cholestasis

胆汁淤积
  • 文章类型: Journal Article
    最近的研究报道,在胆管结扎(BDL)大鼠中,AQP8和AQP9的肝表达下调,而人AQP1在大鼠肝脏中的过表达减轻了胆汁淤积。然而,AQP10在人胆汁淤积中的表达及其调控机制尚不清楚。
    收集34例梗阻性胆汁淤积患者和12例对照患者的血清和肝脏样本。将八周大的雄性C57BL/6J小鼠静脉注射编码由肝细胞特异性Alb启动子(AAV8-Alb启动子-hAQP10)驱动的人AQP10的腺相关病毒8(AAV8),以进行功能研究。AQP10启动子和PLC/PRF/5-ASBT细胞系的构建体用于调节机制研究。
    AQP10在梗阻性胆汁淤积患者中显著下调,与血清总胆汁酸(TBA)水平呈负相关。肝细胞特异性过表达hAQP10可显着减轻BDL小鼠的胆汁淤积性肝损伤和肝内胆汁酸(BA)积累。共轭BA,如TCA和炎症因子TNFα,显著抑制AQP10表达。此外,NFκBp65/p50直接与AQP10启动子结合,并降低其在PLC/RPF/5-ASBT细胞和阻塞性胆汁淤积患者肝脏中的活性。然而,BAY11-7082(NFκB信号的特异性抑制剂)减少了这些变化.
    我们是第一个报道在阻塞性胆汁淤积患者中AQP10明显下降的报告。AQP10过表达可显着减轻BDL小鼠胆汁淤积性肝损伤。因此,hAQP10在肝脏中的过度表达可能是一个有价值的胆汁淤积干预策略。
    UNASSIGNED: Recent studies reported that the hepatic expression of AQP8 and AQP9 was downregulated in bile duct-ligated (BDL) rats and that overexpression of human AQP1 in the rat liver attenuated cholestasis. However, the hepatic expression of AQP10 and its regulatory mechanism in human cholestasis remain unclear.
    UNASSIGNED: Serum and liver samples were collected from 34 patients with obstructive cholestasis and from 12 control patients. Eight-week-old male C57BL/6J mice were intravenously injected with an adeno-associated virus 8 (AAV8) encoding human AQP10 driven by a hepatocyte-specific Alb promotor (AAV8-Alb promotor-hAQP10) for functional studies. Constructs of the AQP10 promoter and PLC/PRF/5-ASBT cell lines were used for regulatory mechanism studies.
    UNASSIGNED: AQP10 was significantly downregulated in patients with obstructive cholestasis and negatively associated with the serum levels of total bile acid (TBA). The hepatocyte-specific overexpression of hAQP10 significantly attenuated the cholestatic liver injury and intrahepatic bile acids (BA) accumulation in BDL mice. Conjugated BAs, such as TCA and inflammatory factor TNFα, significantly repressed AQP10 expression. Furthermore, NFκB p65/p50 directly bound to the AQP10 promotor and decreased its activity in PLC/RPF/5-ASBT cells and in the livers of patients with obstructive cholestasis. However, these changes were diminished by BAY 11-7082 (a specific inhibitor of NFκB signaling).
