Ginkgolide B

银杏内酯 B
  • 文章类型: Journal Article
    神经性疼痛是由神经系统内疼痛信号的异常产生引起的病理状态。银杏内酯B(GB),发现了银杏的一种活性成分.银杏叶,具有神经保护特性。本研究旨在探讨GB对神经病理性疼痛的影响及其机制。在体内研究中,采用大鼠慢性缩窄性损伤模型,结果表明,GB(4mg/kg)治疗可有效减轻大鼠的疼痛感觉,降低小胶质细胞标记物Iba-1的表达,NLRP3炎性体,和炎症因子,如白细胞介素(IL)-1β,在脊髓手术后7天。在体外研究中,我们使用脂多糖(500ng/mL)/三磷酸腺苷(5mM)诱导小胶质细胞炎症,并用GB(10,20和40μM)治疗.GB上调线粒体自噬蛋白的表达,例如PINK1,Parkin,LC3II/I,Tom20和Beclin1,并减少了细胞产生的活性氧。此外,它降低了炎症相关蛋白的表达,如Caspase-1,IL-1β,和NLRP3在小胶质细胞中。然而,ParkinshRNA/siRNA或自噬抑制剂3-甲基腺嘌呤(5mM)逆转了这一效应.这些发现表明,GB通过激活PINK1-Parkin介导的线粒体自噬来减轻神经炎症,从而减轻神经性疼痛。
    Neuropathic pain is a pathological state induced by the aberrant generation of pain signals within the nervous system. Ginkgolide B(GB), an active component found of Ginkgo. biloba leaves, has neuroprotective properties. This study aimed to explore the effects of GB on neuropathic pain and its underlying mechanisms. In the in vivo study, we adopted the rat chronic constriction injury model, and the results showed that GB(4 mg/kg) treatment effectively reduced pain sensation in rats and decreased the expressions of Iba-1 (a microglia marker), NLRP3 inflammasome, and inflammatory factors, such as interleukin (IL)-1β, in the spinal cord 7 days post-surgery. In the in vitro study, we induced microglial inflammation using lipopolysaccharide (500 ng/mL) / adenosine triphosphate (5 mM) and treated it with GB (10, 20, and 40 μM). GB upregulated the expression of mitophagy proteins, such as PINK1, Parkin, LC3 II/I, Tom20, and Beclin1, and decreased the cellular production of reactive oxygen species. Moreover, it lowered the expression of inflammation-related proteins, such as Caspase-1, IL-1β, and NLRP3 in microglia. However, this effect was reversed by Parkin shRNA/siRNA or the autophagy inhibitor 3-methyladenine (5 mM). These findings reveal that GB alleviates neuropathic pain by mitigating neuroinflammation through the activation of PINK1-Parkin-mediated mitophagy.
    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
    用于治疗病毒感染后综合征的纳米医学正处于新兴阶段。尽管对常规抗氧化剂的临床前研究取得了有希望的结果,将其临床转化为治疗COVID后疾病的疗法仍然具有挑战性。这些限制是由于它们的生物利用度低,不稳定性,有限的运输到目标组织,半衰期短,需要频繁和高剂量。在冠状病毒(SARS-CoV-2)感染期间激活免疫系统可导致活性氧(ROS)的产生增加,抗氧化剂储备耗尽,最后,氧化应激和神经炎症。为了解决这个问题,我们开发了一种基于脂质(囊泡型和立方体型)纳米颗粒(LNPs)共封装银杏内酯B和槲皮素的抗氧化纳米疗法。通过冻干混合薄脂质膜的水合,通过自组装方法制备负载抗氧化剂的纳米载体。我们在一个新的体外模型中评估了LNP,用于研究冠状病毒感染中氧化应激引起的神经元功能障碍。我们研究了响应于引起氧化应激介导的神经毒性的过硫酸钾(KPS)而触发的关键下游信号通路。用KPS(50mM)处理神经元来源的细胞(SH-SY5Y)30分钟可显着增加线粒体功能障碍,同时消耗谷胱甘肽过氧化物酶(GSH-Px)和酪氨酸羟化酶(TH)的水平。这导致凋亡和坏死细胞死亡过程的顺序激活,这证实了两种蛋白(GSH-Px和TH)在长COVID综合征中的重要意义。纳米药物介导的银杏内酯B负载的立方体和囊泡LNP治疗显示最小的细胞毒性和完全减弱KPS诱导的细胞死亡过程,细胞凋亡从32.6%(KPS)降至19.0%(MO-GB),12.8%(MO-GB-Quer),14.8%(DMPC-PEG-GB),和23.6%(DMPC-PEG-GB-Quer)通过自由基清除和补充GSH-Px水平。这些发现表明,基于GB-LNP的纳米药物可以通过调节细胞内氧化还原稳态来抵抗KPS诱导的细胞凋亡。
