Cargando…

Nitric Oxide Circumvents Virus-Mediated Metabolic Regulation during Human Cytomegalovirus Infection

Nitric oxide is a versatile and critical effector molecule that can modulate many cellular functions. Although recognized as a regulator of infections, the inhibitory mechanism of nitric oxide against human cytomegalovirus (HCMV) replication remains elusive. We demonstrate that nitric oxide attenuat...

Descripción completa

Detalles Bibliográficos
Autores principales: Mokry, Rebekah L., Schumacher, Megan L., Hogg, Neil, Terhune, Scott S.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: American Society for Microbiology 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7773989/
https://www.ncbi.nlm.nih.gov/pubmed/33323506
http://dx.doi.org/10.1128/mBio.02630-20
_version_ 1783630169481674752
author Mokry, Rebekah L.
Schumacher, Megan L.
Hogg, Neil
Terhune, Scott S.
author_facet Mokry, Rebekah L.
Schumacher, Megan L.
Hogg, Neil
Terhune, Scott S.
author_sort Mokry, Rebekah L.
collection PubMed
description Nitric oxide is a versatile and critical effector molecule that can modulate many cellular functions. Although recognized as a regulator of infections, the inhibitory mechanism of nitric oxide against human cytomegalovirus (HCMV) replication remains elusive. We demonstrate that nitric oxide attenuates viral replication by interfering with HCMV-mediated modulation of several cellular processes. Nitric oxide exposure reduced HCMV genome synthesis and infectious viral progeny with cell-type-dependent differences observed. Mitochondrial respiration was severely reduced in both uninfected and HCMV-infected cells during exposure with little impact on ATP levels indicating changes in cellular metabolism. Metabolomics identified significantly altered small molecules in multiple pathways during nitric oxide exposure including nucleotide biosynthesis, tricarboxylic acid (TCA) cycle, and glutamine metabolism. Glutathione metabolites were increased coinciding with a reduction in the glutathione precursor glutamine. This shift was accompanied by increased antioxidant enzymes. Glutamine deprivation mimicked defects in HCMV replication and mitochondrial respiration observed during nitric oxide exposure. These data suggest that nitric oxide limits glutaminolysis by shuttling glutamine to glutathione synthesis. In addition, lipid intermediates were severely altered, which likely contributes to the observed increase in defective viral particles. Nitric oxide disrupts multiple cellular processes, and we had limited success in rescuing replication defects by supplementing with metabolic intermediates. Our studies indicate that nitric oxide attenuation of HCMV is multifactorial with interference in viral manipulation of cellular metabolism playing a central role.
format Online
Article
Text
id pubmed-7773989
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher American Society for Microbiology
record_format MEDLINE/PubMed
spelling pubmed-77739892021-01-05 Nitric Oxide Circumvents Virus-Mediated Metabolic Regulation during Human Cytomegalovirus Infection Mokry, Rebekah L. Schumacher, Megan L. Hogg, Neil Terhune, Scott S. mBio Research Article Nitric oxide is a versatile and critical effector molecule that can modulate many cellular functions. Although recognized as a regulator of infections, the inhibitory mechanism of nitric oxide against human cytomegalovirus (HCMV) replication remains elusive. We demonstrate that nitric oxide attenuates viral replication by interfering with HCMV-mediated modulation of several cellular processes. Nitric oxide exposure reduced HCMV genome synthesis and infectious viral progeny with cell-type-dependent differences observed. Mitochondrial respiration was severely reduced in both uninfected and HCMV-infected cells during exposure with little impact on ATP levels indicating changes in cellular metabolism. Metabolomics identified significantly altered small molecules in multiple pathways during nitric oxide exposure including nucleotide biosynthesis, tricarboxylic acid (TCA) cycle, and glutamine metabolism. Glutathione metabolites were increased coinciding with a reduction in the glutathione precursor glutamine. This shift was accompanied by increased antioxidant enzymes. Glutamine deprivation mimicked defects in HCMV replication and mitochondrial respiration observed during nitric oxide exposure. These data suggest that nitric oxide limits glutaminolysis by shuttling glutamine to glutathione synthesis. In addition, lipid intermediates were severely altered, which likely contributes to the observed increase in defective viral particles. Nitric oxide disrupts multiple cellular processes, and we had limited success in rescuing replication defects by supplementing with metabolic intermediates. Our studies indicate that nitric oxide attenuation of HCMV is multifactorial with interference in viral manipulation of cellular metabolism playing a central role. American Society for Microbiology 2020-12-15 /pmc/articles/PMC7773989/ /pubmed/33323506 http://dx.doi.org/10.1128/mBio.02630-20 Text en Copyright © 2020 Mokry et al. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Research Article
Mokry, Rebekah L.
Schumacher, Megan L.
Hogg, Neil
Terhune, Scott S.
Nitric Oxide Circumvents Virus-Mediated Metabolic Regulation during Human Cytomegalovirus Infection
title Nitric Oxide Circumvents Virus-Mediated Metabolic Regulation during Human Cytomegalovirus Infection
title_full Nitric Oxide Circumvents Virus-Mediated Metabolic Regulation during Human Cytomegalovirus Infection
title_fullStr Nitric Oxide Circumvents Virus-Mediated Metabolic Regulation during Human Cytomegalovirus Infection
title_full_unstemmed Nitric Oxide Circumvents Virus-Mediated Metabolic Regulation during Human Cytomegalovirus Infection
title_short Nitric Oxide Circumvents Virus-Mediated Metabolic Regulation during Human Cytomegalovirus Infection
title_sort nitric oxide circumvents virus-mediated metabolic regulation during human cytomegalovirus infection
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7773989/
https://www.ncbi.nlm.nih.gov/pubmed/33323506
http://dx.doi.org/10.1128/mBio.02630-20
work_keys_str_mv AT mokryrebekahl nitricoxidecircumventsvirusmediatedmetabolicregulationduringhumancytomegalovirusinfection
AT schumachermeganl nitricoxidecircumventsvirusmediatedmetabolicregulationduringhumancytomegalovirusinfection
AT hoggneil nitricoxidecircumventsvirusmediatedmetabolicregulationduringhumancytomegalovirusinfection
AT terhunescotts nitricoxidecircumventsvirusmediatedmetabolicregulationduringhumancytomegalovirusinfection