Your browser doesn't support javascript.
loading
An aberrant immune-epithelial progenitor niche drives viral lung sequelae.
Narasimhan, Harish; Cheon, In Su; Qian, Wei; Hu, Sheng'en Shawn; Parimon, Tanyalak; Li, Chaofan; Goplen, Nick; Wu, Yue; Wei, Xiaoqin; Son, Young Min; Fink, Elizabeth; de Almeida Santos, Gislane; Tang, Jinyi; Yao, Changfu; Muehling, Lyndsey; Canderan, Glenda; Kadl, Alexandra; Cannon, Abigail; Young, Samuel; Hannan, Riley; Bingham, Grace; Arish, Mohammed; Sen Chaudhari, Arka; Im, Jun Sub; Mattingly, Cameron L R; Pramoonjago, Patcharin; Marchesvsky, Alberto; Sturek, Jeffrey; Kohlmeier, Jacob E; Shim, Yun Michael; Woodfolk, Judith; Zang, Chongzhi; Chen, Peter; Sun, Jie.
  • Narasimhan H; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA.
  • Cheon IS; Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
  • Qian W; Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA.
  • Hu SS; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA.
  • Parimon T; Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
  • Li C; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA.
  • Goplen N; Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
  • Wu Y; Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA.
  • Wei X; Women's Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
  • Son YM; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
  • Fink E; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA.
  • de Almeida Santos G; Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
  • Tang J; Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA.
  • Yao C; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA.
  • Muehling L; Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
  • Canderan G; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA.
  • Kadl A; Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
  • Cannon A; Department of Systems Biotechnology, Chung-Ang University, Anseong, Korea.
  • Young S; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA.
  • Hannan R; Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
  • Bingham G; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA.
  • Arish M; Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
  • Sen Chaudhari A; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA.
  • Im JS; Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
  • Mattingly CLR; Women's Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
  • Pramoonjago P; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
  • Marchesvsky A; Division of Asthma, Allergy and Immunology, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
  • Sturek J; Division of Asthma, Allergy and Immunology, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
  • Kohlmeier JE; Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
  • Shim YM; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA.
  • Woodfolk J; Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
  • Zang C; Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA, USA.
  • Chen P; Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, USA.
  • Sun J; Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA.
Nature ; 2024 Sep 04.
Article en En | MEDLINE | ID: mdl-39232171
ABSTRACT
The long-term physiological consequences of respiratory viral infections, particularly in the aftermath of the COVID-19 pandemic-termed post-acute sequelae of SARS-CoV-2 (PASC)-are rapidly evolving into a major public health concern1-3. While the cellular and molecular aetiologies of these sequelae are poorly defined, increasing evidence implicates abnormal immune responses3-6 and/or impaired organ recovery7-9 after infection. However, the precise mechanisms that link these processes in the context of PASC remain unclear. Here, with insights from three cohorts of patients with respiratory PASC, we established a mouse model of post-viral lung disease and identified an aberrant immune-epithelial progenitor niche unique to fibroproliferation in respiratory PASC. Using spatial transcriptomics and imaging, we found a central role for lung-resident CD8+ T cell-macrophage interactions in impairing alveolar regeneration and driving fibrotic sequelae after acute viral pneumonia. Specifically, IFNγ and TNF derived from CD8+ T cells stimulated local macrophages to chronically release IL-1ß, resulting in the long-term maintenance of dysplastic epithelial progenitors and lung fibrosis. Notably, therapeutic neutralization of IFNγ + TNF or IL-1ß markedly improved alveolar regeneration and pulmonary function. In contrast to other approaches, which require early intervention10, we highlight therapeutic strategies to rescue fibrotic disease after the resolution of acute disease, addressing a current unmet need in the clinical management of PASC and post-viral disease.

Texto completo: 1 Banco de datos: MEDLINE Idioma: En Año: 2024 Tipo del documento: Article

Texto completo: 1 Banco de datos: MEDLINE Idioma: En Año: 2024 Tipo del documento: Article