Blood-Brain Barrier

血脑屏障
  • 文章类型: Journal Article
    内皮细胞中细胞质Ca2+的严格控制对于内皮屏障功能的调节是必不可少的。这里,我们研究了电压门控Ca2+(Cav)通道亚基Cavβ3的作用,在调节脑微血管内皮细胞(BMECs)中的Ca2信号传导以及这如何有助于血脑屏障的完整性。
    我们通过Ca2+成像和蛋白质印迹研究了Cavβ3在BMEC中的功能,检查了体外内皮屏障功能和体内血脑屏障的完整性,并使用Cavβ3-/-(Cavβ3缺陷型)小鼠作为对照,评估了小鼠诱导实验性自身免疫性脑脊髓炎后的病程。
    我们鉴定了BMEC中的Cavβ3蛋白,但是电生理记录没有显示出明显的Cav通道活性。在体内,在不存在Cavβ3的情况下,血脑屏障完整性降低.诱导实验性自身免疫性脑脊髓炎后,Cavβ3-/-小鼠表现出更早的疾病发作,加剧了临床残疾和增加的T细胞浸润。体外,Cavβ3-/-BMEC单层的跨内皮阻力低于野生型BMEC单层,并且连接蛋白ZO-1(闭塞带-1)的组织受损。凝血酶刺激肌醇1,4,5-三磷酸依赖性Ca2+释放,其通过MLC(肌球蛋白轻链)的Ca2+依赖性磷酸化促进细胞收缩并增强内皮屏障通透性。这些影响在Cavβ3-/-中比在野生型BMECs中更明显,而在MLCK(MLC激酶)抑制剂ML-7的存在下,差异被消除。Cacnb3cDNA在Cavβ3-/-BMECs中的表达恢复了野生型表型。共免疫沉淀和质谱表明Cavβ3与肌醇1,4,5-三磷酸受体蛋白的关联。
    独立于其作为Cav通道亚基的功能,Cavβ3与肌醇1,4,5-三磷酸受体相互作用,并参与BMEC中细胞质Ca2和Ca2依赖性MLC磷酸化的严格控制,Cavβ3在BMEC中的这种作用有助于血脑屏障的完整性,并减轻实验性自身免疫性脑脊髓炎疾病的严重程度。
    UNASSIGNED: Tight control of cytoplasmic Ca2+ in endothelial cells is essential for the regulation of endothelial barrier function. Here, we investigated the role of Cavβ3, a subunit of voltage-gated Ca2+ (Cav) channels, in modulating Ca2+ signaling in brain microvascular endothelial cells (BMECs) and how this contributes to the integrity of the blood-brain barrier.
    UNASSIGNED: We investigated the function of Cavβ3 in BMECs by Ca2+ imaging and Western blot, examined the endothelial barrier function in vitro and the integrity of the blood-brain barrier in vivo, and evaluated disease course after induction of experimental autoimmune encephalomyelitis in mice using Cavβ3-/- (Cav β3-deficient) mice as controls.
    UNASSIGNED: We identified Cavβ3 protein in BMECs, but electrophysiological recordings did not reveal significant Cav channel activity. In vivo, blood-brain barrier integrity was reduced in the absence of Cavβ3. After induction of experimental autoimmune encephalomyelitis, Cavβ3-/- mice showed earlier disease onset with exacerbated clinical disability and increased T-cell infiltration. In vitro, the transendothelial resistance of Cavβ3-/- BMEC monolayers was lower than that of wild-type BMEC monolayers, and the organization of the junctional protein ZO-1 (zona occludens-1) was impaired. Thrombin stimulates inositol 1,4,5-trisphosphate-dependent Ca2+ release, which facilitates cell contraction and enhances endothelial barrier permeability via Ca2+-dependent phosphorylation of MLC (myosin light chain). These effects were more pronounced in Cavβ3-/- than in wild-type BMECs, whereas the differences were abolished in the presence of the MLCK (MLC kinase) inhibitor ML-7. Expression of Cacnb3 cDNA in Cavβ3-/- BMECs restored the wild-type phenotype. Coimmunoprecipitation and mass spectrometry demonstrated the association of Cavβ3 with inositol 1,4,5-trisphosphate receptor proteins.
