- Review
- Open access
- Published:
Synthesis of TUDCA from chicken bile: immobilized dual-enzymatic system for producing artificial bear bile substitute
Microbial Cell Factories volume 23, Article number: 326 (2024)
Abstract
Bear bile, a valuable animal-derived medicinal substance primarily composed of tauroursodeoxycholic acid (TUDCA), is widely distributed in the medicinal market across various countries due to its significant therapeutic potential. Given the extreme cruelty involved in bear bile extraction, researchers are focusing on developing synthetic bear bile powder as a more humane alternative. This review presents an industrially practical and environmentally friendly process for producing an artificial substitute for bear bile powder using inexpensive and readily available chicken bile powder through an immobilized 7α-,7β-HSDH dual-enzymatic syste. Current technology has facilitated the industrial production of TUDCA from Tauodeoxycholic acid (TCDCA) using chicken bile powder. The review begins by examining the chemical composition, structure, and properties of bear bile, followed by an outline of the pharmacological mechanisms and manufacturing methods of TUDCA, covering chemical synthesis and biotransformation methods, and a discussion on their respective advantages and disadvantages. Finally, the process of converting chicken bile powder into bear bile powder using an immobilized 7α-Hydroxysteroid Dehydrogenases(7α-HSDH) with 7β- Hydroxysteroid Dehydrogenases (7β-HSDH) dual-enzyme system is thoroughly explained. The main objective of this review is to propose a comprehensive strategy for the complete synthesis of artificial bear bile from chicken bile within a controlled laboratory setting.
Graphical Abstract

Introduction
Animal bile has long been utilized to treat ailments in traditional Chinese medicine(TCM). Bear bile is the most well-known animal bile-based medication. It has a more than 1,000-year therapeutic history and is well-known in many nations [1,2,3,4,5,6,7,8,9]. Bear bile is the dried gallbladder bile of the Asiatic black bear (Selenarctos thibetanus), the brown bear (Ursus arctos) [10, 11] and other bear species, obtained from the Asiatic black bear by "live bile extraction" [6, 12].Although this cruel practice has stimulated a wave of condemnation across the world [6], bear bile, as an ingredient in over 100 traditional Chinese medicinal preparations, has not been effectively replaced by synthetic ursodeoxycholic acid (UDCA) [6], bile from other animals [7, 8] (such as cattle, chickens, and pigs), or herbs from TCM [8, 13, 14] (such as Gardenia jasminoides, Scutellaria baicalensis, Coptis chinensis, Phellodendron amurense, Andrographis paniculata, and Rheum palmatum).
Bile acids, which are synthesized from cholesterol in the liver [15, 16], initially yield chenodeoxycholic acid (CDCA) and cholic acid (CA) (Fig. 1). Following this, bile acids undergo conjugation with glycine or taurine before being stored and concentrated in the gallbladder. These bile acids, synthesized in the liver, are referred to as primary bile acids. Following a meal, bile acids are released into the duodenum to aid in the digestion of dietary fats and oils as well as facilitating the absorption of lipid-soluble vitamins [17,18,19,20]. In the ileum, conjugated bile acids are reabsorbed and transported via the portal blood back to the liver, a process known as enterohepatic circulation (Fig. 2). This process conserves over 95% of the bile acid pool [15]. After being synthesized in the liver, bile acids are secreted into the capillary bile ducts via the bile-salt export pump (BSEP) located in hepatocytes [21,22,23]. Simultaneously, bile acids combine with organic anions, organic cations, and reduced glutathione to form divalent anions. These substances, along with cholesterol and phospholipids, are then transported into the bile ducts by multidrug resistance related protein 2 (MRP2) and multidrug resistance transporter1 (MDRT1), also known as MDR1 [24, 25]. Transporters located on the basolateral side of the hepatocyte basement membrane act as a pathway for the release of bilirubin and bile acids in cases of cholestasis. The primary basolateral transport systems include MRP3 and MRP4 from the multidrug resistance related protein (MRP) family, as well as organic anion transporting peptide 2 (OATP2) and organic solute transporting peptide (OSTP) also known as organic solute transporters alpha/beta (OSTα/β) [24, 26]. These transporters facilitate the movement of bile acids and other organic anions into the systemic circulation. In a healthy state, bile is stored in the gallbladder and released into the intestinal lumen post-meals [27]. It is then reabsorbed at the end of the ileum through the apical sodium-dependent bile acid transporter (ASBT) and transported to the portal vein via the basolateral OSTα/β and MRP3 [28,29,30]. Intracellularly, ileal bile acid binding protein (IBABP) helps transport bile acids to the basement membrane side and reduces their toxicity to ileal cells. MRP2 is located in the luminal membrane of small intestinal cells and is responsible for re-secreting bile acids into the intestine lumen. Once in the portal vein, hepatocytes reabsorb the bile acids through the Na+-dependent taurocholic cotransporting polypeptide (NTCP) and organic anion transport polypeptide 1 (OATP1) [31, 32], before being transported to the luminal membrane of the small intestine. OATP1 is also involved in the re-uptake of bile acids into hepatocytes.
Structure of bile acid.The basic structure of bile acids mainly consists of a steroid core and a five-carbon side chain. The steroid core is composed of three six-carbon rings and one five-carbon ring. When R1 to R8 are combined with different chemical groups, different types of bile acids are formed. For example, UDCA is formed by the isomerization of the seven-position hydroxyl group of CDCA, and different bile acids have different functions and biological activities
Bile acid biosynthesis, transport and metabolism.The synthesis pathways of bile acids involve both the classical pathway and the alternative pathway, with key enzymes such as CYP7A1 and CYP8B1. Once bile acid is produced, it is transported to the capillary bile ducts via bile salt export pumps and further moved with the assistance of various drugs. Bile acids are stored in the gallbladder, reabsorbed through ASBT, and then transported back to the portal vein via OSTα/β and MRP3 in the terminal ileum. In the enterohepatic circulation, IBABP and MRP2 play crucial roles. The synthesis and recycling of bile acids is a complex process involving multiple enzymes and transporters, which is essential for maintaining bile acid balance and bile formation in the body
Daily, approximately 70–82% of bile acids in the bile acid pool are reabsorbed via ileal active transport [32,33,34]. The presence of more hydroxyl groups in bile acids enhances transport speed, with trihydroxy bile acids showing a 6 to 8 times higher uptake rate than monohydroxy bile acids [35, 36]. Conjugated bile acids are transported 4–6 times faster than non-conjugated bile acids, and T-bound bile acids are transported more swiftly than G-bound bile acids [37]. Additionally, there is competitive inhibition among different types of bile acids during the transport process.
Passive transport of bile acids is influenced by intestinal pH and the dissociation coefficient of bile acids. Free bile acids are reabsorbed passively at a faster rate than G-bound bile acids, while ionic T-bound bile acids are primarily reabsorbed through active transport. Moreover, the presence of a higher number of hydroxyl groups leads to lower membrane permeation efficiency [38]; the arrangement of hydroxyl groups also impacts passive reabsorption, with 7α hydroxycholic acid being more readily reabsorbed passively than 7β hydroxycholic acid [39, 40]. The length of the side chain does not affect passive reabsorption.
UDCA has a long history of being used in the treatment of liver disease, with its origins dating back to the Tang Dynasty in China. It was not until 1927 that a Japanese scientist successfully isolated pure UDCA crystals from bear bile and officially named it UDCA(urso, derived from the Latin word for 'bear') [41, 42]. CDCA is the precursor of UDCA. Due to its low hepatotoxicity, Western countries have recognized its medicinal value and started producing it in large quantities. In the late 1980s, with the emergence of laparoscopic cholecystectomy, the use of UDCA gradually declined [43, 44]. However, its medicinal value was rediscovered when it was found to improve biochemical parameters in primary biliary cholangitis and slow down disease progression. Subsequently, UDCA was certified by the U.S. Food and Drug Administration (FDA) as a first-line drug for treating PBC [45].
TUDCA is a unique natural bile acid amide that is produced in the body by combining UDCA and taurine. It stands out from UDCA due to the additional taurine group, which increases its polarity. TUDCA exhibits a faster dissolution rate and higher solubility compared to UDCA. Research indicates that TUDCA is not only safer but also more effective in treating various diseases [46, 47].This compound has shown promise in various pharmaceutical applications, including its positive impact on hepatobiliary diseases, its potential for enhancing treatment outcomes in Alzheimer's and Parkinson's diseases [2, 4, 48, 49], as well as its ability to prevent diseases associated with apoptosis. TCDCA is the precursor of TUDCA, and the two form an isomer pair. Importantly, bear bile contains a relatively high content of TUDCA (23–40%), while the content of TUDCA in other animals’ bile is very low (< 1%) [50,51,52]. Interestingly, the content of TCDCA in the bile of poultry such as chicken, ducks, and geese is higher than 45% [53,54,55], while the content of their other components is very similar to that of bear bile. It is well-known that in vivo TCDCA is converted into TUDCA through a five-step process for the enterohepatic circulation of bile acid [56,57,58,59,60,61,62,63] that involves the deconjugation of TCDCA by bile salt hydrolase (BSH), the 7α- or 7β-dehydroxylation of CDCA, the ligation of UDCA and coenzyme A (CoA) thioesters by bile acid CoA ligase (BAL), and the formation of TUDCA by the catalysis of bile acid CoA:amino acid N-acyltransferases (BAT). In the past, TUDCA was prepared by chemical synthesis, and compared to chemical epimerization, biotransformation of TUDCA from TCDCA is a mild and environmentally friendly process [64,65,66,67].
This review article provides a comprehensive overview of the latest research progress on the discovery, pharmacological mechanisms and biosynthesis of UDCA and TUDCA. The focus was on the biosynthetic pathway of TUDCA prepared from chicken bile powder by a two-step enzymatic method of co-immobilizing 7α-HSDH and 7β-HSDH with chitosan. Initially, a unique dual enzyme coupling system was established to produce artificial bear bile from chicken bile, with the goal of preserving and utilizing scarce medicinal resources within the context of the enterohepatic cycle of bile acids. Subsequently, the emergence of artificial bear bile powder as a potential substitute for natural bear bile powder using readily available materials has become a prominent area of interest in current research and development. Furthermore, this study involved the identification and characterization of several 7α- and 7β-hydroxysteroid dehydrogenases (7α- and 7β-HSDH) responsible for TUDCA biosynthesis, achieved through the construction of the genome of intestinal microbial elements from black bears. The structural elucidation of 7α- and 7β-HSDH enzymes using X-ray crystallography and molecular modification techniques facilitated the discovery of high enzyme activity mutants. Moreover, the development of immobilized 7α- and 7β-HSDH enzymes using chitosan-modified epoxy resin as a carrier enhanced their thermal and cyclic stability. TCDCA was successfully converted into TUDCA from chicken bile, and industrial production has been achieved (Shanghai Kaibao Pharmaceutical Co and Chongqing Jize Biotechnology Co), laying the foundation for the potential development of bile powder alternative drugs [68,69,70].
