urolithin A

尿石素 A
  • 文章类型: Journal Article
    最近的研究证实,焦亡参与了肺动脉高压(PH)的进展,可以促进肺动脉重塑。尿磷脂A(UA),一种肠道菌群代谢产物,包括鞣花酸(EA)和鞣花酸,已被证明在各种病理条件下对焦亡具有抑制作用。然而,其对PH的作用尚未确定。为了研究UA在缓解PH方面的潜力,小鼠暴露于缺氧(10%氧气,4周)诱导PH,有或没有UA治疗。此外,进行了体外实验以进一步揭示潜在的机制。UA的体内治疗通过减轻肺重塑来抑制PH的进展。在PH小鼠模型中,与细胞凋亡相关的基因显着上调,并在施用UA后逆转。据此,UA治疗可通过AMPK/NF-κB/NLRP3途径显著抑制缺氧诱导的肺动脉平滑肌细胞(PASMC)的凋亡。我们的研究结果表明,UA治疗通过抑制PASMC焦凋亡有效缓解PH进展,这代表了一种创新的PH治疗方法。
    Recent studies confirmed that pyroptosis is involved in the progression of pulmonary hypertension (PH), which could promote pulmonary artery remodeling. Urolithin A (UA), an intestinal flora metabolite of ellagitannins (ETs) and ellagic acid (EA), has been proven to possess inhibitory effects on pyroptosis under various pathological conditions. However, its role on PH remained undetermined. To investigate the potential of UA in mitigating PH, mice were exposed to hypoxia (10% oxygen, 4 weeks) to induce PH, with or without UA treatment. Moreover, in vitro experiments were carried out to further uncover the underlying mechanisms. The in vivo treatment of UA suppressed the progression of PH via alleviating pulmonary remodeling. Pyroptosis-related genes were markedly upregulated in mice models of PH and reversed after the administration of UA. In accordance with that, UA treatment significantly inhibited hypoxia-induced pulmonary arterial smooth muscle cell (PASMC) pyroptosis via the AMPK/NF-κB/NLRP3 pathway. Our results revealed that UA treatment effectively mitigated PH progression through inhibiting PASMC pyroptosis, which represents an innovative therapeutic approach for PH.
    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
    昼夜节律在肠道稳态和肠道免疫功能中起重要作用。据报道,昼夜节律失调会导致肠道微生物群失调,肠屏障破坏,引发肠道炎症.然而,肠道微生物代谢产物与肠道屏障昼夜节律的关系尚不清楚。尿磷脂A(UA),一种肠道微生物代谢产物,在这项研究中被选中。成果显示UA对肠上皮细胞时钟基因BMAL1和PER2的表达节律有影响。此外,该研究调查了UA对时钟基因(BMAL1和PER2)和紧密连接(OCLN,TJP1和CLND1),所有这些都因炎症而失调。此外,通过口服给予雌性C57BL/6小鼠UA预处理显示粪便IgA浓度的改善,紧密连接表达式(Clnd1和Clnd4),在使用DSS治疗诱导的DSS诱导的结肠炎模型中和时钟基因表达(Bmal1和Per2)。最后,Nrf2-SIRT1信号通路通过拮抗剂治疗证实参与UA对肠上皮细胞昼夜节律的影响。这项研究还表明,有证据表明,UA喂食对中央时钟有影响,SCN中的昼夜节律。因此,这项研究强调了UA通过改善肠屏障和SCN的昼夜节律失调,在治疗IBD等睡眠障碍疾病方面的潜力。
    Circadian rhythm plays an important role in intestinal homeostasis and intestinal immune function. Circadian rhythm dysregulation was reported to induce intestinal microbiota dysbiosis, intestinal barrier disruption, and trigger intestinal inflammation. However, the relationship between intestinal microbiota metabolites and the circadian rhythm of the intestinal barrier was still unclear. Urolithin A (UA), a kind of intestinal microbial metabolite, was selected in this study. Results showed UA influenced on the expression rhythm of the clock genes BMAL1 and PER2 in intestinal epithelial cells. Furthermore, the study investigated the effects of UA on the expression rhythms of clock genes (BMAL1 and PER2) and tight junctions (OCLN, TJP1, and CLND1), all of which were dysregulated by inflammation. In addition, UA pre-treatment by oral administration to female C57BL/6 mice showed the improvement in the fecal IgA concentrations, tight junction expression (Clnd1 and Clnd4), and clock gene expression (Bmal1 and Per2) in a DSS-induced colitis model induced