urolithin A

尿石素 A
  • 文章类型: Journal Article
    六价铬(Cr(VI))暴露与胃肠道毒性有关,而分子途径和关键靶标仍然难以捉摸。计算毒理学分析预测了蛋白磷酸酶2A(PP2A)与Cr(VI)引起的肠损伤基因之间的相关性。这里,我们建立了肠上皮特异性敲除Ppp2r1a(编码PP2AAα亚基)的小鼠模型,以研究Cr(VI)诱导的小肠毒性的潜在机制。杂合小鼠(HE)和匹配的野生型(WT)同窝以0、5、20、80mg/L连续28天给予Cr(VI)。Cr(VI)治疗导致隐窝增生,上皮细胞凋亡,和肠屏障功能障碍,伴随着WT小鼠杯状细胞计数和Occludin表达的下降。值得注意的是,这些效应在HE小鼠中加重,表明PP2AAα缺乏使小鼠对Cr(VI)引起的肠损伤具有易感性。综合数据分析和生物学实验表明,Cr(VI)暴露可以降低Ser127处的YAP1磷酸化,但增加蛋白质的表达和活性。与升高的TAZ蛋白一起驱动损伤后的上皮隐窝细胞增殖,提示Hippo/YAP1信号通路参与Cr(VI)诱导的肠毒性。然而,HE小鼠中YAP1的磷酸化增强导致肠上皮的增殖/修复缺陷,从而加剧Cr(VI)诱导的肠屏障功能障碍。值得注意的是,通过分子对接和进一步的研究,我们鉴定了尿脂素A,微生物代谢产物,减弱Cr(VI)诱导的肠屏障功能破坏,部分通过调节YAP1的表达和活性。我们的发现揭示了新的分子途径参与了Cr(VI)引起的小肠损伤,尿石素A可以潜在地预防环境危害引起的肠道疾病。
    Hexavalent chromium (Cr(VI)) exposure has been linked with gastrointestinal toxicity, whereas the molecular pathways and key targets remain elusive. Computational toxicology analysis predicted the correlation between protein phosphatase 2A (PP2A) and genes regarding Cr(VI)-induced intestinal injury. Here, we generated a mouse model with intestinal epithelium-specific knock-out of Ppp2r1a (encoding PP2A Aα subunit) to investigate the mechanisms underlying Cr(VI)-induced small intestinal toxicity. Heterozygous mice (HE) and matched wild-type (WT) littermates were administrated with Cr(VI) at 0, 5, 20, 80 mg/L for 28 successive days. Cr(VI) treatment led to crypt hyperplasia, epithelial cell apoptosis, and intestinal barrier dysfunction, accompanied by the decline of goblet cell counts and Occludin expression in WT mice. Notably, these effects were aggravated in HE mice, indicating that PP2A Aα deficiency conferred mice with susceptibility to Cr(VI)-induced intestinal injury. Integrated data analysis and biological experiments revealed Cr(VI) exposure could decrease YAP1 phosphorylation at Ser127 but increase protein expression and activity, together with elevated TAZ protein driving epithelial crypt cells proliferation following damage, suggesting the involvement of Hippo/YAP1 signaling pathway in Cr(VI)-induced intestinal toxicity. Nevertheless, the enhanced phosphorylation of YAP1 in HE mice resulted in proliferation/repair defects in intestinal epithelium, thereby exacerbating Cr(VI)-induced gut barrier dysfunction. Notably, by molecular docking and further studies, we identified Urolithin A, a microbial metabolite, attenuated Cr(VI)-induced disruption of intestinal barrier function, partly by modulating YAP1 expression and activity. Our findings reveal the novel molecular pathways participated in Cr(VI)-caused small intestinal injury and urolithin A could potentially protect against environmental hazards-induced intestinal 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
