Oligodendrocyte progenitor cells

少突胶质祖细胞
  • 文章类型: Journal Article
    脊髓损伤(SCI)可导致损伤水平以下的感觉和运动功能丧失,严重威胁人类的健康和生活质量。神经系统损伤后病理变化的一个重要特征是脱髓鞘,部分导致损伤后神经功能的长期缺陷。中枢神经系统(CNS)的髓鞘再生主要由少突胶质祖细胞(OPCs)介导。许多复杂的细胞内信号和转录因子调节从OPCs到成熟少突胶质细胞(OLs)和髓鞘形成的分化过程。研究表明microRNA(miRNA)在调节OPC功能中的重要性。在这次审查中,本文对SCI后脱髓鞘和髓鞘再生进行了综述,这可能为SCI治疗提供潜在的治疗靶点。
    Spinal cord injury (SCI) can cause loss of sensory and motor function below the level of injury, posing a serious threat to human health and quality of life. One significant characteristic feature of pathological changes following injury in the nervous system is demyelination, which partially contributes to the long-term deficits in neural function after injury. The remyelination in the central nervous system (CNS) is mainly mediated by oligodendrocyte progenitor cells (OPCs). Numerous complex intracellular signaling and transcriptional factors regulate the differentiation process from OPCs to mature oligodendrocytes (OLs) and myelination. Studies have shown the importance of microRNA (miRNA) in regulating OPC functions. In this review, we focus on the demyelination and remyelination after SCI, and summarize the progress of miRNAs on OPC functions and remyelination, which might provide a potential therapeutic target for SCI treatments.
    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:脊髓损伤(SCI)是一种破坏性疾病,可引起少突胶质细胞和神经元的广泛损伤,导致脱髓鞘和轴突变性。在这项研究中,我们共移植了包含来自人诱导多能干细胞(iPSCs)的少突胶质细胞祖细胞(OPCs)与人脐静脉内皮细胞(HUVECs)的细胞移植物,据报道,它们促进了OPCs的生存和迁移,入大鼠挫伤模型以促进SCI后功能恢复。
    方法:OPCs来自iPSCs,并在不同时间点通过免疫荧光进行鉴定。进行体外功能测定以评估HUVECs对增殖的影响。迁移,以及通过共培养和迁移测定OPCs的存活,以及神经元轴突的生长。将OPCs和HUVEC的组合移植到大鼠挫伤模型中。8周后,免疫荧光染色检测移植细胞的安全性,观察神经元修复情况,髓鞘形成,和受伤区域的神经回路重建;功能恢复由Basso评估,Beattie,和Bresnahan开放领域规模,爬梯,SEP,MEP。此外,HUVECs对移植物的影响也在体内测定。
    结果:数据显示HUVECs促进其增殖,迁移,和OPCs在体外和体内的存活。此外,植入后8周,OPCs和HUVECs共同移植的大鼠明显促进了髓鞘再生,增强移植物与宿主之间的功能连接,促进功能恢复。此外,与OPCs单独移植相比,共同移植在病变边界产生了更多的感觉神经元,并显着改善了感觉功能恢复。
    结论:我们的研究表明,OPCs联合HUVEC移植可显著提高SCI后的运动和感觉功能恢复。OPCs联合HUVECs组和OPCs单独组的运动功能恢复无显著性,OPCs联合HUVECs组的感觉功能恢复明显优于其他两组。这些发现为SCI研究领域提供了新的见解。
    BACKGROUND: Spinal cord injury (SCI) is a devastating disease that causes extensive damage to oligodendrocytes and neurons leading to demyelination and axonal degeneration. In this study, we co-transplanted cell grafts containing oligodendrocyte progenitor cells (OPCs) derived from human-induced pluripotent stem cells (iPSCs) combined with human umbilical vein endothelial cells (HUVECs), which were reported to promote OPCs survival and migration, into rat contusion models to promote functional recovery after SCI.