    Nanomedicine for treating post-viral infectious disease syndrome is at an emerging stage. Despite promising results from preclinical studies on conventional antioxidants, their clinical translation as a therapy for treating post-COVID conditions remains challenging. The limitations are due to their low bioavailability, instability, limited transport to the target tissues, and short half-life, requiring frequent and high doses. Activating the immune system during coronavirus (SARS-CoV-2) infection can lead to increased production of reactive oxygen species (ROS), depleted antioxidant reserve, and finally, oxidative stress and neuroinflammation. To tackle this problem, we developed an antioxidant nanotherapy based on lipid (vesicular and cubosomal types) nanoparticles (LNPs) co-encapsulating ginkgolide B and quercetin. The antioxidant-loaded nanocarriers were prepared by a self-assembly method via hydration of a lyophilized mixed thin lipid film. We evaluated the LNPs in a new in vitro model for studying neuronal dysfunction caused by oxidative stress in coronavirus infection. We examined the key downstream signaling pathways that are triggered in response to potassium persulfate (KPS) causing oxidative stress-mediated neurotoxicity. Treatment of neuronally-derived cells (SH-SY5Y) with KPS (50 mM) for 30 min markedly increased mitochondrial dysfunction while depleting the levels of both glutathione peroxidase (GSH-Px) and tyrosine hydroxylase (TH). This led to the sequential activation of apoptotic and necrotic cell death processes, which corroborates with the crucial implication of the two proteins (GSH-Px and TH) in the long-COVID syndrome. Nanomedicine-mediated treatment with ginkgolide B-loaded cubosomes and vesicular LNPs showed minimal cytotoxicity and completely attenuated the KPS-induced cell death process, decreasing apoptosis from 32.6% (KPS) to 19.0% (MO-GB), 12.8% (MO-GB-Quer), 14.8% (DMPC-PEG-GB), and 23.6% (DMPC-PEG-GB-Quer) via free radical scavenging and replenished GSH-Px levels. These findings indicated that GB-LNPs-based nanomedicines may protect against KPS-induced apoptosis by regulating intracellular redox homeostasis.
    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
    The review presents an analysis of experimental data on the study of neurobiological effects of ginkgolide B, which may find application in the therapy of Alzheimer\'s disease (AD). Ginkgolide B is a diterpene trilactone isolated from the leaves of the relict woody plant Ginkgo biloba L., which has been used for thousands of years in traditional Chinese medicine as a neuroprotective agent. In recent years, this compound has attracted attention because of its wide range of neurobiological effects. The neuroprotective effect of ginkgolide B on brain neurons when exposed to various neurotoxins has been established. This compound has also been shown to effectively protect neurons from the effects of beta-amyloid. Studies have revealed the ability of ginkgolide B to reduce microglia activity and regulate neurotransmitter release. In vivo experiments have shown that this substance significantly increases the expression of brain-derived neurotrophic factor (BDNF) and improves cognitive functions, including memory and learning. It is concluded that ginkgolide B, apparently, may find application in the future as a multi-targeted agent of complex therapy of AD.
