生物技术通报, 2023, 39(4): 187-200 doi: 10.13560/j.cnki.biotech.bull.1985.2022-0931

综述与专论

胆汁酸生理功能及其与肠道微生物互作研究进展

熊淑琪,

江西农业大学省部共建猪遗传改良与养殖技术国家重点实验室,南昌 330045

Towards the Understanding on the Physiological Functions of Bile Acids and Interactions with Gut Microbiota

XIONG Shu-qi,

State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang 330045

通讯作者: 熊淑琪同时为本文通讯作者

责任编辑: 张婷婷

收稿日期: 2022-07-26  

Received: 2022-07-26  

作者简介 About authors

熊淑琪,博士研究生,研究方向:肠道微生物;E-mail:1262254945@qq.com

摘要

胆汁酸(bile acids, BAs)是由胆固醇代谢产生的,在肝脏中胆固醇转化为初级胆汁酸,进入肠道后又经肠道菌群代谢为次级胆汁酸,并通过肠肝循环在体内维持稳态。胆汁酸分子既有亲水端又有亲脂端的结构,可发挥洗涤剂的作用,有助于溶解和吸收膳食中的脂类及脂溶性维生素等,这也使得胆汁酸具有一定的抗菌特性。胆汁酸与肠道菌群的互作不仅体现在其自身的生理功能上,而且肠道细菌的组成及数量也调控着胆汁酸代谢,并通过激活不同的受体信号调节糖脂代谢、能量代谢以及免疫炎症反应等。本文综述了胆汁酸的代谢过程及其如何通过FXR、TGR5等受体信号调控宿主生理功能等,为后续通过科学合理地调控肠道菌群及其代谢物来维护动物健康、促进畜禽生产提供参考。

关键词: 胆汁酸; 肠道菌群; 生理功能; 互作

Abstract

Bile acids(BAs)are produced from the metabolism of cholesterol. In the liver, cholesterol is converted into primary bile acids,and they are metabolized into secondary bile acids by gut microbiota when they reache the intestinal tract. Their homeostasis are maintained in the body through the enterohepatic circulation. The molecules of bile acids have both hydrophilic and lipophilic ends, which can play the role of detergent and help to dissolve and absorb dietary lipids and lipid-soluble vitamins. This also endows bile acids with certain antibacterial properties. The interaction between bile acids and gut microbiota is not only reflected in their own physiological functions of bile acids, but also in the regulations of the gut microbial composition and abundances in bile acid metabolism. Bile acids regulate glucose, lipid and energy metabolisms, and immune and inflammatory reactions by activating different receptor signal pathways. This article reviews the metabolic process of bile acids, and discusses how it regulates host physiological functions through FXR, TGR5 and other receptor signal pathways. It also provides references for maintaining animal health and promoting livestock and poultry production performance through scientifically and reasonably regulating gut microbiota and its metabolites.

Keywords: bile acids; gut microbiota; physiological function; interaction

PDF (2423KB) 元数据 多维度评价 相关文章 导出 EndNote| Ris| Bibtex  收藏本文

本文引用格式

熊淑琪. 胆汁酸生理功能及其与肠道微生物互作研究进展[J]. 生物技术通报, 2023, 39(4): 187-200 doi:10.13560/j.cnki.biotech.bull.1985.2022-0931

XIONG Shu-qi. Towards the Understanding on the Physiological Functions of Bile Acids and Interactions with Gut Microbiota[J]. Biotechnology Bulletin, 2023, 39(4): 187-200 doi:10.13560/j.cnki.biotech.bull.1985.2022-0931

人和动物体内寄居着大量的微生物,其中大部分为肠道细菌,与生物体的健康和生长密切相关。越来越多的研究表明,肠道菌群及其代谢物在人类代谢性疾病,如肥胖、二型糖尿病、非酒精性脂肪肝病以及心血管疾病的发生发展中起着至关重要的作用[1]。在畜牧生产中,肠道菌群的组成与饲料利用、生长发育、脂肪沉积和免疫健康等密切相关。肠道菌群的组成和功能受到许多因素的影响,包括饮食、年龄、性别、遗传、生活方式、用药和情绪压力等,其中饮食被认为是塑造肠道菌群最重要的驱动因素。

肠道作为最大的消化器官,复杂多样的肠道细菌通过分解膳食纤维、脂肪、蛋白质等营养物质,产生大量的微生物代谢物,这些代谢物在宿主体内作为代谢底物和信号分子发挥作用,调节宿主的健康与免疫,保护宿主免受外来有害物质的侵袭。因此,明晰肠道菌群与其代谢物之间的关系对于解析肠道菌群与宿主之间互作具有重要意义。随着代谢组学的发展,通过非靶向及靶向代谢组学分析,目前已经确定了上百种常见的菌群代谢物,包括短链脂肪酸、胆汁酸、氨基酸、维生素以及吲哚衍生物等。胆汁酸与肠道微生物之间具有密切的双向调控作用,其互作关系为研究肠道菌群调节宿主代谢奠定了基础。胆汁酸是动物体内胆固醇代谢产生的一类物质,因其可促进脂质及脂溶性物质的消化吸收、保护肝脏免受脂肪沉积造成的负担和损伤、抑制细菌等生物特性,在现代畜禽养殖业中的应用也已经越来越广泛。因此,本文将从胆汁酸的生成和代谢过程,与微生物的互作关系、作用方式,及生理功能等方面进行整理和总结,为胆汁酸的开发利用助力畜禽生产提供理论依据。

1 胆汁酸的合成代谢及其与微生物互作

1.1 胆汁酸的合成途径

胆汁酸是肝脏中胆固醇代谢的终产物,其合成过程涉及多种催化酶。首先,肝脏中的胆固醇通过两条不同的途径代谢合成初级胆汁酸,一条是由胆固醇7α-羟化酶(CYP7A1)介导催化的经典途径,生成两种初级胆汁酸:胆酸(CA)和鹅脱氧胆酸(CDCA),这一途径决定了肝脏中胆汁酸的含量;而另一条是由甾醇27-羟化酶(CYP27A1)催化的替代途径,生成CDCA[2]。成人肝脏中主要靠经典途径合成胆汁酸,而新生儿主要通过替代途径合成胆汁酸,直至断奶后,CYP7A1才开始表达,转为经典途径。不同物种间初级胆汁酸的种类不尽相同(表1),在啮齿动物小鼠中主要胆酸为α-鼠胆酸(α-MCA)和β-鼠胆酸(β-MCA),而猪中特殊的初级胆汁酸为猪胆酸(HCA)[3]。这些初级胆汁酸属于游离型胆汁酸,它们在肝脏中会进一步在胆汁酸辅酶A合成酶(BACS)、氨基酸N-乙酰转移酶(BAAT)的作用下与牛磺酸和甘氨酸结合,形成结合型胆汁酸牛磺胆酸(TCA)、牛磺鹅脱氧胆酸(TCDCA)、甘氨胆酸(GCA)和甘氨鹅脱氧胆酸(GCDCA)等,这一过程增强了胆汁酸的亲水性[4]。在健康成人的胆汁酸池中,主要为甘氨酸结合型胆汁酸,其与牛磺酸结合型胆汁酸的比例约为3∶1;而在小鼠中,约95%为牛磺酸结合型胆汁酸,其中TCA约占60%,Tα-MCA及Tβ-MCA约40%。胆汁酸通过肝脏毛细胆管膜的胆盐输出泵(BSEP)和多药耐药相关蛋白2(MRP2)运输到胆汁中[5],胆盐与胆固醇和磷脂形成混合胶束并储存在胆囊内,直到进食后刺激胆囊收缩,将胆汁酸释放到肠道以促进脂质的消化吸收[2]。绝大多数结合型胆汁酸在回肠通过顶端钠离子依赖胆汁酸转运蛋白(ASBT)主动吸收进入小肠上皮细胞,再与回肠胆汁酸结合蛋白(IBABP)结合至肠基底膜,进入到肠道中的胆汁酸有95%会在基底侧膜的有机溶质转运蛋白α/β(OSTα/OSTβ)的作用下分泌入门静脉,随后在有机阴离子转运多肽(OATPs)和NA+/牛磺胆酸共转运多肽(NTCP)的介导下转运到肝细胞中进行加工,再与肝脏中新生成的胆汁酸由BSEP和MRP2分泌到胆汁中,摄食后再排入肠腔,形成肠肝循环[6]。而剩下5%的胆汁酸会在肠道菌群的修饰下经过解耦联和脱羟基作用转化为次级胆汁酸排出。人体内1 d约会发生6-12次这种循环,这也使得胆汁酸可以重复利用以满足人体的生理需要[7]

表1   各物种主要胆汁酸

Table 1  Composition of bile acids in different species

物种Species初级胆汁酸Primary bile acids次级胆汁酸Secondary bile acids
游离型Free结合型Conjugated游离型Free结合型Conjugated
胆酸、鹅脱氧胆酸甘氨胆酸、
甘氨鹅脱氧胆酸
石胆酸、
脱氧胆酸
甘氨石胆酸、
甘氨脱氧胆酸
啮齿动物(鼠)α-鼠胆酸、β-鼠胆酸、胆酸牛磺α/β-鼠胆酸、牛磺胆酸ω-鼠胆酸、
熊脱氧胆酸、石胆酸
牛磺ω-鼠胆酸、
牛磺熊脱氧胆酸、牛磺石胆酸
猪胆酸甘氨/牛磺猪胆酸猪脱氧胆酸甘氨/牛磺猪脱氧胆酸

新窗口打开| 下载CSV


1.2 胆汁酸的微生物代谢

次级胆汁酸是分泌到肠道中的初级胆汁酸在肠道细菌的作用下代谢而成的,这一环节在胆汁酸代谢中十分重要,有实验证明在无菌或抗生素治疗的大鼠中,胆汁酸多样性会显著降低,其中牛磺酸结合的胆汁酸丰度显著增加[8]。次级胆汁酸代谢的过程主要分为两步:(1)解耦联,与甘氨酸或牛磺酸结合后的胆汁酸在胆盐水解酶(BSHs)作用下去结合,这是胆汁酸进行下一步修饰的“网关反应”[9]。2019年,Song等[10]对来自六大洲11个群体的人类肠道细菌BSHs进行分类鉴定,共有591个菌株含BSHs蛋白序列,其分布在12门的117个属中,分别为Actinobacteria、Bacteroidetes、Chlamydiae、Cyanobacteria、Euryarchaeota、Firmicute、Fusobacteri、Planctomycetes、Proteobacteria、Spirochaetes、Synergistetes、Verrucomicrobia。(2)7α-脱羟化,在肠道中,解耦联后的游离胆汁酸在微生物的作用下进一步经7α-脱羟基转化为次级胆汁酸。与初级胆汁酸类似,不同物种间生成的次级胆汁酸也不尽相同(表1)。人体内CA和CDCA分别转化为脱氧胆酸(DCA)和石胆酸(LCA),小鼠α/β-MCA转化为脱氧鼠胆酸(MDCA),猪HCA转化为猪脱氧胆酸(HDCA)[3]。早在1980年,就有研究发现肠道细菌Clostridium scindens VPI 12708 能将CA进行7α-脱羟基化后转化为DCA[11]。Funabashi等[12]对胆酸脱羟基的信号通路进行解析,发现具有核心bai基因簇的菌株具有诱导脱羟基化的作用,这些菌株主要来自BacteroidesClostridiumEscherichiaEubacte-riumLactobacillus等。

肠道细菌对胆汁酸的分子修饰作用还包括差向异构化,这也是丰富肠道胆汁酸多样性的主要过程。需要特别注意的是,熊脱氧胆酸(UDCA)在小鼠中属于初级胆汁酸,但在人体内是由CDCA经过7α/β-异构化后形成的,这种作用可通过Clostridium介导[7]。在小鼠盲肠中分离出了3株菌(一株是Eubacterium lentum,另外两株属于Fusobacterium)可将β-MCA通过6β-羟基氧化和6α-还原异构化为ω-MCA[13],此外Clostridium也可介导这种转化[14]。β-MCA还可以发生6β-异构化和7β-脱羟转化为HDCA,但具体的作用菌株还未阐明[13]。在胆汁酸的C3、C7、C12位羟基上发生氧化、差向异构化形成一些氧代胆汁酸,如来自ActinobacteriaFirmic-utes的多种肠道细菌通过3α羟类固醇脱氢酶(3α-HSDH)的作用将DCA和LCA转化为3-oxoDCA和3-oxoLCA[15]。3-oxoLCA又可在Gordonibacter pame-laeaeEggerthella lentaClostridium citroniaeRumi-nococcus gnavus等细菌的作用下进一步发生3β-羟基氧化代谢为isoLCA,其中Eggerthella lentaRumi-nococcus gnavus可将3-oxoDCA转化为isoDCA[15]。胆汁酸还可能发生5α/β-差向异构化生成allo-BAs,如Li等[16]从11个细菌属(BacillusBacteroidesBifidobacteriumCatenibacteriumCollinsellaEgge-rthellaLachnospiraLactobacillusParabacteroidesPeptoniphilusMediterraneibacter)中发现16种细菌可将3-oxoLCA转化为3-oxoalloLCA。

1.3 胆汁酸对肠道菌群的调节

肠道微生物群有助于初级胆汁酸向次级胆汁酸的生物转化,改变胆汁酸的组成,进而激活不同的受体信号影响宿主。而当一些胆汁酸不耐受的细菌暴露于胆汁酸时,其细胞膜的完整性会受到破坏,从而可抑制细菌的过度生长[17]。也有证据表明胆汁酸可诱导细菌和哺乳动物的DNA损伤[18],这可能也是胆汁淤积造成肝损伤的原因。Bernstein等[19]在人肝癌细胞系HepG2中测定了胆盐脱氧胆酸钠(NaDOC)对细胞应激相关的特异性基因启动子或反应元件激活的影响,证明胆汁酸可激活与DNA损伤、氧化应激、内质网应激和蛋白质错误折叠相关的基因启动子,影响微生物生长。艰难梭菌的孢子萌发可受到一些初级胆汁酸如CDCA的刺激,而次级胆汁酸(LCA、UDCA)对其有抑制作用[20]。对艰难梭菌感染者进行粪菌移植,可改变其微生物群和胆汁酸的组成,恢复一些可将初级胆汁酸转化为次级胆汁酸的微生物,具有一定的治疗效果[21]。胆汁酸还可以通过核受体调节小肠的抗菌性防御,胆汁酸通过激活FXR诱导参与肠保护的基因表达,有助于抑制细菌过度生长和回肠黏膜损伤[22]。在培养的胆道上皮细胞中,CDCA和UDCA可分别通过两种不同的核受体FXR和VDR诱导抗菌肽的表达,对抗有害微生物入侵胆道上皮[23]。然而,胆汁酸也可以促进某些耐胆汁酸细菌的增殖,如Bilophila wadsworthiaEscherichia coliListeria monocytogenes以及一些表达BSH的乳酸杆菌和双歧杆菌[24]

2 胆汁酸受体及其作用途径

胆汁酸作为一种信号分子,可以通过激活一些细胞受体参与调节物质代谢以及维持宿主内环境稳态等。已知的胆汁酸受体分为两大类,一类属于核受体,包括法尼醇X受体(FXR)、维生素D受体(VDR)、孕烷X受体(PXR)和组成型雄甾烷受体(CAR);另一类是膜受体,主要包含G蛋白偶联受体5(TGR5)和1-磷酸-鞘氨醇受体2(S1PR2)[25]。这些受体在全身各组织部位广泛表达,目前研究最多的是FXR受体和TGR5受体,参与调节胆汁酸代谢以及宿主糖脂和免疫代谢等途径。

2.1 通过FXR受体调节宿主代谢

FXR受体属于一类配体依赖的核受体,胆汁酸是FXR的内源性配体,其中最有效的激活剂是CDCA,其次是CA、DCA和LCA,而UDCA、Tα-MCA和Tβ-MCA被证明是FXR的天然拮抗剂[7]。FXR可在肝脏、肠道、肾和肾上腺等多组织表达,其中表达量最高的是肝脏和肠道[26]。胆汁酸激活FXR在调控胆汁酸代谢和糖脂代谢方面发挥着重要作用(图1表2)。

图1

图1   胆汁酸的主要作用途径及生理功能

Fig. 1   Main pathways and physiological functions of bile acids


表2   胆汁酸受体FXR和TGR5的主要作用途径

Table 2  Functional pathways of bile acid receptors FXR and TGR5

受体Receptor胆汁酸配体Ligand bile acids表达组织 Expressive tissues作用途径Functional pathways
激活剂
Activators
拮抗剂Antagonists
FXRCDCA、CA、T(G)CDCA、T(G)CA、LCA、DCA、HCAsTαMC、TβMCA
UDCA、isoDCA
肝脏胆汁酸代谢FXR—SHP-1—LRH-1—抑制CYP7A1
FXR—IR-1—激活胆汁酸转运载体蛋白
脂代谢FXR—SHP—SREBP-1c—抑制脂肪酸合成酶
FXR—PPARα—促进脂肪酸氧化代谢
FXR—ApoCII—LPL—促进甘油三酯水解
FXR—p-JNK—HNF4α—SRB1—促进HDL-C清除
糖代谢FXR—SHP—抑制糖异生基因
FXR—LPK—抑制糖酵解
肠道胆汁酸代谢FXR—FGF19/15—结合肝脏FGFR4/βKlotho—激活JNK途径—抑制CYP7A1
糖代谢FXR—FGF19/15—结合肝脏FGFR4/βKlotho—抑制肝脏糖异生基因
FXR—抑制GLP-1分泌
免疫细胞炎症反应FXR—抑制促炎细胞因子的分泌
TGR5T(G)LCA、LCA、T(G)DCA、DCA、CDCA、T(G)CDCA、TCA、CA、UDCA、HCAs棕色脂肪组织能量代谢TGR5—cAMP—Dio2—增强能量消耗
骨骼肌
免疫细胞免疫反应TGR5—cAMP—抑制NF-kB信号通路—抑制促炎细胞因子
肠道糖代谢TGR5—cAMP—促进GLP-1分泌
肠上皮稳态调控肠道干细胞自我更新、增殖和分化