    METHODS: OPCs were derived from iPSCs and identified by immunofluorescence at different time points. Functional assays in vitro were performed to evaluate the effect of HUVECs on the proliferation, migration, and survival of OPCs by co-culture and migration assay, as well as on the neuronal axonal growth. A combination of OPCs and HUVECs was transplanted into the rat contusive model. Upon 8 weeks, immunofluorescence staining was performed to test the safety of transplanted cells and to observe the neuronal repairment, myelination, and neural circuit reconstruction at the injured area; also, the functional recovery was assessed by Basso, Beattie, and Bresnahan open-field scale, Ladder climb, SEP, and MEP. Furthermore, the effect of HUVECs on grafts was also determined in vivo.
    RESULTS: Data showed that HUVECs promote the proliferation, migration, and survival of OPCs both in vitro and in vivo. Furthermore, 8 weeks upon engraftment, the rats with OPCs and HUVECs co-transplantation noticeably facilitated remyelination, enhanced functional connection between the grafts and the host and promoted functional recovery. In addition, compared with the OPCs-alone transplantation, the co-transplantation generated more sensory neurons at the lesion border and significantly improved the sensory functional recovery.
    CONCLUSIONS: Our study demonstrates that transplantation of OPCs combined with HUVECs significantly enhances both motor and sensory functional recovery after SCI. No significance was observed between OPCs combined with HUVECs group and OPCs-alone group in motor function recovery, while the sensory function recovery was significantly promoted in OPCs combined with HUVECs groups compared with the other two groups. These findings provide novel insights into the field of SCI research.
    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
    脱髓鞘是各种中枢神经系统疾病中常见的病理特征。它的特征是病变区域少突胶质祖细胞(OPCs)的聚集,在分化为成熟少突胶质细胞(OLG)方面面临困难。OPCs的分化需要Sox10的存在,但其表达在病理条件下降低。因此,我们提出了一种通过将Sox10内源性加载到外泌体中来调节OPCs分化和实现髓鞘修复的治疗策略。要做到这一点,我们产生了一个慢病毒武装的Sox10,可以锚定到外泌体膜的内表面。然后我们感染HEK293细胞以获得高表达Sox10的外泌体(exosomes-Sox10,Exoss)。体外,实验证实,Exos和Exos都可以被OPC吸收,但只有ExOS对OPCs表现出促分化作用。在体内,我们管理PBS,Exos,和ExoSs对Cuprizone诱导的脱髓鞘小鼠。结果表明,与其他组相比,Exoss可以调节PDGFRαOPCs向APCOLG的分化,并减少小鼠大脑call体区域的髓磷脂损伤。进一步的测试表明,Sox10可能通过增强MBP的表达对髓鞘有修复作用,可能是由外泌体将蛋白质递送到病变中促进的。这种内源性负载技术有望作为基于蛋白质的药物治疗脱髓鞘疾病的策略。
    Demyelination is a pathological feature commonly observed in various central nervous system diseases. It is characterized by the aggregation of oligodendrocyte progenitor cells (OPCs) in the lesion area, which face difficulties in differentiating into mature oligodendrocytes (OLGs). The differentiation of OPCs requires the presence of Sox10, but its expression decreases under pathological conditions. Therefore, we propose a therapeutic strategy to regulate OPCs differentiation and achieve myelin repair by endogenously loading Sox10 into exosomes. To accomplish this, we generated a lentivirus-armed Sox10 that could anchor to the inner surface of the exosome membrane. We then infected HEK293 cells to obtain exosomes with high expression of Sox10 (exosomes-Sox10, ExoSs). In vitro, experiments confirmed that both Exos and ExoSs can be uptaken by OPCs, but only ExoSs exhibit a pro-differentiation effect on OPCs. In vivo, we administered PBS, Exos, and ExoSs to cuprizone-induced demyelinating mice. The results demonstrated that ExoSs can regulate the differentiation of PDGFRα+ OPCs into APC+ OLGs and reduce myelin damage in the corpus callosum region of the mouse brain compared to other groups. Further testing suggests that Sox10 may have a reparative effect on the myelin sheath by enhancing the expression of MBP, possibly facilitated by the exosome delivery of the protein into the lesion. This endogenously loaded technology holds promise as a strategy for protein-based drugs in the treatment of demyelinating diseases.