    В обзоре представлен анализ экспериментальных данных по исследованию нейробиологических эффектов гинкголида B, которые могут найти применение в терапии болезни Альцгеймера (БА). Гинкголид B — это дитерпеновый трилактон, выделенный из листьев реликтового древесного растения гинкго двулопастного (Ginkgo biloba L.), которое в течение тысячелетий используется в традиционной китайской медицине. В последние годы это соединение привлекает внимание из-за широкого спектра нейробиологических эффектов. Установлено нейропротективное действие гинкголида B на нейроны головного мозга при воздействии различных нейротоксинов. Показано, что данное соединение также эффективно защищает нейроны от воздействия бета-амилоида. Исследования выявили способность гинкголида B снижать активность микроглии и регулировать выделение нейромедиаторов. В экспериментах in vivo показано, что данное вещество значительно повышает экспрессию нейротрофического фактора мозга (BDNF), а также улучшает когнитивные функции, включая память и способность к обучению. Делается вывод, что гинкголид B, по-видимому, может найти применение в будущем в качестве многоцелевого средства комплексной терапии БА.
    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
    淋巴系统通过促进代谢废物的去除在维持最佳中枢神经系统(CNS)功能中起着至关重要的作用。水通道蛋白4(AQP4)蛋白,主要位于星形胶质细胞末端,是代谢废物排泄的关键途径。β-Dystroglycan(β-DG)可以将AQP4蛋白锚定在星形胶质细胞的足底膜上,并被基质金属蛋白酶(MMP)-9蛋白裂解。研究表明,高血糖会上调神经系统中MMP-9的表达,导致神经性疼痛。银杏内酯B(GB)对MMP-9蛋白有抑制作用。在这项研究中,我们研究了在痛性糖尿病神经病变(PDN)患者中,GB对MMP-9介导的β-DG裂解的抑制是否参与了淋巴系统AQP4极性的调节,并发挥了神经保护作用.通过注射链脲佐菌素(STZ)建立PDN模型。使用磁共振成像(MRI)观察了淋巴系统的功能变化。测量缩爪阈值(PWT)以评估机械性异常性疼痛。MMP-9、β-DG蛋白的表达,免疫印迹法和免疫荧光法检测AQP4。我们的发现表明,大鼠脊髓淋巴系统内造影剂清除效率显着降低,伴随着PWT下降,MMP-9蛋白表达增加,β-DG蛋白表达降低,AQP4极性丧失。值得注意的是,GB治疗证明了通过抑制MMP-9调节AQP4极性来改善脊髓淋巴功能的能力,为PDN提供了一条有前途的治疗途径。
    The glymphatic system plays a crucial role in maintaining optimal central nervous system (CNS) function by facilitating the removal of metabolic wastes. Aquaporin-4 (AQP4) protein, predominantly located on astrocyte end-feet, is a key pathway for metabolic waste excretion. β-Dystroglycan (β-DG) can anchor AQP4 protein to the end-feet membrane of astrocytes and can be cleaved by matrix metalloproteinase (MMP)-9 protein. Studies have demonstrated that hyperglycemia upregulates MMP-9 expression in the nervous system, leading to neuropathic pain. Ginkgolide B (GB) exerts an inhibitory effect on the MMP-9 protein. In this study, we investigated whether inhibition of MMP-9-mediated β-DG cleavage by GB is involved in the regulation of AQP4 polarity within the glymphatic system in painful diabetic neuropathy (PDN) and exerts neuroprotective effects. The PDN model was established by injecting streptozotocin (STZ). Functional changes in the glymphatic system were observed using magnetic resonance imaging (MRI). The paw withdrawal threshold (PWT) was measured to assess mechanical allodynia. The protein expressions of MMP-9, β-DG, and AQP4 were detected by Western blotting and immunofluorescence. Our findings revealed significant decreases in the efficiency of contrast agent clearance within the spinal glymphatic system of the rats, accompanied by decreased PWT, increased MMP-9 protein expression, decreased β-DG protein expression, and loss of AQP4 polarity. Notably, GB treatment demonstrated the capacity to ameliorate spinal cord glymphatic function by modulating AQP4 polarity through MMP-9 inhibition, offering a promising therapeutic avenue for PDN.