新窗口打开| 下载CSV


FXR受体抑制胆汁酸合成酶的表达,负反馈调控胆汁酸的合成。Xu等[27]利用兔模型体内实验证实FXR可以抑制CYP7A1的活性及mRNA的表达,而胆汁酸合成限速酶并没有FXR的结合位点,说明FXR并不直接抑制其转录。在肝脏中,发现胆汁酸激活的FXR可诱导下游靶基因SHP-1的表达,SHP-1能与激活CYP7A1表达的肝受体同源物(LRH-1)形成抑制性复合物,从而阻断CYP7A1的表达[28]。Goodwin等[29]用雄性大鼠为模型也证明FXR可以抑制CYP27A1的转录,并证明了SHP也可以与肝脏核因子4α(HNF4α)结合来抑制CYP27A1的转录。肝脏中FXR的激活可以通过SHP途径来抑制胆汁酸合成酶的表达,然而在SHP缺陷小鼠中胆汁酸合成的反馈抑制作用并没有完全废除,暗示可能还存在独立于SHP的途径可调节胆汁酸的合成[30]。在小鼠的研究中发现,肠道中FXR的激活可以增加成纤维细胞生长因子15(FGF15,人同源FGF19)的表达和分泌,并通过血液循环到达肝脏与FGF受体4(FGFR4)结合,激活JNK途径从而抑制CYP7A1的表达[31]。此外,大量实验证明,转运相关蛋白上具有FXR受体的结合反应元件IR-1,如 FXR可直接与IR结合,激活转运蛋白BESP的表达,从而促进肝脏分泌胆汁酸[32]

脂质代谢失衡容易引起脂肪肝等代谢性疾病。随着对胆汁酸研究的深入,发现胆汁酸及其受体FXR对机体的脂质代谢具有重要调控作用。经研究证实,FXR基因敲除小鼠血浆和肝脏的甘油三酯水平均显著增加[33]。固醇调节元件结合蛋白(SREBPs)是一个膜结合转录因子家族,可激活编码胆固醇和不饱和脂肪酸合成所需酶的基因,是脂肪合成的重要调节因子[34]。被内源性胆汁酸激活的FXR以SHP依赖的方式介导下调SREBP-1c的表达,使脂肪酸合成酶(FAS)的表达降低,进而抑制肝脏脂肪的生成[35]。糖类反应元件结合蛋白(ChREBP)可促进糖类向脂质的转化,而胆汁酸活化FXR后对ChREBP的表达具有下调作用,从而抑制脂肪的合成[36]。过氧化物酶体增殖物激活受体α(PPAR-α)具有调节脂蛋白代谢、脂肪酸氧化的作用,其活性也可受到FXR的调控,导致脂肪酸氧化代谢增加,减轻脂肪堆积[37]。BAs-FXR不仅可以抑制脂肪的生成,其还具有促进甘油三酯转运吸收的作用。在FXR缺陷小鼠中,血清高密度脂蛋白(HDL)胆固醇水平升高,而导致去除血液中HDL胆固醇(HDL-C)的清道夫受体B1(SRB1)在肝脏中表达降低[38]。有研究对FXR调控SRB1的途径进行探究,发现肝脏FXR的激活可导致磷酸化JNK(p-JNK)水平的下降,随后上调肝细胞核因子4α(HNF4α)以及SRB1的表达,进而增强HDL-C的清除[39]。载脂蛋白ApoCII也是FXR的靶基因,其表达增加促进了脂蛋白酯酶(LPL)的表达,导致甘油三酯的水解增加[40]。此外,FXR还可上调磷脂转移蛋白(PLTP)的表达,促进对HDL中脂质的清除[41]。胆汁酸受体FXR通过参与脂质合成、脂质氧化代谢以及脂质的转运吸收等多途径、多靶点调控机体的脂质代谢,从而维持内环境相关脂质的稳态。

肝脏通过调节糖异生、糖原合成和糖酵解途径维持着血糖稳态。在肝脏FXR缺陷小鼠中观察到胰岛素抵抗导致葡萄糖稳态失调的现象[42]。一些研究已经证实肝脏BAs-FXR信号通过降低肝脏糖异生并诱导肝糖原合成来调节血糖水平[43]。磷酸烯醇式丙酮酸羧激酶(PEPCK)、葡萄糖-6-磷酸酶(G6Pase)和果糖-1,6-双磷酸酶(FBP1)是糖异生途径中的3种关键酶。Yamagata等[44]发现胆汁酸能以FXR-SHP依赖的途径与糖异生相关基因启动子区的核转录因子互作并抑制其表达。有研究表明FXR敲除小鼠表现出葡萄糖不耐受和胰岛素不敏感,而激活FXR后可降低G6Pase的表达,抑制肝脏糖异生从而降低小鼠的血糖水平[45]。丙酮酸激酶(LPK)是糖酵解途径中的关键基因,有研究证明FXR与LPK启动子区发生物理互作并降低LPK的表达,抑制糖酵解,从而增加糖原储存导致降糖反应[36]。作为FGF家族的一员,FGF15/19已经成为一种新的代谢稳态调节因子,在过表达FGF15的小鼠中观察到胰岛素敏感性大幅增强,进而导致其对饮食诱导的肥胖抵抗[46]。如前所述,FGF19会受到FXR受体的调节,BA-FXR信号诱导FGF19循环至肝脏与FGFR4及其辅受体βKlotho形成复合物,具有抑制糖异生并促进肝糖原合成的功能,实现降血糖作用[47]。而在肠内分泌L细胞中,被胆汁酸激活的FXR会干扰GLP-1的分泌和胰高血糖素原的表达,这一研究为BAs对血糖的调控提供了一种新的分子机制[48]。可见,不同部位表达的FXR在调控宿主糖代谢的不同途径上出现互相矛盾的结论,在后续对BAs-FXR信号途径的研究中应注意不同组织的作用特异性。

肝脏和肠道的免疫细胞中也检测到FXR的表达,其对免疫炎症反应也具有一定的调节作用。有研究表明巨噬细胞中FXR激活引起的炎症抑制可依赖SHP机制,阻断NF-kB信号传导,抑制促炎基因的启动子[49]。在树突状细胞中,胆汁酸激活FXR可抑制TNF的分泌,而在小鼠的自然杀伤T细胞中FXR的激活对骨桥蛋白(osteopontin)、IL1β、IFNγ的分泌具有抑制作用[5]。患有溃疡性结肠炎的人肠道NF-kB依赖性的细胞因子含量较高,肠道炎症反应严重,这与其FXR的表达降低也有一定的关系[50]。而在IBD小鼠模型中,给药激活FXR受体可保护肠屏障,降低炎症水平[51]

2.2 通过TGR5受体调节宿主代谢

2002年Maruyama等[52]首次报道了一种新的胆汁酸膜受体TGR5,属于G蛋白偶联受体(GPCR)超家族的一员。TGR5在很多组织中都有表达,如肝脏、肠道、胆囊、脂肪组织、肌肉、肾脏等;不同胆汁酸激活TGR5的效力不同,次级胆汁酸LCA、DCA及其结合型胆汁酸对胆汁酸的作用最强,其次是CDCA、CA及其结合胆汁酸[7]。作为胆汁酸受体,其也影响着胆汁酸代谢。胆汁酸激活TGR5可增加环腺苷酸(cAMP)水平,而cAMP可活跃ASBT转运蛋白,促进胆汁酸的吸收,避免细胞胆汁淤积[53]。BAs-TGR5信号途径在调节宿主能量代谢、葡萄糖稳态及炎症反应方面也是极其重要的(图1表2)。

脂肪和肌肉组织中TGR5受体激活与机体的能量消耗有关。Watanab等[54]在小鼠膳食中补充胆汁酸发现棕色脂肪组织和骨骼肌的能量消耗增加,这种作用与BAs-TGR5信号激活导致依赖cAMP的2型脱碘酶(Dio2)的表达增加有关,该酶可促进甲状腺素原(T4)脱碘转化为三碘甲状腺原氨酸(T3),增加耗氧率,从而增强机体的能量消耗。节食减肥后体重易反弹是因为节食在减少能量摄入的同时也降低了能量消耗。对节食前后肠道菌群和胆汁酸变化进行分析,发现有益菌Parabacteroides distasonis和非12α-羟基胆汁酸的比例减少,在饮食中补充益生菌和非12α-羟基胆汁酸UDCA、LCA可改善体重反弹现象。研究发现补充的胆汁酸通过激活TGR5,增加Dio2作用下产生的T3,通过促进解偶联蛋白1(UCP1)基因转录,引起棕色脂肪组织产热增加,从而增加机体的能量消耗[55]

胆汁酸对肠内分泌L细胞释放GLP-1有刺激作用,其作用机制不仅与FXR受体有关,还与TGR5的激活有关,并且两者的介导作用相反。糖尿病患者在接受高剂量的TGR5激动剂的治疗后,GLP-1会明显升高且血糖显著降低[56]。Thomas等[57]对TGR5激活促进GLP-1的机制进行研究,发现TGR5通过增加线粒体氧化磷酸化以及ATP/ADP的比率,使依赖ATP的钾通道(关闭和钙电压通道(CaV)打开,最终导致肠促胰岛素的GLP-1的释放增加,改善肥胖小鼠的血糖稳态。一般情况下,肠L细胞中TGR5激活介导的降血糖可能在摄食后迅速反应,而FXR的激活会出现延迟反应,这种差异导致BAs-TGR5信号对GLP-1的分泌更积极。Zheng等[3]研究发现猪体内有一类特殊的胆汁酸——HCAs能够作用于肠内分泌L细胞,同时激活TGR5且抑制FXR,导致cAMP水平增加,进而有效促进GLP-1的分泌,实现高效降血糖。

TGR5在免疫细胞中也有表达,在调控机体的炎症反应方面发挥着特别的功能。在神经炎症小鼠模型中,使用TUDCA治疗具有明显的抗炎作用,其可激活TGR5受体,抑制小胶质细胞分泌干扰素γ(IFN-γ)和肿瘤坏死因子-α(TNF-α)等促炎因子[58]。Kawamata等[59]在兔模型中发现,胆汁酸能通过TGR5受体影响巨噬细胞的功能以及抑制由脂多糖(LPS)刺激的促炎细胞因子分泌。在大鼠的Kupffer细胞中也有类似报道,BAs-TGR5信号可诱导cAMP的水平并减少炎症细胞因子的产生[60]。Pols等[61]通过在TGR5敲除和TGR5过表达的小鼠中分离巨噬细胞,明确了TGR5通过诱导cAMP的水平而影响NF-kB信号通路,实现对炎症反应的抑制作用。巨噬细胞根据其分泌因子和发挥的功能不同可以分为M1型巨噬细胞和M2型巨噬细胞,M1型主要分泌促炎因子,而M2型表达炎症抑制因子IL-10等[62]。在M1型状态下,TGR5刺激表皮生长因子受体(EGFR)依赖的信号通路的反转录激活,导致NF-κB的激活,增加促炎细胞因子的表达[4]。BAs-TGR5信号诱导产生的cAMP可激活促使cAMP反应元件结合蛋白(CREB)磷酸化的PKA表达,磷酸化后的CREB与cAMP应答元素(CRE)结合调节IL-10的表达[63],可促进巨噬细胞从M1状态分化为M2。巨噬细胞中依赖胆汁酸激活的TGR5信号并不能完全诱导巨噬细胞从M1分化到M2,一般呈现两种亚型的混合状态,但是M2型处于优势地位,表现出明显的免疫抑制[4]。由此可见,BAs-TGR5信号有助于调节抗炎和促炎细胞因子分泌的平衡。除了对肠内分泌细胞和免疫细胞具有调控作用,近期有团队还探究了BAs-TGR5信号对肠道干细胞是否也有影响。从野生型小鼠和TGR5敲除小鼠中提取肠类器官,与BAs或TGR5激动剂共同培养,通过形态学观察、免疫荧光实验以及流式细胞术分析肠道干细胞的数量、增殖和分化,发现TGR5敲除组小鼠肠内分泌细胞和杯状细胞的分化显著降低,且在葡聚糖硫酸钠(DSS)诱导下,加速了结肠炎进程,表明BAs-TGR5调控肠道干细胞的更新和增殖,对肠类器官的生长具有促进作用[64]

3 胆汁酸的生理功能

进食是触发胆汁酸释放、循环的主要因素,胆汁酸具有较强的表面活性,可有效地将脂质乳化,形成乳糜微粒,从而扩大与脂肪水解酶的接触面,同时激活脂肪酶,加速脂质的消化吸收[65]。现在胆汁酸已被视为餐后信使,通过激活不同器官中的FXR和TGR5受体对全身代谢进行调节,例如胆汁酸通过受体信号控制肠内分泌激素GLP-1的分泌调节葡萄糖稳态。一旦这些稳态失衡,都会导致机体发生严重的代谢功能障碍。胆汁酸除了消化代谢功能外,目前对肠道菌群-胆汁酸-宿主轴信号传导的研究已经拓展到各个领域,包括肠肝轴、肠脑轴、肠免疫轴等(图1图2)。

图2

图2   胆汁酸与免疫调节

Fig. 2   Bile acids and immune regulation


3.1 肠肝轴

胆汁酸的合成、运输和代谢失调是引起人类肝脏疾病的常见病因。非酒精性脂肪肝病(NAFLD)是一种从肝脂肪变性到非酒精性脂肪肝炎(NASH),最终发展到肝硬化的疾病,其病理发展的严重程度与血清总胆汁酸水平呈正相关,NASH患者粪便中初级胆汁酸CA和CDCA含量以及结合胆汁酸与非结合胆汁酸的比值显著升高[66]。进一步研究发现NAFLD患者胆汁酸转运蛋白水平以及肝脏对FGF19的反应也出现下降[67],表明这些患者的BAs-FXR信号途径受到破坏。胆汁酸在肝脏中淤积抑制FXR的转录活性,从而减少胆汁酸转运蛋白的表达,引起持续炎症的发生,最终还会促进肝细胞癌(HCC)的发展[4]。妊娠期胆汁淤积是一种由于胆汁酸在肝脏内过量积累影响胆汁酸转运蛋白的表达所引起的肝脏疾病,目前最有效的治疗方法是通过一种天然胆汁酸-熊脱氧胆酸(UDCA)进行治疗改善[68]。UDCA可以降低胆汁酸的细胞毒性从而保护胆管细胞,抑制肝细胞凋亡[69]。此外,UDCA对于减轻NASH的肝损伤也具有一定作用[70]。FXR激动剂也可通过诱导BESP刺激肝脏胆汁酸流出改善胆汁淤积[71]。在肝脏FXR缺乏的小鼠中活化肠细胞中的FXR可以通过激活FGF15恢复胆汁酸稳态,防止肝脏恶性癌变的发生[72]。这些研究也为以胆汁酸及其信号通路为靶向,治疗一些肝脏代谢损伤疾病提供了可能。

3.2 肠脑轴

胆汁酸不仅通过肠肝循环介导肝脏和肠道的互作,还在全身循环,可以通过直接或间接的途径向中枢神经系统传递信号。有研究已经证明大脑中也可以检测到胆汁酸的存在,其中游离型胆汁酸可由外周被动扩散至脑内,结合型胆汁酸通过主动转运穿过血脑屏障[73]。并且胆汁酸转运蛋白及胆汁酸受体都在大脑中有一定的表达。在亨廷顿病、阿尔茨海默症和帕金森等几种神经退行性疾病动物模型中观察到TUDCA具有神经保护作用[74]。TGR5在神经元和星形胶质细胞中均可表达,胆汁酸激活后可降低小胶质细胞的活化并抑制促炎细胞因子IL-1β、IL-6和TNFα的产生,减少神经炎症[75]。FXR基因敲除小鼠的抗忧郁和抗焦虑能力增强,但是认知行为受到损害,检测其海马区的神经递质发现GABA/Glu的平衡被扰乱,小脑中5-HIAA明显升高,表明FXR可调节不同脑区域神经递质的稳态来影响行为[76]。除了与受体结合的直接途径外,胆汁酸激活肠道FXR和TGR5可导致FGF19和GLP-1两种信号分子的释放,间接作用于大脑。穿过血脑屏障的FGF19通过与大脑中的FGFR1-4和β-klotho蛋白结合形成稳定受体复合物发挥作用[77],影响能量代谢和糖代谢。在高脂肪饮食喂养的胰岛素抵抗、瘦素缺乏的两个小鼠模型中,脑室内灌注FGF19可改善血糖状态,降低胰岛素抵抗,增强胰岛素信号在外周的传递[78],而在大脑中使用FGFR抑制剂可阻断这种有益效应[79]。胆汁酸刺激TGR5产生的肠道激素GLP-1也可以从肠道扩散至大脑,其主要通过两种途径:一是通过体循环至大脑与其受体结合,二是通过迷走神经进行信号传导[73]。这些都为胆汁酸信号途径作用于大脑提供证据,但是体内胆汁酸刺激是否足以对中枢神经系统产生实质性影响还有待于研究考证。

