Cargando…

Intravital imaging of three different microvascular beds in SARS-CoV-2–infected mice

Severe acute respiratory syndrome coronavirus–2 (SARS-CoV-2) enters the respiratory tract, where it infects the alveoli epithelial lining. However, patients have sequelae that extend well beyond the alveoli into the pulmonary vasculature and, perhaps, beyond to the brain and other organs. Because of...

Descripción completa

Detalles Bibliográficos
Autores principales: Castanheira, Fernanda V. S., Nguyen, Rita, Willson, Michelle, Davoli-Ferreira, Marcela, David, Bruna A., Kelly, Margaret M., Lee, Woo-Yong, Kratofil, Rachel M., Zhang, Wen X., Bui-Marinos, Maxwell, Corcoran, Jennifer A., Kubes, Paul
Formato: Online Artículo Texto
Lenguaje:English
Publicado: The American Society of Hematology 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10284260/
https://www.ncbi.nlm.nih.gov/pubmed/37307197
http://dx.doi.org/10.1182/bloodadvances.2022009430
_version_ 1785061369263947776
author Castanheira, Fernanda V. S.
Nguyen, Rita
Willson, Michelle
Davoli-Ferreira, Marcela
David, Bruna A.
Kelly, Margaret M.
Lee, Woo-Yong
Kratofil, Rachel M.
Zhang, Wen X.
Bui-Marinos, Maxwell
Corcoran, Jennifer A.
Kubes, Paul
author_facet Castanheira, Fernanda V. S.
Nguyen, Rita
Willson, Michelle
Davoli-Ferreira, Marcela
David, Bruna A.
Kelly, Margaret M.
Lee, Woo-Yong
Kratofil, Rachel M.
Zhang, Wen X.
Bui-Marinos, Maxwell
Corcoran, Jennifer A.
Kubes, Paul
author_sort Castanheira, Fernanda V. S.
collection PubMed
description Severe acute respiratory syndrome coronavirus–2 (SARS-CoV-2) enters the respiratory tract, where it infects the alveoli epithelial lining. However, patients have sequelae that extend well beyond the alveoli into the pulmonary vasculature and, perhaps, beyond to the brain and other organs. Because of the dynamic events within blood vessels, histology does not report platelet and neutrophil behavior. Because of the rapid nontranscriptional response of these cells, neither single-cell RNA sequencing nor proteomics report robustly on their critical behaviors. We used intravital microscopy in level-3 containment to examine the pathogenesis of SARS-CoV-2 within 3 organs in mice expressing human angiotensin converting enzyme 2 (ACE-2) ubiquitously (CAG-AC-70) or on epithelium (K18-promoter). Using a neon-green SARS-CoV-2, we observed both the epithelium and endothelium infected in AC70 mice but only the epithelium in K18 mice. There were increased neutrophils in the microcirculation but not in the alveoli of the lungs of AC70 mice. Platelets formed large aggregates in the pulmonary capillaries. Despite only neurons being infected within the brain, profound neutrophil adhesion forming the nidus of large platelet aggregates were observed in the cerebral microcirculation, with many nonperfused microvessels. Neutrophils breached the brain endothelial layer associated with a significant disruption of the blood-brain-barrier. Despite ubiquitous ACE-2 expression, CAG-AC-70 mice had very small increases in blood cytokine, no increase in thrombin, no infected circulating cells, and no liver involvement suggesting limited systemic effects. In summary, our imaging of SARS-CoV-2–infected mice gave direct evidence that there is a significant perturbation locally in the lung and brain microcirculation induced by local viral infection leading to increased local inflammation and thrombosis in these organs.
format Online
Article
Text
id pubmed-10284260
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher The American Society of Hematology
record_format MEDLINE/PubMed
