Fact-checked by Grok 2 weeks ago

Glucuronidation

Glucuronidation is a major phase II conjugation reaction in mammalian , in which derived from UDP-glucuronic acid (UDPGA) is covalently attached to functional groups—such as hydroxyl, carboxyl, amino, or groups—on substrates including drugs, environmental toxins, and endogenous compounds like and hormones, thereby enhancing their polarity and water solubility to promote renal or biliary excretion. This process is catalyzed by the superfamily of UDP-glucuronosyltransferase (UGT) enzymes, which are membrane-bound proteins primarily localized in the of hepatocytes, enterocytes, and renal cells, with over 20 functional isoforms in humans divided into four families (UGT1, UGT2, UGT3, and UGT8), the most prominent being UGT1A and UGT2B subfamilies that handle the majority of and endogenous substrates. Key isoforms such as UGT1A1 (responsible for conjugation), UGT1A4 (amines and amides), UGT1A9 (phenolics and carboxyls), and UGT2B7 (steroids and opioids like ) collectively metabolize approximately 35-40% of clinically used , underscoring glucuronidation's central role in clearance and . The biological significance of glucuronidation extends beyond mere elimination; it often follows phase I oxidation by enzymes, forming hydrophilic metabolites that are typically inactive but can occasionally be pharmacologically active (e.g., morphine-6-glucuronide, which is more potent than its parent compound) or contribute to toxicity if deconjugated by bacterial in the gut, potentially leading to enterohepatic recirculation and prolonged exposure. Efflux transporters like multidrug resistance-associated proteins (MRP2, MRP3, MRP4) and resistance protein (BCRP) collaborate with UGTs to vectorially transport these conjugates into , , or bloodstream, influencing , , and interindividual variability influenced by genetic polymorphisms—such as UGT1A1*28 associated with , a benign condition of unconjugated hyperbilirubinemia. Clinically, glucuronidation's importance is evident in its impact on therapeutic outcomes and disease states; for instance, it facilitates the detoxification of pollutants like polycyclic aromatic hydrocarbons and dietary flavonoids (e.g., ), while deficiencies or inhibition (e.g., by herbal supplements or disease) can lead to severe disorders like Crigler-Najjar syndrome, characterized by profound unconjugated accumulation and risk in neonates. Overall, this pathway represents a critical barrier against chemical , with ongoing focusing on its regulation by nuclear receptors (e.g., CAR, PXR) and implications for in and .

Introduction

Definition and Biological Role

Glucuronidation is a phase II conjugation reaction in which , derived from glucuronic acid (UDPGA), is covalently attached to functional groups such as hydroxyl, carboxyl, or amino moieties on various substrates, forming water-soluble glucuronides. This process enhances the polarity of lipophilic compounds, promoting their elimination and serving as a primary mechanism in humans. Catalyzed by UDP-glucuronosyltransferases (UGTs), glucuronidation utilizes the activated form of glucuronic acid to facilitate these conjugations efficiently across multiple tissues. Biologically, glucuronidation plays a critical role in protecting against the accumulation of toxic substances by converting hydrophobic xenobiotics and endogenous molecules into excretable forms, primarily through biliary and urinary pathways. It is essential for the of endogenous compounds, including —where conjugation prevents by enabling its hepatic excretion—and steroid hormones like and , which undergo glucuronidation to regulate their levels and facilitate clearance. This pathway thus maintains physiological balance while mitigating potential toxicity from both internal and external sources. As one of the predominant phase II metabolic processes alongside sulfation and , glucuronidation accounts for the biotransformation of approximately 35% of drugs metabolized by phase II conjugation reactions, underscoring its significance in and drug clearance. By increasing solubility without requiring prior oxidation, it provides a versatile route for eliminating a broad range of compounds, contributing substantially to overall metabolic defense.

Historical Background

The discovery of glucuronidation traces back to the late , when early observations of conjugated metabolites in urine laid the groundwork for recognizing as a key component in processes. In 1879, Oswald Schmiedeberg and Hans Meyer isolated from the urine of dogs administered , identifying it as the sugar moiety in a conjugated form of the compound and establishing its role in mammalian metabolism of foreign substances. This finding built on prior isolations of sugar-containing urinary metabolites, such as urochloralic acid in the 1870s by Von Mering and Musculus from human urine after administration, highlighting a pattern of glycoside-like conjugates in response to xenobiotics. These observations marked the initial recognition of glucuronidation as a physiological pathway, though the enzymatic mechanism remained elusive. Significant progress occurred in the mid-20th century with the elucidation of the biochemical basis of glucuronidation. In 1953, Geoffrey J. Dutton and Isabel D. E. Storey isolated uridine diphosphate glucuronic acid (UDPGA) from rat liver extracts, demonstrating its role as the activated donor of glucuronic acid in formation and resolving the cofactor required for the reaction. Concurrently, studies on bilirubin metabolism linked glucuronidation to ; in 1957, Rudi Schmid and colleagues identified direct-reacting in as its glucuronide conjugate, while Billing, Cole, and confirmed the primary excretory form as diglucuronide. By 1958, and Walker synthesized glucuronide using liver preparations, solidifying UDP-glucuronosyltransferases (UGTs) as the mediating enzymes and explaining deficiencies in conditions like . These discoveries, influenced by Luis Leloir's broader work on nucleotide sugars including UDP-glucose oxidation to UDPGA, transformed glucuronidation from an empirical observation to a defined enzymatic pathway. The molecular era began in the 1980s with advances in gene cloning and characterization of UGTs. In 1988, Harding et al. reported the first cloning of a human UGT cDNA (UGT1A1) using expression in COS-7 cells, enabling insights into its structure and substrate specificity for bilirubin. The 1990s saw cloning of the UGT1 and UGT2 gene families, revealing multiple isoforms and genetic polymorphisms affecting drug metabolism, such as UGT1A1*28 associated with Gilbert's syndrome. Nomenclature evolved from descriptive terms like "glucuronyl transferase" to a systematic superfamily classification in 1997 by Mackenzie et al., organizing UGTs into families (UGT1–UGT8) based on sequence homology and evolutionary divergence under IUPHAR guidelines. Structural studies advanced in the , providing atomic-level insights into UGT . In 2007, Miley et al. determined the 1.8 crystal structure of the UDPGA-binding domain of UGT2B7, revealing a GT-A fold with a Rossmann-like domain for binding and highlighting conserved motifs across isoforms. Subsequent work in the , including co-crystal structures like that of UGT2B15 with inhibitors, further delineated substrate-binding sites and informed polymorphism impacts on activity. These milestones underscored glucuronidation's complexity and its implications for pharmacogenetics up to the present.

