Flavanones

黄酮类
  • 文章类型: Journal Article
    近年来,炎症性疾病已成为人类健康的重要问题。通过对抗炎药的持续研究,Alpinetin已显示出有希望的抗炎特性,包括参与表观遗传修饰途径。作为表观遗传修饰的关键调节剂,Mecp2可能在调节Alpinetin的表观遗传效应中发挥作用,可能影响其抗炎特性。为了检验这个假设,两个关键组件,p65(NF-KB家族的成员)和p300(一种共激活剂),通过表达谱微阵列进行筛选,这与小鼠巨噬细胞的LPS刺激强度有很强的相关性。同时,通过破坏p65的合成及其与炎症基因启动子的相互作用,然而它对p300没有表现出类似的影响。此外,Mecp2可以通过附着于Alpetin诱导的甲基化炎症基因启动子来抑制p300的结合,导致启动子乙酰化障碍,随后影响p65的结合,最终增强了alpinetin的抗炎能力。同样,在脓毒症小鼠模型中,观察到过表达Mecp2的纯合子在通过alpetin给药时与杂合子相比显示出更大的器官损伤减少和存活率提高。然而,阻断DNA甲基转移酶3A(DNMT3A)的表达导致Mecp2抗炎辅助作用的丧失。总之,Mecp2可能通过表观遗传的“串扰”来增强Alpinetin的抗炎作用,强调涉及Mecp2和alpinetin的联合治疗策略用于抗炎干预的潜在疗效。
    In recent years, inflammatory disorders have emerged as a significant concern for human health. Through ongoing research on anti-inflammatory agents, alpinetin has shown promising anti-inflammatory properties, including involvement in epigenetic modification pathways. As a crucial regulator of epigenetic modifications, Mecp2 may play a role in modulating the epigenetic effects of alpinetin, potentially impacting its anti-inflammatory properties. To test this hypothesis, two key components, p65 (a member of NF-KB family) and p300 (a type of co-activator), were screened by the expression profiling microarray, which exhibited a strong correlation with the intensity of LPS stimulation in mouse macrophages. Meanwhile, alpinetin demonstrates the anti-inflammatory properties through its ability to disrupt the synthesis of p65 and its interaction with promoters of inflammatory genes, yet it did not exhibit similar effects on p300. Additionally, Mecp2 can inhibit the binding of p300 by attaching to the methylated inflammatory gene promoter induced by alpinetin, leading to obstacles in promoter acetylation and subsequently impacting the binding of p65, ultimately enhancing the anti-inflammatory capabilities of alpinetin. Similarly, in a sepsis mouse model, it was observed that homozygotes overexpressing Mecp2 showed a greater reduction in organ damage and improved survival rates compared to heterozygotes when administered by alpinetin. However, blocking the expression of DNA methyltransferase 3A (DNMT3A) resulted in the loss of Mecp2\'s anti-inflammatory assistance. In conclusion, Mecp2 may augment the anti-inflammatory effects of alpinetin through epigenetic \'crosstalk\', highlighting the potential efficacy of a combined therapeutic strategy involving Mecp2 and alpinetin for anti-inflammatory intervention.
    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,破骨细胞活性受到细胞内pH波动的显著影响。因此,pH敏感的门控纳米药物递送系统代表了减轻破骨细胞过度活性的有希望的治疗方法。我们之前的研究表明,柚皮苷,一种天然类黄酮,有效减轻破骨细胞活性。然而,柚皮苷的口服利用率低,半衰期短,阻碍了其临床应用。我们开发了一种药物递送系统,其中壳聚糖,作为看门人,包覆载有柚皮苷(CS@MSNs-柚皮苷)的介孔二氧化硅纳米颗粒。然而,CS@MSNs-柚皮苷对破骨细胞的抑制作用和潜在机制尚不清楚,保证进一步的研究。
    首先,我们合成了CS@MSNs-柚皮苷,并进行了全面表征。我们还测量了pH梯度溶液中的药物释放速率并验证了其生物安全性。随后,我们研究了CS@MSNs-柚皮苷对骨髓源性巨噬细胞诱导的破骨细胞的影响,在探索潜在机制的同时,重点关注分化和骨吸收活性。最后,我们建立了大鼠双侧临界大小的颅骨缺损模型,其中CS@MSNs-柚皮苷分散在GelMA水凝胶中以实现原位药物递送。我们观察到CS@MSNs-柚皮苷在体内促进骨再生和抑制破骨细胞活性的能力。
    CS@MSNs-柚皮苷表现出高的均匀性和分散性,低细胞毒性(浓度≤120μg/mL),和显著的pH敏感性。体外,与Naringin和MSNs-Naringin相比,CS@MSNs-柚皮苷更有效地抑制破骨细胞的形成和骨吸收活性。这种作用伴随着NF-κB和MAPK信号通路中关键因子的磷酸化减少,细胞凋亡水平增加,以及随后的破骨细胞特异性基因和蛋白质的产生减少。在体内,CS@MSNs-Naringin的表现优于Naringin和MSNs-Naringin,促进新骨形成,同时更大程度地抑制破骨细胞活性。
