Hematoma absorption

  • 文章类型: Journal Article
    背景:脑出血(ICH)是一种破坏性的神经系统疾病,可导致严重的感觉运动功能障碍和认知功能下降,然而,目前尚无有效的治疗策略来缓解这些患者的预后.Mas轴介导的神经保护与各种神经系统疾病的病理有关,然而,Mas受体在ICH中的作用仍有待阐明.
    方法:C57BL/6小鼠纹状体注射胶原酶建立ICH模型。在ICH后鼻内施用Mas受体激动剂AVE0991(0.9mg/kg)。使用行为测试的组合,西方印迹,免疫荧光染色,血肿体积,脑水肿,定量PCR,TUNEL染色,Fluoro-JadeC染色,尼氏染色,和药理学方法,我们研究了鼻内应用AVE0991对ICH后血肿吸收和神经系统结局的影响,并研究了其潜在机制.
    结果:发现Mas受体在活化的小胶质细胞/巨噬细胞中显著表达,Mas受体在小胶质细胞/巨噬细胞中的峰值表达在大约3-5天观察到,随后下降。治疗后AVE0991激活Mas可促进血肿吸收,减少脑水肿,并改善ICH小鼠的短期和长期神经功能。此外,AVE0991治疗可有效减弱神经元凋亡,抑制中性粒细胞浸润,并减少ICH后血肿周围炎症细胞因子的释放。机械上,AVE0991后处理显着促进小胶质细胞/巨噬细胞向抗炎,吞噬,和修复表型,Mas抑制剂A779和Nrf2抑制剂ML385消除了通过Mas激活的小胶质细胞/巨噬细胞的这种功能表型转变。此外,ICH小胶质细胞清除后,AVE0991治疗的血肿清除和神经保护作用被逆转。
    结论:Mas激活可促进血肿吸收,改善神经功能缺损,缓解神经元凋亡,减少神经炎症,并通过Akt/Nrf2信号通路调节ICH后小胶质细胞/巨噬细胞的功能和表型。因此,鼻内应用Mas激动剂ACE0991可能为ICH患者的临床治疗提供有希望的策略。
    BACKGROUND: Intracerebral hemorrhage (ICH) is a devastating neurological disease causing severe sensorimotor dysfunction and cognitive decline, yet there is no effective treatment strategy to alleviate outcomes of these patients. The Mas axis-mediated neuroprotection is involved in the pathology of various neurological diseases, however, the role of the Mas receptor in the setting of ICH remains to be elucidated.
    METHODS: C57BL/6 mice were used to establish the ICH model by injection of collagenase into mice striatum. The Mas receptor agonist AVE0991 was administered intranasally (0.9 mg/kg) after ICH. Using a combination of behavioral tests, Western blots, immunofluorescence staining, hematoma volume, brain edema, quantitative-PCR, TUNEL staining, Fluoro-Jade C staining, Nissl staining, and pharmacological methods, we examined the impact of intranasal application of AVE0991 on hematoma absorption and neurological outcomes following ICH and investigated the underlying mechanism.
    RESULTS: Mas receptor was found to be significantly expressed in activated microglia/macrophages, and the peak expression of Mas receptor in microglia/macrophages was observed at approximately 3-5 days, followed by a subsequent decline. Activation of Mas by AVE0991 post-treatment promoted hematoma absorption, reduced brain edema, and improved both short- and long-term neurological functions in ICH mice. Moreover, AVE0991 treatment effectively attenuated neuronal apoptosis, inhibited neutrophil infiltration, and reduced the release of inflammatory cytokines in perihematomal areas after ICH. Mechanistically, AVE0991 post-treatment significantly promoted the transformation of microglia/macrophages towards an anti-inflammatory, phagocytic, and reparative phenotype, and this functional phenotypic transition of microglia/macrophages by Mas activation was abolished by both Mas inhibitor A779 and Nrf2 inhibitor ML385. Furthermore, hematoma clearance and neuroprotective effects of AVE0991 treatment were reversed after microglia depletion in ICH.
    CONCLUSIONS: Mas activation can promote hematoma absorption, ameliorate neurological deficits, alleviate neuron apoptosis, reduced neuroinflammation, and regulate the function and phenotype of microglia/macrophages via Akt/Nrf2 signaling pathway after ICH. Thus, intranasal application of Mas agonist ACE0991 may provide promising strategy for clinical treatment of ICH patients.
