatments. (B) FACS analysis of surface FasL and TRAIL expression levels in KLRG1 (−) and KLRG1 (+) NK subsets after exposure to various tumor cell culture supernatants. Data represent three independent experiments done in triplicate (mean±SD). *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001; ns, no significance; one-way ANOVA and Tukey-Kramer multiple comparison tests. ANOVA, analysis of variance; DMEM, dulbecco‘s modified eagle medium; FACS, fluorescence activated cell sorting; NKARs, NK cell activation receptors; N-cad Ab, N-cadherin blocking antibody; NK, natural killer; NKIRs, NK cell inhibitory receptor; rhN-cad, recombinant human soluble N-cadherin; shNC, short hairpin negative control; shN-cad, short hairpin N-cadherin.
Figure 5
Figure 5
NK cell-secreted cytokines and fluid shear stress facilitate N-cadherin release from tumor cells by enhancing the activity of ADAM10. (A) ADAM10 expression in SCC9-parental, SCC9-seeded and SCC9-seeded shADAM10 cells. (B) sN-cad levels in the supernatants of SCC9-seeded, ADAM10-silenced SCC9-seeded, SCC9-parental and ADAM10-silenced SCC9-parental cells. (C) NK cell cytotoxicity against K562 cells with the indicated treatments at E/T ratios of 5:1, 10:1 and 20:1. (D) sN-cad levels in the supernatants of SCC9-seeded, ADAM10-silenced SCC9-seeded, N-cadherin-silenced SCC9-seeded, SCC9-parental, ADAM10-silenced SCC9-parental and N-cadherin-silenced SCC9-parental cells after exposure to NK cell supernatants. (E) sN-cad levels in the supernatants of SCC9-seeded, ADAM10-silenced SCC9-seeded, SCC9-parental and ADAM10-silenced SCC9-parental cells with the indicated treatments. (F) ADAM10 activity in SCC9-seeded and SCC9-parental cells with the indicated treatments. (G) Western blots of AKT, phospho-AKT, p70-S6K, phospho-p70-S6K, 4EB-P1, phospho-4EB-P1 and ADAM10 levels in SCC9-seeded cells with the indicated treatments. (H) sN-cad levels in the supernatants of SCC9-seeded cells with the indicated treatments. (I) NK cell cytotoxicity against K562 cells with the indicated treatments at E/T ratios of 5:1, 10:1 and 20:1. (J, K) ADAM10 activity (J) and expression (K) in tumor cells cultured in a shaker incubator for 24 hours. (L) Calcium influx in SCC9-seeded cells cultured in a shaker incubator for 24 hours. GsMTx4, a specific inhibitor of the Piezo1, abrogated the shear stress-induced calcium influx. (M–O) ADAM10 expression (M), sN-cad levels (N) and the effect of cell supernatants on NK cell cytotoxicity (O) in SCC9-seeded cells cultured in a shaker incubator with or without GsMTx4 treatment. (P) sN-cad levels in the supernatants of tumor cells with the indicated treatments. (Q) NK cell cytotoxicity against K562 cells with the indicated treatments at E/T ratios of 5:1, 10:1 and 20:1. (R, S) Gel-like image representation (left) and electropherograms representation (right) of N-cadherin (R) and ADAM10 (S) expression in SCC9-parental and SCC9-seeded cell enriched samples obtained from both Rag1-/- and NK cell-depleted Rag1-/- self-seeding models by Capillary Western analysis. Data represent three independent experiments done in triplicate (mean±SD). *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001; ns, no significance; unpaired Student’s t-test (B, J); two-way ANOVA analysis (C, I, O and Q); one-way ANOVA and Tukey-Kramer multiple comparison tests (D–F, H, L, N and P). ANOVA, analysis of variance; E/T, effector/target; NK, natural killer; shADAM10, short hairpin ADAM10; shNC, short hairpin negative control; shN-cad, short hairpin N-cadherin; sN-cad, Soluble N-cadherin; TNF, tumor necrosis factor.
Figure 6