spelling pubmed-102842602023-06-22 Intravital imaging of three different microvascular beds in SARS-CoV-2–infected mice Castanheira, Fernanda V. S. Nguyen, Rita Willson, Michelle Davoli-Ferreira, Marcela David, Bruna A. Kelly, Margaret M. Lee, Woo-Yong Kratofil, Rachel M. Zhang, Wen X. Bui-Marinos, Maxwell Corcoran, Jennifer A. Kubes, Paul Blood Adv Immunobiology and Immunotherapy Severe acute respiratory syndrome coronavirus–2 (SARS-CoV-2) enters the respiratory tract, where it infects the alveoli epithelial lining. However, patients have sequelae that extend well beyond the alveoli into the pulmonary vasculature and, perhaps, beyond to the brain and other organs. Because of the dynamic events within blood vessels, histology does not report platelet and neutrophil behavior. Because of the rapid nontranscriptional response of these cells, neither single-cell RNA sequencing nor proteomics report robustly on their critical behaviors. We used intravital microscopy in level-3 containment to examine the pathogenesis of SARS-CoV-2 within 3 organs in mice expressing human angiotensin converting enzyme 2 (ACE-2) ubiquitously (CAG-AC-70) or on epithelium (K18-promoter). Using a neon-green SARS-CoV-2, we observed both the epithelium and endothelium infected in AC70 mice but only the epithelium in K18 mice. There were increased neutrophils in the microcirculation but not in the alveoli of the lungs of AC70 mice. Platelets formed large aggregates in the pulmonary capillaries. Despite only neurons being infected within the brain, profound neutrophil adhesion forming the nidus of large platelet aggregates were observed in the cerebral microcirculation, with many nonperfused microvessels. Neutrophils breached the brain endothelial layer associated with a significant disruption of the blood-brain-barrier. Despite ubiquitous ACE-2 expression, CAG-AC-70 mice had very small increases in blood cytokine, no increase in thrombin, no infected circulating cells, and no liver involvement suggesting limited systemic effects. In summary, our imaging of SARS-CoV-2–infected mice gave direct evidence that there is a significant perturbation locally in the lung and brain microcirculation induced by local viral infection leading to increased local inflammation and thrombosis in these organs. The American Society of Hematology 2023-06-14 /pmc/articles/PMC10284260/ /pubmed/37307197 http://dx.doi.org/10.1182/bloodadvances.2022009430 Text en © 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved. https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
spellingShingle Immunobiology and Immunotherapy
Castanheira, Fernanda V. S.
Nguyen, Rita
Willson, Michelle
Davoli-Ferreira, Marcela
David, Bruna A.
Kelly, Margaret M.
Lee, Woo-Yong
Kratofil, Rachel M.
Zhang, Wen X.
Bui-Marinos, Maxwell
Corcoran, Jennifer A.
Kubes, Paul
Intravital imaging of three different microvascular beds in SARS-CoV-2–infected mice
title Intravital imaging of three different microvascular beds in SARS-CoV-2–infected mice
title_full Intravital imaging of three different microvascular beds in SARS-CoV-2–infected mice
title_fullStr Intravital imaging of three different microvascular beds in SARS-CoV-2–infected mice
title_full_unstemmed Intravital imaging of three different microvascular beds in SARS-CoV-2–infected mice
title_short Intravital imaging of three different microvascular beds in SARS-CoV-2–infected mice
title_sort intravital imaging of three different microvascular beds in sars-cov-2–infected mice
topic Immunobiology and Immunotherapy
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10284260/
https://www.ncbi.nlm.nih.gov/pubmed/37307197
http://dx.doi.org/10.1182/bloodadvances.2022009430
work_keys_str_mv AT castanheirafernandavs intravitalimagingofthreedifferentmicrovascularbedsinsarscov2infectedmice
AT nguyenrita intravitalimagingofthreedifferentmicrovascularbedsinsarscov2infectedmice
AT willsonmichelle intravitalimagingofthreedifferentmicrovascularbedsinsarscov2infectedmice
AT davoliferreiramarcela intravitalimagingofthreedifferentmicrovascularbedsinsarscov2infectedmice
AT davidbrunaa intravitalimagingofthreedifferentmicrovascularbedsinsarscov2infectedmice
AT kellymargaretm intravitalimagingofthreedifferentmicrovascularbedsinsarscov2infectedmice
AT leewooyong intravitalimagingofthreedifferentmicrovascularbedsinsarscov2infectedmice
AT kratofilrachelm intravitalimagingofthreedifferentmicrovascularbedsinsarscov2infectedmice
AT zhangwenx intravitalimagingofthreedifferentmicrovascularbedsinsarscov2infectedmice
AT buimarinosmaxwell intravitalimagingofthreedifferentmicrovascularbedsinsarscov2infectedmice
AT corcoranjennifera intravitalimagingofthreedifferentmicrovascularbedsinsarscov2infectedmice
AT kubespaul intravitalimagingofthreedifferentmicrovascularbedsinsarscov2infectedmice