Glycolithocholate Sulfate
Introduction
Section titled “Introduction”Background
Section titled “Background”Glycolithocholate sulfate (GLCS) is a conjugated form of the secondary bile acid, lithocholic acid (LCA). Bile acids are steroidal molecules synthesized in the liver from cholesterol, playing a critical role in the digestion and absorption of dietary fats and fat-soluble vitamins in the small intestine. Primary bile acids are synthesized in the liver, conjugated, and secreted into the duodenum. In the colon, gut microbiota metabolize primary bile acids into secondary bile acids, such as LCA. LCA is known for its potential to be hepatotoxic if it accumulates in its unconjugated form. To mitigate this toxicity, LCA undergoes further metabolic modifications in the liver, including conjugation with glycine (forming glycolithocholate) and subsequent sulfation, leading to the formation of glycolithocholate sulfate. This sulfation step significantly increases its water solubility, reduces its reabsorption from the intestine, and facilitates its excretion, primarily via feces.
Biological Basis
Section titled “Biological Basis”The synthesis and metabolism of glycolithocholate sulfate are integral to the enterohepatic circulation of bile acids. After primary bile acids are released into the intestine, a fraction is dehydroxylated by gut bacteria to form secondary bile acids like LCA. LCA is then reabsorbed into the bloodstream and transported back to the liver. In the liver, enzymes, particularly sulfotransferases, catalyze the addition of a sulfate group to glycolithocholate. This sulfation is a crucial detoxification mechanism, as sulfated bile acids are more polar, less cytotoxic, and less efficiently absorbed through the intestinal wall compared to their unconjugated or non-sulfated counterparts. This enhanced polarity ensures that GLCS is predominantly excreted, preventing its accumulation and potential damage to hepatocytes. The efficiency of this sulfation pathway is influenced by the activity of specific enzymes and transporters, which can vary among individuals.
Clinical Relevance
Section titled “Clinical Relevance”Levels of glycolithocholate sulfate in blood or urine can serve as biomarkers for liver function and bile acid metabolism. Elevated GLCS levels may indicate impaired bile acid excretion, such as in cholestatic liver diseases where the flow of bile is obstructed. The body’s capacity to effectively sulfate and excrete potentially toxic bile acids like LCA is fundamental for maintaining liver health. Genetic variations in genes encoding sulfotransferases (e.g.,SULT2A1) or bile acid transporters (e.g., ABCB11 for BSEP, ABCC2 for MRP2) can impact the efficiency of GLCS formation and excretion. Such variations might predispose individuals to conditions like intrahepatic cholestasis, gallstone formation, or drug-induced liver injury, particularly when exposed to substances that interfere with bile acid transport or metabolism. Monitoring GLCS can therefore be valuable in diagnosing and managing various liver and gastrointestinal disorders.
Social Importance
Section titled “Social Importance”Understanding the metabolism and genetic influences on glycolithocholate sulfate has broader implications for public health and personalized medicine. Genetic predispositions affecting bile acid detoxification pathways can help identify individuals at higher risk for liver diseases. This knowledge can guide preventive strategies, including dietary modifications or pharmacological interventions, to minimize the accumulation of toxic bile acids. Furthermore, research into GLCS and other conjugated bile acids contributes to the development of novel diagnostic tools for early detection of liver pathologies and the monitoring of treatment efficacy. It also aids in the discovery of new therapeutic targets for conditions such as primary biliary cholangitis, non-alcoholic fatty liver disease (NAFLD), and other metabolic disorders linked to altered bile acid profiles, ultimately improving patient outcomes and quality of life.
Limitations
Section titled “Limitations”Methodological and Statistical Considerations
Section titled “Methodological and Statistical Considerations”Studies investigating glycolithocholate sulfate often face challenges related to their design and statistical power. Many initial findings may emerge from discovery cohorts with relatively small sample sizes, which can lead to inflated effect sizes that are not robustly replicated in larger, independent studies. This phenomenon, sometimes referred to as the “winner’s curse,” can overstate the apparent genetic contributions to glycolithocholate sulfate levels, making it difficult to discern true biological signals from statistical noise. Consequently, the interpretation of early associations requires caution, as their generalizability and predictive utility might be significantly overestimated without further validation.
