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

      A spatially restricted fibrotic niche in pulmonary fibrosis is sustained by M-CSF/M-CSFR signalling in monocyte-derived alveolar macrophages

      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

          Ontologically distinct populations of macrophages differentially contribute to organ fibrosis through unknown mechanisms.

          We applied lineage tracing, single-cell RNA sequencing and single-molecule fluorescence in situ hybridisation to a spatially restricted model of asbestos-induced pulmonary fibrosis.

          We demonstrate that tissue-resident alveolar macrophages, tissue-resident peribronchial and perivascular interstitial macrophages, and monocyte-derived alveolar macrophages are present in the fibrotic niche. Deletion of monocyte-derived alveolar macrophages but not tissue-resident alveolar macrophages ameliorated asbestos-induced lung fibrosis. Monocyte-derived alveolar macrophages were specifically localised to fibrotic regions in the proximity of fibroblasts where they expressed molecules known to drive fibroblast proliferation, including platelet-derived growth factor subunit A. Using single-cell RNA sequencing and spatial transcriptomics in both humans and mice, we identified macrophage colony-stimulating factor receptor (M-CSFR) signalling as one of the novel druggable targets controlling self-maintenance and persistence of these pathogenic monocyte-derived alveolar macrophages. Pharmacological blockade of M-CSFR signalling led to the disappearance of monocyte-derived alveolar macrophages and ameliorated fibrosis.

          Our findings suggest that inhibition of M-CSFR signalling during fibrosis disrupts an essential fibrotic niche that includes monocyte-derived alveolar macrophages and fibroblasts during asbestos-induced fibrosis.

          Abstract

          Monocyte-derived alveolar macrophages orchestrate the development of the fibrotic niche, causally related to fibrosis and maintained via M-CSF/M-CSFR signalling http://bit.ly/2nDjS20

          Related collections

          Most cited references33

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

          Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage

          Tissue fibrosis is a major cause of mortality that results from the deposition of matrix proteins by an activated mesenchyme. Macrophages accumulate in fibrosis, but the role of specific subgroups in supporting fibrogenesis has not been investigated in vivo. Here we used single-cell RNA sequencing (scRNA-seq) to characterize the heterogeneity of macrophages in bleomycin-induced lung fibrosis in mice. A novel computational framework for the annotation of scRNA-seq by reference to bulk transcriptomes (SingleR) enabled the subclustering of macrophages and revealed a disease-associated subgroup with a transitional gene expression profile intermediate between monocyte-derived and alveolar macrophages. These CX3CR1+SiglecF+ transitional macrophages localized to the fibrotic niche and had a profibrotic effect in vivo. Human orthologues of genes expressed by the transitional macrophages were upregulated in samples from patients with idiopathic pulmonary fibrosis. Thus, we have identified a pathological subgroup of transitional macrophages that are required for the fibrotic response to injury.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            Single-Cell Transcriptomic Analysis of Human Lung Provides Insights into the Pathobiology of Pulmonary Fibrosis

            Rationale: The contributions of diverse cell populations in the human lung to pulmonary fibrosis pathogenesis are poorly understood. Single-cell RNA sequencing can reveal changes within individual cell populations during pulmonary fibrosis that are important for disease pathogenesis. Objectives: To determine whether single-cell RNA sequencing can reveal disease-related heterogeneity within alveolar macrophages, epithelial cells, or other cell types in lung tissue from subjects with pulmonary fibrosis compared with control subjects. Methods: We performed single-cell RNA sequencing on lung tissue obtained from eight transplant donors and eight recipients with pulmonary fibrosis and on one bronchoscopic cryobiospy sample from a patient with idiopathic pulmonary fibrosis. We validated these data using in situ RNA hybridization, immunohistochemistry, and bulk RNA-sequencing on flow-sorted cells from 22 additional subjects. Measurements and Main Results: We identified a distinct, novel population of profibrotic alveolar macrophages exclusively in patients with fibrosis. Within epithelial cells, the expression of genes involved in Wnt secretion and response was restricted to nonoverlapping cells. We identified rare cell populations including airway stem cells and senescent cells emerging during pulmonary fibrosis. We developed a web-based tool to explore these data. Conclusions: We generated a single-cell atlas of pulmonary fibrosis. Using this atlas, we demonstrated heterogeneity within alveolar macrophages and epithelial cells from subjects with pulmonary fibrosis. These results support the feasibility of discovery-based approaches using next-generation sequencing technologies to identify signaling pathways for targeting in the development of personalized therapies for patients with pulmonary fibrosis.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells.