using DSS treatment. Finally, the Nrf2-SIRT1 signaling pathway was confirmed to be involved in UA\'s effect on the circadian rhythm of intestinal epithelial cells by antagonist treatment. This study also showed evidence that UA feeding showed an impact on the central clock, which are circadian rhythms in SCN. Therefore, this study highlighted the potential of UA in treating diseases like IBD with sleeping disorders by improving the dysregulated circadian rhythms in both the intestinal barrier and the SCN.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: English Abstract
    目的:观察尿石素A(UA)对呼吸道合胞病毒(RSV)诱导的新生小鼠肺部感染的治疗作用并探讨其机制。
    方法:Babl/c小鼠(5-7日龄)经鼻腔滴注RSV,并在感染2小时后腹膜内注射盐水或2.5、5和10mg/kgUA,然后每天一次,持续2周。然后收集支气管肺泡灌洗液(BALF)用于检测炎症细胞和介质,HE染色评价肺部病理。用2.5、5或10μmol/LUA处理RSV感染的BEAS-2B细胞。炎症因子,细胞活力,使用ELISA分析细胞凋亡和自噬,CCK-8测定,TUNEL染色,流式细胞术,Western印迹和免疫荧光染色。使用qRT-PCR检测miR-136和Sirt1mRNA的细胞表达。使用双荧光素酶报告系统来验证miR-136和Sirt1之间的结合。
    结果:在新生Babl/c小鼠中,RSV感染引起明显的肺部病变,促进肺细胞凋亡与LC3-Ⅱ/Ⅰ,Beclin-1和miR-136表达,并增加了细胞总数,BALF中的炎症细胞和因子和p62和Sirt1表达降低。所有这些变化都被UA剂量依赖性地缓解。在BEAS-2B细胞中,RSV感染显著增加细胞凋亡,LC3B阳性细胞和miR-136表达及Sirt1表达降低(P<0.01),其被UA剂量依赖性地减毒。双荧光素酶报告基因测定证实了miR-136和Sirt1之间的结合。在用UA处理的RSV感染的BEAS-2B细胞中,过表达miR-136和Ex527处理均显著增加炎症因子和细胞凋亡,但降低LC3B表达,这些变化通过联合治疗得到了进一步增强。
    结论:UA通过激活miR-136介导的Sirt1信号通路改善RSV诱导的新生小鼠肺部感染。
    OBJECTIVE: To observe the therapeutic effects of urolithin A (UA) on respiratory syncytial virus (RSV)-induced lung infection in neonatal mice and explore the underlying mechanisms.
    METHODS: Babl/c mice (5-7 days old) were subjected to nasal instillation of RSV and received intraperitoneal injection of saline or 2.5, 5 and 10 mg/kg UA 2 h after the infection and then once daily for 2 weeks. Bronchoalveolar lavage fluid (BALF) was then collected for detection of inflammatory cells and mediators, and lung pathology was evaluated with HE staining. RSV-infected BEAS-2B cells were treated with 2.5, 5 or 10 µmol/ L UA. Inflammatory factors, cell viability, apoptosis and autophagy were analyzed using ELISA, CCK-8 assay, TUNEL staining, flow cytometry, Western blotting and immunofluorescence staining. The cellular expressions of miR-136 and Sirt1 mRNAs were detected using qRT-PCR. A dual-luciferase reporter system was used to verify the binding between miR-136 and Sirt1.
    RESULTS: In neonatal Babl/c mice, RSV infection caused obvious lung pathologies, promoted pulmonary cell apoptosis and LC3-Ⅱ/Ⅰ, Beclin-1 and miR-136 expressions, and increased the total cell number, inflammatory cells and factors in the BALF and decreased p62 and Sirt1 expressions. All these changes were alleviated dose-dependently by UA. In BEAS-2B cells, RSV infection significantly increased cell apoptosis, LC3B-positive cells and miR-136 expression and reduced Sirt1 expression (P<0.01), which were dose-dependently attenuated by UA. Dual-luciferase reporter assay confirmed the binding between miR-136 and Sirt1. In RSV-infected BEAS-2B cells with UA treatment, overexpression of miR-136 and Ex527 treatment both significantly increased the inflammatory factors and cell apoptosis but decreased LC3B expression, and these changes were further enhanced by their combined treatment.
