Improved Antitumor Effect of NK Cells Activated by Neutrophils in a Bone Marrow Transplant Model

The licensing process mediated by inhibitory receptors of the Ly49 C-type lectin superfamily that recognizes self-major histocompatibility complex (MHC) class I in mice is essential for the proper antitumor function of natural killer (NK) cells. Several models for NK cell licensing can be exploited...

Full description

Bibliographic Details
Main Authors: Daisuke Nakato, Shotaro Iwamoto, Keishiro Amano, Takahiro Ito, Hidemi Toyoda, Ryo Hanaki, Mari Morimoto, Kaori Niwa, Isao Tawara, Kyoko Imanaka-Yoshida, Masahiro Ogawa, Masahiro Hirayama
Format: Article
Language:English
Published: Hindawi Limited 2023-01-01
Series:Mediators of Inflammation
Online Access:http://dx.doi.org/10.1155/2023/6316581
_version_ 1811165458735300608
author Daisuke Nakato
Shotaro Iwamoto
Keishiro Amano
Takahiro Ito
Hidemi Toyoda
Ryo Hanaki
Mari Morimoto
Kaori Niwa
Isao Tawara
Kyoko Imanaka-Yoshida
Masahiro Ogawa
Masahiro Hirayama
author_facet Daisuke Nakato
Shotaro Iwamoto
Keishiro Amano
Takahiro Ito
Hidemi Toyoda
Ryo Hanaki
Mari Morimoto
Kaori Niwa
Isao Tawara
Kyoko Imanaka-Yoshida
Masahiro Ogawa
Masahiro Hirayama
author_sort Daisuke Nakato
collection DOAJ
description The licensing process mediated by inhibitory receptors of the Ly49 C-type lectin superfamily that recognizes self-major histocompatibility complex (MHC) class I in mice is essential for the proper antitumor function of natural killer (NK) cells. Several models for NK cell licensing can be exploited for adoptive immunotherapy for cancer. However, the appropriate adoptive transfer setting to induce efficient graft versus tumor/leukemia effects remains elusive, especially after hematopoietic stem cell transplantation (HSCT). In our previous experiment, we showed that intraperitoneal neutrophil administration with their corresponding NK receptor ligand-activated NK cells using congenic mice without HSCT. In this experiment, we demonstrate enhanced antitumor effects of licensed NK cells induced by weekly intraperitoneal injections of irradiated neutrophil-enriched peripheral blood mononuclear cells (PBMNCs) in recipient mice bearing lymphoma. Bone marrow transplantation was performed using BALB/c mice (H-2d) as the recipient and B10 mice (H-2b) as the donor. The tumor was A20, a BALB/c-derived lymphoma cell line, which was injected subcutaneously into the recipient at the same time as the HSCT. Acute graft versus host disease was not exacerbated in this murine MHC class I mismatched HSCT setting. The intraperitoneal injection of PBMNCs activated a transient licensing of NK subsets expressed Ly49G2, its corresponding NK receptor ligand to H-2d, and reduced A20 tumor growth in the recipient after HSCT. Pathological examination revealed that increased donor-oriented NK1.1+NK cells migrated into the recipient tumors, depending on neutrophil counts in the administered PBMNCs. Collectively, our data reveal a pivotal role of neutrophils in promoting NK cell effector functions and adoptive immunotherapy for cancer.
first_indexed 2024-04-10T15:37:25Z
format Article
id doaj.art-20b9b5d21a3845a0a85a3b857a70aec6
institution Directory Open Access Journal
issn 1466-1861
language English
last_indexed 2024-04-10T15:37:25Z
publishDate 2023-01-01
publisher Hindawi Limited
record_format Article
series Mediators of Inflammation
spelling doaj.art-20b9b5d21a3845a0a85a3b857a70aec62023-02-13T01:09:01ZengHindawi LimitedMediators of Inflammation1466-18612023-01-01202310.1155/2023/6316581Improved Antitumor Effect of NK Cells Activated by Neutrophils in a Bone Marrow Transplant ModelDaisuke Nakato0Shotaro Iwamoto1Keishiro Amano2Takahiro Ito3Hidemi Toyoda4Ryo Hanaki5Mari Morimoto6Kaori Niwa7Isao Tawara8Kyoko Imanaka-Yoshida9Masahiro Ogawa10Masahiro Hirayama11Department of PediatricsDepartment of PediatricsDepartment of PediatricsDepartment of PediatricsDepartment of PediatricsDepartment of PediatricsDepartment of PediatricsDepartment of PediatricsDepartment of Hematology and OncologyDepartment of Pathology and Matrix BiologyDepartment of