    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
    本研究探讨了银杏内酯B(GB)的有效性,一种来自银杏叶的化合物,在对抗青光眼引起的细胞死亡方面,重点研究线粒体损伤和线粒体通透性转换孔(mPTP)。利用高眼压模型和体外青光眼模拟,这项研究调查了在氧糖剥夺/再灌注(OGD/R)和大鼠青光眼模型中GB对视网膜祖细胞(RPCs)的影响。研究方法包括细胞凋亡评估,通过Westernblot进行凋亡标记分析,线粒体结构和功能评估。研究结果表明,GB显著降低体外暴露于OGD/R的RPCs的细胞凋亡,并减少体内缺血再灌注损伤。GB的保护作用归因于其保持线粒体完整性的能力,保持膜电位,调节钙水平,并抑制mPTP打开。这些结果强调了GB作为急性原发性闭角型青光眼治疗剂的潜力,强调其减轻RPCs和视网膜神经纤维层细胞线粒体损伤和凋亡的能力。
    This research delves into the effectiveness of Ginkgolide B (GB), a compound from Ginkgo biloba, in combating cell death caused by glaucoma, with a focus on mitochondrial impairment and the mitochondrial permeability transition pore (mPTP). Utilizing models of high intraocular pressure and in vitro glaucoma simulations, the study investigates GB\'s impact on retinal progenitor cells (RPCs) under oxygen-glucose deprivation/reperfusion (OGD/R) and in a rat glaucoma model. The study methodologies included apoptosis assessment, apoptotic marker analysis via Western blot, and mitochondrial structure and function evaluation. The findings reveal that GB notably decreases apoptosis in RPCs exposed to OGD/R in vitro, and reduces ischemia-reperfusion damage in vivo. GB\'s protective role is attributed to its ability to preserve mitochondrial integrity, maintain membrane potential, regulate calcium levels, and inhibit mPTP opening. These results underscore GB\'s potential as a therapeutic agent for acute primary angle-closure glaucoma, highlighting its capability to alleviate mitochondrial damage and apoptosis in RPCs and retinal nerve fiber layer cells.
    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
    由于核技术在医学中的广泛应用,大脑暴露于电离辐射的风险逐渐增加,工业,和航空航天领域。放射性脑损伤(RBI)极有可能引起广泛的神经系统并发症,包括精神分裂症,阿尔茨海默病(AD),抑郁症。银杏内酯B(GB)是从银杏叶中提取的有效活性成分之一。对中枢神经系统发挥保护作用,参与了Hippo信号通路的调控。MST1作为Hippo途径的核心激酶之一,参与调节细胞增殖,分化,和凋亡。然而,尚不清楚GB是否减轻放射性脑损伤(RBI),以及GB的辐射防护作用是否涉及MST1信号传导.因此,本研究旨在探讨GB的辐射防护作用和潜在机制。
    用X射线(20Gy)刺激C57BL/6小鼠以建立RBI模型。然后,采用Morris水迷宫试验(MWM)和降压被动回避试验(SDPAT)评价小鼠的学习记忆功能。露天试验(OFT),尾部悬挂试验(TST),和强迫游泳测试(FST)用于评估运动活动和绝望的变化。此外,X射线刺激的SH-SY5Y细胞用于验证GB的辐射防护作用。免疫荧光双重染色,二氢乙锭(DHE),westernblot,和流式细胞术用于探讨DCC/MST1信号在RBI中的作用。
    在这项研究中,X射线治疗的小鼠表现出认知障碍和抑郁样行为,通过GB治疗得到改善。GB还减少了ROS的产生和海马中TUNEL阳性细胞的数量。此外,GB增加了p-AKT和Bcl2的蛋白质水平,而降低了MST1,p-p38,p-JNK的蛋白质水平,体内和体外裂解的caspase-3和Bax。此外,外源性Netrin-1减轻X射线诱导的ROS产生和凋亡,而Netrin-1受体DCC的敲除消除了GB的保护作用。
    氧化应激和MST1介导的神经元凋亡参与了辐射诱导的认知障碍和抑郁样行为,GB对DCC的调控是对RBI的有效干预。
    UNASSIGNED: The risk of brain exposure to ionizing radiation increases gradually due to the extensive application of nuclear technology in medical, industrial, and aerospace fields. Radiation-induced brain injury (RBI) is highly likely to cause a wide range of neurological complications, including schizophrenia, Alzheimer\'s disease (AD), depression. Ginkgolide B (GB) is one of the effective active components extracted from ginkgo biloba leaves, exerts protective effects on CNS, which is involved in the regulation of the Hippo signaling pathway. MST1, as one of the core kinases of the Hippo pathway, participated in regulating cell proliferation, differentiation, and apoptosis. However, it remains unclear whether GB attenuates radiation brain injury (RBI) and whether the radioprotective effect of GB refers to MST1 signaling. Hence, our study aimed to explore the radiation protection effect and the potential mechanism of GB.