    CONCLUSIONS: UA ameliorates RSV-induced lung infection in neonatal mice by activating miR-136-mediated Sirt1 signaling pathway.
    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
    保持粘液层对于先天免疫系统至关重要。尿石素A(UroA)是一种肠道微生物来源的代谢产物;然而,其作为物理屏障对粘蛋白产生的影响尚不清楚。本研究旨在阐明UroA对结肠粘蛋白产生的保护作用。使用野生型小鼠的体内实验,NF-E2相关因子2(Nrf2)缺陷小鼠,和用芳香烃受体(AhR)拮抗剂治疗的野生型小鼠进行研究UroA的生理作用。进行使用产粘蛋白细胞(LS174T)的体外测定以评估UroA处理后的粘液产生。我们发现UroA通过Nrf2和AhR信号传导促进的粘蛋白2表达增强而增厚小鼠结肠粘液,而不改变紧密连接。UroA降低异硫氰酸荧光素-葡聚糖实验中的粘膜通透性并减轻葡聚糖硫酸钠诱导的结肠炎。UroA处理增加了产生短链脂肪酸的细菌和丙酸浓度。LS174T细胞研究证实UroA通过AhR和Nrf2途径促进粘液产生。总之,UroA诱导的增强的肠粘液分泌是通过Nrf-2和AhR的作用介导的,有助于维持肠道屏障功能。
    Maintaining the mucus layer is crucial for the innate immune system. Urolithin A (Uro A) is a gut microbiota-derived metabolite; however, its effect on mucin production as a physical barrier remains unclear. This study aimed to elucidate the protective effects of Uro A on mucin production in the colon. In vivo experiments employing wild-type mice, NF-E2-related factor 2 (Nrf2)-deficient mice, and wild-type mice treated with an aryl hydrocarbon receptor (AhR) antagonist were conducted to investigate the physiological role of Uro A. Additionally, in vitro assays using mucin-producing cells (LS174T) were conducted to assess mucus production following Uro A treatment. We found that Uro A thickened murine colonic mucus via enhanced mucin 2 expression facilitated by Nrf2 and AhR signaling without altering tight junctions. Uro A reduced mucosal permeability in fluorescein isothiocyanate-dextran experiments and alleviated dextran sulfate sodium-induced colitis. Uro A treatment increased short-chain fatty acid-producing bacteria and propionic acid concentration. LS174T cell studies confirmed that Uro A promotes mucus production through the AhR and Nrf2 pathways. In conclusion, the enhanced intestinal mucus secretion induced by Uro A is mediated through the actions of Nrf-2 and AhR, which help maintain intestinal barrier function.
    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:年龄相关性黄斑变性(AMD)是发达国家老年人失明的主要原因,到2040年,受影响的人数预计将几乎翻一番。视网膜是我们体内最高的代谢需求之一,部分或完全由神经视网膜和视网膜色素上皮(RPE)中的线粒体满足。分别。连同其有丝分裂后的状态和来自入射光的持续光氧化损伤,视网膜需要一个涉及自噬的严密调控的内务系统.天然多酚尿石素A(UA)在衰老和年龄相关疾病的几种模型中显示出神经保护作用,主要归因于其诱导线粒体自噬和线粒体生物合成的能力。碘酸钠(SI)给药概括了AMD的晚期阶段,包括地理萎缩和感光细胞死亡。
    方法:体外,使用离体和体内模型来测试SI模型中UA的神经保护潜能。功能测定(OCT,ERGs),细胞分析(流式细胞术,qPCR)和精细共聚焦显微镜(免疫组织化学,串联选择性自噬记者)帮助解决了这个问题。
    结果:UA减轻了SI治疗小鼠的神经变性并保留了视觉功能。同时,我们在SI损伤诱导后观察到严重的蛋白质停滞缺陷,包括自噬体积累,在接受UA的动物中得到解决。UA治疗可恢复自噬通量并触发PINK1/Parkin依赖性线粒体自噬,正如文献中先前报道的那样。由SI引起的自噬阻断是由严重的溶酶体膜透化引起的。虽然UA不诱导溶酶体生物发生,它确实通过嗜血恢复了透化溶酶体的上循环。在SI处理的细胞中,细胞自噬适配器SQSTM1/p62的敲低消除了UA的生存力挽救,加剧溶酶体缺陷并抑制自噬。
    结论:总的来说,这些数据突出显示了UA在AMD治疗中的一种新的推定应用,即它通过促进p62依赖性的细胞自噬来维持蛋白质抑制,从而绕过溶酶体缺陷.