    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: 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
    目的:肝硬化门脉高压(CPH)是导致肝硬化患者死亡的主要原因。超过50%的CPH患者接受了当前的临床药物治疗,由于门静脉压力降低不足,仍然存在静脉曲张破裂出血或有时死亡。升高的肝内血管阻力(IHVR)在增加门静脉压力中起着基本作用。由于其在降低门静脉压力和维持正常门静脉流入以保持肝功能方面的有效作用,降低IHVR被认为是一种最佳的抗CPH策略,但没有临床药物。我们旨在研究微生物来源的尿石素A(UroA)在IHVR和CPH中的保护作用。
    方法:对小鼠进行CCl4或BDL手术以诱导肝纤维化和CPH。16SrRNA基因测序用于微生物分析。转录组学和代谢组学分析用于研究宿主和细胞反应。
    结果:UroA在CPH患者中明显缺乏,与疾病严重程度呈负相关。在CPH小鼠中也证实了UroA缺乏症,并且与产生UroA的细菌菌株(鼠乳杆菌,L.murinus).肝星状细胞(HSC)的谷氨酰胺溶解被鉴定为先前未识别的UroA靶标。UroA抑制谷氨酰胺酶1的活性以抑制谷氨酰胺分解,它通过减轻IHVR来抵消HSC的纤维化和收缩并改善CPH。补充UroA或L.murinus可有效改善小鼠的CPH。
    结论:我们首次确定肠道微生物代谢产物UroA的缺乏是CPH的重要原因。我们证明UroA通过抑制HSC谷氨酰胺分解降低IHVR发挥优异的抗CPH作用,这凸显了其作为CPH新型治疗剂的巨大潜力。
    OBJECTIVE: Cirrhotic portal hypertension (CPH) is the leading cause of mortality in patients with cirrhosis. Over 50% of patients with CPH treated with current clinical pharmacotherapy still present variceal bleeding or sometimes death owing to insufficient reduction in portal pressure. Elevated intrahepatic vascular resistance (IHVR) plays a fundamental role in increasing portal pressure. Because of its potent effect in reducing portal pressure and maintaining normal portal inflow to preserve liver function, lowering the IHVR is acknowledged as an optimal anti-CPH strategy but without clinical drugs. We aimed to investigate the protective effect of microbial-derived Urolithin A (UroA) in IHVR and CPH.
    METHODS: Carbon tetrachloride or bile duct ligation surgery was administered to mice to induce liver fibrosis and CPH. 16S rRNA gene sequencing was used for microbial analysis. Transcriptomics and metabolomics analyses were employed to study the host and cell responses.
    RESULTS: UroA was remarkably deficient in patients with CPH and was negatively correlated with disease severity. UroA deficiency was also confirmed in CPH mice and was associated with a reduced abundance of UroA-producing bacterial strain (Lactobacillus murinus, L. murinus). Glutaminolysis of hepatic stellate cells (HSCs) was identified as a previously unrecognized target of UroA. UroA inhibited the activity of glutaminase1 to suppress glutaminolysis, which counteracted fibrogenesis and contraction of HSCs and ameliorated CPH by relieving IHVR. Supplementation with UroA or L. murinus effectively ameliorated CPH in mice.
    CONCLUSIONS: We for the first time identify the deficiency of gut microbial metabolite UroA as an important cause of CPH. We demonstrate that UroA exerts an excellent anti-CPH effect by suppressing HSC glutaminolysis to lower the IHVR, which highlighted its great potential as a novel therapeutic agent for CPH.