Biochemical Aspects

Reaction Mechanism

Glucuronidation is a phase II metabolic reaction catalyzed by UDP-glucuronosyltransferases (UGTs), in which the moiety from the cofactor uridine 5'-diphospho-α-D- (UDPGA) is transferred to a nucleophilic on a , known as the aglycone, resulting in the formation of a water-soluble conjugate and the release of uridine 5'-diphosphate (UDP). This overall reaction can be represented as: \text{R-XH} + \text{UDPGA} \rightarrow \text{R-X-GlcA} + \text{UDP} where R-XH denotes the aglycone substrate with a nucleophilic group XH (e.g., hydroxyl, carboxyl), and GlcA is β-D-glucuronic acid. The reaction proceeds via a stepwise enzymatic process beginning with the binding of UDPGA to the of the , forming an . Subsequently, the aglycone substrate binds, creating a ternary , followed by a nucleophilic attack from the deprotonated functional group of the aglycone on the anomeric carbon (C1) of the moiety. This transfer occurs through a second-order (SN2) mechanism, which inverts the configuration at C1 and is facilitated by a catalytic dyad involving a residue that acts as a base to abstract a proton from the and an residue that stabilizes the protonated . Finally, the product is released, and dissociates from the . Kinetic studies of glucuronidation typically follow Michaelis-Menten kinetics, reflecting the bisubstrate nature of the reaction involving the aglycone and UDPGA. The is generally sequential, often described as a compulsory ordered bi-bi process where UDPGA binds first, or occasionally random ordered bi-bi, rather than ping-pong, with variations depending on the UGT isoform. The Michaelis constant () for UDPGA typically ranges from 50 to 200 μM across UGT isoforms. For effective glucuronidation, the aglycone must possess a nucleophilic site, such as a hydroxyl (-OH), carboxyl (-COOH), amino (-NH₂), or (-SH) group, capable of attacking the electrophilic C1 of . Planar and hydrophobic structural features in the aglycone often enhance binding to the hydrophobic cleft in the UGT , while at the nucleophilic site influences , determining which is preferentially conjugated.

Enzymes and Isoforms

Glucuronidation is catalyzed by a superfamily of enzymes known as UDP-glucuronosyltransferases (UGTs), which are integral membrane proteins primarily localized to the () of cells. In humans, the UGT superfamily comprises approximately 22 genes encoding functional isoforms divided into four subfamilies: UGT1, UGT2, UGT3, and UGT8. While UGT1 and UGT2 primarily catalyze glucuronidation using UDPGA, UGT3 and UGT8 utilize other UDP-sugars for (e.g., UDP-GlcNAc for UGT3, UDP-galactose for UGT8), though they share structural similarities. The UGT1 subfamily includes nine isoforms (UGT1A1–UGT1A10, excluding UGT1A2 which is a ), generated from a single locus on 2q37 through of unique first exons to four shared exons (exons 2–5), allowing for diverse substrate specificities while maintaining a conserved catalytic core. In contrast, the UGT2 subfamily consists of 13 isoforms clustered on 4q13, subdivided into UGT2A (three isoforms primarily in ) and UGT2B (seven to ten isoforms mainly in liver and other tissues); the UGT3 subfamily has two isoforms (UGT3A1 and UGT3A2) on 5p13, and UGT8 has one functional isoform on 4q26. Structurally, human UGT isoforms are type I transmembrane proteins consisting of 500–550 amino acids, featuring an N-terminal signal peptide, a single transmembrane helix anchoring the protein to the ER membrane with the bulk of the protein oriented toward the lumen, a variable N-terminal aglycone (substrate)-binding domain responsible for isoform-specific recognition, and a conserved C-terminal domain that binds the UDP-glucuronic acid (UDPGA) cofactor. The conserved region includes a signature 44-amino-acid motif (e.g., FXDQXG) critical for UDP-sugar binding and catalysis, while the variable N-terminal domain, encoded by exon 1 in UGT1A isoforms, confers broad but overlapping substrate specificity across the family. This modular architecture enables UGTs to accommodate diverse substrates, from small planar molecules to bulky compounds, by facilitating induced fit in the active site. Catalytically, UGT isoforms exhibit broad substrate promiscuity but with preferences that define their physiological roles; for instance, UGT1A1 is the primary for glucuronidating and certain anticancer drugs like , while UGT2B7 efficiently conjugates , steroids, and planar such as . UGT2B isoforms, in particular, show affinity for and planar structures, contributing to the of environmental toxins and drugs in hepatic and extrahepatic tissues. Isoform-specific activities often overlap, allowing functional redundancy, but variations in expression and kinetics ensure specialized contributions, such as UGT1A4's role in glucuronidation. The UGT superfamily demonstrates strong evolutionary across vertebrates, arising from ancient duplications that diversified the families while preserving core catalytic functions for and . For example, UGT8 shows over 90% sequence identity between and orthologs, reflecting its essential role in galactosylation of ceramides for synthesis, whereas UGT1 and UGT2 subfamilies exhibit adaptive expansions in mammals to handle complex xenobiotics. Structural studies since the , including those in the 2020s using and simulations, have elucidated dynamics and substrate-induced conformational changes in isoforms such as UGT1A1 and UGT2B7, revealing flexible loops that accommodate diverse aglycones. These insights highlight how isoform variability enhances glucuronidation's role in metabolic versatility.

Physiological Distribution

Primary Tissue Sites

Glucuronidation primarily occurs in the liver, where hepatocytes express a diverse array of UDP-glucuronosyltransferase (UGT) isoforms responsible for the majority of whole-body glucuronidation activity. This organ accounts for the bulk of systemic and clearance of endogenous and substrates through high UGT expression levels, including prominent isoforms such as UGT1A1, UGT1A4, UGT2B4, and UGT2B7. Within the liver lobule, UGT activity displays zonal heterogeneity, with pericentral regions exhibiting higher rates for certain isoforms and substrates; for example, glucuronidation of 7-hydroxycoumarin reaches maximal rates of 35 μmol/g/hr in pericentral hepatocytes compared to 9.6 μmol/g/hr in periportal areas. The represents another major site of glucuronidation, localized mainly in enterocytes, where it contributes around 10–15% of total UGT activity based on relative mRNA expression levels equivalent to about one-seventh of hepatic values. This tissue plays a critical role in presystemic , particularly for orally administered compounds, with high expression of isoforms like UGT1A8, UGT1A10, and UGT2B7 facilitating first-pass glucuronidation. The , particularly the proximal tubules, is a key extrahepatic site supporting glucuronidation for renal of conjugates, with isoforms such as UGT1A6, UGT1A9, and UGT2B7 driving activity that can achieve scaled clearances representing 30–43% of hepatic levels for select substrates such as (~33%) and (~30%). Minor contributions arise from tissues including the (UGT1A6 and UGT2B7 for inhaled xenobiotics), (UGT1A6 for neurotransmitters and UGT8 for maintenance), (UGT2B isoforms for local ), and (UGT2B4 and UGT2B7 for handling). During , the expresses UGT2B4, UGT2B15, and UGT2B17 to protect the from maternal toxins, though at relatively low overall activity levels. Developmentally, fetal liver UGT activity is markedly low, particularly for UGT1A1, leading to immature glucuronidation capacity at birth; postnatal maturation rapidly increases hepatic expression and function within weeks to months. Species variations influence distribution, with displaying proportionally higher extrahepatic UGT activity (e.g., in intestine and ) compared to humans, where hepatic dominance is more pronounced.