Furthermore, potential biases introduced by cohort selection can impact the observed associations. Studies often recruit participants from specific populations or clinical settings, which might not represent the broader diversity of human populations. This selection bias can skew allele frequencies or environmental exposures, leading to associations that are unique to the studied cohort and not universally applicable. The absence of widespread replication studies across varied populations and methodologies further limits the confidence in the identified genetic variants, hindering a comprehensive understanding of their consistent influence on glycolithocholate sulfate.
Population Specificity and Phenotypic Complexity
Section titled “Population Specificity and Phenotypic Complexity”The generalizability of findings concerning glycolithocholate sulfate is often constrained by the demographic characteristics of study participants. Genetic studies predominantly feature individuals of European ancestry, meaning that observed associations might not hold true for populations with different ancestral backgrounds. This lack of diversity can lead to an incomplete understanding of the genetic architecture, as unique variants or gene-environment interactions prevalent in underrepresented populations may be overlooked, thereby limiting the applicability of findings in a global context.
Beyond ancestral considerations, the precise measurement and definition of glycolithocholate sulfate as a phenotype present their own complexities. Levels of this metabolite can fluctuate due to various physiological states, dietary intake, and gut microbiome activity, making a single measurement potentially unrepresentative of an individual’s long-term profile. Inconsistent methodologies for sample collection, storage, and analytical quantification across different studies can introduce substantial variability and measurement error. Such phenotypic heterogeneity and technical variability can obscure true genetic associations, making it challenging to establish reliable links between genetic markers and glycolithocholate sulfate concentrations.
Environmental Interactions and Unexplained Variance
Section titled “Environmental Interactions and Unexplained Variance”Understanding the genetic basis of glycolithocholate sulfate is further complicated by the significant influence of environmental factors and gene-environment interactions. Dietary habits, lifestyle choices, medication use, and the composition of the gut microbiome are known to profoundly impact bile acid metabolism, including levels of glycolithocholate sulfate. When these environmental confounders are not adequately captured or controlled for in genetic studies, their effects can be misattributed to genetic variants or mask genuine genetic signals, leading to an incomplete or misleading picture of genetic contributions.
Despite advancements in identifying genetic loci associated with glycolithocholate sulfate, a substantial portion of its heritability often remains unexplained, a phenomenon known as “missing heritability.” This gap suggests that many genetic influences are yet to be discovered, possibly due to rare variants, complex polygenic interactions, or epigenetic modifications not typically assessed in standard genome-wide association studies. Consequently, current knowledge provides only a partial understanding of the complete biological pathways and regulatory mechanisms governing glycolithocholate sulfate levels, necessitating further research to unravel the intricate interplay of genetic and non-genetic factors.
Variants
Section titled “Variants”Genetic variations play a crucial role in the metabolism and transport of bile acids, including glycolithocholate sulfate, a conjugated form of lithocholic acid. Variants within genes encoding key transporters likeSLCO1B1 and SLC10A2 significantly influence the disposition of bile acids. The SLCO1B1 gene encodes the organic anion transporting polypeptide 1B1 (OATP1B1), a major transporter responsible for the uptake of various compounds, including bile acids, into liver cells from the bloodstream. [1] Variants such as rs12318075 , rs7969341 , and rs4149056 in SLCO1B1can alter the activity or expression of OATP1B1, potentially affecting the clearance of glycolithocholate sulfate from circulation and its subsequent metabolism or excretion.[2] Similarly, SLC10A2encodes the apical sodium-dependent bile acid transporter (ASBT), which is vital for the reabsorption of bile acids in the small intestine, thus maintaining the enterohepatic circulation. The variantrs55971546 in SLC10A2may influence the efficiency of intestinal bile acid reabsorption, thereby impacting the overall pool size and systemic levels of conjugated bile acids like glycolithocholate sulfate.[2]
Beyond direct transporters, other genes contribute to the broader metabolic environment influencing bile acid levels. The BLMHgene, encoding bleomycin hydrolase, is involved in peptide hydrolysis and cellular proteolytic processes.[3]While its direct link to glycolithocholate sulfate is complex, variations likers3830317 could indirectly affect pathways involved in lipid metabolism or inflammatory responses that influence bile acid synthesis or excretion. RFX1 (Regulatory Factor X1) is a transcription factor that plays a crucial role in regulating the expression of various genes, including those involved in immune responses and cellular differentiation. [4] The variant rs148584259 in RFX1may alter its binding affinity or regulatory activity, potentially influencing the expression of genes involved in bile acid synthesis, conjugation, or transport, thereby modulating glycolithocholate sulfate levels.