    我们的研究表明,CS@MSNs-Naringin在体外和体内表现出惊人的抗破骨细胞能力,而且促进颅骨缺损的骨再生。
    UNASSIGNED: It is well-established that osteoclast activity is significantly influenced by fluctuations in intracellular pH. Consequently, a pH-sensitive gated nano-drug delivery system represents a promising therapeutic approach to mitigate osteoclast overactivity. Our prior research indicated that naringin, a natural flavonoid, effectively mitigates osteoclast activity. However, naringin showed low oral availability and short half-life, which hinders its clinical application. We developed a drug delivery system wherein chitosan, as gatekeepers, coats mesoporous silica nanoparticles loaded with naringin (CS@MSNs-Naringin). However, the inhibitory effects of CS@MSNs-Naringin on osteoclasts and the underlying mechanisms remain unclear, warranting further research.
    UNASSIGNED: First, we synthesized CS@MSNs-Naringin and conducted a comprehensive characterization. We also measured drug release rates in a pH gradient solution and verified its biosafety. Subsequently, we investigated the impact of CS@MSNs-Naringin on osteoclasts induced by bone marrow-derived macrophages, focusing on differentiation and bone resorption activity while exploring potential mechanisms. Finally, we established a rat model of bilateral critical-sized calvarial bone defects, in which CS@MSNs-Naringin was dispersed in GelMA hydrogel to achieve in situ drug delivery. We observed the ability of CS@MSNs-Naringin to promote bone regeneration and inhibit osteoclast activity in vivo.
    UNASSIGNED: CS@MSNs-Naringin exhibited high uniformity and dispersity, low cytotoxicity (concentration≤120 μg/mL), and significant pH sensitivity. In vitro, compared to Naringin and MSNs-Naringin, CS@MSNs-Naringin more effectively inhibited the formation and bone resorption activity of osteoclasts. This effect was accompanied by decreased phosphorylation of key factors in the NF-κB and MAPK signaling pathways, increased apoptosis levels, and a subsequent reduction in the production of osteoclast-specific genes and proteins. In vivo, CS@MSNs-Naringin outperformed Naringin and MSNs-Naringin, promoting new bone formation while inhibiting osteoclast activity to a greater extent.
    UNASSIGNED: Our research suggested that CS@MSNs-Naringin exhibited the strikingly ability to anti-osteoclasts in vitro and in vivo, moreover promoted bone regeneration in the calvarial bone defect.
    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
    柚皮素(NAR)是一种突出的黄烷酮,已被认为具有促进人牙周膜干细胞(hPDLSC)成骨分化的能力。本研究旨在探讨NAR如何促进hPDLSCs的成骨分化,并评估其在牙槽骨缺损修复中的功效。为此,通过mRNA测序和网络药理学分析,建立了NAR作用的蛋白质-蛋白质相互作用网络。通过逆转录定量和蛋白质印迹评估基因和蛋白质表达水平。采用茜素红和碱性磷酸酶染色观察hPDLSCs的成骨能力,免疫荧光用于检测NAR分子探针和AKT在细胞中的共定位。通过显微计算机断层扫描(micro-CT)评估下颌骨缺损的修复,Masson染色和免疫荧光。此外,计算机模拟对接软件用于确定NAR与靶蛋白的结合亲和力,AKT.结果表明,一氧化氮(NO)-环磷酸鸟苷(cGMP)-蛋白激酶G(PKG)信号通路的激活可以促进hPDLSCs的成骨分化。抑制AKT,内皮型一氧化氮合酶和可溶性鸟苷酸环化酶分别减弱NAR促进hPDLSCs成骨分化的能力。Micro-CT和Masson染色显示,NAR管饲组在缺损部位表现出更多的新骨形成。免疫荧光分析证实了在NAR灌胃组中,Runt相关转录因子2和骨桥蛋白的表达上调。总之,本研究结果表明,NAR通过与AKT结合激活NO‑cGMP‑PKG信号通路,促进hPDLSCs的成骨分化.