Cellular Localization and Expression

UDP-glucuronosyltransferase (UGT) enzymes, responsible for glucuronidation, are primarily localized to the (), where they function as integral membrane proteins with their catalytic domains oriented toward the lumenal side, facilitating the conjugation of substrates with UDP-glucuronic acid in a topologically restricted environment. This localization is conserved across mammalian UGT isoforms, with the di-lysine motif (KKXX or KXKXX) at the anchoring them to the membrane. While the majority of UGT activity is -associated, some isoforms exhibit partial association with the Golgi apparatus and membrane, potentially influencing intracellular trafficking and localization of glucuronides. Expression profiles of UGT isoforms vary significantly across tissues and cell types, reflecting their specialized roles in . In the liver, UGT1A1 and UGT1A4 are highly expressed, contributing substantially to and glucuronidation, while UGT2B7 predominates in both liver and , handling and substrates. In the intestine, UGT1A6 and UGT1A9 show elevated expression, aiding in the first-pass of dietary compounds. Sex- and age-specific patterns further modulate these profiles; for instance, UGT2B isoforms, such as UGT2B17, exhibit approximately four-fold higher expression in males compared to females in the liver, potentially impacting clearance and . Age-related changes, including developmental increases in hepatic UGT1A1 during infancy, also influence expression dynamics. Tissue-specific regulation of UGT expression is driven by distinct promoters for each isoform, particularly within the UGT1A family, where nine unique promoters control and mRNA production to match physiological demands. For example, hepatic UGT1A1 mRNA levels are approximately 10-fold higher than in the intestine, underscoring the liver's dominant role in systemic glucuronidation while intestinal expression supports local . These promoters respond to liver-enriched transcription factors like HNF-1α and HNF-4α, ensuring isoform-specific transcription in organs. Methods for mapping UGT cellular localization and expression include for protein detection in tissue sections, quantitative PCR (qPCR) for precise mRNA quantification, and more recently, single-cell RNA sequencing (scRNA-seq) to uncover intra-tissue heterogeneity. has visualized UGT1A1 in hepatocytes and enterocytes, while qPCR has quantified isoform abundance across samples; scRNA-seq studies from the 2020s reveal cell-type-specific variations, such as differential UGT2B expression in hepatic non-parenchymal cells, highlighting microenvironmental influences on glucuronidation capacity. The ER localization of UGTs enables functional coupling with phase I cytochrome P450 oxidases, which are also ER-embedded, allowing sequential oxidation and glucuronidation of lipophilic substrates within the same compartment to enhance solubility and excretion efficiency.

Substrates

Endogenous Substrates

Glucuronidation serves as a key for the conjugation and elimination of numerous endogenous compounds, enhancing their solubility to prevent toxicity and maintain physiological . These substrates include heme-derived products, hormones, vitamins, and mediators, primarily processed by UDP-glucuronosyltransferase (UGT) enzymes in the liver and other tissues. Bilirubin, a breakdown product of heme from senescent red blood cells, represents a major endogenous substrate for glucuronidation, predominantly catalyzed by UGT1A1 in the liver. This enzyme conjugates bilirubin with to form bilirubin diglucuronide, which is highly soluble and excreted into , preventing the accumulation of toxic unconjugated bilirubin that can lead to conditions like in neonates by crossing the blood-brain barrier. In healthy adults, approximately 250–300 mg of bilirubin is produced and glucuronidated daily, accounting for about 80% of total bilirubin turnover from hemoglobin catabolism. Steroid hormones undergo glucuronidation to regulate their activity and facilitate clearance, contributing to endocrine balance. Estrogens such as , estrone, and are conjugated primarily at phenolic or hydroxyl groups by multiple UGT isoforms, including UGT1A1, UGT1A3, UGT2B7, and UGT2B15, forming metabolites like estradiol-17β-glucuronide that are excreted in and bile. Androgens like testosterone and are mainly glucuronidated by UGT2B15 and UGT2B17 at the 17β-hydroxyl position, aiding in their inactivation and elimination. , including thyroxine (T4) and (T3), are also substrates, with glucuronidation occurring on phenolic hydroxyl or carboxyl groups via UGT1A1 and other isoforms, representing a significant pathway for their alongside deiodination and sulfation. Retinoic acid, a bioactive derivative of , is glucuronidated to all-trans-retinoyl-β-D-glucuronide, an that promotes excretion while retaining some biological activity, primarily mediated by hepatic UGTs. Similarly, vitamin D metabolites, such as 25-hydroxyvitamin D3, undergo glucuronidation at the 3β-hydroxyl position by UGT1A3 and UGT1A4, forming monoglucuronides that are secreted into for fecal elimination. These processes help control and signaling in and calcium . Other endogenous substrates include minor pathways for bile acids, which are glucuronidated at hydroxyl groups by UGT2A1 and UGT2B isoforms to reduce , though this is less prominent than sulfation in humans. Leukotrienes, such as , an inflammatory , form glucuronides via UGT1A1, UGT1A3, and UGT2B7, aiding in their urinary excretion. Fatty acids and their alcohols are also conjugated, with long-chain fatty alcohols serving as substrates for extrahepatic UGTs, supporting . Additionally, certain with endogenous origins, such as catecholamine metabolites, link dietary phenolics to glucuronidation pathways. The physiological significance of glucuronidating these substrates lies in maintaining homeostasis to avoid and , modulating levels to prevent endocrine disruptions, and regulating signaling for and immunity. Emerging research highlights interactions with the gut , where bacterial β-glucuronidases deconjugate these glucuronides, reactivating substrates like and influencing host-microbe and estrogen homeostasis in conditions such as hormone-related cancers.