Advances in bile acid research: mechanisms and pharmacological effects of UDCA
Bile acids are crucial for the digestion and absorption of lipids and also play a role in modulating various physiological functions in the body. UDCA, a particular bile acid known for its distinctive characteristics, has garnered considerable interest in medical research. This review highlights the latest developments in bile acid studies, particularly emphasizing the mechanisms and therapeutic effects of ursodeoxycholic acid (UDCA). UDCA has been recognized as an important therapeutic compound exhibiting various actions across several disorders. Grasping its mechanisms of action and pharmacological properties is vital for enhancing its clinical use.
Advances in bile acid research and pharmaceutical applications
Bile acids were first isolated in 1838, but their precise chemical structures were not elucidated until almost a century later [68]. The synthesis of cortisone by H. Sarett's [71,72,73] using deoxycholic acid (DCA) and the observation by rheumatologist Philip Hench [74,75,76] that cortisone alleviated symptoms of rheumatoid arthritis played pivotal roles in advancing bile acid research. Subsequent discovery that fungi could synthesize significant quantities of cortisone from plant saponins led to a decline in DCA-related studies [77,78,79]. Despite fluctuations in interest over time, bile acid research remains crucial in the pharmaceutical field. Ongoing scientific investigations continue to focus on bile acids, with the potential to pave the way for novel avenues in drug development.
Research on bile acids has seen a resurgence with advancements in chromatography and mass spectrometry technology [80,81,82,83,84]. A study revealed that CDCA effectively reduces cholesterol saturation and dissolves gallstones, sparking renewed interest in bile acids. Initially, UDCA was favored over CDCA for its ability to dissolve cholesterol stones, but the rise of laparoscopic cholecystectomy led to UDCA's decline [85]. However, UDCA's therapeutic potential was later recognized in improving biochemical indicators of primary biliary cholangitis [86,87,88] and slowing disease progression [89], establishing its enduring medicinal value.
The role of bile acids in metabolic regulation: FXR and TGR5 signaling
In recent years, research on bile acids has expanded, uncovering their role as signaling molecules that interact with the nuclear receptor farnesoid X receptor (FXR) and the membrane receptor TGR5 [90, 91]. These molecules not only regulate homeostasis but also impact various physiological activities, including sugar, fat, and energy metabolism. The mechanisms involved include blocking NTCP to prevent bile acid uptake into hepatocytes, protecting hepatocytes from bile acid-induced ER stress and mitochondrial damage, and creating cytokine receptor inhibitors to hinder neutrophil chemotaxis (Fig. 3). Obeticholic acid [91, 92], a derivative of CDCA and an FXR agonist, has been shown to significantly improve blood sugar, serum triglyceride, and total cholesterol levels [93]. Studies indicate that it reduces bile acid pools by inhibiting CYP7A1, leading to a decrease in intracellular bile acid levels and a reduced risk of triggering an inflammatory response. Additionally, modified versions of FGF19 and all-trans retinoic acid seem to play a role in inhibiting CYP7A1 function [92, 94].
Bile acid signaling and control of metabolism.Bile acids are actively reabsorbed in the intestine through ASBT, with the majority being reabsorbed in the distal ileum. ASBT shows a preference for transporting T- or G-conjugated bile acids, particularly dihydroxy bile acids. Furthermore, ASBT is negatively regulated by FXR and SHP in both humans and mice, with bile acids inhibiting ASBT expression via the FXR-SHP pathway. Additionally, FGF15 plays a role in regulating the inhibitory effects on ASBT
Traditional Chinese medicine claims that bear bile can clear heat, promote choleretics, calm the liver, and detoxify, among other effects, and may have a beneficial impact on liver protection and vision enhancement. Research indicates that UDCA, when combined with glycine or taurine, can inhibit the intestinal FXR signaling pathway [95, 96], leading to increased bile acid reabsorption and faster enterohepatic circulation. Furthermore, studies show that reduced levels of FGF15/19 [97,98,99] can enhance the expression of hepatic bile acid transporters, decrease bile acid pools, and facilitate bile acid excretion through feces, thus maintaining bile acid balance. These findings broaden our understanding of UDCA's pharmacological mechanisms and offer new avenues for future research on bile acid metabolism.
The role of UDCA in enhancing bile efflux and transport in liver cells
UDCA can enhance the function and expression of Cl-/HCO3- cotransporter AE2, leading to improved bile efflux [100]. TUDCA can elevate calcium ion levels in liver cells, triggering PKC-α activation and facilitating bile outflow [101]. Moreover, TUDCA can enhance the expression of bile transporters on the bile capillary membrane by activating MAPK. Additionally, UDCA can upregulate the expression of bile acid transporters in hepatocytes, thereby enhancing bile acid efflux [2, 102]. Overall, UDCA plays a crucial role in regulating bile efflux and bile acid transport, facilitating bile extracellular secretion and vesicle exocytosis.
The protective effect of continuous UDCA administration on liver toxicity
Accumulation of bile in the liver to a toxic concentration can result in severe damage and potentially progress to cirrhosis [103, 104]. However, Continuous administration of UDCA can improve bile acid composition, thereby reducing the toxicity of endogenous bile acids [105, 106]. This treatment can effectively improve the bile acid composition and protect the liver.
The immunomodulatory effects of UDCA in patients with primary biliary cholangitis and primary sclerosing cholangitis
Research indicates [107, 108] that the major histocompatibility complex (MHC) is expressed in liver cells of patients with primary biliary cholangitis (PBC) or primary sclerosing cholangitis (PSC), and UDCA can decrease this expression. UDCA functions by activating the glucocorticoid receptor (GR) and inhibiting nitric oxide synthase activit [109, 110]. Clinical trials [111,112,113] have demonstrated that following UDCA treatment, there is a reduction in various antibodies levels in patients, such as serum immunoglobulin M13 and anti-mitochondrial antibodies. Consequently, UDCA exhibits anti-inflammatory and immunomodulatory properties in the management of patients with PBC or PSC.
The cytoprotective effects of UDCA on cell membrane stability and apoptosis
UDCA is a compound known for its cytoprotective effects, primarily focused on maintaining cell membrane stability and anti-apoptotic properties [114,115,116]. Research indicates that UDCA can effectively counteract cell membrane damage caused by hydrophobic bile acids through potential binding to various regions of the cell membrane [117]. Furthermore, UDCA has been observed to reduce mitochondrial membrane permeability [118], leading to an anti-apoptotic impact. Both in vitro and in vivo studies have demonstrated the significant protective and anti-apoptotic capabilities of UDCA, positioning it as a promising treatment option for preventing cell membrane damage and apoptosis. Moreover, UDCA has been shown to inhibit the formation of MPTP, decrease ROS production, and elevate GSH levels, thereby enhancing the antioxidant response in liver cells [119, 120]. Additionally, UDCA can impede P53 activity and promote proteasomal degradation to reduce P53's half-life. Furthermore, TUDCA can alleviate ER stress in cells and hinder apoptosis progression. Interestingly, some studies suggest that UDCA may have a pro-apoptotic effect on liver cancer cells by inducing alkaline sphingomyelinase expression, ultimately restraining cell proliferation [121, 122]. Overall, UDCA exhibits multifaceted effects on regulating cell apoptosis and proliferation, positioning it as a potential therapeutic candidate for liver diseases and liver cancer.
Comparative study on artificial and natural bear bile powder composition and pharmacological effects
Bile acids, which are essential components of bile, are synthesized by the liver from cholesterol through two main pathways: the classical pathway and the alternative pathway [123, 124]. The key enzyme that controls the rate of bile acid synthesis in the classical pathway is cholesterol 7α-hydroxylase [125,126,127], located in the endoplasmic reticulum of hepatocytes. This pathway primarily produces CA) and CDCA.On the other hand, the alternative pathway, known as the acidic pathway, converts cholesterol to CDCA with the help of CYP27A1 and CYP7B1 enzymes [128, 129].
Currently, artificial bear bile powder is primarily prepared using chemical compounding and biological transformation methods. The chemical compounding method prepares artificial bear bile powder by mixing different chemical ingredients [130]. This method can more accurately control the proportion of ingredients, making the final product closer to the ingredients of natural bear bile. Biotransformation methods include enzymatic methods and microbial transformation methods. The enzyme catalysis method achieves the synthesis of chemical substances through enzymatic reactions [131, 132]. For instance, Wang Bochu and others [133] pointed out that chenodeoxycholic acid can be converted into UDCA under the action of 7α-hydroxysteroid dehydrogenase and oxidative coenzyme. Cholic acid and the microbial transformation method uses microorganisms to perform such transformations, such as using immobilized cells of anaerobic bacteria [134] to synthesize ursodeoxycholic acid.
Enzymatic synthesis and microbial transformation are considered safer alternatives to chemical synthesis due to their avoidance of toxic chemical reagents [135]. Moreover, these methods can establish a simulated enterohepatic circulation expression system to produce bile acid's active ingredients, resulting in higher medicinal efficacy. As a result, enzymatic synthesis and microbial transformation are generally preferred for synthesizing pharmaceutical ingredients in bile [136]. Despite enzyme synthesis being more environmentally friendly and healthier than chemical synthesis, improving enzyme catalytic performance through enzyme engineering presents a difficult and intricate challenge.
The chemical composition of artificial bear bile powder closely resembles that of natural bear bile powder, allowing its pharmacological effects to match those of traditional bear bile [137]. Research indicates that artificial bear bile powder contains similar levels of TUDCA,TCDCA other key components, leading to comparable sedative and choleretic effects as natural bear bile powder [138]. Based on pharmacodynamic study findings, it can be inferred that artificial bear bile powder can serve as a viable substitute for natural bear bile powder.
Understanding the synthesis of UDCA and TUDCA: enzyme cascade and chemical approaches in enterohepatic circulation
This review article discusses the synthetic methods of UDCA and TUDCA. UDCA is a widely utilized drug for the treatment of hepatobiliary diseases, while TUDCA exhibits distinct pharmacological activities[139]. The article elaborates on the synthesis pathways for both substances, detailing the synthesis of UDCA from animal cholic acid and steroid compounds as starting materials, as well as the chemical and biological synthesis methods for TUDCA.