    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
    这项工作是研究四种益生元糖阿拉伯树胶(GA)的作用,低聚果糖(FOS),魔芋葡甘聚糖(KGM),和菊粉(INU)掺入对包封效率(EE)的影响,物理化学稳定性,和尿石素A-负载脂质体(UroA-LP)的体外消化。还通过体外结肠发酵研究了脂质体对肠道菌群的调节。结果表明,用GA包被的脂质体表现出最佳的EE,生物可及性,储存和热稳定性,生物可及性是UroA-LP的1.67倍。用FOS涂覆的UroA-LP显示出最佳的冻融稳定性和转化。同时,糖的添加显著提高了拟杆菌的相对丰度,降低了变形杆菌和放线菌的丰度。涂有FOS的UroA-LP,INU,和GA表现出最高的副杆菌属有益菌丰度,Monoglobus,和结核分枝杆菌,分别。FOS还可以降低有害细菌Collinsella和肠球菌的丰度,增加乙酸的含量,丁酸和异丁酸。因此,益生元糖可以改善EE,物理化学稳定性,UroA-LP的肠道菌群调节,并促进UroA的生物可利用性,但是效率因糖类类型而异,这为UroA在食品工业中的应用和提高其生物活性奠定了基础。
    This work was to investigate the effect of four prebiotic saccharides gum arabic (GA), fructooligosaccharide (FOS), konjac glucomannan (KGM), and inulin (INU) incorporation on the encapsulation efficiency (EE), physicochemical stability, and in vitro digestion of urolithin A-loaded liposomes (UroA-LPs). The regulation of liposomes on gut microbiota was also investigated by in vitro colonic fermentation. Results indicated that liposomes coated with GA showed the best EE, bioaccessibility, storage and thermal stability, the bioaccessibility was 1.67 times of that of UroA-LPs. The UroA-LPs coated with FOS showed the best freeze-thaw stability and transformation. Meanwhile, saccharides addition remarkably improved the relative abundance of Bacteroidota, reduced the abundances of Proteobacteria and Actinobacteria. The UroA-LPs coated with FOS, INU, and GA exhibited the highest beneficial bacteria abundance of Parabacteroides, Monoglobus, and Phascolarctobacterium, respectively. FOS could also decrease the abundance of harmful bacteria Collinsella and Enterococcus, and increase the levels of acetic acid, butyric acid and iso-butyric acid. Consequently, prebiotic saccharides can improve the EE, physicochemical stability, gut microbiota regulation of UroA-LPs, and promote the bioaccessibility of UroA, but the efficiency varied based on saccharides types, which can lay a foundation for the application of UroA in foods industry and for the enhancement of its bio-activities.
    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
    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
    尿磷脂A(UroA),一种膳食植物化学物质,由富含天然多酚的水果的肠道细菌产生。人体内ET代谢为UroA的效率取决于肠道微生物群。UroA已显示出多种药理活性。在这项研究中,我们研究了UroA对动脉粥样硬化病变发展和稳定性的影响。采用高脂高胆固醇饮食喂养载脂蛋白E缺陷(ApoE-/-)小鼠3个月建立动脉粥样硬化模型。同时给予小鼠UroA(50mg·kg-1·d-1,i.g.)。我们显示UroA给药显著减少了饮食诱导的头臂动脉粥样硬化病变,斑块中的巨噬细胞含量,内皮粘附分子的表达,斑块内出血和坏死核心的大小,同时增加了平滑肌肌动蛋白的表达和纤维帽的厚度,暗示斑块稳定的特征。使用TNF-α刺激的人内皮细胞阐明了潜在的机制。UroA(10、25、50μM)预处理剂量依赖性地抑制TNF-α诱导的内皮细胞活化和单核细胞粘附。然而,UroA对TNF-α刺激的人脐静脉内皮细胞(HUVECs)的抗炎作用与NF-κBp65通路无关。我们进行了RNA测序谱分析,以确定与血管功能相关的基因(DEG)的差异表达,炎症反应,UroA预处理的HUVECs中的细胞粘附和血栓形成。人类疾病富集分析表明,DEGs与心血管疾病显着相关。我们证明UroA预处理通过促进NO产生和降低TNF-α刺激的HUVECs中YAP/TAZ蛋白表达和TEAD转录活性来减轻内皮炎症。另一方面,我们发现UroA在ApoE-/-小鼠肝脏中调节脂肪转录因子SREBP1/2的转录和裂解,从而改善胆固醇代谢。本研究为预防动脉粥样硬化的新饮食治疗方案提供了实验依据。
