OST, organic solute transporter

  • 文章类型: Journal Article
    未经批准:有机溶质转运蛋白(OST)亚基OSTα和OSTβ促进胆汁酸从肠细胞流出到门静脉循环。OSTα或OSTβ缺乏的患者在胆汁酸吸收不良水平方面表现出相当大的差异,慢性腹泻,和胆汁淤积的迹象。在这里,我们产生并表征了OSTβ缺乏的小鼠模型。
    未经证实:使用CRISR/Cas9产生Ostβ-/-小鼠,并与野生型和Ostα-/-小鼠进行比较。使用腺相关病毒血清型8载体在Ostβ-/-小鼠的肝脏中重新表达OSTβ。在两种模型中都通过胆管结扎(BDL)或3.5-二乙氧基羰基-1.4-二氢可利丁(DDC)喂养诱导胆汁淤积。
    未经授权:类似于Ostα-/-小鼠,Ostβ-/-小鼠表现出细长的小肠,绒毛变钝,隐窝深度增加。Ostβ-/-小鼠中回肠Fgf15表达水平的增加和Asbt表达的降低表明胆汁酸在肠上皮细胞中的积累。与Ostα-/-小鼠相比,通过BDL或DDC饮食诱导Ostβ-/-小鼠胆汁淤积,导致存活率降低和体重严重下降,而是肝脏表型的改善.通过腺相关病毒介导的过表达恢复肝脏Ostβ表达并不能挽救Ostβ-/-小鼠的表型。
    未经证实:OSTβ是回肠胆汁酸转运的关键,其缺乏导致类似于Ostα-/-小鼠的肠道表型,但它对生存和肝脏表型有不同的影响,独立于其在肝脏中的表达。我们的发现为OSTα和OSTβ缺陷患者的可变临床表现提供了见解。
    未经证实:有机溶质转运蛋白(OST)亚基OSTα和OSTβ共同促进结合胆汁酸流入门静脉循环。Ostα敲除小鼠的小肠更长,更厚,并且在很大程度上可以防止实验性胆汁淤积性肝损伤。在这里,我们首次产生并表征了Ostβ基因敲除小鼠。Ostα和Ostβ敲除小鼠在正常条件下具有相似的表型。然而,在胆汁淤积中,OSTβ基因敲除小鼠具有恶化的整体表型,这表明OSTβ的单独和特定作用,可能是其他肠道蛋白质的相互作用伙伴。
    UNASSIGNED: Organic solute transporter (OST) subunits OSTα and OSTβ facilitate bile acid efflux from the enterocyte into the portal circulation. Patients with deficiency of OSTα or OSTβ display considerable variation in the level of bile acid malabsorption, chronic diarrhea, and signs of cholestasis. Herein, we generated and characterized a mouse model of OSTβ deficiency.
    UNASSIGNED: Ostβ -/- mice were generated using CRISR/Cas9 and compared to wild-type and Ostα -/- mice. OSTβ was re-expressed in livers of Ostβ -/- mice using adeno-associated virus serotype 8 vectors. Cholestasis was induced in both models by bile duct ligation (BDL) or 3.5-diethoxycarbonyl-1.4-dihydrocollidine (DDC) feeding.
    UNASSIGNED: Similar to Ostα -/- mice, Ostβ -/- mice exhibited elongated small intestines with blunted villi and increased crypt depth. Increased expression levels of ileal Fgf15, and decreased Asbt expression in Ostβ -/- mice indicate the accumulation of bile acids in the enterocyte. In contrast to Ostα -/- mice, induction of cholestasis in Ostβ -/- mice by BDL or DDC diet led to lower survival rates and severe body weight loss, but an improved liver phenotype. Restoration of hepatic Ostβ expression via adeno-associated virus-mediated overexpression did not rescue the phenotype of Ostβ -/- mice.
    UNASSIGNED: OSTβ is pivotal for bile acid transport in the ileum and its deficiency leads to an intestinal phenotype similar to Ostα -/- mice, but it exerts distinct effects on survival and the liver phenotype, independent of its expression in the liver. Our findings provide insights into the variable clinical presentation of patients with OSTα and OSTβ deficiencies.