    Naringenin (NAR) is a prominent flavanone that has been recognized for its capacity to promote the osteogenic differentiation of human periodontal ligament stem cells (hPDLSCs). The present study aimed to explore how NAR promotes the osteogenic differentiation of hPDLSCs and to assess its efficacy in repairing alveolar bone defects. For this purpose, a protein‑protein interaction network of NAR action was established by mRNA sequencing and network pharmacological analysis. Gene and protein expression levels were evaluated by reverse transcription‑quantitative and western blotting. Alizarin red and alkaline phosphatase staining were also employed to observe the osteogenic capacity of hPDLSCs, and immunofluorescence was used to examine the co‑localization of NAR molecular probes and AKT in cells. The repair of mandibular defects was assessed by micro‑computed tomography (micro‑CT), Masson staining and immunofluorescence. Additionally, computer simulation docking software was utilized to determine the binding affinity of NAR to the target protein, AKT. The results demonstrated that activation of the nitric oxide (NO)‑cyclic guanosine monophosphate (cGMP)‑protein kinase G (PKG) signaling pathway could promote the osteogenic differentiation of hPDLSCs. Inhibition of AKT, endothelial nitric oxide synthase and soluble guanylate cyclase individually attenuated the ability of NAR to promote the osteogenic differentiation of hPDLSCs. Micro‑CT and Masson staining revealed that the NAR gavage group exhibited more new bone formation at the defect site. Immunofluorescence assays confirmed the upregulated expression of Runt‑related transcription factor 2 and osteopontin in the NAR gavage group. In conclusion, the results of the present study suggested that NAR promotes the osteogenic differentiation of hPDLSCs by activating the NO‑cGMP‑PKG signaling pathway through its binding to AKT.
    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)与高复发和不良预后相关。黄芩苷具有多种药理作用,包括抗炎和抗增殖活性。这里,我们研究了黄芩素对OSCC转移的影响及其潜在的作用机制。
    方法:用不同浓度的黄芩素处理SCC-4和CAL-27细胞。通过甲基噻唑基二苯基-溴化四唑(MTT)测定评估OSCC细胞的增殖。至于迁移和转移,将黄芩素处理的OSCC细胞用于伤口愈合测定和Transwell测定。上皮-间质转化相关蛋白(E-cadherin,N-钙黏着蛋白,波形蛋白)和细胞外调节蛋白激酶(ERK)/ETS转录因子ELK1(ELK-1)/Snail信号通路相关蛋白在黄芩素处理的OSCC细胞中通过蛋白质印迹进行评估。
    结果:细胞增殖和迁移的速率,连同转移潜力,黄芩素处理的细胞显着低于对照组(p<0.05),效果是浓度依赖性的。此外,与对照相比,黄芩素显着降低SCC-4和CAL-27细胞中N-钙黏着蛋白和波形蛋白的水平,并增加E-cadherin水平(p<0.05)。机械上,黄芩素下调p-ERK1/2,磷酸-ETS转录因子ELK1(p-ELK-1)的水平,和蜗牛(p<0.05)。最后,ERK/ELK-1/Snail通路抑制剂(U0126)可促进黄芩素抑制OSCC细胞迁移和侵袭的作用(p<0.05)。
    结论:黄芩素减轻迁移,入侵,OSCC细胞通过ERK/ELK-1/Snail信号通路转移。本研究为开发黄芩素作为治疗OSCC的化合物提供了依据。
    BACKGROUND: Oral squamous cell carcinoma (OSCC) is associated with high recurrence and poor prognosis. Baicalin has multiple pharmacological effects, including anti-inflammatory and anti-proliferative activities. Here, we examine the effect of baicalein on OSCC metastasis and its potential mechanism of action.
    METHODS: SCC-4 and CAL-27 cells were treated with different concentrations of baicalein. The proliferation of OSCC cells was evaluated by Methylthiazolyldiphenyl-tetrazolium bromide (MTT) assay. As for migration and metastasis, baicalein-treated OSCC cells were used for wound healing assay and Transwell assay. The levels of epithelial-mesenchymal transition-related proteins (E-cadherin, N-cadherin, vimentin) and extracellular regulated protein kinases (ERK)/ETS Transcription Factor ELK1 (ELK-1)/Snail signaling pathway-related proteins in baicalein-treated OSCC cells were evaluated by western blotting.