              Blockade of colony-stimulating factor-1 (CSF-1) limits macrophage infiltration and improves response of mammary carcinomas to chemotherapy. Herein we identify interleukin (IL)-10 expression by macrophages as the critical mediator of this phenotype. Infiltrating macrophages were the primary source of IL-10 within tumors, and therapeutic blockade of IL-10 receptor (IL-10R) was equivalent to CSF-1 neutralization in enhancing primary tumor response to paclitaxel and carboplatin. Improved response to chemotherapy was CD8(+) T cell-dependent, but IL-10 did not directly suppress CD8(+) T cells or alter macrophage polarization. Instead, IL-10R blockade increased intratumoral dendritic cell expression of IL-12, which was necessary for improved outcomes. In human breast cancer, expression of IL12A and cytotoxic effector molecules were predictive of pathological complete response rates to paclitaxel.
                Bookmark

                Author and article information

                Journal
                Eur Respir J
                Eur. Respir. J
                ERJ
                erj
                The European Respiratory Journal
                European Respiratory Society
                0903-1936
                1399-3003
                January 2020
                16 January 2020
                : 55
                : 1
                : 1900646
                Affiliations
                [1 ]Division of Pulmonary and Critical Care Medicine, Dept of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
                [2 ]Dept of Respiratory Medicine, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
                [3 ]Dept of Medicine, Section of Pulmonary and Critical Care, The University of Chicago, Chicago, IL, USA
                [4 ]Dept of Medicine, Division of Pulmonary and Critical Care Medicine, Jesse Brown VA Medical Center, Chicago, IL, USA
                [5 ]Center for Advanced Microscopy, Robert H. Lurie Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
                [6 ]Division of Rheumatology, Dept of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
                [7 ]These authors contributed equally to this work
                [8 ]These authors contributed equally to this work
                Author notes
                Alexander V. Misharin, Division of Pulmonary and Critical Care Medicine, Dept of Medicine, Feinberg School of Medicine, Northwestern University, Simpson Querrey Biomedical Research Center, 303 E. Superior Street, Chicago, IL 60611, USA. E-mail: a-misharin@ 123456northwestern.edu
                Author information
                https://orcid.org/0000-0002-6435-6001
                https://orcid.org/0000-0003-0912-7617
                Article
                ERJ-00646-2019
                10.1183/13993003.00646-2019
                6962769
                31601718
                b7c61409-bb64-4e59-af88-53d323837cd8
                Copyright ©ERS 2020

                This article is open access and distributed under the terms of the Creative Commons Attribution Non-Commercial Licence 4.0.

                History
                : 01 April 2019
                : 26 September 2019
                Funding
                Funded by: National Institute on Aging, open-funder-registry 10.13039/100000049;
                Award ID: AG049665
                Funded by: National Heart, Lung, and Blood Institute, open-funder-registry 10.13039/100000050;
                Award ID: HL135124
                Funded by: The Veterans Administration
                Award ID: BX000201
                Funded by: National Institute of Allergy and Infectious Diseases, open-funder-registry 10.13039/100000060;
                Award ID: AI135964
                Funded by: U.S. Department of Defense, open-funder-registry 10.13039/100000005;
                Award ID: PR141319
                Categories
                Original Articles
                Interstitial Lung Disease and Basic Science
                13

                Respiratory medicine
                Respiratory medicine

                Comments

                Comment on this article