Xenobiotic Substrates

Xenobiotic substrates of glucuronidation include diverse foreign compounds from environmental, dietary, and industrial sources, which are conjugated by UDP-glucuronosyltransferase (UGT) enzymes to enhance their and facilitate elimination. These processes primarily occur in the liver and intestine, serving as a critical barrier against toxic exposures. Unlike endogenous substrates, often require prior phase I , such as oxidation, to generate suitable nucleophilic groups for conjugation. Environmental toxins represent major xenobiotic substrates, particularly polycyclic aromatic hydrocarbons (PAHs) and pesticides. Phenolic metabolites of benzopyrene, a prevalent PAH in tobacco smoke and polluted air, are efficiently glucuronidated by UGT1A9 in human hepatic and extrahepatic tissues, contributing to their detoxification. Similarly, the pesticide carbaryl undergoes initial hydroxylation followed by glucuronidation, yielding significant metabolites like 5,6-dihydro-5,6-dihydroxycarbaryl glucuronide in rats, which aids in urinary excretion. Dietary compounds, especially plant-derived polyphenols, are extensively glucuronidated upon absorption. such as , abundant in fruits and , are rapidly converted to conjugates in the human intestinal mucosa and liver, with multiple UGT isoforms including UGT1A1 and UGT1A9 facilitating this process. Phenolic s from plants, like hydroxycinnamic acids, undergo glucuronidation at their hydroxyl groups primarily by intestinal and hepatic UGTs, which influences their and antioxidant efficacy. Industrial chemicals also serve as key substrates, with (BPA) and being prominent examples. BPA, a widespread , is predominantly metabolized via glucuronidation in the liver to form BPA-glucuronide, which is rapidly excreted in with a of approximately 2 hours. Phthalates, such as di(2-ethylhexyl) phthalate (DEHP), are conjugated through glucuronidation of their monoester metabolites, enhancing water solubility and promoting urinary elimination in humans. UGT isoforms display structural specificity for xenobiotic substrates, with preferences for planar versus bulky molecules. UGT1A enzymes, such as UGT1A9, often accommodate planar aromatics like PAH phenols, while UGT2B isoforms, including UGT2B7 and UGT2B17, favor bulkier structures, as exemplified by the glucuronidation of complex like and galangin. This selectivity ensures broad coverage of xenobiotic diversity. In , glucuronidation plays a pivotal role in detoxifying carcinogens by inactivating reactive intermediates and preventing formation, as observed with PAH metabolites. However, some glucuronides, such as those of dietary phenolics like genistin, can undergo enterohepatic recirculation, where deconjugate them via , leading to reabsorption and prolonged systemic exposure.

Regulation and Factors

Genetic and Epigenetic Influences

Genetic polymorphisms in the UDP-glucuronosyltransferase (UGT) gene family significantly influence glucuronidation activity by altering enzyme expression and function. A prominent example is the UGT1A128 variant, characterized by an extra TA dinucleotide repeat in the promoter region (A(TA)7TAA instead of A(TA)6TAA), which reduces transcriptional efficiency and thereby decreases conjugation capacity. This polymorphism is strongly associated with , a benign condition marked by mild unconjugated hyperbilirubinemia due to impaired hepatic glucuronidation of . Similarly, the UGT2B72 (rs7439366, c.802C>T, p.His268Tyr) modifies the enzyme's catalytic properties, potentially altering glucuronidation of certain substrates such as and other opioids, with mixed effects on efficacy and profiles in clinical settings. Epigenetic modifications provide another layer of for UGT expression, often silencing or enhancing activity without altering the DNA . Hypermethylation of the UGT1A1 promoter , for instance, represses transcription by recruiting repressive complexes, a mechanism observed in cells where it contributes to reduced detoxification capacity and increased irinotecan toxicity. Histone acetylation, conversely, promotes an open state that facilitates UGT1A1 transcription; deacetylation by histone deacetylases (HDACs) correlates with developmental silencing of the in non-hepatic tissues. MicroRNAs (miRNAs) also play a posttranscriptional role, with miR-122, a liver-enriched miRNA, upregulating UGT1A1 protein expression by targeting repressors or stabilizing mRNA, thereby supporting hepatic glucuronidation . Population-level variations in UGT polymorphisms underscore pharmacogenomic implications for . The UGT1A1*28 has a frequency of approximately 30-40% in Caucasian populations, compared to lower rates (around 10-20%) in Asian groups, leading to higher prevalence of and increased risk of adverse drug reactions, such as severe from , in carriers. These differences necessitate genotype-guided dosing to optimize therapeutic outcomes and minimize toxicity across diverse patient cohorts. Recent advances in the 2020s have leveraged CRISPR-Cas9 technology to edit UGT genes, offering potential curative strategies for disorders like Crigler-Najjar syndrome. In a 2023 study, somatic correction of a Ugt1a1 one-base deletion in a model using CRISPR-Cas9 reduced bilirubin levels and improved survival, demonstrating precise restoration of function without off-target effects. As of 2025, the ICH M12 guideline emphasizes evaluation of UGT1A1, UGT1A4, and UGT2B7 in drug-drug interaction studies to better predict pharmacokinetic variability. Recent research also highlights PI3K/AKT signaling enhancing glucuronidation in colon cancer cells to evade . Epigenetic therapies, such as HDAC inhibitors (e.g., ), are also emerging to modulate UGT expression; by inhibiting deacetylation, these agents enhance at UGT promoters, potentially augmenting glucuronidation in conditions like cancer where silencing occurs. Such interventions highlight the therapeutic promise of targeting epigenetic marks to fine-tune UGT activity.

Physiological and Environmental Factors

Glucuronidation activity undergoes significant changes throughout human development. In neonates, hepatic UGT1A1 expression and activity are markedly reduced, contributing to physiologic through impaired conjugation and excretion. This immaturity typically resolves postnatally as UGT1A1 levels increase, reaching peak expression and function in adulthood. In the elderly, UGT activity is relatively preserved compared to phase I metabolism, though it may be slightly reduced in some individuals due to decreased hepatic mass and altered enzyme expression, potentially impacting drug clearance minimally. Hormonal factors play a key role in modulating UGT expression. Estrogens, such as , upregulate UGT2B isoforms, including UGT2B15, in estrogen receptor-positive tissues like cells, enhancing the glucuronidation of substrates like androgens and xenobiotics. Circadian rhythms also influence hepatic UGT expression; for instance, the clock component Rev-erbα regulates the diurnal of UGT1A9 in mouse liver, affecting glucuronidation rates that peak during the active phase. Environmental exposures can induce or inhibit glucuronidation. Polycyclic aromatic hydrocarbons (PAHs) in cigarette smoke upregulate UGT1A1 via the pathway, increasing and conjugation in smokers. Conversely, in , such as and naringenin, potently inhibit multiple UGT isoforms (e.g., UGT1A1, UGT1A3, UGT1A9, UGT2B7) with IC₅₀ values below 10 μM, potentially leading to herb-drug interactions by reducing glucuronidation of substrates like or analgesics. Disease states often downregulate UGT activity. Acute suppresses hepatic UGT1A1 and UGT2B expression through pro-inflammatory cytokines like IL-6 and TNF-α, decreasing glucuronidation of and drugs during or . Obesity alters UGT profiles, with hepatic associated with increased UGT mRNA in some mouse models but reduced activity for isoforms like UGT1A9 in human liver, contributing to dysregulated metabolism of endogenous and exogenous compounds. Nutritional factors influence UGT function, particularly in the liver and gut. High-fat diets suppress hepatic UGT1A1 activity, as seen in non-alcoholic models where accumulation inhibits glucuronidation and exacerbates hyperbilirubinemia. Emerging microbiome research highlights how modulate gut-associated glucuronidation; by altering β-glucuronidase activity in the , like species can reduce deconjugation of glucuronides, indirectly enhancing net host UGT efficiency for endobiotics such as estrogens.