The complex enzymatic cascade involved in UDCA and TUDCA biosynthesis in enterohepatic circulation
Intestinal microorganisms can provide key enzymes for the bile acid synthesis pathway-enterohepatic circulation [16]. Enterohepatic circulation represents a sophisticated physiological process involving the synthesis of primary bile acids, which are produced by hepatocytes using cholesterol as a foundational substrate [140]. Once synthesized, these primary bile acids undergo metabolism within the liver, where they are conjugated with either glycine or taurine. The resultant conjugated bile acids then move through the cellular structures known as microtubules to be stored in the gallbladder. In this organ, bile acids are concentrated and eventually released into the small intestine. These bile acids play a crucial role in aiding the digestion and absorption of dietary fats and fat-soluble vitamins by facilitating their solubilization. In the small intestine, bile acids are actively engaged in the processes of fat digestion and the absorption of essential fat-soluble vitamins. In particular, within the terminal ileum, these bile acids are taken up through active transport mechanisms. It is estimated that around 95% of the bile acids that are absorbed are subsequently transported mixed with plasma proteins back to the liver, illustrating the efficiency of enterohepatic circulation. This complex process involves numerous enzymes, including bile salt hydrolase (BSH) and several hydroxysteroid dehydrogenases (3α-HSDH, 3β-HSDH, 7α-HSDH, 7β-HSDH, 12α-HSDH, and 12β-HSDH), which perform critical roles in modifying bile acids. Specifically, BSH is responsible for the hydrolysis of conjugated bile acids into their free acid forms, while the hydroxysteroid dehydrogenases mentioned facilitate the isomerization of hydroxyl groups present in the bile acids, a series of reactions that are characterized by their reversibility. The in vivo synthesis of TUDCA nvolves a sequence of five distinct enzymatic steps, each facilitated by specific enzymes, as depicted in Fig. 4. During this process, bile salt hydrolase (BSH) acts on TCDCA to convert it into CDCA, which undergoes further enzymatic modification by 7α-HSDH and 7β-HSDH to yield UDCA. This UDCA is then transformed into TUDCA through the actions of bile acid-CoA ligase (BAL) and bile acid-N-acetyltransferase [141]. Additionally, literature suggests that 7α-HSDH exhibits activity against TCDCA in vitro, highlighting its importance. Notably, the in vitro reactions do not necessitate the involvement of cellular membrane crossing, allowing for a direct catalytic transformation of TCDCA into TUDCA through hydroxyl isomerization, irrespective of hydrolysis and independent of taurine's binding to the 7α- and 7β-HSDH enzymes [9].
Chemical synthesis method of TUDCA: mixed anhydride-phenol ester method and condensation agent method
The mixed anhydride-phenolic ester method has historically been one of the prevalent techniques for synthesizing TUDCA in prior research endeavors [142,143,144]. This method operates on a core principle that entails the initial transformation of UDCA into a mixed anhydride or an activated phenolic ester. Following this, the converted compound is then reacted with taurine, resulting in the formation of TUDCA. Despite its common use, this synthesis approach requires the employment of highly toxic reagents, including sulfoxide dichloride and trifluoroacetic anhydride, which pose significant safety hazards.Moreover, the mixed anhydride-phenolic ester method is characterized by an intricate sequence of numerous reaction steps, contributing to its complexity and inefficiency. This convoluted process often yields a relatively low overall output of the desired product, which is a considerable drawback. Additionally, the substantial generation of by-products during the reaction exacerbates the challenges faced during the subsequent separation and purification stages, further complicating the synthesis process. These factors collectively highlight the limitations of this method in the efficient and safe production of TUDCA.
The condensation agent method involves the direct combination of UDCAwith Taurine, resulting in the formation of an amide bond that ultimately synthesizes TUDCA [46, 145]. While this approach is considered straightforward in terms of procedure, it comes with significant drawbacks. One major concern is the high cost associated with the condensation agents required for the reaction. Additionally, there is a risk of side reactions occurring, particularly racemization, which can compromise both the purity and yield of the final product. These factors highlight the need for careful consideration when employing this method in the synthesis of TUDCA.
Synthesis of UDCA from androstenedione
The synthesis of UDCA, is achieved by employing steroidal compounds like androstenedione as initial raw materials [146]. This approach involves a series of multi-step chemical reactions that transform these starting materials into the desired compound. One notable advantage of this method is the use of easily accessible raw materials, which can facilitate the overall synthesis process. However, the complexity of this method cannot be understated, as it involves multiple reaction steps that require careful and precise control of the reaction conditions. Additionally, the process necessitates elaborate separation and purification techniques to ensure the final product is of high purity and quality.
Conversion of TCDCA to TUDCA in chicken bile: insights from enzyme modification and optimization in gut microbiota
TUDCA is a secondary bile acid formed from UDCA and taurine, known for its low toxicity, strong hydrophilicity, and high bioavailability [147, 148]. It plays a significant pharmacological role in addressing various diseases associated with metabolic abnormalities. Research indicates that TUDCA can enhance Alzheimer's disease [149,150,151], avert atherosclerosis [152], and manage hepatobiliary diseases [153, 154], while also alleviating intestinal inflammation [155] and retinal degeneration [156, 157], ultimately promoting overall health benefits for individuals.
The process of TUDCA synthesis in the body primarily involves several steps. Initially, liver cells utilize cholesterol to synthesize the primary bile acid CDCA, which is then combined with glycine and taurine to form GCDCA and TCDCA [158, 159]. These compounds are subsequently excreted from the digestive tract. Upon reaching the intestine, GCDCA and TCDCA are enzymatically converted back into CDCA, which is further transformed into UDCA by microorganisms [160]. UDCA then enters the bloodstream through reabsorption. Subsequently, UDCA is taken back into liver cells, where it is converted into TUDCA and GUDCA before being secreted into the digestive tract to initiate a new enterohepatic circulation process (Fig. 1).
Differences in bile acid functions among different animals are primarily due to variations in bile acid composition [161]. Bear bile powder is highly valued for its medicinal properties due to its high content of TUDCA and TCDCA, whereas poultry and chicken gall predominantly contain TCDCA. Hyodeoxycholic acid (HDCA) is a unique component found in pig gallbladders. Interestingly, our analysis revealed that TCDCA and TUDCA share a very similar structure, differing only in 7 key components [138, 162, 163]. Furthermore, the non-bile acid content of poultry chicken bile powder closely resembles that of bear bile powder. Although chicken bile powder has been historically used for its medicinal benefits, it is now less commonly utilized [164]. Therefore, repurposing discarded chicken bile powder as the primary material for producing analogues of bear bile powder offers a sustainable approach to resource utilization, effectively transforming waste into valuable substances.
TUDCA has demonstrated effectiveness in treating various diseases. The in vivo production of TUDCA involves a complex five-step process catalyzed by five enzymes [165] within the enterohepatic circulation of bile salts. In 2015, Ji Qingzhi and colleagues [68] introduced a novel in vitro method for preparing TUDCA. This method entails the direct epimerization of TCDCA to TUDCA catalyzed by immobilized 7α- HSDH with 7β-HSDH in a dual-enzyme coupling system. 7α- HSDH with 7β-HSDH were immobilized on modified chitosan microspheres using single and co-immobilization techniques. A comparison of the TUDCA yields from the two methods revealed that the batch reaction catalyzed by the dual-enzyme coupling system achieved a high TUDCA yield of 62.3%. This enzymatic technology underscores the in vitro synthesis of TUDCA via a dual-enzyme coupling system, offering potential applications for the synthesis of TUDCA and other valuable bile acid derivatives. Since the inception of in vitro TUDCA synthesis using a dual-enzyme coupling system, numerous researchers [166,167,168] have dedicated their efforts to refining and optimizing this method. In recent years, there has been significant progress in enhancing the catalytic efficiency of key enzymes, particularly focusing on the molecular structures of 7α- HSDH with 7β-HSDH to optimize their activity (Fig. 5). Through the discovery of new enzymes and molecular modifications, a more efficient dual-enzyme coupling system was developed, successfully extracted from chicken bile. TCDCA was successfully converted into TUDCA from chicken bile, and industrial production has been achieved. These accomplishments have established a strong foundation for the development of alternative medicines to replace bear bile powder.
Converting TCDCA to TUDCA in chicken bile. A Schematic diagram of two-step enzymatic conversion of TUDCA [61, 70, 165, 168]. TCDCA is catalyzedby 7α- and 7β-hydroxysteroid dehydrogenases, which facilitate an epimerization reaction of the C7 hydroxyl group, resulting in the conversion to TUDCA. By screening or molecular modification to identify 7α- and 7β-hydroxysteroid dehydrogenases with enhanced catalytic activity, and by determining their optimal reaction conditions, the conversion efficiency of TCDCA can be improved.54. B Immobilized 7α-, 7β-HSDH coupled system [68, 175]: Schematic diagram of a two-step enzymatic synthesis system: Utilizing knowledge from enzyme engineering, pharmaceutical engineering, and biochemical reaction engineering, the key enzymes involved in bile acid metabolism, namely 7α- and 7β-hydroxysteroid dehydrogenase, were successfully immobilized. A dual-enzyme coupling system comprising both 7α- and 7β-HSDH was constructed. Furthermore, optimizing the immobilization conditions can enhance the catalytic efficiency of this dual-enzyme coupling system. C Two-step enzymatic method successfully converted TUDCA into chicken bile powder: based on A and B, the optimal coupled immobilized enzymes of 7α- and 7β-HSDH were obtained. This two-step enzyme coupling system was employed to catalyze the complex substrate, chicken bile powder, resulting in a TUDCA content that closely approximates that found in bear bile powder
Metagenomic analysis of black bear fecal samples reveals novel 7α-HSDH with 7β- HSDH
The research explored the gut microbiome of black bears in China by analyzing fecal samples from regions like Heilongjiang, Sichuan, and Yunnan using metagenomic sequencing. The sequencing data is accessible in the NCBI Short Reads Archive under accession number SRP079591 [169]. This study sheds light on the microbial communities and metabolic functions in black bear feces. Scientists identified new 7α- and 7β-HSDHs in Asian black bear feces through metagenomics, expanding knowledge of their gut microbiome. Enzymes responsible for these discoveries were cloned and expressed in E. coli, revealing eight 7α-HSDHs and one 7β-HSDH. Among these, J-1–1 showed significant activity towards TCDCA, indicating its role in bile acid metabolism. Y1-b-1 (7β-HSDH) exhibited optimal pH and temperature at 9.0 and 30 °C, respectively, highlighting its efficiency with TCDCA. These HSDHs were categorized as basophilic and mesophilic enzymes, with St-2–1 identified as an acidophilic mesophilic enzyme with an optimum pH of 5.5, showcasing the diverse functionalities of these enzymes in the Asian black bear gut microbiome [9] (Fig. 5A).
Enhancing thermostability and activity of steroid dehydrogenases
The CA 7α-HSDH [10] crystal structure contains the enzyme, coenzyme NADP+, and substrate TCDCA, while the CA 7β-HSDH structure includes only the enzyme and coenzyme [163]. In a different study [170], researchers used AutoDock software to dock TUDCA to CA 7β-HSDH [171], revealing an interesting spatial orientation difference compared to TCDCA in CA 7α-HSDH [172, 173]. Molecular dynamics simulations identified flexible sites for improving thermal stability, leading to the selection of Asp201 in the LOOP structure of CA 7β-HSDH for saturation mutagenesis. Among the tested mutants, only the Asp201Cys mutant showed a significant increase in activity and Tm value. Researchers have also engineered mutants of 7α-HSDH to enhance catalytic activity, with notable success in the presence of Mg2+. Additionally, mutants like A26V, I222V, and P212A were developed to improve thermostability for industrial applications, showing promising results in activity retention and Tm value enhancement.