3.3 肠免疫轴

早前就有一系列研究证实肠道共生菌及其代谢物能够调节Treg的产生[80-81],直至2019年,连续几项研究发现胆汁酸也可以影响T细胞的分化和活性,从而与免疫系统互作。在肠道固有层中表达一种独特的Treg细胞群—— FoxP3+ Tregs,其表达转录因子RORγ,也被称为RORγ+ Tregs,有益于控制肠道炎症及维持稳态[82]。Song等[83]通过在小鼠饮用水中加入不同的胆汁酸盐实验,发现一些去结合的初级胆汁酸混合物(如CA/CDCA/UDCA)以及某些次级胆汁酸混合物(LCA/3-oxoLCA)可通过胆汁酸受体VDR调节RORγ+ Treg的水平。VDR是一种核受体,能被LCA、3-oxo-LCA、glyco-LCA、6-oxo-LCA以及两种合成衍生物LCA-acetate和LCA-propionate激活,作用于肠细胞发挥抗炎功能[84-86]。LCA和3-oxoLCA也是胆汁酸受体PXR的主要激动剂[87],在DSS诱导的结肠炎小鼠中激活PXR可以抑制NF-kB信号,减少趋化细胞因子的表达,发挥抗炎作用[88]。在小鼠免疫细胞实验中,还发现isoDCA和ω-MCA可以显著促进调节性T细胞(Treg)的分化但不影响辅助性T 细胞17(Th17)的分化。在一系列体外实验中对这种作用机制进行探究,发现这种调节作用需要树突状细胞的参与,且是通过抑制树突状细胞中的炎症反应降低对T细胞启动的免疫刺激性,增强Treg的生成[89]。然而,Hang等[90]在小鼠中发现3-oxoLCA通过直接与RORγt发生物理互作并抑制其转录活性来抑制Th17分化;另一种LCA衍生物isoalloLCA可促进Treg的分化,并通过实验证实这种作用机制与线粒体活性氧的增加以及FoxP3启动子区的H3K27乙酰化有关。此外,与3-oxoLCA类似,其经过3β-羟基氧化后生成的isoLCA也可以与RORγt互作抑制Th17分化[15]。这些研究阐明了肠道细菌产生的次级胆汁酸代谢物调节T细胞功能的新途径,但目前大多数研究都只集中于胆汁酸对Treg和Th17的分化调节,后续还需要更多地研究探索胆汁酸对其他T细胞亚型的影响,进而更全面系统地研究肠道菌群与宿主免疫互作的机制。

4 胆汁酸在生物医学和动物生产中的应用

胆汁酸与肠道微生物群通过多种信号通路与宿主相互作用,发挥其生理作用,现在很多人体上的研究也证实了在小鼠模型中发现的结果,并加深了我们对胆汁酸与疾病之间关系的理解,为通过调控胆汁酸受体FXR和TGR5信号通路治疗肠道炎症和肿瘤发生奠定基础。

如原发性胆管炎(PBC)是一种慢性胆汁淤积性自身免疫性疾病,可进展为终末期肝病。美国食品和药物管理局(FDA)批准的第一个治疗PBC的药物是UDCA,可以减缓该疾病的进展,但是UDCA对约30%-40%的患者无效[91]。而在所有的合成胆汁酸衍生物中,OCA以FXR激动剂的形式被FDA批准用于临床治疗对UDCA无反应的患者[92]。UDCA作为一种治疗性胆汁酸还可以性别特异性地影响肠道微生物群落组成,降低男性晚期腺瘤的风险,而对女性无效[93]

胆汁酸及其衍生物,除了用于人类疾病的治疗,还可用于动物的生产上。随着集约化养殖业的发展,高脂肪、高能量配合饲料的使用有益于满足养殖动物快速生长的需求,但同时也会导致脂肪的过度沉积,对畜禽肉质及商业价值造成严重影响。目前,胆汁酸已成为一种新型饲料添加剂,可有效提高饲粮中脂质的吸收利用率,改善畜禽的生长性能[94]。断奶仔猪常因饲料成分的改变影响肠道屏障诱发腹泻,影响仔猪的生长速度,有试验发现在断奶仔猪的日粮中添加胆汁酸可通过改变肠道菌群组成,显著提高断奶仔猪的平均日增重、降低料肉比,改善其生产性能和肠道炎症[95]。已有试验证实,在早期断奶仔猪饲粮中补充初级胆汁酸CDCA具有提高血浆中促肠道生长营养因子胰高血糖素样肽2(GLP-2)的趋势,提高断奶后肠道屏障的完整性,改善肠道黏膜的保护潜力[96]。母猪妊娠期胆汁酸失调对胎儿的生长具有有害影响,而通过调节肠道有益菌改善胆汁酸组成,从而提高母猪的繁殖性能[97]。还有一些研究表明在泌乳母猪饲粮中添加胆汁酸可显著降低母乳中胆固醇含量并提升总蛋白含量,改善初乳质量,从而显著提高断奶仔猪重及血清胰岛素、甲状腺激素的水平,促进仔猪的发育成熟。胆汁酸对动物生产的积极作用已受到越来越多的重视,但目前大部分试验都倾向于在饲粮中直接补充,使用剂量和使用时间的把控是当前研究中面临的主要问题。而通过内源性调节肠道菌群的组成来维持体内胆汁酸稳态,可成为无抗时代促进畜禽健康生长的养殖新思路。

5 总结与展望

胆汁酸是一种具有两亲性的类固醇分子,其组成及多样性受到宿主和微生物的共同代谢调控。胆汁酸除了通过肠肝循环促进膳食中脂类物质的消化吸收,还可以到达几乎身体的所有器官,作为强信号分子通过不同的受体和细胞信号与宿主相互作用,调节机体脂质、葡萄糖、能量代谢以及免疫炎症等。因此,本综述通过系统地探讨肠道微生物-胆汁酸的互作及其发挥生理功能的作用机制,以期在无抗背景下靶向肠道菌群及胆汁酸代谢物对动物的营养健康进行调控,并在后期畜禽生产的应用研究中提供一定的参考。

肠道菌群可以通过产生的胆汁酸代谢物为桥梁建立与宿主生理之间的联系,并且随着宿主健康状态的改变,胆汁酸在机体内的分布也会发生变化。肠道微生态一旦失调,就可能会通过影响胆汁酸的解耦联、脱羟及结构修饰等过程破坏正常胆汁酸代谢,导致肠道上皮炎症增加,进一步破坏肠道屏障。此外,已有较多的研究证实宿主遗传可在一定程度上影响肠道菌群的功能,而在后续的一些研究中也可从微生物胆汁酸代谢物的角度出发,系统地探究影响肠道菌群和胆汁酸组成的遗传因素,为菌群与宿主互作的研究提供崭新视角。

近些年来,随着对胆汁酸合成、代谢及其功能研究的不断深入,胆汁酸及其多种受体可能成为治疗脂肪肝、糖尿病等代谢综合征以及免疫炎症的新方向,但知悉其作用机理并向临床转化还需要进行更系统、更深入的实践探索。值得注意的是,在研究胆汁酸信号影响宿主的作用机制时,应考虑不同物种间胆汁酸的组成谱以及不同组织部位上受体作用途径的差异性。在未来还可通过结合肠道宏基因组、培养组学等技术进一步挖掘不同状态下肠道菌群与胆汁酸之间的互作关系。

参考文献

Wu JY, Wang K, Wang XM, et al.

The role of the gut microbiome and its metabolites in metabolic diseases

[J]. Protein Cell, 2021, 12(5): 360-373.

DOI:10.1007/s13238-020-00814-7      [本文引用: 1]

It is well known that an unhealthy lifestyle is a major risk factor for metabolic diseases, while in recent years, accumulating evidence has demonstrated that the gut microbiome and its metabolites also play a crucial role in the onset and development of many metabolic diseases, including obesity, type 2 diabetes, nonalcoholic fatty liver disease, cardiovascular disease and so on. Numerous microorganisms dwell in the gastrointestinal tract, which is a key interface for energy acquisition and can metabolize dietary nutrients into many bioactive substances, thus acting as a link between the gut microbiome and its host. The gut microbiome is shaped by host genetics, immune responses and dietary factors. The metabolic and immune potential of the gut microbiome determines its significance in host health and diseases. Therefore, targeting the gut microbiome and relevant metabolic pathways would be effective therapeutic treatments for many metabolic diseases in the near future. This review will summarize information about the role of the gut microbiome in organism metabolism and the relationship between gut microbiome-derived metabolites and the pathogenesis of many metabolic diseases. Furthermore, recent advances in improving metabolic diseases by regulating the gut microbiome will be discussed.

Chiang JYL.

Bile acid metabolism and signaling

[J]. Compr Physiol, 2013, 3(3): 1191-1212.

DOI:10.1002/cphy.c120023      PMID:23897684      [本文引用: 2]

Bile acids are important physiological agents for intestinal nutrient absorption and biliary secretion of lipids, toxic metabolites, and xenobiotics. Bile acids also are signaling molecules and metabolic regulators that activate nuclear receptors and G protein-coupled receptor (GPCR) signaling to regulate hepatic lipid, glucose, and energy homeostasis and maintain metabolic homeostasis. Conversion of cholesterol to bile acids is critical for maintaining cholesterol homeostasis and preventing accumulation of cholesterol, triglycerides, and toxic metabolites, and injury in the liver and other organs. Enterohepatic circulation of bile acids from the liver to intestine and back to the liver plays a central role in nutrient absorption and distribution, and metabolic regulation and homeostasis. This physiological process is regulated by a complex membrane transport system in the liver and intestine regulated by nuclear receptors. Toxic bile acids may cause inflammation, apoptosis, and cell death. On the other hand, bile acid-activated nuclear and GPCR signaling protects against inflammation in liver, intestine, and macrophages. Disorders in bile acid metabolism cause cholestatic liver diseases, dyslipidemia, fatty liver diseases, cardiovascular diseases, and diabetes. Bile acids, bile acid derivatives, and bile acid sequestrants are therapeutic agents for treating chronic liver diseases, obesity, and diabetes in humans.© 2013 American Physiological Society.

Zheng XJ, Chen TL, Jiang RQ, et al.

Hyocholic acid species improve glucose homeostasis through a distinct TGR5 and FXR signaling mechanism

[J]. Cell Metab, 2021, 33(4): 791-803.e7.

DOI:10.1016/j.cmet.2020.11.017      PMID:33338411      [本文引用: 3]

Hyocholic acid (HCA) and its derivatives are found in trace amounts in human blood but constitute approximately 76% of the bile acid (BA) pool in pigs, a species known for its exceptional resistance to type 2 diabetes. Here, we show that BA depletion in pigs suppressed secretion of glucagon-like peptide-1 (GLP-1) and increased blood glucose levels. HCA administration in diabetic mouse models improved serum fasting GLP-1 secretion and glucose homeostasis to a greater extent than tauroursodeoxycholic acid. HCA upregulated GLP-1 production and secretion in enteroendocrine cells via simultaneously activating G-protein-coupled BA receptor, TGR5, and inhibiting farnesoid X receptor (FXR), a unique mechanism that is not found in other BA species. We verified the findings in TGR5 knockout, intestinal FXR activation, and GLP-1 receptor inhibition mouse models. Finally, we confirmed in a clinical cohort, that lower serum concentrations of HCA species were associated with diabetes and closely related to glycemic markers.Copyright © 2020 Elsevier Inc. All rights reserved.

Jia W, Xie GX, Jia WP.

Bile acid-microbiota crosstalk in gastrointestinal inflammation and carcinogenesis

[J]. Nat Rev Gastroenterol Hepatol, 2018, 15(2): 111-128.

DOI:10.1038/nrgastro.2017.119      PMID:29018272      [本文引用: 4]

Emerging evidence points to a strong association between the gut microbiota and the risk, development and progression of gastrointestinal cancers such as colorectal cancer (CRC) and hepatocellular carcinoma (HCC). Bile acids, produced in the liver, are metabolized by enzymes derived from intestinal bacteria and are critically important for maintaining a healthy gut microbiota, balanced lipid and carbohydrate metabolism, insulin sensitivity and innate immunity. Given the complexity of bile acid signalling and the direct biochemical interactions between the gut microbiota and the host, a systems biology perspective is required to understand the liver-bile acid-microbiota axis and its role in gastrointestinal carcinogenesis to reverse the microbiota-mediated alterations in bile acid metabolism that occur in disease states. An examination of recent research progress in this area is urgently needed. In this Review, we discuss the mechanistic links between bile acids and gastrointestinal carcinogenesis in CRC and HCC, which involve two major bile acid-sensing receptors, farnesoid X receptor (FXR) and G protein-coupled bile acid receptor 1 (TGR5). We also highlight the strategies and cutting-edge technologies to target gut-microbiota-dependent alterations in bile acid metabolism in the context of cancer therapy.

Fuchs CD, Trauner M.

Role of bile acids and their receptors in gastrointestinal and hepatic pathophysiology

[J]. Nat Rev Gastroenterol Hepatol, 2022, 19(7): 432-450.

DOI:10.1038/s41575-021-00566-7      PMID:35165436      [本文引用: 2]

Bile acids (BAs) can regulate their own metabolism and transport as well as other key aspects of metabolic homeostasis via dedicated (nuclear and G protein-coupled) receptors. Disrupted BA transport and homeostasis results in the development of cholestatic disorders and contributes to a wide range of liver diseases, including nonalcoholic fatty liver disease and hepatocellular and cholangiocellular carcinoma. Furthermore, impaired BA homeostasis can also affect the intestine, contributing to the pathogenesis of irritable bowel syndrome, inflammatory bowel disease, and colorectal and oesophageal cancer. Here, we provide a summary of the role of BAs and their disrupted homeostasis in the development of gastrointestinal and hepatic disorders and present novel insights on how targeting BA pathways might contribute to novel treatment strategies for these disorders.© 2022. Springer Nature Limited.

Alrefai WA, Gill RK.

Bile acid transporters: structure, function, regulation and pathophysiological implications

[J]. Pharm Res, 2007, 24(10): 1803-1823.

DOI:10.1007/s11095-007-9289-1      URL     [本文引用: 1]

Wahlström A, Sayin SI, Marschall HU, et al.

Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism

[J]. Cell Metab, 2016, 24(1): 41-50.

DOI:10.1016/j.cmet.2016.05.005      PMID:27320064      [本文引用: 4]

The gut microbiota is considered a metabolic "organ" that not only facilitates harvesting of nutrients and energy from the ingested food but also produces numerous metabolites that signal through their cognate receptors to regulate host metabolism. One such class of metabolites, bile acids, is produced in the liver from cholesterol and metabolized in the intestine by the gut microbiota. These bioconversions modulate the signaling properties of bile acids via the nuclear farnesoid X receptor and the G protein-coupled membrane receptor 5, which regulate numerous metabolic pathways in the host. Conversely, bile acids can modulate gut microbial composition both directly and indirectly through activation of innate immune genes in the small intestine. Thus, host metabolism can be affected through microbial modifications of bile acids, which lead to altered signaling via bile acid receptors, but also by altered microbiota composition.Copyright © 2016 Elsevier Inc. All rights reserved.

Swann JR, Want EJ, Geier FM, et al.

Systemic gut microbial modulation of bile acid metabolism in host tissue compartments

[J]. Proc Natl Acad Sci USA, 2011, 108(Suppl 1): 4523-4530.

DOI:10.1073/pnas.1006734107      URL     [本文引用: 1]

\n We elucidate the detailed effects of gut microbial depletion on the bile acid sub-metabolome of multiple body compartments (liver, kidney, heart, and blood plasma) in rats. We use a targeted ultra-performance liquid chromatography with time of flight mass-spectrometry assay to characterize the differential primary and secondary bile acid profiles in each tissue and show a major increase in the proportion of taurine-conjugated bile acids in germ-free (GF) and antibiotic (streptomycin/penicillin)-treated rats. Although conjugated bile acids dominate the hepatic profile (97.0 ± 1.5%) of conventional animals, unconjugated bile acids comprise the largest proportion of the total measured bile acid profile in kidney (60.0 ± 10.4%) and heart (53.0 ± 18.5%) tissues. In contrast, in the GF animal, taurine-conjugated bile acids (especially taurocholic acid and tauro-β-muricholic acid) dominated the bile acid profiles (liver: 96.0 ± 14.5%; kidney: 96 ± 1%; heart: 93 ± 1%; plasma: 93.0 ± 2.3%), with unconjugated and glycine-conjugated species representing a small proportion of the profile. Higher free taurine levels were found in GF livers compared with the conventional liver (5.1-fold;\n P\n < 0.001). Bile acid diversity was also lower in GF and antibiotic-treated tissues compared with conventional animals. Because bile acids perform important signaling functions, it is clear that these chemical communication networks are strongly influenced by microbial activities or modulation, as evidenced by farnesoid X receptor-regulated pathway transcripts. The presence of specific microbial bile acid co-metabolite patterns in peripheral tissues (including heart and kidney) implies a broader signaling role for these compounds and emphasizes the extent of symbiotic microbial influences in mammalian homeostasis.\n

Jones BV, Begley M, Hill C, et al.

Functional and comparative metagenomic analysis of bile salt hydrolase activity in the human gut microbiome

[J]. Proc Natl Acad Sci USA, 2008, 105(36): 13580-13585.

DOI:10.1073/pnas.0804437105      PMID:18757757      [本文引用: 1]

Bile salt hydrolases (BSHs) catalyze the "gateway" reaction in a wider pathway of bile acid modification by the gut microbiota. Because bile acids function as signaling molecules regulating their own biosynthesis, lipid absorption, cholesterol homeostasis, and local mucosal defenses in the intestine, microbial BSH activity has the potential to greatly influence host physiology. However, the function, distribution, and abundance of BSH enzymes in the gut community are unknown. Here, we show that BSH activity is a conserved microbial adaptation to the human gut environment with a high level of redundancy in this ecosystem. Through metagenomic analyses we identified functional BSH in all major bacterial divisions and archaeal species in the gut and demonstrate that BSH is enriched in the human gut microbiome. Phylogenetic analysis illustrates that selective pressure in the form of conjugated bile acid has driven the evolution of members of the Ntn_CGH-like family of proteins toward BSH activity in gut-associated species. Furthermore, we demonstrate that BSH mediates bile tolerance in vitro and enhances survival in the murine gut in vivo. Overall, we demonstrate the use of function-driven metagenomics to identify functional anchors in complex microbial communities, and dissect the gut microbiome according to activities relevant to survival in the mammalian gastrointestinal tract.

Song ZW, Cai YY, Lao XZ, et al.

Taxonomic profiling and populational patterns of bacterial bile salt hydrolase(BSH)genes based on worldwide human gut microbiome

[J]. Microbiome, 2019, 7(1): 9.