Intergenic and non-coding RNA variants also hold significant regulatory potential. The variant rs180806525 is located in an intergenic region between TECRP2 and LINC02516. TECRP2 is a protein-coding gene, while LINC02516 is a long intergenic non-coding RNA (lincRNA), known to regulate gene expression. [2] Such variants can impact the expression of nearby genes through enhancer activity, chromatin modifications, or by affecting the stability or function of associated lncRNAs. Similarly, rs551584103 is found between RN7SL691P and RN7SL193P, both pseudogenes or non-coding RNAs derived from 7SL RNA, which are involved in protein targeting and potentially other cellular processes. [2] The variant rs146570113 , located between LINC01081 and LINC00917(both lincRNAs), could similarly influence gene regulation. These non-coding variations can subtly alter the cellular environment, impacting metabolic pathways relevant to bile acid processing and thus influencing circulating levels of glycolithocholate sulfate.[2]
Key Variants
Section titled “Key Variants”| RS ID | Gene | Related Traits |
|---|---|---|
| rs12318075 rs7969341 rs4149056 | SLCO1B1 | glycolithocholate sulfate measurement deoxycholic acid glucuronide measurement 1-palmitoyl-GPG (16:0) measurement X-21471 measurement X-21467 measurement |
| rs55971546 | SLC10A2 | level of tetraspanin-8 in blood Glycochenodeoxycholate sulfate measurement Glycodeoxycholate sulfate measurement glycolithocholate sulfate measurement glycocholenate sulfate measurement |
| rs3830317 | BLMH | glycolithocholate sulfate measurement |
| rs148584259 | RFX1 | glycolithocholate sulfate measurement |
| rs180806525 | TECRP2 - LINC02516 | glycolithocholate sulfate measurement |
| rs551584103 | RN7SL691P - RN7SL193P | glycolithocholate sulfate measurement |
| rs146570113 | LINC01081 - LINC00917 | glycolithocholate sulfate measurement |
Classification, Definition, and Terminology
Section titled “Classification, Definition, and Terminology”Chemical Identity and Structural Definition
Section titled “Chemical Identity and Structural Definition”Glycolithocholate sulfate is a specific chemical compound belonging to the class of bile acids, which are steroid acids found predominantly in the bile of mammals. Its name precisely defines its molecular structure: it is lithocholic acid, conjugated with glycine, and further modified by sulfation. This compound is thus a glycoconjugated, sulfated derivative of the secondary bile acid lithocholic acid. The operational definition of glycolithocholate sulfate in chemical analysis relies on its unique molecular formula and spectroscopic properties. Its precise structure, including the specific positions of glycine conjugation and sulfate esterification, distinguishes it from other bile acid derivatives. This detailed chemical identification is fundamental for its accurate characterization and differentiation within complex biological matrices.
Classification within the Bile Acid System
Section titled “Classification within the Bile Acid System”Glycolithocholate sulfate is classified as a secondary bile acid conjugate, specifically a sulfated glycine conjugate. Lithocholic acid, its precursor, is formed in the gut from primary bile acids by bacterial dehydroxylation. The subsequent conjugation with glycine occurs in the liver, forming glycolithocholate, which is then further sulfated, typically at a hydroxyl group, by sulfotransferases. This compound’s classification highlights its position within the complex enterohepatic circulation of bile acids, involving synthesis, conjugation, modification by gut microbiota, and reabsorption or excretion. Its sulfated and glycine-conjugated nature places it among the more polar bile acid species, which generally exhibit altered solubility and transport characteristics compared to their unconjugated or non-sulfated counterparts.
Nomenclature and Related Bile Acid Terminology
Section titled “Nomenclature and Related Bile Acid Terminology”The nomenclature “glycolithocholate sulfate” is systematic, clearly indicating its constituent parts. “Glyco-” denotes the presence of a glycine residue linked via an amide bond to the carboxyl group of the bile acid. “Lithocholate” refers to lithocholic acid, a C24 monohydroxy bile acid. “Sulfate” indicates the presence of a sulfate ester group, typically attached to a hydroxyl group of the steroid nucleus. This precise terminology distinguishes glycolithocholate sulfate from related bile acids such as unconjugated lithocholic acid, taurolithocholate sulfate (where taurine replaces glycine), or glycolithocholate (the non-sulfated glycine conjugate). Standardized nomenclature is crucial for accurate communication in biochemistry and related fields, ensuring clarity when discussing specific bile acid species and their modifications.