    UNASSIGNED: Organic solute transporter (OST) subunits OSTα and OSTβ together facilitate the efflux of conjugated bile acids into the portal circulation. Ostα knockout mice have longer and thicker small intestines and are largely protected against experimental cholestatic liver injury. Herein, we generated and characterized Ostβ knockout mice for the first time. Ostα and Ostβ knockout mice shared a similar phenotype under normal conditions. However, in cholestasis, Ostβ knockout mice had a worsened overall phenotype which indicates a separate and specific role of OSTβ, possibly as an interacting partner of other intestinal proteins.
    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
    慢性肝病(CLD)是许多国家残疾调整寿命的主要原因之一。最近对核胆汁酸受体途径的理解越来越关注肠道之间串扰的影响,胆汁酸,和肝脏对肝脏病理的影响。虽然通常用于胆汁淤积症和溶解胆结石,胆汁酸对肠道微生物组和人体代谢的影响的发现为其在早期和晚期肝病中的应用提供了独特的潜力,因为其病因多样。基于这些发现,使用基于胆汁酸的分子的临床前研究在解决肝脏炎症和纤维化方面显示出令人鼓舞的结果。新出现的数据还表明,胆汁酸谱在肝病的各种原因中具有明显的变化。我们总结了与胆汁酸在健康和疾病相关的当前知识和证据,并讨论了胆汁酸衍生物在CLD中的最终和正在进行的治疗试验。在不久的将来,这方面的进一步证据可能有助于临床医生更好地发现和管理肝脏疾病.
    Chronic liver disease (CLD) is one of the leading causes of disability-adjusted life years in many countries. A recent understanding of nuclear bile acid receptor pathways has increased focus on the impact of crosstalk between the gut, bile acids, and liver on liver pathology. While conventionally used in cholestatic disorders and to dissolve gallstones, the discovery of bile acids\' influence on the gut microbiome and human metabolism offers a unique potential for their utility in early and advanced liver diseases because of diverse etiologies. Based on these findings, preclinical studies using bile acid-based molecules have shown encouraging results at addressing liver inflammation and fibrosis. Emerging data also suggest that bile acid profiles change distinctively across various causes of liver disease. We summarize the current knowledge and evidence related to bile acids in health and disease and discuss culminated and ongoing therapeutic trials of bile acid derivatives in CLD. In the near future, further evidence in this area might help clinicians better detect and manage liver diseases.
    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
    通过FXR和TGR5信令,胆汁酸(BAs)调节脂质和葡萄糖代谢,炎症和纤维化。因此,肠道分泌后,BAs返回肝脏,修饰和重吸收可能与非酒精性脂肪性肝炎(NASH)的发病机制有关。在这里,我们在NASH的临床前模型中表征了BAs的肠肝谱和信号传导,并探讨了BA成分实验操作的后果。
    我们使用高脂饮食(HFD)喂养的foz/foz和高果糖西方饮食喂养的C57BL/6J小鼠,并将它们与各自的控件进行比较。小鼠接受补充有脱氧胆酸(DCA)的饮食以调节BA组成。
    与对照组相比,NASH小鼠的门静脉血液和胆汁中的BAs浓度较低,而全身BA浓度没有显著改变。值得注意的是,次级BAs的浓度,尤其是DCA,在NASH小鼠的胆汁和门静脉血中,继发性与原发性BA的比例显着降低。因此,门静脉血中FXR和TGR5配体含量低,并在NASH小鼠中赋予较差的抗炎保护。NASH肝脏中增强的一级BA合成和二级BA向一级BA的转化有助于二级BA的消耗。HFD喂养的foz/foz小鼠的膳食DCA补充恢复了门静脉血液中的BA浓度,增加TGR5和FXR信号,改善了代谢异常状态,免受脂肪变性和肝细胞膨胀,减少巨噬细胞浸润。
    肝肠循环中的BA组成,但不是在全身循环中,在NASH的临床前模型中发生了深刻的变化,在二级BA中具有特定的消耗。从NASH保护的BA配置文件的饮食校正,支持肠肝性BA在NASH发病机制中的作用。
    这项研究清楚地表明,在相关的临床前模型中,肝肠胆汁酸的改变极大地促进了非酒精性脂肪性肝炎的发展。的确,胆汁酸组成的实验调节恢复了受干扰的FXR和TGR5信号传导,并预防了非酒精性脂肪性肝炎和相关的代谢紊乱。
    UNASSIGNED: Through FXR and TGR5 signaling, bile acids (BAs) modulate lipid and glucose metabolism, inflammation and fibrosis. Hence, BAs returning to the liver after enteric secretion, modification and reabsorption may contribute to the pathogenesis of non-alcoholic steatohepatitis (NASH). Herein, we characterized the enterohepatic profile and signaling of BAs in preclinical models of NASH, and explored the consequences of experimental manipulation of BA composition.