    UNASSIGNED: C57BL/6 mice were stimulated with an X-ray (20 Gy) to establish an RBI model. Then, morris water maze test (MWM) and step-down passive avoidance test (SDPAT) were used to assess the learning and memory function of mice. The open field test (OFT), tail suspension test (TST), and forced swimming test (FST) were used to assess changes in locomotor activity and hopelessness. Besides, X-ray-stimulated SH-SY5Y cells were used to verify the radioprotective effect of GB. Immunofluorescence double staining, Dihydroethidium (DHE), western blot, and flow cytometry were used to explore the role of DCC/MST1 signaling in RBI.
    UNASSIGNED: In this study, X-ray-treated mice exhibited cognitive impairment and depression-like behavior, which was ameliorated by GB treatment. GB also reduced the ROS production and the number of TUNEL-positive cells in the hippocampus. Moreover, GB increased the protein levels of p-AKT and Bcl2, while decreased the protein levels of MST1, p-p38, p-JNK, cleaved-caspase-3 and Bax both in vivo and in vitro. Additionally, exogenous Netrin-1 alleviated X-ray-induced ROS production and apoptosis, whereas knockout of Netrin-1 receptor DCC abolished the protective effect of GB.
    UNASSIGNED: Oxidative stress and MST1-mediated neuronal apoptosis participated in radiation-induced cognitive impairment and depression-like behaviors, and modulation of DCC by GB was an effective intervention against RBI.
    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
    银杏内酯B(GB),已被证明是最有效的天然存在的血小板活化因子(PAF)拮抗剂,被广泛用于心血管和脑血管疾病的管理。然而,其有限的口服生物利用度因其在胃酸中的低溶解度和在肠液中的稳定性不足而受到阻碍,从而制约了其实际应用。本研究旨在开发GB纳米晶体(GB-NC)和GB纳米晶体自稳定的皮克林纳米乳液(GB-NSSPNE)使用小型化的湿珠研磨方法。在体内和体外进行比较评价以评估其有效性。调查结果显示,GB-NSSPNE,其完整的纳米粒子缓慢释放和吸收,与GB-NC的快速释放和吸收相比,在提高GB的口服生物利用度方面更有效。这一发现表明了口服GB的潜在新策略。
    Ginkgolide B (GB), which has been demonstrated as the most efficacious naturally occurring platelet-activating factor (PAF) antagonist, is extensively utilized for the management of cardiovascular and cerebrovascular ailments. Nevertheless, its limited oral bioavailability is hindered by its low solubility in gastric acid and inadequate stability in intestinal fluid, thereby constraining its practical application. This study aimed to develop GB nanocrystals (GB-NCs) and GB nanocrystals self-stabilized Pickering nano-emulsion (GB-NSSPNE) using a miniaturized wet bead milling method. Comparative evaluations were conducted in vivo and in vitro to assess their effectiveness. The findings revealed that GB-NSSPNE, with its intact nanoparticle slow release and absorption, was more effective in enhancing the oral bioavailability of GB compared to the rapid release and absorption of GB-NCs. This finding suggests a potential novel strategy for the oral delivery of GB.