The study delved into the effects of St-2–2 mutations at the carboxyl terminus (C-terminal), including various mutations such as K262R, K261Q, A259L, I255Q, K262R/K261Q/A259L, and K262R/K261Q/A259L/I255Q, on enzyme function. Results indicated that the I255Q mutant displayed increased activity towards certain substrates but decreased activity towards others. Conversely, the three-site directed mutant K262R/K261Q/A259L showed enhanced activity compared to the wild type, with no significant differences observed for specific substrates. These findings suggest that modifications in the C-terminal region of SDRs members can significantly impact substrate specificity, influencing preferences for certain substrates.Moreover, certain mutants (K262R, A259L, I255Q, and K262R/K261Q/A259L) demonstrated improved thermostability, especially the I255Q mutant which exhibited higher activity levels after heat treatment compared to the wild type. Overall, the study stresses the crucial role of the C-terminal in determining substrate specificity and enzyme activity in St-2–2 mutations. It emphasizes how targeted modifications in this region could potentially improve enzymatic function and stability, offering insights into enhancing enzyme performance for various applications. Continued research in this field could lead to the development of more efficient and stable enzymes through targeted alterations in the C-terminus of SDRs members, further advancing our understanding of the impact of mutations on enzyme efficiency (Fig. 5A).
Optimization of immobilized 7α-HSDH with 7β-HSDH double enzyme carriers based on multi-parameter collaboration in microenvironment and its application
A double-enzyme-coupled system was established by researchers [11], utilizing immobilized 7α-HSDH with 7β-HSDH on carriers like chitosan and epoxy resin. Both enzymes were co-immobilized in the carrier, with 7α-HSDH catalyzing the reaction of TCDCA with NADP+ to produce T-7-KLC and NADPH, which were then converted to TUDCA and NADP+ by 7β-HSDH. This coupling of reactions allowed for the regeneration of cofactors (NADP+ and NADPH) in the process [68].
The double-enzyme-coupled system includes two forms: one consisting of a mix of immobilized 7α-HSDH microspheres and 7β-HSDH microspheres, and the other containing co-immobilized 7α- and 7β-HSDH microspheres. The latter form shows higher productivity and yield of TUDCA, partially explained by the close proximity of 7α- and 7β-HSDH, reducing diffusional limitations during the reaction. Immobilized enzyme carriers were designed with varying concentrations of chitosan-modified epoxy resins to create different microenvironments with medium hydrophobicity, crowding, and charge conditions. Manipulating these conditions can enhance the activities and thermal stabilities of immobilized 7α-HSDH and 7β-HSDH. Increasing crowding by reducing pore size of the carrier improves enzyme activity, as well as enhances thermal stability and cycling stability.In particular, among the chitosan-modified epoxy resin carriers, the catalytic efficiency of double enzyme immobilized in EP-0.5-C was found to be least affected after 7 successive reaction cycles [12]. The conversion rate of TCDCA only decreased from 85.45 ± 0.36% to 84.32 ± 0.55%, while the yield of TUDCA decreased from 55.02 ± 2.07% to 51.54 ± 0.67% [68]. This indicates that carefully controlling the microenvironment of immobilized enzymes through different carrier compositions can have a significant impact on enzyme activity and stability over multiple reaction cycles [174].
Dual-enzyme co-immobilization for efficient TCDCA conversion
In the study conducted by Ji Qingzhi and colleagues [68], three methods for immobilization were put forward: firstly, step-by-step catalysis with immobilized enzymes; secondly, mixed catalysis using immobilized enzymes; and thirdly, co-immobilization of dual enzymes for catalysis. The findings indicated that co-immobilization of dual enzymes yielded the most promising results, as assessed by the yield of TUDCA and the conversion rate of TCDCA. A comparative analysis of the three techniques—step-by-step catalysis with immobilized enzymes, mixed catalysis with immobilized enzymes, and dual-enzyme co-immobilization—showcased the following results: step-by-step catalysis achieved a conversion rate of 72.76% for TCDCA and a yield of just 22.08% for TUDCA. In comparison, the immobilized enzyme mixture and dual-enzyme co-immobilization techniques attained TCDCA conversion rates of 80.12% and 90.40%, with corresponding TUDCA yields of 41.23% and 62.49%. Therefore, the dual-enzyme co-immobilization technique is recognized as the most efficient approach for immobilizing 7α-, 7β-HSDH in facilitating the conversion of TCDCA. As shown in Fig. 5B.
Large-scale production of artificial bear bile powder using co-immobilized enzymes
The production of Artificial Bear Bile Powder (ABBP) involved using a chicken bile powder solution with 7α- and 7β-HSDHs co-immobilized on EP-0.5-C to create a bear bile powder analogue [175,176,177]. TCDCA was successfully converted into TUDCA from chicken bile, and industrial production has been achieved (Shanghai Kaibao Pharmaceutical Co and Chongqing Jize Biotechnology Co), laying the foundation for the potential development of bile powder alternative drugs [68, 178, 179]. By carefully controlling the ratio of 7α-HSDH to 7β-HSDH, a quantitative transformation of TCDCA to TUDCA was achieved in the complex substrate of chicken bile powder solution [178]. Notably, the intermediate T-7-KLCA was not detected in the reaction process, showcasing the efficiency of the transformation. Furthermore, the results of the production process showed that the levels of TCDCA, TUDCA, total protein, and bilirubin in the reaction product closely resembled those found in natural bear bile powder (NBBP) [137]. However, it was observed that the levels of total amino acids and total cholesterol in the reaction product were higher compared to NBBP. This indicates that while the production of ABBP successfully replicated the key components of natural bear bile powder, there were slight differences in certain parameters. Overall, the study demonstrated the feasibility of efficiently producing ABBP using enzymatic transformation processes. Relevant studies have demonstrated that there is no significant difference in the pharmacodynamics of ABBP and NBBP [180, 181]. Overall, the study introduced a innovative dual-enzyme system for the mass production of synthetic bear bile using chicken bile. This was achieved through a combination of technologies including metagenomics, enzyme crystallography, enzyme design, and enzyme manipulation. The synthetic bear bile derived from chicken bile is comparable to natural bear bile in terms of both chemical composition and pharmacodynamics. The advancement in biotechnology is immensely advantageous in efforts to decrease the practice of "live bear bile extraction" and to safeguard the Asian black bear population. As shown in Fig. 5C.
Conclusions
This article provides a comprehensive review of the latest research advancements concerning the discovery, pharmacological mechanisms, and biosynthesis of ursodeoxycholic acid (UDCA) and TUDCA. The study specifically investigates the biosynthetic pathway of TUDCA utilizing chicken bile powder as the raw material, and employs chitosan for the co-immobilization of 7α-hydroxysteroid dehydrogenase (7α-HSDH) and 7β-hydroxysteroid dehydrogenase (7β-HSDH) through a two-step enzymatic method. This review outlines the identification and characterization of several 7α- and 7β-HSDH enzymes that are pivotal for TUDCA biosynthesis. It highlights that, based on the X-ray crystal structures of these enzymes, molecular modification techniques have yielded mutant 7α- and 7β-HSDH enzymes exhibiting enhanced catalytic activity. Furthermore, the immobilized 7α- and 7β-HSDH enzymes, developed using chitosan-modified epoxy resin as a carrier, were employed to construct a stable and efficient dual-enzyme coupling system capable of biotransforming complex substrates in situ. A product resembling natural bear bile powder was synthesized from a chicken bile powder solution. The dual-enzyme coupling system, as described in this review, demonstrates significant efficacy in converting taurochenodeoxycholic acid (TCDCA) into TUDCA, achieving a high conversion rate while maintaining an appropriate immobilization ratio and conditions, thereby promoting the production of the target product. Finally, the process of transforming chicken bile powder into bear bile powder using the immobilized 7α-HSDH and 7β-HSDH dual-enzyme system is elaborated in detail. In conclusion, this review presents a biopreparative method based on a dual-enzyme coupling system (immobilized 7α- and 7β-HSDH) as a sustainable approach for the complete synthesis of artificial bear bile from chicken bile within a controlled laboratory environment.
Availability of data and materials
No datasets were generated or analysed during the current study.
References
Zheng M, Li YY, Wang GF, Jin JY, Wang YH, Wang TM, Yang L, Liu SY, Wu JS, Wang ZT, Ma YM. Protective effect of cultured bear bile powder against dimethylnitrosamine-induced hepatic fibrosis in rats. Biomed Pharmacother. 2019;112:108701.
Zangerolamo L, Vettorazzi JF, Rosa LRO, Carneiro EM, Barbosa HCL. The bile acid TUDCA and neurodegenerative disorders: an overview. Life Sci. 2021;272:119252.
Watanabe S, Kamei T, Tanaka K, Kawasuji K, Yoshioka T, Ohno M. Roles of bile acid conjugates and phospholipids in in vitro activation of pancreatic lipase by bear bile and cattle bile. J Ethnopharmacol. 2009;125:203–6.
Huang F, Pariante CM, Borsini A. From dried bear bile to molecular investigation: a systematic review of the effect of bile acids on cell apoptosis, oxidative stress and inflammation in the brain, across pre-clinical models of neurological, neurodegenerative and neuropsychiatric disorders. Brain Behav Immun. 2022;99:132–46.
Hinsley A, Wan AKY, Garshelis D, Hoffmann M, Hu S, Lee TM, Meginnis K, Moyle B, Qiu Y, Ruan X, Milner-Gulland EJ. Understanding why consumers in China switch between wild, farmed and synthetic bear bile products. Conserv Biol. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/cobi.13895.
Sheng X, Zhang H, Weng Q. Traditional Chinese medicine: China’s bear farms prompt public outcry. Nature. 2012;484:455.
Feng Y, Siu K, Wang N, Ng K, Tsao S, Nagamatsu T, Tong Y. Bear bile: dilemma of traditional medicinal use and animal protection. J Ethnobiol Ethnomed. 2009;5:2–2.
Sandra SA, Revitt DM, Huw J, Milan V, Monique SJS, Celia MB. Antiinflammatory and hepatoprotective medicinal herbs as potential substitutes for bear bile. Int Rev Neurobiol. 2017. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/bs.irn.2017.02.008.
Ferrandi EE, Bertolesi GM. In search of sustainable chemical processes: cloning, recombinant expression, and functional characterization of the 7α- and 7β-hydroxysteroid dehydrogenases from Clostridium absonum. Appl Microbiol Biotechnol. 2012;95:1221–33.