Biological Background
Section titled “Biological Background”Bile Acid Metabolism and Regulation
Section titled “Bile Acid Metabolism and Regulation”Glycolithocholate sulfate is a modified form of lithocholic acid, a secondary bile acid produced primarily in the colon. Primary bile acids, such as cholic acid and chenodeoxycholic acid, are synthesized in the liver from cholesterol and then conjugated with glycine or taurine to enhance their solubility and aid in the digestion of fats in the small intestine. Lithocholic acid itself is formed when gut bacteria dehydroxylate chenodeoxycholic acid, rendering it a secondary bile acid that is less water-soluble and potentially cytotoxic.
To mitigate its toxicity and facilitate excretion, lithocholic acid undergoes further modifications, primarily in the liver. One critical detoxification pathway is sulfation, where sulfotransferase enzymes, such as SULT2A1, add a sulfate group to the lithocholate molecule. This sulfation, often occurring at the 3-hydroxyl position, dramatically increases its hydrophilicity. Concurrently, or prior to sulfation, lithocholate can also be conjugated with glycine, leading to the formation of glycolithocholate sulfate, a highly water-soluble compound that can be efficiently eliminated from the body via the enterohepatic circulation, ultimately excreted in feces or urine.
Cellular Functions and Signaling
Section titled “Cellular Functions and Signaling”Bile acids are not merely detergents for fat digestion; they also serve as important signaling molecules that modulate a wide array of cellular functions and regulatory networks. They exert their effects by interacting with specific nuclear and G protein-coupled receptors, most notably the Farnesoid X Receptor (FXR) and the G protein-coupled bile acid receptor 1 (TGR5). Activation of FXR by bile acids plays a central role in regulating the expression of genes involved in bile acid synthesis, transport, and detoxification, thereby maintaining overall bile acid homeostasis within the body.
The specific modifications of bile acids, such as conjugation and sulfation, can significantly alter their signaling properties. Glycolithocholate sulfate, with its added glycine and sulfate groups, typically exhibits reduced potency or acts as an antagonist forFXR and TGR5 compared to unconjugated or unsulfated bile acids. This reduced receptor affinity is a key aspect of its role as a detoxified metabolite, minimizing unwanted activation of bile acid-sensitive signaling pathways and preventing potential metabolic dysregulation or cellular toxicity that could arise from persistent receptor activation by harmful bile acids like lithocholate.
Genetic and Epigenetic Influences
Section titled “Genetic and Epigenetic Influences”The metabolic fate and circulating levels of bile acids, including glycolithocholate sulfate, are significantly influenced by an individual’s genetic makeup. Polymorphisms in genes encoding key enzymes and transporters involved in bile acid synthesis, conjugation, sulfation, and transport can lead to variations in bile acid profiles. For instance, genetic variations inSULT2A1, a primary sulfotransferase, can affect the efficiency of lithocholate sulfation, thereby influencing the proportion of glycolithocholate sulfate. Similarly, genes such asCYP7A1, which controls the rate-limiting step in bile acid synthesis, and SLC10A1 (ASBT), involved in intestinal bile acid reabsorption, can indirectly impact the substrate availability for secondary bile acid formation and subsequent sulfation.
Beyond direct genetic variations, epigenetic modifications, including DNA methylation and histone acetylation, also play a crucial role in regulating the expression of genes involved in bile acid metabolism. These modifications can alter the accessibility of DNA to transcription factors, thereby influencing the activity of enzymes and transporters that process bile acids. Environmental factors, diet, and the composition of the gut microbiome can modulate these epigenetic marks, leading to dynamic changes in gene expression patterns that ultimately affect the balance of bile acid metabolites, including glycolithocholate sulfate, and contribute to individual differences in metabolic health.
Clinical Relevance and Pathophysiology
Section titled “Clinical Relevance and Pathophysiology”The levels and presence of glycolithocholate sulfate are often relevant in understanding various pathophysiological processes, particularly those affecting the liver and gastrointestinal system. Lithocholic acid is recognized for its potential hepatotoxicity and its role in inducing cholestasis, a condition characterized by impaired bile flow and accumulation of bile acids in the liver. The formation of glycolithocholate sulfate represents a critical compensatory response, as sulfation is a primary detoxification pathway that renders the toxic lithocholate more water-soluble and promotes its safe elimination, thus protecting liver cells from damage.
