9
views
0
recommends
+1 Recommend
2 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      SARS-CoV-2 viral persistence in lung alveolar macrophages is controlled by IFN-γ and NK cells

      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

          Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA generally becomes undetectable in upper airways after a few days or weeks postinfection. Here we used a model of viral infection in macaques to address whether SARS-CoV-2 persists in the body and which mechanisms regulate its persistence. Replication-competent virus was detected in bronchioalveolar lavage (BAL) macrophages beyond 6 months postinfection. Viral propagation in BAL macrophages occurred from cell to cell and was inhibited by interferon-γ (IFN-γ). IFN-γ production was strongest in BAL NKG2r +CD8 + T cells and NKG2A lo natural killer (NK) cells and was further increased in NKG2A lo NK cells after spike protein stimulation. However, IFN-γ production was impaired in NK cells from macaques with persisting virus. Moreover, IFN-γ also enhanced the expression of major histocompatibility complex (MHC)-E on BAL macrophages, possibly inhibiting NK cell-mediated killing. Macaques with less persisting virus mounted adaptive NK cells that escaped the MHC-E-dependent inhibition. Our findings reveal an interplay between NK cells and macrophages that regulated SARS-CoV-2 persistence in macrophages and was mediated by IFN-γ.

          Abstract

          Huot et al. show that interferon-γ (IFN-γ) regulates the persistence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in bronchoalveolar macrophages from cynomolgus macaques up to 18 months postinfection.

          Related collections

          Most cited references69

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

          Pathological inflammation in patients with COVID-19: a key role for monocytes and macrophages

          The COVID-19 pandemic caused by infection with SARS-CoV-2 has led to more than 200,000 deaths worldwide. Several studies have now established that the hyperinflammatory response induced by SARS-CoV-2 is a major cause of disease severity and death in infected patients. Macrophages are a population of innate immune cells that sense and respond to microbial threats by producing inflammatory molecules that eliminate pathogens and promote tissue repair. However, a dysregulated macrophage response can be damaging to the host, as is seen in the macrophage activation syndrome induced by severe infections, including in infections with the related virus SARS-CoV. Here we describe the potentially pathological roles of macrophages during SARS-CoV-2 infection and discuss ongoing and prospective therapeutic strategies to modulate macrophage activation in patients with COVID-19.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            HLA-E binds to natural killer cell receptors CD94/NKG2A, B and C.

            The protein HLA-E is a non-classical major histocompatibility complex (MHC) molecule of limited sequence variability. Its expression on the cell surface is regulated by the binding of peptides derived from the signal sequence of some other MHC class I molecules. Here we report the identification of ligands for HLA-E. We constructed tetramers in which recombinant HLA-E and beta2-microglobulin were refolded with an MHC leader-sequence peptide, biotinylated, and conjugated to phycoerythrin-labelled Extravidin. This HLA-E tetramer bound to natural killer (NK) cells and a small subset of T cells from peripheral blood. On transfectants, the tetramer bound to the CD94/NKG2A, CD94/NKGK2B and CD94/NKG2C NK cell receptors, but did not bind to the immunoglobulin family of NK cell receptors (KIR). Surface expression of HLA-E was enough to protect target cells from lysis by CD94/NKG2A+ NK-cell clones. A subset of HLA class I alleles has been shown to inhibit killing by CD94/NKG2A+ NK-cell clones. Only the HLA alleles that possess a leader peptide capable of upregulating HLA-E surface expression confer resistance to NK-cell-mediated lysis, implying that their action is mediated by HLA-E, the predominant ligand for the NK cell inhibitory receptor CD94/NKG2A.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Comparative pathogenesis of COVID-19, MERS, and SARS in a nonhuman primate model

              The current pandemic coronavirus, SARS-CoV-2, was recently identified in patients with an acute respiratory syndrome, COVID-19. To compare its pathogenesis with that of previously emerging coronaviruses, we inoculated cynomolgus macaques with SARS-CoV-2 or MERS-CoV and compared the pathology and virology with historical reports of SARS-CoV infections. In SARS-CoV-2-infected macaques, virus was excreted from nose and throat in the absence of clinical signs, and detected in type I and II pneumocytes in foci of diffuse alveolar damage and in ciliated epithelial cells of nasal, bronchial, and bronchiolar mucosae. In SARS-CoV-infection, lung lesions were typically more severe, while they were milder in MERS-CoV infection, where virus was detected mainly in type II pneumocytes. These data show that SARS-CoV-2 causes COVID-19-like disease in macaques, and provides a new model to test preventive and therapeutic strategies.
                Bookmark