Clinical Implications

Drug Metabolism and Interactions

Glucuronidation plays a significant role in the of numerous pharmaceuticals, facilitating their detoxification and elimination by conjugating them with , primarily through UDP-glucuronosyltransferase (UGT) enzymes. This phase II process accounts for the clearance of approximately 10% of the top 200 prescribed drugs, underscoring its importance in . For instance, analgesics like are primarily metabolized via UGT2B7 to form active morphine-6-glucuronide, which contributes to analgesia, and inactive morphine-3-glucuronide. Nonsteroidal anti-inflammatory drugs (NSAIDs) such as naproxen undergo acyl glucuronidation, mainly by UGT2B7, to produce a that is excreted in and , aiding in the resolution of without significant accumulation. Antivirals, including , are glucuronidated by UGT2B7 to form the major inactive 5'-O-glucuronyl zidovudine, which constitutes about 60-75% of the dose in , enhancing its solubility for renal clearance. The kinetics of glucuronidation vary by substrate and enzyme isoform, influencing efficacy and safety. , an antiepileptic, is predominantly cleared through UGT1A4-mediated N-glucuronidation, with over 90% of the dose metabolized to the inactive 2-N-glucuronide, leading to a clearance of approximately 0.9-1.3 mL/min/kg in adults. This pathway's efficiency is evident in the drug's of 25-33 hours, but it can accelerate during physiological changes like due to upregulated UGT expression. Overall, glucuronidation's contribution to clearance is substrate-specific, with some compounds like relying on UGT1A1 for the formation of the SN-38-glucuronide, where impaired activity prolongs SN-38 exposure and heightens toxicity risk. Drug-drug interactions mediated by glucuronidation are increasingly recognized, particularly in scenarios common in chronic disease management. Inhibitors such as atazanavir, an protease inhibitor, potently suppress UGT1A1 activity, reducing glucuronidation and causing asymptomatic hyperbilirubinemia in up to 50% of patients, though this rarely leads to discontinuation. Conversely, inducers like rifampin upregulate UGT1A9 expression via the pregnane X receptor, accelerating the metabolism of substrates like and potentially necessitating dose adjustments to maintain therapeutic levels. In , these interactions amplify risks, as co-administration of multiple UGT substrates or modulators can alter exposure by 20-50%, contributing to suboptimal efficacy or adverse events. Pharmacogenomic variations in UGT genes further modulate drug responses, guiding personalized dosing. The UGT1A128 polymorphism, characterized by a TA repeat expansion in the promoter, reduces activity by 70-80% in homozygotes, increasing irinotecan-induced and risk by 2-4 fold, prompting FDA-recommended dose reductions from 350 mg/m² to 200-250 mg/m². Similar variants in UGT2B7 affect opioid glucuronidation, with the UGT2B72 linked to higher morphine-6-glucuronide levels and enhanced analgesia in some populations. These genetic insights highlight the need for preemptive testing in high-risk therapies. Recent 2025 analyses have strengthened associations between UGT inhibition and drug-induced (DILI), showing that non-CYP inhibitors, including UGT modulators, independently predict high DILI concern in over 30% of cases, emphasizing their role in polypharmacy-related .

Associated Diseases and Disorders

is a benign resulting from mild deficiency in the UDP-glucuronosyltransferase 1A1 (UGT1A1) , primarily due to promoter such as the TA repeat polymorphism (UGT1A1*28). This leads to reduced glucuronidation of , causing intermittent unconjugated hyperbilirubinemia without liver damage or , often triggered by , , or infections. The condition has a prevalence of approximately 5-10% in the general population and is typically asymptomatic, though it may exacerbate in combination with other factors. Crigler-Najjar syndrome encompasses severe inherited defects in UGT1A1, classified into type I (complete enzyme absence due to biallelic null mutations) and type II (partial activity from missense mutations, retaining 5-25% function). Both types cause profound unconjugated hyperbilirubinemia, with type I presenting in neonates and posing a high risk of —a neurotoxic from deposition in the —while type II manifests later with milder symptoms. Type I requires lifelong intensive phototherapy to isomerize for excretion, often supplemented by , whereas type II responds to induction of residual UGT1A1 activity, reducing levels by 25-50%. Liver transplantation remains curative for type I, but trials using adeno-associated viral vectors to deliver functional UGT1A1 have shown promising reductions in early 2020s phase I/II studies, with durable effects up to two years post-administration. As of 2025, updates from trials such as GNT-018-IDES demonstrate successful AAV in patients with pre-existing anti-AAV antibodies using imlifidase pretreatment, further advancing treatment options for this rare disorder. Dysregulation of UGT enzymes plays a dual role in cancer progression and therapy . In , UGT polymorphisms, such as UGT2B7*2 (His268Tyr), reduce glucuronidation of active metabolites like endoxifen, potentially elevating their levels and altering or risk, though clinical mechanisms remain complex due to variable metabolite clearance. Conversely, UGT overexpression in tumors contributes to by accelerating inactivation of chemotherapeutics; for instance, GLI1 induction upregulates UGT1A isoforms in resistant cells, enhancing glucuronidation of drugs like cytarabine in and Hsp90 inhibitors in , as demonstrated in studies from 2014-2015. Recent analyses (2015-2025) confirm GLI1-mediated UGT elevation stabilizes enzymes via inhibition, promoting broad-spectrum reversible by GLI1 inhibitors like vismodegib. UGT1A1 variants, including rs4148323 (G71R) and promoter polymorphisms, are associated with prolonged neonatal unconjugated hyperbilirubinemia and increased risk, contributing to hazardous levels in up to 20-30% of affected infants in certain populations, often requiring phototherapy to prevent . In cholestatic liver diseases, impaired biliary hinders glucuronide elimination, leading to accumulation of detoxified bile acids and exacerbating , as glucuronidation serves as a compensatory urinary clearance pathway under severe . Therapeutic strategies for glucuronidation-related disorders emphasize enzyme induction and replacement. effectively induces residual UGT1A1 in Crigler-Najjar type II, normalizing in 70-80% of cases, while adjunct therapies like fenofibrate upregulate UGT2B7 for bile acid glucuronidation in models. Emerging advances include hepatocyte transplantation for partial enzyme replacement and CRISPR-based gene editing in preclinical trials, alongside ongoing AAV-mediated gene therapies that have achieved 20-50% reduction in Crigler-Najjar patients without transplantation.