    UNASSIGNED: We are the first to report that AQP10 was significantly decreased in patients with obstructive cholestasis. AQP10 overexpression significantly attenuated cholestatic liver injury in BDL mice. Therefore, overexpression of hAQP10 in the liver may be a valuable strategy for cholestasis 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
    有机阴离子转运多肽(OATPs)在胆汁酸和胆红素的转运中起着至关重要的作用。在我们之前的研究中,白细胞介素6(IL-6)降低胆汁淤积性疾病中的OATP1B3水平。然而,目前尚不清楚IL-6是否抑制胆汁淤积性疾病中OATP1B1的表达.本研究旨在研究IL-6能否抑制OATP1B1的表达并探讨其作用机制。
    使用RT-qPCR和酶联免疫吸附测定在胆汁淤积小鼠模型中研究了干扰素基因刺激因子(STING)信号传导对炎症因子的影响。探讨炎症因子对肝细胞癌中OATP1B1表达的影响,我们用TNF-α处理PLC/PRF/5细胞后,通过RT-qPCR和WesternBlot分析OATP1B1的表达,IL-1β,IL-6为了阐明IL-6抑制OATP1B1表达的机制,我们使用RT-qPCR和Western印迹检测了OATP1B1调节因子TCF4在PLC/PRF/5和HepG2细胞中的表达。通过siRNA转染敲低β-catenin/TCF4,研究了β-catenin/TCF4与OATP1B1之间的相互作用机制。
    STING抑制剂降低胆汁淤积小鼠模型的炎症因子水平,IL-6对OATP1B1表现出最有效的抑制作用。IL-6下调β-catenin/TCF4,导致OATP1B1表达降低。敲低β-连环蛋白/TCF4抵消了β-连环蛋白/TCF4介导的OATP1B1抑制。
    STING介导的IL-6上调可抑制OATP1B1,导致OATP1B1对胆汁酸和胆红素的转运减少。这可能有助于改变患有与IL-6产生增加相关的疾病的患者的药代动力学。
    UNASSIGNED: Organic anion-transporting polypeptides (OATPs) play a crucial role in the transport of bile acids and bilirubin. In our previous study, interleukin 6 (IL-6) reduced OATP1B3 levels in cholestatic disease. However, it remains unclear whether IL-6 inhibits OATP1B1 expression in cholestatic diseases. This study aimed to investigate whether IL-6 can inhibit OATP1B1 expression and explore the underlying mechanisms.
    UNASSIGNED: The effect of stimulator of interferon genes (STING) signaling on inflammatory factors was investigated in a cholestatic mouse model using RT-qPCR and enzyme-linked immunosorbent assay. To assess the impact of inflammatory factors on OATP1B1 expression in hepatocellular carcinoma, we analyzed OATP1B1 expression by RT-qPCR and Western Blot after treating PLC/PRF/5 cells with TNF-α, IL-1β, and IL-6. To elucidate the mechanism by which IL-6 inhibits OATP1B1 expression, we examined the expression of the OATP1B1 regulator TCF4 in PLC/PRF/5 and HepG2 cells using RT-qPCR and Western Blot. The interaction mechanism between β-catenin/TCF4 and OATP1B1 was investigated by knocking down β-catenin/TCF4 through siRNA transfection.
    UNASSIGNED: The STING inhibitor decreased inflammatory factor levels in the cholestatic mouse model, with IL-6 exhibiting the most potent inhibitory effect on OATP1B1. IL-6 downregulated β-catenin/TCF4, leading to decreased OATP1B1 expression. Knocking-down β-catenin/TCF4 counteracted the β-catenin/TCF4-mediated repression of OATP1B1.
    UNASSIGNED: STING-mediated IL-6 up-regulation may inhibit OATP1B1, leading to reduced transport of bile acids and bilirubin by OATP1B1. This may contribute to altered pharmacokinetics in patients with diseases associated with increased IL-6 production.
    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
    暂无摘要。
    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:胆汁淤积性肝病引起局部和全身高凝,中性粒细胞胞外陷阱(NET)作为主要驱动因素。这些NETs与阻塞性黄疸患者肝功能下降有关。然而,NETs对胆汁淤积性肝病肝脏高凝的影响尚不清楚.
    方法:我们利用胆管结扎来创建实验小鼠,并分析肝脏中的NETs形成。纤维蛋白沉积,组织因子表达,通过蛋白质印迹和免疫组织化学技术观察肝脏的炎症。LSEC与分离的NET孵育,我们使用凝血蛋白产生测定法和测量内皮通透性来检测内皮促凝血活性。在体内和体外环境中,DNaseI用于阐明NETs对肝内高凝状态的影响,肝毒性,LSEC,和巨噬细胞活化或损伤。
    结果:胆管结扎小鼠肝组织中NETs水平显著升高,伴有中性粒细胞浸润,组织坏死,纤维蛋白沉积,和血栓形成倾向与假小鼠相比。值得注意的是,NET导致LSEC上的磷脂酰丝氨酸和组织因子暴露,增强凝血因子Xa和凝血酶的产生。增强的促凝血活性可以通过用DNaseI降解NETs来逆转。NET诱导的LSEC渗透率变化,以VE-钙粘蛋白表达和F-肌动蛋白回缩增加为特征,这可以通过DNaseI来拯救。同时,NET的形成与KC活化和炎症因子的形成有关。
    结论:NET促进肝内凝血和炎症的激活,导致肝组织损伤。针对NET形成的策略可能为治疗胆汁淤积性肝病提供潜在的治疗方法。
    BACKGROUND: Cholestatic liver diseases induce local and systemic hypercoagulation, with neutrophil extracellular traps (NETs) serving as major drivers. These NETs have been linked to decreased liver function in patients with obstructive jaundice. However, the impact of NETs on liver hypercoagulation in cholestatic liver disease remains unknown.