DOI:10.1186/s40168-019-0628-3      [本文引用: 1]

White BA, Lipsky RL, Fricke RJ, et al.

Bile acid induction specificity of 7 alpha-dehydroxylase activity in an intestinal Eubacterium species

[J]. Steroids, 1980, 35(1): 103-109.

PMID:7376208      [本文引用: 1]

The addition of cholic acid to growing cultures of Eubacterium species V.P.I. 12708 caused a 25 and 46-fold increase in 7 alpha-dehydroxylation activity using cell extracts or whole cell suspensions, respectively. Bile acid conversion rates using either [14C]-cholic acid or [14C]-chenodeoxycholic acid as substrates increased at approximately the same rate when either cholic or chenodeoxycholic acid was added to growing cultures as inducer. The induction of 7 alpha-dehydroxylase activity was highly specific requiring a free C-24-carboxyl group and an unhindered 7 alpha-hydroxy group on the B ring of the steroid nucleus. Unexpectedly, cholic acid also rapidly induced NADH:flavin oxidoreductase activity in growing cultures of this bacterium.

Funabashi M, Grove TL, Wang M, et al.

A metabolic pathway for bile acid dehydroxylation by the gut microbiome

[J]. Nature, 2020, 582(7813): 566-570.

DOI:10.1038/s41586-020-2396-4      [本文引用: 1]

Eyssen H, de Pauw G, Stragier J, et al.

Cooperative formation of Omega-muricholic acid by intestinal microorganisms

[J]. Appl Environ Microbiol, 1983, 45(1): 141-147.

DOI:10.1128/aem.45.1.141-147.1983      URL     [本文引用: 2]

Three anaerobic bacteria, isolated from the ceca of rats and mice, converted, through a concerted mechanism, beta-muricholic acid, the predominant bile acid in germfree rats, into omega-muricholic acid. One isolate was a Eubacterium lentum strain; the second and third isolates were tentatively identified as atypical Fusobacterium sp. strains. The conversion of beta-muricholic acid into omega-muricholic acid proceeded in two steps: E. lentum oxidized the 6 beta-hydroxyl group of beta-muricholic acid to a 6-oxo group, which was reduced by either of the two other species to a 6 alpha-hydroxyl group, yielding omega-muricholic acid. This transformation occurred both in vitro and in gnotobiotic rats. Monoassociation of germfree rats with the E. lentum strain gave rise to an unidentified fecal bile acid, probably a derivative of beta-muricholic acid having a double bond in the side chain.

Sacquet EC, Raibaud PM, Mejean C, et al.

Bacterial formation of Omega-muricholic acid in rats

[J]. Appl Environ Microbiol, 1979, 37(6): 1127-1131.

DOI:10.1128/aem.37.6.1127-1131.1979      URL     [本文引用: 1]

In the feces of conventional rats, the amount of omega-muricholic and hyodeoxycholic acids vary according to the diet. To understand this phenomenon, we investigated the bacterial formation of these bile acids. The present paper reports the first isolation, from conventional rat feces, of a strain of Clostridium group III which transforms beta-muricholic acid, the main bile acid in germfree rats, into omega-muricholic acid.

Paik D, Yao LN, Zhang YC, et al.

Human gut bacteria produce ΤΗ17-modulating bile acid metabolites

[J]. Nature, 2022, 603(7903): 907-912.

DOI:10.1038/s41586-022-04480-z      [本文引用: 3]

Li W, Hang SY, Fang Y, et al.

A bacterial bile acid metabolite modulates Treg activity through the nuclear hormone receptor NR4A1

[J]. Cell Host Microbe, 2021, 29(9): 1366-1377.e9.

DOI:10.1016/j.chom.2021.07.013      URL     [本文引用: 1]

Noh DO, Gilliland SE.

Influence of bile on cellular integrity and β-galactosidase activity of Lactobacillus acidophilus

[J]. J Dairy Sci, 1993, 76(5): 1253-1259.

PMID:8505417      [本文引用: 1]

The influence of bile on beta-galactosidase activity, cellular integrity, cellular retention of beta-galactosidase, and cellular permeability of five strains of Lactobacillus acidophilus was investigated. The five strains were also compared for bile tolerance. Two strains, 223 and 4356, were significantly less resistant to bile than the others (107, NCFM, and 606). beta-Galactosidase activity of all five strains was significantly higher in the presence of.3% oxgall than in its absence. Strain 107 showed the highest increase of enzyme activity in the presence of oxgall. Cells were not lysed in the presence of.3% oxgall, and beta-galactosidase was retained inside the cell even after extended incubation (60 min) in the presence of.3% oxgall. However, material that absorbed light at 260 nm leaked from the cells in the presence of oxgall. We concluded that, in the presence of bile, the permeability of cells of L. acidophilus increased, permitting more substrate to enter the cells, thus increasing the beta-galactosidase activity of whole cells.

Kandell RL, Bernstein C.

Bile salt/acid induction of DNA damage in bacterial and mammalian cells: implications for colon cancer

[J]. Nutr Cancer, 1991, 16(3/4): 227-238.

DOI:10.1080/01635589109514161      URL     [本文引用: 1]

Bernstein H, Payne CM, Bernstein C, et al.

Activation of the promoters of genes associated with DNA damage, oxidative stress, ER stress and protein malfolding by the bile salt, deoxycholate

[J]. Toxicol Lett, 1999, 108(1): 37-46.

DOI:10.1016/s0378-4274(99)00113-7      PMID:10472808      [本文引用: 1]

Toxic bile salts, retained within the liver because of impaired biliary excretion, are considered to play a major role in liver injury during cholestasis. Bile salts cause cellular stresses that may result in apoptosis. To better understand such cellular stresses, the effect of the bile salt sodium deoxycholate (NaDOC) on activation of 13 specific gene promoters or response elements associated with different cellular stresses was measured in the transformed human hepatoma line, HepG2. NaDOC was found to activate transcription factors and induce or activate the promoters of genes that respond to protein malfolding (grp78 and hsp70), DNA damage (gadd153, hsp70 and c-fos), oxidative stress (NF-kappaB, c-fos, hsp70 and gadd153), ER stress (grp78) and Ca++ imbalance (grp78).

Sorg JA, Sonenshein AL.

Inhibiting the initiation of Clostridium difficile spore germination using analogs of chenodeoxycholic acid, a bile acid

[J]. J Bacteriol, 2010, 192(19): 4983-4990.

DOI:10.1128/JB.00610-10      URL     [本文引用: 1]

\n To cause disease,\n Clostridium difficile\n spores must germinate in the host gastrointestinal tract. Germination is initiated upon exposure to glycine and certain bile acids, e.g., taurocholate. Chenodeoxycholate, another bile acid, inhibits taurocholate-mediated germination. By applying Michaelis-Menten kinetic analysis to\n C. difficile\n spore germination, we found that chenodeoxycholate is a competitive inhibitor of taurocholate-mediated germination and appears to interact with the spores with greater apparent affinity than does taurocholate. We also report that several analogs of chenodeoxycholate are even more effective inhibitors. Some of these compounds resist 7α-dehydroxylation by\n Clostridium scindens\n, a core member of the normal human colonic microbiota, suggesting that they are more stable than chenodeoxycholate in the colonic environment.\n

Weingarden AR, Chen C, Bobr A, et al.

Microbiota transplantation restores normal fecal bile acid composition in recurrent Clostridium difficile infection

[J]. Am J Physiol Gastrointest Liver Physiol, 2014, 306(4): G310-G319.

DOI:10.1152/ajpgi.00282.2013      URL     [本文引用: 1]

Fecal microbiota transplantation (FMT) has emerged as a highly effective therapy for refractory, recurrent Clostridium difficile infection (CDI), which develops following antibiotic treatments. Intestinal microbiota play a critical role in the metabolism of bile acids in the colon, which in turn have major effects on the lifecycle of C. difficile bacteria. We hypothesized that fecal bile acid composition is altered in patients with recurrent CDI and that FMT results in its normalization. General metabolomics and targeted bile acid analyses were performed on fecal extracts from patients with recurrent CDI treated with FMT and their donors. In addition, 16S rRNA gene sequencing was used to determine the bacterial composition of pre- and post-FMT fecal samples. Taxonomic bacterial composition of fecal samples from FMT recipients showed rapid change and became similar to the donor after the procedure. Pre-FMT fecal samples contained high concentrations of primary bile acids and bile salts, while secondary bile acids were nearly undetectable. In contrast, post-FMT fecal samples contained mostly secondary bile acids, as did non-CDI donor samples. Therefore, our analysis showed that FMT resulted in normalization of fecal bacterial community structure and metabolic composition. Importantly, metabolism of bile salts and primary bile acids to secondary bile acids is disrupted in patients with recurrent CDI, and FMT corrects this abnormality. Since individual bile salts and bile acids have pro-germinant and inhibitory activities, the changes suggest that correction of bile acid metabolism is likely a major mechanism by which FMT results in a cure and prevents recurrence of CDI.

Inagaki T, Moschetta A, Lee YK, et al.

Regulation of antibacterial defense in the small intestine by the nuclear bile acid receptor

[J]. Proc Natl Acad Sci USA, 2006, 103(10): 3920-3925.

DOI:10.1073/pnas.0509592103      PMID:16473946      [本文引用: 1]

Obstruction of bile flow results in bacterial proliferation and mucosal injury in the small intestine that can lead to the translocation of bacteria across the epithelial barrier and systemic infection. These adverse effects of biliary obstruction can be inhibited by administration of bile acids. Here we show that the farnesoid X receptor (FXR), a nuclear receptor for bile acids, induces genes involved in enteroprotection and inhibits bacterial overgrowth and mucosal injury in ileum caused by bile duct ligation. Mice lacking FXR have increased ileal levels of bacteria and a compromised epithelial barrier. These findings reveal a central role for FXR in protecting the distal small intestine from bacterial invasion and suggest that FXR agonists may prevent epithelial deterioration and bacterial translocation in patients with impaired bile flow.

D'Aldebert E, Biyeyeme Bi Mve MJ, Mergey M, et al.

Bile salts control the antimicrobial peptide cathelicidin through nuclear receptors in the human biliary epithelium

[J]. Gastroenterology, 2009, 136(4): 1435-1443.

DOI:10.1053/j.gastro.2008.12.040      PMID:19245866      [本文引用: 1]

Under normal conditions, the biliary tract is a microbial-free environment. The absence of microorganisms has been attributed to various defense mechanisms that include the physicochemical and signaling actions of bile salts. Here, we hypothesized that bile salts may stimulate the expression of a major antimicrobial peptide, cathelicidin, through nuclear receptors in the biliary epithelium.The expression of cathelicidin was analyzed in human liver samples by immunostaining and reverse-transcription quantitative polymerase chain reaction. The regulation of cathelicidin expression by the endogenous bile salt, chenodeoxycholic acid, and by the therapeutic bile salt, ursodeoxycholic acid (UDCA), was assessed in human biliary epithelial cells in which endogenous nuclear receptor expression was blunted by siRNA or dominant-negative strategies.In the human liver, biliary epithelial cells show intense immunoreactivity for cathelicidin and for the vitamin D receptor. In cultured biliary epithelial cells, chenodeoxycholic acid and UDCA induce cathelicidin expression through 2 different nuclear receptors: the farnesoid X receptor and the vitamin D receptor, respectively. Importantly, vitamin D further increases the induction of cathelicidin expression by both bile salts. In a prototypical inflammatory biliary disease (ie, primary biliary cirrhosis), we document that hepatic expressions of the vitamin D receptor and of cathelicidin significantly increased with UDCA therapy.Our results indicate that bile salts may contribute to biliary tract sterility by controlling epithelial cell innate immunity. They further suggest that in inflammatory biliary diseases, which involve bacterial factors, a strategy systematically combining UDCA with vitamin D would increase therapeutic efficacy.

Nie YF, Hu J, Yan XH.

Cross-talk between bile acids and intestinal microbiota in host metabolism and health

[J]. J Zhejiang Univ Sci B, 2015, 16(6): 436-446.

DOI:10.1631/jzus.B1400327      URL     [本文引用: 1]

Ticho AL, Malhotra P, Dudeja PK, et al.

Bile acid receptors and gastrointestinal functions

[J]. Liver Res, 2019, 3(1): 31-39.

DOI:10.1016/j.livres.2019.01.001      PMID:32368358      [本文引用: 1]

Bile acids modulate several gastrointestinal functions including electrolyte secretion and absorption, gastric emptying, and small intestinal and colonic motility. High concentrations of bile acids lead to diarrhea and are implicated in the development of esophageal, gastric and colonic cancer. Alterations in bile acid homeostasis are also implicated in the pathophysiology of irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD). Our understanding of the mechanisms underlying these effects of bile acids on gut functions has been greatly enhanced by the discovery of bile acid receptors, including the nuclear receptors: farnesoid X receptor (FXR), vitamin D receptor (VDR), pregnane X receptor (PXR), and constitutive androstane receptor (CAR); and the G protein-coupled receptors: Takeda G protein-coupled receptor (TGR5), sphingosine-1-phosphate receptor 2 (S1PR2), and muscarinic acetylcholine receptor M3 (M3R).. For example, various studies provided evidence demonstrating the anti-inflammatory effects FXR and TGR5 activation in models of intestinal inflammation. In addition, TGR5 activation in enteric neurons was recently shown to increase colonic motility, which may lead to bile acid-induced diarrhea. Interestingly, TGR5 induces the secretion of glucagon-like peptide-1 (GLP-1) from L-cells to enhance insulin secretion and modulate glucose metabolism. Because of the importance of these receptors, agonists of TGR5 and intestine-specific FXR agonists are currently being tested as an option for the treatment of diabetes mellitus and primary bile acid diarrhea, respectively. This review summarizes current knowledge of the functional roles of bile acid receptors in the gastrointestinal tract.

Teodoro JS, Rolo AP, Palmeira CM.

Hepatic FXR: key regulator of whole-body energy metabolism

[J]. Trends Endocrinol Metab, 2011, 22(11): 458-466.

DOI:10.1016/j.tem.2011.07.002      URL     [本文引用: 1]

Xu GR, Pan LX, Erickson SK, et al.

Removal of the bile acid pool upregulates cholesterol 7alpha-hydroxylase by deactivating FXR in rabbits

[J]. J Lipid Res, 2002, 43(1): 45-50.

PMID:11792721      [本文引用: 1]

We investigated the role of the orphan nuclear receptor farnesoid X receptor (FXR) in the regulation of cholesterol 7alpha-hydroxylase (CYP7A1), using an in vivo rabbit model, in which the bile acid pool, which includes high affinity ligands for FXR, was eliminated. After 7 days of bile drainage, the enterohepatic bile acid pool, in both New Zealand White and Watanabe heritable hyperlipidemic rabbits, was depleted. CYP7A1 activity and mRNA levels increased while FXR was deactivated as indicated by reduced FXR protein and changes in the expression of target genes that served as surrogate markers of FXR activation in the liver and ileum, respectively. Hepatic bile salt export pump mRNA levels and ileal bile acid-binding protein decreased while sterol 12alpha-hydroxylase and sodium/taurocholate cotransporting polypeptide mRNA levels increased in the liver. In addition, hepatic FXR mRNA levels decreased significantly. The data, taken together, indicate that FXR was deactivated when the bile acid pool was depleted such that CYP7A1 was upregulated. Further, lack of the high affinity ligand supply was associated with downregulation of hepatic FXR mRNA levels.

Lu TT, Makishima M, Repa JJ, et al.

Molecular basis for feedback regulation of bile acid synthesis by nuclear receptors

[J]. Mol Cell, 2000, 6(3): 507-515.

DOI:10.1016/s1097-2765(00)00050-2      PMID:11030331      [本文引用: 1]

The catabolism of cholesterol into bile acids is regulated by oxysterols and bile acids, which induce or repress transcription of the pathway's rate-limiting enzyme cholesterol 7alpha-hydroxylase (CYP7A1). The nuclear receptor LXRalpha binds oxysterols and mediates feed-forward induction. Here, we show that repression is coordinately regulated by a triumvirate of nuclear receptors, including the bile acid receptor, FXR; the promoter-specific activator, LRH-1; and the promoter-specific repressor, SHP. Feedback repression of CYP7A1 is accomplished by the binding of bile acids to FXR, which leads to transcription of SHP. Elevated SHP protein then inactivates LRH-1 by forming a heterodimeric complex that leads to promoter-specific repression of both CYP7A1 and SHP. These results reveal an elaborate autoregulatory cascade mediated by nuclear receptors for the maintenance of hepatic cholesterol catabolism.

Goodwin B, Jones SA, Price RR, et al.

A regulatory cascade of the nuclear receptors FXR, SHP-1, and LRH-1 represses bile acid biosynthesis

[J]. Mol Cell, 2000, 6(3): 517-526.

DOI:10.1016/s1097-2765(00)00051-4      PMID:11030332      [本文引用: 1]

Bile acids repress the transcription of cytochrome P450 7A1 (CYP7A1), which catalyzes the rate-limiting step in bile acid biosynthesis. Although bile acids activate the farnesoid X receptor (FXR), the mechanism underlying bile acid-mediated repression of CYP7A1 remained unclear. We have used a potent, nonsteroidal FXR ligand to show that FXR induces expression of small heterodimer partner 1 (SHP-1), an atypical member of the nuclear receptor family that lacks a DNA-binding domain. SHP-1 represses expression of CYP7A1 by inhibiting the activity of liver receptor homolog 1 (LRH-1), an orphan nuclear receptor that is known to regulate CYP7A1 expression positively. This bile acid-activated regulatory cascade provides a molecular basis for the coordinate suppression of CYP7A1 and other genes involved in bile acid biosynthesis.

Kerr TA, Saeki S, Schneider M, et al.

Loss of nuclear receptor SHP impairs but does not eliminate negative feedback regulation of bile acid synthesis

[J]. Dev Cell, 2002, 2(6): 713-720.

