Cargando…

The ESCRT-III pathway facilitates cardiomyocyte release of cBIN1-containing microparticles

Microparticles (MPs) are cell–cell communication vesicles derived from the cell surface plasma membrane, although they are not known to originate from cardiac ventricular muscle. In ventricular cardiomyocytes, the membrane deformation protein cardiac bridging integrator 1 (cBIN1 or BIN1+13+17) creat...

Descripción completa

Detalles Bibliográficos
Autores principales: Xu, Bing, Fu, Ying, Liu, Yan, Agvanian, Sosse, Wirka, Robert C., Baum, Rachel, Zhou, Kang, Shaw, Robin M., Hong, TingTing
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5570487/
https://www.ncbi.nlm.nih.gov/pubmed/28806752
http://dx.doi.org/10.1371/journal.pbio.2002354
_version_ 1783259186565480448
author Xu, Bing
Fu, Ying
Liu, Yan
Agvanian, Sosse
Wirka, Robert C.
Baum, Rachel
Zhou, Kang
Shaw, Robin M.
Hong, TingTing
author_facet Xu, Bing
Fu, Ying
Liu, Yan
Agvanian, Sosse
Wirka, Robert C.
Baum, Rachel
Zhou, Kang
Shaw, Robin M.
Hong, TingTing
author_sort Xu, Bing
collection PubMed
description Microparticles (MPs) are cell–cell communication vesicles derived from the cell surface plasma membrane, although they are not known to originate from cardiac ventricular muscle. In ventricular cardiomyocytes, the membrane deformation protein cardiac bridging integrator 1 (cBIN1 or BIN1+13+17) creates transverse-tubule (t-tubule) membrane microfolds, which facilitate ion channel trafficking and modulate local ionic concentrations. The microfold-generated microdomains continuously reorganize, adapting in response to stress to modulate the calcium signaling apparatus. We explored the possibility that cBIN1-microfolds are externally released from cardiomyocytes. Using electron microscopy imaging with immunogold labeling, we found in mouse plasma that cBIN1 exists in membrane vesicles about 200 nm in size, which is consistent with the size of MPs. In mice with cardiac-specific heterozygous Bin1 deletion, flow cytometry identified 47% less cBIN1-MPs in plasma, supporting cardiac origin. Cardiac release was also evidenced by the detection of cBIN1-MPs in medium bathing a pure population of isolated adult mouse cardiomyocytes. In human plasma, osmotic shock increased cBIN1 detection by enzyme-linked immunosorbent assay (ELISA), and cBIN1 level decreased in humans with heart failure, a condition with reduced cardiac muscle cBIN1, both of which support cBIN1 release in MPs from human hearts. Exploring putative mechanisms of MP release, we found that the membrane fission complex endosomal sorting complexes required for transport (ESCRT)-III subunit charged multivesicular body protein 4B (CHMP4B) colocalizes and coimmunoprecipitates with cBIN1, an interaction enhanced by actin stabilization. In HeLa cells with cBIN1 overexpression, knockdown of CHMP4B reduced the release of cBIN1-MPs. Using truncation mutants, we identified that the N-terminal BAR (N-BAR) domain in cBIN1 is required for CHMP4B binding and MP release. This study links the BAR protein superfamily to the ESCRT pathway for MP biogenesis in mammalian cardiac ventricular cells, identifying elements of a pathway by which cytoplasmic cBIN1 is released into blood.
format Online
Article
Text
id pubmed-5570487
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-55704872017-08-28 The ESCRT-III pathway facilitates cardiomyocyte release of cBIN1-containing microparticles Xu, Bing Fu, Ying Liu, Yan Agvanian, Sosse Wirka, Robert C. Baum, Rachel Zhou, Kang Shaw, Robin M. Hong, TingTing PLoS Biol Research Article Microparticles (MPs) are cell–cell communication vesicles derived from the cell surface plasma membrane, although they are not known to originate from cardiac ventricular muscle. In ventricular cardiomyocytes, the membrane deformation protein cardiac bridging integrator 1 (cBIN1 or BIN1+13+17) creates transverse-tubule (t-tubule) membrane microfolds, which facilitate ion channel trafficking and modulate local ionic concentrations. The microfold-generated microdomains continuously reorganize, adapting in response to stress to modulate the calcium signaling apparatus. We explored the possibility that cBIN1-microfolds are externally released from cardiomyocytes. Using electron microscopy imaging with immunogold labeling, we found in mouse plasma that cBIN1 exists in