Cargando…

Myeloid deletion of phosphoinositide-dependent kinase-1 enhances NK cell-mediated antitumor immunity by mediating macrophage polarization

A large number of heterogeneous macrophages can be observed in solid tumor lesions. Classically activated M1 macrophages are a powerful killer of cancer cells. In contrast, tumor-associated macrophages (TAMs) are often referred to as M2 phenotype and usually impair tumor immunity mediated by cytotox...

Descripción completa

Detalles Bibliográficos
Autores principales: He, Yuexi, Du, Juan, Dong, Zhongjun
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Taylor & Francis 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7458637/
https://www.ncbi.nlm.nih.gov/pubmed/32923133
http://dx.doi.org/10.1080/2162402X.2020.1774281
_version_ 1783576239646179328
author He, Yuexi
Du, Juan
Dong, Zhongjun
author_facet He, Yuexi
Du, Juan
Dong, Zhongjun
author_sort He, Yuexi
collection PubMed
description A large number of heterogeneous macrophages can be observed in solid tumor lesions. Classically activated M1 macrophages are a powerful killer of cancer cells. In contrast, tumor-associated macrophages (TAMs) are often referred to as M2 phenotype and usually impair tumor immunity mediated by cytotoxic lymphocytes, natural killer (NK) cells and CD8(+) T cells. Therefore, orchestrating M2 to M1 reprogramming will provide a promising approach to tumor immunotherapy. Here we used a PyMT-induced spontaneous breast cancer model in which M2-polarized macrophages were abundant. This M2 phenotype was closely related to tumor progression and immune dysfunction of NK cells and CD8(+) T cells. We then found that these TAMs showed increased energy expenditure and over-activation of two kinases, Akt and mammalian target of rapamycin (mTOR). Myeloid inactivation of phosphoinositide-dependent kinase-1 (PDK1), the upstream regulator for Akt and mTOR signaling, significantly reduced excessive metabolic activation of macrophages. Notably, the loss of PDK1 significantly led to regression of breast cancer and prevented lung metastasis. Mechanistically, PDK1 deficiency mainly inhibited the activation of mTOR complex 1 (mTORC1), transforming TAMs into M1 phenotype, thereby reversing tumor-related dysfunction of T cells and NK cells. Therefore, targeting PDK1 may be a new approach for M2 macrophage-enriched solid tumor immunotherapy.
format Online
Article
Text
id pubmed-7458637
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher Taylor & Francis
record_format MEDLINE/PubMed
spelling pubmed-74586372020-09-11 Myeloid deletion of phosphoinositide-dependent kinase-1 enhances NK cell-mediated antitumor immunity by mediating macrophage polarization He, Yuexi Du, Juan Dong, Zhongjun Oncoimmunology Original Research A large number of heterogeneous macrophages can be observed in solid tumor lesions. Classically activated M1 macrophages are a powerful killer of cancer cells. In contrast, tumor-associated macrophages (TAMs) are often referred to as M2 phenotype and usually impair tumor immunity mediated by cytotoxic lymphocytes, natural killer (NK) cells and CD8(+) T cells. Therefore, orchestrating M2 to M1 reprogramming will provide a promising approach to tumor immunotherapy. Here we used a PyMT-induced spontaneous breast cancer model in which M2-polarized macrophages were abundant. This M2 phenotype was closely related to tumor progression and immune dysfunction of NK cells and CD8(+) T cells. We then found that these TAMs showed increased energy expenditure and over-activation of two kinases, Akt and mammalian target of rapamycin (mTOR). Myeloid inactivation of phosphoinositide-dependent kinase-1 (PDK1), the upstream regulator for Akt and mTOR signaling, significantly reduced excessive metabolic activation of macrophages. Notably, the loss of PDK1 significantly led to regression of breast cancer and prevented lung metastasis. Mechanistically, PDK1 deficiency mainly inhibited the activation of mTOR complex 1 (mTORC1), transforming TAMs into M1 phenotype, thereby reversing tumor-related dysfunction of T cells and NK cells. Therefore, targeting PDK1 may be a new approach for M2 macrophage-enriched solid tumor immunotherapy. Taylor & Francis 2020-06-03 /pmc/articles/PMC7458637/ /pubmed/32923133 http://dx.doi.org/10.1080/2162402X.2020.1774281 Text en © 2020 The Author(s). Published with license by Taylor & Francis Group, LLC. https://creativecommons.org/licenses/by-nc/4.0/This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (http://creativecommons.org/licenses/by-nc/4.0/ (https://creativecommons.org/licenses/by-nc/4.0/) ), which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Original Research
He, Yuexi
Du, Juan
Dong, Zhongjun
Myeloid deletion of phosphoinositide-dependent kinase-1 enhances NK cell-mediated antitumor immunity by mediating macrophage polarization
title Myeloid deletion of phosphoinositide-dependent kinase-1 enhances NK cell-mediated antitumor immunity by mediating macrophage polarization
title_full Myeloid deletion of phosphoinositide-dependent kinase-1 enhances NK cell-mediated antitumor immunity by mediating macrophage polarization
title_fullStr Myeloid deletion of phosphoinositide-dependent kinase-1 enhances NK cell-mediated antitumor immunity by mediating macrophage polarization
title_full_unstemmed Myeloid deletion of phosphoinositide-dependent kinase-1 enhances NK cell-mediated antitumor immunity by mediating macrophage polarization
title_short Myeloid deletion of phosphoinositide-dependent kinase-1 enhances NK cell-mediated antitumor immunity by mediating macrophage polarization
title_sort myeloid deletion of phosphoinositide-dependent kinase-1 enhances nk cell-mediated antitumor immunity by mediating macrophage polarization
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7458637/
https://www.ncbi.nlm.nih.gov/pubmed/32923133
http://dx.doi.org/10.1080/2162402X.2020.1774281
work_keys_str_mv AT heyuexi myeloiddeletionofphosphoinositidedependentkinase1enhancesnkcellmediatedantitumorimmunitybymediatingmacrophagepolarization
AT dujuan myeloiddeletionofphosphoinositidedependentkinase1enhancesnkcellmediatedantitumorimmunitybymediatingmacrophagepolarization
AT dongzhongjun myeloiddeletionofphosphoinositidedependentkinase1enhancesnkcellmediatedantitumorimmunitybymediatingmacrophagepolarization