    BACKGROUND: Age-related macular degeneration (AMD) is the leading cause of blindness in elderly people in the developed world, and the number of people affected is expected to almost double by 2040. The retina presents one of the highest metabolic demands in our bodies that is partially or fully fulfilled by mitochondria in the neuroretina and retinal pigment epithelium (RPE), respectively. Together with its post-mitotic status and constant photooxidative damage from incoming light, the retina requires a tightly-regulated housekeeping system that involves autophagy. The natural polyphenol Urolithin A (UA) has shown neuroprotective benefits in several models of aging and age-associated disorders, mostly attributed to its ability to induce mitophagy and mitochondrial biogenesis. Sodium iodate (SI) administration recapitulates the late stages of AMD, including geographic atrophy and photoreceptor cell death.
    METHODS: A combination of in vitro, ex vivo and in vivo models were used to test the neuroprotective potential of UA in the SI model. Functional assays (OCT, ERGs), cellular analysis (flow cytometry, qPCR) and fine confocal microscopy (immunohistochemistry, tandem selective autophagy reporters) helped address this question.
    RESULTS: UA alleviated neurodegeneration and preserved visual function in SI-treated mice. Simultaneously, we observed severe proteostasis defects upon SI damage induction, including autophagosome accumulation, that were resolved in animals that received UA. Treatment with UA restored autophagic flux and triggered PINK1/Parkin-dependent mitophagy, as previously reported in the literature. Autophagy blockage caused by SI was caused by severe lysosomal membrane permeabilization. While UA did not induce lysosomal biogenesis, it did restore upcycling of permeabilized lysosomes through lysophagy. Knockdown of the lysophagy adaptor SQSTM1/p62 abrogated viability rescue by UA in SI-treated cells, exacerbated lysosomal defects and inhibited lysophagy.
    CONCLUSIONS: Collectively, these data highlight a novel putative application of UA in the treatment of AMD whereby it bypasses lysosomal defects by promoting p62-dependent lysophagy to sustain proteostasis.
    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
    酒精相关性肝病(ALD)被认为是全球健康危机,导致大约20%的肝癌相关死亡。肠道微生物群的菌群失调与ALD的发展有关,与肠道微生物代谢产物尿石素A(UA)表现出缓解肝脏症状的潜力。然而,UA对ALD的保护功效及其由微生物群介导的潜在机制仍然难以捉摸。在这项研究中,我们提供的证据表明,UA通过主要尿蛋白1(MUP1)介导的特定肠道-微生物群-肝轴有效改善酒精诱导的代谢紊乱和肝内质网(ER)应激.此外,UA表现出通过富集细菌的丰度来恢复酒精诱导的肠道微生物群生态失调的潜力(B.sartorii),双杆菌属(P.distasonis),和阿克曼西亚粘虫(A.粘液虫),以及它们的代谢产物丙酸.在使用抗生素耗尽肠道微生物区后,观察到UA发挥的肝保护作用的部分减弱。随后,进行了粪便微生物群移植(FMT)实验,以评估UA在ALD中的微生物群依赖性作用。来自UA处理的小鼠的FMT表现出与直接UA处理相当的功效。因为它通过MUP1的调制有效地减弱了ER应力。值得注意的是,在肝脏MUP1、肠道微生物组、和受UA影响的代谢组概况。有趣的是,口服富含UA的B.sartorii,P.Distasonis,和粘蛋白乳杆菌可以通过MUP1增强丙酸的产生以有效地抑制ER应激,模拟UA治疗。总的来说,这些发现阐明了UA通过肠道-微生物群-肝轴缓解ALD的因果机制.这种独特的机制为开发针对ALD的新型微生物组靶向治疗策略提供了启示。
    Alcohol-related liver disease (ALD) is recognized as a global health crisis, contributing to approximately 20% of liver cancer-associated fatalities. Dysbiosis of the gut microbiome is associated with the development of ALD, with the gut microbial metabolite urolithin A (UA) exhibiting a potential for alleviating liver symptoms. However, the protective efficacy of UA against ALD and its underlying mechanism mediated by microbiota remain elusive. In this study, we provide evidence demonstrating that UA effectively ameliorates alcohol-induced metabolic disorders and hepatic endoplasmic reticulum (ER) stress through a specific gut-microbiota-liver axis mediated by major urinary protein 1 (MUP1). Moreover, UA exhibited the potential to restore alcohol-induced dysbiosis of the intestinal microbiota by enriching the abundance of Bacteroides sartorii (B. sartorii), Parabacteroides distasonis (P. distasonis), and Akkermansia muciniphila (A. muciniphila), along with their derived metabolite propionic acid. Partial attenuation of the hepatoprotective effects exerted by UA was observed upon depletion of gut microbiota using antibiotics. Subsequently, a fecal microbiota transplantation (FMT) experiment was conducted to evaluate the microbiota-dependent effects of UA in ALD. FMT derived from mice treated with UA exhibited comparable efficacy to direct UA treatment, as it effectively attenuated ER stress through modulation of MUP1. It was noteworthy that strong associations were observed among the hepatic MUP1, gut microbiome, and metabolome profiles affected by UA. Intriguingly, oral administration of UA-enriched B. sartorii, P. distasonis, and A. muciniphila can enhance propionic acid production to effectively suppress ER stress via MUP1, mimicking UA treatment. Collectively, these findings elucidate the causal mechanism that UA alleviated ALD through the gut-microbiota-liver axis. This unique mechanism sheds light on developing novel microbiome-targeted therapeutic strategies against ALD.