    METHODS: We utilized bile duct ligation to create experimental mice and analyzed NETs formation in the liver. Fibrin deposition, tissue factor expression, and inflammation in the liver were visualized through western blot and immunohistochemical techniques. LSECs were incubated with isolated NETs, and we detected endothelial procoagulant activity using coagulation protein production assays and measuring endothelial permeability. In both in vivo and in vitro settings, DNase I was applied to clarify the effect of NETs on intrahepatic hypercoagulability, hepatotoxicity, LSEC, and macrophage activation or injury.
    RESULTS: Bile duct ligation mice exhibited significantly increased levels of NETs in liver tissue, accompanied by neutrophil infiltration, tissue necrosis, fibrin deposition, and thrombophilia compared to sham mice. Notably, NETs resulted in phosphatidylserine and tissue factor exposure on LSEC, enhancing coagulation Factor Xa and thrombin production. The enhanced procoagulant activity could be reversed by degrading NETs with DNase I. Additionally, NETs-induced permeability changes in LSECs, characterized by increased VE-cadherin expression and F-actin retraction, which could be rescued by DNase I. Meanwhile, NET formation is associated with KC activation and the formation of inflammatory factors.
    CONCLUSIONS: NETs promote intrahepatic activation of coagulation and inflammation, leading to liver tissue injury. Strategies targeting NET formation may offer a potential therapeutic approach for treating cholestatic liver disease.
    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
    长期服用抗结核药物会引起药物性胆汁淤积性肝损伤。利福平导致的肝损伤可能与胆汁酸核受体法尼醇X受体(FXR)有关。为了调查这一点,通过连续7天的灌胃给药利福平(200mg/kg),在野生型(C57BL/6N)小鼠和FXR基因敲除(FXR-null)小鼠中均可诱导胆汁淤积。与C57BL/6N小鼠比拟,FXR-null小鼠在利福平给药后表现出更严重的肝损伤,以肝脏尺寸增大为特征,转氨酶升高,增加炎症。此外,在利福平治疗下,FXR敲除会损害脂质分泌并加剧肝脏脂肪变性。重要的是,代谢分子BSEP的表达增加,C57BL/6N小鼠服用利福平后NTCP和CYP7A1下降,而这些变化在FXR敲除小鼠中不存在。此外,利福平治疗C57BL/6N和FXR-null小鼠与c-JunN末端激酶磷酸化(p-JNK)水平升高有关,在FXR无效小鼠中具有更明显的升高。我们的研究表明,利福平引起的肝损伤,脂肪变性,胆汁淤积与FXR功能障碍和胆汁酸代谢改变有关,并且JNK信号通路部分参与了这种损伤。基于这些结果,我们认为FXR可能是解决药物性肝损伤的新治疗靶点。
    Antituberculosis drugs induce pharmacologic cholestatic liver injury with long-term administration. Liver injury resulting from rifampicin is potentially related to the bile acid nuclear receptor Farnesoid X Receptor (FXR). To investigate this, cholestasis was induced in both wild-type (C57BL/6N) mice and FXR knockout (FXR-null) mice through administration of rifampicin (200 mg/kg) via gavage for 7 consecutive days. Compared with C57BL/6N mice, FXR-null mice exhibited more severe liver injury after rifampicin administration, characterized by enlarged liver size, elevated transaminases, and increased inflammation. Moreover, under rifampicin treatment, FXR knockout impairs lipid secretion and exacerbates hepatic steatosis. Significantly, the expression of metabolism molecules BSEP increased, while NTCP and CYP7A1 decreased following rifampicin administration in C57BL/6N mice, whereas these changes were absent in FXR knockout mice. Furthermore, rifampicin treatment in both C57BL/6N and FXR-null mice was associated with elevated c-Jun N-terminal kinase phosphorylation (p-JNK) levels, with a more pronounced elevation in FXR-null mice. Our study suggests that rifampicin-induced liver injury, steatosis, and cholestasis are associated with FXR dysfunction and altered bile acid metabolism, and that the JNK signaling pathway is partially implicated in this injury. Based on these results, we propose that FXR might be a novel therapeutic target for addressing drug-induced liver injury.