    Urolithin A (UroA), a dietary phytochemical, is produced by gut bacteria from fruits rich in natural polyphenols ellagitannins (ETs). The efficiency of ETs metabolism to UroA in humans depends on gut microbiota. UroA has shown a variety of pharmacological activities. In this study we investigated the effects of UroA on atherosclerotic lesion development and stability. Apolipoprotein E-deficient (ApoE-/-) mice were fed a high-fat and high-cholesterol diet for 3 months to establish atherosclerosis model. Meanwhile the mice were administered UroA (50 mg·kg-1·d-1, i.g.). We showed that UroA administration significantly decreased diet-induced atherosclerotic lesions in brachiocephalic arteries, macrophage content in plaques, expression of endothelial adhesion molecules, intraplaque hemorrhage and size of necrotic core, while increased the expression of smooth muscle actin and the thickness of fibrous cap, implying features of plaque stabilization. The underlying mechanisms were elucidated using TNF-α-stimulated human endothelial cells. Pretreatment with UroA (10, 25, 50 μM) dose-dependently inhibited TNF-α-induced endothelial cell activation and monocyte adhesion. However, the anti-inflammatory effects of UroA in TNF-α-stimulated human umbilical vein endothelial cells (HUVECs) were independent of NF-κB p65 pathway. We conducted RNA-sequencing profiling analysis to identify the differential expression of genes (DEGs) associated with vascular function, inflammatory responses, cell adhesion and thrombosis in UroA-pretreated HUVECs. Human disease enrichment analysis revealed that the DEGs were significantly correlated with cardiovascular diseases. We demonstrated that UroA pretreatment mitigated endothelial inflammation by promoting NO production and decreasing YAP/TAZ protein expression and TEAD transcriptional activity in TNF-α-stimulated HUVECs. On the other hand, we found that UroA administration modulated the transcription and cleavage of lipogenic transcription factors SREBP1/2 in the liver to ameliorate cholesterol metabolism in ApoE-/- mice. This study provides an experimental basis for new dietary therapeutic option to prevent atherosclerosis.
    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
    焦虑和社交退缩在阿尔茨海默病(AD)患者中非常普遍。然而,这些症状背后的神经回路机制仍然难以捉摸,并且需要有效的预防策略。
    本研究旨在阐明AD中社交焦虑的神经回路机制。
    我们利用5xFAD小鼠进行了一系列实验,包括光遗传学操作,串联质谱标记蛋白质组分析,行为评估,和免疫荧光染色。
    在5xFAD小鼠中,我们观察到基底外侧杏仁核(aBLA)前部有明显的淀粉样β(Aβ)积累。行为上,6个月大的5xFAD小鼠在社交互动过程中表现出过度的社交回避。同时,从aBLA到腹侧海马CA1(vCA1)的通路被显著激活,并在社会互动过程中表现出混乱的放电模式.通过光遗传学抑制aBLA-vCA1通路,我们有效地提高了5xFAD小鼠的社交能力。在存在Aβ积累的情况下,我们确定了aBLA内蛋白质网络的明显变化。在服用尿石素A(UA)一个月后,我们观察到aBLA内异常蛋白网络的显著恢复。UA治疗也减弱了aBLA-vCA1途径的无序放电,导致社交能力的提高。
    aBLA-vCA1回路是在AD进展过程中响应Aβ积累的脆弱途径,在Aβ引起的社交焦虑中起着至关重要的作用。针对aBLA-vCA1回路和UA给药都是改善Aβ受损社交能力的有效策略。
    UNASSIGNED: Anxiety and social withdrawal are highly prevalent among patients with Alzheimer\'s disease (AD). However, the neural circuit mechanisms underlying these symptoms remain elusive, and there is a need for effective prevention strategies.
    UNASSIGNED: This study aims to elucidate the neural circuitry mechanisms underlying social anxiety in AD.
    UNASSIGNED: We utilized 5xFAD mice and conducted a series of experiments including optogenetic manipulation, Tandem Mass Tag-labeled proteome analysis, behavioral assessments, and immunofluorescence staining.