    UNASSIGNED: Independent of its function as a subunit of Cav channels, Cavβ3 interacts with the inositol 1,4,5-trisphosphate receptor and is involved in the tight control of cytoplasmic Ca2+ and Ca2+-dependent MLC phosphorylation in BMECs, and this role of Cavβ3 in BMECs contributes to blood-brain barrier integrity and attenuates the severity of experimental autoimmune encephalomyelitis disease.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的:由于治疗选择有限,缺血性卒中在医学研究中仍然是一个挑战。重组人组织纤溶酶原激活剂(rtPA)是再通的主要治疗方法。然而,近50%的患者出现并发症,导致无效的再灌注。导致无效再灌注的确切因素仍不清楚;然而,最近的研究表明,免疫细胞,特别是中性粒细胞,可能通过中性粒细胞胞外陷阱的形成等机制影响rtPA溶栓的结果。本研究旨在探讨rtPA对中性粒细胞的非溶栓作用,并强调其对无效再灌注的贡献。
    方法:我们评估了rtPA治疗对大鼠大脑中动脉闭塞的影响。我们还评估了大量脑缺血(MCI)患者中rtPA治疗后的中性粒细胞浸润和活化。
    结果:rtPA增加了中性粒细胞向脑微血管的浸润,并恶化了缺血期间的血脑屏障损伤。它还增加了MCI患者的中性粒细胞计数。
    结论:中性粒细胞在促进缺血性损伤和血脑屏障破坏中起关键作用,使它们成为潜在的治疗目标。
    OBJECTIVE: Ischemic stroke remains a challenge in medical research because of the limited treatment options. Recombinant human tissue plasminogen activator (rtPA) is the primary treatment for recanalization. However, nearly 50% of the patients experience complications that result in ineffective reperfusion. The precise factors contributing to ineffective reperfusion remain unclear; however, recent studies have suggested that immune cells, notably neutrophils, may influence the outcome of rtPA thrombolysis via mechanisms such as the formation of neutrophil extracellular traps. This study aimed to explore the nonthrombolytic effects of rtPA on neutrophils and highlight their contribution to ineffective reperfusion.
    METHODS: We evaluated the effects of rtPA treatment on middle cerebral artery occlusion in rats. We also assessed neutrophil infiltration and activation after rtPA treatment in vitro and in vivo in a small cohort of patients with massive cerebral ischemia (MCI).
    RESULTS: rtPA increased neutrophil infiltration into the brain microvessels and worsened blood-brain barrier damage during ischemia. It also increased the neutrophil counts of the patients with MCI.
    CONCLUSIONS: Neutrophils play a crucial role in promoting ischemic injury and blood-brain barrier disruption, making them potential therapeutic targets.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的探讨创伤性脑损伤(TBI)与高级别胶质瘤共同的分子发病机制,探讨异丙酚(PF)作为潜在保护剂的作用机制。通过分析中国胶质瘤基因组图谱(CGGA)和癌症基因组图谱(TCGA)数据库,我们比较了高级别胶质瘤和TBI患者的转录组数据,以确定共同的病理机制.通过生物信息学分析,体外实验和体内TBI模型,我们研究了在氧化应激下PF通过Prrx1对细胞外基质(ECM)相关基因的调节作用。使用双层BBB模型研究了PF对氧化应激下BBB完整性的影响,我们探讨了PF对TBI后小鼠紧密连接蛋白和ECM相关基因的保护作用。研究发现,高级别胶质瘤和TBI具有ECM不稳定性的重要分子病理机制。PF通过直接结合Prrx1或通过miRNA间接调节Prrx1来稳定ECM并保护BBB。此外,PF在氧化应激下降低细胞内钙离子和ROS水平,从而保持BBB完整性。在TBI小鼠模型中,PF通过上调紧密连接蛋白保护BBB的完整性并稳定ECM相关基因的表达。我们的研究揭示了TBI和胶质母细胞瘤之间共同的分子发病机制,并证明了PF作为BBB保护剂的潜力。这为新型神经创伤治疗药物的开发提供了新的靶点和途径。
