neonatal brain injury

新生儿脑损伤
  • 文章类型: Editorial
    暂无摘要。
    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
    引言补体反应在新生儿缺氧缺血性脑病(HIE)的再灌注时激活,并导致过度的神经炎症和更差的结果。C5a是一种强大的过敏毒素,对每个补体途径至关重要,它与C5aR1的参与与脑损伤和神经元死亡直接相关。推断C5aR1拮抗作用可以减少过度的神经炎症,从而改善神经和功能结果,我们用抑制C5a-C5aR1相互作用的小分子PMX205在HIE大鼠模型中检验了这一假设.方法采用Vannucci方法对足月等效幼崽(P10-12)进行轻中度HIE,并用PMX205治疗。我们将运动和认知结果与两个行为测试进行了比较[食物处理和加速;新颖的对象识别(NOR)和开放领域],以提高我们结论的准确性。结果观察到精细运动功能的改善,平衡和探索行为,但识别记忆和粗大运动功能几乎没有改善。病变面积和组织学评估显示,对治疗的皮质神经保护作用很强,但对海马CA1区的持续损伤。治疗1天后,观察到更好的结构和功能结果,提示C5aR1拮抗作用超过潜在损伤阶段可能会损害恢复。在剂量反应实验中,仅在雌性大鼠中,损伤造成的大脑面积损失得到改善,提示补体反应中潜在的性二态。结论这些结果证明了用PMX205在新生儿HIE中靶向C5aR1信号传导的概念验证,并强调了性别在缺氧缺血性损伤中的作用。
    BACKGROUND: The complement response activates upon reperfusion in neonatal hypoxic-ischemic encephalopathy (HIE) and contributes to excessive neuroinflammation and worse outcomes. C5a is a powerful anaphylatoxin central to each of the complement pathways, and its engagement with C5aR1 is directly tied to brain injury and neuronal death. Reasoning C5aR1 antagonism can decrease excessive neuroinflammation and thereby improve neurological and functional outcomes, we tested this hypothesis in a rat model of HIE with PMX205, a small molecule that inhibits C5a-C5aR1 interaction.
    METHODS: Term-equivalent pups (P10-12) were subjected to mild-moderate HIE by Vannucci\'s method and treated with PMX205. We compared motor and cognitive outcomes with two behavioral tests each (food handling and accelerod; novel object recognition [NOR] and open field) to improve the accuracy of our conclusions.
    RESULTS: Improvements were observed in fine motor function, balance, and exploratory behaviors, but little to no improvement in recognition memory and gross motor function. Lesion area and histological assessments showed robust cortical neuroprotection from treatment but persistent injury to the CA1 region of the hippocampus. Better structural and functional outcomes were seen within 1 day of treatment, suggesting C5aR1 antagonism beyond the latent injury phase may impair recovery. In a dose-response experiment, cerebral area loss from injury was improved only in female rats, suggesting underlying sexual dimorphisms in the complement response.
    CONCLUSIONS: These results demonstrate proof-of-concept for targeting C5aR1 signaling in neonatal HIE with PMX205 and underscore the role of sex in hypoxic-ischemic injury.
    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
    缺氧缺血(HI)是新生儿脑损伤的主要原因之一。线粒体自噬与新生儿脑HI损伤后受损线粒体的降解和细胞存活有关。Pleckstrin同源结构域家族A成员1(PHLDA1)在各种疾病的进展中起着至关重要的作用,包括氧化应激的调节,免疫反应和细胞凋亡。在本研究中,我们研究了PHLDA1在HI诱导的神经元损伤中的作用,并进一步探索了体内和体外PHLDA1调节线粒体自噬的机制。通过结扎左颈总动脉并暴露于具有8%O2和92%N2的缺氧室,在新生大鼠中建立HI模型。在经历氧和葡萄糖剥夺/复氧(OGD/R)的原代海马神经元中进行体外研究。我们表明,在HI新生大鼠的海马和OGD/R处理的原代神经元中,PHLDA1的表达显着上调。通过慢病毒载体敲除新生大鼠PHLDA1不仅显着改善HI诱导的海马神经元损伤,而且显着改善长期认知功能结果,而通过慢病毒载体在新生大鼠中过度表达PHLDA1会加重这些结果。原代神经元中的PHLDA1敲低可显着逆转细胞活力的降低和细胞内活性氧(ROS)水平的增加,并减轻OGD诱导的线粒体功能障碍,而PHLDA1的过表达降低了这些参数。在OGD/R处理的原代海马神经元中,我们发现,PHLDA1敲低可通过激活FUNDC1增强线粒体自噬,而FUNDC1敲低或用线粒体自噬抑制剂Mdivi-1(25μM)预处理可消除线粒体自噬.值得注意的是,Mdivi-1预处理或FUNDC1敲低不仅增加了脑梗塞体积,而且还取消了PHLDA1敲低对HI新生大鼠的神经保护作用。一起,这些结果表明,PHLDA1通过抑制FUNDC1介导的神经元线粒体自噬,有助于新生儿HI诱导的脑损伤.