PMID:12062084      [本文引用: 1]

The in vivo role of the nuclear receptor SHP in feedback regulation of bile acid synthesis was examined. Loss of SHP in mice caused abnormal accumulation and increased synthesis of bile acids due to derepression of rate-limiting CYP7A1 and CYP8B1 hydroxylase enzymes in the biosynthetic pathway. Dietary bile acids induced liver damage and restored feedback regulation. A synthetic agonist of the nuclear receptor FXR was not hepatotoxic and had no regulatory effects. Reduction of the bile acid pool with cholestyramine enhanced CYP7A1 and CYP8B1 expression. We conclude that input from three negative regulatory pathways controls bile acid synthesis. One is mediated by SHP, and two are SHP independent and invoked by liver damage and changes in bile acid pool size.

Inagaki T, Choi M, Moschetta A, et al.

Fibroblast growth factor 15 functions as an enterohepatic signal to regulate bile acid homeostasis

[J]. Cell Metab, 2005, 2(4): 217-225.

DOI:10.1016/j.cmet.2005.09.001      PMID:16213224      [本文引用: 1]

The liver and intestine play crucial roles in maintaining bile acid homeostasis. Here, we demonstrate that fibroblast growth factor 15 (FGF15) signals from intestine to liver to repress the gene encoding cholesterol 7alpha-hydroxylase (CYP7A1), which catalyzes the first and rate-limiting step in the classical bile acid synthetic pathway. FGF15 expression is stimulated in the small intestine by the nuclear bile acid receptor FXR and represses Cyp7a1 in liver through a mechanism that involves FGF receptor 4 (FGFR4) and the orphan nuclear receptor SHP. Mice lacking FGF15 have increased hepatic CYP7A1 mRNA and protein levels and corresponding increases in CYP7A1 enzyme activity and fecal bile acid excretion. These studies define FGF15 and FGFR4 as components of a gut-liver signaling pathway that synergizes with SHP to regulate bile acid synthesis.

Chiang JYL.

Bile acid regulation of gene expression: roles of nuclear hormone receptors

[J]. Endocr Rev, 2002, 23(4): 443-463.

DOI:10.1210/er.2000-0035      PMID:12202460      [本文引用: 1]

Bile acids derived from cholesterol and oxysterols derived from cholesterol and bile acid synthesis pathways are signaling molecules that regulate cholesterol homeostasis in mammals. Many nuclear receptors play pivotal roles in the regulation of bile acid and cholesterol metabolism. Bile acids activate the farnesoid X receptor (FXR) to inhibit transcription of the gene for cholesterol 7alpha-hydroxylase, and stimulate excretion and transport of bile acids. Therefore, FXR is a bile acid sensor that protects liver from accumulation of toxic bile acids and xenobiotics. Oxysterols activate the liver orphan receptors (LXR) to induce cholesterol 7alpha-hydroxylase and ATP-binding cassette family of transporters and thus promote reverse cholesterol transport from the peripheral tissues to the liver for degradation to bile acids. LXR also induces the sterol response element binding protein-1c that regulates lipogenesis. Therefore, FXR and LXR play critical roles in coordinate control of bile acid, cholesterol, and triglyceride metabolism to maintain lipid homeostasis. Nuclear receptors and bile acid/oxysterol-regulated genes are potential targets for developing drug therapies for lowering serum cholesterol and triglycerides and treating cardiovascular and liver diseases.

Moschetta A, Bookout AL, Mangelsdorf DJ.

Prevention of cholesterol gallstone disease by FXR agonists in a mouse model

[J]. Nat Med, 2004, 10(12): 1352-1358.

DOI:10.1038/nm1138      PMID:15558057      [本文引用: 1]

Cholesterol gallstone disease is characterized by several events, including cholesterol precipitation in bile, increased bile salt hydrophobicity and gallbladder inflammation. Here, we describe the same phenotype in mice lacking the bile acid receptor, FXR. Furthermore, in susceptible wild-type mice that recapitulate human cholesterol gallstone disease, treatment with a synthetic FXR agonist prevented sequelae of the disease. These effects were mediated by FXR-dependent increases in biliary bile salt and phospholipid concentrations, which restored cholesterol solubility and thereby prevented gallstone formation. Taken together, these results indicate that FXR is a promising therapeutic target for treating or preventing cholesterol gallstone disease.

Debose-Boyd RA, Ye J.

SREBPs in lipid metabolism, insulin signaling, and beyond

[J]. Trends Biochem Sci, 2018, 43(5): 358-368.

DOI:10.1016/j.tibs.2018.01.005      URL     [本文引用: 1]

Watanabe M, Houten SM, Wang L, et al.

Bile acids lower triglyceride levels via a pathway involving FXR, SHP, and SREBP-1c

[J]. J Clin Invest, 2004, 113(10): 1408-1418.

DOI:10.1172/JCI21025      PMID:15146238      [本文引用: 1]

We explored the effects of bile acids on triglyceride (TG) homeostasis using a combination of molecular, cellular, and animal models. Cholic acid (CA) prevents hepatic TG accumulation, VLDL secretion, and elevated serum TG in mouse models of hypertriglyceridemia. At the molecular level, CA decreases hepatic expression of SREBP-1c and its lipogenic target genes. Through the use of mouse mutants for the short heterodimer partner (SHP) and liver X receptor (LXR) alpha and beta, we demonstrate the critical dependence of the reduction of SREBP-1c expression by either natural or synthetic farnesoid X receptor (FXR) agonists on both SHP and LXR alpha and LXR beta. These results suggest that strategies aimed at increasing FXR activity and the repressive effects of SHP should be explored to correct hypertriglyceridemia.

Duran-Sandoval D, Cariou B, Percevault F, et al.

The farnesoid X receptor modulates hepatic carbohydrate metabolism during the fasting-refeeding transition

[J]. J Biol Chem, 2005, 280(33): 29971-29979.

DOI:10.1074/jbc.M501931200      PMID:15899888      [本文引用: 2]

The liver plays a central role in the control of blood glucose homeostasis by maintaining a balance between glucose production and utilization. The farnesoid X receptor (FXR) is a bile acid-activated nuclear receptor. Hepatic FXR expression is regulated by glucose and insulin. Here we identify a role for FXR in the control of hepatic carbohydrate metabolism. When submitted to a controlled fasting-refeeding schedule, FXR(-/-) mice displayed an accelerated response to high carbohydrate refeeding with an accelerated induction of glycolytic and lipogenic genes and a more pronounced repression of gluconeogenic genes. Plasma insulin and glucose levels were lower in FXR(-/-) mice upon refeeding the high-carbohydrate diet. These alterations were paralleled by decreased hepatic glycogen content. Hepatic insulin sensitivity was unchanged in FXR(-/-) mice. Treatment of isolated primary hepatocytes with a synthetic FXR agonist attenuated glucose-induced mRNA expression as well as promoter activity of L-type pyruvate kinase, acetyl-CoA carboxylase 1, and Spot14. Moreover, activated FXR interfered negatively with the carbohydrate response elements regions. These results identify a novel role for FXR as a modulator of hepatic carbohydrate metabolism.

Li TG, Chiang JYL.

Regulation of bile acid and cholesterol metabolism by PPARs

[J]. PPAR Res, 2009, 2009: 501739.

[本文引用: 1]

Lambert G, Amar MJA, Guo G, et al.

The farnesoid X-receptor is an essential regulator of cholesterol homeostasis

[J]. J Biol Chem, 2003, 278(4): 2563-2570.

DOI:10.1074/jbc.M209525200      PMID:12421815      [本文引用: 1]

To address the importance of the farnesoid X-receptor (FXR; NR1H4) for normal cholesterol homeostasis, we evaluated the major pathways of cholesterol metabolism in the FXR-deficient (-/-) mouse model. Compared with wild-type, FXR(-/-) mice have increased plasma high density lipoprotein (HDL) cholesterol and a markedly reduced rate of plasma HDL cholesterol ester clearance. Concomitantly, FXR(-/-) mice exhibit reduced expression of hepatic genes involved in reverse cholesterol transport, most notably, that for scavenger receptor BI. FXR(-/-) mice also have increased: (i) plasma non-HDL cholesterol and triglyceride levels, (ii) apolipoprotein B-containing lipoprotein synthesis, and (iii) intestinal cholesterol absorption. Surprisingly, biliary cholesterol elimination was increased in FXR(-/-) mice, despite decreased expression of hepatic genes thought to be involved in this process. These data demonstrate that FXR is a critical regulator of normal cholesterol metabolism and that genetic changes affecting FXR function have the potential to be pro-atherogenic.

Zhang YQ, Yin LY, Anderson J, et al.

Identification of novel pathways that control farnesoid X receptor-mediated hypocholesterolemia

[J]. J Biol Chem, 2010, 285(5): 3035-3043.

DOI:10.1074/jbc.M109.083899      PMID:19996107      [本文引用: 1]

Farnesoid X receptor (FXR) plays important regulatory roles in bile acid, lipoprotein, and glucose homeostasis. Here, we have utilized Fxr(-/-) mice and mice deficient in scavenger receptor class B type I (SR-BI), together with an FXR-specific agonist and adenovirus expressing hepatocyte nuclear factor 4alpha or constitutively active FXR, to identify the mechanisms by which activation of FXR results in hypocholesterolemia. We identify a novel pathway linking FXR to changes in hepatic p-JNK, hepatocyte nuclear factor 4alpha, and finally SR-BI. Importantly, we demonstrate that the FXR-dependent increase in SR-BI results in both hypocholesterolemia and an increase in reverse cholesterol transport, a process involving the transport of cholesterol from peripheral macrophages to the liver for excretion into the feces. In addition, we demonstrate that FXR activation also induces an SR-BI-independent increase in reverse cholesterol transport and reduces intestinal cholesterol absorption. Together, these data indicate that FXR is a promising therapeutic target for treatment of hypercholesterolemia and coronary heart disease.

Kast HR, Nguyen CM, Sinal CJ, et al.

Farnesoid X-activated receptor induces apolipoprotein C-II transcription: a molecular mechanism linking plasma triglyceride levels to bile acids

[J]. Mol Endocrinol, 2001, 15(10): 1720-1728.

PMID:11579204      [本文引用: 1]

The farnesoid X-activated receptor (FXR; NR1H4), a member of the nuclear hormone receptor superfamily, induces gene expression in response to several bile acids, including chenodeoxycholic acid. Here we used suppression subtractive hybridization to identify apolipoprotein C-II (apoC-II) as an FXR target gene. Retroviral expression of FXR in HepG2 cells results in induction of the mRNA encoding apoC-II in response to several FXR ligands. EMSAs demonstrate that recombinant FXR and RXR bind to two FXR response elements that are contained within two important distal enhancer elements (hepatic control regions) that lie 11 kb and 22 kb upstream of the transcription start site of the apoC-II gene. A luciferase reporter gene containing the hepatic control region or two copies of the wild-type FXR response element was activated when FXR-containing cells were treated with FXR ligands. In addition, we report that hepatic expression of both apoC-II and phospholipid transfer protein mRNAs increases when mice are fed diets supplemented with cholic acid, an FXR ligand, and this induction is attenuated in FXR null mice. Finally, we observed decreased plasma triglyceride levels in mice fed cholic acid- containing diets. These results identify a mechanism whereby FXR and its ligands lower plasma triglyceride levels. These findings may have important implications in the clinical management of hyperlipidemias.

Jiang XC, Jin WJ, Hussain MM.

The impact of phospholipid transfer protein(PLTP)on lipoprotein metabolism

[J]. Nutr Metab(Lond), 2012, 9(1): 75.

[本文引用: 1]

Ma K, Saha PK, Chan L, et al.

Farnesoid X receptor is essential for normal glucose homeostasis

[J]. J Clin Invest, 2006, 116(4): 1102-1109.

DOI:10.1172/JCI25604      PMID:16557297      [本文引用: 1]

The bile acid receptor farnesoid X receptor (FXR; NR1H4) is a central regulator of bile acid and lipid metabolism. We show here that FXR plays a key regulatory role in glucose homeostasis. FXR-null mice developed severe fatty liver and elevated circulating FFAs, which was associated with elevated serum glucose and impaired glucose and insulin tolerance. Their insulin resistance was confirmed by the hyperinsulinemic euglycemic clamp, which showed attenuated inhibition of hepatic glucose production by insulin and reduced peripheral glucose disposal. In FXR-/- skeletal muscle and liver, multiple steps in the insulin signaling pathway were markedly blunted. In skeletal muscle, which does not express FXR, triglyceride and FFA levels were increased, and we propose that their inhibitory effects account for insulin resistance in that tissue. In contrast to the results in FXR-/- mice, bile acid activation of FXR in WT mice repressed expression of gluconeogenic genes and decreased serum glucose. The absence of this repression in both FXR-/- and small heterodimer partner-null (SHP-/-) mice demonstrated that the previously described FXR-SHP nuclear receptor cascade also targets glucose metabolism. Taken together, our results identify a link between lipid and glucose metabolism mediated by the FXR-SHP cascade.

Shapiro H, Kolodziejczyk AA, Halstuch D, et al.

Bile acids in glucose metabolism in health and disease

[J]. J Exp Med, 2018, 215(2): 383-396.

DOI:10.1084/jem.20171965      URL     [本文引用: 1]

Bile acids (BAs) are cholesterol-derived metabolites that facilitate the intestinal absorption and transport of dietary lipids. Recently, BAs also emerged as pivotal signaling molecules controlling glucose, lipid, and energy metabolism by binding to the nuclear hormone farnesoid X receptor (FXR) and Takeda G protein receptor 5 (TGR5) in multiple organs, leading to regulation of intestinal incretin secretion, hepatic gluconeogenesis, glycogen synthesis, energy expenditure, inflammation, and gut microbiome configuration. Alterations in BA metabolism and signaling are associated with obesity and type 2 diabetes mellitus (T2DM), whereas treatment of T2DM patients with BA sequestrants, or bariatric surgery in morbidly obese patients, results in a significant improvement in glycemic response that is associated with changes in the BA profile and signaling. Herein, we review the roles of BAs in glucose metabolism in health and disease; highlight the limitations, unknowns, and challenges in understanding the impact of BAs on the glycemic response; and discuss how this knowledge may be harnessed to develop innovative therapeutic approaches for the treatment of hyperglycemia and diabetes.

Yamagata K, Daitoku H, Shimamoto Y, et al.

Bile acids regulate gluconeogenic gene expression via small heterodimer partner-mediated repression of hepatocyte nuclear factor 4 and Foxo1

[J]. J Biol Chem, 2004, 279(22): 23158-23165.

DOI:10.1074/jbc.M314322200      PMID:15047713      [本文引用: 1]

Bile acid homeostasis is tightly controlled by the feedback mechanism in which an atypical orphan nuclear receptor (NR) small heterodimer partner (SHP) inactivates several NRs such as liver receptor homologue-1 and hepatocyte nuclear factor 4. Although NRs have been implicated in the transcriptional regulation of gluconeogenic genes, the effect of bile acids on gluconeogenic gene expression remained unknown. Here, we report that bile acids inhibit the expression of gluconeogenic genes, including glucose-6-phosphatase (G6Pase), phosphoenolpyruvate carboxykinase, and fructose 1,6-bis phosphatase in an SHP-dependent fashion. Cholic acid diet decreased the mRNA levels of these gluconeogenic enzymes, whereas those of SHP were increased. Reporter assays demonstrated that the promoter activity of phosphoenolpyruvate carboxykinase and fructose 1,6-bis phosphatase via hepatocyte nuclear factor 4, or that of G6Pase via the forkhead transcription factor Foxo1, was down-regulated by treatment with chenodeoxicholic acid and with transfected SHP. Remarkably, Foxo1 interacted with SHP in vivo and in vitro, which led to the repression of Foxo1-mediated G6Pase transcription by competition with a coactivator cAMP response element-binding protein-binding protein. These findings reveal a novel mechanism by which bile acids regulate gluconeogenic gene expression via an SHP-dependent regulatory pathway.

Zhang YQ, Lee FY, Barrera G, et al.

Activation of the nuclear receptor FXR improves hyperglycemia and hyperlipidemia in diabetic mice

[J]. Proc Natl Acad Sci USA, 2006, 103(4): 1006-1011.

DOI:10.1073/pnas.0506982103      PMID:16410358      [本文引用: 1]

Farnesoid X receptor (FXR) plays an important role in maintaining bile acid and cholesterol homeostasis. Here we demonstrate that FXR also regulates glucose metabolism. Activation of FXR by the synthetic agonist GW4064 or hepatic overexpression of constitutively active FXR by adenovirus-mediated gene transfer significantly lowered blood glucose levels in both diabetic db/db and wild-type mice. Consistent with these data, FXR null mice exhibited glucose intolerance and insulin insensitivity. We further demonstrate that activation of FXR in db/db mice repressed hepatic gluconeogenic genes and increased hepatic glycogen synthesis and glycogen content by a mechanism that involves enhanced insulin sensitivity. In view of its central roles in coordinating regulation of both glucose and lipid metabolism, we propose that FXR agonists are promising therapeutic agents for treatment of diabetes mellitus.

Fu L, John LM, Adams SH, et al.

Fibroblast growth factor 19 increases metabolic rate and reverses dietary and leptin-deficient diabetes

[J]. Endocrinology, 2004, 145(6): 2594-2603.