    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:围产期缺氧缺血性脑病(HIE)是一种可导致长期认知,电机,和新生儿的行为障碍。尽管脑低温疗法目前是HIE的标准治疗方法,它不能提供完整的神经保护。因此,有必要探索额外的治疗方法以提高治疗效果.本研究旨在探讨银杏内酯B(GB)在促进HIE后神经可塑性和促进自发恢复中的潜在作用。
    方法:在本研究中,我们采用新生大鼠HIE模型研究GB对自然恢复的影响。在缺氧后24小时开始GB治疗并连续给药14天。我们评估了治疗期后的几个结果指标,包括自发的行为恢复和大脑修复。此外,我们量化了HIE发生后血浆和缺血周围区的netrin-1水平.
    结果:我们发现GB治疗显著促进了HIE幼崽的自发行为恢复。此外,认知功能恢复,脑组织修复有明显的加速.我们观察到脑室下细胞增殖增加,地层,和亚粒区。特别感兴趣的是,我们观察到GB治疗后血浆和缺血半影区的netrin-1水平升高.
    结论:我们的研究结果表明,GB促进受HIE影响的新生儿的神经可塑性和自发恢复。观察到的netrin-1的上调可能对介导这些作用至关重要。这些结果凸显了GB作为HIE后治疗的潜力,特别是在促进自发恢复和改善长期结果方面。
    OBJECTIVE: Perinatal hypoxic-ischemic encephalopathy (HIE) is a condition that can lead to long-term cognitive, motor, and behavioral impairments in newborns. Although brain hypothermia therapy is currently the standard treatment for HIE, it does not provide complete neuroprotection. As a result, there is a need to explore additional therapies to enhance treatment outcomes. This study aims to investigate the potential role of Ginkgolide B (GB) in promoting neuroplasticity and facilitating spontaneous recovery after HIE.
    METHODS: In this study, we employed a neonatal rat model of HIE to investigate the effects of GB on spontaneous recovery. GB treatment was initiated 24 h after hypoxia and administered continuously for a duration of 14 days. We evaluated several outcome measures after the treatment period, including spontaneous behavioral recovery and brain repair. Additionally, we quantified the levels of netrin-1 in both plasma and the peri-ischemic zone after the occurrence of HIE.
    RESULTS: We found that GB treatment significantly facilitated spontaneous behavioral recovery in the HIE pups. Furthermore, cognitive function was restored, and brain tissue repair had a noticeable acceleration. We observed increased cell proliferation in the subventricular, stratum, and subgranular zones. Of particular interest, we observed elevated levels of netrin-1 in both plasma and the ischemic penumbra following GB treatment.
    CONCLUSIONS: Our findings suggest that GB promotes neuroplasticity and enhances spontaneous recovery in newborns affected by HIE. The observed upregulation of netrin-1 may be crucial in mediating these effects. These results highlight the promising potential of GB as a post-HIE therapy, particularly in enhancing spontaneous recovery and improving long-term outcomes.
