CX3CR1-Expressing Immune Cells Infiltrate the Tumor Microenvironment and Promote Radiation Resistance in a Mouse Model of Lung Cancer

Introduction: Chemokine (C-X3-C Motif) Receptor 1 (CX3CR1) is present in a subset of the immune cells in the tumor microenvironment (TME) and plays an essential and diverse role in cancer progression. However, its potential function in the irradiated TME remains unknown. Materials and Methods: A mou...

Full description

Bibliographic Details
Main Authors: Tamar Ben-Mordechai, Yaacov R. Lawrence, Zvi Symon, Ariel Shimoni-Sebag, Uri Amit
Format: Article
Language:English
Published: MDPI AG 2023-11-01
Series:Cancers
Subjects:
Online Access:https://www.mdpi.com/2072-6694/15/22/5472
_version_ 1797459880263548928
author Tamar Ben-Mordechai
Yaacov R. Lawrence
Zvi Symon
Ariel Shimoni-Sebag
Uri Amit
author_facet Tamar Ben-Mordechai
Yaacov R. Lawrence
Zvi Symon
Ariel Shimoni-Sebag
Uri Amit
author_sort Tamar Ben-Mordechai
collection DOAJ
description Introduction: Chemokine (C-X3-C Motif) Receptor 1 (CX3CR1) is present in a subset of the immune cells in the tumor microenvironment (TME) and plays an essential and diverse role in cancer progression. However, its potential function in the irradiated TME remains unknown. Materials and Methods: A mouse lung cancer model was performed by subcutaneously inoculating Lewis Lung Carcinoma (LLC) cells expressing luciferase (Luc-2) and mCherry cells in CX3CR1<sup>GFP/GFP</sup>, CX3CR1<sup>DTR/+</sup>, and wild–type (WT) mice. Bioluminescence imaging, clonogenic assay, and flow cytometry were used to assess tumor progression, proliferation, and cell composition after radiation. Results: Radiation provoked a significant influx of CX3CR1-expressing immune cells, notably monocytes and macrophages, into the TME. Co-culturing irradiated LLC cells with CX3CR1-deficient monocytes, and macrophages resulted in reduced clonogenic survival and increased apoptosis of the cancer cells. Interestingly, deficiency of CX3CR1 in macrophages led to a redistribution of the irradiated LLC cells in the S-phase, parallel to increased expression of cyclin E1, required for cell cycle G1/S transition. In addition, the deficiency of CX3CR1 expression in macrophages altered the cytokine secretion with a decrease in interleukin 6, a crucial mediator of cancer cell survival and proliferation. Next, LLC cells were injected subcutaneously into CX3CR1<sup>DTR/+</sup> mice, sensitive to diphtheria toxin (DT), and WT mice. After injection, tumors were irradiated with 8 Gy, and mice were treated with DT, leading to conditional ablation of CX3CR1-expressing cells. After three weeks, CX3CR1-depleted mice displayed reduced tumor progression. Furthermore, combining the S-phase-specific chemotherapeutic gemcitabine with CX3CR1 cell ablation resulted in additional attenuation of tumor progression. Conclusions: CX3CR1-expressing mononuclear cells invade the TME after radiation therapy in a mouse lung cancer model. CX3CR1 cell depletion attenuates tumor progression following radiation and sensitizes the tumor to S–phase-specific chemotherapy. Thus, we propose a novel strategy to improve radiation sensitivity by targeting the CX3CR1-expressing immune cells.
first_indexed 2024-03-09T16:57:14Z
format Article
id doaj.art-fd18f8d0a2e441e28d4d40057f6f506c
institution Directory Open Access Journal
issn 2072-6694
language English
last_indexed 2024-03-09T16:57:14Z
publishDate 2023-11-01
publisher MDPI AG
record_format Article
series Cancers
spelling doaj.art-fd18f8d0a2e441e28d4d40057f6f506c2023-11-24T14:34:41ZengMDPI AGCancers2072-66942023-11-011522547210.3390/cancers15225472CX3CR1-Expressing Immune Cells Infiltrate the Tumor Microenvironment and Promote Radiation Resistance in a Mouse Model of Lung CancerTamar Ben-Mordechai0Yaacov R. Lawrence1Zvi Symon2Ariel Shimoni-Sebag3Uri Amit4Radiation Oncology Department, Chaim Sheba Medical Center, Ramat Gan 52621, IsraelRadiation Oncology Department, Chaim Sheba Medical Center, Ramat Gan 52621, IsraelRadiation Oncology Department, Chaim Sheba Medical Center, Ramat Gan 52621, IsraelRadiation Oncology Department, Chaim Sheba Medical Center, Ramat Gan 52621, IsraelRadiation Oncology Department, Tel Aviv Medical Center, Tel Aviv 