Abstract
Glucuronidation is a well-recognized phase II metabolic pathway for a variety of chemicals including drugs and endogenous substances. Although it is usually the secondary metabolic pathway for a compound preceded by phase I hydroxylation, glucuronidation alone could serve as the dominant metabolic pathway compounds, including some with high aqueous solubility. Glucuronidation involves the metabolism of parent compound by UDP-glucuronosyltransferases (UGTs) into hydrophilic and negatively charged glucuronides that cannot exit the cell without the aid of efflux transporters. Therefore, elimination of parent compound via glucuronidation in a metabolic active cell is controlled by two driving forces; the formation of glucuronides by UGT enzymes and the (polarized) excretion of these glucuronides by efflux transporters located on the cell surfaces in various drug disposition organs.
Contrary to the common assumption that the glucuronides reaching the systemic circulation were destined for urinary excretion, recent evidences suggest that hepatocytes are capable of highly efficient biliary clearance of the gut-generated glucuronides. Furthermore, the biliary- and enteric-eliminated glucuronides participate into recycling schemes involving intestinal microbes, which often prolong their local and systemic exposure, albeit at low systemic concentrations. Taken together, these recent research advances indicate that though UGT determines the rate and extent of glucuronide generation, the efflux and uptake transporters determine the distribution of these glucuronides into blood and then to various organs for elimination. Recycling schemes impact the apparent plasma half-life of parent compounds and their glucuronides that reach intestinal lumen, in addition to prolonging their gut and colon exposure.
Keywords: Glucuronidation, glucuronides, UGT, disposition, recycling, interplay, efflux transporters, uptake transporters
1. Introduction
Glucuronidation Process.
Glucuronidation is an enzyme reaction process catalyzed by UDP-glucuronosyltransferases (i.e., UGTs) in different animals including humans, as well as plants and bacteria (Mackenzie et al., 2003) (Nagar and Blanchard, 2006; Nagar and Remmel, 2006). Glucuronidation process attaches a glucuronide moiety to a substrate making a product that is highly hydrophilic (Radominska-Pandya et al., 1998; Tukey and Strassburg, 2000; Mackenzie et al., 2003; Nagar and Blanchard, 2006; Nagar and Remmel, 2006; Perera et al., 2008). The glucuronides are then often eliminated via bile or urine. Therefore, glucuronidation is considered to be a detoxification process or a defense mechanism that helps humans remove unwanted substances including endogenous substances (e.g., bilirubin), drugs (e.g., SN-38) and other xenobiotics (e.g., environmental toxins) from the body. For example, genetic deficiency related to UGT1A1 could result in hyperbilirubinemia, a disease called Gilbert’s syndrome (Radominska-Pandya et al., 1998; Tukey and Strassburg, 2000; Mackenzie et al., 2003; Nagar and Blanchard, 2006; Nagar and Remmel, 2006; Perera et al., 2008). Hence, glucuronidation is an essential biological process in humans, protecting us from excessive accumulation of toxic substances in the body.
Study of the glucuronidation processes started about the same time as the study of cytochrome P450 (or CYP). Initial report of human cytochrome P450 was in 1960s (Reynolds, 1966) and the first human CYP isoform was cloned in 1985 (actually a partial cloning of human CYP2A6) (Phillips et al., 1985). Similarly, human UGT catalyzed reaction was also first reported in the 1960s (Pogell and Leloir, 1961) and the first human UGT was cloned in 1988 (Harding et al., 1988). However, there are major differences between these two enzyme superfamilies (UGTs and CYPs) with respect to the volume of research. A PubMed search, conducted on July 16 of 2016, using the keyword combination of “human cytochrome P450” generated 46,604 hits, whereas the same search using the keyword combination of “human glucuronide” only generated 7257 hits (a keyword combination of “human glucuronidation” only generated 2984 hits). Therefore, we often and quite accurately believe that we collectively know more about CYPs than UGTs.
Glucuronidation of Drugs and Endogenous Substances.
Despite apparent limitations relative to CYP studies, significant amount of information exists on glucuronidation, especially glucuronidation of drugs by UGTs. Specifically, these enzymes are broadly but unevenly distributed throughout various cells, tissues and organs with heavy concentrations in the first-pass metabolism organs (i.e., liver and intestine) as well as the major elimination organ (i.e., kidney). Glucuronidation serves as the primary elimination pathway for a variety of drugs on the market (Table 1). However, in contrast to a relatively small number of drugs with glucuronidation as primary elimination pathway, for a vast majority of drugs, glucuronidation often occurs as a secondary step after the primary metabolites are produced by phase I reaction such as hydrolysis, hydroxylation, dealkylation, etc. As shown in Table 2, the range of chemical structure that undergoes glucuronidation as secondary step is quite diverse.
Table 1.