    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
    活性氧(ROS)的积累,尤其是脂质,诱导神经元和少突胶质祖细胞(OPCs)大量细胞死亡,并导致中风后严重的神经功能缺损。虽然小化合物,比如去铁胺,已经证明,Lipostatin-1和Ferratin-1可以有效减少脂质ROS,内源性保护性分子对抗脂质ROS积累和随后细胞死亡的机制尚不清楚,尤其是在OPC中,这对于维持脑白质完整性和改善卒中后的长期结局至关重要.这里,使用小鼠初级OPC培养物,我们证明白细胞介素-10(IL-10),细胞因子在减少神经炎症和促进血肿清除中起作用,显着减少出血诱导的脂质ROS积累和随后在OPCs中的铁凋亡。机械上,IL-10激活IL-10R/STAT3信号通路并上调DLK1/AMPK/ACC轴。随后,IL-10重新编程脂质代谢并减少脂质ROS积累。此外,在自体血液注射脑出血中风(ICH)小鼠模型中,内源性IL-10缺乏,特异性敲除OPCs中的Il10r或Dlk1,或ACC抑制剂的施用与OPC细胞死亡增加有关,脱髓鞘,轴突发芽,以及ICH慢性期的认知缺陷,反之亦然。这些数据表明,IL-10通过减少脂质ROS保护免受OPC损失和白质损伤,支持进一步开发潜在的临床应用,以使中风和相关疾病患者受益。
    Accumulation of reactive oxygen species (ROS), especially on lipids, induces massive cell death in neurons and oligodendrocyte progenitor cells (OPCs) and causes severe neurologic deficits post stroke. While small compounds, such as deferoxamine, lipostatin-1, and ferrostatin-1, have been shown to be effective in reducing lipid ROS, the mechanisms by which endogenously protective molecules act against lipid ROS accumulation and subsequent cell death are still unclear, especially in OPCs, which are critical for maintaining white matter integrity and improving long-term outcomes after stroke. Here, using mouse primary OPC cultures, we demonstrate that interleukin-10 (IL-10), a cytokine playing roles in reducing neuroinflammation and promoting hematoma clearance, significantly reduced hemorrhage-induced lipid ROS accumulation and subsequent ferroptosis in OPCs. Mechanistically, IL-10 activated the IL-10R/STAT3 signaling pathway and upregulated the DLK1/AMPK/ACC axis. Subsequently, IL-10 reprogrammed lipid metabolism and reduced lipid ROS accumulation. In addition, in an autologous blood injection intracerebral hemorrhagic stroke (ICH) mouse model, deficiency of the endogenous Il-10, specific knocking out Il10r or Dlk1 in OPCs, or administration of ACC inhibitor was associated with increased OPC cell death, demyelination, axonal sprouting, and the cognitive deficits during the chronic phase of ICH and vice versa. These data suggest that IL-10 protects against OPC loss and white matter injury by reducing lipid ROS, supporting further development of potential clinical applications to benefit patients with stroke and related disorders.