    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
    背景:脑缺血/再灌注(I/R)损伤是神经元损伤和死亡的主要原因。银杏内酯B(GB)已被证明在各种脑损伤模型中表现出神经保护作用。
    目的:研究的目的是探讨GB在保护脑I/R损伤中的潜在作用并探讨其潜在机制。
    方法:成年雄性Sprague-Dawley大鼠暴露于短暂的大脑中动脉闭塞(tMCAO),然后再灌注以引发脑I/R损伤。用不同剂量的GB处理大鼠,载体对照或阳性药物。神经功能,梗死体积,和铁凋亡标志物的水平进行了评估。用OGD/R诱导的PC12细胞进行体外实验,以进一步研究GB对铁凋亡的影响及其机制。此外,分子对接,进行了微尺度热泳(MST)测定,以探索GB和NCOA4的组合。
    结果:梗死体积减少和神经功能增强是GB治疗对脑I/R损伤的剂量依赖性保护的迹象。此外,GB处理对氧化应激和铁凋亡标志物的水平有影响,包括活性氧(ROS),丙二醛(MDA),超氧化物歧化酶(SOD),IR损伤时脑环境中的Fe2+。此外,相关铁中毒的关键因子如ACSL4、GPX4、FTH1和NCOA4可受GB调控。在OGD/R诱导的PC12细胞中,GB通过抑制自噬和破坏NCOA4-FTH1的相互作用来防止铁凋亡。
    结论:我们的研究结果表明,GB可能通过破坏NCOA4-FTH1相互作用抑制铁凋亡来对抗脑I/R损伤。GB对于脑I/R损伤具有潜在的治疗应用,并进一步研究潜在的机制和临床试验是必要的。
    BACKGROUND: Cerebral ischemia/reperfusion (I/R) injury is a major cause of neuronal damage and death. Ginkgolide B (GB) has been shown to exhibit neuroprotective effects in various brain injury models.
    OBJECTIVE: The aim of study was to investigate the potential role of GB in protecting against cerebral I/R injury and explore the underlying mechanisms.
    METHODS: Adult male Sprague-Dawley rats were exposed to transient middle cerebral artery occlusion (tMCAO) followed by reperfusion in order to trigger cerebral I/R injury. The rats were treated with different doses of GB, vehicle control or positive drug. Neurological function, infarct volume, and levels of ferroptosis markers were evaluated. In vitro experiments were performed using OGD/R-induced PC12 cells to further investigate the effects of GB on ferroptosis and its mechanisms. In addition, molecular docking, and microscale thermophoresis (MST) assay were conducted to explore the combination of GB and NCOA4.
    RESULTS: Reduced infarct volume and enhanced neurological function were signs of dose-dependent protection from cerebral I/R injury by GB therapy. Additionally, GB treatment had an impact on the levels of oxidative stress and ferroptosis markers, including reactive oxygen species (ROS), malondialdehyde (MDA), superoxide dismutase (SOD), and Fe2+ in the cerebral environment during IR injury. Moreover, relevant ferroptosis key factors such as ACSL4, GPX4, FTH1, and NCOA4 can be regulated by GB. In OGD/R-induced PC12 cells, GB protected against ferroptosis by inhibiting autophagy and disrupting the interaction of NCOA4-FTH1.
    CONCLUSIONS: Our findings suggest that GB may protect against cerebral I/R injury by inhibiting ferroptosis through disrupting NCOA4-FTH1 interaction. GB has potential therapeutic applications for cerebral I/R injury, and further investigation of the underlying mechanisms and clinical trials are warranted.