    Hypoxia-ischemia (HI) is one of the main causes of neonatal brain injury. Mitophagy has been implicated in the degradation of damaged mitochondria and cell survival following neonatal brain HI injury. Pleckstrin homology-like domain family A member 1 (PHLDA1) plays vital roles in the progression of various disorders including the regulation of oxidative stress, the immune responses and apoptosis. In the present study we investigated the role of PHLDA1 in HI-induced neuronal injury and further explored the mechanisms underlying PHLDA1-regulated mitophagy in vivo and in vitro. HI model was established in newborn rats by ligation of the left common carotid artery plus exposure to an oxygen-deficient chamber with 8% O2 and 92% N2. In vitro studies were conducted in primary hippocampal neurons subjected to oxygen and glucose deprivation/-reoxygenation (OGD/R). We showed that the expression of PHLDA1 was significantly upregulated in the hippocampus of HI newborn rats and in OGD/R-treated primary neurons. Knockdown of PHLDA1 in neonatal rats via lentiviral vector not only significantly ameliorated HI-induced hippocampal neuronal injury but also markedly improved long-term cognitive function outcomes, whereas overexpression of PHLDA1 in neonatal rats via lentiviral vector aggravated these outcomes. PHLDA1 knockdown in primary neurons significantly reversed the reduction of cell viability and increase in intracellular reactive oxygen species (ROS) levels, and attenuated OGD-induced mitochondrial dysfunction, whereas overexpression of PHLDA1 decreased these parameters. In OGD/R-treated primary hippocampal neurons, we revealed that PHLDA1 knockdown enhanced mitophagy by activating FUNDC1, which was abolished by FUNDC1 knockdown or pretreatment with mitophagy inhibitor Mdivi-1 (25 μM). Notably, pretreatment with Mdivi-1 or the knockdown of FUNDC1 not only increased brain infarct volume, but also abolished the neuroprotective effect of PHLDA1 knockdown in HI newborn rats. Together, these results demonstrate that PHLDA1 contributes to neonatal HI-induced brain injury via inhibition of FUNDC1-mediated neuronal mitophagy.
    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
    含有连接结构域的蛋白聚糖修饰细胞外基质(ECM)以调节细胞稳态,并且还可以使组织/器官对损伤和应激敏感。缺氧缺血性(H-I)损伤通过激活炎症并减弱再生和修复途径来破坏细胞稳态。在大脑中,ECM的主要成分是糖胺聚糖(GAG),透明质酸(HA),但是ECM的HA修饰是否调节细胞稳态和对H-I损伤的反应尚不清楚。在这份报告中,同时雇用雄性和雌性老鼠,我们证明了含有蛋白聚糖的连接域,TNFα刺激基因-6(TSG-6),从出生开始在大脑中活跃,并在离散的神经发育窗中差异修饰ECMHA。TSG-6的ECMHA修饰使其能够作为发育开关来调节Hippo途径效应蛋白的活性,是的,成熟的大脑中的相关蛋白1(YAP1)和对H-I损伤的反应。缺乏TSG-6表达的小鼠表现出YAP1靶标的表达失调,兴奋性氨基酸转运蛋白1(EAAT1;GLAST)和2(EAAT2;GLT-1)。TSG-6-/-小鼠中YAP1激活的失调与大脑对H-I损伤的年龄和性别依赖性致敏一致,因此1周大的新生儿表现出抗炎反应,而不是3个月大的成年男性而不是女性的促炎损伤反应。因此,我们的发现支持小鼠大脑中年龄和性别依赖性H-I损伤反应的关键调节因子是TSG-6依赖性ECM修饰对Hippo-YAP1途径的调节。意义声明缺氧缺血性(H-I)损伤是全球发病率和死亡率的常见原因。许多基因组和蛋白质组筛选已经确定细胞外基质(ECM)的变化是最常见的发现,然而,下游机制的见解仍然模糊。我们发现蛋白聚糖,TNF刺激的基因-6(TSG-6)依赖性ECM修饰调节Hippo途径效应子的活性,RNA转录共激活因子,是相关蛋白1(YAP1)。TSG-6-/-小鼠中YAP1激活的失调与大脑对H-I损伤的年龄和性别依赖性致敏同时发生,因此与成人中增强的促炎损伤反应相反,新生儿表现出抗炎反应。因此,我们的研究结果将TSG-6依赖性ECM修饰确立为年龄和性别依赖性H-I损伤反应的关键调节因子.
