urolithin A

尿石素 A
  • 文章类型: Journal Article
    尿石素A(UA)是来源于鞣花酸的肠道代谢产物。这项系统评价评估了UA对人类的潜在保护作用。在包括250名健康个体的五项研究中,UA(10-1000mg/天),持续时间为28天至4个月,显示剂量依赖性抗炎作用,并上调一些线粒体基因,自噬的标志物,和脂肪酸氧化。它不影响线粒体最大三磷酸腺苷的产生,生物发生,动力学,或肠道菌群组成。UA增加肌肉力量和耐力,然而,对人体测量没有影响,心血管结果,和身体功能。不相关的不良事件为轻度或中度。需要对更多的生理系统和更长的干预期进行进一步的研究。
    Urolithin A (UA) is a gut metabolite derived from ellagic acid. This systematic review assesses the potential geroprotective effect of UA in humans. In five studies including 250 healthy individuals, UA (10 - 1000mg/day) for a duration ranging from 28 days to 4 months, showed a dose-dependent anti-inflammatory effect and upregulated some mitochondrial genes, markers of autophagy, and fatty acid oxidation. It did not affect mitochondrial maximal adenosine triphosphate production, biogenesis, dynamics, or gut microbiota composition. UA increased muscle strength and endurance, however, had no effect on anthropometrics, cardiovascular outcomes, and physical function. Unrelated adverse events were mild or moderate. Further research across more physiological systems and longer intervention periods is required.
    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
    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
    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
    健康长寿医学旨在通过针对整个生命周期的衰老过程来优化健康。解决加速衰老涉及适应生活方式和使用老年保护药物和补充剂,包括营养补充剂和生物活性化合物。食品和药物管理局,根据《膳食补充剂健康和教育法》,将生物活性化合物和医药产品归类为膳食补充剂。虽然许多公司出售可以被认为是老虎机的成分,他们的质量控制监督有限。政府安全当局只核实违禁化合物的存在,不是标签上列出的成分的准确性。这里,烟酰胺单核苷酸和尿石素A补充剂,易于在线或在药店访问,进行了活性成分含量测试。结果显示与标记量有显著偏差,范围从+28.6%到-100%。这表明老年保护补充剂的质量存在相当大的差异。为了解决这种可变性,代表医疗保健专业人员的社会之间和内部的合作,行业和监管机构必须确保老年保护补充剂的质量。
    Healthy Longevity Medicine aims to optimize health by targeting aging processes across the lifespan. Addressing accelerated aging involves adaptation of lifestyle and the use of geroprotective drugs and supplements, including nutritional supplements and bioactive compounds. The Food and Drug Administration, under the Dietary Supplement Health and Education Act, categorizes bioactive compounds and medicinal products as dietary supplements. While numerous companies sell ingredients that can be deemed geroprotectors, there\'s limited oversight in their quality control. Governmental safety authorities only verify the presence of prohibited compounds, not the accuracy of ingredients listed on labels.Here, Nicotinamide mononucleotide and Urolithin A supplements, easily accessible online or in pharmacies, were tested for their active ingredient content. Results showed a significant deviation from the labeled amounts, ranging from + 28.6% to -100%. This indicates a considerable disparity in the quality of geroprotective supplements.To address this variability, collaboration between and within societies representing healthcare professionals, industry and regulatory bodies is imperative to ensure the quality of geroprotective supplements.
    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: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
    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
    背景:口腔鳞状细胞癌(OSCC)是世界上最常见的恶性肿瘤,死亡率惊人。尽管在治疗策略和药物开发方面取得了进展,总体生存率仍然很低。因此,必须开发具有最小脱靶效应的替代或免费抗癌药物。尿磷脂A,由人类肠道微生物组内源性产生的鞣花酸和鞣花单宁的微生物代谢产物被认为具有抗癌活性。然而,尿石素A在OSCC中的抗肿瘤作用尚未阐明。在这项研究中,我们研究了尿石素A是否在OSCC细胞系中抑制细胞生长并诱导细胞凋亡和自噬依赖性细胞死亡。
    目的:本研究旨在评估尿石素A在体外和体内小鼠模型中抑制OSCC的潜力及其对OSCC增殖和侵袭的调节作用。
    方法:我们在体外和体内OSCC小鼠模型中评估尿石素A是否可以诱导细胞死亡。
    结果:对尿石素A处理的OSCC细胞系的流式细胞术和免疫印迹分析显示尿石素A通过诱导内质网应激和随后的AKT和mTOR信号传导的抑制显著诱导OSCC的细胞死亡,如磷酸化mTOR和4EBP1水平降低所证明的。这进一步揭示了凋亡和自噬信号通路之间可能的串扰。体内研究表明,尿石素A治疗可减小肿瘤大小,并显示mTOR减少,ERK1/2和Akt水平随着增殖标志物的降低,Ki67.一起来看,体外以及我们的体内数据表明,尿石素A是一种潜在的抗癌剂,而AKT/mTOR/ERK信号的抑制对于尿石素A诱导的口腔癌生长抑制至关重要.
    结论:尿石素A通过诱导OSCC细胞凋亡和自噬途径发挥其抗肿瘤活性。我们的发现表明,尿石素A通过诱导内质网应激和随后的AKT和mTOR信号传导的抑制显着诱导口腔鳞状细胞癌的细胞死亡,磷酸化mTOR和4EBP1水平降低证明了这一点。尿石素A在体外和体内小鼠模型中均显着抑制肿瘤生长,表明其作为预防和治疗OSCC的抗癌剂的潜力。从今以后,我们的研究结果为尿石素A在预防和治疗OSCC方面的治疗潜力提供了新的见解.