    RESULTS: The rates of cell proliferation and migration, along with the metastatic potential, of baicalein-treated cells were significantly lower than those of the control (p < 0.05), and the effects were concentration-dependent. Furthermore, compared to the control, baicalein significantly decreased the levels of N-cadherin and vimentin in SCC-4 and CAL-27 cells, and increased the E-cadherin level (p < 0.05). Mechanistically, baicalein downregulated the levels of p-ERK1/2, phospho-ETS Transcription Factor ELK1 (p-ELK-1), and Snail (p < 0.05). Finally, the ERK/ELK-1/Snail pathway inhibitor (U0126) promoted the effect of baicalein in inhibiting the migration and invasion of OSCC cells (p < 0.05).
    CONCLUSIONS: Baicalein abates the migration, invasion, and metastasis of OSCC cells through the ERK/ELK-1/Snail signaling pathway. This study provides a basis for the development of baicalein as a compound for the 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
    骨髓炎是一种侵入性骨感染,可导致严重疼痛甚至残疾,给骨科手术带来挑战。柚皮苷可以减轻骨相关的炎症。本研究旨在阐明柚皮苷在金黄色葡萄球菌诱导的小鼠骨髓炎模型中的作用及机制。在给予柚皮苷后收集金黄色葡萄球菌感染的小鼠的股骨,并进行显微计算机断层扫描以分析皮质骨破坏和骨丢失。还评估了股骨中的细菌生长。使用酶联免疫吸附测定法测量小鼠股骨中的促炎细胞因子水平。采用苏木精和伊红染色和抗酒石酸酸性磷酸酶染色分析病理变化和骨吸收,分别。定量逆转录聚合酶链反应和蛋白质印迹分析用于定量股骨成骨分化相关基因的信使RNA和蛋白质表达。使用细胞计数试剂盒-8测定人骨髓来源的干细胞(hBMSC)的活力。进行茜素红S染色和碱性磷酸酶染色以评估矿化结节的形成和体外骨形成。使用蛋白质印迹分析评估股骨组织和hBMSCs中的Notch信号传导相关蛋白水平。实验结果表明,柚皮苷通过增加骨体积/总体积比减轻金黄色葡萄球菌诱导的小鼠皮质骨破坏和骨丢失。柚皮苷抑制金黄色葡萄球菌诱导的股骨细菌生长和炎症。此外,它缓解了组织病理学变化,抑制骨吸收,并增加了骨髓小鼠成骨标志物的表达。它在体外增加了hBMSCs的活力并促进其分化和骨矿化。此外,柚皮苷通过上调模型小鼠股骨和金黄色葡萄球菌刺激的hBMSCs中Notch1,Jagged1和Hes1的蛋白质水平来激活Notch信号。总之,柚皮苷减少细菌生长,炎症,和骨吸收,同时通过激活Notch信号上调金黄色葡萄球菌感染的小鼠和hBMSCs中成骨标志物的表达。
    Osteomyelitis is an invasive bone infection that can lead to severe pain and even disability, posing a challenge for orthopedic surgery. Naringin can reduce bone-related inflammatory conditions. This study aimed to elucidate the function and mechanism of naringin in a Staphylococcus aureus-induced mouse model of osteomyelitis. Femurs of S. aureus-infected mice were collected after naringin administration and subjected to microcomputed tomography to analyze cortical bone destruction and bone loss. Bacterial growth in femurs was also assessed. Proinflammatory cytokine levels in mouse femurs were measured using enzyme-linked immunosorbent assays. Pathological changes and bone resorption were analyzed using hematoxylin and eosin staining and tartrate-resistant acid phosphatase staining, respectively. Quantitative reverse transcription polymerase chain reaction and western blot analysis were used to quantify the messenger RNA and protein expression of osteogenic differentiation-associated genes in the femurs. The viability of human bone marrow-derived stem cells (hBMSCs) was determined using cell counting kit-8. Alizarin Red S staining and alkaline phosphatase staining were performed to assess the formation of mineralization nodules and bone formation in vitro. Notch signaling-related protein levels in femur tissues and hBMSCs were assessed using western blot analysis. Experimental results revealed that naringin alleviated S. aureus-induced cortical bone destruction and bone loss in mice by increasing the bone volume/total volume ratio. Naringin suppressed S. aureus-induced bacterial growth and inflammation in femurs. Moreover, it alleviated histopathological changes, inhibited bone resorption, and increased the expression of osteogenic markers in osteomyelitic mice. It increased the viability of hBMSCs and promoted their differentiation and bone mineralization in vitro. Furthermore, naringin activated Notch signaling by upregulating the protein levels of Notch1, Jagged1, and Hes1 in the femurs of model mice and S. aureus-stimulated hBMSCs. In conclusion, naringin reduces bacterial growth, inflammation, and bone resorption while upregulating the expression of osteogenic markers in S. aureus-infected mice and hBMSCs by activating Notch signaling.