    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
    少突胶质前体细胞(OPCs)存在于成人中枢神经系统(CNS)中,并且它们分化为髓鞘少突胶质细胞(OL)的能力受损可导致多发性硬化症(MS)患者脱髓鞘,伴有神经功能缺损和认知障碍。外泌体,细胞释放的小囊泡,已知通过携带生物活性分子促进细胞间通讯。在这项研究中,我们利用来自人脐带间充质干细胞(HUMSCs-Exos)的外泌体。我们对外泌体处理的细胞进行了测序和生物信息学分析,以证明HUMSCs-Exos可以刺激OPC中的髓磷脂基因表达。功能研究表明,HUMSCs-Exos激活Pi3k/Akt通路,并通过miR-23a-3p的转移调节Tbr1/Wnt信号分子,促进OLs分化和增强髓鞘相关蛋白的表达。在实验性自身免疫性脑脊髓炎(EAE)模型中,HUMSCs-Exos治疗可显着改善神经功能并促进髓鞘再生。这项研究提供了对使用无细胞外泌体治疗与多发性硬化症相关的中枢神经系统脱髓鞘的细胞和分子见解,证明了其治疗脱髓鞘和神经退行性疾病的巨大潜力。
    Oligodendrocyte precursor cells are present in the adult central nervous system, and their impaired ability to differentiate into myelinating oligodendrocytes can lead to demyelination in patients with multiple sclerosis, accompanied by neurological deficits and cognitive impairment. Exosomes, small vesicles released by cells, are known to facilitate intercellular communication by carrying bioactive molecules. In this study, we utilized exosomes derived from human umbilical cord mesenchymal stem cells (HUMSCs-Exos). We performed sequencing and bioinformatics analysis of exosome-treated cells to demonstrate that HUMSCs-Exos can stimulate myelin gene expression in oigodendrocyte precursor cells. Functional investigations revealed that HUMSCs-Exos activate the Pi3k/Akt pathway and regulate the Tbr1/Wnt signaling molecules through the transfer of miR-23a-3p, promoting oligodendrocytes differentiation and enhancing the expression of myelin-related proteins. In an experimental autoimmune encephalomyelitis model, treatment with HUMSCs-Exos significantly improved neurological function and facilitated remyelination. This study provides cellular and molecular insights into the use of cell-free exosome therapy for central nervous system demyelination associated with multiple sclerosis, demonstrating its great potential for treating demyelinating and neurodegenerative diseases.
    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
    少突胶质祖细胞(OPCs)在髓鞘形成和吞噬作用中起关键作用,与邻近细胞通信并有助于血脑屏障(BBB)的完整性。然而,在阿尔茨海默病(AD)的病理情况下,大脑的微环境经历了有害的变化,显着影响OPC和它们的功能。从OPC功能开始,我们深入研究了OPCs向产生髓鞘的少突胶质细胞的转化,与中枢神经系统(CNS)中其他细胞的复杂信号相互作用,以及令人着迷的吞噬过程,影响OPCs的功能并影响CNS稳态。此外,我们讨论了OPCs在BBB形成中的重要作用,并强调了OPCs在形成CNS保护性屏障中的关键贡献。在AD的背景下,讨论了大脑局部微环境的恶化,主要集中在神经炎症,氧化应激,和有毒蛋白质的积累。有害的变化扰乱了大脑的微妙平衡,影响OPCs的再生能力并损害髓磷脂的完整性。在病理条件下,OPCs在迁移和增殖方面经历了重大变化,导致分化受损和产生成熟少突胶质细胞的能力降低。此外,髓鞘变性和形成在AD中变得越来越活跃,导致进行性神经变性。最后,我们总结了目前针对OPCs的AD治疗方法。振兴OPC衰老的策略,调节信号通路以增强OPC分化,并探索其他潜在的治疗途径有望减轻AD对OPCs和CNS功能的影响。总之,这篇综述强调了OPCs在CNS功能中不可或缺的作用及其在AD发病机制中的作用。OPCs和AD脑微环境之间复杂的相互作用强调了神经退行性疾病的复杂性。在病理条件下研究OPC的见解为针对OPC和促进神经变性的创新治疗策略提供了基础。未来的研究将促进我们对神经退行性疾病的理解和管理,最终为AD和相关疾病患者提供有效治疗和改善生活质量的希望。
    Oligodendrocyte progenitor cells (OPCs) play pivotal roles in myelin formation and phagocytosis, communicating with neighboring cells and contributing to the integrity of the blood-brain barrier (BBB). However, under the pathological circumstances of Alzheimer\'s disease (AD), the brain\'s microenvironment undergoes detrimental changes that significantly impact OPCs and their functions. Starting with OPC functions, we delve into the transformation of OPCs to myelin-producing oligodendrocytes, the intricate signaling interactions with other cells in the central nervous system (CNS), and the fascinating process of phagocytosis, which influences the function of OPCs and affects CNS homeostasis. Moreover, we discuss the essential role of OPCs in BBB formation and highlight the critical contribution of OPCs in forming CNS-protective barriers. In the