Aggravated pulmonary injury after subarachnoid hemorrhage in PDGF-Bret/ret mice

Abstract Background Recent advances in surgical and neuroprotective strategies could effectively manage the pathophysiological progression of subarachnoid hemorrhage (SAH). However, pulmonary dysfunction frequently occurs in SAH patients with an increased risk of unsatisfactory outcomes. Based on th...

Full description

Bibliographic Details
Main Authors: Pengyu Pan, Jie Qu, Qiang Li, Rongwei Li, Yang Yang, Shilun Zuo, Xin Liu, Hua Feng, Yujie Chen
Format: Article
Language:English
Published: BMC 2020-06-01
Series:Chinese Neurosurgical Journal
Subjects:
Online Access:http://link.springer.com/article/10.1186/s41016-020-00193-2
id doaj-a85d05abc53a46ffa9e0d7154bd20d9e
record_format Article
spelling doaj-a85d05abc53a46ffa9e0d7154bd20d9e2020-11-25T03:54:20ZengBMCChinese Neurosurgical Journal2057-49672020-06-01611910.1186/s41016-020-00193-2Aggravated pulmonary injury after subarachnoid hemorrhage in PDGF-Bret/ret micePengyu Pan0Jie Qu1Qiang Li2Rongwei Li3Yang Yang4Shilun Zuo5Xin Liu6Hua Feng7Yujie Chen8Department of Neurosurgery, Southwest Hospital, Third Military Medical UniversityDepartment of Neurosurgery, Southwest Hospital, Third Military Medical UniversityDepartment of Neurosurgery, Southwest Hospital, Third Military Medical UniversityDepartment of Neurosurgery, Southwest Hospital, Third Military Medical UniversityDepartment of Neurosurgery, Southwest Hospital, Third Military Medical UniversityDepartment of Neurosurgery, Southwest Hospital, Third Military Medical UniversityDepartment of Neurosurgery, Southwest Hospital, Third Military Medical UniversityDepartment of Neurosurgery, Southwest Hospital, Third Military Medical UniversityDepartment of Neurosurgery, Southwest Hospital, Third Military Medical UniversityAbstract Background Recent advances in surgical and neuroprotective strategies could effectively manage the pathophysiological progression of subarachnoid hemorrhage (SAH). However, pulmonary dysfunction frequently occurs in SAH patients with an increased risk of unsatisfactory outcomes. Based on the similar microvascular structures in the blood-air barrier and blood-brain barrier and possible brain-lung crosstalks, we believe that pericytes may be involved in both neurological and pulmonary dysfunction after SAH. Methods In our experiments, platelet-derived growth factor B (PDGF-B) retention motif knockout (PDGF-Bret/ret) mice and adeno-associated virus PDGF-B were employed to show the involvement of pericyte deficiency and PDGF-B expression. Neurological score, SAH grade, hematoxylin-eosin staining, and PaO2/FiO2 ratio analysis were performed to evaluate the neurological deficits and pulmonary functions in endovascular perforation SAH models at 24 h after surgery, as well as western blotting and immunofluorescence staining for underlying molecular expressions. Results We found that neonatal PDGF-Bret/ret mice exhibited pulmonary atelectasis 12 h after birth. Further investigation showed a decrease in PaO2/FiO2 and lung-specific surfactant proteins in adult PDGF-Bret/ret mice. These dysfunctions were much worse than those in wild-type mice at 24 h after SAH. PDGF-B overexpression alleviated pulmonary dysfunction after SAH. Conclusions These results suggested pulmonary dysfunction after SAH and the pivotal role of PDGF-B signaling for the pathophysiological process and future therapeutic targets of pulmonary injury treatment after SAH. Further studies are needed for pathophysiological investigations and translational studies on pulmonary injuries after SAH.http://link.springer.com/article/10.1186/s41016-020-00193-2Platelet-derived growth factor BPulmonary injurySubarachnoid hemorrhageLung-specific surfactant protein
collection DOAJ
language English
format Article
sources DOAJ
author Pengyu Pan
Jie Qu
Qiang Li
Rongwei Li
Yang Yang
