Cargando…

Embolus extravasation is an alternative mechanism for cerebral microvascular recanalization

Cerebral microvascular occlusion is a common phenomenon throughout life1,2 that could be an underappreciated mechanism of brain pathology. Failure to promptly recanalize microvessels may lead to disruption of brain circuits and significant functional deficits3. Hemodynamic forces and the fibrinolyti...

Descripción completa

Detalles Bibliográficos
Autores principales: Lam, Carson K., Yoo, Taehwan, Hiner, Bennett, Liu, Zhiqiang, Grutzendler, Jaime
Formato: Texto
Lenguaje:English
Publicado: 2010
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2879083/
https://www.ncbi.nlm.nih.gov/pubmed/20505729
http://dx.doi.org/10.1038/nature09001
_version_ 1782181903262023680
author Lam, Carson K.
Yoo, Taehwan
Hiner, Bennett
Liu, Zhiqiang
Grutzendler, Jaime
author_facet Lam, Carson K.
Yoo, Taehwan
Hiner, Bennett
Liu, Zhiqiang
Grutzendler, Jaime
author_sort Lam, Carson K.
collection PubMed
description Cerebral microvascular occlusion is a common phenomenon throughout life1,2 that could be an underappreciated mechanism of brain pathology. Failure to promptly recanalize microvessels may lead to disruption of brain circuits and significant functional deficits3. Hemodynamic forces and the fibrinolytic system4 are considered the principal mechanisms responsible for recanalization of occluded cerebral capillaries and terminal arterioles. However, using high resolution fixed tissue microscopy and two photon imaging in living mice we found that a large fraction of occluding microemboli failed to be lysed and washed out within 48 hours after internal carotid infusion. Surprisingly, emboli were instead found to translocate outside the vessel lumen within 2-7 days leading to complete re-establishment of blood flow and sparing of the vessel. Recanalization occurred by a previously unknown mechanism of microvascular plasticity involving the rapid envelopment of emboli by endothelial membrane projections which subsequently form a new vessel wall. This was followed by the formation of an endothelial opening through which emboli translocated into the perivascular parenchyma. The rate of embolus extravasation was significantly reduced by pharmacological inhibition of matrix metalloproteinase 2/9 activity. In aged mice, extravasation was markedly delayed, resulting in persistent tissue hypoxia, synaptic damage and cell death. Our study identifies a novel cellular mechanism that may be critical for recanalization of occluded microvessels. Alterations in the efficiency of this protective mechanism may have important implications in microvascular pathology, stroke recovery, and age-related cognitive decline.
format Text
id pubmed-2879083
institution National Center for Biotechnology Information
language English
publishDate 2010
record_format MEDLINE/PubMed
spelling pubmed-28790832010-11-27 Embolus extravasation is an alternative mechanism for cerebral microvascular recanalization Lam, Carson K. Yoo, Taehwan Hiner, Bennett Liu, Zhiqiang Grutzendler, Jaime Nature Article Cerebral microvascular occlusion is a common phenomenon throughout life1,2 that could be an underappreciated mechanism of brain pathology. Failure to promptly recanalize microvessels may lead to disruption of brain circuits and significant functional deficits3. Hemodynamic forces and the fibrinolytic system4 are considered the principal mechanisms responsible for recanalization of occluded cerebral capillaries and terminal arterioles. However, using high resolution fixed tissue microscopy and two photon imaging in living mice we found that a large fraction of occluding microemboli failed to be lysed and washed out within 48 hours after internal carotid infusion. Surprisingly, emboli were instead found to translocate outside the vessel lumen within 2-7 days leading to complete re-establishment of blood flow and sparing of the vessel. Recanalization occurred by a previously unknown mechanism of microvascular plasticity involving the rapid envelopment of emboli by endothelial membrane projections which subsequently form a new vessel wall. This was followed by the formation of an endothelial opening through which emboli translocated into the perivascular parenchyma. The rate of embolus extravasation was significantly reduced by pharmacological inhibition of matrix metalloproteinase 2/9 activity. In aged mice, extravasation was markedly delayed, resulting in persistent tissue hypoxia, synaptic damage and cell death. Our study identifies a novel cellular mechanism that may be critical for recanalization of occluded microvessels. Alterations in the efficiency of this protective mechanism may have important implications in microvascular pathology, stroke recovery, and age-related cognitive decline. 2010-05-27 /pmc/articles/PMC2879083/ /pubmed/20505729 http://dx.doi.org/10.1038/nature09001 Text en Users may view, print, copy, download and text and data- mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use: http://www.nature.com/authors/editorial_policies/license.html#terms
spellingShingle Article
Lam, Carson K.
Yoo, Taehwan
Hiner, Bennett
Liu, Zhiqiang
Grutzendler, Jaime
Embolus extravasation is an alternative mechanism for cerebral microvascular recanalization
title Embolus extravasation is an alternative mechanism for cerebral microvascular recanalization
title_full Embolus extravasation is an alternative mechanism for cerebral microvascular recanalization
title_fullStr Embolus extravasation is an alternative mechanism for cerebral microvascular recanalization
title_full_unstemmed Embolus extravasation is an alternative mechanism for cerebral microvascular recanalization
title_short Embolus extravasation is an alternative mechanism for cerebral microvascular recanalization
title_sort embolus extravasation is an alternative mechanism for cerebral microvascular recanalization
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2879083/
https://www.ncbi.nlm.nih.gov/pubmed/20505729
http://dx.doi.org/10.1038/nature09001
work_keys_str_mv AT lamcarsonk embolusextravasationisanalternativemechanismforcerebralmicrovascularrecanalization
AT yootaehwan embolusextravasationisanalternativemechanismforcerebralmicrovascularrecanalization
AT hinerbennett embolusextravasationisanalternativemechanismforcerebralmicrovascularrecanalization
AT liuzhiqiang embolusextravasationisanalternativemechanismforcerebralmicrovascularrecanalization
AT grutzendlerjaime embolusextravasationisanalternativemechanismforcerebralmicrovascularrecanalization