context of AD, the deterioration of the local microenvironment in the brain is discussed, mainly focusing on neuroinflammation, oxidative stress, and the accumulation of toxic proteins. The detrimental changes disturb the delicate balance in the brain, impacting the regenerative capacity of OPCs and compromising myelin integrity. Under pathological conditions, OPCs experience significant alterations in migration and proliferation, leading to impaired differentiation and a reduced ability to produce mature oligodendrocytes. Moreover, myelin degeneration and formation become increasingly active in AD, contributing to progressive neurodegeneration. Finally, we summarize the current therapeutic approaches targeting OPCs in AD. Strategies to revitalize OPC senescence, modulate signaling pathways to enhance OPC differentiation, and explore other potential therapeutic avenues are promising in alleviating the impact of AD on OPCs and CNS function. In conclusion, this review highlights the indispensable role of OPCs in CNS function and their involvement in the pathogenesis of AD. The intricate interplay between OPCs and the AD brain microenvironment underscores the complexity of neurodegenerative diseases. Insights from studying OPCs under pathological conditions provide a foundation for innovative therapeutic strategies targeting OPCs and fostering neurodegeneration. Future research will advance our understanding and management of neurodegenerative diseases, ultimately offering hope for effective treatments and improved quality of life for those affected by AD and related disorders.
    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
    脑白质损伤(WMI)是早产儿最常见的脑损伤,导致运动和发育缺陷,通常伴有认知障碍。然而,没有有效的治疗方法。治疗早产WMI的一种有希望的方法是细胞替代疗法。其中丢失的细胞可以被外源少突胶质细胞祖细胞(OPCs)替代。
    这项研究开发了一种将人类神经干细胞(hNSC)分化为人类OPCs(hOPCs)的方法。出生后第3天建立早产WMI动物模型,出生后第10天富集OLIG2/NG2/PDGFRα/O4hOPCs并移植到call体中。然后,组织学分析和电子显微镜用于检测病变结构;进行行为测定以检测认知功能。
    移植的hOPCs在整个主要白质区域存活并迁移。观察到移植的hOPCs的形态分化。组织学分析显示病变区域的结构修复。电子显微镜证实了call体轴突的髓鞘再生。Morris水迷宫测试显示认知功能恢复。
    我们的研究表明,外源性hOPCs可以在WMI大鼠脑内分化为CC1+OLS,提高他们的认知功能。
    UNASSIGNED: Cerebral white matter injury (WMI) is the most common brain injury in preterm infants, leading to motor and developmental deficits often accompanied by cognitive impairment. However, there is no effective treatment. One promising approach for treating preterm WMI is cell replacement therapy, in which lost cells can be replaced by exogenous oligodendrocyte progenitor cells (OPCs).
    UNASSIGNED: This study developed a method to differentiate human neural stem cells (hNSCs) into human OPCs (hOPCs). The preterm WMI animal model was established in rats on postnatal day 3, and OLIG2+/NG2+/PDGFRα+/O4+ hOPCs were enriched and transplanted into the corpus callosum on postnatal day 10. Then, histological analysis and electron microscopy were used to detect lesion structure; behavioral assays were performed to detect cognitive function.
    UNASSIGNED: Transplanted hOPCs survived and migrated throughout the major white matter tracts. Morphological differentiation of transplanted hOPCs was observed. Histological analysis revealed structural repair of lesioned areas. Re-myelination of the axons in the corpus callosum was confirmed by electron microscopy. The Morris water maze test revealed cognitive function recovery.
    UNASSIGNED: Our study showed that exogenous hOPCs could differentiate into CC1+ OLS in the brain of WMI rats, improving their cognitive functions.