Zahoor B, Liu X, Wu P, Sun W, Jia X, Lv Z, Zhao X, He X, He B, Cai Q, Songer M. Activity pattern study of Asiatic black bear (Ursus thibetanus) in the Qinling Mountains, China, by using infrared camera traps. Environ Sci Pollut Res Int. 2021;28:25179–86.
Hagey LR, Crombie DL, Espinosa E, Carey MC, Hofmann AF. Ursodeoxycholic acid in the Ursidae: biliary bile acids of bears, pandas, and related carnivores. J Lipid Res. 1911;1993:34.
Sukanan D, Anthony BP. Community attitudes towards bears, bear bile use, and bear conservation in Luang Prabang, Lao PDR. J Ethnobiol Ethnomed. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13002-019-0292-5.
Wang N, Feng Y, Cheung F, Chow OY, Wang X, Su W, Tong Y. A comparative study on the hepatoprotective action of bear bile and coptidis rhizoma aqueous extract on experimental liver fibrosis in rats. BMC Complementary Altern Med. 2012. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1472-6882-12-239.
Appiah SS, Paul B, Michael H, Tetsuo K, Msj S, Chaim MB. Herbal alternatives to bear bile: effects of Scutellaria baicalensis Georgi on IL-6 promoter and CYP3A4 activities. Focus Altern Complementary Ther. 2006. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.2042-7166.2006.tb04718.x.
Wahlström A, Sayin Sama I, Marschall HU, Bäckhed F. Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism. Cell Metab. 2016;24:41–50.
Larusso NF, Korman MG, Hoffman NE, Hofmann AF. Dynamics of the enterohepatic circulation of bile acids. Postprandial serum concentrations of conjugates of cholic acid in health, cholecystectomized patients, and patients with bile acid malabsorption. N Engl J Med. 1974;291:689.
Tandon P, Rowe BH, Vandermeer B, Bain VG. The efficacy and safety of bile acid binding agents, opioid antagonists, or rifampin in the treatment of cholestasis-associated pruritus. Am J Gastroenterol. 2007;102:1528–36.
Miettinen TA. Relationship between faecal bile acids, absorption of fat and vitamin B12, and serum lipids in patients with ileal resections. Eur J Clin Investig. 1971. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1365-2362.1971.tb00557.x.
Ewa O, Eoin COB, Michael F, Ed CL, Aideen L. Bile acids induce IL-1α and drive NLRP3 inflammasome-independent production of IL-1β in murine dendritic cells. Front Immunol. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2023.1285357.
Portincasa P, Ciaula AD, Garruti G, Vacca M, Wang QH. Bile acids and GPBAR-1: dynamic interaction involving genes, environment and gut microbiome. Nutrients. 2020;12:3709.
Jan S, Carola D, Marianne W, Philippski P, Constanze W, Helmut H, Dieter H, Verena K. Assessment of bile salt export pump (BSEP) inhibition by BSEP-reactive immunoglobulins from Antibody-induced BSEP deficiency patients using a novel, cell-based assay. J Hepatol. 2017. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0168-8278(17)30633-5.
Paloma J, Loreto H, Pilar M-F, Alvarez-Doforno R, Francisca Y, Maria Carmen GD, Camarena C, Adl V, Frauca E, Gema M-B, et al. Recurrence of bile salt export pump deficiency after liver transplantation. N Engl J Med. 2009. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJMoa0901075.
Claudia F, Emmanuel Dauda D, Philipp K, Veronika M, Hubert S, Tatjana S, Reiberger T, Michael T. Loss of bile salt export pump (BSEP/ABCB11) protects mice from development of carbon tetrachloride (CCl4) induced hepatic fibrosis. J Hepatol. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1055/s-0039-1691935.
Trauner M, Boyer JL. Bile salt transporters: molecular characterization, function, and regulation. Physiol Rev. 2003;83:633.
Jun S, Jing L, Baoxiang Y, Heidi LW, Evers BM. 1099 Neurotensin deficiency improves bile acid homeostasis disrupted by high-fat diet feeding through regulation of fxr and bile acid transporters in ileum. Gastroenterology. 2020;158:214.
James JB, Kim LRB, Melina MM. Novel insights into the organic solute transporter alpha/beta, OSTα/β: from the bench to the bedside. Pharmacol Ther. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.pharmthera.2020.107542.
Kullak-Ublick GA, Becker MB. Regulation of drug and bile salt transporters in liver and intestine. Drug Metab Rev. 2003;35:305–17.
Zollner G, Marschall HU, Wagner M, Trauner M. Role of nuclear receptors in the adaptive response to bile acids and cholestasis: pathogenetic and therapeutic considerations. Mol Pharm. 2006;3:231–51.
Kruh GD, Rao AC, Haywood SJ, Belinsky MG, Craddock HL, Dawson TP. The organic solute transporter alpha-beta, Ostalpha-Ostbeta, is essential for intestinal bile acid transport and homeostasis. Proc Natl Acad Sci USA. 2008;105:3891.
Ferrebee CB, Rao A, Haywood J, Pachura K, Dawson PA. 224 organic solute transporter alpha-beta (Osta-Ostβ) protects small intestine from potential bile acid-induced injury. Gastroenterology. 2016;150:S1020.
Ogawa K, Suzuki H, Hirohashi T, Ishikawa T, Sugiyama Y. Characterization of inducible nature of MRP3 in rat liver. AJP Gastrointest Liver Physiol. 2000;278:G438-446.
Hata S, Wang P, Eftychiou N, Ananthanarayanan M, Batta A, Salen G, Pang KS, Wolkoff AW. Substrate specificities of rat oatp1 and ntcp: implications for hepatic organic anion uptake. Amjphysiolgastrointest Liver Physiol. 2003;285:G829.
Peters AM, Walters JRF. Recycling rate of bile acids in the enterohepatic recirculation as a major determinant of whole body 75SeHCAT retention. Eur J Nucl Med Mol Imaging. 2013;40:1618–21.
El-Mir MY. Effect of maternal cholestasis on biliary lipid and bile acid secretion in the infant rat. Hepatology. 1997;26:527–36.
Li T, Francl JM, Boehme S, Chiang JYL. Regulation of cholesterol and bile acid homeostasis by the CYP7A1/SREBP2/miR-33a axis. Hepatology. 2013;58:1111.
Lanzini. Intestinal absorption of the bile acid analogue 75Se-homocholic acid-taurine is increased in primary biliary cirrhosis, and reverts to normal during ursodeoxycholic acid administration. Gut. 2003;52:1371–5.
Cowen AE, Korman MG, Hofmann AF, Thomas PJ. Plasma disappearance of radioactivity after intravenous injection of labeled bile acids in man. Gastroenterology. 1975;68:1567–73.
Dietschy JM. Mechanisms for the intestinal absorption of bile acids. J Lipid Res. 1968;9:297.
Salen G, Shefer S, Setoguchi T, Mosbach EH. Bile alcohol metabolism in man. Conversion of 5β cholestane 3α, 7α, 12α, 25 tetrol to cholic acid. J Clin Investig. 1975;56:226–31.
Duane WC, Javitt NB. Conversion of 7α-hydroxycholesterol to bile acid in human subjects: is there an alternate pathway favoring cholic acid synthesis? J Lab Clin Med. 2002;139:109–15.
Tilles G, Wallach D. Ta?Eb A: Topical therapy of atopic dermatitis: Controversies from Hippocrates to topical immunomodulators. J Am Acad Dermatol. 2007;56:295–301.
Makino I, Tanaka H. From a choleretic to an immunomodulator: historical review of ursodeoxycholic acid as a medicament. J Gastroenterol Hepatol. 1998. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1440-1746.1998.tb00707.x.
Ying J, Dai S, Fu R, Hong J, Dai C, Jin Q. Effect of ursodeoxycholic acid on gallstone formation after bariatric surgery: An updated meta-analysis. Obesity. 2022;30:1170.
Diculescu M, Iacob S, Iacob R, Sclifos D, Oproiu A. Is oral bile acid dissolution a better alternative to laparoscopic cholecystectomy for patients with gallbladder stones? Ann Fundeni Hosp. 2001;6:18–27.
Kotb MA. Molecular mechanisms of ursodeoxycholic acid toxicity & side effects: ursodeoxycholic acid freezes regeneration & induces hibernation mode. Int J Mol Sci. 2012. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms13078882.
Denk GU, Maitz S, Wimmer R, Rust C, Invernizzi P, Ferdinandusse S, Kulik W, Fuchsbichler A, Fickert P, Trauner M. Conjugation is essential for the anticholestatic effect of NorUrsodeoxycholic acid in taurolithocholic acid-induced cholestasis in rat liver. Hepatology. 2010;52:1758.
Brevini T, Maes M, Webb GJ, John BV, Fuchs CD, Buescher G, Wang L, Griffiths C, Brown ML, Scott WE, et al. FXR inhibition may protect from SARS-CoV-2 infection by reducing ACE2. Nature. 2023;615:134–42.
Khalaf K, Tornese P, Cocco A, Albanese A. Tauroursodeoxycholic acid: a potential therapeutic tool in neurodegenerative diseases. Transl Neurodegener. 2022;11:33.
Cortez LM, Campeau J, Norman G, Kalayil M, Van der Merwe J, McKenzie D, Sim VL. Bile acids reduce prion conversion, reduce neuronal loss, and prolong male survival in models of prion disease. J Virol. 2015;89:7660–72.
Li L, Liu C, Mao W, Tumen B, Li P. Taurochenodeoxycholic acid inhibited AP-1 activation via stimulating glucocorticoid receptor. Molecules. 2019;24:4513.
Li L, Liu C, Liu M, Shi L, Liu Q, Guan H, Li P. Taurochenodeoxycholic acid induces apoptosis of fibroblast-like synoviocytes. Eur J Pharmacol. 2013;706:36–40.
Xiong F, Wu SG, Zhang J, Jakovli I, Li WX. Dietary bile salt types influence the composition of biliary bile acids and gut microbiota in grass carp. Front Microbiol. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fmicb.2018.02209.
Yixin Z, Jiaojiao W, Linnan L, Yamin L, Shuai S, Xu L, Shaoyong L, Zhengtao W, Li Y. Rapid identification of bear bile powder from other bile sources using chip-based nano-electrospray ionization tandem mass spectrometry. Rapid Commun Mass Spectrom. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/rcm.9326.
Yingpeng X, Li Y, Shujuan Z, Zhengtao W. Large-scale production of tauroursodeoxycholic acid products through fermentation optimization of engineered Escherichia coli cell factory. Microb Cell Fact. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12934-019-1076-2.
Jie S, Jie W, Lu Y, Li Y, Shujuan Z, Zhengtao W. Rapidly directional biotransformation of tauroursodeoxycholic acid through engineered Escherichia coli. J Ind Microbiol Biotechnol. 2017. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10295-017-1935-y.
Tao W, Moon-Sik Y, Hanchen X, Lei W, Huafeng W, Guang J. Serum bile acid profiles improve clinical prediction of nonalcoholic fatty liver in T2DM patients. J Proteome Res. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/acs.jproteome.1c00104.