PediatricsDepartment of PediatricsThe licensing process mediated by inhibitory receptors of the Ly49 C-type lectin superfamily that recognizes self-major histocompatibility complex (MHC) class I in mice is essential for the proper antitumor function of natural killer (NK) cells. Several models for NK cell licensing can be exploited for adoptive immunotherapy for cancer. However, the appropriate adoptive transfer setting to induce efficient graft versus tumor/leukemia effects remains elusive, especially after hematopoietic stem cell transplantation (HSCT). In our previous experiment, we showed that intraperitoneal neutrophil administration with their corresponding NK receptor ligand-activated NK cells using congenic mice without HSCT. In this experiment, we demonstrate enhanced antitumor effects of licensed NK cells induced by weekly intraperitoneal injections of irradiated neutrophil-enriched peripheral blood mononuclear cells (PBMNCs) in recipient mice bearing lymphoma. Bone marrow transplantation was performed using BALB/c mice (H-2d) as the recipient and B10 mice (H-2b) as the donor. The tumor was A20, a BALB/c-derived lymphoma cell line, which was injected subcutaneously into the recipient at the same time as the HSCT. Acute graft versus host disease was not exacerbated in this murine MHC class I mismatched HSCT setting. The intraperitoneal injection of PBMNCs activated a transient licensing of NK subsets expressed Ly49G2, its corresponding NK receptor ligand to H-2d, and reduced A20 tumor growth in the recipient after HSCT. Pathological examination revealed that increased donor-oriented NK1.1+NK cells migrated into the recipient tumors, depending on neutrophil counts in the administered PBMNCs. Collectively, our data reveal a pivotal role of neutrophils in promoting NK cell effector functions and adoptive immunotherapy for cancer.http://dx.doi.org/10.1155/2023/6316581
spellingShingle Daisuke Nakato
Shotaro Iwamoto
Keishiro Amano
Takahiro Ito
Hidemi Toyoda
Ryo Hanaki
Mari Morimoto
Kaori Niwa
Isao Tawara
Kyoko Imanaka-Yoshida
Masahiro Ogawa
Masahiro Hirayama
Improved Antitumor Effect of NK Cells Activated by Neutrophils in a Bone Marrow Transplant Model
Mediators of Inflammation
title Improved Antitumor Effect of NK Cells Activated by Neutrophils in a Bone Marrow Transplant Model
title_full Improved Antitumor Effect of NK Cells Activated by Neutrophils in a Bone Marrow Transplant Model
title_fullStr Improved Antitumor Effect of NK Cells Activated by Neutrophils in a Bone Marrow Transplant Model
title_full_unstemmed Improved Antitumor Effect of NK Cells Activated by Neutrophils in a Bone Marrow Transplant Model
title_short Improved Antitumor Effect of NK Cells Activated by Neutrophils in a Bone Marrow Transplant Model
title_sort improved antitumor effect of nk cells activated by neutrophils in a bone marrow transplant model
url http://dx.doi.org/10.1155/2023/6316581
work_keys_str_mv AT daisukenakato improvedantitumoreffectofnkcellsactivatedbyneutrophilsinabonemarrowtransplantmodel
AT shotaroiwamoto improvedantitumoreffectofnkcellsactivatedbyneutrophilsinabonemarrowtransplantmodel
AT keishiroamano improvedantitumoreffectofnkcellsactivatedbyneutrophilsinabonemarrowtransplantmodel
AT takahiroito improvedantitumoreffectofnkcellsactivatedbyneutrophilsinabonemarrowtransplantmodel
AT hidemitoyoda improvedantitumoreffectofnkcellsactivatedbyneutrophilsinabonemarrowtransplantmodel
AT ryohanaki improvedantitumoreffectofnkcellsactivatedbyneutrophilsinabonemarrowtransplantmodel
AT marimorimoto improvedantitumoreffectofnkcellsactivatedbyneutrophilsinabonemarrowtransplantmodel
AT kaoriniwa improvedantitumoreffectofnkcellsactivatedbyneutrophilsinabonemarrowtransplantmodel
AT isaotawara improvedantitumoreffectofnkcellsactivatedbyneutrophilsinabonemarrowtransplantmodel
AT kyokoimanakayoshida improvedantitumoreffectofnkcellsactivatedbyneutrophilsinabonemarrowtransplantmodel
AT masahiroogawa improvedantitumoreffectofnkcellsactivatedbyneutrophilsinabonemarrowtransplantmodel
AT masahirohirayama improvedantitumoreffectofnkcellsactivatedbyneutrophilsinabonemarrowtransplantmodel