Compound | ACS Number | Chemical Structure | Pathway | Metabolite found in | Species | References |
---|---|---|---|---|---|---|
Quercetin | 117-39-5 | About 8.5% | Blood, Urine | Rat, mice, gerbil, human | (Manach et al., 1995; Hollman et al., 1997; Morand et al., 1998; Yeh et al., 2016) | |
Apigenin | 520-36-5 | Primary, Immature 10.0–31.6% mature 4.9% | Urine, feces, blood | Human, rat, | (Gradolatto et al., 2005; Chen et al., 2007) | |
Baicalein | 491-67-8 | Minor, less than 2.9% | Urine, feces, blood | Human, rat, mice, monkey | (Lai et al., 2003b; Tian et al., 2012) | |
Resveratrol | 501-36-0 | Primary, greater than 9–16% | Urine, blood, | Human, mice, rat | (Meng et al., 2004; Walle et al., 2004) | |
Daidzein | 486-66-8 | Primary, greater than 11% | Urine, feces, blood | Cat, rat, human, mice | (King and Bursill, 1998; Redmon et al., 2016) | |
Biochanin A | 491-80-5 | About 30.8% | Blood, urine, fece | Rat, human, mice | (Jia et al., 2004; Moon and Morris, 2007) | |
Formononetin | 485-72-3 | Minor, less than 0.18% | Bile | Rat, human, mice | (Jeong et al., 2005a) (Singh et al., 2011b) | |
Luteolin | 491-70-3 | Primary, greater than 48.78% | Blood, urine, feces | Rat, human, mice | (Shimoi et al., 1998; Lin et al., 2015) | |
Raloxifene | 84449-90-1 | Primary, greater than 11% | Blood, bile, feces, urine | Human, rat, dog | (Kosaka et al., 2011; Trdan et al., 2011) | |
Naringenin | 480-41-1 | Primary, greater than 86% | Blood, urine | Rat, human, mice | (Peng et al., 1998; Xu et al., 2009) | |
3-Hydroxyflavone | 577-85-5 | Primary, about 95% | Blood | Mice | (Xu et al., 2013) | |
Maackiain | 19908-48-6 | Primary, about 58% | Blood | Mice | (Gao et al., 2011) | |
Chrysin | 480-40-0 | Minor, less than 1% | Blood, urine, bile, feces | Humans, rats, mice | (Walle et al., 2001; Ge et al., 2015) | |
Emodin | 518-82-1 | About 40% | Blood, urine, feces, bile | Rats, | (Bachmann and Schlatter, 1981; Shia et al., 2010) | |
Troglitazone | 97322-87-7 | Minor, less than 5% | Blood, bile, urine, feces | Rats, humans, mice, dogs, cats | (Kawai et al., 1997; Loi et al., 1999; Michels et al., 2000) | |
Propranolol | 525-66-6 | About 17% | Bile, urine, blood, feces | Dogs, humans, rats, sheep | (Routledge and Shand, 1979; Marshall et al., 1981; Lo et al., 1984; Walle et al., 1985; Motheova et al., 1986) | |
SN-38 | 130194-92-2 | Primary, about 40% | Bile, blood, urine | Rats, humans | (Lokiec et al., 1995; Itoh et al., 2004; Kato et al., 2011) | |
Morphine | 57-27-2 | Primary, Greater than 50% | Blood, urine, cerebrospinal, bile | Humans, Rats, Dogs | (Glare and Walsh, 1991; Wolff et al., 1995; Andersen et al., 1997; Christrup, 1997; Andersen et al., 2003) | |
Mycophenolic acid | 24280-93-1 | Primary, about 87% | Blood, bile, urine | Humans, | (Joy et al., 2009) | |
Wogonin | 632-85-9 | Minor, about 5.9%, 5.7% were metabolites of its sulfates, | Blood, urine | Humans, rats, | (Lai et al., 2003 a) (Talbi et al., 2014) | |
Oroxylin A | - | Blood, urine | Rats, Humans | (Li et al., 1998; Fong et al., 2014) | ||
Fraxetin | 574-84-5 | - | Blood | Rats | (Zhao et al., 2016) | |
Estradiol | 50-27-1 | Primary, about 53% | Urine, Blood, bile | Humans, Sheep, dogs, baboon, rabbits | (Miyazaki et al., 1972; Musey et al., 1973; Collins et al., 1976; Nambara and Kawarada, 1977; Miyazaki et al., 1980) | |
Arbidol | 131707-25-0 | Minor, about 3.6% | Blood, feces, urine | Humans, Rats | (Liu et al., 2012b; Pan et al., 2013) | |
Ornidazole | 16773-42-5 | About 37.3% | Blood, urine | Humans, rats, dog | (Schwartz et al., 1979; Du et al., 2013; Du et al., 2014) | |
Magnolol | 528-43-8 | Primary, about 18% | Urine, blood, feces, bile | Rats, humans | (Hattori et al., 1986; Nakazawa et al., 2003) |
Table 2.
Compounds | ACS Number | Chemical Structures | References |
---|---|---|---|
MeOP | 67023-02-3 | (Meyer et al., 2015) | |
TPN729 | - | (Zhu et al., 2016) | |
Profluthrin | 223419-20-3 | (Beyerle et al., 2015) | |
DALT | 60676-77-9 | (Michely et al., 2015) | |
Alpelisib | 1217486-61-7 | (James et al., 2015) | |
Miltirone | 27210-57-7 | (Guo et al., 2015) | |
Rupestonic acid | 115473-63-7 | (Gu et al., 2015) | |
2C-P | 207740-22-5 | (Wink et al., 2015) | |
Praziquantel | 55268-74-1 | (Wang et al., 2014) | |
ABT-894 | 799279-80-4 | (Liu et al., 2014) | |
Tofacitinib | 477600-75-2 | (Dowty et al., 2014) | |
4-MA | 64-11-9 | (Welter et al., 2014) | |
Brucine | 357-57-3 | (Tian et al., 2014) | |
Praeruptorin A | 73069-27-9 | (Song et al., 2014) | |
Etamicastat | 760173-05-5 | (Loureiro et al., 2014) | |
GLS4 | - | (Zhou et al., 2013) | |
Apatinib | 811803-05-1 | (Ding et al., 2013) | |
Glaucine | 475-81-0 | (Meyer et al., 2013) | |
GDC-0152 | 873652-48-3 | (Yue et al., 2013) | |
Vismodegib | 879085-55-9 | (Yue et al., 2011) | |
Benfluron | 78250-23-4 | (Jirasko et al., 2011) | |
BIBF 1120 | 928326-83-4 | (Stopfer et al., 2011) | |
Anastrozole | 120511-73-1 | (Kamdem et al., 2010) | |
Vabicaserin | 620948-93-8 | (Tong et al., 2010) | |
Dicentrine | 517-66-8 | (Lai et al., 2010) | |
Flumatinib | 895519-91-2 | (Gong et al., 2010) | |
Anacetrapib | 875446-37-0 | (Tan et al., 2010) | |
2-Amino-3-methylimidazo[4,5-f]quinolone | 76180-96-6 | (Lakshmi et al., 2009) | |
Berberine | 2086-83-1 | (Liu et al., 2009) | |
Mazapertine | 134208-17-6 | (Wu et al., 2007) | |
Loratadine | 79794-75-5 | (Ramanathan et al., 2007) |
Majority of the glucuronides are pharmacologically inactive, however, in certain incidences glucuronides have been shown to be equally or more effective than the parent drug. For example, morphine-6-glucuronide is reported to be 45–61 folds more potent (Frances et al., 1990; Stone et al., 2003) and ezetimibe-glucuronide is reported to be 2–11 folds more potent (Ghosal et al., 2004; Kosoglou et al., 2005; Oswald et al., 2007) than their respective parent compounds. Additionally, some glucuronides can be toxic. For example, many acyl glucuronides have been shown to have high potential toxicities in vitro and in vivo (Shipkova et al., 2003).