Koichi S, Seiichiro K, Hiroshi S. Differential effects between tauroursodeoxycholic and taurochenodeoxycholic acids in hepatic fibrosis: an assessment by primary cultured Ito and Kupffer cells from the rat liver*. J Gastroenterol Hepatol. 1996. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1440-1746.1996.tb00290.x.
Yifei L, Mingmei S, Caiyun Z, Hongjiao X, Junmin W, Tao W, Guang J. Kaempferol attenuates nonalcoholic steatohepatitis by regulating serum and liver bile acid metabolism. Front Pharmacol. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fphar.2022.946360.
Dempsey JL, Wang D, Siginir G, Fei Q, Raftery D, Gu H, Yue Cui J. Pharmacological activation of PXR and CAR downregulates distinct bile acid-metabolizing intestinal bacteria and alters bile acid homeostasis. Toxicol Sci. 2019;168:40–60.
Azzaroli F, Mehal W, Soroka CJ, Wang L, Lee J, Crispe IN, Boyer JL. Ursodeoxycholic acid diminishes Fas-ligand-induced apoptosis in mouse hepatocytes. Hepatology. 2002;36:49–54.
Yinping P, Shuang T, Minghai Z, Fanglin A, Zhuozhou T, Lijun Z, Deshuai L, Jun T, Bochu W. A novel NADP(H)-Dependent 7alpha-HSDH: discovery and construction of substrate selectivity mutant by C-terminal truncation. Catalysts. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/catal12070781.
Ridlon JM, Devendran S, Alves JM, Doden H, Wolf PG, Pereira GV, Ly L, Volland A, Takei H, Nittono H, et al. The “in vivo lifestyle” of bile acid 7alpha-dehydroxylating bacteria: comparative genomics, metatranscriptomic, and bile acid metabolomics analysis of a defined microbial community in gnotobiotic mice. Gut Microbes. 2020;11:381–404.
Marion S, Studer N, Desharnais L, Menin L, Escrig S, Meibom A, Hapfelmeier S, Bernier-Latmani R. In vitro and in vivo characterization of Clostridium scindens bile acid transformations. Gut Microbes. 2019;10:481–503.
Studer N, Desharnais L, Beutler M, Brugiroux S, Terrazos MA, Menin L, Schurch CM, McCoy KD, Kuehne SA, Minton NP, et al. Functional intestinal bile acid 7alpha-dehydroxylation by clostridium scindens associated with protection from clostridium difficile infection in a gnotobiotic mouse model. Front Cell Infect Microbiol. 2016;6:191.
Ridlon JM, Kang DJ, Hylemon PB. Bile salt biotransformations by human intestinal bacteria. J Lipid Res. 2006;47:241–59.
Reed AD, Theriot CM. Contribution of inhibitory metabolites and competition for nutrients to colonization resistance against clostridioides difficile by commensal clostridium. Microorganisms. 2021;9:371.
Doden H, Sallam LA, Devendran S, Ly L, Doden G, Daniel SL, Alves JMP, Ridlon JM. Metabolism of oxo-bile acids and characterization of recombinant 12alpha-hydroxysteroid dehydrogenases from bile acid 7alpha-dehydroxylating human gut bacteria. Appl Environ Microbiol. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.1128/AEM.00235-18.
Qingzhi J, Jun T, Lingyu Z, Deshuai L, Bochu W. Preparing tauroursodeoxycholic acid (TUDCA) using a double-enzyme-coupled system. Biochem Eng J. 2016. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bej.2015.08.005.
Youchao Q, Linkai S, Guozhen D, Yonggui M, Peifeng L. Taurochenodeoxycholic acid increases cAMP content via specially interacting with bile acid receptor TGR5. Molecules. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/molecules26237066.
Yinping P, Shuang T, Liancai Z, Deshuai L, Jun T, Bochu W. Design of St-2–2 7α-HSDH mutants for altering substrate preference and thermostability. Mol Catal. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.mcat.2023.113423.
Sarett LH. Partial synthesis OF pregnene-4-TRIOL-17(β),20(β),21-DIONE-3,11 AND PREGNENE-4-DIOL-17(β),21-TRIONE-3,11,20 monoacetate. J Biol Chem. 1946;162:601–31.
Small DM. Recent advances in bile acid research. New York: Raven Press; 1986. p. 327.
Jooho P, Jeong Uk C, Kwangmeyung K, Youngro B. Bile acid transporter mediated endocytosis of oral bile acid conjugated nanocomplex. Biomaterials 2017;147:145-54.
Neeck G. Fifty years of experience with cortisone therapy in the study and treatment of rheumatoid arthritis. Ann N Y Acad Sci. 2010;966:28–38.
Platt WD, Steinberg IH. Prednisone alone and in combination with salicylates and phenylbutazone in the treatment of rheumatoid arthritis. N Engl J Med. 1957;256:823–7.
Glyn JH. The discovery of cortisone: a personal memory. Bmj British Med J. 1998;317:822–3.
Okeke CN, Gugnani HC. Studies on pathogenic dematiaceous fungi. 1. Isolation from natural sources. Mycopathologia. 1986;94:19–25.
Gugnani HC, Obiefuna MN, Ikerionwu SE. Studies on pathogenic dematiaceous fungi, II. Pathogenicity of fonsecaea pedrosoi and phialophora verrucosa for laboratory mice: untersuchungen über pathogene dematiaceen II. Pathogenitt von fonsecaea pedrosoi und phialophora verrucosa bei labormusen. Mycoses. 1986. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1439-0507.1986.tb03952.x.
Felger CE, Lorraine F. Experimental cerebral chromoblastomycosis. J Infect Dis. 1962;111:1–7.
Thistle JL, Schoenfield LJ. Induced alterations in composition of bile of persons having cholelithiasis. Gastroenterology. 1971;61:488–96.
Duncan Bell G, Whitney B, Hermon Dowling R. Gallstone dissolution in man using chenodeoxycholic acid. Lancet. 1972;300:1213–6.
Danzinger RG, Hofmann AF, Schoenfield LJ, Thistle JL. Dissolution of cholesterol gallstones by chenodeoxycholic acid. N Engl J Med. 1972;286:1–8.
Waters B. The antibiotic action of saponin II I. Saponins as plant fungistatic compounds. Planta. 1968;79:77–83.
Massoumi H, Pulicottil M, Kokkat A, Ricci M, Kiyici N, Hertan H. Triple trouble after laparoscopic cholecystectomy: dropped stone, bile leak and abscess. Am J Gastroenterol. 2004;99:S196.
Sackmann M, Pauletzki J, Aydemir U, Holl J, Sauerbruch T, Hasford J, Paumgartner G. Efficacy and safety of ursodeoxycholic acid for dissolution of gallstone fragments: comparison with the combination of ursodeoxycholic acid and chenodeoxycholic acid. Hepatology. 2010;14:1136–41.
Poupon RE, Chrétien Y, Poupon R. Paumgartner : serum bile acids in primary biliary cirrhosis: effect of ursodeoxycholic acid therapy. Hepatology. 1993;17:599–604.
Leuschner U, Kurtz W. Treatment of primary biliary cirrhosis and cholestatic disorders with ursodeoxycholic acid. Lancet. 1987;2:508.
Chazouillères O, Poupon R, Capron JP, Metman EH, Dhumeaux D, Amouretti M, Couzigou P, Labavle D, Trinchet JC. Is ursodeoxycholic acid an effective treatment for primary sclerosing cholangitis? J Hepatol. 1989;9:S138–S138.
Fujinaga Y, Namisaki T, Moriya K, Kitade M, Kawaratani H, Kaji K, Okura Y, Seki K, Takaya H, Sawada Y. Biochemical response to ursodeoxycholic acid predicts histologic primary biliary cholangitis progression. Hepatology. 2017;66:196.
Wang H, Chen J, Hollister K, Sowers LC, Forman BM. Endogenous bile acids are ligands for the nuclear receptor FXR/BAR. Mol Cell. 1999;3:543.
Hameed B, Terrault NA, Gill RM, Loomba R, Chalasani N, Hoofnagle JH, Van Natta ML, Crn N. Clinical and metabolic effects associated with weight changes and obeticholic acid in non-alcoholic steatohepatitis. Aliment Pharmacol Ther. 2018;47:645.
Lefebvre P, Cariou B, Lien F, Kuipers F, Staels B. Role of bile acids and bile acid receptors in metabolic regulation. Physiol Rev. 2009;89:147–91.
Liu L, Liu Z, Li H, Cao Z, Liu Y. Naturally occurring TPE-CA maintains gut microbiota and bile acids homeostasis via FXR signaling modulation of the liver-gut axis. Front Pharmacol. 2020;11:12.
Staels B, Fonseca VA. Bile acids and metabolic regulation: mechanisms and clinical responses to bile acid sequestration. Diabetes Care. 2009. https://doiorg.publicaciones.saludcastillayleon.es/10.2337/dc09-S355.
Modica S, Murzilli S, Salvatore L, Schmidt DR, Moschetta A. Nuclear bile acid receptor FXR protects against intestinal tumorigenesis. Cancer Res. 2008. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.CAN-08-1791.
Zhao Z, Yang LL, Wang QL, Du JF, Zheng ZG, Jiang Y, Li P, Li HJ. Baohuoside I inhibits FXR signaling pathway to interfere with bile acid homeostasis via targeting ER α degradation. Cell Biol Toxicol. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10565-022-09737-x.
Vergnes L, Lee JM, Chin RG, Auwerx J, Reue K. Diet1 functions in the FGF15/19 enterohepatic signaling axis to modulate bile acid and lipid levels. Cell Metab. 2013;17:916–28.
Hartmann P, Hochrath K, Horvath A, Chen P, Seebauer CT, Llorente C, Wang L, Alnouti Y, Fouts DE, StRkel P. Modulation of the intestinal bile acid-FXR-FGF15 axis improves alcoholic liver disease in mice. Hepatology. 2017;67:2150.
Wang Y, Gunewardena S, Li F, Matye DJ, Li T. An FGF15/19-TFEB regulatory loop controls hepatic cholesterol and bile acid homeostasis. Nat Commun. 2020;11:3612.
Ikegami T, Matsuzaki Y. Ursodeoxycholic acid: mechanism of action and novel clinical applications. Hepatol Res. 2008. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/j.1872-034X.2007.00297.x.
Zhang Y, Jiang R, Zheng X, Lei S, Huang F, Xie G, Kwee S, Yu H, Farrar C, Sun B. Ursodeoxycholic acid accelerates bile acid enterohepatic circulation. British J Pharmacol. 2019;176:2848.
Barone M, Francavilla A, Polimeno L, Ierardi E, Romanelli D, Berloco P, Leo AD, Panella C. Modulation of rat hepatocyte proliferation by bile salts: in vitro and in vivo studies. Hepatology. 1996;23:1159.