membrane vesicles about 200 nm in size, which is consistent with the size of MPs. In mice with cardiac-specific heterozygous Bin1 deletion, flow cytometry identified 47% less cBIN1-MPs in plasma, supporting cardiac origin. Cardiac release was also evidenced by the detection of cBIN1-MPs in medium bathing a pure population of isolated adult mouse cardiomyocytes. In human plasma, osmotic shock increased cBIN1 detection by enzyme-linked immunosorbent assay (ELISA), and cBIN1 level decreased in humans with heart failure, a condition with reduced cardiac muscle cBIN1, both of which support cBIN1 release in MPs from human hearts. Exploring putative mechanisms of MP release, we found that the membrane fission complex endosomal sorting complexes required for transport (ESCRT)-III subunit charged multivesicular body protein 4B (CHMP4B) colocalizes and coimmunoprecipitates with cBIN1, an interaction enhanced by actin stabilization. In HeLa cells with cBIN1 overexpression, knockdown of CHMP4B reduced the release of cBIN1-MPs. Using truncation mutants, we identified that the N-terminal BAR (N-BAR) domain in cBIN1 is required for CHMP4B binding and MP release. This study links the BAR protein superfamily to the ESCRT pathway for MP biogenesis in mammalian cardiac ventricular cells, identifying elements of a pathway by which cytoplasmic cBIN1 is released into blood. Public Library of Science 2017-08-14 /pmc/articles/PMC5570487/ /pubmed/28806752 http://dx.doi.org/10.1371/journal.pbio.2002354 Text en © 2017 Xu et al http://creativecommons.org/licenses/by/4.0/ This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/) , which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Article
Xu, Bing
Fu, Ying
Liu, Yan
Agvanian, Sosse
Wirka, Robert C.
Baum, Rachel
Zhou, Kang
Shaw, Robin M.
Hong, TingTing
The ESCRT-III pathway facilitates cardiomyocyte release of cBIN1-containing microparticles
title The ESCRT-III pathway facilitates cardiomyocyte release of cBIN1-containing microparticles
title_full The ESCRT-III pathway facilitates cardiomyocyte release of cBIN1-containing microparticles
title_fullStr The ESCRT-III pathway facilitates cardiomyocyte release of cBIN1-containing microparticles
title_full_unstemmed The ESCRT-III pathway facilitates cardiomyocyte release of cBIN1-containing microparticles
title_short The ESCRT-III pathway facilitates cardiomyocyte release of cBIN1-containing microparticles
title_sort escrt-iii pathway facilitates cardiomyocyte release of cbin1-containing microparticles
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5570487/
https://www.ncbi.nlm.nih.gov/pubmed/28806752
http://dx.doi.org/10.1371/journal.pbio.2002354
work_keys_str_mv AT xubing theescrtiiipathwayfacilitatescardiomyocytereleaseofcbin1containingmicroparticles
AT fuying theescrtiiipathwayfacilitatescardiomyocytereleaseofcbin1containingmicroparticles
AT liuyan theescrtiiipathwayfacilitatescardiomyocytereleaseofcbin1containingmicroparticles
AT agvaniansosse theescrtiiipathwayfacilitatescardiomyocytereleaseofcbin1containingmicroparticles
AT wirkarobertc theescrtiiipathwayfacilitatescardiomyocytereleaseofcbin1containingmicroparticles
AT baumrachel theescrtiiipathwayfacilitatescardiomyocytereleaseofcbin1containingmicroparticles
AT zhoukang theescrtiiipathwayfacilitatescardiomyocytereleaseofcbin1containingmicroparticles
AT shawrobinm theescrtiiipathwayfacilitatescardiomyocytereleaseofcbin1containingmicroparticles
AT hongtingting theescrtiiipathwayfacilitatescardiomyocytereleaseofcbin1containingmicroparticles
AT xubing escrtiiipathwayfacilitatescardiomyocytereleaseofcbin1containingmicroparticles
AT fuying escrtiiipathwayfacilitatescardiomyocytereleaseofcbin1containingmicroparticles
AT liuyan escrtiiipathwayfacilitatescardiomyocytereleaseofcbin1containingmicroparticles
AT agvaniansosse escrtiiipathwayfacilitatescardiomyocytereleaseofcbin1containingmicroparticles
AT wirkarobertc escrtiiipathwayfacilitatescardiomyocytereleaseofcbin1containingmicroparticles
AT baumrachel escrtiiipathwayfacilitatescardiomyocytereleaseofcbin1containingmicroparticles
AT zhoukang escrtiiipathwayfacilitatescardiomyocytereleaseofcbin1containingmicroparticles
AT shawrobinm escrtiiipathwayfacilitatescardiomyocytereleaseofcbin1containingmicroparticles
AT hongtingting escrtiiipathwayfacilitatescardiomyocytereleaseofcbin1containingmicroparticles