Shilun Zuo
Xin Liu
Hua Feng
Yujie Chen
spellingShingle Pengyu Pan
Jie Qu
Qiang Li
Rongwei Li
Yang Yang
Shilun Zuo
Xin Liu
Hua Feng
Yujie Chen
Aggravated pulmonary injury after subarachnoid hemorrhage in PDGF-Bret/ret mice
Chinese Neurosurgical Journal
Platelet-derived growth factor B
Pulmonary injury
Subarachnoid hemorrhage
Lung-specific surfactant protein
author_facet Pengyu Pan
Jie Qu
Qiang Li
Rongwei Li
Yang Yang
Shilun Zuo
Xin Liu
Hua Feng
Yujie Chen
author_sort Pengyu Pan
title Aggravated pulmonary injury after subarachnoid hemorrhage in PDGF-Bret/ret mice
title_short Aggravated pulmonary injury after subarachnoid hemorrhage in PDGF-Bret/ret mice
title_full Aggravated pulmonary injury after subarachnoid hemorrhage in PDGF-Bret/ret mice
title_fullStr Aggravated pulmonary injury after subarachnoid hemorrhage in PDGF-Bret/ret mice
title_full_unstemmed Aggravated pulmonary injury after subarachnoid hemorrhage in PDGF-Bret/ret mice
title_sort aggravated pulmonary injury after subarachnoid hemorrhage in pdgf-bret/ret mice
publisher BMC
series Chinese Neurosurgical Journal
issn 2057-4967
publishDate 2020-06-01
description Abstract Background Recent advances in surgical and neuroprotective strategies could effectively manage the pathophysiological progression of subarachnoid hemorrhage (SAH). However, pulmonary dysfunction frequently occurs in SAH patients with an increased risk of unsatisfactory outcomes. Based on the similar microvascular structures in the blood-air barrier and blood-brain barrier and possible brain-lung crosstalks, we believe that pericytes may be involved in both neurological and pulmonary dysfunction after SAH. Methods In our experiments, platelet-derived growth factor B (PDGF-B) retention motif knockout (PDGF-Bret/ret) mice and adeno-associated virus PDGF-B were employed to show the involvement of pericyte deficiency and PDGF-B expression. Neurological score, SAH grade, hematoxylin-eosin staining, and PaO2/FiO2 ratio analysis were performed to evaluate the neurological deficits and pulmonary functions in endovascular perforation SAH models at 24 h after surgery, as well as western blotting and immunofluorescence staining for underlying molecular expressions. Results We found that neonatal PDGF-Bret/ret mice exhibited pulmonary atelectasis 12 h after birth. Further investigation showed a decrease in PaO2/FiO2 and lung-specific surfactant proteins in adult PDGF-Bret/ret mice. These dysfunctions were much worse than those in wild-type mice at 24 h after SAH. PDGF-B overexpression alleviated pulmonary dysfunction after SAH. Conclusions These results suggested pulmonary dysfunction after SAH and the pivotal role of PDGF-B signaling for the pathophysiological process and future therapeutic targets of pulmonary injury treatment after SAH. Further studies are needed for pathophysiological investigations and translational studies on pulmonary injuries after SAH.
topic Platelet-derived growth factor B
Pulmonary injury
Subarachnoid hemorrhage
Lung-specific surfactant protein
url http://link.springer.com/article/10.1186/s41016-020-00193-2
work_keys_str_mv AT pengyupan aggravatedpulmonaryinjuryaftersubarachnoidhemorrhageinpdgfbretretmice
AT jiequ aggravatedpulmonaryinjuryaftersubarachnoidhemorrhageinpdgfbretretmice
AT qiangli aggravatedpulmonaryinjuryaftersubarachnoidhemorrhageinpdgfbretretmice
AT rongweili aggravatedpulmonaryinjuryaftersubarachnoidhemorrhageinpdgfbretretmice
AT yangyang aggravatedpulmonaryinjuryaftersubarachnoidhemorrhageinpdgfbretretmice
AT shilunzuo aggravatedpulmonaryinjuryaftersubarachnoidhemorrhageinpdgfbretretmice
AT xinliu aggravatedpulmonaryinjuryaftersubarachnoidhemorrhageinpdgfbretretmice
AT huafeng aggravatedpulmonaryinjuryaftersubarachnoidhemorrhageinpdgfbretretmice
AT yujiechen aggravatedpulmonaryinjuryaftersubarachnoidhemorrhageinpdgfbretretmice
_version_ 1724474211956686848