Although not frequently reported, hydrophilic molecules are sometimes also glucuronidated. These molecules are often conjugated with a highly hydrophilic group (e.g., sugar or sulfate) but that did not appear to prevent them from getting glucuronidated. Several flavonoid glycosides are conjugated into glucuronides as reported in Table 3. Furthermore, it is difficult to separate the highly hydrophilic glycosides from their glucuronides in the reverse-phase chromatographic column, which is frequently used to separate drug from its glucuronide(s) during sample analysis. Therefore, mass spectrophotometry is usually employed to analyze glycoside and their glucuronides in the in vitro and in vivo experimental studies. This need for LC-MS/MS might have been the reason why hydrophilic UGT substrates are more difficult to identify, especially when the hydrophilic moiety is a sugar that is often deconjugated in the ion source of mass spectrometer.
Table 3.
Compound | ACS Number | Chemical Structure | Glucuronidation Pathway | Metabolite found in | Species studied | References |
---|---|---|---|---|---|---|
Genistein | 446-72-0 | Primary, greater than 50% | Urine, feces and blood | Mice, rats, humans | (Adlercreutz et al., 1993; Bloedon et al., 2002; Busby et al., 2002; Chen et al., 2003a; An and Morris, 2011; Yang et al., 2012) | |
EGCG | 989-51-5 | Minor, about 7.5% | Blood, urine, bile, | Rats, mice, humans, dogs | (Vaidyanathan and Walle, 2002) | |
Catechin | 88191-48-4 | Primary, greater than 50% | Blood, bile, urine, | Rats, mice, dogs, monkeys, humans | (Kuhnle et al., 2000) | |
Rutin | 153-18-4 | Rarely, about 2%. Most were metabolites of its aglycone | Blood, urine, | Rats, humans, filamentous fungi | (Jaganath et al., 2006) | |
Puerarin | 3681-99-0 | Minor, about 5%. Some were metabolites of its aglycone | Blood, urine, bile | Rats, humans | (Luo et al., 2012) | |
Epicatechin | 35323-91-2 | Primary, greater than 45% | Blood, | Rats, humans, dogs, | (Crespy et al., 2004) | |
Calycosin-7-O-Glucuronide | 20633-67-4 | Minor, about 5% | Blood, | Rats | (Shi et al., 2015) | |
Protocatechuic Aldehyde | 139-85-5 | greater than 30% | Urine | Rats, pigs | (Liu et al., 2008; Zhao et al., 2013) | |
Dopamine | 51-61-6 | greater than 25% | Brain | Rats, mice | (Uutela et al., 2009) | |
Bilirubin | 635-65-4 | greater than 50% | Liver | Human | (Ma et al., 2014) | |
Etoposide | 33419-42-0 | greater than 50% | Blood, urine, bile | Rats, rabbits, humans | (Hande et al., 1988a; Hande et al., 1988b; Wen et al., 2007) | |
5-Hydroxytryptophol | 154-02-9 | greater than 50% | Urine, | Humans, | (Stephanson et al., 2005) | |
4-Nitrophenol | 100-02-7 | About 40% | Urine, bile | Rats, | (Meerman et al., 1987) | |
Edaravone | 89-25-8 | greater than 70% | Urine | humans | (Ma et al., 2012) | |
Zidovudine | 30516-87-1 | greater than 50% | Blood, | Humans, | (Nadal et al., 1996) | |
Morphine | 57-27-2 | greater than 50% | Blood, muscle, fat, brain | Humans, | (Frost et al., 2015) | |
Nicotine | 22083-74-5 | Minor, about 5–10% | Blood, | Humans, | (Kaivosaari et al., 2007) | |
Imipramine | 50-49-7 | greater than 20% | Blood, urine | Humans, rabbits | (Qian and Zeng, 2006) | |
Benzotriazole | 95-14-7 | greater than 20% | Blood | Rats, | (Town et al., 1993) | |
Chloramphenicol | 56-75-7 | greater than 50% | Liver | bovine, equine, porcine | (Fedeniuk et al., 2015) |
When compared to CYP-catalyzed reactions, an important distinction of glucuronidation is that the metabolites produced are highly hydrophilic molecules that cannot penetrate the cell membrane via passive permeation. Rather, they need the action of various efflux transporters to pump them out of the cells (Jeong et al., 2005b). Hence, the driving force for glucuronidation is different from that of CYP catalyzed reaction. Specifically, it is driven by the twin forces of UGT enzyme present in the cellular endothelial reticulum and efflux transporters present on the cell surface. The efflux transporters can act as a driving force for glucuronidation by controlling the rate of efflux of glucuronides from the cells, which in turn can affect the formation rate. The faster efflux rate can cause the rate of glucuronide formation to increase due to the removal of potentially inhibitory glucuronides. Whereas, the slower efflux rate can cause lower glucuronidation output when compared to the actual glucuronidation capacity based on sub-cellular fractions, possibly using a product-inhibition feedback mechanism. In vast majority of the cases, these reactions occur in a polarized cell, making the glucuronide excretion polarized and dependent on the distribution of efflux transporter on two polar surfaces of a differentiated cell (Fig 1). For most orally administered drugs, these twin forces are in full effect in intestinal and liver cells, and together they determine the dispositional fate of a glucuronide.
Another distinction of the glucuronidation process is that the corresponding metabolites produced can be reconverted back to the original compound (or aglycone). This process could occasionally occur in mammalian tissues (usually at a very slow rate), but reconversion is extraordinary rapid in the colon when they are in contact with intestinal microflora, which produces a large quantity and variety of glucuronidases that can readily convert glucuronides into aglycones. The aglycones can then be re-absorbed to complete the process of recycling or recirculation. Hence, for glucuronides that are excreted back to the intestinal lumen, they often become bioavailable again following reconversion to the original compound.