    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:胆汁淤积导致严重的肝损伤和随后的肝纤维化。然而,对肝纤维化与胆汁淤积引起的肠道和纤维化肝组织以及肠道微生物群代谢产物变化之间的关系的全面了解是不够的。
    方法:采用胆总管结扎法(BDL)建立26天小鼠胆汁淤积性肝纤维化模型。收集纤维化肝组织和肠内容物。进行非靶向代谢组学以测定肠内容物和肝组织中的代谢物。采用宏基因组学来探索肠道微生物群。
    结果:正常和胆汁淤积性肝纤维化小鼠的肠道内容物和肝组织中的代谢物高度不同。β-丙氨酸代谢和谷胱甘肽代谢在BDL组的肠道中下调。半乳糖代谢,不饱和脂肪酸的生物合成,ABC转运蛋白在BDL组的肠道中上调,在肝脏中下调。精氨酸生物合成,牛磺酸和下牛磺酸代谢,精氨酸和脯氨酸代谢,BDL组的肠道和初级胆汁酸生物合成下调,肝脏上调。宏基因组分析显示,BDL组微生物群的α多样性降低。BDL组中肠道菌群结构的改变导致重要代谢途径的功能减退(例如叶酸生物合成,组氨酸代谢,硫胺素代谢,生物素代谢,和苯丙氨酸,酪氨酸和色氨酸生物合成)和酶(如NADH,DNA解旋酶,和DNA指导的DNA聚合酶)。相关分析表明,某些肠道微生物与肠道和肝脏代谢产物有关。
    结论:非靶向代谢组学和宏基因组学提供了胆汁淤积性肝纤维化小鼠肠道和肝脏代谢以及肠道菌群的全面信息。因此,显著改变的细菌和代谢物可能有助于在未来提供一些抗胆汁淤积性肝纤维化的靶点。
    OBJECTIVE: Cholestasis induces severe liver injury and subsequent liver fibrosis. However, a comprehensive understanding of the relationships between liver fibrosis and cholestasis-induced changes in metabolites in the gut and fibrotic liver tissue and in the gut microbiota is insufficient.
    METHODS: Common bile duct ligation (BDL) was employed to establish a cholestatic liver fibrosis model in mice for 26 days. Fibrotic liver tissue and the gut contents were collected. Untargeted metabolomics was conducted for the determination of metabolites in the gut contents and liver tissues. Metagenomics was adopted to explore the gut microbiota.
    RESULTS: The metabolites in the gut contents and liver tissues between normal and cholestatic liver fibrosis mice were highly distinct. Beta-alanine metabolism and glutathione metabolism were downregulated in the gut of the BDL group. Galactose metabolism, biosynthesis of unsaturated fatty acids, and ABC transporters were upregulated in the gut and downregulated in the liver of the BDL group. Arginine biosynthesis, taurine and hypotaurine metabolism, arginine and proline metabolism, and primary bile acid biosynthesis were downregulated in the gut and upregulated in the liver of the BDL group. Metagenomic analysis revealed that the alpha diversity of the microbiota in the BDL group decreased. The altered structure of the gut microbiota in the BDL group led to the hypofunction of important metabolic pathways (such as folate biosynthesis, histidine metabolism, thiamine metabolism, biotin metabolism, and phenylalanine, tyrosine and tryptophan biosynthesis) and enzymes (such as NADH, DNA helicase, and DNA-directed DNA polymerase). Correlation analyses indicated that certain gut microbes were associated with gut and liver metabolites.