    The purpose of this study is to explore the shared molecular pathogenesis of traumatic brain injury (TBI) and high-grade glioma and investigate the mechanism of propofol (PF) as a potential protective agent. By analyzing the Chinese glioma genome atlas (CGGA) and The Cancer Genome Atlas (TCGA) databases, we compared the transcriptomic data of high-grade glioma and TBI patients to identify common pathological mechanisms. Through bioinformatics analysis, in vitro experiments and in vivo TBI model, we investigated the regulatory effect of PF on extracellular matrix (ECM)-related genes through Prrx1 under oxidative stress. The impact of PF on BBB integrity under oxidative stress was investigated using a dual-layer BBB model, and we explored the protective effect of PF on tight junction proteins and ECM-related genes in mice after TBI. The study found that high-grade glioma and TBI share ECM instability as an important molecular pathological mechanism. PF stabilizes the ECM and protects the BBB by directly binding to Prrx1 or indirectly regulating Prrx1 through miRNAs. In addition, PF reduces intracellular calcium ions and ROS levels under oxidative stress, thereby preserving BBB integrity. In a TBI mouse model, PF protected BBB integrity through up-regulated tight junction proteins and stabilized the expression of ECM-related genes. Our study reveals the shared molecular pathogenesis between TBI and glioblastoma and demonstrate the potential of PF as a protective agent of BBB. This provides new targets and approaches for the development of novel neurotrauma therapeutic drugs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    猪链球菌(S.suis)2型(SS2)是一种重要的人畜共患病原体,可引起猪的严重神经感染,对公众健康造成严重威胁。炎症小体激活在宿主抵抗微生物感染中起重要作用,但很少研究炎症小体激活在猪链球菌感染期间血脑屏障(BBB)完整性中的作用。这项研究调查了猪链球菌诱导的NLRP3炎性体激活导致BBB破坏的机制。我们的结果表明,猪链球菌感染激活了脑微血管内皮细胞(BMECs)中的NLRP3炎性体,导致促炎细胞因子(IL-1β,IL-6和TNF-α)和趋化因子(CCL-2和CXCL-2)以及GasderminD(GSDMD)的裂解被炎症体抑制剂MCC950显着减弱。此外,猪链球菌感染显着下调了紧密连接(TJs)蛋白和跨内皮电阻(TEER)的表达,而NLRP3抑制挽救了猪链球菌诱导的TJs蛋白降解,并显着减少了跨BBB的猪链球菌数量。感染模型。此外,重组IL-1β加剧了BMECs中TJs蛋白的减少。在鼠猪链球菌感染模型中,MCC950降低了小鼠脑内的细菌负荷和过度的炎症反应。此外,在抑制NLRP3炎性体后,BBB的完整性受到TJ蛋白表达增加和病理损伤减少的保护,表明NLRP3炎性体在猪链球菌引起的脑膜炎中起破坏性作用。我们的研究扩展了对NLRP3炎性体在细菌性脑膜炎中的作用的理解,为开发针对NLRP3的抗感染药物治疗细菌性脑膜炎提供了有价值的信息。
    Streptococcus suis (S. suis) type 2 (SS2) is an important zoonotic pathogen causing severe neural infections in pigs and causes serious threat to public health. Inflammasome activation plays an important role in the host against microbial infection but the role of inflammasome activation in the blood-brain barrier (BBB) integrity during S. suis infection is rarely studied. This study investigated the mechanism by which S. suis-induced NLRP3 inflammasome activation led to BBB disruption. Our results showed that S. suis infection activated NLRP3 inflammasome in brain microvascular endothelial cells (BMECs) leading to the secretion of pro-inflammatory cytokines (IL-1β, IL-6 and TNF-α) and chemokines (CCL-2 and CXCL-2) as well as the cleavage of Gasdermin D (GSDMD) which were significantly attenuated by inflammasome inhibitor MCC950. Furthermore, S. suis infection significantly downregulated expression of tight junctions (TJs) proteins and trans-endothelial electrical resistance (TEER) while NLRP3 inhibition rescued S. suis-induced degradation of TJs proteins and significantly reduced the number of S. suis crossing BBB in transwell infection model. Moreover, recombinant IL-1β exacerbated the reduction of TJs proteins in BMECs. In murine S. suis-infection model, MCC950 reduced the