    Proteoglycans containing link domains modify the extracellular matrix (ECM) to regulate cellular homeostasis and can also sensitize tissues/organs to injury and stress. Hypoxic-ischemic (H-I) injury disrupts cellular homeostasis by activating inflammation and attenuating regeneration and repair pathways. In the brain, the main component of the ECM is the glycosaminoglycan hyaluronic acid (HA), but whether HA modifications of the ECM regulate cellular homeostasis and response to H-I injury is not known. In this report, employing both male and female mice, we demonstrate that link-domain-containing proteoglycan, TNFα-stimulated gene-6 (TSG-6), is active in the brain from birth onward and differentially modifies ECM HA during discrete neurodevelopmental windows. ECM HA modification by TSG-6 enables it to serve as a developmental switch to regulate the activity of the Hippo pathway effector protein, yes-associated protein 1 (YAP1), in the maturing brain and in response to H-I injury. Mice that lack TSG-6 expression display dysregulated expression of YAP1 targets, excitatory amino acid transporter 1 (EAAT1; glutamate-aspartate transporter) and 2 (EAAT2; glutamate transporter-1). Dysregulation of YAP1 activation in TSG-6-/- mice coincides with age- and sex-dependent sensitization of the brain to H-I injury such that 1-week-old neonates display an anti-inflammatory response in contrast to an enhanced proinflammatory injury reaction in 3-month-old adult males but not females. Our findings thus support that a key regulator of age- and sex-dependent H-I injury response in the mouse brain is modulation of the Hippo-YAP1 pathway by TSG-6-dependent ECM modifications.
    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
    脑损伤与早产高度相关。早产并发症,包括自发性或坏死性小肠结肠炎(NEC)相关肠穿孔,与终身神经功能缺损有关,然而,对机制知之甚少。早产脑损伤的早期诊断仍然是一个重大挑战。这里,在肠穿孔后的早产儿中,我们通过头颅超声检测出脑室下区回声(SVE).SVE的发展与2年的运动障碍显着相关。在肠穿孔的新生小鼠模型中复制SVE。对鼠回声室下区(SVZ)的检查显示,多纤毛FoxJ1室管膜细胞中的NLRP3-炎性体组装,并且在该出生后干细胞壁龛中室管膜边界丢失。这些数据表明,尚未充分考虑到SVZ局部早产脑损伤的机制。超声检测SVE可作为早产儿炎症性疾病后神经发育障碍的早期生物标志物。
    Brain injury is highly associated with preterm birth. Complications of prematurity, including spontaneous or necrotizing enterocolitis (NEC)-associated intestinal perforations, are linked to lifelong neurologic impairment, yet the mechanisms are poorly understood. Early diagnosis of preterm brain injuries remains a significant challenge. Here, we identified subventricular zone echogenicity (SVE) on cranial ultrasound in preterm infants following intestinal perforations. The development of SVE was significantly associated with motor impairment at 2 years. SVE was replicated in a neonatal mouse model of intestinal perforation. Examination of the murine echogenic subventricular zone (SVZ) revealed NLRP3-inflammasome assembly in multiciliated FoxJ1+ ependymal cells and a loss of the ependymal border in this postnatal stem cell niche. These data suggest a mechanism of preterm brain injury localized to the SVZ that has not been adequately considered. Ultrasound detection of SVE may serve as an early biomarker for neurodevelopmental impairment after inflammatory disease in preterm infants.