There are considerable differences between UGTs and CYPs enzyme systems in terms of the similarity among their respective isoforms and the clinical significance of their genetic polymorphism. UGT enzymes are quite different from CYP in that each UGT subfamily tends to be clustered closely to each other, sometimes sharing the same gene. For example, the human UGT1A family shares the same gene (Guillemette, 2003; Kiang et al., 2005; Mackenzie et al., 2005; Bosch, 2008; Nies et al., 2008; Ginsberg et al., 2010), and utilizes alternative splicing to produce 9 active isoforms (UGT1A1, 1A3, 1A4, 1A5, 1A6, 1A7, 1A8, 1A9, 1A10). In contrast to CYPs, clinical significance of polymorphism in UGT isoforms remains mostly unclear; perhaps due to the fact the glucuronide production is dependent on both formation and efflux of glucuronide.
Therefore, the purpose of the present review is to outline how these twin forces of glucuronidation affect the production and biodistribution of glucuronides in various body compartments including intestine, liver, blood, and urine. Discussions related to recently- identified efficient hepatocytes-mediated uptake of glucuronides and various recycling schemes are included because they have impact on the disposition of glucuronides in vivo.
2. Nomenclature, Mechanisms of Glucuronidation and Organ-specific Distribution of UGTs
Human UGT Nomenclature.
Based on the amino acid sequence identity, four different families of UGTs are observed in human, namely UGT1, UGT2, UGT3, and UGT8 (Mackenzie et al., 2005). Among these UGT isoforms, UGT1 (UGT1A) and UGT2 (UGT2A and UGT2B) subfamilies are to be considered of paramount importance in terms of imparting drug conjugation ability. Currently, a total of 19 human UGT isoforms are known from both subfamily UGT1 and UGT2. Experimental studies reported that human UGT1A contains 13 distinct individual promoters in chromosome 2q37, which spans approximately 200 kb; whereas human UGT1B contains six individual promoters on the chromosome 4q13 (Mackenzie et al., 2005).
Mechanisms of Glucuronidation.
Glucuronidation is one of the most important phase II conjugative reactions, which eliminates predominantly drugs, dietary substances, toxins and endogenous substances. This particular reaction involves the transfer of the glucuronic group from uridine 5’-diphospho-glucuronic acid (UDPGA) to different substrate molecules containing oxygen, nitrogen, sulfur or carboxyl functional groups to generate relatively polar/hydrophilic glucuronide conjugate. In 2010, it was discovered that human UGTs mediate glucuronidation reaction by using a serine hydrolase-like mechanism, which involves two key amino acids histidine and aspartic acid (so-called “catalytic dyad” or “acid base pair”) (Radominska-Pandya et al., 2010). Investigations later showed that the glucuronidation reaction involves the formation of a ternary complex of enzyme, substrate and the co-factor UDPGA prior to the formation of ultimate conjugate (Luukkanen et al., 2005).
In addition, based on the inhibition studies using expressed recombinant human UGT isoforms, a compulsory ordered bi bi (i.e., two substrates and two products) kinetic mechanism was proposed (Luukkanen et al., 2005) where the co-factor UDPGA first binds with particular UGT enzyme and then forms a complex with substrate. On the other hand, alternative mechanisms such as random ordered bi bi mechanism were also reported, where binding of the substrate to the enzyme does not require prior binding to UDPGA (Yin et al., 1994). Based on the analysis of Luukkanen et al., these conflicting results were largely observed owing to the presence of multiple UGT enzymes and/or inactivated UGT enzyme in the latter study (Luukkanen et al., 2005).
Tissue Distribution of UGTs.
Generally UGT is present in humans, other animals (except cat), plants, and bacteria (Court and Greenblatt, 2000). In case of human, it is primarily distributed in different metabolic organs i.e., liver, kidney and intestine etc. (Uchaipichat et al., 2006). Studies showed that approximately 15%, 20% and 35% of marketed drugs are metabolized by three important human UGT isoforms, namely, UGT1A1, 1A4 and 2B7, respectively (Williams et al., 2004). Different human UGT isoforms such as UGT1A1, 1A3, 1A4, 1A6, 1A9, 2B4, 2B7, 2B10, 2B15, and 2B17 etc. have been reported to be present in liver (Tukey and Strassburg, 2000; Izukawa et al., 2009). After comparing the mRNA expressions of different UGT isoforms it has been found that, compared to UGT1A isoforms, UGT2Bs are more abundant in the human liver (Ohno and Nakajin, 2009). Among UGT2B subfamily, UGT2B4 and UGT2B15 are the highest expressed UGT isoforms present in liver. In case of UGT1A subfamily, UGT1A1 and UGT1A9 are the most abundant isoform present in liver. Apart from liver, UGT isoforms such as UGT1A8, UGT1A10 and UGT2B17 are predominantly expressed in human colon and intestine. The same study indicated that UGT1A7 is only present in the proximal tissues of the gastrointestinal tract (mainly the esophagus and stomach) (Ohno and Nakajin, 2009).
Since protein expression is considered to more accurately reflect the activity of enzymes, the absolute protein expression levels of some UGT isoforms in human liver were determined using LC-MS/MS in a recent study. According to their report, among all measured isoforms, UGT1A6 had the highest expression level in human liver microsomes. It was also found that there was a low correlation between protein and mRNA quantities for most of the UGT isoforms analyzed except UGT1A6 (high correlation with r2 of more than 0.6 (Ohtsuki et al., 2012). The absolute quantification of different UGT isoforms in rat tissue has not been reported yet.