    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
    本研究旨在探讨柚皮苷在氧化应激下促进MC3T3-E1成骨分化的潜能。它探讨了Nar与Wnt/β-catenin和PI3K/Akt信号通路的联系。最初,分析了2911个OP相关基因,揭示与PI3K/Akt和Wnt途径以及氧化应激密切相关。通过各种数据库和分子对接研究确定了Nar的潜在靶标ESR1,HSP90AA1和ESR2,证实了Nar与ESR1和HSP90AA1的亲和力。实验确定了Nar和H2O2的最佳浓度。0.3mmol/LH2O2损伤MC3T3-E1细胞,由0.1μmol/LNar缓解。通过DCFH-DA探针和NO检测证实了氧化应激模型的成功建立。Nar表现出增强成骨分化的能力,抵消氧化损伤。在氧化应激下,它显着增加了MC3T3-E1细胞中成骨细胞相关蛋白的表达。研究发现Nar对GSK-3β磷酸化有积极影响,β-连环蛋白积累,和通路相关蛋白表达,都是促进成骨分化的关键。研究结论:Nar能有效促进MC3T3-E1细胞在氧化应激下的成骨分化。它通过激活Wnt/β-catenin和PI3K/Akt途径实现了这一点,促进GSK-3β磷酸化,并增强β-连环蛋白的积累,在成骨过程中至关重要。
    This study aimed to explore naringin\'s potential to promote the osteogenic differentiation of MC3T3-E1 under oxidative stress. It delved into Nar\'s connection with the Wnt/β-catenin and PI3K/Akt signaling pathways. Initially, 2911 OP-related genes were analyzed, revealing close ties with the PI3K/Akt and Wnt pathways alongside oxidative stress. Nar\'s potential targets-ESR1, HSP90AA1, and ESR2-were identified through various databases and molecular docking studies confirmed Nar\'s affinity with ESR1 and HSP90AA1. Experiments established optimal concentrations for Nar and H2O2. H2O2 at 0.3 mmol/L damaged MC3T3-E1 cells, alleviated by 0.1 µmol/L Nar. Successful establishment of oxidative stress models was confirmed by DCFH-DA probe and NO detection. Nar exhibited the ability to enhance osteogenic differentiation, counteracting oxidative damage. It notably increased osteoblast-related protein expression in MC3T3-E1 cells under oxidative stress. The study found Nar\'s positive influence on GSK-3β phosphorylation, β-catenin accumulation, and pathway-related protein expression, all critical in promoting osteogenic differentiation. The research concluded that Nar effectively promotes osteogenic differentiation in MC3T3-E1 cells under oxidative stress. It achieved this by activating the Wnt/β-catenin and PI3K/Akt pathways, facilitating GSK-3β phosphorylation, and enhancing β-catenin accumulation, pivotal in osteogenesis.
    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:尽管已在细胞过程中广泛研究了AMP激活的蛋白激酶(AMPK),对其底物的理解,下游功能,对细胞命运和结直肠癌(CRC)进展的贡献仍然不完整。
    目的:本研究旨在探讨柚皮素对CRC的作用及其机制。
    方法:在CRC中评估柚皮素的生物学和细胞特性及其抗癌活性。此外,评估了柚皮素和5-氟尿嘧啶联合治疗对体外和体内肿瘤生长的影响。
    结果:本研究发现柚皮素抑制CRC的增殖并促进其凋亡。与柚皮素组相比,柚皮素联合5-氟尿嘧啶对抑制细胞增殖和促进细胞凋亡有显著作用。结果表明柚皮素激活CRC中的AMPK磷酸化和线粒体融合。柚皮素联合5-氟尿嘧啶可显着降低5-氟尿嘧啶对皮下CRC肿瘤裸鼠的心脏毒性和肝损伤,并减轻氧化偶氮甲烷/DSS硫酸葡聚糖(AOM/DSS)诱导的CRC中的结直肠损伤。这两种药物的组合通过增加活性氧(ROS)水平和降低线粒体膜电位(MMP)来改变线粒体功能。从而刺激AMPK/mTOR信号传导。线粒体动力学因此通过激活AMPK/p-AMPK途径来调节,和线粒体稳态是通过增加线粒体融合和减少裂变来协调的,以激活癌细胞的凋亡。
    结论:我们的数据表明柚皮素对抑制CRC增殖很重要,可能是通过AMPK途径,在CRC中调节线粒体功能并诱导细胞凋亡。
    BACKGROUND: Although AMP-activated protein kinase (AMPK) has been extensively studied in cellular processes, the understanding of its substrates, downstream functions, contributions to cell fate and colorectal cancer (CRC) progression remains incomplete.
