7
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      IFNAR1 Deficiency Impairs Immunostimulatory Properties of Neutrophils in Tumor-Draining Lymph Nodes

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Tumor-draining lymph nodes (TDLNs) are the first organs where the metastatic spread of different types of cancer, including head and neck cancer (HNC), occurs and have therefore high prognostic relevance. Moreover, first anti-cancer immune responses have been shown to be initiated in such LNs via tumor-educated myeloid cells. Among myeloid cells present in TDLNs, neutrophils represent a valuable population and considerably participate in the activation of effector lymphocytes there. Tumor-supportive or tumor-inhibiting activity of neutrophils strongly depends on the surrounding microenvironment. Thus, type I interferon (IFN) availability has been shown to prime anti-tumor activity of these cells. In accordance, mice deficient in type I IFNs show elevated tumor growth and metastatic spread, accompanied by the pro-tumoral neutrophil bias. To reveal the mechanism responsible for this phenomenon, we have studied here the influence of defective type I IFN signaling on the immunoregulatory activity of neutrophils in TDLNs. Live imaging of such LNs was performed using two-photon microscopy in a transplantable murine HNC model. Catchup IVM-red and Ifnar1 -/- (type I IFN receptor- deficient) Catchup IVM-red mice were used to visualize neutrophils and to assess their interaction with T-cells in vivo. We have evaluated spatiotemporal patterns of neutrophil/T-cell interactions in LNs in the context of type I interferon receptor (IFNAR1) availability in tumor-free and tumor-bearing animals. Moreover, phenotypic and functional analyses were performed to further characterize the mechanisms regulating neutrophil immunoregulatory capacity. We demonstrated that inactive IFNAR1 leads to elevated accumulation of neutrophils in TDLNs. However, these neutrophils show significantly impaired capacity to interact with and to stimulate T-cells. As a result, a significant reduction of contacts between neutrophils and T lymphocytes is observed, with further impairment of T-cell proliferation and activation. This possibly contributes to the enhanced tumor growth in Ifnar1 -/- mice. In agreement with this, IFNAR1-independent activation of downstream IFN signaling using IFN-λ improved the immunostimulatory capacity of neutrophils in TDLNs and contributed to the suppression of tumor growth. Our results suggest that functional type I IFN signaling is essential for neutrophil immunostimulatory capacity and that stimulation of this signaling may provide a therapeutic opportunity in head and neck cancer patients.

          Related collections

          Most cited references56

          • Record: found
          • Abstract: found
          • Article: not found

          Neutrophils in cancer: neutral no more.

          Neutrophils are indispensable antagonists of microbial infection and facilitators of wound healing. In the cancer setting, a newfound appreciation for neutrophils has come into view. The traditionally held belief that neutrophils are inert bystanders is being challenged by the recent literature. Emerging evidence indicates that tumours manipulate neutrophils, sometimes early in their differentiation process, to create diverse phenotypic and functional polarization states able to alter tumour behaviour. In this Review, we discuss the involvement of neutrophils in cancer initiation and progression, and their potential as clinical biomarkers and therapeutic targets.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Polarization of tumor-associated neutrophil phenotype by TGF-beta: "N1" versus "N2" TAN.

            TGF-beta blockade significantly slows tumor growth through many mechanisms, including activation of CD8(+) T cells and macrophages. Here, we show that TGF-beta blockade also increases neutrophil-attracting chemokines, resulting in an influx of CD11b(+)/Ly6G(+) tumor-associated neutrophils (TANs) that are hypersegmented, more cytotoxic to tumor cells, and express higher levels of proinflammatory cytokines. Accordingly, following TGF-beta blockade, depletion of these neutrophils significantly blunts antitumor effects of treatment and reduces CD8(+) T cell activation. In contrast, in control tumors, neutrophil depletion decreases tumor growth and results in more activated CD8(+) T cells intratumorally. Together, these data suggest that TGF-beta within the tumor microenvironment induces a population of TAN with a protumor phenotype. TGF-beta blockade results in the recruitment and activation of TANs with an antitumor phenotype.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors.

              Spontaneous T cell responses against tumors occur frequently and have prognostic value in patients. The mechanism of innate immune sensing of immunogenic tumors leading to adaptive T cell responses remains undefined, although type I interferons (IFNs) are implicated in this process. We found that spontaneous CD8(+) T cell priming against tumors was defective in mice lacking stimulator of interferon genes complex (STING), but not other innate signaling pathways, suggesting involvement of a cytosolic DNA sensing pathway. In vitro, IFN-? production and dendritic cell activation were triggered by tumor-cell-derived DNA, via cyclic-GMP-AMP synthase (cGAS), STING, and interferon regulatory factor 3 (IRF3). In the tumor microenvironment in vivo, tumor cell DNA was detected within host antigen-presenting cells, which correlated with STING pathway activation and IFN-? production. Our results demonstrate that a major mechanism for innate immune sensing of cancer occurs via the host STING pathway, with major implications for cancer immunotherapy. Copyright © 2014 Elsevier Inc. All rights reserved.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Immunol
                Front Immunol
                Front. Immunol.
                Frontiers in Immunology
                Frontiers Media S.A.
                1664-3224
                27 June 2022
                2022
                : 13
                : 878959
                Affiliations
                [1] 1 Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen , Essen, Germany
                [2] 2 Institute for Experimental Immunology and Imaging, University Duisburg-Essen , Essen, Germany
                [3] 3 Institute for Laboratory Animal Science, Institute of Immunology, Hannover Medical School , Hannover, Germany
                [4] 4 Institute for Virology, University Hospital Essen, University Duisburg-Essen , Essen, Germany
                [5] 5 Biospectroscopy Research Department, Institut für Analytische Wissenschaften (ISAS) e.V. , Dortmund, Germany
                [6] 6 German Cancer Consortium (DKTK) partner site Düsseldorf/Essen , Essen, Germany
                Author notes

                Edited by: Susanna Mandruzzato, University Hospital of Padua, Italy

                Reviewed by: Giovanna Schiavoni, National Institute of Health (ISS), Italy; Xianda Zhao, University of Minnesota, United States; Carlotta Tacconi, University of Milan, Italy

                *Correspondence: Jadwiga Jablonska, jadwiga.jablonska@ 123456uk-essen.de

                †These authors have contributed equally to this work and share the first authorship

                This article was submitted to Cancer Immunity and Immunotherapy, a section of the journal Frontiers in Immunology

                Article
                10.3389/fimmu.2022.878959
                9271705
                35833131
                99b6f173-ebb4-4682-bd46-e8a8ae6a3b2d
                Copyright © 2022 Hussain, Domnich, Bordbari, Pylaeva, Siakaeva, Spyra, Ozel, Droege, Squire, Lienenklaus, Sutter, Hasenberg, Gunzer, Lang and Jablonska

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 18 February 2022
                : 30 May 2022
                Page count
                Figures: 5, Tables: 0, Equations: 1, References: 56, Pages: 13, Words: 7628
                Funding
                Funded by: Deutsche Forschungsgemeinschaft , doi 10.13039/501100001659;
                Award ID: DFG/JA 2461/5-1, DFG/JA 2461/2-1
                Funded by: Deutsche Krebshilfe , doi 10.13039/501100005972;
                Award ID: 70113112
                Categories
                Immunology
                Original Research

                Immunology
                ifnar1,interferons,neutrophils,tumor-draining lymph nodes,neutrophils/t-cells interactions,intravital microscopy,hnc (head and neck cancer)

                Comments

                Comment on this article