    UNASSIGNED: We used high-fat diet (HFD)-fed foz/foz and high-fructose western diet-fed C57BL/6J mice, and compared them to their respective controls. Mice received a diet supplemented with deoxycholic acid (DCA) to modulate BA composition.
    UNASSIGNED: Compared to controls, mice with NASH had lower concentrations of BAs in their portal blood and bile, while systemic BA concentrations were not significantly altered. Notably, the concentrations of secondary BAs, and especially of DCA, and the ratio of secondary to primary BAs were strikingly lower in bile and portal blood of mice with NASH. Hence, portal blood was poor in FXR and TGR5 ligands, and conferred poor anti-inflammatory protection in mice with NASH. Enhanced primary BAs synthesis and conversion of secondary to primary BAs in NASH livers contributed to the depletion in secondary BAs. Dietary DCA supplementation in HFD-fed foz/foz mice restored the BA concentrations in portal blood, increased TGR5 and FXR signaling, improved the dysmetabolic status, protected from steatosis and hepatocellular ballooning, and reduced macrophage infiltration.
    UNASSIGNED: BA composition in the enterohepatic cycle, but not in systemic circulation, is profoundly altered in preclinical models of NASH, with specific depletion in secondary BAs. Dietary correction of the BA profile protected from NASH, supporting a role for enterohepatic BAs in the pathogenesis of NASH.
    UNASSIGNED: This study clearly demonstrates that the alterations of enterohepatic bile acids significantly contribute to the development of non-alcoholic steatohepatitis in relevant preclinical models. Indeed, experimental modulation of bile acid composition restored perturbed FXR and TGR5 signaling and prevented non-alcoholic steatohepatitis and associated metabolic disorders.
    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
    回肠胆汁酸吸收是通过顶端钠依赖性胆汁酸转运蛋白(ASBT)的摄取介导的,并通过基底外侧异聚有机溶质转运蛋白α-β(OSTα-OSTβ)输出。在这项研究中,我们研究了肠细胞胆汁酸淤滞对Ostα-/-小鼠的细胞毒性作用,包括肠损伤与胆汁酸肝肠循环启动之间的时间关系。
    回肠组织形态计量学,组织学,细胞增殖的标志物,基因,在出生后第5、10、15、20和30天,在雄性和雌性野生型和Ostα-/-小鼠中分析蛋白质表达。产生并分析Ostα-/-Asbt-/-小鼠。在果蝇中研究了肠道Nrf2激活途径的胆汁酸激活。
    早在第5天,Ostα-/-小鼠每长度回肠重量显着增加,绒毛高度降低,上皮细胞增殖增加。这与新生Ostα-/-小鼠中Asbt的过早表达和胆汁酸激活的法尼醇X受体靶基因的诱导有关。在这些出生后时间点,新生Ostα-/-小鼠中烟酰胺腺嘌呤二核苷酸磷酸氧化酶-1和Nrf2抗氧化反应基因的表达显着增加。胆汁酸还激活果蝇肠细胞中的Nrf2,并且Nrf2的肠细胞特异性敲除增加了果蝇对胆汁酸诱导的毒性的敏感性。Asbt的失活阻止了Ostα-/-小鼠回肠形态的变化和抗氧化反应基因的诱导。
    出生后发育早期,Ostα的丢失导致胆汁酸积累,氧化应激,和回肠的恢复原状反应。除了其在维持胆汁酸稳态方面的重要作用外,Ostα-Ostβ保护回肠上皮免受胆汁酸诱导的损伤。NCBI基因表达综合:GSE99579。
    OBJECTIVE: Ileal bile acid absorption is mediated by uptake via the apical sodium-dependent bile acid transporter (ASBT), and export via the basolateral heteromeric organic solute transporter α-β (OSTα-OSTβ). In this study, we investigated the cytotoxic effects of enterocyte bile acid stasis in Ostα-/- mice, including the temporal relationship between intestinal injury and initiation of the enterohepatic circulation of bile acids.