Many of the studies have been performed to determine the mRNA levels of UGTs in different species such as rat and mouse (Shelby et al., 2003; Mackenzie et al., 2005; Owens et al., 2005; Buckley and Klaassen, 2007). In case of rats, both Ugt1a and Ugt2b subfamilies have been observed to be present. In contrast to human, Ugt1a subfamily containing 10 different isoforms (Ugt1a1, 1a2, 1a3, 1a4, 1a5, 1a6, 1a7, 1a8, 1a9, and 1a10), while Ugt2b subfamily consists of six members, namely, Ugt2b1, 2b2, 2b3, 2b6, 2b8, and 2b12 (Mackenzie et al., 2005; Owens et al., 2005). After comparing the mRNA expression of rat Ugt1 isoforms in different tissues, it was found out that rat Ugt1a isoforms are more prevalent in both liver and intestine compared to other tissues (Shelby et al., 2003). In liver, different isoforms of Ugt1a and Ugt2b subfamilies such as Ugt1a1, 1a3, 1a5, 1a8, 2b1, 2b2, 2b3, 2b6, and 2b12 were present, whereas Ugt1a1, 1a2, 1a3, 1a6 and 1a7 were present in rat intestine. The same study also concluded that only a few Ugt2b subfamily members (Ugt2b3, 2b8 and 2b12) were found in rat intestine (Shelby et al., 2003).
There are similarities and differences in expression of UGTs between rat and mouse. In case of mice, studies indicated that the Ugt1a subfamily contains 14 first exons (Buckley and Klaassen, 2007). Among them only nine enzymes were coded (Ugt1a1, 2, 5, 6a, 6b, 7c, 8, 9, and 10); whereas five were pseudogenes (Ugt1a3, 4, 7a, 7b, and 11). In case of Ugt2b subfamily, only seven Ugt2b genes (Ugt2b1, 2b5, 2b34, 2b35, 2b36, 2b37, and 2b38) were observed to be present in mice. Like rat, mouse liver was also shown to express different Ugt1a isoforms such as Ugt1a1, 1a5, 1a6, 1a9 as well as all Ugt 2b members. In addition, different Ugt subfamily such as Ugt1a6, 1a7c, 2a3, 2b34, and 2b35 are present in mouse gastrointestinal tract (Buckley and Klaassen, 2007).
3. Efflux Transporters Involved in Glucuronide Excretion
Phase II metabolites of drugs such as glucuronides, once formed, uses efflux transporters to exit the cell. Since the glucuronides are the substrate of efflux transporters, the two kinetic processes (glucuronidation by UGT and the excretion of glucuronides by efflux transporters) interplay with each other. These interplays are necessary, in large part, due to highly hydrophilic and charged properties of the metabolites, which requires the action of the efflux transporters (e.g., MRPs, BCRP) to exit cells.
MRPs.
The MRPs are the major efflux transporter family for phase II metabolites, and these transporters are expressed in many epithelial cells (Bera et al., 2002; Meyer zu Schwabedissen and Kroemer, 2011). MRPs share approximately 15% sequence similarities to that of P-gp (Mdr1), and these efflux transporters are predicted to form a large central hydrophobic core in their active binding region. The core region has two nucleotide-binding domains (NBD1 and NBD2); two membrane-spanning domains (MSD1 and MSD2); six trans-membrane spanning helices; and a linker segment called L1. Some of the MRP subfamily members also contain a membrane-spanning domain zero (MSD0), trans-membrane helices, and a linker zero (L0). The MSD0 and L0 are additional extensions that dangle at the N-terminus that extend extracellularly. Nine of the thirteen members of the multidrug resistant MRP/ABCC family are capable of effluxing both endogenous and exogenous organic anion compounds. Some MRPs, such as MRP1, are additionally capable of transporting neutral organic compounds in the presence of free glutathione (Kruh and Belisnsly, 2003). Collectively, the MRP family of efflux transporter is first known to convene resistance to anticancer agents (Kock and Brouwer, 2012). More recently, the Mrp family of efflux transporters has become known to efflux sulfate, glucuronide, and glutathione metabolites into the interstitial space, bile duct, intestinal lumen and basolateral surface of hepatocytes and enterocytes (Keppler and Konig, 2000; Jemnitz et al., 2010; Keppler, 2011; Kock and Brouwer, 2012).
Among the MRPs, Mrp1, Mrp3, Mrp5, and Mrp6 are densely expressed at the basolateral membrane of an epithelial cell, whereas Mrp2 is found in the apical side of the cell. Mrp4 expression and location depends on the tissue and species (Klaassen and Aleksunes, 2010) (van der Deen et al., 2005). For example, Mrp4 is found in the basolateral membrane of human prostatic glandular cell, but has also been localized to the apical membrane of rat kidney tubule cells. For Mrp7, Mrp8, and Mrp9, their specific locations of expression are less clear and appear to be random (Cai and Gros, 2003; Marquez et al., 2009).
Mrp1 was the first of MRPs to be discovered. It is ubiquitous in epithelial vesicular tissue such as lung and blood-tissue barriers. Mrp1 effluxes glutathione and glucuronide conjugates into the tissue underlying the membrane instead of effluxing these metabolites into the interstitial space. Mrp1 is highly expressed in the intestine and certain other organs such as liver (Cherrington et al., 2002), however, the expression level of Mrp1 varies between rats.
Mrp2 is probably the most widely studied MRP transporter, primarily because at least two MRP2-deficient rat models (Eisai hyperbilirubinuria rats and TR- rats) are available to study the role of MRP2 in drug disposition (Chen and Tiwari, 2011). MRP2 is believed to be responsible for transporting bile acid conjugates. Hepatic deficiency of Mrp2 in rats is linked to low bile acid conjugate excretion. There are various reports of a linkage between disease severity and expression level in both humans and rats. MRP2 is shown to be deficient in the hereditary condition known as Dubin-Johnson syndrome, which causes chronic conjugated hyperbilirubinemia and hepatocytes are not able to excrete conjugated organic anions into bile (Keppler and Konig, 1997). MRP2 in the intestine serves as the first barrier against all toxins entering into the systemic circulation (Jansen et al., 1985; Kamisako et al., 1999; Kamisako et al., 2000; Zamek-Gliszczynski et al., 2011). Mrp2 effluxes organic anions that are conjugated to glutathione, glucuronic acid, or sulfate into the intestinal lumen, thereby eliminating/reducing toxicity exposures to epithelial cells at the tip of the villus part of the jejunum (Cherrington et al., 2002; Wittgen et al., 2012) (Meyer zu Schwabedissen and Kroemer, 2011).