                Author and article information

                Contributors
                nicolas.huot@pasteur.fr
                Journal
                Nat Immunol
                Nat Immunol
                Nature Immunology
                Nature Publishing Group US (New York )
                1529-2908
                1529-2916
                2 November 2023
                2 November 2023
                2023
                : 24
                : 12
                : 2068-2079
                Affiliations
                [1 ]GRID grid.508487.6, ISNI 0000 0004 7885 7602, Institut Pasteur, , Université Paris-Cité, HIV, Inflammation and Persistence Unit, ; Paris, France
                [2 ]GRID grid.508487.6, ISNI 0000 0004 7885 7602, Institut Pasteur, , Université Paris Cité, INSERM U1222, Humoral Immunology Unit, ; Paris, France
                [3 ]GRID grid.7429.8, ISNI 0000000121866389, Université Paris-Saclay, INSERM, CEA, Immunologie des Maladies Virales, Auto-Immunes, Hématologiques et Bactériennes (IMVA-HB/IDMIT/UMR1184), ; Fontenay-aux-Roses & Kremlin Bicêtre, France
                [4 ]GRID grid.508487.6, ISNI 0000 0004 7885 7602, Institut Pasteur, , Université Paris-Cité, Structural Virology Unit, CNRS UMR3569, ; Paris, France
                [5 ]GRID grid.38142.3c, ISNI 000000041936754X, Center for Virology and Vaccine Research, , Beth Israel Deaconess Medical Center, Harvard Medical School, ; Boston, MA USA
                [6 ]GRID grid.26009.3d, ISNI 0000 0004 1936 7961, Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Department of Surgery, , Duke University School of Medicine, ; Durham, NC USA
                [7 ]GRID grid.32224.35, ISNI 0000 0004 0386 9924, Ragon Institute of Massachusetts General Hospital, MIT, ; Cambridge, MA USA
                [8 ]GRID grid.412100.6, ISNI 0000 0001 0667 3730, Duke Research and Discovery at RTP, , Duke University Health System, ; Durham, NC USA
                Author information
                http://orcid.org/0000-0001-5982-7991
                http://orcid.org/0000-0002-1896-5092
                http://orcid.org/0000-0002-9953-7988
                http://orcid.org/0000-0003-3157-2557
                http://orcid.org/0000-0002-4928-4484
                Article
                1661
                10.1038/s41590-023-01661-4
                10681903
                37919524
                144526f8-dd72-4de5-b104-31ff86d81eed
                © The Author(s) 2023

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 15 April 2022
                : 26 September 2023
                Funding
                Funded by: FundRef https://doi.org/10.13039/100009060, Sidaction;
                Funded by: FundRef https://doi.org/10.13039/501100001665, Agence Nationale de la Recherche (French National Research Agency);
                Award ID: #20RR028-00
                Award ID: 11-INBS-0008
                Award ID: 10-EQPX-02-01
                Award ID: 10–INSB–04
                Award ID: #101003589
                Award ID: ANR-11-INBS-0008
                Award ID: ANR-10-EQPX-02-01
                Award Recipient :
                Funded by: FundRef https://doi.org/10.13039/501100004431, Fondation de France;
                Award ID: #00106077
                Award ID: #00106077
                Award Recipient :
                Funded by: FundRef https://doi.org/10.13039/100000002, U.S. Department of Health & Human Services | National Institutes of Health (NIH);
                Award ID: R01DK130472
                Award ID: R01AI143457
                Award ID: R01AI143457
                Award Recipient :
                Categories
                Article
                Custom metadata
                © Springer Nature America, Inc. 2023

                Immunology
                infection,sars-cov-2
                Immunology
                infection, sars-cov-2

                Comments

                Comment on this article

                scite_

                Similar content294

                Cited by6

                Most referenced authors1,214