    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
    簇集蛋白(CLU)基因中rs11136000变体的C等位基因代表了晚发性阿尔茨海默病的第三强已知遗传风险因素。然而,这种单核苷酸多态性(SNP)是否具有功能性,其潜在机制尚不清楚.在这项研究中,CLUrs11136000SNP通过小规模CRISPR-Cas9筛选被鉴定为功能性变体。源自携带CLUrs11136000SNP的“C”或“T”等位基因的等基因诱导多能干细胞(iPSC)的星形胶质细胞表现出不同的CLU表达水平。TARDNA结合蛋白-43(TDP-43)优先与“C”等位基因结合,以促进CLU表达并加剧炎症。干扰素应答和CXCL10表达在细胞因子处理的C/C星形胶质细胞中升高,导致少突胶质细胞祖细胞(OPC)增殖和髓鞘形成的抑制。因此,在C/C携带者的人脑中检测到升高的CLU和CXCL10但降低的髓磷脂碱性蛋白(MBP)表达。我们的研究揭示了在CLUrs11136000风险“C”等位基因携带者中观察到的白质完整性降低的潜在机制。
    The C allele of rs11136000 variant in the clusterin (CLU) gene represents the third strongest known genetic risk factor for late-onset Alzheimer\'s disease. However, whether this single-nucleotide polymorphism (SNP) is functional and what the underlying mechanisms are remain unclear. In this study, the CLU rs11136000 SNP is identified as a functional variant by a small-scale CRISPR-Cas9 screen. Astrocytes derived from isogenic induced pluripotent stem cells (iPSCs) carrying the \"C\" or \"T\" allele of the CLU rs11136000 SNP exhibit different CLU expression levels. TAR DNA-binding protein-43 (TDP-43) preferentially binds to the \"C\" allele to promote CLU expression and exacerbate inflammation. The interferon response and CXCL10 expression are elevated in cytokine-treated C/C astrocytes, leading to inhibition of oligodendrocyte progenitor cell (OPC) proliferation and myelination. Accordingly, elevated CLU and CXCL10 but reduced myelin basic protein (MBP) expression are detected in human brains of C/C carriers. Our study uncovers a mechanism underlying reduced white matter integrity observed in the CLU rs11136000 risk \"C\" allele carriers.
    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
    脊髓损伤(SCI)是一种破坏性的神经系统疾病,导致行动不便,感官,和自主神经功能障碍。少突胶质祖细胞(OPCs)的损失,可以分化为成熟的少突胶质细胞和再髓鞘损伤的轴突,与SCI患者恢复较差有关。然而,抑制OPCs损失一直是一个难以克服的问题。在这项研究中,我们证明了槲皮素的抗铁凋亡作用是Erastin诱导OPC铁凋亡的机制。槲皮素改善OPCs中爱司汀诱导的铁凋亡,如铁浓度降低所示,活性氧(ROS)的产生,谷胱甘肽(GSH)含量增加,线粒体形态更正常。与擦除素诱导的OPC相比,在槲皮素处理的OPCs中,髓鞘碱性蛋白(MBP)阳性髓鞘和NF200阳性轴突显著增加.此外,槲皮素通过下调转铁蛋白改善了erastin诱导的铁凋亡以及OPCs的髓鞘和轴突损失。具有转铁蛋白过表达质粒的转染OPCs显着废除了槲皮素在OPC铁凋亡中的保护作用。使用ChIP-qPCR,发现转铁蛋白与其上游基因Id2的直接相互作用。Id2的过表达逆转槲皮素对OPC铁凋亡的影响。体内研究发现槲皮素大大减少了损伤面积,提高SCI后BBB评分。此外,在SCI模型中,槲皮素显著下调Id2和转铁蛋白表达,同时显著上调GPX4和PTGS2表达。总之,槲皮素通过抑制Id2/转铁蛋白途径防止OPCs的铁凋亡。这些发现强调了槲皮素作为一种抗铁凋亡剂用于治疗或预防脊髓损伤。
    Spinal cord injury (SCI) is a destructive neurological disorder that causes impaired mobility, sensory, and autonomic dysfunctions. The loss of oligodendrocyte progenitor cells (OPCs), which can differentiate into mature oligodendrocytes and re-myelinate damaged axons, is related to poorer recovery for SCI patients. However, inhibiting OPCs loss has always been a difficult problem to overcome. In this study, we demonstrated the anti-ferroptosis effects of quercetin as a mechanism in erastin-induced OPC ferroptosis. Quercetin ameliorated erastin-induced ferroptosis in OPCs, as indicated by decreased iron concentration, reactive oxygen