bacterial load and the excessive inflammatory response in mice brain. In addition, the integrity of the BBB was protected with increased TJ proteins expression and decreased pathological injury after the inhibition of NLRP3 inflammasome, indicating NLRP3 inflammasome plays a destructive role in meningitis induced by S. suis. Our study expands the understanding on the role of NLRP3 inflammasome in bacterial meningitis, which provide the valuable information for the development of anti-infective agents targeting NLRP3 to treat bacterial meningitis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    血脑屏障(BBB)是中枢神经系统中至关重要的物理过滤结构。这里,我们研究了特定的星形胶质细胞亚群在调节BBB完整性中的作用。我们表明,表达Dmp1的星形胶质细胞通过其末端脚将线粒体转移到内皮细胞,以维持BBB完整性。在表达Dmp1的星形胶质细胞中缺失Mitofusin2(Mfn2)基因会抑制线粒体转移并导致BBB渗漏。此外,星形胶质细胞中MFN2的减少有助于与年龄相关的线粒体转移效率降低,从而损害BBB的完整性.一起,我们描述了星形胶质细胞通过线粒体转移调节BBB完整性的机制。我们的发现为支持与衰老和疾病相关的BBB进行性分解的细胞框架提供了创新的见解。
    The blood-brain barrier (BBB) acts as the crucial physical filtration structure in the central nervous system. Here, we investigate the role of a specific subset of astrocytes in the regulation of BBB integrity. We showed that Dmp1-expressing astrocytes transfer mitochondria to endothelial cells via their endfeet for maintaining BBB integrity. Deletion of the Mitofusin 2 (Mfn2) gene in Dmp1-expressing astrocytes inhibited the mitochondrial transfer and caused BBB leakage. In addition, the decrease of MFN2 in astrocytes contributes to the age-associated reduction of mitochondrial transfer efficiency and thus compromises the integrity of BBB. Together, we describe a mechanism in which astrocytes regulate BBB integrity through mitochondrial transfer. Our findings provide innnovative insights into the cellular framework that underpins the progressive breakdown of BBB associated with aging and disease.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    神经炎症已成为癫痫和认知障碍的共同分子机制,为免疫反应和大脑功能之间复杂的相互作用提供了新的见解。证据显示高迁移率族蛋白1(HMGB1)参与血脑屏障破坏,并与癫痫严重程度和耐药性相关。虽然抗炎治疗显示出希望,翻译这些发现在阐明机制和开发可靠的生物标志物方面面临挑战。然而,战略性靶向神经炎症和HMGB1介导的炎症具有治疗潜力。这篇综述综合了关于癫痫和认知障碍中HMGB1和相关生物标志物的知识,以塑造针对这些复杂炎症过程的未来研究和治疗。
    Neuroinflammation has emerged as a shared molecular mechanism in epilepsy and cognitive impairment, offering new insights into the complex interplay between immune responses and brain function. Evidence reveals involvement of High mobility group box 1 (HMGB1) in blood-brain barrier disruption and correlations with epilepsy severity and drug resistance. While anti-inflammatory treatments show promise, translating these discoveries faces challenges in elucidating mechanisms and developing reliable biomarkers. However, strategically targeting neuroinflammation and HMGB1-mediated inflammation holds therapeutic potential. This review synthesises knowledge on HMGB1 and related biomarkers in epilepsy and cognitive impairment to shape future research and treatments targeting these intricate inflammatory processes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    血脑屏障(BBB)是大脑解剖学的重要组成部分,保护屏障,确保适当的营养运输到大脑,最终导致调节适当的大脑功能。然而,由于这种选择性,它在提供药物治疗中枢神经系统(CNS)疾病方面提出了重大挑战。已经设计了各种不同的载体来输送药物通过这个屏障来治疗神经退行性疾病,极大地影响了患者的生活质量。用于穿过BBB的两种主要类型的载体是聚合物和脂质体,这两者都封装药物以允许它们胞吞BBB的细胞。对于阿尔茨海默病,帕金森病,多发性硬化症,胶质母细胞瘤和脑癌,有许多不同的纳米颗粒治疗方法正在开发中,它们增加了现有药物的生物利用度和靶向能力或新的药物靶标,以减少这些疾病的症状。通过这些系统,纳米医学提供了一种靶向特定组织的新方法,尤其是中枢神经系统,和治疗疾病没有全身毒性,通常与目前使用的药物。