64239, IsraelIntroduction: Chemokine (C-X3-C Motif) Receptor 1 (CX3CR1) is present in a subset of the immune cells in the tumor microenvironment (TME) and plays an essential and diverse role in cancer progression. However, its potential function in the irradiated TME remains unknown. Materials and Methods: A mouse lung cancer model was performed by subcutaneously inoculating Lewis Lung Carcinoma (LLC) cells expressing luciferase (Luc-2) and mCherry cells in CX3CR1<sup>GFP/GFP</sup>, CX3CR1<sup>DTR/+</sup>, and wild–type (WT) mice. Bioluminescence imaging, clonogenic assay, and flow cytometry were used to assess tumor progression, proliferation, and cell composition after radiation. Results: Radiation provoked a significant influx of CX3CR1-expressing immune cells, notably monocytes and macrophages, into the TME. Co-culturing irradiated LLC cells with CX3CR1-deficient monocytes, and macrophages resulted in reduced clonogenic survival and increased apoptosis of the cancer cells. Interestingly, deficiency of CX3CR1 in macrophages led to a redistribution of the irradiated LLC cells in the S-phase, parallel to increased expression of cyclin E1, required for cell cycle G1/S transition. In addition, the deficiency of CX3CR1 expression in macrophages altered the cytokine secretion with a decrease in interleukin 6, a crucial mediator of cancer cell survival and proliferation. Next, LLC cells were injected subcutaneously into CX3CR1<sup>DTR/+</sup> mice, sensitive to diphtheria toxin (DT), and WT mice. After injection, tumors were irradiated with 8 Gy, and mice were treated with DT, leading to conditional ablation of CX3CR1-expressing cells. After three weeks, CX3CR1-depleted mice displayed reduced tumor progression. Furthermore, combining the S-phase-specific chemotherapeutic gemcitabine with CX3CR1 cell ablation resulted in additional attenuation of tumor progression. Conclusions: CX3CR1-expressing mononuclear cells invade the TME after radiation therapy in a mouse lung cancer model. CX3CR1 cell depletion attenuates tumor progression following radiation and sensitizes the tumor to S–phase-specific chemotherapy. Thus, we propose a novel strategy to improve radiation sensitivity by targeting the CX3CR1-expressing immune cells.https://www.mdpi.com/2072-6694/15/22/5472radiationimmunotherapycytokineslung cancer
spellingShingle Tamar Ben-Mordechai
Yaacov R. Lawrence
Zvi Symon
Ariel Shimoni-Sebag
Uri Amit
CX3CR1-Expressing Immune Cells Infiltrate the Tumor Microenvironment and Promote Radiation Resistance in a Mouse Model of Lung Cancer
Cancers
radiation
immunotherapy
cytokines
lung cancer
title CX3CR1-Expressing Immune Cells Infiltrate the Tumor Microenvironment and Promote Radiation Resistance in a Mouse Model of Lung Cancer
title_full CX3CR1-Expressing Immune Cells Infiltrate the Tumor Microenvironment and Promote Radiation Resistance in a Mouse Model of Lung Cancer
title_fullStr CX3CR1-Expressing Immune Cells Infiltrate the Tumor Microenvironment and Promote Radiation Resistance in a Mouse Model of Lung Cancer
title_full_unstemmed CX3CR1-Expressing Immune Cells Infiltrate the Tumor Microenvironment and Promote Radiation Resistance in a Mouse Model of Lung Cancer
title_short CX3CR1-Expressing Immune Cells Infiltrate the Tumor Microenvironment and Promote Radiation Resistance in a Mouse Model of Lung Cancer
title_sort cx3cr1 expressing immune cells infiltrate the tumor microenvironment and promote radiation resistance in a mouse model of lung cancer
topic radiation
immunotherapy
cytokines
lung cancer
url https://www.mdpi.com/2072-6694/15/22/5472
work_keys_str_mv AT tamarbenmordechai cx3cr1expressingimmunecellsinfiltratethetumormicroenvironmentandpromoteradiationresistanceinamousemodeloflungcancer
AT yaacovrlawrence cx3cr1expressingimmunecellsinfiltratethetumormicroenvironmentandpromoteradiationresistanceinamousemodeloflungcancer
AT zvisymon cx3cr1expressingimmunecellsinfiltratethetumormicroenvironmentandpromoteradiationresistanceinamousemodeloflungcancer
AT arielshimonisebag cx3cr1expressingimmunecellsinfiltratethetumormicroenvironmentandpromoteradiationresistanceinamousemodeloflungcancer
AT uriamit cx3cr1expressingimmunecellsinfiltratethetumormicroenvironmentandpromoteradiationresistanceinamousemodeloflungcancer