    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
    功能性聚合物-脂质混合纳米颗粒(H-NP)是一类有前途的纳米载体,结合了聚合物和脂质纳米颗粒的好处,提供生物相容性,结构稳定性,高承载能力,and,最重要的是,优越的表面功能化。这里,我们报道了对转铁蛋白受体(TfR)具有特异性的高功能H-NP的合成和设计,使用小分子配体,藤黄酸(GA)。荧光研究揭示了H-NP的分子取向,脂质致密的核心被聚合物包围,用GA功能化。尿磷脂A,免疫调节剂和抗炎剂,作为通过传统的基于乳液的技术制备H-NP的模型药物样化合物,其中与GA缀合的聚合物纳米颗粒(P-NP)(157nm和52%的包封效率)和固体脂质纳米颗粒(L-NP)(186nm和29%的包封效率)相比,H-NP导致更小的颗粒(132nm)和更高的包封效率(10%的载药量为70%)。与使用人小肠上皮(FHs74)细胞的单个NP相比,H-NP显示出优异的细胞内积累。通过流式细胞术分析证明了体外功效,其中,在顺铂诱导的健康人近端肾小管细胞(HK2)模型中,通过降低TLR4,NF-κβ,和IL-β表达。这项初步工作强调了H-NP作为新型功能性聚合物-脂质药物递送系统的潜力,为未来研究其在解决癌症患者化疗引起的急性肾损伤方面的治疗潜力奠定了基础。
    Functional polymer-lipid hybrid nanoparticles (H-NPs) are a promising class of nanocarriers that combine the benefits of polymer and lipid nanoparticles, offering biocompatibility, structural stability, high loading capacity, and, most importantly, superior surface functionalization. Here, we report the synthesis and design of highly functional H-NPs with specificity toward the transferrin receptor (TfR), using a small molecule ligand, gambogic acid (GA). A fluorescence study revealed the molecular orientation of H-NPs, where the lipid-dense core is surrounded by a polymer exterior, functionalized with GA. Urolithin A, an immunomodulator and anti-inflammatory agent, served as a model drug-like compound to prepare H-NPs via traditional emulsion-based techniques, where H-NPs led to smaller particles (132 nm) and superior entrapment efficiencies (70 % at 10 % drug loading) compared to GA-conjugated polymeric nanoparticles (P-NPs) (157 nm and 52 % entrapment efficiency) and solid lipid nanoparticles (L-NPs) (186 nm and 29 % entrapment efficiency). H-NPs showed superior intracellular accumulation compared to individual NPs using human small intestinal epithelial (FHs 74) cells. The in vitro efficacy was demonstrated by flow cytometry analysis, in which UA-laden H-NPs showed excellent anti-inflammatory properties in cisplatin-induced injury in healthy human proximal tubular cell (HK2) model by decreasing the TLR4, NF-κβ, and IL-β expression. This preliminary work highlights the potential of H-NPs as a novel functional polymer-lipid drug delivery system, establishing the foundation for future research on its therapeutic potential in addressing chemotherapy-induced acute kidney injury in cancer 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
    背景:自噬受损,包括线粒体自噬和溶酶体功能受损,在阿尔茨海默病(AD)中起着关键作用。尿石素A(UA)是刺激线粒体自噬的鞣花酸的肠道微生物代谢产物。UA长期治疗AD的效果和作用机制尚不清楚。
    方法:我们在三个AD小鼠模型中解决了这些问题,电生理学,生物化学,和生物信息学方法。
    结果:长期UA治疗显著改善学习,记忆,不同AD转基因小鼠的嗅觉功能。UA还降低了淀粉样蛋白β(Aβ)和tau病理,并增强了长期增强作用。UA通过增加溶酶体功能诱导线粒体自噬。UA改善细胞溶酶体功能和正常溶酶体组织蛋白酶,主要是组织蛋白酶Z,为了恢复AD的溶酶体功能,表明组织蛋白酶在UA诱导的AD治疗作用中的关键作用。
    结论:我们的研究强调了溶酶体功能障碍在AD病因学中的重要性,并指出UA的高翻译潜力。
    结论:长期尿石素A(UA)治疗可改善学习,记忆,阿尔茨海默病(AD)小鼠的嗅觉功能。UA部分通过调节组织蛋白酶Z(Ctsz)蛋白恢复溶酶体功能。UA调节免疫应答和AD特异性病理生理途径。
    Compromised autophagy, including impaired mitophagy and lysosomal function, plays pivotal roles in Alzheimer\'s disease (AD). Urolithin A (UA) is a gut microbial metabolite of ellagic acid that stimulates mitophagy. The effects of UA\'s long-term treatment of AD and mechanisms of action are unknown.