Nowak G, Noren U, Marschall HU, Moller L, Wernerson A, Ericzon B. Protective effect of UDCA on rat liver against ischemia/reperfusion injury. J Hepatol. 2003;38:1.
Fickert P. Ursodeoxycholic acid (UDCA) feeding aggravates liver injury in bile duct-ligated and MDR2 kockout mice. J Hepatol. 2001;34:182.
Bidault-Jourdainne V, Merlen G, Gnon I, Garcin I, Tordjmann T. TGR5 controls bile acid composition and gallbladder function to protect the liver from bile acid overload. JHEP Rep. 2020;56:25.
Kimura A, Yoneda M, Nakamura K, Tamori K, Kato T, Akiyama K, Makino I. Metabolisms of N-acetylglucosaminides bile acids during ursodeoxycholic acid (UDCA) treatment in patients with liver diseases. Gastroenterology. 1995;108:A1097.
Miloshevski M, Vladimir S, Trajanovski D, Vasilevski V, Joksimovic N. Effect of long-term therapy of cholestatic liver diseases with ursodeoxycholic acid. Gastroenterology. 2000. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/S0016-5085(00)81780-1.
Festi D, Montagnani M, Azzaroli F, Lodato F, Mazzella G, Roda A, Biase AR, Roda E, Simoni P, Colecchia A. Clinical efficacy and effectiveness of ursodeoxycholic acid in cholestatic liver diseases. Curr Clin Pharmacol. 2007. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/157488407780598171.
Chazouillères O, Poupon R, Capron JP, Metman EH, Dhumeaux D, Amouretti M, Couzigou P, Labayle D, Trinchet JC. Ursodeoxycholic acid for primary sclerosing cholangitis. J Hepatol. 1990;11:120–3.
Olsson R, Boberg KM, de Muckadell OS, Lindgren S, Hultcrantz R, Folvik G, Bell H, Gangsøy-Kristiansen M, Matre J, Rydning A, Wikman O. High-dose ursodeoxycholic acid in primary sclerosing cholangitis: a 5-year multicenter, randomized, controlled study. Digest World Core Med J. 2006;129:1464.
Corpechot C, Carrat F, Bonnand AM, Poupon RE, Poupon R. The effect of ursodeoxycholic acid therapy on liver fibrosis progression in primary biliary cirrhosis. Hepatology. 2000;32:1196–9.
Chung SW, Lee JH, Kim MA, Leem G, Kim SW, Chang Y, Lee HY, Yoon JS, Park JY, Lee YB. Additional fibrate treatment in UDCA-refractory PBC patients. Liver Int. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/liv.14165.
Aliya G, Joseph JL, Brian DJ, Elizabeth JA, Craig L, Konstantinos L. Mo1004 predictors and durability of biochemical response to ursodeoxycholic acid (UDCA) in patients with primary biliary cirrhosis (PBC): a long-term follow-up study. Gastroenterology 2015;148(4):S-1061.
Tanikawa K, Shotaro S, Etsuo N, Hiroshi A. Cytoprotective effects of cytotoxic chenodeoxycholic acid (CDCA), ursodeoxycholic acid (UDCA), polyene phosphatidylcholine (PPC), and prostaglandin E1 in cultured hepatocytes. Hepatology 1984.
Amaral JD, Viana RJS, Ramalho RM, Steer CJ, Rodrigues CMP. Bile acids: regulation of apoptosis by ursodeoxycholic acid. J Lipid Res. 2009;50:1721.
Fickert P, Wagner M, Marschall HU, Fuchsbichler A, Zollner G, Tsybrovskyy O, Zatloukal K, Liu J, Waalkes MP, Cover C. 24-norUrsodeoxycholic acid is superior to ursodeoxycholic acid in the treatment of sclerosing cholangitis in Mdr2 (Abcb4) knockout mice. Gastroenterology. 2006;130:465–81.
Peters SMA, Jong MDD, Bindels RJM, Os CHV, Wetzels JFM. Effects of renal cytoprotective agents on erythrocyte membrane stability. Life ences. 1998;63:975–83.
Yu H, Fu QR, Huang ZJ, Lin JY, Chen QX, Wang Q, Shen DY. Apoptosis induced by ursodeoxycholic acid in human melanoma cells through the mitochondrial pathway. Oncol Rep. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.3892/or.2018.6828.
Metalli VD, Mancino MG, Mancino A, Torrice A, Alvaro D. Bile salts regulate proliferation and apoptosis of liver cells by modulating the IGF1 system. Dig Liver Dis. 2007;39:654–62.
Rodrigues CM, Steer CJ. The therapeutic effects of ursodeoxycholic acid as an anti-apoptotic agent. Expert Opin Investig Drugs. 2001;10:1243–53.
Brasitus TA. Primary chemoprevention strategies for colorectal cancer: ursodeoxycholic acid and other agents. Gastroenterology. 1995;109:2036–8.
Heffernan A, Duplancic D, Kumric M, Ticinovic Kurir T, Bozic J. Metabolic crossroads: unveiling the complex interactions between obstructive sleep apnoea and metabolic syndrome. Int J Mol Sci. 2024;25:3243.
Pandak WM, Heuman DM, Hylemon PB, Vlahcevic ZR. Regulation of bile acid synthesis. IV. Interrelationship between cholesterol and bile acid biosynthesis pathways. J Lipid Res. 1990;31:79.
Chiang JYL. Regulation of bile acid synthesis: pathways, nuclear receptors, and mechanisms. J Hepatol. 2004;40:539–51.
Twisk J, Hoekman MFM, Lehmann EM, Meijer P, Mager WH, Princen HMG. Insulin suppresses bile acid synthesis in cultured rat hepatocytes by down-regulation of cholesterol 27-hydroxylase and sterol 7α-hydroxylase gene transcription. Hepatology. 1995. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/hep.1840210235.
Song KH, Ellis E, Strom S, Chiang JYL. Hepatocyte growth factor signaling pathway inhibits cholesterol 7-hydroxylase and bile acid synthesis in human hepatocytes. Hepatology. 2007. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/hep.21878.
Sarah S, Betsy TK, Gerald S, Clifford JS, Lien N, Thomas C, Tint GS, Ashok KB. Regulation of bile acid synthesis by deoxycholic acid in the rat: different effects on cholesterol 7?-hydroxylase and sterol 27-hydroxylase*1. Hepatology. 1995. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/0270-9139(95)90631-2.
Field FJ, Kam NT, Mathur SN. Regulation of cholesterol metabolism in the intestine. Gastroenterology. 1990;99:539.
Dietschy JM, Wilson JD. Regulation of cholesterol metabolism. N Engl J Med. 1970. https://doiorg.publicaciones.saludcastillayleon.es/10.1056/NEJM197005142822005.
Long-Hai J, Hong-Liang W, Xiu-Hong M, Jing X, Ke W, Shen JI. Determination of fingerprint of bear bile powder in Tanreqing Injection and its main components. Chin Tradit Patent Med. 2013;35:109.
Champagne L, Lévaray N, Zhu XX. Two-step enzymatic synthesis of biocompatible polymers made from cholic acid. Am Chem Soc. 2017. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/acssuschemeng.6b02043.
Alawadhi S, Oommen S, Afza M. 16α-hydroxycholic acid: microbial transformation product of cholic acid. British J Pharm Res. 2013;3:374.
Qingzhi J, Jiamin C, Luping Z, Ruiyao W, Bochu W. The impact of bilirubin on 7α- and 7β-hydroxysteroid dehydrogenases: spectra and docking analysis. Catalysts. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/catal13060965.
Archer RH, Maddox IS, Chong R. Transformation of cholic acid by Clostridium bifermentans. J Appl Microbiol. 2008;52:49–56.
Boyd GS, Percy-Robb IW. Enzymatic regulation of bile acid synthesis. Am J Med. 1971;51:580–7.
Gerd J, Ann-Christine M, Arne N, Tore M. Intestinal microbial bile acid transformation in healthy infants. J Pediatr Gastroenterol Nutr. 1995. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/j.1536-4801.1995.tb11578.x.
Xi W, Xiaoyan Q, Maoying T, Zhaowei D, Chao W, Guo GH, Qinwan H, Jin W. In vivo and network pharmacological studies of natural bear bile powder against hyperlipidemia. ChemistrySelect. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/slct.202300435.
Bi D, Xing-Yun C, Yue-Lin S, Lei Y, Peng-Fei T. Novel bile acids from bear bile powder and bile of geese. Chem Pharm Bull. 2010;57:528–31.
Wimmer R, Hohenester S, Pusl T, Denk GU. Tauroursodeoxycholic acid exerts anticholestatic effects by a cooperative cPKCα-/PKA-dependent mechanism in rat liver. Gut. 2008. https://doiorg.publicaciones.saludcastillayleon.es/10.1136/gut.2007.140871.
Oelkers P, Kirby LC, Heubi JE, Dawson PA. Primary bile acid malabsorption caused by mutations in the ileal sodium-dependent bile acid transporter gene (SLC10A2). J Clin Investig. 1997;99:1880–7.
Costa MA, Bedgar DL, Moinuddin SGA, Kim KW, Cardenas CL, Cochrane FC, Shockey JM, Helms GL, Amakura Y, Takahashi H. Characterization in vitro and in vivo of the putative multigene 4-coumarate:CoA ligase network in Arabidopsis: syringyl lignin and sinapate/sinapyl alcohol derivative formation. Phytochemistry. 2005;66:2072–91.
Nagy RA, Van Montfoort APA, Dikkers A, Van Echten-Arends J, Homminga I, Land JA, Hoek A, Tietge UJF. Presence of bile acids in human follicular fluid and their relation with embryo development in modified natural cycle IVF. Human Reprod. 2015;30:1102.
Leese HJ, Lenton EA. Glucose and lactate in human follicular fluid: concentrations and interrelationships. Human Reprod. 1990;5:915–9.
Kreisberg RA, Siegal AM, Crawford OW. Glucose-lactate interrelationships: effect of ethanol. J Clin Investig. 1971;50:175–85.
Dilger K, Hohenester S, Winkler-Budenhofer U, Bastiaansen BAJ, Schaap FG, Rust C, Beuers U. Effect of ursodeoxycholic acid on bile acid profiles and intestinal detoxification machinery in primary biliary cirrhosis and health. J Hepatol. 2012;57:133–40.
Varela C, da Silva EJT, Amaral C, Georgina CDS, Baptista T, Alcaro S, Costa G, Carvalho RA, Teixeira NAA, Roleira FMF. New structure-activity relationships of A- and D-ring modified steroidal aromatase inhibitors: design, synthesis, and biochemical evaluation. J Med Chem. 2012;55:3992–4002.
Valgarður S, Hiroyuki T, Svetlana S, Višnja R, Roman G, Kavitha S, Leeb-Lundberg LMF, Takashi I, Hiroshi N, Kenichi M. Bile acids protect expanding hematopoietic stem cells from unfolded protein stress in fetal liver. Blood. 2015. https://doiorg.publicaciones.saludcastillayleon.es/10.1182/blood.V126.23.897.897.