    UNASSIGNED: In 5xFAD mice, we observed significant amyloid-β (Aβ) accumulation in the anterior part of basolateral amygdala (aBLA). Behaviorally, 6-month-old 5xFAD mice displayed excessive social avoidance during social interaction. Concurrently, the pathway from aBLA to ventral hippocampal CA1 (vCA1) was significantly activated and exhibited a disorganized firing patterns during social interaction. By optogenetically inhibiting the aBLA-vCA1 pathway, we effectively improved the social ability of 5xFAD mice. In the presence of Aβ accumulation, we identified distinct changes in the protein network within the aBLA. Following one month of administration of Urolithin A (UA), we observed significant restoration of the abnormal protein network within the aBLA. UA treatment also attenuated the disorganized firings of the aBLA-vCA1 pathway, leading to an improvement in social ability.
    UNASSIGNED: The aBLA-vCA1 circuit is a vulnerable pathway in response to Aβ accumulation during the progression of AD and plays a crucial role in Aβ-induced social anxiety. Targeting the aBLA-vCA1 circuit and UA administration are both effective strategies for improving the Aβ-impaired social ability.
    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
    重症急性胰腺炎(SAP),以胰腺腺泡细胞死亡为特征,目前缺乏有效的靶向治疗。鞣花酸(EA),富含石榴,在SAP治疗中显示出有希望的抗炎和抗氧化作用。然而,其他形式的EA的角色,例如从石榴中提取的植物细胞外囊泡(EV),和尿磷脂A(UA),从EA通过体内肠道微生物群代谢转化,尚未明确阐明。我们的研究旨在比较石榴衍生的EV(P-EV)和UA在SAP治疗中的作用,以筛选有效的制剂并探讨其保护SAP腺泡细胞的机制。通过比较P-EV和UA对受损腺泡细胞的保护作用,UA的治疗效果优于P-EV。随后,我们进一步讨论了UA缓解SAP炎症的机制。体内动物实验发现,UA不仅能改善SAP小鼠胰腺组织的炎症环境和外周血循环,而且通过胰腺组织转录组学和透射电镜等结果揭示了UA改善SAP的机制可能与线粒体和内质网(ER)有关。进一步研究发现,UA可以调节ER-线粒体钙通道,减少胰腺组织坏死。小鼠胰腺类器官和腺泡细胞的体外实验也证实,UA可以通过调节ER-线粒体钙通道和坏死途径蛋白来改善胰腺炎症。这项研究不仅探索了植物EV对SAP的治疗作用,而且还揭示了UA可以通过调节ER-线粒体钙通道和减少腺泡细胞坏死来减轻SAP。提供对SAP发病机制和潜在治疗的见解。
    Severe acute pancreatitis (SAP), characterized by pancreatic acinar cell death, currently lacks effective targeted therapies. Ellagic acid (EA), rich in pomegranate, shows promising anti-inflammatory and antioxidant effects in SAP treatment. However, the roles of other forms of EA, such as plant extracellular vesicles (EVs) extracted from pomegranate, and Urolithin A (UA), converted from EA through gut microbiota metabolism in vivo, have not been definitively elucidated. Our research aimed to compare the effects of pomegranate-derived EVs (P-EVs) and UA in the treatment of SAP to screen an effective formulation and to explore its mechanisms in protecting acinar cells in SAP. By comparing the protective effects of P-EVs and UA on injured acinar cells, UA showed superior therapeutic effects than P-EVs. Subsequently, we further discussed the mechanism of UA in alleviating SAP inflammation. In vivo animal experiments found that UA could not only improve the inflammatory environment of pancreatic tissue and peripheral blood circulation in SAP mice but also revealed that the mechanism of UA in improving SAP might be related to mitochondria and endoplasmic reticulum (ER) through the results including pancreatic tissue transcriptomics and transmission electron microscopy. Further research found that UA could regulate ER-mitochondrial calcium channels and reduce pancreatic tissue necroptosis. In vitro experiments of mouse pancreatic organoids and acinar cells also confirmed that UA could improve pancreatic inflammation by regulating the ER-mitochondrial calcium channel and necroptosis pathway proteins. This study not only explored the therapeutic effect of plant EVs on SAP but also revealed that UA could alleviate SAP by regulating ER-mitochondrial calcium channel and reducing acinar cell necroptosis, providing insights into the pathogenesis and potential treatment of SAP.
    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: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)

公众号