References

  1. [1]
    Glucuronidation: Driving Factors and Their Impact ... - PubMed Central
    Glucuronidation is a well-recognized phase II metabolic pathway for a variety of chemicals including drugs and endogenous substances.
  2. [2]
    Glucuronidation - an overview | ScienceDirect Topics
    Glucuronidation is a process where glucose is used for conjugation, playing a significant role in endogenous and xenobiotic metabolism.
  3. [3]
    New Perspectives on Acyl Glucuronide Risk Assessment in Drug ...
    Glucuronidation is the conjugation of glucuronic acid to various functional groups, catalyzed by UDP-glucuronosyl-transferases (UGTs) in the presence of ...3. Results · 3.2. Reactivity · 4. Discussion<|separator|>
  4. [4]
  5. [5]
    Oswald Schmiedeberg (1838–1921) - PubMed Central - NIH
    Jul 23, 2024 · Together with Schmiedeberg, he discovered glucuronic acid as the most important reaction partner of drugs. He also described the relationship ...
  6. [6]
    Glucuronidation - International Society for the Study of Xenobiotics
    It wasn't until 1953 that this process was elucidated through the discovery of uridine diphosphoglucuronic acid (UDPGA) by Dutton and Storey and their ...
  7. [7]
  8. [8]
  9. [9]
    Luis Federico Leloir, 6 September 1906 - 3 December 1987
    UDPG was also found to undergo enzymic oxidation to UDP-glucuronic acid which is the donor of the glucuronyl residue in the synthesis of glucuronides. Other.
  10. [10]
  11. [11]
  12. [12]
  13. [13]
    Crystal Structure of the Cofactor-Binding Domain of the Human ...
    We report the 1.8 Å resolution apo crystal structure of the UDP-glucuronic acid binding domain of human UGT isoform 2B7 (UGT2B7), which catalyzes the ...
  14. [14]
    Enzyme Kinetics of Uridine Diphosphate Glucuronosyltransferases ...
    Glucuronidation is a bisubstrate reaction that requires the aglycone and the cofactor, UDP-GlcUA. Accumulating evidence suggests that the bisubstrate reaction ...
  15. [15]
    The crystal structure of human UDP-glucuronosyltransferase 2B7 C ...
    GT-A proteins consist of a single α/β/α sandwich that resembles a Rossman-like fold and contains a divalent metal that is important for donor ligand binding. GT ...
  16. [16]
  17. [17]
    The UDP-Glycosyltransferase (UGT) Superfamily: New Members ...
    Feb 6, 2019 · UGT1A6 displays high activity towards planar phenols and arylamines (55), and major xenobiotic substrates include 1-naphthol, 4 ...
  18. [18]
    Glucuronidation of 7-hydroxycoumarin in periportal and pericentral ...
    Maximal rates of glucuronidation in periportal and pericentral regions of the liver lobule calculated with this new method were 9.6 and 35 mumoles/g/hr, ...Missing: zonal distribution UGT
  19. [19]
    Use of Isoform-Specific UGT Metabolism to Determine and Describe ...
    ... glucuronidation rate of the three individual UGTs (2.44 nmol/min/mg). Similarly, the glucuronidation rates at 10 µM were 4.91±0.13 nmol/min/mg, which was ...
  20. [20]
    Challenges and Opportunities with Predicting in Vivo Phase II ...
    Glucuronidation is the most important phase II metabolic pathway which is responsible for the clearance of many endogenous and exogenous compounds.Missing: definition | Show results with:definition
  21. [21]
    Characterization of In Vitro Glucuronidation Clearance of a Range of ...
    This study characterized glucuronidation clearance (CLint, UGT) in human kidney, liver, and intestinal microsomes in the presence and absence of bovine serum ...
  22. [22]
    Investigation of the Endoplasmic Reticulum Localization of UDP ...
    UDP-Glucuronosyltransferase (UGT) is one of the major drug-metabolizing enzymes catalyzing glucuronidation, and 22 isoforms have been identified so far.
  23. [23]
    Regulation of UDP-Glucuronosyltransferase 1A1 Expression ... - NIH
    Each UGT1A gene is regulated by an individual promoter, driving tissue-specific expression. UGTs 1A1, 1A3, 1A4, 1A5, 1A6, and 1A9 are the primary hepatic ...
  24. [24]
    Quantitative Analysis of UDP-Glucuronosyltransferase Ugt1a and ...
    Ugt2b mRNA levels represented approximately 80% of total Ugt mRNA levels in the liver, whereas Ugt1a mRNA levels accounted for almost 90% in the small intestine ...
  25. [25]
    Sex Differences in UDP-Glucuronosyltransferase 2B17 Expression ...
    Men have about 4-fold higher UGT2B17 expression and higher glucuronidation activity than women, potentially affecting carcinogen detoxification.
  26. [26]
    Quantitative analysis of the UDP‐glucuronosyltransferase ...
    Nov 20, 2023 · However, in normal tissues, the tissue-specific expression profiles and transcriptional regulation of the UGTs are still not fully understood.
  27. [27]
    The regulation of UDP-glucuronosyltransferase genes by tissue ...
    The factors controlling UGT gene expression in the liver include the liver-enriched transcription factors, HNF-1alpha and HNF-4alpha, several members of the ...
  28. [28]
    Quantification of Hepatic UDP Glucuronosyltransferase 1A Splice ...
    The nested qPCR developed in this study was used to determine the relative expression of UGT1A1 v1 and UGT1A1 v2/v3 in total RNA isolated from the liver ...
  29. [29]
    Integrated analysis of single-cell RNA-seq and spatial ... - Frontiers
    Jun 25, 2025 · Methods: We employed functional enrichment algorithms, including AUCell and UCell, to assess lactylation activity in GBM cancer cells.
  30. [30]
    Physiology, Bilirubin - StatPearls - NCBI Bookshelf - NIH
    Many different isoforms of UDPGT exist, but the physiologically important isoform in bilirubin glucuronidation is UDPGT1A1. The enzyme esterifies 2 glucuronide ...
  31. [31]
    Bilirubin is an Endogenous Antioxidant in Human Vascular ... - NIH
    Jul 6, 2016 · Approximately 250–300 mg bilirubin/day is formed in a normal adult. Most of bilirubin arises in the spleen from the catabolism of ...
  32. [32]
    Glucuronides in the gut: Sugar-driven symbioses between microbe ...
    Apr 7, 2017 · Thus, bacterial GUS enzymes appear to play an important role in health and disease by metabolizing glucuronides in the gut.Missing: definition | Show results with:definition
  33. [33]
    Metabolism of Thyroid Hormone - Endotext - NCBI Bookshelf
    Jan 1, 2017 · The remainder of T4 is metabolized by different pathways, in particular glucuronidation and sulfation. T3 is further metabolized largely by IRD ...CLINICAL SUMMARY · THYROID HORMONE... · Type III iodothyronine...
  34. [34]
    Retinoic acid metabolism and mechanism of action: a review
    Retinoic acid (RA) acts through RAR and RXR receptors, and derives from retinol via oxidation. RA is then metabolized by CYPs and glucuronidation.