Mantopoulos D, Murakami Y, Comander J, Thanos A, Roh M, Miller JW, Vavvas DG. Tauroursodeoxycholic Acid (TUDCA) protects photoreceptors from cell death after experimental retinal detachment. PLoS ONE. 2010;6:e24245.
Rodrigues CMP, Sola S, Nan Z, Castro RE, Ribeiro PS, Low WC, Steer CJ. Tauroursodeoxycholic acid reduces apoptosis and protects against neurological injury after acute hemorrhagic stroke in rats. Proc Natl Acad Sci USA. 2003;100:6087.
Pierre G. Neurodegenerative disorders and metabolic disease. Arch Dis Child. 2013;98:618–24.
Khalaf K, Tornese P, Cocco A, Albanese A. Tauroursodeoxycholic acid: a potential therapeutic tool in neurodegenerative diseases. Transl Neurodegener. 2022;11:1–17.
Bode N, Grebe A, Kerksiek A, Lütjohann D, Werner N, Nickenig G, Latz E, Zimmer S. Ursodeoxycholic acid impairs atherogenesis and promotes plaque regression by cholesterol crystal dissolution in mice. Biochem Biophys Res Commun. 2016;478:356–62.
Leonardo B, Angélico M, Stanley RG, Gene L, Gianfranco A. Ursodeoxycholate and tauroursodeoxycholate inhibition of cholangiocyte proliferative and secretive functions is associated with activation of Ca2+-dependent PKC alpha. Dig Liver Dis. 2001.
Baiocchi L, Tisone G, Russo MA, Longhi C, Palmieri G, Volpe A, Almerighi C, Telesca C, Carbone M, Toti L. TUDCA prevents cholestasis and canalicular damage induced by ischemia-reperfusion injury in the rat, modulating PKCα–ezrin pathway. Transpl Int. 2010;21:792–800.
Patricia M-M, Isabel RC, Pilar R, Cristina HC, Carlos JA, Raquel Luengo G, Antonio Z, Marı́a Dolores Z, Olga MA, Marin JJG. Dose-dependent antiinflammatory effect of ursodeoxycholic acid in experimental colitis. Int Immunopharmacol. 2013. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.intimp.2012.11.017.
Xie Q, Khaoustov VI, Chung CC, Sohn J, Krishnan B, Lewis DE, Yoffe B. Effect of tauroursodeoxycholic acid on endoplasmic reticulum stress-induced caspase-12 activation. Hepatology. 2002;36:592.
Rosa AI, Fonseca I, Nunes MJ, Moreira S, Rodrigues E, Carvalho AN, Rodrigues CM, Gama MJ, Castro-Caldas M. Novel insights into the antioxidant role of tauroursodeoxycholic acid in experimental models of Parkinson’s disease. Biochimica Et Biophysica Acta Molecular Basis of Disease Bba. 2017. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.bbadis.2017.06.004.
Michelle S, Brice B, Julie R, Kim J, Robert WP, James MB, Jay SK. P5707An evaluation of the use of propensity scores in cardiovascular literature: a systematic review and recommendations. Eur Heart J. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/eurheartj/ehz746.0648.
Hanson RF, Pries JM. Synthesis and enterohepatic circulation of bile salts. Gastroenterology. 1977;73:611–8.
Hofmann AF, Molino G, Milanese M, Belforte G. Description and simulation of a physiological pharmacokinetic model for the metabolism and enterohepatic circulation of bile acids in man. Cholic acid in healthy man. J Clin Investig. 1983;71:1003–22.
Jolley CD, Dietschy JM, Turley SD. Induction of bile acid synthesis by cholesterol and cholestyramine feeding is unimpaired in mice deficient in apolipoprotein AI. Hepatology. 2010;32:1309–16.
Sara S, Amit L, Christina S, Jennifer H, Bo A, Mats R. Murine bile acids explain species differences in the regulation of bile acid and cholesterol metabolism. Atherosclerosis. 2021;331:e123.
Whiting MJ, Watts JM. Prediction of the bile acid composition of bile from serum bile acid analysis during gallstone dissolution therapy. Gastroenterology. 1980;78:220–5.
Keiko K, Teruo H, Kazumi Y, Yoshikazu A. Studies on bile acids in bear bile. J Biochem. 1973;74:489.
Bochu W, Bochu W, Jun T, Lingyu Z. Carboxyl-terminal and Arg38 are essential for activity of the 7α-hydroxysteroid dehydrogenase from clostridium absonum. Protein Pept Lett. 2014;21:894.
Shuang T, Yinping P, Deshuai L, Shunlin J, Lingyu Z, Jun T, Na Q, Qiong Y, Zhang Z, Bin Y, et al. Structural and functional characterization of a novel acidophilic 7α-hydroxysteroid dehydrogenase. Protein Sci. 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/pro.3599.
Miettinen MM, Poutanen MH, Vihko RK. Characterization of estrogen-dependent growth of cultured MCF-7 human breast-cancer cells expressing 17beta-hydroxysteroid dehydrogenase type 1. Int J Cancer. 1996;68:600–4.
Shunlin J, Yinping P, Lingyu Z, Jun T, Shuang T, Qiong Y, Zhang Z, Deshuai L, Bochu W. A novel 7α-hydroxysteroid dehydrogenase: magnesium ion significantly enhances its activity and thermostability. Int J Biol Macromol. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ijbiomac.2021.02.082.
Can S, Bochu W, Jun T, Lingyu Z, Deshuai L. Discovery of tauroursodeoxycholic acid biotransformation enzymes from the gut microbiome of black bears using metagenomics. Sci Rep. 2017. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/srep45495.
Hongqin Y, Yanmei H, Jiuyang L, Peisong T, Qiaomei S, Xiaopeng X, Bing T, Jiawei H, Hui L. Binding modes of environmental endocrine disruptors to human serum albumin: insights from STD-NMR, ITC, spectroscopic and molecular docking studies. Sci Rep. 2017. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-017-11604-3.
Di W, Yan Z, Jin Y, Kailin X, Qing W, Yuanzhi L, Hui L. Binding mechanism of tauroursodeoxycholic acid to human serum albumin: insights from NMR relaxation and docking simulations. RSC Adv. 2015;5:11036.
Deshuai L, Xi L, Jun T. An overview of 7α- and 7β-hydroxysteroid dehydrogenases: structure, specificity and practical application. Protein Pept Lett. 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/0929866528666210816114032.
Deshuai L, Jun T, Lingyu Z, Shunlin J, Shuang T, Kaiyi Y, Jingxuan H, Bochu W. Engineering clostridium absonum 7α-hydroxysteroid dehydrogenase for enhancing thermostability based on flexible site and ΔΔG prediction. Protein Pept Lett. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/0929866524666171113113100.
Qiong Y, Bochu W, Zhi Z, Deshuai L, Jun T, Lingyu Z. The effects of macromolecular crowding and surface charge on the properties of an immobilized enzyme: activity, thermal stability, catalytic efficiency and reusability. RSC Adv. 2017. https://doiorg.publicaciones.saludcastillayleon.es/10.1039/C7RA06544B.
Qiong Y, Bochu W, Zhi Z, Deshuai L, Jun T, Lingyu Z. The effects of macromolecular crowding and surface charge on the properties of an immobilized enzyme: activity, thermal stability, catalytic efficiency and reusability. RSC Adv. 2017;7:38028.
Qiong Y, Liuying L, Bochu W, Lingyu Z, Jun T. Modifying the microenvironment of epoxy resin to improve the activity of immobilized 7α-hydroxysteroid dehydrogenases. Appl Biochem Biotechnol. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s12010-020-03473-w.
Qing Y, Bochu W, Deshuai L, Jing T, Lijun Z. Chitosan-modified epoxy resin for improving the performance of an immobilized enzyme carrier. Sci Adv Mater. 2018. https://doiorg.publicaciones.saludcastillayleon.es/10.1166/sam.2018.3151.
Qingzhi J, Bochu W, Li-Der C, Jinglan H, Wenjing F. Co-immobilised 7α- and 7β-HSDH as recyclable biocatalyst: high-performance production of TUDCA from waste chicken bile. RSC Adv. 2018;8:34192.
Qingzhi J, Bochu W, Jun T, Lingyu Z, Liuying L. Immobilized multienzymatic systems for catalysis of cascade reactions. Process Biochem. 2016. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.procbio.2016.06.004.
Xiaoshu S, Haoyu X, Bin Z, Yining Z, Yuanyuan L, Tianming W, Jiasheng W, Shaoyong L, Zhengtao W, Rong S, Li Y. Anti-convulsant effects of cultures bear bile powder in febrile seizure via regulation of neurotransmission and inhibition of neuroinflammation. J Ethnopharmacol. 2020. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jep.2020.112998.
Jingyi C, Jiasheng W, Su F, Shaoyong L, Tianming W, Yuanyuan L, Juan Z, Rong S, Zhengtao W, Li Y. Cultured bear bile powder ameliorates acute liver injury in cholestatic mice via inhibition of hepatic inflammation and apoptosis. J Ethnopharmacol. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jep.2021.114829.
Acknowledgements
In the process of finding and summarising the article, we received valuable help from , Tang Shijing, Pan Yingping, Yang Qiong, Lou Deshuai, whose work contributed to this article. In addition, we would like to thank Zhu Liancai, Tan Jun, Liu Shaoyong,Wang Bochu for their valuable advice and guidance, whose expertise and and experience had a significant impact on the direction of our research.
Funding
This research was funded by Chongqing Natural Science Foundation Innovation Development Joint Fund(CSTB2022NSCQ-LZX0053); Chongqing Entrepreneurship;Innovation Support Program for Overseas Returnees(CX2023011); Graduate Research and Innovation Foundation of Chongqing, China (CYB21072);the Scientific and Technological Research Program of Chongqing Municipal Education Commission (KJQN202001427).
Author information
Authors and Affiliations
Contributions
CRediT authorship contribution statement ShijinTang: Investigation, Methodology, Data curation, Formal analysis, Writing-original draft;Yinping Pan: Data curation, Methodology; Qiong Yang: Writing-review & editing; Deshuai Lou: Writing-review & editing;Jun Tan: Writing-review & editing; Liancai Zhu: Supervision, Conceptualization, Methodology; Shaoyong Liu: Supervision, Conceptualization, Methodology; Bochu Wang: Supervision, Methodology, Funding acquisition.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
All authors have read and agreed to the published version of the manuscript.
Competing interests
The authors declare no competing interests.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Shijing, T., Yinping, P., Qiong, Y. et al. Synthesis of TUDCA from chicken bile: immobilized dual-enzymatic system for producing artificial bear bile substitute. Microb Cell Fact 23, 326 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12934-024-02592-x
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12934-024-02592-x