    The blood-brain barrier (BBB) is a crucial part of brain anatomy as it is a specialized, protective barrier that ensures proper nutrient transport to the brain, ultimately leading to regulating proper brain function. However, it presents a major challenge in delivering pharmaceuticals to treat central nervous system (CNS) diseases due to this selectivity. A variety of different vehicles have been designed to deliver drugs across this barrier to treat neurodegenerative diseases, greatly impacting the patient\'s quality of life. The two main types of vehicles used to cross the BBB are polymers and liposomes, which both encapsulate pharmaceuticals to allow them to transcytose the cells of the BBB. For Alzheimer\'s disease, Parkinson\'s disease, multiple sclerosis, and glioblastoma brain cancer, there are a variety of different nanoparticle treatments in development that increase the bioavailability and targeting ability of existing drugs or new drug targets to decrease symptoms of these diseases. Through these systems, nanomedicine offers a new way to target specific tissues, especially for the CNS, and treat diseases without the systemic toxicity that often comes with medications used currently.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    异双功能小分子降解剂是靶向蛋白质降解剂(TPDs)的一个子集,由通过接头连接的两个配体组成,以诱导目标蛋白的蛋白酶体降解。与传统的小分子相比,这些化合物通常表现出膨胀的物理化学性质,这可能需要创新的配方策略,使其能够交付和发挥药效学作用。血脑屏障(BBB)在人体生理中起着重要的作用,但它的存在需要先进的方法来治疗中枢神经系统(CNS)疾病。通过将TPDs等新兴模式与传统的药物递送概念相结合,可以开发克服BBB的新策略。在可用的路线中,基于脂质和聚合物的长效递送似乎是最适合TPDs的,由于它们能够包封亲脂性货物,并且有可能被功能化以进行靶向递送。另一个关键考虑因素是了解E3连接酶在大脑不同区域的表达。新的脑或CNS疾病特异性E3连接酶的发现可以帮助克服目前与TPD的CNS递送相关的一些障碍。这篇综述讨论了克服和改善TPDs向CNS的治疗性递送的现有策略。
    Heterobifunctional small molecule degraders are a subset of targeted protein degraders (TPDs), consisting of two ligands joined by a linker to induce proteasomal degradation of a target protein. As compared to traditional small molecules these compounds generally demonstrate inflated physicochemical properties, which may require innovative formulation strategies to enable their delivery and exert pharmacodynamic effect. The blood brain barrier (BBB) serves an essential function in human physiology, but its presence requires advanced approaches for treating central nervous system (CNS) diseases. By integrating emerging modalities like TPDs with conventional concepts of drug delivery, novel strategies to overcome the BBB can be developed. Amongst the available routes, lipid and polymer-based long-acting delivery seems to be the most amenable to TPDs, due to their ability to encapsulate lipophilic cargo and potential to be functionalized for targeted delivery. Another key consideration will be understanding E3 ligase expression in the different regions of the brain. Discovery of new brain or CNS disease specific E3 ligases could help overcome some of the barriers currently associated with CNS delivery of TPDs. This review discusses the current strategies that exist to overcome and improve therapeutic delivery of TPDs to the CNS.