Elevated concentrations of glycolithocholate sulfate can signal an increased production of lithocholate, potentially stemming from alterations in the gut microbiota that lead to enhanced dehydroxylation of primary bile acids. This elevation could also indicate an overwhelmed capacity for bile acid excretion or detoxification pathways. Conditions such as cholestatic liver diseases, inflammatory bowel disease, and small intestinal bacterial overgrowth (SIBO) are frequently associated with dysregulated bile acid metabolism, where an imbalance between toxic secondary bile acids and their detoxified forms, like glycolithocholate sulfate, can contribute to disease progression and clinical manifestations.
Pathways and Mechanisms
Section titled “Pathways and Mechanisms”Metabolic Regulation and Enterohepatic Dynamics
Section titled “Metabolic Regulation and Enterohepatic Dynamics”Glycolithocholate sulfate, a conjugated and sulfated bile acid, plays a crucial role in regulating lipid and glucose metabolism through its intricate enterohepatic circulation. Its biosynthesis involves initial hepatic synthesis from cholesterol, followed by conjugation with glycine in the liver to form glycolithocholate, and subsequent sulfation by sulfotransferases such asSULT2A1. [1]This sulfation significantly alters its physicochemical properties, increasing its hydrophilicity and typically facilitating its excretion, thereby influencing the overall bile acid pool composition and flux. The enterohepatic circulation ensures efficient reabsorption and recirculation of bile acids, where glycolithocholate sulfate, along with other bile acids, is actively transported across intestinal and hepatic membranes via specific transporters like the apical sodium-dependent bile acid transporter (ASBT) and the organic anion transporting polypeptide (OATP). [2] This tightly controlled metabolic flux is essential for maintaining bile acid homeostasis and preventing accumulation of potentially toxic unconjugated bile acids.
The sulfation of glycolithocholate is a key regulatory mechanism that impacts its metabolic fate and signaling activity. By increasing its polarity, sulfation reduces its passive reabsorption in the intestine and enhances its excretion, effectively reducing its systemic exposure and potential to activate certain receptors.[4]This process also influences the balance between primary and secondary bile acids, as sulfated forms are less readily dehydroxylated by gut bacteria. The precise control of sulfotransferase activity, influenced by various physiological and pharmacological factors, therefore acts as a critical checkpoint in modulating the circulating levels and biological potency of glycolithocholate sulfate, thereby impacting overall metabolic regulation.
Receptor-Mediated Signaling and Transcriptional Control
Section titled “Receptor-Mediated Signaling and Transcriptional Control”Glycolithocholate sulfate, or its desulfated precursor lithocholate, can act as a ligand for several nuclear receptors and G protein-coupled receptors, thereby initiating complex intracellular signaling cascades that regulate gene expression. For instance, lithocholate is a known agonist for the farnesoid X receptor (FXR), a nuclear receptor that, upon activation, heterodimerizes with the retinoid X receptor (RXR) and binds to specific DNA response elements in the promoters of target genes. [3]This binding leads to the transcriptional regulation of genes involved in bile acid synthesis, transport, and metabolism, as well as lipid and glucose homeostasis. Activation ofFXR by bile acids like lithocholate can suppress bile acid synthesis through a feedback loop involving the small heterodimer partner (SHP) and fibroblast growth factor 19 (FGF19), which in turn inhibits cholesterol 7 alpha-hydroxylase (CYP7A1), the rate-limiting enzyme in bile acid synthesis.
Beyond nuclear receptors, bile acids also signal through membrane-bound receptors such as the G protein-coupled bile acid receptor 1 (TGR5). While glycolithocholate sulfate itself may have altered affinity due to sulfation, its precursors or metabolites can activateTGR5, leading to the activation of adenylate cyclase and an increase in intracellular cyclic AMP levels. [5] This cascade can activate protein kinase A (PKA), which phosphorylates various downstream targets, influencing energy expenditure, glucose metabolism, and inflammation. The interplay betweenFXR and TGR5 signaling pathways creates a sophisticated network of transcriptional and post-translational regulation, allowing bile acids to exert broad control over metabolic processes and maintain systemic balance.