  35. [35]
    Human UGT1A4 and UGT1A3 Conjugate 25-Hydroxyvitamin D3 - NIH
    Two isozymes, UGT1A4 and UGT1A3, were identified as the principal catalysts of 25OHD3 glucuronidation in human liver. Three 25OHD3 monoglucuronides (25OHD3-25- ...
  36. [36]
    Why bile acid glucuronidation is a minor pathway for conjugation of ...
    Why bile acid glucuronidation is a minor pathway for conjugation of endogenous bile acids in man.
  37. [37]
    Quantification of leukotriene B4 glucuronide in human urine - PubMed
    We have developed a method for measuring leukotriene B4 glucuronide, a marker of systemic leukotriene B4 biosynthesis, in human urine.
  38. [38]
    Regulation of Sulfotransferase and UDP-Glucuronosyltransferase ...
    Endogenous UGT substrates include bilirubin, neutral steroids, bile acids, fatty acids, and retinoids [74, 78, 79].
  39. [39]
    Gut microbial beta-glucuronidase: a vital regulator in female ...
    Aug 9, 2023 · Gut microbial beta-glucuronidase (gmGUS) is a key factor regulating host estrogen metabolism, maintaining physiological estrogen levels.
  40. [40]
    Characterization of benzo(a)pyrene-trans-7,8-dihydrodiol ... - PubMed
    Apr 1, 2002 · These results suggest that several UGTs may play an important role in the overall glucuronidation of BPD in humans, with UGT1A1, UGT1A7, UGT1A9, ...
  41. [41]
    5,6-Dihydro-5,6-dihydroxycarbaryl glucuronide as a significant ...
    5,6-Dihydro-5,6-dihydroxycarbaryl glucuronide as a significant metabolite of carbaryl in the rat | Journal of Agricultural and Food Chemistry.
  42. [42]
    METABOLIC CONVERSION OF DIETARY FLAVONOIDS ALTERS ...
    Quercetin is rapidly metabolized in the human intestinal mucosa and liver into glucuronide and sulphate conjugates with or without methylation (Scheme III).
  43. [43]
    In vitro and in vivo conjugation of dietary hydroxycinnamic acids by ...
    The glucuronidation and sulfation of xenobiotics in the intestine and liver are carried out by UDP-glucuronosyltransferases (UGTs) and sulfotransferases (SULTs) ...
  44. [44]
    An insight into bisphenol A, food exposure and its adverse effects on ...
    Bisphenol A is highly metabolized and secreted into urine, primarily as a glucuronide conjugate with a half-life of 2 h (53). BPA's half-life also depends on ...
  45. [45]
    Glucuronidation Activity of the UGT2B17 Enzyme toward Xenobiotics
    Evidence showed that galangin, chrysin, 7hydroxyflavone and naringin were substrates for UGT2B17, among which galangin had the highest conversion value, whereas ...
  46. [46]
    Enterohepatic Circulation and Pharmacokinetics of Genistin and ...
    Oct 24, 2019 · In general, enterohepatic recirculation may prolong the pharmacol. effect of certain drugs and drug metabolites. Of particular importance is the ...<|separator|>
  47. [47]
    very important pharmacogene information for UGT1A1 - PMC - NIH
    Gilbert's syndrome can be caused by a variety of genetic changes, but within White and African-American populations it is most commonly attributed to the UGT1A1 ...
  48. [48]
    the UGT1A1*28 promoter polymorphism as a biomarker of ... - PubMed
    Gilbert syndrome belongs to the group of the most common human metabolic disorders and is characterized by an elevated level of bilirubin in blood serum.
  49. [49]
    Association between UGT2B7 gene polymorphisms and fentanyl ...
    The metabolism of morphine by glucuronidation is known to be influenced by polymorphisms of the UGT2B7 gene. Some metabolic products of fentanyl are reportedly ...
  50. [50]
    Irinotecan inactivation is modulated by epigenetic silencing of ...
    Mar 15, 2006 · We conclude that DNA methylation represses UGT1A1 expression in colon cancer and that this process may contribute to the level of tumoral ...
  51. [51]
    Epigenetics and microRNAs in UGT1As - PMC - NIH
    May 25, 2021 · Epigenetics regulates UGT1As pretranscriptionally through DNA methylation and histone modification, and miRNAs are considered the key mechanism of ...
  52. [52]
  53. [53]
    Association Between the UGT1A1*28 Allele, Bilirubin Levels, and ...
    Sep 25, 2006 · The allele frequency of this polymorphism among the white population is extraordinarily high, 34% to 40%, and the frequency of individuals ...
  54. [54]
    Distribution of the UGT1A1*28 polymorphism in Caucasian and ...
    In conclusion, consistent with previous reports, the UGT1A1 (TA)7/7 genotype was significantly more common in Caucasians than in Asians. UGT1A1 (TA)n/n genotype ...
  55. [55]
    CRISPR-Cas9-mediated somatic correction of a one-base deletion ...
    Nov 19, 2023 · We reduced plasma bilirubin to safe levels and partially rescued neonatal lethality by correcting the mutant Ugt1a1 gene of a Crigler-Najjar mouse model.Missing: 2020s | Show results with:2020s
  56. [56]
    Developmental hyperbilirubinemia and CNS toxicity in mice ... - PNAS
    High levels of unconjugated bilirubin (UCB) in newborn children is associated with a reduction in hepatic UDP glucuronosyltransferase (UGT) 1A1 activity ...
  57. [57]
    Age-Related Changes in Hepatic Activity and Expression of ... - NIH
    We hypothesize that the decrease in UGT activity observed in this study may be caused by the loss of smooth endoplasmic reticulum in hepatocytes, reduced ...
  58. [58]
    Estrogen Regulation of the Glucuronidation Enzyme UGT2B15 in ...
    UGT2B15 is the only UGT2B enzyme up-regulated by estrogen, and marked estradiol stimulation of UGT2B15 mRNA levels is observed, in a time- and dose-dependent ...
  59. [59]
    Circadian Clock Component Rev-erb α Regulates Diurnal Rhythm of ...
    Jun 11, 2020 · Loss of Rev-erbα (a circadian clock component) downregulated the Ugt1a9 expression and blunted its rhythm in mouse liver. Accordingly, propofol ...
  60. [60]
    Cigarette Smoking and Irinotecan Treatment - ASCO Publications
    PAHs are also known to induce some isoforms of the uridine diphosphate glucuronosyltransferase (UGT) family, which includes important enzymes involved in ...
  61. [61]
    Biol. Pharm. Bull. 39(12): 2052-2059 (2016) - J-Stage
    modestly inhibit UGT isoforms. ... These data should be useful for guiding applications of orange juice or grapefruit juice, HET or NGR and related pharmaceutical ...
  62. [62]
    Effects of Pro-Inflammatory Cytokines on Hepatic Metabolism ... - NIH
    Nov 28, 2022 · Among the UGT family, UGT1A1 is involved in the glucuronidation of various endogenous substrates such as bilirubin and estradiol, and drugs such ...
  63. [63]
    UDP-Glucuronosyltransferase Expression in Mouse Liver Is ...