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    可以选择性去除大脑中的流氓蛋白的抗体是治疗神经退行性疾病(ND)的明显选择,但是经过几十年的努力,只有两种治疗阿尔茨海默病的抗体被批准,几十个处于测试阶段,一个人被撤回,另一个停了下来,可能是由于功效问题。然而,这些结果应该是明显的,因为这些抗体由于血脑屏障(BBB)保护剂而不能充分进入大脑.然而,所有产品都可以通过与转铁蛋白结合来恢复活力,最好是较小的片段。该模型可以快速且低成本地进行测试,应应用于bapineuzumab,solanezumab,克雷珠单抗,gantenerumab,aducanumab,lecanemab,Donanemab,Cinpanemab,和Gantenerumab,和他们的碎片。本文证明与转铁蛋白缀合不会改变与脑蛋白如淀粉样蛋白-β(Aβ)和α-突触核蛋白的结合。我们还提出了缀合物设计的选择,其将允许在进入脑后裂解以防止其胞吐作用,同时保持片段连接以实现与蛋白质的最佳结合。所识别的产品可以容易地进行测试并以最低的监管成本和延迟返回给患者。这些工程抗体可以通过重组工程制造,优选通过mRNA技术,作为一种更实惠的解决方案,以满足有效治疗神经退行性疾病的迫切需要。
    Antibodies that can selectively remove rogue proteins in the brain are an obvious choice to treat neurodegenerative disorders (NDs), but after decades of efforts, only two antibodies to treat Alzheimer\'s disease are approved, dozens are in the testing phase, and one was withdrawn, and the other halted, likely due to efficacy issues. However, these outcomes should have been evident since these antibodies cannot enter the brain sufficiently due to the blood-brain barrier (BBB) protectant. However, all products can be rejuvenated by binding them with transferrin, preferably as smaller fragments. This model can be tested quickly and at a low cost and should be applied to bapineuzumab, solanezumab, crenezumab, gantenerumab, aducanumab, lecanemab, donanemab, cinpanemab, and gantenerumab, and their fragments. This paper demonstrates that conjugating with transferrin does not alter the binding to brain proteins such as amyloid-β (Aβ) and α-synuclein. We also present a selection of conjugate designs that will allow cleavage upon entering the brain to prevent their exocytosis while keeping the fragments connected to enable optimal binding to proteins. The identified products can be readily tested and returned to patients with the lowest regulatory cost and delays. These engineered antibodies can be manufactured by recombinant engineering, preferably by mRNA technology, as a more affordable solution to meet the dire need to treat neurodegenerative disorders effectively.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    血清淀粉样蛋白A(SAA)蛋白是高度保守的脂蛋白,众所周知,它与急性期反应和系统性淀粉样变性有关。但是它们的生物学功能还不完全清楚。最近的研究表明,SAA蛋白可以通过穿过完整的血脑屏障(BBB)进入大脑,并且它们会损害BBB功能。一旦进入中枢神经系统(CNS),SAA蛋白可以同时具有保护作用和有害作用,这对中枢神经系统疾病有重要意义。在对SAA主题系列的回顾中,我们讨论了将SAA与神经炎症和中枢神经系统疾病相关的现有文献,以及血脑屏障在这些关系中的可能作用。
    Serum amyloid A (SAA) proteins are highly conserved lipoproteins that are notoriously involved in the acute phase response and systemic amyloidosis, but their biological functions are incompletely understood. Recent work has shown that SAA proteins can enter the brain by crossing the intact blood-brain barrier (BBB), and that they can impair BBB functions. Once in the central nervous system (CNS), SAA proteins can have both protective and harmful effects, which have important implications for CNS disease. In this review of the thematic series on SAA, we discuss the existing literature that relates SAA to neuroinflammation and CNS disease, and the possible roles of the BBB in these relations.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号