    CONCLUSIONS: Untargeted metabolomics and metagenomics provided comprehensive information on gut and liver metabolism and gut microbiota in mice with cholestatic liver fibrosis. Therefore, significantly altered bacteria and metabolites may help provide some targets against cholestatic liver fibrosis 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
    成纤维细胞生长因子21(FGF21)和丁酸钠(NaB)对胆汁淤积诱导的肝纤维化的保护作用尚不清楚。本研究旨在探讨FGF21和NaB对胆管结扎(BDL)诱导的肝纤维化的影响。
    野生型(WT)和FGF21敲除(KO)小鼠接受BDL手术14天。通过Masson染色在mRNA或蛋白质水平上的纤维化标志物表达来评估肝纤维化。针对BDL损伤评估WT小鼠中腺病毒介导的FGF21过表达。在施用磷酸盐缓冲盐水或NaB的WT和FGF21KO小鼠中进行BDL手术。使用稳定代谢检测和16SrRNA基因测序评估NaB对能量代谢和肠道微生物群的影响。
    BDL诱导的WT小鼠肝纤维化伴随着FGF21的高诱导。与WT小鼠相比,FGF21KO小鼠表现出BDL诱导的更严重的肝纤维化。FGF21过表达保护BDL诱导的肝纤维化,正如在mRNA和蛋白质水平上α-SMA的降低所证明的那样。NaB给药增强了葡萄糖和能量代谢,并重塑了肠道微生物群。NaB减轻了WT小鼠中BDL诱导的肝纤维化,但在FGF21KO小鼠中加重了这一点。
    FGF21在减轻胆汁淤积引起的肝损伤和纤维化中起关键作用。NaB以FGF21依赖性方式对胆汁淤积具有有益作用。因此,NaB施用可以是用于通过增强FGF21信号传导和调节肠道微生物群治疗胆汁淤积的新型营养疗法。
    UNASSIGNED: The beneficial effects of fibroblast growth factor 21 (FGF21) and sodium butyrate (NaB) on protection against cholestasis-induced liver fibrosis are not well known. This study aimed to explore the effects of FGF21 and NaB on bile duct ligation (BDL)-induced liver fibrosis.
    UNASSIGNED: Wild-type (WT) and FGF21 knockout (KO) mice received BDL surgery for 14 days. Liver fibrosis was assessed by Masson\'s staining for fibrosis marker expressions at the mRNA or protein levels. Adenovirus-mediated FGF21 overexpression in the WT mice was assessed against BDL damage. BDL surgeries were performed in WT and FGF21 KO mice that were administered either phosphate-buffered saline or NaB. The effects of NaB on the energy metabolism and gut microbiota were assessed using stable metabolism detection and 16S rRNA gene sequencing.
    UNASSIGNED: BDL-induced liver fibrosis in the WT mice was accompanied by high induction of FGF21. Compared to the WT mice, the FGF21 KO mice showed more severe liver fibrosis induced by BDL. FGF21 overexpression protected against BDL-induced liver fibrosis, as proved by the decreasing α-SMA at both the mRNA and protein levels. NaB administration enhanced the glucose and energy metabolisms as well as remodeled the gut microbiota. NaB alleviated BDL-induced liver fibrosis in the WT mice but aggravated the same in FGF21 KO mice.
    UNASSIGNED: FGF21 plays a key role in alleviating cholestasis-induced liver damage and fibrosis. NaB has beneficial effects on cholestasis in an FGF21-dependent manner. NaB administration can thus be a novel nutritional therapy for treating cholestasis via boosting FGF21 signaling and regulating the gut microbiota.
    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
    患儿 男,1月11日龄,因发现皮肤黄染1个月余就诊,肝功能示转氨酶升高、胆汁淤积。因患儿经治疗后粪便色浅,肝功能无改善,肝脏穿刺病理示细胆管增生,遂行胆道探查术,但胆道通畅。术后继续给予护肝利胆治疗。患儿3月龄时发现其在黄疸消退后仍存在显著增高的胆汁酸水平,最终行基因检测确诊钠牛磺胆酸共转运多肽缺陷病。.