DOI:10.1210/en.2003-1671      PMID:14976145      [本文引用: 1]

Hormonal control of metabolic rate can be important in regulating the imbalance between energy intake and expenditure that underlies the development of obesity. In mice fed a high-fat diet, human fibroblast growth factor 19 (FGF19) increased metabolic rate [1.53 +/- 0.06 liters O(2)/h.kg(0.75) (vehicle) vs. 1.93 +/- 0.05 liters O(2)/h.kg(0.75) (FGF19); P < 0.001] and decreased respiratory quotient [0.82 +/- 0.01 (vehicle) vs. 0.80 +/- 0.01 (FGF19); P < 0.05]. In contrast to the vehicle-treated mice that gained weight (0.14 +/- 0.05 g/mouse.d), FGF19-treated mice lost weight (-0.13 +/- 0.03 g/mouse.d; P < 0.001) without a significant change in food intake. Furthermore, in addition to a reduction in weight gain, treatment with FGF19 prevented or reversed the diabetes that develops in mice made obese by genetic ablation of brown adipose tissue or genetic absence of leptin. To explore the mechanisms underlying the FGF19-mediated increase in metabolic rate, we profiled the FGF19-induced gene expression changes in the liver and brown fat. In brown adipose tissue, chronic exposure to FGF19 led to a gene expression profile that is consistent with activation of this tissue. We also found that FGF19 acutely increased liver expression of the leptin receptor (1.8-fold; P < 0.05) and decreased the expression of acetyl coenzyme A carboxylase 2 (0.6-fold; P < 0.05). The gene expression changes were consistent with the experimentally determined increase in fat oxidation and decrease in liver triglycerides. Thus, FGF19 is able to increase metabolic rate concurrently with an increase in fatty acid oxidation.

Kir S, Beddow SA, Samuel VT, et al.

FGF19 as a postprandial, insulin-independent activator of hepatic protein and glycogen synthesis

[J]. Science, 2011, 331(6024): 1621-1624.

DOI:10.1126/science.1198363      PMID:21436455      [本文引用: 1]

Fibroblast growth factor (FGF) 19 is an enterokine synthesized and released when bile acids are taken up into the ileum. We show that FGF19 stimulates hepatic protein and glycogen synthesis but does not induce lipogenesis. The effects of FGF19 are independent of the activity of either insulin or the protein kinase Akt and, instead, are mediated through a mitogen-activated protein kinase signaling pathway that activates components of the protein translation machinery and stimulates glycogen synthase activity. Mice lacking FGF15 (the mouse FGF19 ortholog) fail to properly maintain blood concentrations of glucose and normal postprandial amounts of liver glycogen. FGF19 treatment restored the loss of glycogen in diabetic animals lacking insulin. Thus, FGF19 activates a physiologically important, insulin-independent endocrine pathway that regulates hepatic protein and glycogen metabolism.

Trabelsi MS, Daoudi M, Prawitt J, et al.

Farnesoid X receptor inhibits glucagon-like peptide-1 production by enteroendocrine L cells

[J]. Nat Commun, 2015, 6: 7629.

DOI:10.1038/ncomms8629      [本文引用: 1]

Biagioli M, Carino A.

Signaling from intestine to the host: how bile acids regulate intestinal and liver immunity

[J]. Handb Exp Pharmacol, 2019, 256: 95-108.

DOI:10.1007/164_2019_225      PMID:31119464      [本文引用: 1]

Primary bile acids (BAs) are generated in the liver as the end products of cholesterol catabolism; they are then conjugated and accumulated in the gallbladder. After a meal ingestion, BAs are reversed into the duodenum to facilitate the lipid absorption. At the intestinal level, the 95% of BAs are reabsorbed and redirected into enterohepatic circulation; indeed only a small amount of them are then subjected to chemical modifications by the intestinal microbiota, which plays a very important role in the generation of secondary bile acids and in regulating host's metabolism and activity of the immune system. Behind their role in nutrients absorption, bile acids act as signaling molecules, activating several receptors, known as bile acid-activated receptors (BARs), including the farnesoid-X-receptor (FXR) and the G protein-coupled bile acid receptor 1 (GPBAR1 or Takeda G-protein receptor 5). Both receptors appear to contribute to maintain the tolerogenic state of the liver and intestine immunity. In particular, FXR and GPBAR1 are highly expressed in cells of innate immunity including intestinal and liver macrophages, dendritic cells, and natural killer T cells. In this chapter, we provide an overview on mechanisms through which FXR and GPBAR1 modulate the signaling between microbiota and intestinal and liver innate immune system. This overview could help to explain beneficial effects exerted by GPBAR1 and FXR agonists in the treatment of metabolic and immuno-mediated diseases.

Vavassori P, Mencarelli A, Renga B, et al.

The bile acid receptor FXR is a modulator of intestinal innate immunity

[J]. J Immunol, 2009, 183(10): 6251-6261.

DOI:10.4049/jimmunol.0803978      PMID:19864602      [本文引用: 1]

The farnesoid X receptor (FXR) is a bile acid-regulated nuclear receptor expressed in enterohepatic tissues. In this study we investigated whether FXR is expressed by cells of innate immunity and regulates inflammation in animal models of colitis. Acute (7 days) and chronic (8 wk) colitis were induced in wild-type and FXR(-/-) mice by intrarectal administration of trinitrobenzensulfonic acid or by 7-day administration of 5% dextran sulfate in drinking water. The results of this experiment demonstrate that FXR is expressed by and exerts counterregulatory effects on cells of innate immunity. Exposure of LPS-activated macrophages to 6-ethyl chenodeoxycholic acid (6E-CDCA; INT-747) a synthetic FXR ligand, results in a reciprocal regulation of NF-kappaB dependent-genes (TNF-alpha, IL-1beta, IL-6, COX-1, COX-2, and iNOS) and induction of SHP, a FXR-regulated gene. FXR activation stabilizes the nuclear corepressor NCoR on the NF-kappaB responsive element on the IL-1beta promoter. Colon inflammation in Crohn's disease patients and in rodent models of colitis is associated with a reduced expression of FXR mRNA. Using two rodent models of colon inflammation, we show that progression of these immune-mediated disorders is exacerbated in FXR(-/-) mice (p < 0.01). In vivo treatment with INT-747 attenuates organ injury and immune cell activation. FXR activation increased the colon expression of I-BABP, FXR, and SHP while reducing IL-1beta, IL-2, IL-6, TNF-alpha, and IFN-gamma mRNA expression and attenuating disease severity. In aggregate, these findings provide evidence that FXR is an essential component of a network of nuclear receptors that regulate intestinal innate immunity and homeostasis.

Gadaleta RM, van Erpecum KJ, Oldenburg B, et al.

Farnesoid X receptor activation inhibits inflammation and preserves the intestinal barrier in inflammatory bowel disease

[J]. Gut, 2011, 60(4): 463-472.

DOI:10.1136/gut.2010.212159      PMID:21242261      [本文引用: 1]

Inflammatory bowel disease (IBD) is characterised by chronic intestinal inflammation, resulting from dysregulation of the mucosal immune system and compromised intestinal epithelial barrier function. The bile salt, nuclear farnesoid X receptor (FXR), was recently implicated in intestinal antibacterial defence and barrier function. The aim of this study was to investigate the therapeutic potential of FXR agonists in the treatment of intestinal inflammation in complementary in vivo and in vitro models.Colitis was induced in wild-type (WT) and Fxr-null mice using dextran sodium sulfate, and in WT mice using trinitrobenzenesulfonic acid. Mice were treated with vehicle or the FXR agonist INT-747, and colitis symptoms were assessed daily. Epithelial permeability assays and cytokine expression analysis were conducted in mouse colon and enterocyte-like cells (Caco-2/HT29) treated with medium or INT-747. Inflammatory cytokine secretion was determined by ELISA in various human immune cell types.INT-747-treated WT mice are protected from DSS- and TNBS-induced colitis, as shown by significant reduction of body weight loss, epithelial permeability, rectal bleeding, colonic shortening, ulceration, inflammatory cell infiltration and goblet cell loss. Furthermore, Fxr activation in intestines of WT mice and differentiated enterocyte-like cells downregulates expression of key proinflammatory cytokines and preserves epithelial barrier function. INT-747 significantly decreases tumour necrosis factor α secretion in activated human peripheral blood mononuclear cells, purified CD14 monocytes and dendritic cells, as well as in lamina propria mononuclear cells from patients with IBD.FXR activation prevents chemically induced intestinal inflammation, with improvement of colitis symptoms, inhibition of epithelial permeability, and reduced goblet cell loss. Furthermore, FXR activation inhibits proinflammatory cytokine production in vivo in the mouse colonic mucosa, and ex vivo in different immune cell populations. The findings provide a rationale to explore FXR agonists as a novel therapeutic strategy for IBD.

Maruyama T, Miyamoto Y, Nakamura T, et al.

Identification of membrane-type receptor for bile acids(M-BAR)

[J]. Biochem Biophys Res Commun, 2002, 298(5): 714-719.

DOI:10.1016/S0006-291X(02)02550-0      URL     [本文引用: 1]

Xia XF, Francis H, Glaser S, et al.

Bile acid interactions with cholangiocytes

[J]. World J Gastroenterol, 2006, 12(22): 3553-3563.

DOI:10.3748/wjg.v12.i22.3553      URL     [本文引用: 1]

Watanabe M, Houten SM, Mataki C, et al.

Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation

[J]. Nature, 2006, 439(7075): 484-489.

DOI:10.1038/nature04330      [本文引用: 1]

Li MC, Wang SL, Li YT, et al.

Gut microbiota-bile acid crosstalk contributes to the rebound weight gain after calorie restriction in mice

[J]. Nat Commun, 2022, 13(1): 2060.

DOI:10.1038/s41467-022-29589-7      PMID:35440584      [本文引用: 1]

Calorie restriction (CR) and fasting are common approaches to weight reduction, but the maintenance is difficult after resuming food consumption. Meanwhile, the gut microbiome associated with energy harvest alters dramatically in response to nutrient deprivation. Here, we reported that CR and high-fat diet (HFD) both remodeled the gut microbiota with similar microbial composition, Parabacteroides distasonis was most significantly decreased after CR or HFD. CR altered microbiota and reprogramed metabolism, resulting in a distinct serum bile acid profile characterized by depleting the proportion of non-12α-hydroxylated bile acids, ursodeoxycholic acid and lithocholic acid. Downregulation of UCP1 expression in brown adipose tissue and decreased serum GLP-1 were observed in the weight-rebound mice. Moreover, treatment with Parabacteroides distasonis or non-12α-hydroxylated bile acids ameliorated weight regain via increased thermogenesis. Our results highlighted the gut microbiota-bile acid crosstalk in rebound weight gain and Parabacteroides distasonis as a potential probiotic to prevent rapid post-CR weight gain.© 2022. The Author(s).

Hodge RJ, Nunez DJ.

Therapeutic potential of takeda-G-protein-receptor-5(TGR5)agonists. hope or hype?

[J]. Diabetes Obes Metab, 2016, 18(5): 439-443.

DOI:10.1111/dom.12636      PMID:26818602      [本文引用: 1]

The gastrointestinal tract regulates glucose and energy metabolism, and there is increasing recognition that bile acids function as key signalling molecules in these processes. For example, bile acid changes that occur after bariatric surgery have been implicated in the effects on satiety, lipid and cholesterol regulation, glucose and energy metabolism, and the gut microbiome. In recent years, Takeda-G-protein-receptor-5 (TGR5), a bile acid receptor found in widely dispersed tissues, has been the target of significant drug discovery efforts in the hope of identifying effective treatments for metabolic diseases including type 2 diabetes, obesity, atherosclerosis, fatty liver disease and cancer. Although the benefits of targeting the TGR5 receptor are potentially great, drug development work to date has identified risks that include histopathological changes, tumorigenesis, gender differences, and questions about the translation of animal data to humans. The present article reviews the noteworthy challenges that must be addressed along the path of development of a safe and effective TGR5 agonist therapy. © 2016 John Wiley & Sons Ltd.

Thomas C, Gioiello A, Noriega L, et al.

TGR5-mediated bile acid sensing controls glucose homeostasis

[J]. Cell Metab, 2009, 10(3): 167-177.

DOI:10.1016/j.cmet.2009.08.001      PMID:19723493      [本文引用: 1]

TGR5 is a G protein-coupled receptor expressed in brown adipose tissue and muscle, where its activation by bile acids triggers an increase in energy expenditure and attenuates diet-induced obesity. Using a combination of pharmacological and genetic gain- and loss-of-function studies in vivo, we show here that TGR5 signaling induces intestinal glucagon-like peptide-1 (GLP-1) release, leading to improved liver and pancreatic function and enhanced glucose tolerance in obese mice. In addition, we show that the induction of GLP-1 release in enteroendocrine cells by 6alpha-ethyl-23(S)-methyl-cholic acid (EMCA, INT-777), a specific TGR5 agonist, is linked to an increase of the intracellular ATP/ADP ratio and a subsequent rise in intracellular calcium mobilization. Altogether, these data show that the TGR5 signaling pathway is critical in regulating intestinal GLP-1 secretion in vivo, and suggest that pharmacological targeting of TGR5 may constitute a promising incretin-based strategy for the treatment of diabesity and associated metabolic disorders.

Yanguas-Casás N, Barreda-Manso MA, Nieto-Sampedro M, et al.

TUDCA: an agonist of the bile acid receptor GPBAR1/TGR5 with anti-inflammatory effects in microglial cells

[J]. J Cell Physiol, 2017, 232(8): 2231-2245.

DOI:10.1002/jcp.25742      PMID:27987324      [本文引用: 1]

Bile acids are steroid acids found in the bile of mammals. The bile acid conjugate tauroursodeoxycholic acid (TUDCA) is neuroprotective in different animal models of stroke and neurological diseases. We have previously shown that TUDCA has anti-inflammatory effects on glial cell cultures and in a mouse model of acute neuroinflammation. We show now that microglial cells (central nervous system resident macrophages) express the G protein-coupled bile acid receptor 1/Takeda G protein-coupled receptor 5 (GPBAR1/TGR5) in vivo and in vitro. TUDCA binding to GPBAR1/TGR5 caused an increase in intracellular cAMP levels in microglia that induced anti-inflammatory markers, while reducing pro-inflammatory ones. This anti-inflammatory effect of TUDCA was inhibited by small interference RNA for GPBAR1/TGR5 receptor, as well as by treatment with a protein kinase A (PKA) inhibitor. In the mouse model of acute neuroinflammation, treating the animals with TUDCA was clearly anti-inflammatory. TUDCA biased the microglial phenotype in vivo and in vitro toward the anti-inflammatory. The bile acid receptor GPBAR1/TGR5 could be a new therapeutic target for pathologies coursing with neuroinflammation and microglia activation, such as traumatic brain injuries, stroke, or neurodegenerative diseases. TUDCA and other GPBAR1/TGR5 agonists need to be further investigated, to determine their potential in attenuating the neuropathologies associated with microglia activation. J. Cell. Physiol. 232: 2231-2245, 2017. © 2016 Wiley Periodicals, Inc.© 2016 Wiley Periodicals, Inc.

Kawamata Y, Fujii R, Hosoya M, et al.

A G protein-coupled receptor responsive to bile acids

[J]. J Biol Chem, 2003, 278(11): 9435-9440.

DOI:10.1074/jbc.M209706200      PMID:12524422      [本文引用: 1]

So far some nuclear receptors for bile acids have been identified. However, no cell surface receptor for bile acids has yet been reported. We found that a novel G protein-coupled receptor, TGR5, is responsive to bile acids as a cell-surface receptor. Bile acids specifically induced receptor internalization, the activation of extracellular signal-regulated kinase mitogen-activated protein kinase, the increase of guanosine 5'-O-3-thio-triphosphate binding in membrane fractions, and intracellular cAMP production in Chinese hamster ovary cells expressing TGR5. Our quantitative analyses for TGR5 mRNA showed that it was abundantly expressed in monocytes/macrophages in human and rabbit. Treatment with bile acids was found to suppress the functions of rabbit alveolar macrophages including phagocytosis and lipopolysaccharide-stimulated cytokine productions. We prepared a monocytic cell line expressing TGR5 by transfecting a TGR5 cDNA into THP-1 cells that did not express TGR5 originally. Treatment with bile acids suppressed the cytokine productions in the THP-1 cells expressing TGR5, whereas it did not influence those in the original THP-1 cells, suggesting that TGR5 is implicated in the suppression of macrophage functions by bile acids.

Keitel V, Donner M, Winandy S, et al.

Expression and function of the bile acid receptor TGR5 in Kupffer cells

[J]. Biochem Biophys Res Commun, 2008, 372(1): 78-84.

DOI:10.1016/j.bbrc.2008.04.171      URL     [本文引用: 1]

Pols TWH, Nomura M, Harach T, et al.

TGR5 activation inhibits atherosclerosis by reducing macrophage inflammation and lipid loading

[J]. Cell Metab, 2011, 14(6): 747-757.

DOI:10.1016/j.cmet.2011.11.006      PMID:22152303      [本文引用: 1]

The G protein-coupled receptor TGR5 has been identified as an important component of the bile acid signaling network, and its activation has been linked to enhanced energy expenditure and improved glycemic control. Here, we demonstrate that activation of TGR5 in macrophages by 6α-ethyl-23(S)-methylcholic acid (6-EMCA, INT-777), a semisynthetic BA, inhibits proinflammatory cytokine production, an effect mediated by TGR5-induced cAMP signaling and subsequent NF-κB inhibition. TGR5 activation attenuated atherosclerosis in Ldlr(-/-)Tgr5(+/+) mice but not in Ldlr(-/-)Tgr5(-/-) double-knockout mice. The inhibition of lesion formation was associated with decreased intraplaque inflammation and less plaque macrophage content. Furthermore, Ldlr(-/-) animals transplanted with Tgr5(-/-) bone marrow did not show an inhibition of atherosclerosis by INT-777, further establishing an important role of leukocytes in INT-777-mediated inhibition of vascular lesion formation. Taken together, these data attribute a significant immune modulating function to TGR5 activation in the prevention of atherosclerosis, an important facet of the metabolic syndrome.Copyright © 2011 Elsevier Inc. All rights reserved.

Cook J, Hagemann T.

Tumour-associated macrophages and cancer

[J]. Curr Opin Pharmacol, 2013, 13(4): 595-601.

