Glycolytic interference blocks influenza A virus propagation by impairing viral polymerase-driven synthesis of genomic vRNA

Influenza A virus (IAV), like any other virus, provokes considerable modifications of its host cell’s metabolism. This includes a substantial increase in the uptake as well as the metabolization of glucose. Although it is known for quite some time that suppression of glucose metabolism restricts vir...

Verfasser: Kleinehr, Jens
Schöfbänker, Michael
Daniel, Katharina
Günl, Franziska
Mohamed, Fakry Fahmy
Janowski, Josua
Brunotte, Linda
Börgeling, Yvonne
Liebmann, Marie
Behrens, Matthias
Gerdemann, Andrea
Klotz, Luisa Hildegard
Esselen, Melanie
Humpf, Hans-Ulrich
Ludwig, Stephan
Hrincius, Eike-Roman
FB/Einrichtung:FB 05: Medizinische Fakultät
FB 13: Biologie
Dokumenttypen:Artikel
Medientypen:Text
Erscheinungsdatum:2023
Publikation in MIAMI:28.08.2023
Datum der letzten Änderung:28.08.2023
Angaben zur Ausgabe:[Electronic ed.]
Quelle:PLoS Pathogens 19 (2023) 7, e1010986, 1-34
Fachgebiet (DDC):610: Medizin und Gesundheit
Lizenz:CC BY 4.0
Sprache:English
Förderung:Finanziert durch den Open-Access-Publikationsfonds der Westfälischen Wilhelms-Universität Münster (WWU Münster).
Format:PDF-Dokument
URN:urn:nbn:de:hbz:6-09938603761
Weitere Identifikatoren:DOI: 10.17879/09938611393
Permalink:https://nbn-resolving.de/urn:nbn:de:hbz:6-09938603761
Verwandte Dokumente:
Onlinezugriff:10.1371_journal.ppat.1010986.pdf

Influenza A virus (IAV), like any other virus, provokes considerable modifications of its host cell’s metabolism. This includes a substantial increase in the uptake as well as the metabolization of glucose. Although it is known for quite some time that suppression of glucose metabolism restricts virus replication, the exact molecular impact on the viral life cycle remained enigmatic so far. Using 2-deoxy-d-glucose (2-DG) we examined how well inhibition of glycolysis is tolerated by host cells and which step of the IAV life cycle is affected. We observed that effects induced by 2-DG are reversible and that cells can cope with relatively high concentrations of the inhibitor by compensating the loss of glycolytic activity by upregulating other metabolic pathways. Moreover, mass spectrometry data provided information on various metabolic modifications induced by either the virus or agents interfering with glycolysis. In the presence of 2-DG viral titers were significantly reduced in a dose-dependent manner. The supplementation of direct or indirect glycolysis metabolites led to a partial or almost complete reversion of the inhibitory effect of 2-DG on viral growth and demonstrated that indeed the inhibition of glycolysis and not of N-linked glycosylation was responsible for the observed phenotype. Importantly, we could show via conventional and strand-specific qPCR that the treatment with 2-DG led to a prolonged phase of viral mRNA synthesis while the accumulation of genomic vRNA was strongly reduced. At the same time, minigenome assays showed no signs of a general reduction of replicative capacity of the viral polymerase. Therefore, our data suggest that the significant reduction in IAV replication by glycolytic interference occurs mainly due to an impairment of the dynamic regulation of the viral polymerase which conveys the transition of the enzyme's function from transcription to replication.