    OBJECTIVE: The aim of this study was to investigate the effects and mechanisms of naringenin on CRC.
    METHODS: The biological and cellular properties of naringenin and its anticancer activity were evaluated in CRC. In addition, the effect of combined treatment with naringenin and 5-fluorouracil on tumor growth in vitro and in vivo was evaluated.
    RESULTS: The present study found that naringenin inhibits the proliferation of CRC and promote its apoptosis. Compared with the naringenin group, naringenin combined with 5-fluorouracil had significant effect on inhibiting cell proliferation and promoting its apoptosis. It is showed that naringenin activates AMPK phosphorylation and mitochondrial fusion in CRC. Naringenin combined with 5-fluorouracil significantly reduces cardiotoxicity and liver damage induced by 5-fluorouracil in nude mice bearing subcutaneous CRC tumors, and attenuates colorectal injuries in azoxymethane/DSS dextran sulfate (AOM/DSS)-induced CRC. The combination of these two drugs alters mitochondrial function by increasing reactive oxygen species (ROS) levels and decreasing the mitochondrial membrane potential (MMP), thereby stimulating AMPK/mTOR signaling. Mitochondrial dynamics are thereby regulated by activating the AMPK/p-AMPK pathway, and mitochondrial homeostasis is coordinated through increased mitochondrial fusion and reduced fission to activate apoptosis in cancer cells.
    CONCLUSIONS: Our data suggest that naringenin is important for inhibiting CRC proliferation, possibly through the AMPK pathway, to regulate mitochondrial function and induce apoptosis in CRC.
    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:本研究旨在探讨肝爽颗粒(GSG)对二甲基亚硝胺(DMN)诱导的大鼠肝纤维化的保护作用及其机制。
    方法:通过DMN给药实验诱发大鼠肝纤维化,并采用不同剂量的GSG作为干预措施。通过测量血清转氨酶和胆红素水平来评估肝细胞损伤,伴随着肝组织的组织病理学检查。通过酶联免疫吸附测定(ELISA)定量血小板衍生生长因子(PDGF)和转化生长因子-β1(TGF-β1)的肝脏浓度。使用免疫组织化学技术评估肝组织中α-平滑肌肌动蛋白(α-SMA)的表达。肝干扰素-γ(IFN-γ)的水平,肿瘤坏死因子-α(TNF-α),和白介素(IL-2,IL-4,IL-6,IL-10)的光谱通过定量实时PCR(qRT-PCR)进行定量。此外,肝星状细胞(HSC)在体外培养,并暴露于TNF-α在柚皮苷的存在下,GSG的主要组成部分。也通过qRT-PCR定量这些细胞中金属蛋白酶-1(TIMP-1)和基质金属肽酶-1(MMP-1)的组织抑制剂的基因表达水平。通过细胞计数试剂盒-8测定评价HSC的增殖活性。最后,通过Western印迹分析Smad蛋白表达的改变。
    结果:在纤维化大鼠中施用GSG导致血清转氨酶和胆红素水平降低,随着组织病理学肝损伤的减轻。此外,用GSG处理的纤维化大鼠表现出肝TGF-β1,PDGF,和TNF-α水平。此外,GSG治疗导致IFN-γ的mRNA水平增加,IL-2和IL-4以及α-SMA在肝脏中的表达降低。此外,用柚皮苷治疗,GSG的关键提取物,与对照组相比,TNF-α刺激的HSCs中MMP-1的表达升高,TIMP-1的水平降低。此外,柚皮苷给药导致HSC内Smad表达减少。
    结论:GSG具有通过调节炎症和纤维化因子减轻DMN诱导的大鼠模型纤维化的潜力。值得注意的是,柚皮苷,GSG的主要提取物,可能在调节TGF-β-Smad信号通路中起关键作用。
    OBJECTIVE: The present study aims to investigate the specific protective effects and underlying mechanisms of Ganshuang granule (GSG) on dimethylnitrosamine (DMN)-induced hepatic fibrosis in rat models.