Mrp3 is very important for basolateral clearance of drugs and other substances in organs such as liver, gut, adrenals, pancreas, and kidney (Zelcer et al., 2001; Kock and Brouwer, 2012). It facilitates the excretion of organic anions, especially bile acids. Importantly, Mrp3 aids in the excretion of glucuronide metabolites into sinusoidal blood within the liver and into portal vein from enterocytes. Studies have shown that while expression level of Mrp3 was unrelated to the excretion of sulfate conjugates, it was proportional to the basolateral clearance of glucuronide conjugates in rats (Borst and Elferink, 2002).
Fewer studies have been conducted on other MRPs. Mrp4 appears to have high affinity for sulfate conjugates of bile acids and steroids (Kock and Brouwer, 2012). Glucuronide and glutathione metabolites also interact with Mrp4 in the liver; however, they have lower affinity in the presence of sulfate conjugates (Russel et al., 2008). Mrp4 is expressed in the prostate, lung, muscle, and pancreas (Klaassen and Aleksunes, 2010). Mrp5 expression is relatively low in a healthy liver. However, in hepatic cholestasis condition, Mrp5 mRNA is up regulated (Kock and Brouwer, 2012). Mrp5 selectively binds to glutathione conjugates and cyclic nucleotides, which are also substrate for Mrp4. Mrp5 is also the main anionic conjugate efflux transporter at the basolateral side and is ubiquitous in many organs (Klaassen and Aleksunes, 2010; Chen and Tiwari, 2011; Kock and Brouwer, 2012).
The physiologic function and the potential involvement in drug resistance of the other Mrps are still under investigation. Mrp6 localizes in the kidney and the basolateral surface of hepatocytes and is mainly involved in transporting glutathione conjugates and substrates such as BQ123, a cyclic-pentapeptide endothelin receptor antagonist (Kool et al., 1999; Madon et al., 2000; Belinsky et al., 2002). Specific transporting mechanisms of Mrp5 and Mrp6 are unclear. Mrp7 is a potassium channel regulator and therefore can be found in various organs, such as pancreas, testis, colon, spinal cord, tonsils, lung, trachea, and skin (Klaassen and Aleksunes, 2010). Mrp7 is capable of effluxing various amphipathic anions such as 17-beta-estradiol-(17-beta-d-glururonide) (Chen et al., 2003b). Mrp8 is ubiquitous throughout many organs and is responsible for nervous responses (Bortfeld et al., 2006). Mrp8 is known to efflux dehydropiandrosteron-3-sulfate, an endogenous precursor of many sex hormones (Chen et al., 2005b; Bortfeld et al., 2006). Mrp9 sequence is about 45–55% similar to that of Mrp5 sequence (Meyer zu Schwabedissen and Kroemer, 2011), but, its specific substrate and organ expression remain unclear so far (Bera et al., 2002).
BCRP.
The breast cancer resistance protein (Bcrp, ABCG2) was also identified as a hepatic canaliculus drug efflux transport protein, although its role in cancer multidrug resistance has been known for decades (Nakanishi and Ross, 2012). BCRP is densely expressed in the liver, kidney, blood-brain, intestine, and placental barriers (Doyle and Ross, 2003). BCRP’s role in biliary excretion of phase II conjugates is just beginning to be understood, partly because it covers a wide range of substrates. Common to the P-gp and MRP family, a single Transmembrane domain (TMD) binds to the terminus of the Nucleotide-binding domain (NBD) in BCRP. Since there are two TMD and two NBD repeats, the binding of a TMD to a single NBD is considered as a homodimer or half-transporters. The BCRP half-transporter NBD contains a single nucleotide that binds to the TMD. BCRP exist as either homodimer or homotetramers. As part of an ATP-dependent half-transporter, BCRP becomes functional upon dimerization (Biemans-Oldehinkel et al., 2006). It excretes anthracyclines, the active metabolite of CPT-11 (SN-38), mitoxanthrone, doxorubicin, as well as many sulfate and glucuronide conjugates into bile (Ni et al., 2010; Alvarez et al., 2011). BCRP appears to be responsible for the non-Mrp2 mediated component of biliary excretion of sulfate metabolites. The role of BCRP in the biliary excretion of sulfate conjugates has been demonstrated in vitro, and in rat and mice organ perfusion studies (Zamek-Gliszczynski et al., 2006c; Zamek-Gliszczynski et al., 2008; Zhu et al., 2010). Negligible excretion of glutathione conjugates in Mrp2-deficient rat livers indicate BCRP’s minor role in the excretion of these metabolites. BCRP transports many hydrophilic conjugated organic anions instead of hydrophobic compounds that are most likely substrates of P-gp. Working synergistically, Bcrp, Mrp-2, and P-gp eliminate a multitude of drugs absorbed across tissue barriers (Chang et al., 2011).
BSEP.
Bile salt export pump’s (BSEP, ABCB11) is another member of the Mdr family. BSEP is more than 80% homologous in human, rat, mouse, and dog. The variations are found mostly in the encoding gene of transmembrane loops (Yabuuchi et al., 2008). The exact structure of BSEP is not known (Wakabayashi et al., 2004) but it is believed that BSEP contains two trans-membrane domains composed of six helices connected between the cytoplasmic domain that is homologous to the structure of p-glycoprotein (van Den Elsen et al., 1999). However, BSEP is believed to also have four putative N-linked glycosylation sites for post-translational modifications (Borst and Elferink, 2002). BSEP is exclusively found in the canalicular membrane of the hepatocytes and has very specific substrates compared to p-glycoprotein (Kock and Brouwer, 2012). BSEP exclusively effluxes monoanionic conjugated bile acids, and whether it will excrete any other anions such as glucuronides remains to be determined. Currently, only a small number of drugs such as pravastatin and vinblastine are known to be effluxed by BSEP (Hirano et al., 2005; Yabuuchi et al., 2008). Studies have shown that deficiency in expression or function of this protein leads to intra-hepatic cholestasis and even liver injury (Balistreri et al., 2005; Hirano et al., 2006).