    METHODS: Ileal tissue morphometry, histology, markers of cell proliferation, gene, and protein expression were analyzed in male and female wild-type and Ostα-/- mice at postnatal days 5, 10, 15, 20, and 30. Ostα-/-Asbt-/- mice were generated and analyzed. Bile acid activation of intestinal Nrf2-activated pathways was investigated in Drosophila.
    RESULTS: As early as day 5, Ostα-/- mice showed significantly increased ileal weight per length, decreased villus height, and increased epithelial cell proliferation. This correlated with premature expression of the Asbt and induction of bile acid-activated farnesoid X receptor target genes in neonatal Ostα-/- mice. Expression of reduced nicotinamide adenine dinucleotide phosphate oxidase-1 and Nrf2-anti-oxidant responsive genes were increased significantly in neonatal Ostα-/- mice at these postnatal time points. Bile acids also activated Nrf2 in Drosophila enterocytes and enterocyte-specific knockdown of Nrf2 increased sensitivity of flies to bile acid-induced toxicity. Inactivation of the Asbt prevented the changes in ileal morphology and induction of anti-oxidant response genes in Ostα-/- mice.
    CONCLUSIONS: Early in postnatal development, loss of Ostα leads to bile acid accumulation, oxidative stress, and a restitution response in ileum. In addition to its essential role in maintaining bile acid homeostasis, Ostα-Ostβ functions to protect the ileal epithelium against bile acid-induced injury. NCBI Gene Expression Omnibus: GSE99579.
    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
    胆汁酸的经典功能包括充当洗涤剂以促进肠内营养的消化和吸收。此外,胆汁酸也作为信号分子调节葡萄糖稳态,脂质代谢和能量消耗。在诸如体循环的区室中胆汁酸的信号传导潜力部分地由有效的肠肝循环调节,该肠肝循环起到保存和引导肠和肝胆区室中的胆汁酸库的作用。肝胆和肠胆汁酸转运的变化可以改变成分,尺寸,和胆汁酸库的分布。这些改变反过来会对胆汁酸信号及其下游代谢靶标产生显著影响。这篇综述讨论了我们对胆汁酸的肠肝循环与通过胆汁酸激活受体的信号传导的代谢后果之间的相互关系的理解的最新进展。例如法尼醇X核受体(FXR)和G蛋白偶联胆汁酸受体(TGR5)。
    The classical functions of bile acids include acting as detergents to facilitate the digestion and absorption of nutrients in the gut. In addition, bile acids also act as signaling molecules to regulate glucose homeostasis, lipid metabolism and energy expenditure. The signaling potential of bile acids in compartments such as the systemic circulation is regulated in part by an efficient enterohepatic circulation that functions to conserve and channel the pool of bile acids within the intestinal and hepatobiliary compartments. Changes in hepatobiliary and intestinal bile acid transport can alter the composition, size, and distribution of the bile acid pool. These alterations in turn can have significant effects on bile acid signaling and their downstream metabolic targets. This review discusses recent advances in our understanding of the inter-relationship between the enterohepatic cycling of bile acids and the metabolic consequences of signaling via bile acid-activated receptors, such as farnesoid X nuclear receptor (FXR) and the G-protein-coupled bile acid receptor (TGR5).
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    这篇综述着重于胆汁酸信号传导与胆管细胞有关的各种成分。还探讨了它们作为潜在治疗胆管疾病的靶标的作用。虽然许多因素参与这些复杂的信号通路,本文综述了跨膜G蛋白偶联受体(TGR5)的作用,法尼醇X受体(FXR),熊去氧胆酸(UDCA)和碳酸氢盐伞。根据胆管细胞和胆汁酸的一般背景,我们将扩大本综述,包括最近(5-7年内)关于胆汁酸信号和胆管细胞功能领域的部分.这些发现都表明胆汁酸会影响胆道功能,反过来,调节病理事件期间的胆管细胞反应。
    This review focuses on various components of bile acid signaling in relation to cholangiocytes. Their roles as targets for potential therapies for cholangiopathies are also explored. While many factors are involved in these complex signaling pathways, this review emphasizes the roles of transmembrane G protein coupled receptor (TGR5), farnesoid X receptor (FXR), ursodeoxycholic acid (UDCA) and the bicarbonate umbrella. Following a general background on cholangiocytes and bile acids, we will expand the review and include sections that are most recently known (within 5-7 years) regarding the field of bile acid signaling and cholangiocyte function. These findings all demonstrate that bile acids influence biliary functions which can, in turn, regulate the cholangiocyte response during pathological events.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号