CYP19A1 mediates severe SARS-CoV-2 disease outcome in males

  • Stephanie Stanelle-Bertram (Geteilte/r Erstautor/in)
  • Sebastian Beck (Geteilte/r Erstautor/in)
  • Nancy Kouassi Mounogou (Geteilte/r Erstautor/in)
  • Berfin Schaumburg (Geteilte/r Erstautor/in)
  • Fabian Stoll
  • Amirah Al Jawazneh
  • Zoé Schmal
  • Tian Bai
  • Martin Zickler
  • Georg Beythien
  • Kathrin Becker
  • Madeleine de la Roi
  • Fabian Heinrich
  • Claudia Schulz
  • Martina Sauter
  • Susanne Krasemann
  • Philine Lange
  • Axel Heinemann
  • Debby van Riel
  • Lonneke Leijten
  • Lisa Bauer
  • Thierry P P van den Bosch
  • Boaz Lopuhaä
  • Tobias Busche
  • Daniel Wibberg
  • Dirk Schaudien
  • Torsten Goldmann
  • Anna Lüttjohann
  • Jenny Ruschinski
  • Hanna Jania
  • Zacharias Müller
  • Vinicius Pinho Dos Reis
  • Vanessa Krupp-Buzimkic
  • Martin Wolff
  • Chiara Fallerini
  • Margherita Baldassarri
  • Simone Furini
  • Katrina Norwood
  • Christopher Käufer
  • Nina Schützenmeister
  • Maren von Köckritz-Blickwede
  • Maria Schroeder
  • Dominik Jarczak
  • Axel Nierhaus
  • Tobias Welte
  • Stefan Kluge
  • Alice C McHardy
  • Frank Sommer
  • Jörn Kalinowski
  • Susanne Krauss-Etschmann
  • Franziska Richter
  • Jan von der Thüsen
  • Wolfgang Baumgärtner
  • Karin Klingel
  • Benjamin Ondruschka
  • Alessandra Renieri
  • Gülsah Gabriel
  • GEN-COVID Multicenter Study Group

Abstract

Male sex represents one of the major risk factors for severe COVID-19 outcome. However, underlying mechanisms that mediate sex-dependent disease outcome are as yet unknown. Here, we identify the CYP19A1 gene encoding for the testosterone-to-estradiol metabolizing enzyme CYP19A1 (also known as aromatase) as a host factor that contributes to worsened disease outcome in SARS-CoV-2-infected males. We analyzed exome sequencing data obtained from a human COVID-19 cohort (n = 2,866) using a machine-learning approach and identify a CYP19A1-activity-increasing mutation to be associated with the development of severe disease in men but not women. We further analyzed human autopsy-derived lungs (n = 86) and detect increased pulmonary CYP19A1 expression at the time point of death in men compared with women. In the golden hamster model, we show that SARS-CoV-2 infection causes increased CYP19A1 expression in the lung that is associated with dysregulated plasma sex hormone levels and reduced long-term pulmonary function in males but not females. Treatment of SARS-CoV-2-infected hamsters with a clinically approved CYP19A1 inhibitor (letrozole) improves impaired lung function and supports recovery of imbalanced sex hormones specifically in males. Our study identifies CYP19A1 as a contributor to sex-specific SARS-CoV-2 disease outcome in males. Furthermore, inhibition of CYP19A1 by the clinically approved drug letrozole may furnish a new therapeutic strategy for individualized patient management and treatment.

Bibliografische Daten

OriginalspracheEnglisch
ISSN2666-3791
DOIs
StatusVeröffentlicht - 19.09.2023
PubMed 37572667