Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2021

Open Access 01-12-2021 | Melanoma | Research

MITF induces escape from innate immunity in melanoma

Authors: Luis Sánchez-del-Campo, Román Martí-Díaz, María F. Montenegro, Rebeca González-Guerrero, Trinidad Hernández-Caselles, Enrique Martínez-Barba, Antonio Piñero-Madrona, Juan Cabezas-Herrera, Colin R. Goding, José Neptuno Rodríguez-López

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2021

Login to get access

Abstract

Background

The application of immune-based therapies has revolutionized cancer treatment. Yet how the immune system responds to phenotypically heterogeneous populations within tumors is poorly understood. In melanoma, one of the major determinants of phenotypic identity is the lineage survival oncogene MITF that integrates diverse microenvironmental cues to coordinate melanoma survival, senescence bypass, differentiation, proliferation, invasion, metabolism and DNA damage repair. Whether MITF also controls the immune response is unknown.

Methods

By using several mouse melanoma models, we examine the potential role of MITF to modulate the anti-melanoma immune response. ChIP-seq data analysis, ChIP-qPCR, CRISPR-Cas9 genome editing, and luciferase reporter assays were utilized to identify ADAM10 as a direct MITF target gene. Western blotting, confocal microscopy, flow cytometry, and natural killer (NK) cytotoxicity assays were used to determine the underlying mechanisms by which MITF-driven phenotypic plasticity modulates melanoma NK cell-mediated killing.

Results

Here we show that MITF regulates expression of ADAM10, a key sheddase that cleaves the MICA/B family of ligands for NK cells. By controlling melanoma recognition by NK-cells MITF thereby controls the melanoma response to the innate immune system. Consequently, while melanoma MITFLow cells can be effectively suppressed by NK-mediated killing, MITF-expressing cells escape NK cell surveillance.

Conclusion

Our results reveal how modulation of MITF activity can impact the anti-melanoma immune response with implications for the application of anti-melanoma immunotherapies.
Appendix
Available only for authorised users
Literature
2.
go back to reference García-Jimenéz C, Goding CR. Starvation and pseudo-starvation as drivers of cancer metastasis through translation reprogramming. Cell Metab. 2019;29:258–67.CrossRef García-Jimenéz C, Goding CR. Starvation and pseudo-starvation as drivers of cancer metastasis through translation reprogramming. Cell Metab. 2019;29:258–67.CrossRef
24.
go back to reference Foerster F, Boegel S, Heck, Pickert G, Rüssel N, Rosigkeit S, et al. Enhanced protection of C57 BL/6 vs Balb/c mice to melanoma liver metastasis is mediated by NK cells. Oncoimmunology. 2017;7:e1409929.CrossRefPubMedPubMedCentral Foerster F, Boegel S, Heck, Pickert G, Rüssel N, Rosigkeit S, et al. Enhanced protection of C57 BL/6 vs Balb/c mice to melanoma liver metastasis is mediated by NK cells. Oncoimmunology. 2017;7:e1409929.CrossRefPubMedPubMedCentral
26.
go back to reference Montenegro MF, González-Guerrero R, Sánchez-del-Campo L, Piñero-Madrona A, Cabezas-Herrera J, Rodríguez-López JN. Targeting the epigenetics of the DNA damage response in breast cancer. Cell Death Dis. 2016;7:e2180.CrossRefPubMedPubMedCentral Montenegro MF, González-Guerrero R, Sánchez-del-Campo L, Piñero-Madrona A, Cabezas-Herrera J, Rodríguez-López JN. Targeting the epigenetics of the DNA damage response in breast cancer. Cell Death Dis. 2016;7:e2180.CrossRefPubMedPubMedCentral
34.
go back to reference Ferrari de Andrade L, Tay RE, Pan D, Luoma AM, Ito Y, Badrinath S, et al. Antibody-mediated inhibition of MICA and MICB shedding promotes NK cell–driven tumor immunity. Science. 2018;359:1537–42.CrossRefPubMedPubMedCentral Ferrari de Andrade L, Tay RE, Pan D, Luoma AM, Ito Y, Badrinath S, et al. Antibody-mediated inhibition of MICA and MICB shedding promotes NK cell–driven tumor immunity. Science. 2018;359:1537–42.CrossRefPubMedPubMedCentral
36.
go back to reference López-Cobo S, Pieper N, Campos-Silva C, García-Cuesta EM, Reyburn HT, Paschen A, et al. Impaired NK cell recognition of vemurafenib-treated melanoma cells is overcome by simultaneous application of histone deacetylase inhibitors. Oncoimmunology. 2017;7:e1392426.CrossRefPubMedPubMedCentral López-Cobo S, Pieper N, Campos-Silva C, García-Cuesta EM, Reyburn HT, Paschen A, et al. Impaired NK cell recognition of vemurafenib-treated melanoma cells is overcome by simultaneous application of histone deacetylase inhibitors. Oncoimmunology. 2017;7:e1392426.CrossRefPubMedPubMedCentral
38.
go back to reference Maurer S, Kropp KN, Klein G, Steinle A, Haen SP, Walz JS, et al. Platelet-mediated shedding of NKG2D ligands impairs NK cell immune-surveillance of tumor cells. Oncoimmunology. 2017;7:e1364827.CrossRefPubMedPubMedCentral Maurer S, Kropp KN, Klein G, Steinle A, Haen SP, Walz JS, et al. Platelet-mediated shedding of NKG2D ligands impairs NK cell immune-surveillance of tumor cells. Oncoimmunology. 2017;7:e1364827.CrossRefPubMedPubMedCentral
39.
go back to reference Donizy P, Zietek M, Leskiewicz M, Halon A, Matkowski R. High percentage of ADAM-10 positive melanoma cells correlates with paucity of tumor-infiltrating lymphocytes but does not predict prognosis in cutaneous melanoma patients. Anal Cell Pathol. 2015;2015:1–7. https://doi.org/10.1155/2015/975436.CrossRef Donizy P, Zietek M, Leskiewicz M, Halon A, Matkowski R. High percentage of ADAM-10 positive melanoma cells correlates with paucity of tumor-infiltrating lymphocytes but does not predict prognosis in cutaneous melanoma patients. Anal Cell Pathol. 2015;2015:1–7. https://​doi.​org/​10.​1155/​2015/​975436.CrossRef
51.
go back to reference Vivas-García Y, Falletta P, Liebing J, Louphrasitthiphol P, Feng Y, Chauhan J, et al. Lineage-restricted regulation of SCD and fatty acid saturation by MITF controls melanoma phenotypic plasticity. Mol Cell. 2020;77:120–39.CrossRefPubMed Vivas-García Y, Falletta P, Liebing J, Louphrasitthiphol P, Feng Y, Chauhan J, et al. Lineage-restricted regulation of SCD and fatty acid saturation by MITF controls melanoma phenotypic plasticity. Mol Cell. 2020;77:120–39.CrossRefPubMed
Metadata
Title
MITF induces escape from innate immunity in melanoma
Authors
Luis Sánchez-del-Campo
Román Martí-Díaz
María F. Montenegro
Rebeca González-Guerrero
Trinidad Hernández-Caselles
Enrique Martínez-Barba
Antonio Piñero-Madrona
Juan Cabezas-Herrera
Colin R. Goding
José Neptuno Rodríguez-López
Publication date
01-12-2021
Publisher
BioMed Central
Keywords
Melanoma
Melanoma
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2021
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-021-01916-8

Other articles of this Issue 1/2021

Journal of Experimental & Clinical Cancer Research 1/2021 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine