CITCO, 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime

CI TCO,6 - (4 - 氯苯基) 咪唑并 [2, 1 - b] [1, 3] 噻唑 - 5 - 甲醛 O - (3, 4 - 二氯苄基) 肟
  • 文章类型: Journal Article
    众所周知,外源性生物敏感核受体孕烷X受体(PXR)和组成型雄甾烷受体(CAR)的药理激活可增加药物代谢并减少炎症。关于它们在肠道微生物组中的生理功能知之甚少。在这项研究中,我们发现了使用基因工程小鼠调节肠道微生物组丰富度的PXR/CAR的二价功能。PXR或CAR的缺乏增加了微生物的丰富度,两种受体的缺失协同增加了微生物的丰富度。PXR和CAR缺乏增加了促炎细菌螺旋杆菌科和螺杆菌。PXR和CAR的缺乏增加了乳酸菌的相对丰度,具有胆盐水解酶活性,对应于粪便中初级牛磺酸结合胆汁酸(BAs)的减少,这可能导致更高的牛磺酸和未结合的BAs的内部负担,两者都与炎症有关,氧化应激,和细胞毒性。PXR/CAR对肠道微生物组的基础作用不同于这些受体的药理和毒理学激活。确定了常见的PXR/CAR靶向细菌,其中大部分被这些受体抑制。与野生型小鼠相比,hPXR-TG小鼠具有不同的微生物谱。这项研究首次揭示了PXR和CAR对肠道微生物组的基础功能。
    Pharmacological activation of the xenobiotic-sensing nuclear receptors pregnane X receptor (PXR) and constitutive androstane receptor (CAR) is well-known to increase drug metabolism and reduce inflammation. Little is known regarding their physiological functions on the gut microbiome. In this study, we discovered bivalent hormetic functions of PXR/CAR modulating the richness of the gut microbiome using genetically engineered mice. The absence of PXR or CAR increased microbial richness, and absence of both receptors synergistically increased microbial richness. PXR and CAR deficiency increased the pro-inflammatory bacteria Helicobacteraceae and Helicobacter. Deficiency in both PXR and CAR increased the relative abundance of Lactobacillus, which has bile salt hydrolase activity, corresponding to decreased primary taurine-conjugated bile acids (BAs) in feces, which may lead to higher internal burden of taurine and unconjugated BAs, both of which are linked to inflammation, oxidative stress, and cytotoxicity. The basal effect of PXR/CAR on the gut microbiome was distinct from pharmacological and toxicological activation of these receptors. Common PXR/CAR-targeted bacteria were identified, the majority of which were suppressed by these receptors. hPXR-TG mice had a distinct microbial profile as compared to wild-type mice. This study is the first to unveil the basal functions of PXR and CAR on the gut microbiome.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    组成型雄甾烷受体(CAR,NR3I1)属于核受体超家族。据报道,CAR激动剂TCPOBOP诱导肝肿大,但潜在的机制仍然很大程度上未知。Yes相关蛋白(YAP)是器官大小的有效调节剂。本研究的目的是探讨YAP在CAR激活诱导的肝肿大和肝再生中的作用。在野生型(WT)小鼠中评估TCPOBOP诱导的CAR活化对肝肿大和肝再生的影响,肝脏特异性YAP缺陷小鼠,和部分肝切除术(PHx)小鼠。结果表明,TCPOBOP可以增加野生型小鼠和PHx小鼠的肝体重比。观察到中央静脉(CV)区域周围的肝细胞增大,同时,门静脉(PV)区域周围的KI67细胞数量增加证明了肝细胞的增殖。在TCPOBOP处理的小鼠中,YAP及其下游靶标的蛋白质水平上调,并且可以通过CAR激活诱导YAP易位。免疫共沉淀结果表明了CAR和YAP的潜在蛋白质-蛋白质相互作用。然而,在肝脏特异性YAP缺陷(Yap-/-)小鼠中仍然可以观察到CAR活化诱导的肝肿大。总之,CAR激活部分通过诱导YAP易位和与YAP信号通路相互作用促进肝肿大和肝再生,这为进一步理解CAR的生理功能提供了新的见解。
    The constitutive androstane receptor (CAR, NR3I1) belongs to nuclear receptor superfamily. It was reported that CAR agonist TCPOBOP induces hepatomegaly but the underlying mechanism remains largely unknown. Yes-associated protein (YAP) is a potent regulator of organ size. The aim of this study is to explore the role of YAP in CAR activation-induced hepatomegaly and liver regeneration. TCPOBOP-induced CAR activation on hepatomegaly and liver regeneration was evaluated in wild-type (WT) mice, liver-specific YAP-deficient mice, and partial hepatectomy (PHx) mice. The results demonstrate that TCPOBOP can increase the liver-to-body weight ratio in wild-type mice and PHx mice. Hepatocytes enlargement around central vein (CV) area was observed, meanwhile hepatocytes proliferation was promoted as evidenced by the increased number of KI67+ cells around portal vein (PV) area. The protein levels of YAP and its downstream targets were upregulated in TCPOBOP-treated mice and YAP translocation can be induced by CAR activation. Co-immunoprecipitation results suggested a potential protein-protein interaction of CAR and YAP. However, CAR activation-induced hepatomegaly can still be observed in liver-specific YAP-deficient (Yap -/-) mice. In summary, CAR activation promotes hepatomegaly and liver regeneration partially by inducing YAP translocation and interaction with YAP signaling pathway, which provides new insights to further understand the physiological functions of CAR.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号