    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
    MIcroglia/巨噬细胞介导的红细胞吞噬作用在脑出血后血肿清除中起关键作用。动态细胞骨架变化伴随吞噬作用。然而,这些变化是否以及如何与小胶质细胞/巨噬细胞介导的红细胞吞噬作用相关尚不清楚.在这项研究中,我们研究了乙酰化α-微管蛋白的功能,稳定的微管形式,在体外和体内脑出血后的小胶质细胞/巨噬细胞红细胞吞噬中。我们首先使用与BV2小胶质细胞或RAW264.7巨噬细胞系共培养的原代DiOGFP标记的红细胞评估乙酰化α-微管蛋白在红细胞吞噬作用中的功能。在红细胞吞噬过程中,BV2和RAW264.7细胞中乙酰化α-微管蛋白的表达显着降低。此外,沉默α-微管蛋白乙酰转移酶1(ATAT1),一种新发现的α-微管蛋白乙酰转移酶,BV2和RAW264.7细胞降低了Ac-α-tub水平并增强了红细胞吞噬作用。与这些发现一致,在ATAT1-/-小鼠中,我们观察到血肿周围红细胞的离子化钙结合衔接分子1(Iba1)和Perls阳性小胶质细胞/巨噬细胞吞噬细胞增加,脑出血小鼠血肿体积减少.此外,敲除ATAT1减轻血肿周围神经元凋亡和促炎细胞因子和增加抗炎细胞因子,最终改善脑出血后小鼠的神经功能恢复。这些发现表明,ATAT1缺乏会加速小胶质细胞/巨噬细胞的红细胞吞噬作用和脑出血后的血肿吸收。这些结果为血肿清除机制提供了新的见解,并表明ATAT1是治疗脑出血的潜在靶标。
    MIcroglia/macrophage-mediated erythrophagocytosis plays a crucial role in hematoma clearance after intracerebral hemorrhage. Dynamic cytoskeletal changes accompany phagocytosis. However, whether and how these changes are associated with microglia/macrophage-mediated erythrophagocytosis remain unclear. In this study, we investigated the function of acetylated α-tubulin, a stabilized microtubule form, in microglia/macrophage erythrophagocytosis after intracerebral hemorrhage both in vitro and in vivo. We first assessed the function of acetylated α-tubulin in erythrophagocytosis using primary DiO GFP-labeled red blood cells co-cultured with the BV2 microglia or RAW264.7 macrophage cell lines. Acetylated α-tubulin expression was significantly decreased in BV2 and RAW264.7 cells during erythrophagocytosis. Moreover, silencing α-tubulin acetyltransferase 1 (ATAT1), a newly discovered α-tubulin acetyltransferase, decreased Ac-α-tub levels and enhanced the erythrophagocytosis by BV2 and RAW264.7 cells. Consistent with these findings, in ATAT1-/- mice, we observed increased ionized calcium binding adapter molecule 1 (Iba1) and Perls-positive microglia/macrophage phagocytes of red blood cells in peri-hematoma and reduced hematoma volume in mice with intracerebral hemorrhage. Additionally, knocking out ATAT1 alleviated neuronal apoptosis and pro-inflammatory cytokines and increased anti-inflammatory cytokines around the hematoma, ultimately improving neurological recovery of mice after intracerebral hemorrhage. These findings suggest that ATAT1 deficiency accelerates erythrophagocytosis by microglia/macrophages and hematoma absorption after intracerebral hemorrhage. These results provide novel insights into the mechanisms of hematoma clearance and suggest ATAT1 as a potential target for the treatment of intracerebral hemorrhage.