    The purpose of this study was to determine whether steatosis induced by obesity or fasting could affect liver Ugt mRNA expression and activity. ... The findings ...
  64. [64]
    Suppressing Hepatic UGT1A1 Increases Plasma Bilirubin, Lowers ...
    In this study, male C57BL/6J mice were fed a high-fat diet (HFD, 60%) for 30 weeks to induce NAFLD and were treated subcutaneously with GNUR or sham (CTRL) once ...
  65. [65]
    CHAPTER 13: Glucuronidation - Books
    Aug 27, 2021 · Glucuronidation is a major metabolic pathway for both endogenous chemicals and drugs. About 10% of the top 200 prescribed drugs are metabolized ...
  66. [66]
    Human UGT2B7 Catalyzes Morphine Glucuronidation - ScienceDirect
    UGT2B7 catalyzed morphine glucuronidation at the 3- and 6-hydroxy positions and also mediated the formation of codeine-6-glucuronide from codeine.
  67. [67]
    Naproxen Pathway, Pharmacokinetics - ClinPGx
    The parent drug can be glucuronidated, mainly by UGT2B7, and the desmethyl metabolite can be glucuronidated to form either the phenolic glucuronide or the acyl ...
  68. [68]
    Induction of zidovudine glucuronidation and amination pathways by ...
    Uridine diphosphate-glucuronosyl transferase (UGT) in the liver catalyses the glucuronidation of zidovudine [7]. The formation of AMT appears to be mediated by ...
  69. [69]
    Toward Precision Dosing of Lamotrigine During Pregnancy ...
    Feb 24, 2025 · Lamotrigine is 90% metabolized via the glucuronidation pathway, with the remaining 10% excreted in the urine as a prototype [6]. The ...
  70. [70]
    Model-based lamotrigine clearance changes during pregnancy - NIH
    The objective of the study was to characterize changes in the oral clearance (CL/F) of lamotrigine (LTG) over the course of pregnancy and the postpartum period.
  71. [71]
    Atazanavir Therapy and UGT1A1 Genotype - NCBI - NIH
    Oct 17, 2023 · Atazanavir inhibits the uridine diphosphate-glucuronosyltransferase 1A1 (UGT1A1) enzyme—the only enzyme that conjugates bilirubin for ...
  72. [72]
    In vitro inhibition of UDP glucuronosyltransferases by atazanavir and ...
    These results suggest that a direct inhibition of UGT1A1-mediated bilirubin glucuronidation may provide a mechanism for the reversible hyperbilirubinemia.
  73. [73]
    Effect of Rifampin on the Pharmacokinetics of Ertugliflozin in Healthy ...
    Rifampin induces UGT1A9, UGT2B7, CYP3A4, and CYP3A5. Because concurrent ... UGT and CYP inducer, such as rifampin. Keywords: coadministration ...
  74. [74]
    Irinotecan Therapy and UGT1A1 Genotype - NCBI - NIH
    May 27, 2015 · Homozygous UGT1A1*28 patients have a greatly increased risk of hematological toxicity (neutropenia) compared with other genotypes, ...
  75. [75]
    All You Need to Know About UGT1A1 Genetic Testing for Patients ...
    Dec 3, 2021 · A model-based clinical simulation study demonstrated that the risk of severe irinotecan toxicity in patients carrying UGT1A1*28 could be reduced ...
  76. [76]
    Effect of UGT2B7 -900G>A (-842G>A; rs7438135) on morphine ...
    Our small pilot study illustrates that in addition to gestational and postnatal age, the UGT2B7 -900G>A polymorphism significantly alters morphine ...
  77. [77]
    Drug interaction with UDP-Glucuronosyltransferase (UGT) enzymes ...
    Our study revealed that drugs that inhibit non-CYP enzymes are significantly associated with high DILI concern.
  78. [78]
    Gilbert Syndrome - StatPearls - NCBI Bookshelf - NIH
    Oct 5, 2024 · This autosomal recessive condition leads to mild to moderate unconjugated hyperbilirubinemia, often presenting as recurrent episodes of jaundice ...
  79. [79]
    The Genetic Basis of the Reduced Expression of Bilirubin UDP ...
    Nov 2, 1995 · People with Gilbert's syndrome have mild, chronic unconjugated hyperbilirubinemia in the absence of liver disease or overt hemolysis.
  80. [80]
    Crigler-Najjar Syndrome - StatPearls - NCBI Bookshelf - NIH
    Feb 12, 2024 · Neonatal jaundice and prematurity: Physiologic jaundice in newborns can result from various factors like liver dysfunction, infection, ...
  81. [81]
    Gene Therapy in Patients with the Crigler–Najjar Syndrome
    Aug 16, 2023 · Prolonged daily phototherapy partially and transiently controls the jaundice, but the only definitive cure is liver transplantation.
  82. [82]
    Crigler Najjar Syndrome - Symptoms, Causes, Treatment | NORD
    Aug 2, 2016 · Crigler-Najjar syndrome is a rare genetic disorder characterized by an inability to properly convert and clear bilirubin from the body.
  83. [83]
    Potential Role of UGT Pharmacogenetics in Cancer Treatment ... - NIH
    Tamoxifen (TAM) is a selective estrogen receptor modulator that is widely used in the prevention and treatment of estrogen receptor-positive (ER+) breast cancer ...Missing: GLI1 | Show results with:GLI1
  84. [84]
    GLI1-Induced Drug Glucuronidation in Resistant Cancer Cells - NIH
    Mar 25, 2015 · We demonstrate that overexpression of UGTs can contribute to oncogenesis by promoting drug resistance.Missing: tamoxifen breast
  85. [85]
    Emerging roles for UDP-glucuronosyltransferases in drug resistance ...
    Feb 12, 2020 · UGTs are membrane-bound enzymes. They predominantly localise in the endoplasmic reticulum (ER), where the glucuronidation reaction takes place ...Ugts As Mediators Of Drug... · Inducible Ugt Expression And... · Ugts And Cancer Progression
  86. [86]
    UGT1A1 mutation association with increased bilirubin levels and ...
    Jun 1, 2021 · Our study demonstrated that UGT1A1 variants contributed to the increased bilirubin level and risk of developing hazardous neonatal ...
  87. [87]
    Should We Consider UGT1A1 Mutation Analysis in Evaluating the ...
    Feb 29, 2024 · Neonates with prolonged unconjugated hyperbilirubinemia may have a higher chance of UGT1A1 mutation than expected. Analysis of UGT1A1 mutations ...
  88. [88]
    Urinary Elimination of Bile Acid Glucuronides under Severe ...
    In conclusion, the present study evidences the major impact that bile flow restoration exerts on the circulating and urinary bile acid-glucuronide profiles and ...
  89. [89]
    Crigler Najjar Syndrome - an overview | ScienceDirect Topics
    In Crigler–Najjar syndrome type 2, the activity of UDP-glucuronosyl transferase can be increased by giving phenobarbital (which induces synthesis of the enzyme) ...Drug Metabolism · Clinical Trials And... · Methods Aimed At Replacing...
  90. [90]
    Adjunct Fenofibrate Up‐regulates Bile Acid Glucuronidation...
    This study investigates the efficacy of adjunct fenofibrate therapy to up‐regulate BA‐glucuronidation and reduce serum BA toxicity during cholestasis.