Your browser doesn't support javascript.
loading
Host Derivation of Sindbis Virus Influences Mammalian Type I Interferon Response to Infection.
Crawford, John M; Buechlein, Aaron M; Moline, Davis A; Rusch, Douglas B; Hardy, Richard W.
Afiliación
  • Crawford JM; Department of Biology, Indiana University, Bloomington, IN 47405, USA.
  • Buechlein AM; Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN 47405, USA.
  • Moline DA; Department of Biology, Indiana University, Bloomington, IN 47405, USA.
  • Rusch DB; Center for Genomics and Bioinformatics, Indiana University, Bloomington, IN 47405, USA.
  • Hardy RW; Department of Biology, Indiana University, Bloomington, IN 47405, USA.
Viruses ; 15(8)2023 08 03.
Article en En | MEDLINE | ID: mdl-37632027
ABSTRACT
Arboviruses are defined by their ability to replicate in both mosquito vectors and mammalian hosts. There is good evidence that arboviruses "prime" their progeny for infection of the next host, such as via differential glycosylation of their outer glycoproteins or packaging of host ribosomal subunits. We and others have previously shown that mosquito-derived viruses more efficiently infect mammalian cells than mammalian-derived viruses. These observations are consistent with arboviruses acquiring host-specific adaptations, and we hypothesized that a virus derived from either the mammalian host or mosquito vector elicits different responses when infecting the mammalian host. Here, we perform an RNA-sequencing analysis of the transcriptional response of Human Embryonic Kidney 293 (HEK-293) cells to infection with either mosquito (Aedes albopictus, C7/10)- or mammalian (Baby Hamster Kidney, BHK-21)-derived Sindbis virus (SINV). We show that the C7/10-derived virus infection leads to a more robust transcriptional response in HEK-293s compared to infection with the BHK-derived virus. Surprisingly, despite more efficient infection, we found an increase in interferon-ß (IFN-ß) and interferon-stimulated gene (ISG) transcripts in response to the C7/10-derived virus infection versus the BHK-derived virus infection. However, translation of interferon-stimulated genes was lower in HEK-293s infected with the C7/10-derived virus, starkly contrasting with the transcriptional response. This inhibition of ISG translation is reflective of a more rapid overall shut-off of host cell translation following infection with the C7/10-derived virus. Finally, we show that the C7/10-derived virus infection of HEK-293 cells leads to elevated levels of phosphorylated eukaryotic translation elongation factor-2 (eEF2), identifying a potential mechanism leading to the more rapid shut-off of host translation. We postulate that the rapid shut-off of host translation in mammalian cells infected with the mosquito-derived virus acts to counter the IFN-ß-stimulated transcriptional response.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Interferón Tipo I / Aedes Tipo de estudio: Prognostic_studies Límite: Animals / Humans / Infant Idioma: En Revista: Viruses Año: 2023 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Interferón Tipo I / Aedes Tipo de estudio: Prognostic_studies Límite: Animals / Humans / Infant Idioma: En Revista: Viruses Año: 2023 Tipo del documento: Article País de afiliación: Estados Unidos