Cargando…

Activated NK cells reprogram MDSCs via NKG2D-NKG2DL and IFN-γ to modulate antitumor T-cell response after cryo-thermal therapy

BACKGROUND: Myeloid‐derived suppressor cells (MDSCs) can potently inhibit T-cell activity, promote growth and metastasis of tumor and contribute to resistance to immunotherapy. Targeting MDSCs to alleviate their protumor functions and immunosuppressive activities is intimately associated with cancer...

Descripción completa

Detalles Bibliográficos
Autores principales: Peng, Peng, Lou, Yue, Wang, Shicheng, Wang, Junjun, Zhang, Zelu, Du, Peishan, Zheng, Jiamin, Liu, Ping, Xu, Lisa X
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BMJ Publishing Group 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9756281/
https://www.ncbi.nlm.nih.gov/pubmed/36521929
http://dx.doi.org/10.1136/jitc-2022-005769
_version_ 1784851600026632192
author Peng, Peng
Lou, Yue
Wang, Shicheng
Wang, Junjun
Zhang, Zelu
Du, Peishan
Zheng, Jiamin
Liu, Ping
Xu, Lisa X
author_facet Peng, Peng
Lou, Yue
Wang, Shicheng
Wang, Junjun
Zhang, Zelu
Du, Peishan
Zheng, Jiamin
Liu, Ping
Xu, Lisa X
author_sort Peng, Peng
collection PubMed
description BACKGROUND: Myeloid‐derived suppressor cells (MDSCs) can potently inhibit T-cell activity, promote growth and metastasis of tumor and contribute to resistance to immunotherapy. Targeting MDSCs to alleviate their protumor functions and immunosuppressive activities is intimately associated with cancer immunotherapy. Natural killer (NK) cells can engage in crosstalk with multiple myeloid cells to alter adaptive immune responses, triggering T-cell immunity. However, whether the NK-cell-MDSC interaction can modulate the T-cell immune response requires further study. Cryo-thermal therapy could induce the maturation of MDSCs by creating an acute inflammatory environment to elicit a CD4(+) Th1-dominant immune response, but the mechanism regulating this process remains unclear. METHODS: NK cells were depleted and NKG2D was blocked with monoclonal antibodies in vivo. MDSCs, NK cells and T cells were assessed by flow cytometry and isolated by magnetic-activated cell sorting (MACS). MDSCs and NK cells were cocultured with T cells to determine their immunological function. The transcriptional profiles of MDSCs were measured by qRT-PCR and RNA-sequencing. Isolated NK cells and MDSCs by MACS were cocultured to study the viability and maturation of MDSCs regulated by NK cells. TIMER was used to comprehensively examine the immunological, clinical, and genomic features of tumors. RESULTS: NK-cell activation after cryo-thermal therapy decreased MDSC accumulation and reprogrammed immunosuppressive MDSCs toward a mature phenotype to promote T cell antitumor immunity. Furthermore, we discovered that NK cells could kill MDSCs via the NKG2D-NKG2DL axis and promote MDSC maturation by interferon gamma (IFN-γ) in response to NKG2D. In addition, CD4(+) Th1-dominant antitumor immune response was dependent on NKG2D, which promoted the major histocompatibility complex Ⅱ pathway of MDSCs. High activated NK-cell infiltration and NKG2D level in tumors were positively correlated with better clinical outcomes. CONCLUSIONS: Cryo-thermal therapy induces effective CD4(+) Th1-dominant antitumor immunity by activating NK cells to reprogram MDSCs, providing a promising therapeutic strategy for cancer immunotherapy.
format Online
Article
Text
id pubmed-9756281
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BMJ Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-97562812022-12-17 Activated NK cells reprogram MDSCs via NKG2D-NKG2DL and IFN-γ to modulate antitumor T-cell response after cryo-thermal therapy Peng, Peng Lou, Yue Wang, Shicheng Wang, Junjun Zhang, Zelu Du, Peishan Zheng, Jiamin Liu, Ping Xu, Lisa X J Immunother Cancer Basic Tumor Immunology BACKGROUND: Myeloid‐derived suppressor cells (MDSCs) can potently inhibit T-cell activity, promote growth and metastasis of tumor and contribute to resistance to immunotherapy. Targeting MDSCs to alleviate their protumor functions and immunosuppressive activities is intimately associated with cancer immunotherapy. Natural killer (NK) cells can engage in crosstalk with multiple myeloid cells to alter adaptive immune responses, triggering T-cell immunity. However, whether the NK-cell-MDSC interaction can modulate the T-cell immune response requires further study. Cryo-thermal therapy