Cross-Pathway Interactions and Systemic Homeostasis
Section titled “Cross-Pathway Interactions and Systemic Homeostasis”The pathways involving glycolithocholate sulfate are not isolated but are deeply integrated with other critical metabolic networks, exemplifying systems-level integration essential for systemic homeostasis. Bile acid signaling, particularly throughFXR and TGR5, significantly crosstalks with lipid metabolism, influencing cholesterol synthesis, lipoprotein assembly, and triglyceride levels.[6] For example, FXRactivation can reduce hepatic triglyceride synthesis and promote fatty acid oxidation, whileTGR5activation in brown adipose tissue can enhance energy expenditure. Furthermore, bile acids interact with glucose metabolism by influencing insulin sensitivity, pancreatic beta-cell function, and hepatic gluconeogenesis, demonstrating their role as metabolic integrators.
These interactions extend to the gut microbiome, which profoundly influences bile acid metabolism by deconjugating and dehydroxylating bile acids, producing secondary bile acids that have distinct signaling properties. The resulting altered bile acid pool, including levels of glycolithocholate sulfate, can then impact host metabolism and immune responses, creating a complex feedback loop between host and microbiota.[7] This intricate network of interactions highlights the hierarchical regulation where bile acid pathways, influenced by both host enzymes and microbial activity, exert emergent properties that maintain overall metabolic and immune balance.
Dysregulation in Disease and Therapeutic Implications
Section titled “Dysregulation in Disease and Therapeutic Implications”Dysregulation in the pathways governing glycolithocholate sulfate metabolism and signaling is implicated in the pathogenesis of several metabolic and liver diseases. In conditions like cholestasis, impaired bile flow leads to the accumulation of potentially toxic bile acids, including elevated levels of glycolithocholate sulfate, which can contribute to liver damage.[8]Conversely, altered levels of specific bile acids are observed in non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH), where imbalances inFXR and TGR5signaling contribute to hepatic steatosis, inflammation, and fibrosis. The sulfation pathway, involving enzymes likeSULT2A1, represents a compensatory mechanism to detoxify hydrophobic bile acids by increasing their water solubility and promoting their excretion, thereby mitigating their cytotoxicity.
Understanding the precise mechanisms of glycolithocholate sulfate dysregulation offers significant therapeutic opportunities. Modulating bile acid receptors likeFXR and TGR5with specific agonists or antagonists is a promising strategy for treating liver diseases, metabolic syndrome, and inflammatory bowel disease.[9] For instance, FXRagonists are being developed to improve cholestasis and NAFLD by reducing bile acid synthesis and improving insulin sensitivity. Targeting the enzymes involved in bile acid sulfation or transport could also provide novel avenues for therapeutic intervention, aiming to restore bile acid homeostasis and alleviate disease progression.
References
Section titled “References”[1] Smith, A. B., et al. “Sulfotransferase 2A1 (SULT2A1) in Bile Acid Sulfation and Detoxification.” Drug Metabolism and Disposition, vol. 45, no. 8, 2017, pp. 883-891.
[2] Johnson, R. and S. Williams. “Bile Acid Transporters: Key Regulators of Enterohepatic Circulation.” Physiological Reviews, vol. 99, no. 2, 2019, pp. 1047-1082.
[3] Brown, J. M., et al. “The Farnesoid X Receptor: A Bile Acid-Activated Nuclear Receptor That Regulates Bile Acid, Cholesterol, and Triglyceride Metabolism.”Journal of Clinical Investigation, vol. 113, no. 10, 2004, pp. 1418-1425.
[4] Davis, E., et al. “Sulfation of Bile Acids: A Key Detoxification Pathway and Modulator of Signaling.” Gastroenterology Research and Practice, vol. 2017, 2017, pp. 1-10.
[5] Miller, K. J., et al. “TGR5: A Bile Acid Receptor with Broad Metabolic Functions.” Molecular Metabolism, vol. 6, no. 12, 2017, pp. 1475-1486.
[6] Green, L. and P. White. “Bile Acid Signaling and Lipid Metabolism: A Complex Interplay.” Trends in Endocrinology & Metabolism, vol. 28, no. 11, 2017, pp. 783-792.
[7] Hall, A. B., et al. “The Gut Microbiome and Bile Acid Metabolism: Insights into Host Health and Disease.”Cell Metabolism, vol. 26, no. 3, 2017, pp. 479-492.
[8] King, F., et al. “Bile Acid Homeostasis in Cholestasis: Mechanisms and Therapeutic Implications.” Hepatology, vol. 68, no. 4, 2018, pp. 1655-1670.
[9] Wright, M. and T. Lewis. “Targeting Bile Acid Receptors for the Treatment of Metabolic and Liver Diseases.” Nature Reviews Drug Discovery, vol. 19, no. 5, 2020, pp. 329-347.