4. Possible and Likely Uptake Transporters Involved in Glucuronides Transport
Owing to their hydrophilic nature, the glucuronides also use uptake transporters, in addition to efflux transporters, to cross the biological membrane. Once formed in the intestinal epithelial cells, glucuronides are excreted into the portal vein by the efflux transporters in enterocytes, from where they enter hepatocytes with the aid of hepatic uptake transporters to be excreted into bile. The three major uptake transporter families involved in drug disposition, organic anion transporting polypeptides (OATPs), organic anion transporters (OATs), and the organic cation transporters (OCTs), likely belongs to the two solute carrier (SLC) superfamilies (Roth et al., 2012). SLC transporters are widely distributed in small intestine, kidney, liver and even brain, and are responsible for the uptake of many drugs and/or their glucuronides. These transporters are mostly expressed on the apical membrane of enterocytes, and the basolateral membranes of hepatocytes. However, in kidney, different SLC transporters are expressed on both apical and basolateral sides (Roth et al., 2012).
The OATP family is the predominant hepatic drug uptake transporter superfamily among SLCs in liver (Vasilyeva et al., 2015). 11 isoforms of OATPs are classified into 6 families (from OATP1 to 6) and several subfamilies based on similarities in their amino acid sequences (Vasilyeva et al., 2015). OATP1B1, OATP1B3 and OATP2B1 are three of the major isoforms expressed in the hepatic basolateral membrane (van de Steeg et al., 2012; Badee et al., 2015; Rowland et al., 2015), which are responsible for most of the hepatic uptake of drugs and their metabolites.
OATP transporters show varied expression levels on the apical or basolateral side of various human tissues. OATP1B1 and 1B3 are exclusively present on the sinusoidal membrane of liver (van de Steeg et al., 2012; Vasilyeva et al., 2015), whereas OATP2B1 is ubiquitously distributed throughout human body. It is not only expressed in liver (sinusoidal membrane) but also in other organs like brain, kidney and intestine (gut lumen) (van de Steeg et al., 2012; Rowland et al., 2015). Other isoforms such as OATP1A2, OATP2A1 are expressed at a relatively low concentration. OATP1A2 is exclusively expressed in cholangiocytes, the epithelial cells of the bile duct and may be involved in the reabsorption of xenobiotics excreted into the bile. Additionally, OATP1A2 is expressed on the apical side in the intestinal epithelial cells (Roth et al., 2012). Though they participate in the uptake process, their role in drug disposition is not yet understood (van de Steeg et al., 2012; Rowland et al., 2015; Vasilyeva et al., 2015).
OATPs share a similar transmembrane domain organization, which contains 12 predicted transmembrane domains and a large fifth extracellular loop. Based on a comparison among multiple species, all OATPs/Oatps (rat) transport using a rocker-switch type of mechanism, where substrate passes through a central, positively charged pore (Roth et al., 2012; Rowland et al., 2015). OATP-mediated uptake of drugs and glucuronides is an ATP- and sodium- independent transport process, however, the exact driving force for transporters is still not clear (Roth et al., 2012). One hypothesis offered a possible explanation to the mechanism. A pharmacophore model developed for OATP1B1 base on published apparent Km values of OATP substrates suggests that substrates contain two hydrogen bond acceptors, one hydrogen bond donor and two hydrophobic regions (van de Steeg et al., 2012; Rowland et al., 2015). However, there are different mechanism among different model substrates and even different OATPs (e.g. OATP 1B1 and 1B3) have slightly different mechanism.
The pharmacological importance of OATP transporters is owed to their broad substrate specificities (Roberts et al., 2002; Roth et al., 2012). As electrogenic transporters, OATP1B1 and 1B3 facilitate transport of many drugs and their metabolites. Many anticancer drugs like irinotecan and its active metabolite SN-38, methotrexate are their substrates (Roberts et al., 2002; Kalliokoski and Niemi, 2009). Though OATP1B1 and 1B3 share most of their substrates list, OATP1B3 seems to be the only hepatic OATP responsible for the uptake of digoxin, paclitaxel and docetaxel (Kalliokoski and Niemi, 2009). Moreover, statins are also transported by OATPs (Ming, 2008; Peng et al., 2015). As the top ranked anti-hyperlipidemia drug, statins have special importance in human health, which makes the study of OATPs more relevant and impactful. Many studies of drug–drug interactions focused on drug metabolism enzymes, such as CYPs, only. However, the wide range of the substrates as a “perpetrator” or a “victim” makes OATPs a potential target of drug-drug interactions. Pinpointing a single uptake transporter responsible for drug-drug interactions is still challenging due to overlapping substrate-specificity, which could be further impacted by the change in internal drug concentrations by metabolism enzymes.
Apart from the uptake of these exogenous substrates, OATPs are also responsible for the detoxification of endogenous compounds (Iusuf et al., 2012). For example, OATP1B1 and 1B3 play a major role in the metabolism of bilirubin and disposition of the resulting glucuronides. Together with MRP3, OATP1B1 and 1B3 establish a liver-blood shuttling loop for the transport of bilirubin and its glucuronides (van de Steeg et al., 2012; Vasilyeva et al., 2015). The functional deficiency of these two uptake transporters would lead to the blockage of the hepatocyte hopping, which causes the Rotor Syndrome (RS), a rare hereditary hyperbilirubinemia (Marin, 2012; van de Steeg et al., 2012; Rowland et al., 2015). Also, the homeostatic equilibrium of other endogenous substances such as bile acids, conjugated steroids and thyroid hormones are affected by OATPs because of their involvement in recycling of these substances (Kalliokoski and Niemi, 2009; Marin, 2012).
Organic anion transporters (OATs) are another transporter family related to the hepatic uptake. The substrates with one or two carboxylate groups are favored by the transporters (Roth et al., 2012). Though several isoforms have been detected in human liver, OAT2 is the most abundant one expressed in liver. (Jonker and Schinkel, 2004; Peng et al., 2015). Interestingly, sinusoidal membrane of hepatocyte is commonly regarded as the membrane for polarized distribution of OAT2, however, the subcellular localization of this protein in liver has only been demonstrated in rat but not in human (Jonker and Schinkel, 2004). Similar to OATPs, OAT2 is a sodium independent transporter. It works as a multi-specific organic anion exchanger by effluxing glutamate during the anion exchange. In human kidneys, OAT1, OAT2, and OAT3 are localized in the basolateral cell membrane, whereas OAT4, OAT10, and URAT1 in the apical cell membrane of proximal tubule cells, respectively. Studies have shown that OAT1 and OAT3 are involved in the elimination of various classes of drugs and conjugates of endogenous and exogenous substances from blood to urine in renal proximal tubules (Emami Riedmaier et al., 2012; Roth et al., 2012).