DOI:10.1016/j.coph.2013.05.017      PMID:23773801      [本文引用: 1]

Our understanding of the complex roles and functions of tumour-associated myeloid cells has improved vastly over the last few years. Alternatively activated macrophages, TAMs, are an abundant part of solid and haematological malignancies and have been linked with progression, metastasis and resistance to therapy. Still, characterisation and TAM targeting is hindered by a lack of TAM specific markers, but advances in next generation technologies are rapidly increasing our understanding of the sheer diversity of myeloid differentiation and phenotypic regulation. These technologies help to shed light on the heterogeneous phenotypic states of myeloid cells within the tumour. Alternative approaches to influence the myeloid compartment within cancers surround inhibition of myeloid recruitment or 're-education' of the plastic TAM phenotype. Our knowledge continuously grows on how even 'established' therapies might influence the myeloid compartment within tumours. Now the promising results from elegant pre-clinical studies at first translate into the clinic and use combination therapies with myeloid inhibitors and standard chemotherapy. Copyright © 2013 Elsevier Ltd. All rights reserved.

Priyanka S, Medhamurthy R.

Characterization of cAMP/PKA/CREB signaling cascade in the bonnet monkey corpus luteum: expressions of inhibin-alpha and StAR during different functional status

[J]. Mol Hum Reprod, 2007, 13(6): 381-390.

PMID:17430983      [本文引用: 1]

Luteinizing hormone mediates its nuclear action primarily by activating cAMP/Protein kinase A (PKA) pathway leading to phosphorylation of cAMP response element binding (CREB) family of transcription factors. Earlier studies have documented altered cAMP responsiveness of luteal cells during maturation, and in the rhesus monkey, extinction of CREB expression following luteinization and ovulation. In the course of studies aimed at characterizing LH-cAMP signaling pathway, we serendipitously discovered that CREB is after all present in the monkey corpus luteum (CL). The present experiments were carried out to examine the PKA activity, CREB expression and RT-PCR expression of inhibin-alpha (Inh-alpha) subunit and steroidogenic acute regulatory protein (StAR) in CL obtained from a variety of model systems. PKA activity in the CL was maintained throughout the luteal phase. Messenger RNA expression by RT-PCR and Northern analyses and protein levels employing antibodies specific to total- and phospho-forms demonstrated presence of CREB in the CL. Additionally, immuno-histo/cytochemical analyses, Electrophoretic mobility shift assays and chromatin immunoprecipitation assays for Inh-alpha and StAR genes further confirmed the presence of CREB in the CL. The present study, contrary to an earlier report, demonstrates the presence of CREB (both transcript and protein) in the monkey CL. Also, analysis of expression of Inh-alpha and StAR genes (considered to be cAMP responsive), during different functional status of CL suggests that LH regulates their expression perhaps by cAMP/PKA/CREB pathway.

Sorrentino G, Perino A, Yildiz E, et al.

Bile acids signal via TGR5 to activate intestinal stem cells and epithelial regeneration

[J]. Gastroenterology, 2020, 159(3): 956-968.e8.

DOI:S0016-5085(20)34739-9      PMID:32485177      [本文引用: 1]

Renewal and patterning of the intestinal epithelium is coordinated by intestinal stem cells (ISCs); dietary and metabolic factors provide signals to the niche that control ISC activity. Bile acids (BAs), metabolites in the gut, signal nutrient availability by activating the G protein-coupled bile acid receptor 1 (GPBAR1, also called TGR5). TGR5 is expressed in the intestinal epithelium, but it is not clear how its activation affects ISCs and regeneration of the intestinal epithelium. We studied the role of BAs and TGR5 in intestinal renewal, and regulation of ISC function in mice and intestinal organoids.We derived intestinal organoids from wild-type mice and Tgr5 mice, incubated them with BAs or the TGR5 agonist INT-777, and monitored ISC function by morphologic analyses and colony-forming assays. We disrupted Tgr5 specifically in Lgr5-positive ISCs in mice (Tgr5 mice) and analyzed ISC number, proliferation, and differentiation by flow cytometry, immunofluorescence, and organoid assays. Tgr5 mice were given cholecystokinin; we measured the effects of BA release into the intestinal lumen and on cell renewal. We induced colitis in Tgr5 mice by administration of dextran sulfate sodium; disease severity was determined based on body weight, colon length, and histopathology analysis of colon biopsies.BAs and TGR5 agonists promoted growth of intestinal organoids. Administration of cholecystokinin to mice resulted in acute release of BAs into the intestinal lumen and increased proliferation of the intestinal epithelium. BAs and Tgr5 expression in ISCs were required for homeostatic intestinal epithelial renewal and fate specification, and for regeneration after colitis induction. Tgr5 mice developed more severe colitis than mice without Tgr5 disruption in ISCs. ISCs incubated with INT-777 increased activation of yes-associated protein 1 (YAP1) and of its upstream regulator SRC. Inhibitors of YAP1 and SRC prevented organoid growth induced by TGR5 activation.BAs promote regeneration of the intestinal epithelium via activation of TGR5 in ISCs, resulting in activation of SRC and YAP and activation of their target genes. Release of endogenous BAs in the intestinal lumen is sufficient to promote ISC renewal and drives regeneration in response to injury.Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.

Borgstroem B.

Influence of bile salt, ph, and time on the action of pancreatic lipase; physiological implications

[J]. J Lipid Res, 1964, 5: 522-531.

PMID:14221095      [本文引用: 1]

Gottlieb A, Bechmann L, Canbay A.

The presence and severity of nonalcoholic steatohepatitis is associated with specific changes in circulating bile acids

[J]. Ann Hepatol, 2018, 17(3): 340-341.

DOI:10.5604/01.3001.0011.7378      PMID:29735794      [本文引用: 1]

<空>

Schreuder TCMA, Marsman HA, Lenicek M, et al.

The hepatic response to FGF19 is impaired in patients with nonalcoholic fatty liver disease and insulin resistance

[J]. Am J Physiol Gastrointest Liver Physiol, 2010, 298(3): G440-G445.

DOI:10.1152/ajpgi.00322.2009      URL     [本文引用: 1]

Intestinal FGF19 has emerged as a novel endocrine regulator of hepatic bile salt and lipid metabolism. In patients with nonalcoholic fatty liver disease (NAFLD) hepatic lipid metabolism is deranged. A possible role of FGF19 in NAFLD has not been reported yet. In this study, we assessed intestinal FGF19 production and the hepatic response to FGF19 in NAFLD patients with and without insulin resistance [homeostasis model of assessment (HOMA) score ≥2.5 ( n = 12) and HOMA score &lt;2.5 ( n = 8), respectively]. To this end, NAFLD patients received a standardized oral fat challenge. Postprandial excursions of triglycerides, bile salts, and FGF19 were monitored, and plasma levels of a marker for bile salt synthesis (7α-hydroxy-4-cholesten-3-one) were determined. Fasted FGF19 levels were comparable in a control group of healthy volunteers ( n = 15) and in NAFLD patients (0.26 ± 0.28 vs. 0.18 ± 0.09 ng/ml, respectively, P = 0.94). Postprandial FGF19 levels in both controls and NAFLD patients peaked between 3–4 h and were three times higher than baseline levels. The areas under the postprandial FGF19 curve were similar in controls and in the HOMA score-based NAFLD subgroups. In NAFLD patients with HOMA score &lt;2.5, the postprandial increase in plasma FGF19 was accompanied by a lowering of plasma levels of 7α-hydroxy-4-cholesten-3-one (−30%, P = 0.015). This anticipated decline was not observed in insulin-resistant NAFLD patients (+10%, P = 0.22). In conclusion, patients with NAFLD show an unimpaired intestinal FGF19 production. However, the hepatic response to FGF19 is impaired in NAFLD patients with insulin resistance (HOMA score ≥2.5). This impaired hepatic response to FGF19 may contribute to the dysregulation of lipid homeostasis in NAFLD.

Beuers U, Trauner M, Jansen P, et al.

New paradigms in the treatment of hepatic cholestasis: from UDCA to FXR, PXR and beyond

[J]. J Hepatol, 2015, 62(1 Suppl): S25-S37.

DOI:10.1016/j.jhep.2015.02.023      URL     [本文引用: 1]

Solá S, Amaral JD, Aranha MM, et al.

Modulation of hepatocyte apoptosis: cross-talk between bile acids and nuclear steroid receptors

[J]. Curr Med Chem, 2006, 13(25): 3039-3051.

PMID:17073645      [本文引用: 1]

The efficient removal of unwanted cells, such as senescent, damaged, mutated or infected cells is crucial for the maintenance of normal liver function. In fact, apoptosis has emerged as a potential contributor to the pathogenesis of a number of hepatic disorders, such as viral hepatitis, autoimmune diseases, ethanol-induced injury, cholestasis, and hepatocellular carcinoma. In contrast to the effect of cytotoxic bile acids in the liver, ursodeoxycholic acid (UDCA) has increasingly been used for the treatment of various liver disorders. The clinical efficacy of this hydrophilic bile acid was first recognized by its use in traditional Asian medicine. However, many studies have subsequently confirmed that UDCA improves liver function by three major mechanisms of action, including protection of cholangiocytes against the cytotoxicity of hydrophobic bile acids, stimulation of hepatobiliary secretion, and inhibition of liver cell apoptosis. UDCA acts as a potent inhibitor of the classical mitochondrial pathway of apoptosis, but also interferes with alternate and upstream molecular targets such as the E2F-1/p53 pathway. Together, there is growing evidence that this hydrophilic bile acid may modulate gene expression to prevent cell death. Curiously, as a cholesterol-derived molecule, UDCA interacts with nuclear steroid receptors, such as the glucocorticoid receptor. Nuclear steroid receptors play crucial roles in mediating steroid hormone signaling involved in many biological processes, including apoptosis. Here, we review the anti-apoptotic mechanisms of UDCA in hepatic cells, and discuss a potential involvement of nuclear steroid receptors in mediating the survival effects of UDCA.

Ratziu V, de Ledinghen V, Oberti F, et al.

A randomized controlled trial of high-dose ursodesoxycholic acid for nonalcoholic steatohepatitis

[J]. J Hepatol, 2011, 54(5): 1011-1019.

DOI:10.1016/j.jhep.2010.08.030      PMID:21145828      [本文引用: 1]

Nonalcoholic steatohepatitis (NASH) is a prevalent liver disease associated with increased morbidity and mortality. Ursodeoxycholic acid (UDCA) may have antioxidant, anti-inflammatory, and antifibrotic properties and may reduce liver injury in NASH. To date, no studies have assessed the efficacy and safety of high-dose UDCA (HD-UDCA) in patients with NASH.We conducted a 12-month, randomized, double-blind, placebo-controlled multicenter trial to evaluate the efficacy and safety of HD-UDCA (28-35 mg/kg per day) in 126 patients with biopsy-proven NASH and elevated alanine aminotransferase (ALT) levels. The primary study end point was reduction in ALT levels from baseline in patients treated with HD-UDCA compared with placebo. Secondary study end points were the proportion of patients with ALT normalization, relative reduction in the scores of serum markers of fibrosis and hepatic inflammation, and safety and tolerability.HD-UDCA significantly reduced mean ALT levels -28.3% from baseline after 12 months compared with -1.6% with placebo (p<0.001). At the end of the trial, ALT levels normalized (≤35 IU/L) in 24.5% of patients treated with HD-UDCA and in 4.8% of patients who received placebo (p=0.003). Both results were not accounted for by changes in weight during the trial. HD-UDCA significantly reduced the FibroTest® serum fibrosis marker (p<0.001) compared with placebo. HD-UDCA also significantly improved markers of glycemic control and insulin resistance. There were no safety issues in this population.Treatment with HD-UDCA was safe, improved aminotransferase levels, serum fibrosis markers, and selected metabolic parameters. Studies with histologic end points are warranted.Copyright © 2011. Published by Elsevier B.V.

Wagner M, Zollner G, Trauner M.

Nuclear receptors in liver disease

[J]. Hepatology, 2011, 53(3): 1023-1034.

DOI:10.1002/hep.24148      PMID:21319202      [本文引用: 1]

Nuclear receptors are ligand-activated transcriptional regulators of several key aspects of hepatic physiology and pathophysiology. As such, nuclear receptors control a large variety of metabolic processes including hepatic lipid metabolism, drug disposition, bile acid homeostasis, as well as liver regeneration, inflammation, fibrosis, cell differentiation, and tumor formation. Derangements of nuclear receptor regulation and genetic variants may contribute to the pathogenesis and progression of liver diseases. This places nuclear receptors into the frontline for novel therapeutic approaches for a broad range of hepatic disorders and diseases including cholestatic and fatty liver disease, drug hepatotoxicity, viral hepatitis, liver fibrosis, and cancer.Copyright © 2011 American Association for the Study of Liver Diseases.

Degirolamo C, Modica S, Vacca M, et al.

Prevention of spontaneous hepatocarcinogenesis in farnesoid X receptor-null mice by intestinal-specific farnesoid X receptor reactivation

[J]. Hepatology, 2015, 61(1): 161-170.

DOI:10.1002/hep.27274      PMID:24954587      [本文引用: 1]

Farnesoid X receptor (FXR) is the master regulator of bile acid (BA) homeostasis because it controls BA synthesis, influx, efflux, and detoxification in the gut/liver axis. Deregulation of BA homeostasis has been linked to hepatocellular carcinoma (HCC), and spontaneous hepatocarcinogenesis has been observed in FXR-null mice. This dreaded liver neoplasm has been associated with both FXR gene deletion and BA-mediated metabolic abnormalities after inactivation of FXR transcriptional activity. In the present study, we addressed the hypothesis that intestinal selective FXR reactivation would be sufficient to restore the fibroblast growth factor 15 (FGF15)/cholesterol-7alpha-hydroxylase (Cyp7a1) enterohepatic axis and eventually provide protection against HCC. To this end, we generated FXR-null mice with re-expression of constitutively active FXR in enterocytes (FXR(-/-)iVP16FXR) and corresponding control mice (FXR(-/-)iVP16). In FXR-null mice, intestinal selective FXR reactivation normalized BA enterohepatic circulation along with up-regulation of intestinal FXR transcriptome and reduction of hepatic BA synthesis. At 16 months of age, intestinal FXR reactivation protected FXR-null mice from spontaneous HCC development that occurred in otherwise FXR-null mice. Activation of intestinal FXR conferred hepatoprotection by restoring hepatic homeostasis, limiting cellular proliferation through reduced cyclinD1 expression, decreasing hepatic inflammation and fibrosis (decreased signal transducer and activator of transcription 3 activation and curtailed collagen deposition).Intestinal FXR is sufficient to restore BA homeostasis through the FGF15 axis and prevent progression of liver damage to HCC even in the absence of hepatic FXR. Intestinal-selective FXR modulators could stand as potential therapeutic intervention to prevent this devastating hepatic malignancy, even if carrying a somatic FXR mutation.© 2014 by the American Association for the Study of Liver Diseases.

Mertens KL, Kalsbeek A, Soeters MR, et al.

Bile acid signaling pathways from the enterohepatic circulation to the central nervous system

[J]. Front Neurosci, 2017, 11: 617.

DOI:10.3389/fnins.2017.00617      URL     [本文引用: 2]

McMillin M, DeMorrow S.

Effects of bile acids on neurological function and disease

[J]. FASEB J, 2016, 30(11): 3658-3668.

PMID:27468758      [本文引用: 1]

Bile acids are synthesized from cholesterol and are known to be involved with the emulsification and digestion of dietary lipids and fat-soluble vitamins. Outside of this role, bile acids can act as cell signaling effectors through binding and activating receptors on both the cell membrane and nucleus. Numerous reports have investigated these signaling pathways in conditions where the liver is damaged. More recently, effort has been made to investigate the role of bile acids in diseases outside of those associated with liver damage. This review summarizes recent findings on the influences that bile acids can exert in normal neurological function and their contribution to diseases of the nervous system, with the intent of highlighting the role of these metabolites as potential players in neurological disorders.-McMillin, M., DeMorrow, S. Effects of bile acids on neurological function and disease.© FASEB.

McMillin M, Frampton G, Tobin R, et al.

TGR5 signaling reduces neuroinflammation during hepatic encephalopathy

[J]. J Neurochem, 2015, 135(3): 565-576.

DOI:10.1111/jnc.13243      PMID:26179031      [本文引用: 1]

Hepatic encephalopathy (HE) is a serious neurological complication of acute and chronic liver failure. Expression of the neurosteroid/bile acid receptor Takeda G protein-coupled receptor 5 (TGR5) has been demonstrated in the brain and is thought to be neuroprotective. However, it is unknown how TGR5 signaling can influence the progression and associated neuroinflammation of HE. HE was induced in C57Bl/6 mice via intraperitoneal injection of azoxymethane (AOM) and tissue was collected throughout disease progression. TGR5 expression was elevated in the frontal cortex following AOM injection in mice. The cellular localization of TGR5 was found in both neurons and microglia in the cortex of C57Bl/6 mice. Central infusion of the TGR5 agonist, betulinic acid, prior to AOM injection delayed neurological decline, increased cortical cyclic adenosine monophosphate concentrations, reduced microglia activation and proliferation, and reduced proinflammatory cytokine production. Betulinic acid treatment in vitro reduced the neuronal expression of chemokine ligand 2, a chemokine previously demonstrated to contribute to HE pathogenesis. Lastly, treatment of the microglia cell line EOC-20 with conditioned media from betulinic acid-treated primary neurons decreased phagocytic activity and cytokine production. Together, these data identify that activation of TGR5, which is up-regulated during HE, alleviates neuroinflammation and improves outcomes of AOM-treated mice through neuron and microglia paracrine signaling.© 2015 International Society for Neurochemistry.

Huang F, Wang TT, Lan YY, et al.

Deletion of mouse FXR gene disturbs multiple neurotransmitter systems and alters neurobehavior

[J]. Front Behav Neurosci, 2015, 9: 70.