    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
    背景:随着年龄的增长,组织-组织串扰的逐渐恶化会导致组织稳态和功能的显着损害,特别是在肌肉骨骼系统中。据报道,通过异慢性共生和运动等干预措施使全身和局部环境恢复活力,可以改善老年生物的肌肉骨骼稳态。我们已经证明银杏内酯B(GB),一个来自银杏的小分子,通过恢复局部和全身通讯改善老年小鼠的骨骼稳态,暗示着维持骨骼肌稳态和增强再生的潜力。在这项研究中,我们研究了GB对老年小鼠骨骼肌再生的治疗效果。
    方法:通过将氯化钡诱导到20月龄小鼠(老年小鼠)的后肢和C2C12来源的肌管中,建立肌肉损伤模型。通过组织化学染色评估每日给药GB(12mg/kg体重)和骨钙蛋白(50μg/kg体重)对肌肉再生的治疗效果,基因表达,流式细胞术,离体肌肉功能试验和旋转试验。RNA测序用于探索GB对肌肉再生的作用机制。随后的体外和体内实验验证了这些发现。
    结果:GB在老年小鼠中的给药改善了肌肉再生(肌肉质量,P=0.0374;肌纤维数/场,P=0.0001;中心核,胚胎肌球蛋白重链阳性肌纤维区,P=0.0144),促进肌肉收缩特性的恢复(强直力,P=0.0002;抽搐力,P=0.0005)和运动性能(旋转性能,P=0.002),和减少肌肉纤维化(胶原蛋白沉积,P<0.0001)和炎症(巨噬细胞浸润,P=0.03)。GB逆转了衰老相关的骨钙蛋白表达下降(P<0.0001),一种成骨细胞特异性激素,促进肌肉再生。外源性骨钙蛋白补充足以改善肌肉再生(肌肉质量,P=0.0029;肌纤维数/场,P<0.0001),功能恢复(强直力,P=0.0059;抽搐力,P=0.07;转杆性能,P<0.0001)和纤维化(胶原沉积,P=0.0316)在老年小鼠中,不会增加异位骨化的风险。
    结论:GB治疗恢复了骨-肌肉内分泌轴,以逆转与衰老相关的肌肉再生下降,因此代表了一种管理肌肉损伤的创新和可行的方法。我们的结果揭示了骨钙蛋白-GPRC6A介导的骨-肌肉通讯在肌肉再生中的关键和新作用,这为功能性肌肉再生提供了有希望的治疗途径。
    The progressive deterioration of tissue-tissue crosstalk with aging causes a striking impairment of tissue homeostasis and functionality, particularly in the musculoskeletal system. Rejuvenation of the systemic and local milieu via interventions such as heterochronic parabiosis and exercise has been reported to improve musculoskeletal homeostasis in aged organisms. We have shown that Ginkgolide B (GB), a small molecule from Ginkgo biloba, improves bone homeostasis in aged mice by restoring local and systemic communication, implying a potential for maintaining skeletal muscle homeostasis and enhancing regeneration. In this study, we investigated the therapeutic efficacy of GB on skeletal muscle regeneration in aged mice.
    Muscle injury models were established by barium chloride induction into the hind limb of 20-month-old mice (aged mice) and into C2C12-derived myotubes. Therapeutic efficacy of daily administrated GB (12 mg/kg body weight) and osteocalcin (50 μg/kg body weight) on muscle regeneration was assessed by histochemical staining, gene expression, flow cytometry, ex vivo muscle function test and rotarod test. RNA sequencing was used to explore the mechanism of GB on muscle regeneration, with subsequent in vitro and in vivo experiments validating these findings.
    GB administration in aged mice improved muscle regeneration (muscle mass, P = 0.0374; myofiber number/field, P = 0.0001; centre nucleus, embryonic myosin heavy chain-positive myofiber area, P = 0.0144), facilitated the recovery of muscle contractile properties (tetanic force, P = 0.0002; twitch force, P = 0.0005) and exercise performance (rotarod performance, P = 0.002), and reduced muscular fibrosis (collagen deposition, P < 0.0001) and inflammation (macrophage infiltration, P = 0.03). GB reversed the aging-related decrease in the expression of osteocalcin (P < 0.0001), an osteoblast-specific hormone, to promote muscle regeneration. Exogenous osteocalcin supplementation was sufficient to improve muscle regeneration (muscle mass, P = 0.0029; myofiber number/field, P < 0.0001), functional recovery (tetanic force, P = 0.0059; twitch force, P = 0.07; rotarod performance, P < 0.0001) and fibrosis (collagen deposition, P = 0.0316) in aged mice, without an increased risk of heterotopic ossification.
    GB treatment restored the bone-to-muscle endocrine axis to reverse aging-related declines in muscle regeneration and thus represents an innovative and practicable approach to managing muscle injuries. Our results revealed the critical and novel role of osteocalcin-GPRC6A-mediated bone-to-muscle communication in muscle regeneration, which provides a promising therapeutic avenue in functional muscle regeneration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号