Figure 6
N-cadherin and ADAM10 expression affects the tumor self-seeding process. (A, B) Seeded tumors were visualized by fluorescence microscopy at 60 days after inoculation using contralateral seeding model, and fluorescence-based quantification was measured (n=6 per group). SCC9-seeded shN-Cad cells (A) and SCC9-seeded shADAM10 cells (B) were inoculated as donor cells. Bars: 500 µm. (C, D) Seeded tumors were visualized by fluorescence microscopy at 60 days after inoculation using NK cell-depleted contralateral seeding model, and fluorescence-based quantification was measured (n=6 per group). SCC9-seeded shN-Cad cells (C) and SCC9-seeded shADAM10 cells (D) were inoculated as donor cells. Bars: 500 µm. (E, F) Seeded tumors were visualized by fluorescence microscopy at 60 days (E) or 30 days (F) after inoculation using contralateral seeding model, and fluorescence-based quantification was measured (n=6 per group). SCC9 oeN-Cad cells were inoculated as donor cells. Bars: 500 µm. Data represent two independent experiments with 6 mice per group (mean±SD). **p<0.01, ***p<0.001, ****p<0.0001; unpaired Student’s t-test. RFP, red fluorescent protein; GFP, green fluorescent protein; shN-cad, short hairpin N-cadherin; shADAM10, short hairpin ADAM10; oeN-cad, overexpress N-cadherin.
Figure 7
Figure 7
N-cadherin and ADAM10 expression facilitates survival of CTCs and elevated N-cadherin expression in recurrent oral cancer samples. (A–H) CTCs were detected by IVFC in indicated groups of mouse models, each fluorescence peak corresponded to a single surviving CTC. (n=5 per group) (I) Quantification of surviving CTCs in each group. (J) Freshly NK cells were ioslated from each group and FACS analysis of cytolytic effector molecules (granzymes and perforin) expression, cytokines (IFN-γ and TNF-α) expression, NK cell activation receptors (NKG2D and NKp46) expression and NK cell inhibitory receptors (NKG2A and KLRG1) expression in in NK cells of indicated groups. (K, L) Representative images of N-cadherin-stained (K) and ADAM10-stained (L) sections and quantification of immunohistochemical staining in both primary and matched recurrent tumor tissues (n=24 paired samples). Bars: 100 µm. Data represent three independent experiments done in triplicate with 5 mice per group (A–J) or with 24 paired samples (K, L) (mean±SD). *p<0.05, **p<0.01, ***p<0.001, ns, no significance; one-way ANOVA and Tukey-Kramer multiple comparison tests (I, J), paired Student’s t-test (K, L). shN-cad, short hairpin N-cadherin; shADAM10, short hairpin ADAM10; oeN-cad, overexpress N-cadherin; PBS, phosphate buffered saline.

Similar articles

Cited by

References

    1. Yu M, Bardia A, Wittner BS, et al. . Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science 2013;339:580–4. 10.1126/science.1228522 - DOI - PMC - PubMed
    1. Chemi F, Mohan S, Guevara T, et al. . Early dissemination of circulating tumor cells: biological and clinical insights. Front Oncol 2021;11:672195. 10.3389/fonc.2021.672195 - DOI - PMC - PubMed
    1. Aceto N, Bardia A, Miyamoto DT, et al. . Circulating tumor cell clusters are oligoclonal precursors of breast cancer metastasis. Cell 2014;158:1110–22. 10.1016/j.cell.2014.07.013 - DOI - PMC - PubMed
    1. Nagrath S, Sequist LV, Maheswaran S, et al. . Isolation of rare circulating tumour cells in cancer patients by microchip technology. Nature 2007;450:1235–9. 10.1038/nature06385 - DOI - PMC - PubMed
    1. Massagué J, Obenauf AC. Metastatic colonization by circulating tumour cells. Nature 2016;529:298–306. 10.1038/nature17038 - DOI - PMC - PubMed

Publication types

-