    We addressed these questions in three mouse models of AD with behavioral, electrophysiological, biochemical, and bioinformatic approaches.
    Long-term UA treatment significantly improved learning, memory, and olfactory function in different AD transgenic mice. UA also reduced amyloid beta (Aβ) and tau pathologies and enhanced long-term potentiation. UA induced mitophagy via increasing lysosomal functions. UA improved cellular lysosomal function and normalized lysosomal cathepsins, primarily cathepsin Z, to restore lysosomal function in AD, indicating the critical role of cathepsins in UA-induced therapeutic effects on AD.
    Our study highlights the importance of lysosomal dysfunction in AD etiology and points to the high translational potential of UA.
    Long-term urolithin A (UA) treatment improved learning, memory, and olfactory function in Alzheimer\'s disease (AD) mice. UA restored lysosomal functions in part by regulating cathepsin Z (Ctsz) protein. UA modulates immune responses and AD-specific pathophysiological pathways.
    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
    众所周知,长期太空飞行会导致昼夜节律中断,由位于下丘脑视交叉上核(SCN)的中央起搏器驱动,但潜在的分子机制仍不清楚。这里,我们开发了一种大鼠模型,通过尾部悬吊和隔离(TSI)模拟微重力和隔离环境。我们发现TSI环境会对核心体温造成昼夜节律破坏,心率,和大鼠的运动活动节律,尤其是这些节奏的幅度。在TSI模型大鼠中,核心昼夜节律基因NR1D1显示出更高的蛋白质而不是mRNA水平,随着BMAL1水平的降低,这表明NR1D1可以通过翻译后调节来调节。自噬体标记物LC3可以通过LC3相互作用区(LIR)基序直接与NR1D1结合,并以线粒体自噬依赖性方式诱导NR1D1降解。线粒体自噬的缺陷导致NR1D1降解的逆转,从而抑制BMAL1的表达。在TSI模型的SCN中观察到线粒体自噬缺陷和随后的线粒体功能障碍。尿磷脂A(UA),线粒体自噬激活剂,表现出增强核心体温振幅的能力,心率,通过促进线粒体自噬诱导降解NR1D1来实现运动活动节律。累计,我们的结果表明,线粒体自噬通过调节NR1D1降解来发挥昼夜节律控制,揭示线粒体自噬是长期太空飞行以及SCN昼夜节律紊乱疾病的潜在靶标。
    Long-term spaceflight is known to induce disruptions in circadian rhythms, which are driven by a central pacemaker located in the suprachiasmatic nucleus (SCN) of the hypothalamus, but the underlying molecular mechanisms remain unclear. Here, we developed a rat model that simulated microgravity and isolation environments through tail suspension and isolation (TSI). We found that the TSI environment imposed circadian disruptions to the core body temperature, heart rate, and locomotor-activity rhythms of rats, especially in the amplitude of these rhythms. In TSI model rats\' SCNs, the core circadian gene NR1D1 showed higher protein but not mRNA levels along with decreased BMAL1 levels, which indicated that NR1D1 could be regulated through post-translational regulation. The autophagosome marker LC3 could directly bind to NR1D1 via the LC3-interacting region (LIR) motifs and induce the degradation of NR1D1 in a mitophagy-dependent manner. Defects in mitophagy led to the reversal of NR1D1 degradation, thereby suppressing the expression of BMAL1. Mitophagy deficiency and subsequent mitochondrial dysfunction were observed in the SCN of TSI models. Urolithin A (UA), a mitophagy activator, demonstrated an ability to enhance the amplitude of core body temperature, heart rate, and locomotor-activity rhythms by prompting mitophagy induction to degrade NR1D1. Cumulatively, our results demonstrate that mitophagy exerts circadian control by regulating NR1D1 degradation, revealing mitophagy as a potential target for long-term spaceflight as well as diseases with SCN circadian disruption.