    METHODS: Hepatic fibrosis was experimentally evoked in rats by DMN administration, and varying dosages of GSG were employed as an intervention. Hepatocellular damage was assessed by measuring serum levels of aminotransferase and bilirubin, accompanied by histopathological examinations of hepatic tissue. The hepatic concentrations of platelet-derived growth factor (PDGF) and transforming growth factor-β1 (TGF-β1) were quantitated via enzyme-linked immunosorbent assay (ELISA). The expression of α-smooth muscle actin (α-SMA) within hepatic tissue was evaluated using immunohistochemical techniques. The levels of hepatic interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and a spectrum of interleukins (IL-2, IL-4, IL-6, IL-10) were quantified by quantitative real-time PCR (qRT-PCR). Additionally, hepatic stellate cells (HSCs) were cultured in vitro and exposed to TNF-α in the presence of naringin, a principal component of GSG. The gene expression levels of tissue inhibitor of metalloproteinase-1 (TIMP-1) and matrix metallopeptidase-1 (MMP-1) in these cells were also quantified by qRT-PCR. Proliferative activity of HSCs was evaluated by the Cell Counting Kit-8 assay. Finally, alterations in Smad protein expression were analyzed through Western blotting.
    RESULTS: Administration of GSG in rats with fibrosis resulted in reduced levels of serum aminotransferases and bilirubin, along with alleviation of histopathological liver injury. Furthermore, the fibrosis rats treated with GSG exhibited significant downregulation of hepatic TGF-β1, PDGF, and TNF-α levels. Additionally, GSG treatment led to increased mRNA levels of IFN-γ, IL-2, and IL-4, as well as decreased expression of α-SMA in the liver. Furthermore, treatment with naringin, a pivotal extract of GSG, resulted in elevated expression of MMP-1 and decreased levels of TIMP-1 in TNF-α-stimulated HSCs when compared to the control group. Additionally, naringin administration led to a reduction in Smad expression within the HSCs.
    CONCLUSIONS: GSG has the potential to mitigate fibrosis induced by DMN in rat models through the regulation of inflammatory and fibrosis factors. Notably, naringin, the primary extract of GSG, may exert a pivotal role in modulating the TGF-β-Smad 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
    问题,如放化疗的毒副作用和耐药性,靶向治疗和免疫疗法伴随着当前的抗癌治疗,已经成为限制患者临床获益的瓶颈。因此,迫切需要找到疗效更高,副作用更小的有希望的抗癌策略。黄芩素,从中药黄芩中提取的黄酮类成分,已广泛研究了其在分子和细胞水平上对多种类型恶性肿瘤的显着抗癌活性。黄芩素通过抑制血管生成发挥抗肿瘤作用,入侵和迁移,诱导细胞凋亡和细胞周期阻滞,以及调节细胞自噬,新陈代谢,肿瘤微环境和肿瘤干细胞无明显毒副作用。经典信号通路的作用,如PI3K/AKT/mTOR,MAPK,AMPK,Wnt/β-catenin,JAK/STAT3,MMP-2/-9已被强调为黄芩素发挥其抗恶性潜力的主要靶标。此外,黄芩素可以调节相关的非编码RNA,比如lncRNAs,miRNAs和circ-RNAs,抑制肿瘤的发生和发展.除了上述的共性,黄芩素在某些特定的癌症类型中显示出一些特定的抗肿瘤特性。此外,黄芩素联合放化疗的临床前研究为黄芩素作为放化疗的辅助治疗方法的发展铺平了道路。我们的目的是在体外和体内实验的基础上总结黄芩素在不同类型癌症中的作用及其机制。希望为今后黄芩素作为一种有效、安全的临床治疗癌症的化合物提供证据。
    Problems, such as toxic side effects and drug resistance of chemoradiotherapy, target therapy and immunotherapy accompanying the current anti-cancer treatments, have become bottlenecks limiting the clinical benefit for patients. Therefore, it is urgent to find promising anti-cancer strategies with higher efficacy and lesser side effects. Baicalein, a flavonoid component derived from the Chinese medicine scutellaria baicalensis, has been widely studied for its remarkable anti-cancer activity in multiple types of malignancies both at the molecular and cellular levels. Baicalein exerts its anti-tumor effects by inhibiting angiogenesis, invasion and migration, inducing cell apoptosis and cell cycle arrest, as well as regulating cell autophagy, metabolism, the tumor microenvironment and cancer stem cells with no obvious toxic side effects. The role of classic signaling pathways, such as PI3K/AKT/mTOR, MAPK, AMPK, Wnt/β-catenin, JAK/STAT3, MMP-2/-9, have been highlighted as the major targets for baicalein exerting its anti-malignant potential. Besides, baicalein can regulate the relevant non-coding RNAs, such as lncRNAs, miRNAs and circ-RNAs, to inhibit tumorigenesis and progression. In addition to the mentioned commonalities, baicalein shows some specific anti-tumor characteristics in some specific cancer types. Moreover, the preclinical studies of the combination of baicalein and chemoradiotherapy pave the way ahead for developing baicalein as an adjunct treatment with chemoradiotherapy. Our aim is to summary the role of baicalein in different types of cancer with its mechanisms based on in vitro and in vivo experiments, hoping providing proof for baicalein serving as an effective and safe compound for cancer treatment in clinic in the future.