    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
    肠道微生物组与患有脑损伤的极早产儿的病理性神经生理演变有关。婴儿的确切潜在机制及其相关代谢特征尚未完全了解。为了破译与新生儿脑损伤相关的代谢物谱,我们调查了在几个时间点从51例极度早产儿队列中获得的粪便和血浆代谢组,使用基于液相色谱(LC)-高分辨率质谱(MS)的非靶向代谢组学和基于LC-MS/MS的靶向分析来研究胆汁酸和酰胺化胆汁酸缀合物。数据与16SrRNA基因扩增子肠道微生物组谱以及患者细胞因子整合,生长因子,和T细胞图谱。我们发现有和没有脑损伤的婴儿之间神经活性代谢物的早期分化。我们在血浆和粪便中检测到几种细菌来源的胆汁酸氨基酸缀合物。这些结果提供了对极早产儿早期代谢组的见解。
    The gut microbiome is associated with pathological neurophysiological evolvement in extremely premature infants suffering from brain injury. The exact underlying mechanism and its associated metabolic signatures in infants are not fully understood. To decipher metabolite profiles linked to neonatal brain injury, we investigate the fecal and plasma metabolome of samples obtained from a cohort of 51 extremely premature infants at several time points, using liquid chromatography (LC)-high-resolution mass spectrometry (MS)-based untargeted metabolomics and LC-MS/MS-based targeted analysis for investigating bile acids and amidated bile acid conjugates. The data are integrated with 16S rRNA gene amplicon gut microbiome profiles as well as patient cytokine, growth factor, and T cell profiles. We find an early onset of differentiation in neuroactive metabolites between infants with and without brain injury. We detect several bacterially derived bile acid amino acid conjugates in plasma and feces. These results provide insights into the early-life metabolome of extremely premature infants.
    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
    蛋白酶体功能丧失,蛋白质病,和蛋白毒性可能导致神经变性在人类的整个生命周期中的几种形式的脑损伤和疾病。因此,体内激活脑蛋白酶体的药物可能在神经病学中具有广泛的治疗作用。
    使用猪,一种临床相关的大型动物,具有功能房室性脑皮层,我们评估了脑蛋白酶体的定位和生化活性,并测试了小分子激活脑蛋白酶体的能力。
    通过蛋白质印迹,蛋白酶体蛋白亚基PSMB5和PSMA3水平在不同猪脑区域相似。PSMB5的免疫组织化学显示位于细胞质(弥散和颗粒)和细胞核(细胞质<细胞核)中。某些PSMB5免疫反应性与线粒体(电压门控阴离子通道和亲环素D)和细胞死亡(Aven)蛋白共定位在猪和人脑新皮层的神经元体细胞和神经纤维中。在细胞核中,PSMB5免疫反应性弥漫性,颗粒,并聚集,包括核仁周围的装饰。通过荧光分析,在八个不同的猪脑区域中,粗组织可溶性部分中的蛋白酶体胰凝乳蛋白酶样活性(CTL)通常相似。海马中的蛋白酶体CTL活性与鼻粘膜活检中的活性相关。在猪大脑皮层亚细胞部分的初步分析中,蛋白酶体CTL活性在细胞质中最高,然后在核部分中降低约50%;约15-20%的总CTL活性在纯线粒体部分中。用凝胶内活性测定,26S-单端和双端蛋白酶体是猪大脑皮层中的主要形式。通过一种新颖的原位组织化学活性测定,MG132抑制蛋白酶体CTL活性定位于神经纤维,作为马赛克,和细胞体,原子核,和类似中心体的核周卫星。在24小时内用吡唑啉酮衍生物和氯丙嗪静脉注射的仔猪中,脑蛋白酶体CTL活性适度增加。
    这项研究表明,猪脑中的蛋白酶体具有相对的区域均匀性,具有催化活性的突出的核和核周存在,线粒体与活动的联系,26S-单上限优势,和适应症从小分子全身给药吡唑啉酮衍生物和氯丙嗪脑蛋白酶体功能似乎安全激活。
    UNASSIGNED: Loss of proteasome function, proteinopathy, and proteotoxicity may cause neurodegeneration across the human lifespan in several forms of brain injury and disease. Drugs that activate brain proteasomes in vivo could thus have a broad therapeutic impact in neurology.
    UNASSIGNED: Using pigs, a clinically relevant large animal with a functionally compartmental gyrencephalic cerebral cortex, we evaluated the localization and biochemical activity of brain proteasomes and tested the ability of small molecules to activate brain proteasomes.