    BACKGROUND: Oral squamous cell carcinoma (OSCC) is the most prevalent malignancy in the world with an alarming rate of mortality. Despite the advancement in treatment strategies and drug developments, the overall survival rate remains poor. Therefore, it is imperative to develop alternative or complimentary anti cancer drugs with minimum off target effects. Urolithin A, a microbial metabolite of ellagic acid and ellagitannins produced endogenously by human gut micro biome is considered to have anti-cancerous activity. However anti tumorigenic effect of urolithin A in OSCC is yet to be elucidated. In this study, we examined whether urolithin A inhibits cell growth and induces both apoptosis and autophagy dependent cell death in OSCC cell lines.
    OBJECTIVE: The present study aims to evaluate the potential of urolithin A to inhibit OSCC and its regulatory effect on OSCC proliferation and invasion in vitro and in vivo mouse models.
    METHODS: We evaluated whether urolithin A could induce cell death in OSCC in vitro and in vivo mouse models.
    RESULTS: Flow cytometric and immunoblot analysis on Urolithin A treated OSCC cell lines revealed that urolithin A markedly induced cell death of OSCC via the induction of endoplasmic reticulum stress and subsequent inhibition of AKT and mTOR signaling as evidenced by decreased levels of phosphorylated mTOR and 4EBP1. This further revealed a possible cross talk between apoptotic and autophagic signaling pathways. In vivo study demonstrated that urolithin A treatment reduced tumor size and showed a decrease in mTOR, ERK1/2 and Akt levels along with a decrease in proliferation marker, Ki67. Taken together, in vitro as well as our in vivo data indicates that urolithin A is a potential anticancer agent and the inhibition of AKT/mTOR/ERK signalling is crucial in Urolithin A induced growth suppression in oral cancer.
    CONCLUSIONS: Urolithin A exerts its anti tumorigenic activity through the induction of apoptotic and autophagy pathways in OSCC. Our findings suggest that urolithin A markedly induced cell death of oral squamous cell carcinoma via the induction of endoplasmic reticulum stress and subsequent inhibition of AKT and mTOR signaling as evidenced by decreased levels of phosphorylated mTOR and 4EBP1. Urolithin A remarkably suppressed tumor growth in both in vitro and in vivo mouse models signifying its potential as an anticancer agent in the prevention and treatment of OSCC. Henceforth, our findings provide a new insight into the therapeutic potential of urolithin A in the prevention and treatment of OSCC.
    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
    NOD样受体家族含pyrin结构域的蛋白3(NLRP3)炎性体触发白介素1β(IL-1β)的成熟,并参与各种炎性疾病的发病机理。尿磷脂A,鞣花酸的肠道微生物代谢产物,据报道在体外和体内发挥抗炎作用。然而,尿石素A是否抑制NLRP3炎性体激活尚不清楚.在这项研究中,尿石素A抑制NLRP3炎性体激动剂诱导的caspase-1的裂解,IL-1β的成熟,和激活脂多糖引发的小鼠骨髓源性巨噬细胞的焦亡。尿石素A减少了细胞内和线粒体活性氧的产生,并限制了硫氧还蛋白相互作用蛋白与NLRP3之间的相互作用,从而减弱了NLRP3炎性体的激活。尿石素A给药可预防尿酸单钠诱导的小鼠腹膜炎。总的来说,这些发现表明尿石素A抑制NLRP3炎性体激活,至少部分地,通过抑制细胞内和线粒体活性氧的产生。
    The NOD-like receptor family pyrin domain-containing protein 3 (NLRP3) inflammasome triggers the maturation of interleukin-1β (IL-1β) and is implicated in the pathogenesis of various inflammatory diseases. Urolithin A, a gut microbial metabolite of ellagic acid, reportedly exerts antiinflammatory effects in vitro and in vivo. However, whether urolithin A suppresses NLRP3 inflammasome activation is unclear. In this study, urolithin A inhibited the cleavage of NLRP3 inflammasome agonist-induced caspase-1, maturation of IL-1β, and activation of pyroptosis in lipopolysaccharide-primed mouse bone marrow-derived macrophages. Urolithin A reduced generation of intracellular and mitochondrial reactive oxygen species and restricted the interaction between thioredoxin-interacting protein and NLRP3, which attenuated NLRP3 inflammasome activation. Urolithin A administration prevented monosodium urate-induced peritonitis in mice. Collectively, these findings indicate that urolithin A suppresses NLRP3 inflammasome activation, at least partially, by repressing the generation of intracellular and mitochondrial reactive oxygen species.
    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
    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

    求助全文

公众号