当前位置: X-MOL 学术Acta Neuropathol. › 论文详情
Our official English website, www.x-mol.net, welcomes your feedback! (Note: you will need to create a separate account there.)
Microglial-mediated PDGF-CC activation increases cerebrovascular permeability during ischemic stroke.
Acta Neuropathologica ( IF 12.7 ) Pub Date : 2017-07-19 , DOI: 10.1007/s00401-017-1749-z
Enming Joseph Su 1 , Chunzhang Cao 2 , Linda Fredriksson 3 , Ingrid Nilsson 3 , Christina Stefanitsch 3 , Tamara K Stevenson 4 , Juanjuan Zhao 2 , Margret Ragsdale 1 , Yu-Yo Sun 5 , Manuel Yepes 6 , Chia-Yi Kuan 5 , Ulf Eriksson 3 , Dudley K Strickland 2 , Daniel A Lawrence 1, 4 , Li Zhang 2
Affiliation  

Treatment of acute ischemic stroke with the thrombolytic tissue plasminogen activator (tPA) can significantly improve neurological outcomes; however, thrombolytic therapy is associated with an increased risk of intra-cerebral hemorrhage (ICH). Previously, we demonstrated that during stroke tPA acting on the parenchymal side of the neurovascular unit (NVU) can increase blood-brain barrier (BBB) permeability and ICH through activation of latent platelet-derived growth factor-CC (PDGF-CC) and signaling by the PDGF receptor-α (PDGFRα). However, in vitro, activation of PDGF-CC by tPA is very inefficient and the mechanism of PDGF-CC activation in the NVU is not known. Here, we show that the integrin Mac-1, expressed on brain microglia/macrophages (denoted microglia throughout), acts together with the endocytic receptor LRP1 in the NVU to promote tPA-mediated activation of PDGF-CC. Mac-1-deficient mice (Mac-1-/-) are protected from tPA-induced BBB permeability but not from permeability induced by intracerebroventricular injection of active PDGF-CC. Immunofluorescence analysis demonstrates that Mac-1, LRP1, and the PDGFRα all localize to the NVU of arterioles, and following middle cerebral artery occlusion (MCAO) Mac-1-/- mice show significantly less PDGFRα phosphorylation, BBB permeability, and infarct volume compared to wild-type mice. Bone-marrow transplantation studies indicate that resident CD11b+ cells, but not bone-marrow-derived leukocytes, mediate the early activation of PDGF-CC by tPA after MCAO. Finally, using a model of thrombotic stroke with late thrombolysis, we show that wild-type mice have an increased incidence of spontaneous ICH following thrombolysis with tPA 5 h after MCAO, whereas Mac-1-/- mice are resistant to the development of ICH even with late tPA treatment. Together, these results indicate that Mac-1 and LRP1 act as co-factors for the activation of PDGF-CC by tPA in the NVU, and suggest a novel mechanism for tightly regulating PDGFRα signaling in the NVU and controlling BBB permeability.

中文翻译:

小胶质细胞介导的PDGF-CC激活增加缺血性中风期间的脑血管通透性。

用溶栓组织纤溶酶原激活剂(tPA)治疗急性缺血性中风可以显着改善神经系统预后。但是,溶栓治疗会增加脑内出血(ICH)的风险。先前,我们证明了在卒中期间,tPA作用于神经血管单元(NVU)的实质侧,可以通过激活潜在的血小板衍生生长因子-CC(PDGF-CC)和信号传导来增加血脑屏障(BBB)的通透性和ICH由PDGF受体-α(PDGFRα)产生。但是,在体外,tPA激活PDGF-CC的效率非常低,并且NVU中PDGF-CC激活的机制尚不清楚。在这里,我们显示了在大脑小胶质细胞/巨噬细胞(在整个过程中均表示为小胶质细胞)上表达的整合素Mac-1,与NVU中的内吞受体LRP1共同起作用,以促进tPA介导的PDGF-CC活化。Mac-1缺陷小鼠(Mac-1-/-)不受tPA诱导的BBB渗透性的保护,但不受脑室内注射活性PDGF-CC诱导的渗透性的保护。免疫荧光分析表明,Mac-1,LRP1和PDGFRα均定位于小动脉的NVU,并且在大脑中动脉闭塞(MCAO)之后,Mac-1-/-小鼠的PDGFRα磷酸化,BBB通透性和梗死体积明显减少野生型小鼠。骨髓移植研究表明,MCAO后,tPA可以驻留CD11b +细胞,而不是源自骨髓的白细胞,从而介导PDGF-CC的早期活化。最后,使用血栓性中风伴晚期溶栓的模型,我们显示,野生型小鼠在MCAO后5小时用tPA溶栓后,自发性ICH发生率增加,而Mac-1-/-小鼠即使在tPA治疗后期也对ICH产生抵抗力。在一起,这些结果表明Mac-1和LRP1充当NVPA中tPA激活PDGF-CC的辅助因子,并提出了一种严格调节NVU中PDGFRα信号传导并控制BBB渗透性的新机制。
更新日期:2017-07-19
down
wechat
bug