    UNASSIGNED: By Western blotting, proteasome protein subunit PSMB5 and PSMA3 levels were similar in different pig brain regions. Immunohistochemistry for PSMB5 showed localization in the cytoplasm (diffuse and particulate) and nucleus (cytoplasm < nucleus). Some PSMB5 immunoreactivity was colocalized with mitochondrial (voltage-gated anion channel and cyclophilin D) and cell death (Aven) proteins in the neuronal soma and neuropil in the neocortex of pig and human brains. In the nucleus, PSMB5 immunoreactivity was diffuse, particulate, and clustered, including perinucleolar decorations. By fluorogenic assay, proteasome chymotrypsin-like activities (CTL) in crude tissue soluble fractions were generally similar within eight different pig brain regions. Proteasome CTL activity in the hippocampus was correlated with activity in nasal mucosa biopsies. In pilot analyses of subcellular fractions of pig cerebral cortex, proteasome CTL activity was highest in the cytosol and then ~50% lower in nuclear fractions; ~15-20% of total CTL activity was in pure mitochondrial fractions. With in-gel activity assay, 26S-singly and -doubly capped proteasomes were the dominant forms in the pig cerebral cortex. With a novel in situ histochemical activity assay, MG132-inhibitable proteasome CTL activity was localized to the neuropil, as a mosaic, and to cell bodies, nuclei, and centrosome-like perinuclear satellites. In piglets treated intravenously with pyrazolone derivative and chlorpromazine over 24 h, brain proteasome CTL activity was modestly increased.
    UNASSIGNED: This study shows that the proteasome in the pig brain has relative regional uniformity, prominent nuclear and perinuclear presence with catalytic activity, a mitochondrial association with activity, 26S-single cap dominance, and indications from small molecule systemic administration of pyrazolone derivative and chlorpromazine that brain proteasome function appears safely activable.
    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
    由于神经干细胞(NSC)的存在,生发活动在出生后前脑的心室-心室下区(V-SVZ)中持续存在。越来越多的证据表明,早期脑损伤后这些细胞被募集,并表明它们对操纵的适应性。我们使用慢性缺氧作为早期脑损伤的啮齿动物模型,以研究出生后皮质祖细胞的再激活。我们的结果表明,背侧V-SVZ中谷氨酸能祖细胞的增殖和产生增加。V-SVZNSC的命运作图证明了它们对从头皮质神经发生的贡献。谷氨酸能祖细胞的转录分析显示甲基转移酶14(Mettl14)和Wnt/β-catenin信号传导的平行变化。在协议中,通过Mettl14和Wnt/β-catenin途径的遗传和药理激活进行操作,分别,诱导神经发生并促进新形成的细胞成熟。最后,年轻成年NSC的标记表明,药理NSC激活对V-SVZNSC的储库及其生发活性没有不利影响。
    Germinal activity persists throughout life within the ventricular-subventricular zone (V-SVZ) of the postnatal forebrain due to the presence of neural stem cells (NSCs). Accumulating evidence points to a recruitment for these cells following early brain injuries and suggests their amenability to manipulations. We used chronic hypoxia as a rodent model of early brain injury to investigate the reactivation of cortical progenitors at postnatal times. Our results reveal an increased proliferation and production of glutamatergic progenitors within the dorsal V-SVZ. Fate mapping of V-SVZ NSCs demonstrates their contribution to de novo cortical neurogenesis. Transcriptional analysis of glutamatergic progenitors shows parallel changes in methyltransferase 14 (Mettl14) and Wnt/β-catenin signaling. In agreement, manipulations through genetic and pharmacological activation of Mettl14 and the Wnt/β-catenin pathway, respectively, induce neurogenesis and promote newly-formed cell maturation. Finally, labeling of young adult NSCs demonstrates that pharmacological NSC activation has no adverse effects on the reservoir of V-SVZ NSCs and on their germinal activity.
