Your browser doesn't support javascript.
loading
Oxysterols drive inflammation via GPR183 during influenza virus and SARS-CoV-2 infection
Cheng Xiang Foo; Stacey Bartlett; Keng Yih Chew; Minh Dao Ngo; Helle Bielefeldt-Ohmann; Buddhika Jayakody Arachchige; Benjamin Matthews; Sarah Reed; Ran Wang; Matthew J. Sweet; Lucy Burr; Jane E. Sinclair; Rhys Parry; Alexander Khromykh; Kirsty R. Short; Mette M. Rosenkilde; Katharina Ronacher.
Affiliation
  • Cheng Xiang Foo; Mater Research Institute - The University of Queensland
  • Stacey Bartlett; Mater Research Institute - The University of Queensland
  • Keng Yih Chew; The University of Queensland
  • Minh Dao Ngo; Mater Research Institute - The University of Queensland
  • Helle Bielefeldt-Ohmann; The University of Queensland
  • Buddhika Jayakody Arachchige; The University of Queensland
  • Benjamin Matthews; The University of Queensland
  • Sarah Reed; The University of Queensland
  • Ran Wang; Mater Research Institute - The University of Queensland
  • Matthew J. Sweet; The University of Queensland
  • Lucy Burr; Mater Research Institute - The University of Queensland
  • Jane E. Sinclair; The University of Queensland
  • Rhys Parry; The University of Queensland
  • Alexander Khromykh; The University of Queensland
  • Kirsty R. Short; The University of Queensland
  • Mette M. Rosenkilde; University of Copenhagen
  • Katharina Ronacher; Mater Research Institute - The University of Queensland
Preprint in English | bioRxiv | ID: ppbiorxiv-496214
ABSTRACT
RationaleSevere viral respiratory infections are often characterized by extensive myeloid cell infiltration and activation and persistent lung tissue injury. However, the immunological mechanisms driving excessive inflammation in the lung remain elusive. ObjectivesTo identify the mechanisms that drive immune cell recruitment in the lung during viral respiratory infections and identify novel drug targets to reduce inflammation and disease severity. MethodsPreclinical murine models of influenza virus and SARS-CoV-2 infection. ResultsOxidized cholesterols and the oxysterol-sensing receptor GPR183 were identified as drivers of monocyte-macrophage infiltration to the lung during influenza virus (IAV) and SARS-CoV-2 infections. Both IAV and SARS-CoV-2 infections upregulated the enzymes cholesterol 25-hydroxylase (CH25H) and cytochrome P450 family 7 subfamily member B1 (CYP7B1) in the lung, resulting in local production of the oxidized cholesterols 25-hydroxycholesterol and 7,25-dihydroxycholesterol (7,25-OHC). Loss-of-function mutation of GPR183, or treatment with a GPR183 antagonist, reduced macrophage infiltration and inflammatory cytokine production in the lungs of IAV- or SARS-CoV-2-infected mice. The GPR183 antagonist also significantly attenuated the severity of SARS-CoV-2 infection by reducing weight loss and viral loads. ConclusionThis study demonstrates that oxysterols drive inflammation in the lung and provides the first preclinical evidence for therapeutic benefit of targeting GPR183 during severe viral respiratory infections. Author SummaryViral infections trigger oxysterol production in the lung, attracting macrophages via GPR183. Blocking GPR183 reduced inflammation and disease severity in SARS-CoV-2 infection, making GPR183 a putative target for therapeutic intervention.
License
cc_by_nc_nd
Full text: Available Collection: Preprints Database: bioRxiv Type of study: Prognostic study Language: English Year: 2022 Document type: Preprint
Full text: Available Collection: Preprints Database: bioRxiv Type of study: Prognostic study Language: English Year: 2022 Document type: Preprint
...