    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
    背景:脑出血(ICH)后血肿导致进行性神经功能缺损和不良预后。早期清除血肿被广泛认为是限制损伤和改善临床预后的必要治疗方法。小胶质细胞上的CD163,别名血红蛋白(Hb)清道夫受体,在血肿吸收中起着关键作用,但是神经元上的CD163允许Hb摄取并导致神经毒性。在这项研究中,我们关注如何特别促进小胶质细胞而不是神经元CD163介导的Hb摄取和血肿吸收。
    方法:RNA测序用于探索参与ICH进展的潜在分子,并通过磁共振成像(MRI)检测到血肿。应用Westernblot和免疫荧光技术评价脑出血后fractalkine(FKN)的表达和定位。通过红细胞吞噬试验研究FKN在血肿清除中的具体作用机制。利用小干扰RNA(siRNA)转染,探讨过氧化物酶体增殖物激活受体-γ(PPAR-γ)对血肿吸收的影响。采用酶联免疫吸附试验(ELISA)测定ICH患者血清FKN浓度。
    结果:在ICH后的小鼠模型中,发现血肿周围的FKN明显增加。以其在小胶质细胞中的独特受体CX3CR1,FKN显著降低血肿大小和Hb含量,和改善体内神经功能缺损。Further,FKN可以通过CD163/血红素加氧酶-1(HO-1)轴增强体外小胶质细胞的红细胞吞噬作用,而AZD8797(一种特定的CX3CR1抑制剂)逆转了这种作用。此外,发现PPAR-γ介导小胶质细胞中FKN诱导的CD163/HO-1轴表达和红细胞吞噬作用的增加。值得注意的是,在ICH患者中,较高的血清FKN水平与更好的血肿消退相关.
    结论:我们系统确定FKN可能是改善血肿吸收的潜在治疗靶点,并阐明ICH治疗。
    BACKGROUND: Hematoma leads to progressive neurological deficits and poor outcomes after intracerebral hemorrhage (ICH). Early clearance of hematoma is widely recognized as an essential treatment to limit the damage and improve the clinical prognosis. CD163, alias hemoglobin (Hb) scavenger receptor on microglia, plays a pivotal role in hematoma absorption, but CD163 on neurons permits Hb uptake and results in neurotoxicity. In this study, we focus on how to specially promote microglial but not neuronal CD163 mediated-Hb uptake and hematoma absorption.
    METHODS: RNA sequencing was used to explore the potential molecules involved in ICH progression, and hematoma was detected by magnetic resonance imaging (MRI). Western blot and immunofluorescence were used to evaluate the expression and location of fractalkine (FKN) after ICH. Erythrophagocytosis assay was performed to study the specific mechanism of action of FKN in hematoma clearance. Small interfering RNA (siRNA) transfection was used to explore the effect of peroxisome proliferator-activated receptor-γ (PPAR-γ) on hematoma absorption. Enzyme-linked immunosorbent assay (ELISA) was used to determine the serum FKN concentration in ICH patients.
    RESULTS: FKN was found to be significantly increased around the hematoma in a mouse model after ICH. With its unique receptor CX3CR1 in microglia, FKN significantly decreased the hematoma size and Hb content, and improved neurological deficits in vivo. Further, FKN could enhance erythrophagocytosis of microglia in vitro via the CD163/ hemeoxygenase-1 (HO-1) axis, while AZD8797 (a specific CX3CR1 inhibitor) reversed this effect. Moreover, PPAR-γ was found to mediate the increase in the CD163/HO-1 axis expression and erythrophagocytosis induced by FKN in microglia. Of note, a higher serum FKN level was found to be associated with better hematoma resolution in ICH patients.
    CONCLUSIONS: We systematically identified that FKN may be a potential therapeutic target to improve hematoma absorption and we shed light on ICH treatment.
    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
    Exosomes are small (30-150 nm diameter) lipid bilayer-enclosed vesicles found in all bodily fluids. We investigated whether exosomes play a role in chronic subdural hematoma (CSDH). Exosomes were identified and characterized using transmission electron microscopy and NanoSight particle tracking. The functions of hematoma-derived exosomes were evaluated in a rat model of acute subdural hematoma (SDH). The hematoma-derived exosomes inhibited hematoma absorption and exacerbated neurological deficits in SDH rats. We examined the effects of the exosomes on angiogenesis and cell permeability in human umbilical vein endothelial cells (HUVECs). Co-culture of exosomes with HUVECs revealed that the hematoma-derived exosomes were taken-in by the HUVECs, resulting in enhanced tube formation and vascular permeability. Additionally, there was a concomitant increase in ANG-2 expression and decrease in ANG-1 expression. Exosomes were enriched with microRNAs including miR-144-5p, which they could deliver to HUVECs to promote angiogenesis and increase membrane permeability. Overexpression of miR-144-5p in HUVECs and in SDH rats promoted abnormal angiogenesis and reduced hematoma absorption, which mimicked the effects of the hematoma-derived exosomes both in vitro and in vivo. Thus, hematoma-derived exosomes promote abnormal angiogenesis with high permeability and inhibit hematoma absorption through miR-144-5p in CSDH.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号