    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
    (1)研究背景:新生儿脑损伤可导致永久性神经发育障碍。值得注意的是,抑制炎症途径可以减少损伤。确定神经炎症在新生儿脑损伤进展中的作用,我们研究了免疫抑制剂他克莫司在两个时间点治疗新生大鼠幼崽的效果:缺氧缺血(HI)诱导的损伤之前和之后。(2)方法:诱发HI损伤,出生后一天(PND)10只大鼠幼崽接受单颈动脉结扎,然后缺氧(8%氧气,90分钟)。幼犬在PND7的HI前3天开始每日服用他克莫司(或车辆)(HI前),或HI后12小时(后HI)。测试四种剂量:0.025、0.05、0.1或0.25mg/kg/天。幼仔在PND17或PND50处安乐死。(3)结果:所有他克莫司剂量给予pre-HI显著减少脑梗塞面积和神经元丢失,增加了静息小胶质细胞的数量并减少了细胞凋亡(与对照组相比,p<0.05)。相比之下,只有最高剂量的他克莫司在HI后给药(0.25mg/kg/天)可减少脑梗死面积(p<0.05).与对照相比,所有剂量的他克莫司降低了幼崽的重量。(4)结论:他克莫司给药3天前HI是神经保护性的,可能是通过神经炎症和细胞死亡途径介导的。HI后他克莫司减少脑损伤的能力可能有限,更高的剂量会增加老鼠幼崽的死亡率。这项工作强调了在急性损伤期靶向神经炎症的益处。更具体的靶向神经炎症,例如,通过T细胞,需要进一步调查。
    (1) Background: Neonatal brain injury can lead to permanent neurodevelopmental impairments. Notably, suppressing inflammatory pathways may reduce damage. To determine the role of neuroinflammation in the progression of neonatal brain injury, we investigated the effect of treating neonatal rat pups with the immunosuppressant tacrolimus at two time points: before and after hypoxic-ischaemic (HI)-induced injury. (2) Methods: To induce HI injury, postnatal day (PND) 10 rat pups underwent single carotid artery ligation followed by hypoxia (8% oxygen, 90 min). Pups received daily tacrolimus (or a vehicle) starting either 3 days before HI on PND 7 (pre-HI), or 12 h after HI (post-HI). Four doses were tested: 0.025, 0.05, 0.1 or 0.25 mg/kg/day. Pups were euthanised at PND 17 or PND 50. (3) Results: All tacrolimus doses administered pre-HI significantly reduced brain infarct size and neuronal loss, increased the number of resting microglia and reduced cellular apoptosis (p < 0.05 compared to control). In contrast, only the highest dose of tacrolimus administered post-HI (0.25 mg/kg/day) reduced brain infarct size (p < 0.05). All doses of tacrolimus reduced pup weight compared to the controls. (4) Conclusions: Tacrolimus administration 3 days pre-HI was neuroprotective, likely mediated through neuroinflammatory and cell death pathways. Tacrolimus post-HI may have limited capacity to reduce brain injury, with higher doses increasing rat pup mortality. This work highlights the benefits of targeting neuroinflammation during the acute injurious period. More specific targeting of neuroinflammation, e.g., via T-cells, warrants further investigation.
    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
    尽管成年人的大脑疤痕已经得到了广泛的研究,关于新生儿期瘢痕形成的可用数据较少,在新生儿中,外周免疫细胞在这一过程中的参与仍未被探索。使用新生儿缺氧缺血性脑病(HIE)的小鼠模型和共聚焦显微镜,我们对瘢痕形成过程进行了表征,并检查了损伤后1年外周免疫细胞向皮质和海马瘢痕的募集。观察到瘢痕形成的区域差异,包括皮质中存在网状纤维化网络和海马血管周围纤维化。我们确定了在两个地区都有慢性升高水平的趋化因子,通过基于共生的策略,淋巴细胞的募集,中性粒细胞,和单核细胞来源的巨噬细胞在新生儿损伤后几周出现疤痕。一年后,然而,疤痕中没有中性粒细胞和淋巴细胞。我们的数据表明,外周免疫细胞是HIE诱导的脑疤痕的瞬时成分,为后期治疗干预开辟了新的可能性。
    Although brain scars in adults have been extensively studied, there is less data available regarding scar formation during the neonatal period, and the involvement of peripheral immune cells in this process remains unexplored in neonates. Using a murine model of neonatal hypoxic-ischemic encephalopathy (HIE) and confocal microscopy, we characterized the scarring process and examined the recruitment of peripheral immune cells to cortical and hippocampal scars for up to 1 year post-insult. Regional differences in scar formation were observed, including the presence of reticular fibrotic networks in the cortex and perivascular fibrosis in the hippocampus. We identified chemokines with chronically elevated levels in both regions and demonstrated, through a parabiosis-based strategy, the recruitment of lymphocytes, neutrophils, and monocyte-derived macrophages to the scars several weeks after the neonatal insult. After 1 year, however, neutrophils and lymphocytes were absent from the scars. Our data indicate that peripheral immune cells are transient components of HIE-induced brain scars, opening up new possibilities for late therapeutic interventions.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号