could induce the maturation of MDSCs by creating an acute inflammatory environment to elicit a CD4(+) Th1-dominant immune response, but the mechanism regulating this process remains unclear. METHODS: NK cells were depleted and NKG2D was blocked with monoclonal antibodies in vivo. MDSCs, NK cells and T cells were assessed by flow cytometry and isolated by magnetic-activated cell sorting (MACS). MDSCs and NK cells were cocultured with T cells to determine their immunological function. The transcriptional profiles of MDSCs were measured by qRT-PCR and RNA-sequencing. Isolated NK cells and MDSCs by MACS were cocultured to study the viability and maturation of MDSCs regulated by NK cells. TIMER was used to comprehensively examine the immunological, clinical, and genomic features of tumors. RESULTS: NK-cell activation after cryo-thermal therapy decreased MDSC accumulation and reprogrammed immunosuppressive MDSCs toward a mature phenotype to promote T cell antitumor immunity. Furthermore, we discovered that NK cells could kill MDSCs via the NKG2D-NKG2DL axis and promote MDSC maturation by interferon gamma (IFN-γ) in response to NKG2D. In addition, CD4(+) Th1-dominant antitumor immune response was dependent on NKG2D, which promoted the major histocompatibility complex Ⅱ pathway of MDSCs. High activated NK-cell infiltration and NKG2D level in tumors were positively correlated with better clinical outcomes. CONCLUSIONS: Cryo-thermal therapy induces effective CD4(+) Th1-dominant antitumor immunity by activating NK cells to reprogram MDSCs, providing a promising therapeutic strategy for cancer immunotherapy. BMJ Publishing Group 2022-12-15 /pmc/articles/PMC9756281/ /pubmed/36521929 http://dx.doi.org/10.1136/jitc-2022-005769 Text en © Author(s) (or their employer(s)) 2022. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ. https://creativecommons.org/licenses/by-nc/4.0/This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See http://creativecommons.org/licenses/by-nc/4.0/ (https://creativecommons.org/licenses/by-nc/4.0/) .
spellingShingle Basic Tumor Immunology
Peng, Peng
Lou, Yue
Wang, Shicheng
Wang, Junjun
Zhang, Zelu
Du, Peishan
Zheng, Jiamin
Liu, Ping
Xu, Lisa X
Activated NK cells reprogram MDSCs via NKG2D-NKG2DL and IFN-γ to modulate antitumor T-cell response after cryo-thermal therapy
title Activated NK cells reprogram MDSCs via NKG2D-NKG2DL and IFN-γ to modulate antitumor T-cell response after cryo-thermal therapy
title_full Activated NK cells reprogram MDSCs via NKG2D-NKG2DL and IFN-γ to modulate antitumor T-cell response after cryo-thermal therapy
title_fullStr Activated NK cells reprogram MDSCs via NKG2D-NKG2DL and IFN-γ to modulate antitumor T-cell response after cryo-thermal therapy
title_full_unstemmed Activated NK cells reprogram MDSCs via NKG2D-NKG2DL and IFN-γ to modulate antitumor T-cell response after cryo-thermal therapy
title_short Activated NK cells reprogram MDSCs via NKG2D-NKG2DL and IFN-γ to modulate antitumor T-cell response after cryo-thermal therapy
title_sort activated nk cells reprogram mdscs via nkg2d-nkg2dl and ifn-γ to modulate antitumor t-cell response after cryo-thermal therapy
topic Basic Tumor Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9756281/
https://www.ncbi.nlm.nih.gov/pubmed/36521929
http://dx.doi.org/10.1136/jitc-2022-005769
work_keys_str_mv AT pengpeng activatednkcellsreprogrammdscsviankg2dnkg2dlandifngtomodulateantitumortcellresponseaftercryothermaltherapy
AT louyue activatednkcellsreprogrammdscsviankg2dnkg2dlandifngtomodulateantitumortcellresponseaftercryothermaltherapy
AT wangshicheng activatednkcellsreprogrammdscsviankg2dnkg2dlandifngtomodulateantitumortcellresponseaftercryothermaltherapy
AT wangjunjun activatednkcellsreprogrammdscsviankg2dnkg2dlandifngtomodulateantitumortcellresponseaftercryothermaltherapy
AT zhangzelu activatednkcellsreprogrammdscsviankg2dnkg2dlandifngtomodulateantitumortcellresponseaftercryothermaltherapy
AT dupeishan activatednkcellsreprogrammdscsviankg2dnkg2dlandifngtomodulateantitumortcellresponseaftercryothermaltherapy
AT zhengjiamin activatednkcellsreprogrammdscsviankg2dnkg2dlandifngtomodulateantitumortcellresponseaftercryothermaltherapy
AT liuping activatednkcellsreprogrammdscsviankg2dnkg2dlandifngtomodulateantitumortcellresponseaftercryothermaltherapy
AT xulisax activatednkcellsreprogrammdscsviankg2dnkg2dlandifngtomodulateantitumortcellresponseaftercryothermaltherapy