species (ROS) production, and increased content of glutathione (GSH) as well as more normal mitochondria morphology. Compared with erastin-induced OPCs, the myelin basic protein (MBP)-positive myelin and NF200-positive axonal was remarkably increased in quercetin-treated OPCs. Furthermore, quercetin ameliorated the erastin-induced ferroptosis as well as the myelin and axon loss of OPCs by downregulating transferrin. Transfected OPCs with transferrin overexpression plasmids significantly abrogated the protective role of quercetin in OPC ferroptosis. Using ChIP-qPCR, a direct interaction of transferrin with its upstream gene Id2 was found. The overexpression of Id2 reversed the effect of quercetin on OPC ferroptosis. In vivo study found that quercetin greatly decreased the area of injury, and enhanced the BBB score after SCI. Furthermore, in the SCI model, quercetin significantly downregulated Id2 and transferrin expression, while significantly up-regulated GPX4 and PTGS2 expression. In conclusion, quercetin prevents the ferroptosis of OPCs by inhibiting the Id2/transferrin pathway. These findings highlight quercetin as an anti-ferroptosis agent for the treatment or prevention of spinal cord 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
    已报道激活素A(ActA)在体外促进少突胶质祖细胞(OPC)分化并改善成年小鼠的神经学结果。然而,A行为在早产脑损伤中的作用和作用机制尚不清楚。在本研究中,对P5大鼠进行缺氧缺血以建立新生儿白质损伤(WMI)模型,并通过侧脑室注射ActA。病理特征,OPC区分,髓鞘形成,并对神经系统表现进行了分析。Further,本研究探讨了Noggin/BMP4/Id2信号通路参与ActA在WMI中的作用。A行动减轻了病理损伤,促进OPC差异化,增强髓鞘和有髓鞘的轴突形成,改善WMI大鼠的神经功能。此外,行动增强了noggin表达,which,反过来,抑制骨形态发生蛋白4(BMP4)和DNA结合抑制剂2(Id2)的表达。此外,Id2的上调完全消除了A法案在WMI大鼠中的挽救作用。总之,本研究结果表明,ActA通过靶向一种新的Noggin/BMP4/Id2信号通路来挽救早产脑损伤.
    Activin A (Act A) has been reported to promote oligodendrocyte progenitor cell (OPC) differentiation in vitro and improve neurological outcomes in adult mice. However, the roles and mechanisms of action of Act A in preterm brain injury are unknown. In the present study, P5 rats were subjected to hypoxia‑ischemia to establish a neonatal white matter injury (WMI) model and Act A was injected via the lateral ventricle. Pathological characteristics, OPC differentiation, myelination, and neurological performance were analyzed. Further, the involvement of the Noggin/BMP4/Id2 signaling pathway in the roles of Act A in WMI was explored. Act A attenuated pathological damage, promoted OPC differentiation, enhanced myelin sheath and myelinated axon formation, and improved neurological performance of WMI rats. Moreover, Act A enhanced noggin expression, which, in turn, inhibited the expression of bone morphogenetic protein 4 (BMP4) and inhibitor of DNA binding 2 (Id2). Furthermore, upregulation of Id2 completely abolished the rescue effects of Act A in WMI rats. In conclusion, the present findings suggested that Act A rescues preterm brain injury via targeting a novel Noggin/BMP4/Id2 signaling pathway.
    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