    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:糖尿病肾病(DN)是一种危及生命的肾脏疾病,需要紧急治疗。Wogonin在DN中具有肾脏保护作用。本研究旨在探讨汉黄芩素调控高糖诱导的肾细胞损伤的机制。
    方法:糖尿病小鼠(db/db),对照db/m小鼠,和正常葡萄糖(NG)-或HG处理的人小管上皮细胞(HK-2)用于评估细胞因子信号抑制因子3(SOCS3)的水平,Toll样受体4(TLR4),炎症和纤维化。慢病毒用于调节SOCS3和TLR4的表达。在db/db小鼠中口服wogonin(10mg/kg)或媒介物后,组织学形态,血糖,尿蛋白,血清肌酐值(Scr),血尿素氮(BUN),超氧化物歧化酶(SOD),谷胱甘肽(GSH),和活性氧(ROS)进行了评估。RT-qPCR和Western印迹评估炎症和纤维化相关分子。
    结果:HG暴露导致高血糖,严重的肾损伤,高血清Src和BUN,低SOD和GSH,增加ROS。HG下调SOCS3,但上调TLR4和JAK/STAT,纤维化,和与炎症相关的蛋白质.Wogonin通过降低细胞因子减轻HG诱导的肾损伤,ROS,Src,和MDA和增加SOD和GSH。同时,Wogonin在HG条件下上调SOCS3并下调TLR4。Wogonin诱导的SOCS3过表达直接降低TLR4水平,减轻JAK/STAT信号通路相关的炎症和纤维化,但SOCS3敲除显著拮抗汉黄芩素的保护作用。然而,TLR4敲除减少SOCS3敲除诱导的肾损伤。
    结论:Wogonin通过上调SOCS3抑制TLR4和JAK/STAT通路减轻肾脏炎症和纤维化。
    BACKGROUND: Diabetic nephropathy (DN) is a life-threatening renal disease and needs urgent therapies. Wogonin is renoprotective in DN. This study aimed to explore the mechanism of how wogonin regulated high glucose (HG)-induced renal cell injury.
    METHODS: Diabetic mice (db/db), control db/m mice, and normal glucose (NG)- or HG-treated human tubule epithelial cells (HK-2) were used to evaluate the levels of suppressor of cytokine signaling 3 (SOCS3), Toll-like receptor 4 (TLR4), inflammation and fibrosis. Lentivirus was used to regulate SOCS3 and TLR4 expressions. After oral gavage of wogonin (10 mg/kg) or vehicle in db/db mice, histological morphologies, blood glucose, urinary protein, serum creatinine values (Scr), blood urea nitrogen (BUN), superoxide dismutase (SOD), glutathione (GSH), and reactive oxygen species (ROS) were assessed. RT-qPCR and Western blot evaluated inflammation and fibrosis-related molecules.
    RESULTS: HG exposure induced high blood glucose, severe renal injuries, high serumal Src and BUN, low SOD and GSH, and increased ROS. HG downregulated SOCS3 but upregulated TLR4 and JAK/STAT, fibrosis, and inflammasome-related proteins. Wogonin alleviated HG-induced renal injuries by decreasing cytokines, ROS, Src, and MDA and increasing SOD and GSH. Meanwhile, wogonin upregulated SOCS3 and downregulated TLR4 under HG conditions. Wogonin-induced SOCS3 overexpression directly decreased TLR4 levels and attenuated JAK/STAT signaling pathway-related inflammation and fibrosis, but SOCS3 knockdown significantly antagonized the protective effects of wogonin. However, TLR4 knockdown diminished SOCS3 knockdown-induced renal injuries.
    CONCLUSIONS: Wogonin attenuates renal inflammation and fibrosis by upregulating SOCS3 to inhibit TLR4 and JAK/STAT pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号