DOI:10.3389/fnbeh.2015.00070      PMID:25870546      [本文引用: 1]

Farnesoid X receptor (FXR) is a nuclear hormone receptor involved in bile acid synthesis and homeostasis. Dysfunction of FXR is involved in cholestasis and atherosclerosis. FXR is prevalent in liver, gallbladder, and intestine, but it is not yet clear whether it modulates neurobehavior. In the current study, we tested the hypothesis that mouse FXR deficiency affects a specific subset of neurotransmitters and results in an unique behavioral phenotype. The FXR knockout mice showed less depressive-like and anxiety-related behavior, but increased motor activity. They had impaired memory and reduced motor coordination. There were changes of glutamatergic, GABAergic, serotoninergic, and norepinephrinergic neurotransmission in either hippocampus or cerebellum. FXR deletion decreased the amount of the GABA synthesis enzyme GAD65 in hippocampus but increased GABA transporter GAT1 in cerebral cortex. FXR deletion increased serum concentrations of many bile acids, including taurodehydrocholic acid, taurocholic acid, deoxycholic acid (DCA), glycocholic acid (GCA), tauro-a-muricholic acid, tauro-omega-muricholic acid, and hyodeoxycholic acid (HDCA). There were also changes in brain concentrations of taurocholic acid, taurodehydrocholic acid, tauro-w-muricholic acid, tauro-beta-muricholic acid, deoxycholic acid, and lithocholic acid (LCA). Taken together, the results from studies with FXR knockout mice suggest that FXR contributes to the homeostasis of multiple neurotransmitter systems in different brain regions and modulates neurobehavior. The effect appears to be at least partially mediated by bile acids that are known to cross the blood-brain barrier (BBB) inducing potential neurotoxicity.

Xie MH, Holcomb I, Deuel B, et al.

FGF-19, a novel fibroblast growth factor with unique specificity for FGFR4

[J]. Cytokine, 1999, 11(10): 729-735.

DOI:10.1006/cyto.1999.0485      PMID:10525310      [本文引用: 1]

We have identified a novel fibroblast growth factor, FGF-19, the most distant member of the FGF family described to date. FGF-19 is a high affinity, heparin dependent ligand for FGFR4 and is the first member of the FGF family to show exclusive binding to FGFR4. Human FGF-19 maps to chromosome 11 q13.1, a region associated with an osteoporosis-pseudoglioma syndrome of skeletal and retinal defects. FGF-19 message is expressed in several tissues including fetal cartilage, skin, and retina, as well as adult gall bladder and is overexpressed in a colon adenocarcinoma cell line.Copyright 1999 Academic Press.

Marcelin G, Jo YH, Li XS, et al.

Central action of FGF19 reduces hypothalamic AGRP/NPY neuron activity and improves glucose metabolism

[J]. Mol Metab, 2013, 3(1): 19-28.

DOI:10.1016/j.molmet.2013.10.002      URL     [本文引用: 1]

Morton GJ, Matsen ME, Bracy DP, et al.

FGF19 action in the brain induces insulin-independent glucose lowering

[J]. J Clin Invest, 2013, 123(11): 4799-4808.

DOI:10.1172/JCI70710      URL     [本文引用: 1]

Furusawa Y, Obata Y, Fukuda S, et al.

Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells

[J]. Nature, 2013, 504(7480): 446-450.

DOI:10.1038/nature12721      [本文引用: 1]

Arpaia N, Campbell C, Fan XY, et al.

Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation

[J]. Nature, 2013, 504(7480): 451-455.

DOI:10.1038/nature12726      [本文引用: 1]

Sefik E, Geva-Zatorsky N, Oh S, et al.

MUCOSAL IMMUNOLOGY. Individual intestinal symbionts induce a distinct population of RORγ+ regulatory T cells

[J]. Science, 2015, 349(6251): 993-997.

DOI:10.1126/science.aaa9420      PMID:26272906      [本文引用: 1]

T regulatory cells that express the transcription factor Foxp3 (Foxp3(+) T(regs)) promote tissue homeostasis in several settings. We now report that symbiotic members of the human gut microbiota induce a distinct T(reg) population in the mouse colon, which constrains immuno-inflammatory responses. This induction—which we find to map to a broad, but specific, array of individual bacterial species—requires the transcription factor Rorγ, paradoxically, in that Rorγ is thought to antagonize FoxP3 and to promote T helper 17 (T(H)17) cell differentiation. Rorγ's transcriptional footprint differs in colonic T(regs) and T(H)17 cells and controls important effector molecules. Rorγ, and the T(regs) that express it, contribute substantially to regulating colonic T(H)1/T(H)17 inflammation. Thus, the marked context-specificity of Rorγ results in very different outcomes even in closely related cell types.Copyright © 2015, American Association for the Advancement of Science.

Song XY, Sun XM, Oh SF, et al.

Microbial bile acid metabolites modulate gut RORγ+ regulatory T cell homeostasis

[J]. Nature, 2020, 577(7790): 410-415.

DOI:10.1038/s41586-019-1865-0      [本文引用: 1]

Makishima M, Lu TT, Xie W, et al.

Vitamin D receptor as an intestinal bile acid sensor

[J]. Science, 2002, 296(5571): 1313-1316.

DOI:10.1126/science.1070477      PMID:12016314      [本文引用: 1]

The vitamin D receptor (VDR) mediates the effects of the calcemic hormone 1alpha,25-dihydroxyvitamin D3 [1,25(OH)2D3]. We show that VDR also functions as a receptor for the secondary bile acid lithocholic acid (LCA), which is hepatotoxic and a potential enteric carcinogen. VDR is an order of magnitude more sensitive to LCA and its metabolites than are other nuclear receptors. Activation of VDR by LCA or vitamin D induced expression in vivo of CYP3A, a cytochrome P450 enzyme that detoxifies LCA in the liver and intestine. These studies offer a mechanism that may explain the proposed protective effects of vitamin D and its receptor against colon cancer.

Adachi R, Honma Y, Masuno H, et al.

Selective activation of vitamin D receptor by lithocholic acid acetate, a bile acid derivative

[J]. J Lipid Res, 2005, 46(1): 46-57.

DOI:10.1194/jlr.M400294-JLR200      PMID:15489543      [本文引用: 1]

The vitamin D receptor (VDR), a member of the nuclear receptor superfamily, mediates the biological actions of the active form of vitamin D, 1alpha,25-dihydroxyvitamin D(3). It regulates calcium homeostasis, immunity, cellular differentiation, and other physiological processes. Recently, VDR was found to respond to bile acids as well as other nuclear receptors, farnesoid X receptor (FXR) and pregnane X receptor (PXR). The toxic bile acid lithocholic acid (LCA) induces its metabolism through VDR interaction. To elucidate the structure-function relationship between VDR and bile acids, we examined the effect of several LCA derivatives on VDR activation and identified compounds with more potent activity than LCA. LCA acetate is the most potent of these VDR agonists. It binds directly to VDR and activates the receptor with 30 times the potency of LCA and has no or minimal activity on FXR and PXR. LCA acetate effectively induced the expression of VDR target genes in intestinal cells. Unlike LCA, LCA acetate inhibited the proliferation of human monoblastic leukemia cells and induced their monocytic differentiation. We propose a docking model for LCA acetate binding to VDR. The development of VDR agonists derived from bile acids should be useful to elucidate ligand-selective VDR functions.

Ishizawa M, Matsunawa M, Adachi R, et al.

Lithocholic acid derivatives act as selective vitamin D receptor modulators without inducing hypercalcemia

[J]. J Lipid Res, 2008, 49(4): 763-772.

DOI:10.1194/jlr.M700293-JLR200      PMID:18180267      [本文引用: 1]

1alpha,25-Dihydroxyvitamin D(3) [1,25(OH)(2)D(3)], a vitamin D receptor (VDR) ligand, regulates calcium homeostasis and also exhibits noncalcemic actions on immunity and cell differentiation. In addition to disorders of bone and calcium metabolism, VDR ligands are potential therapeutic agents in the treatment of immune disorders, microbial infections, and malignancies. Hypercalcemia, the major adverse effect of vitamin D(3) derivatives, limits their clinical application. The secondary bile acid lithocholic acid (LCA) is an additional physiological ligand for VDR, and its synthetic derivative, LCA acetate, is a potent VDR agonist. In this study, we found that an additional derivative, LCA propionate, is a more selective VDR activator than LCA acetate. LCA acetate and LCA propionate induced the expression of the calcium channel transient receptor potential vanilloid type 6 (TRPV6) as effectively as that of 1alpha,25-dihydroxyvitamin D(3) 24-hydroxylase (CYP24A1), whereas 1,25(OH)(2)D(3) was more effective on TRPV6 than on CYP24A1 in intestinal cells. In vivo experiments showed that LCA acetate and LCA propionate effectively induced tissue VDR activation without causing hypercalcemia. These bile acid derivatives have the ability to function as selective VDR modulators.

Staudinger JL, Goodwin B, Jones SA, et al.

The nuclear receptor PXR is a lithocholic acid sensor that protects against liver toxicity

[J]. Proc Natl Acad Sci USA, 2001, 98(6): 3369-3374.

DOI:10.1073/pnas.051551698      PMID:11248085      [本文引用: 1]

The pregnane X receptor (PXR) is the molecular target for catatoxic steroids such as pregnenolone 16alpha-carbonitrile (PCN), which induce cytochrome P450 3A (CYP3A) expression and protect the body from harmful chemicals. In this study, we demonstrate that PXR is activated by the toxic bile acid lithocholic acid (LCA) and its 3-keto metabolite. Furthermore, we show that PXR regulates the expression of genes involved in the biosynthesis, transport, and metabolism of bile acids including cholesterol 7alpha-hydroxylase (Cyp7a1) and the Na(+)-independent organic anion transporter 2 (Oatp2). Finally, we demonstrate that activation of PXR protects against severe liver damage induced by LCA. Based on these data, we propose that PXR serves as a physiological sensor of LCA, and coordinately regulates gene expression to reduce the concentrations of this toxic bile acid. These findings suggest that PXR agonists may prove useful in the treatment of human cholestatic liver disease.

Shah YM, Ma XC, Morimura K, et al.

Pregnane X receptor activation ameliorates DSS-induced inflammatory bowel disease via inhibition of NF-kappaB target gene expression

[J]. Am J Physiol Gastrointest Liver Physiol, 2007, 292(4): G1114-G1122.

DOI:10.1152/ajpgi.00528.2006      URL     [本文引用: 1]

Pregnane X receptor (PXR) expression was shown to be protective in inflammatory bowel disease (IBD). However, the mechanism by which PXR provides protection remains unclear. Wild-type and Pxr-null mice were treated with the PXR agonist pregnenolone-16α-carbonitrile or vehicle and administered 2.5% dextran sulfate sodium (DSS) in drinking water to induce IBD. Typical clinical symptoms were evaluated on a daily basis. In vivo intestinal permeability assays and proinflammatory cytokine analysis were performed. PXR agonist-treated mice were protected from DSS-induced colitis compared with vehicle-treated mice, as defined by body weight loss, diarrhea, rectal bleeding, colon length, and histology. Pregnenolone-16α-carbonitrile did not decrease the severity of IBD in Pxr-null mice. PXR agonist treatment did not increase epithelial barrier function but did decrease mRNA expression of several NF-κB target genes in a PXR-dependent manner. The present study clearly demonstrates a protective role for PXR agonist in DSS-induced IBD. The data suggest that PXR-mediated repression of NF-κB target genes in the colon is a critical mechanism by which PXR activation decreases the susceptibility of mice to DSS-induced IBD.

Campbell C, McKenney PT, Konstantinovsky D, et al.

Bacterial metabolism of bile acids promotes generation of peripheral regulatory T cells

[J]. Nature, 2020, 581(7809): 475-479.

DOI:10.1038/s41586-020-2193-0      [本文引用: 1]

Hang SY, Paik D, Yao LN, et al.

Bile acid metabolites control TH17 and Treg cell differentiation

[J]. Nature, 2019, 576(7785): 143-148.

DOI:10.1038/s41586-019-1785-z      [本文引用: 1]

Floreani A, Mangini C.

Primary biliary cholangitis: old and novel therapy

[J]. Eur J Intern Med, 2018, 47: 1-5.

DOI:S0953-6205(17)30264-9      PMID:28669591      [本文引用: 1]

Primary biliary cholangitis (PBC), formerly called primary biliary cirrhosis, is a chronic cholestatic liver disease that progresses slowly to end-stage liver disease. The first Food and Drug Administration (FDA)-approved treatment for PBC was ursodeoxycholic acid (UDCA). This treatment slows the progress of the disease, but approximatively 30-40% of patients fail to respond to UDCA. A number of options are under investigation as second line treatment. Obeticholic acid (OCA), a Farnesoid X Receptor agonist, has been approved in May 2017 by FDA for patients non responders or intolerant to UDCA. The results of a randomized, double blind, phase 3 study of OCA (mg or 10mg) compared to placebo, showed that approximatively 50% of patients reached a significant reduction in serum alkaline phosphatase, a marker predictive of disease progression, liver transplantation or death. Other emerging therapies include: agents targeting fibrosis, inflammation, or immunological response. Indeed, after 30years of UDCA therapy as unique choice for PBC patients, a number of targets, derived from a deeper knowledge of the pathophysiology of the disease, has been discovered and they offer different and new therapeutic approaches that are now under evaluation.Copyright © 2017 European Federation of Internal Medicine. Published by Elsevier B.V. All rights reserved.

Trauner M, Nevens F, Shiffman ML, et al.

Long-term efficacy and safety of obeticholic acid for patients with primary biliary cholangitis: 3-year results of an international open-label extension study

[J]. Lancet Gastroenterol Hepatol, 2019, 4(6): 445-453.

DOI:10.1016/S2468-1253(19)30094-9      URL     [本文引用: 1]

Pearson T, Caporaso JG, Yellowhair M, et al.

Effects of ursodeoxycholic acid on the gut microbiome and colorectal adenoma development

[J]. Cancer Med, 2019, 8(2): 617-628.

DOI:10.1002/cam4.2019.8.issue-2      URL     [本文引用: 1]

Ge XK, Wang AA, Ying ZX, et al.

Effects of diets with different energy and bile acids levels on growth performance and lipid metabolism in broilers

[J]. Poult Sci, 2019, 98(2): 887-895.

DOI:10.3382/ps/pey434      URL     [本文引用: 1]

Udomprasert P, Rukkwamsuk T.

Effect of an exogenous emulsifier on growth performance in weanling pigs

[J]. Kasetsart Journal, 2006, 40(3): p.652-656.

[本文引用: 1]

Jain AK, Stoll B, Burrin DG, et al.

Enteral bile acid treatment improves parenteral nutrition-related liver disease and intestinal mucosal atrophy in neonatal pigs

[J]. Am J Physiol Gastrointest Liver Physiol, 2012, 302(2): G218-G224.

DOI:10.1152/ajpgi.00280.2011      URL     [本文引用: 1]

Total parenteral nutrition (TPN) is essential for patients with impaired gut function but leads to parenteral nutrition-associated liver disease (PNALD). TPN disrupts the normal enterohepatic circulation of bile acids, and we hypothesized that it would decrease intestinal expression of the newly described metabolic hormone fibroblast growth factor-19 (FGF19) and also glucagon-like peptides-1 and -2 (GLP-1 and GLP-2). We tested the effects of restoring bile acids by treating a neonatal piglet PNALD model with chenodeoxycholic acid (CDCA). Neonatal pigs received enteral feeding (EN), TPN, or TPN + CDCA for 14 days, and responses were assessed by serum markers, histology, and levels of key regulatory peptides. Cholestasis and steatosis were demonstrated in the TPN group relative to EN controls by elevated levels of serum total and direct bilirubin and also bile acids and liver triglyceride (TG) content. CDCA treatment improved direct bilirubin levels by almost fourfold compared with the TPN group and also normalized serum bile acids and liver TG. FGF19, GLP-1, and GLP-2 were decreased in plasma of the TPN group compared with the EN group but were all induced by CDCA treatment. Intestinal mucosal growth marked by weight and villus/crypt ratio was significantly reduced in the TPN group compared with the EN group, and CDCA treatment increased both parameters. These results suggest that decreased circulating FGF19 during TPN may contribute to PNALD. Moreover, we show that enteral CDCA not only resolves PNALD but acts as a potent intestinal trophic agent and secretagogue for GLP-2.

Wu XY, Yin SN, Cheng CS, et al.

Inclusion of soluble fiber during gestation regulates gut microbiota, improves bile acid homeostasis, and enhances the reproductive performance of sows

[J]. Front Vet Sci, 2021, 8: 756910.

DOI:10.3389/fvets.2021.756910      URL     [本文引用: 1]

Interaction between the dietary fiber and the gut microbes can regulate host bile acid metabolism. This study sought to explore the effects of guar gum combined with pregelatinized waxy maize starch (GCW) in a gestation diet on reproductive performance, gut microbiota composition, and bile acid homeostasis of sows. A total of 61 large white sows were randomly grouped into the control (n = 33) and 2% GCW (n = 28) groups during gestation. GCW diet increased birth-weight of piglets, and decreased the percentage of intrauterine growth restriction (IUGR) piglets. In addition, dietary GCW reduced gut microbial diversity and modulated gut microbial composition in sows on day 109 of gestation. The relative abundance of bile salt hydrolase (BSH) gene-encoding bacteria, Lactobacillus and Bacteroides decreased after GCW administration, whereas no significant difference was observed in the fecal level of total glycine-conjugated and taurine-conjugated bile acids between the two groups. Dietary GCW increased the relative abundance of Ruminococcaceae (one of few taxa comprising 7α-dehydroxylating bacteria), which was associated with elevated fecal deoxycholic acid (DCA) in the GCW group. GCW administration lowered the concentrations of plasma total bile acid (TBA) and 7α-hydroxy-4-cholesten-3-one (C4) (reflecting lower hepatic bile acid synthesis) at day 90 and day 109 of gestation compared with the control diet. Furthermore, the levels of plasma glycoursodeoxycholic acid (GUDCA), tauroursodeoxycholic acid (TUDCA) and glycohyocholic acid (GHCA) were lower in the GCW group compared with the control group. Spearman correlation analysis showed alterations in the composition of the gut microbiota by GCW treatment was associated with improved bile acid homeostasis and reproductive performance of sows. In conclusion, GCW-induced improves bile acid homeostasis during gestation which may enhance reproductive performance of sows.

/