    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
    大黄素(EMO)具有抗α-萘基异硫氰酸盐(ANIT)诱导的胆汁淤积作用。但其机制尚不清楚。法尼醇X受体(Fxr)是掌握胆汁酸的核受体。最近的研究报道Sirtuin1(Sirt1)可以调节Fxr的活性。本研究的目的是基于Sirt1/Fxr信号通路探讨EMO抗ANIT诱导的肝损伤的机制。在有或没有EMO治疗的情况下使用ANIT诱导的胆汁淤积大鼠。血清生化指标,以及肝脏组织病理学变化进行了检查。Sirt1,Fxr,Shp,检测到Bsep和Mrp2。体外研究了Sirt1、Fxr及其下游相关基因的表达。结果表明,EMO能明显减轻ANIT诱导的大鼠肝损伤,并增加Sirt1、Fxr、Shp,Bsep和Mrp2基因在肝脏中的表达,同时降低了Cyp7a1的表达。EMO显著激活Fxr,而Sirt1抑制剂和Sirt1基因沉默在体外显著降低了Fxr活性。总的来说,正确剂量的EMO对ANIT引起的肝损伤具有保护作用,其机制可能是通过Sirt1激活Fxr,从而调节胆汁酸代谢,减少肝细胞胆汁酸负荷。
    Emodin (EMO) has the effect of anti-cholestasis induced by alpha-naphthylisothiocyanate (ANIT). But its mechanism is still unclear. The farnesoid X receptor (Fxr) is the master bile acid nuclear receptor. Recent studies have reported that Sirtuin 1 (Sirt1) can regulate the activities of Fxr. The purpose of the current study was to investigate the mechanism of EMO against ANIT-induced liver injury based on Sirt1/Fxr signaling pathway. The ANIT-induced cholestatic rats were used with or without EMO treatment. Serum biochemical indicators, as well as liver histopathological changes were examined. The genes expressions of Sirt1, Fxr, Shp, Bsep and Mrp2 were detected. The expressions of Sirt1, Fxr and their downstream related genes were investigated in vitro. The results showed that EMO significantly alleviated ANIT-induced liver injury in rats, and increased Sirt1, Fxr, Shp, Bsep and Mrp2 gene expression in liver, while decreased the expression of Cyp7a1. EMO significantly activated Fxr, while Sirt1 inhibitor and Sirt1 gene silencing significantly reduced Fxr activity in vitro. Collectively, EMO in the right dose has a protective effect on liver injury induced by ANIT, and the mechanism may be through activation of Fxr by Sirt1, thus regulating bile acid metabolism, and reducing bile acid load in hepatocytes.
    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
    胆汁淤积性肝病(CLD)的特征是正常胆汁流量受损,最终导致有毒胆汁酸的过度积累。大多数CLD患者最终进展为肝硬化和肝衰竭,由于缺乏有效的治疗,需要肝移植。最近的研究强调了肠道微生物群-胆汁酸轴通过各种途径在肝纤维化进展中的关键作用。胆汁引流阻塞可引起肠道菌群菌群失调,破坏肠黏膜屏障,导致细菌移位。微生物易位激活免疫应答并促进肝纤维化进展。确定用于调节肠道微生物群-胆汁酸轴的治疗靶标代表了改善或可能逆转CLD中肝纤维化的有希望的策略。本文综述了CLD中肠道微生物群-胆汁酸轴的机制,并强调了潜在的治疗靶点。旨在为创新治疗方法奠定基础。
    Cholestatic liver diseases (CLD) are characterized by impaired normal bile flow, culminating in excessive accumulation of toxic bile acids. The majority of patients with CLD ultimately progress to liver cirrhosis and hepatic failure, necessitating liver transplantation due to the lack of effective treatment. Recent investigations have underscored the pivotal role of the gut microbiota-bile acid axis in the progression of hepatic fibrosis via various pathways. The obstruction of bile drainage can induce gut microbiota dysbiosis and disrupt the intestinal mucosal barrier, leading to bacteria translocation. The microbial translocation activates the immune response and promotes liver fibrosis progression. The identification of therapeutic targets for modulating the gut microbiota-bile acid axis represents a promising strategy to ameliorate or perhaps reverse liver fibrosis in CLD. This review focuses on the mechanisms in the gut microbiota-bile acids axis in CLD and highlights potential therapeutic targets, aiming to lay a foundation for innovative treatment approaches.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号