    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
    神经炎症可以由各种刺激引发,包括病毒感染.病毒可以直接侵入大脑并感染神经元细胞,或间接在外周引发“细胞因子风暴”,最终导致大脑中的小胶质细胞活化。虽然小胶质细胞的这种初始激活对于病毒清除很重要,慢性激活导致过度的炎症和氧化应激,可能有神经毒性.值得注意的是,最近的研究表明,某些病毒,如甲型流感病毒,冠状病毒,疱疹病毒和EB病毒可能参与帕金森病等神经退行性疾病的发展,老年痴呆症,和多发性硬化症。因此,寻找针对由病毒感染引发的慢性神经炎症的治疗策略非常重要.这里,我们研究了尿石素A(UA)对病毒模拟物体外诱导的小胶质细胞活化的影响,聚I:C,在神经元的三重共培养系统中,星形胶质细胞和小胶质细胞。免疫细胞化学用于对小胶质细胞的形态变化进行全面的单细胞分析,以指示其反应状态。用UA治疗可显著防止聚I:C诱导的小胶质细胞反应状态,其特征在于小胶质细胞活化标志物CD68和IBA-1的表达增加。UA通过恢复小胶质细胞分枝来恢复聚I:C诱导的形态。此外,UA能够减少促炎介质CCL2,TNF-α的释放,和IL-1β,并在polyI:C处理的培养物中显示出细胞ROS产生减弱的趋势。总的来说,这项研究表明,UA作为健康饮食的组成部分,可能有助于预防病毒诱导的神经炎症,并且在未来的研究中可能具有治疗潜力,通过靶向相关的神经炎症过程来预防或治疗神经退行性疾病.
    Neuroinflammation can be triggered by various stimuli, including viral infections. Viruses can directly invade the brain and infect neuronal cells or indirectly trigger a \"cytokine storm\" in the periphery that eventually leads to microglial activation in the brain. While this initial activation of microglial cells is important for viral clearance, chronic activation leads to excessive inflammation and oxidative stress, which can be neurotoxic. Remarkebly, recent studies have shown that certain viruses such as influenza A virus, coronavirus, herpes virus and Epstein-Barr virus may be involved in the development of neurodegenerative diseases such as Parkinson\'s disease, Alzheimer\'s disease, and multiple sclerosis. Therefore, it is important to find therapeutic strategies against chronic neuroinflammation triggered by viral infections. Here, we investigated the effects of urolithin A (UA) on microglial activation in vitro induced by a viral mimetic, poly I:C, in a triple co-culture system of neurons, astrocytes and microglial cells. Immunocytochemistry was used to perform a comprehensive single-cell analysis of the morphological changes of microglia as an indicator of their reactive state. Treatment with UA significantly prevented the poly I:C-induced reactive state of microglia, which was characterized by increased expression of the microglial activation markers CD68 and IBA-1. UA restored the poly I:C-induced morphology by restoring microglial ramification. In addition, UA was able to reduce the release of the pro-inflammatory mediators CCL2, TNF-α, and IL-1β and showed a trend toward attenuation of cellular ROS production in poly I:C-treated cultures. Overall, this study suggests that UA as a component of a healthy diet may help prevent virus-induced neuroinflammation and may have therapeutic potential for future studies to prevent or treat neurodegenerative diseases by targeting the associated neuroinflammatory processes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号