Compared to OAT1 and OAT3, a relatively small number of drugs are transported by OAT2. Some antiviral drugs, ACE inhibitors, angiotensin II receptor antagonists, HMG-CoA reductase inhibitors, NSAIDs, and antitumor drugs (paclitaxel, methotrexate etc.) are its substrates (Jonker and Schinkel, 2004; Estudante et al., 2013). In addition, a number of endogenous substances including sulfated steroid hormones (estrone-3-sulphate etc.), second messengers (Cyclic GMP and Cyclic AMP) and nucleosides (adenine, cytidine, guanosine etc.) are transported by OAT2 (Jonker and Schinkel, 2004).
5. Driving Forces for the Systemic Distribution of Metabolites
Polarized Excretion of Glucuronides.
In major metabolic organs such as intestine, liver and kidney, the primary cells responsible for metabolism are polarized, and as such efflux transporters located on the basolateral membrane are mostly distinct from those located on the apical membrane. Assuming stable UGT expression and functionality, transporters located at the basolateral membrane of enterocytes and hepatocytes, function as the driving forces for glucuronides to be distributed into blood. On the other hand, hepatic uptake transporters are able to extract extra-hepatic generated glucuronides, limiting their systemic exposure and facilitating their elimination via biliary excretion.
Role of Basolateral Efflux Transporters.
MRP3 is an important transporter located at the basolateral side of enterocytes and sinusoidal membrane of hepatocytes. Various glucuronide conjugates have been identified as substrates of MRP3, and MRP3 facilitates their entry into the mesenteric blood or the general circulation. In vesicular transport and ATPase activity assays, it was shown that estradiol 17-β-d-glucuronide, 7-hydroxycoumarin glucuronide, morphine glucuronide and bisphenol A glucuronide are substrates of MRP3 (Zelcer et al., 2001; Zamek-Gliszczynski et al., 2011; Mazur et al., 2012; Wittgen et al., 2012). Studies in everted intestinal sacs showed that the serosal efflux rate of 4-methylumbelliferone glucuronide decreased significantly in the small intestine of Mrp3 knockout mice (Kitamura et al., 2010). In Mrp3 knockout mice, the levels of resveratrol-3-glucuronide were up to 10-fold lower in plasma and urine (van de Wetering et al., 2009), and levels of morphine-3-glucuronide were up to 50-fold lower in plasma (Zelcer et al., 2005), owing to the lack of Mrp3 in the basolateral membrane of the enterocytes. In addition to MRP3, MRP4 is responsible for driving the intracellular glucuronides into the circulation from hepatocytes. In sandwich-cultured human hepatocytes and membrane vesicle uptake assays, the involvement of MRP3 and MRP4 in hepatic transport of mycophenolic acid glucuronide into the circulation was reported (Matsunaga et al., 2014).
Apical efflux transporters do not directly contribute towards the systemic exposure of glucuronides, however they can affect the outcome indirectly. The reduce expression of apical efflux transporter MRP2 is often associated with up-regulation of basolateral efflux transporter MRP3 to limit hepatic toxicity (Keppler and Konig, 2000; Dietrich et al., 2001; Roberts et al., 2002; Kubo et al., 2009). Owing to this, the systemic exposure of endogenous substrates and xenobiotic can be greatly altered due to the altered expression level or inhibition of apical efflux transporters.
Role of Basolateral Uptake Transporters.
When the extra-hepatic generated glucuronides reach portal vein, they have a chance to be taken up by the hepatocytes. OATP transporters expressed on the sinusoidal membrane of the hepatocytes play an important role in hepatic uptake of many endogenous and exogenous conjugates. For instance, studies in Oatp1a/1b knockout mice suggested that Oatp1a/1b transporters are involved in hepatic uptake of glucuronidated bilirubin (van de Steeg et al., 2010). There is also evidence showing that OATP1 plays important role in mediating the uptake of estradiol 17β-d-glucuronide by hepatocytes (Kouzuki et al., 1999). Moreover, OATP2B1 was demonstrated to be a major transporter involved in hepatic uptake of scutellarein glucuronides, which was suggested as a predominant process for their pharmacokinetic behaviors (Gao et al., 2012). Daidzein-7- glucuronide was reported as a substrate of OATP2B1 as well (Grosser et al., 2015). More detailed discussions can be found in Section 4.
In the liver, the presence of uptake transporters (e.g. OATPs) provides a potential pathway for extrahepatically generated glucuronides to be excreted into bile and therefore enhances the enterohepatic circulation of glucuronide conjugates. The circulation half-life of a glucuronide can be prolonged as the result of the increased enterohepatic recycling. On the other hand, the OATP uptake transporters also work in concert with sinusoidal export transporters (e.g.MRP3). Van de Steeg et al. proposed a theory they called “hepatocyte hopping” based on their observations with glucuronidated bilirubin in Oatp1a/1b mice. They found that the plasma level of bilirubin glucuronide in the Oatp1a/1b-deficient mice is remarkably increased. It was hypothesized that in the presence of both Oatp1a/1b and Mrp3 transporters, bilirubin glucuronide secreted in blood by Mrp3 can be taken up again by Oatp1a/1b located in the neighboring hepatocytes and hence have another chance to be excreted into bile. They claimed that this hepatocyte shuttling process can efficiently prevent the buildup of bilirubin glucuronide in the circulation (van de Steeg et al., 2010; Iusuf et al., 2012).
An important contributor to the systemic distribution of metabolites is recycling of glucuronides that are eliminated via bile or directly into the intestinal lumen. Because of the presence of microflora, glucuronides can be reconverted back into aglycone, which can then be reabsorbed, completing the recycling loop. More detailed discussion can be found in Section 6 below.
6. Recycling Mechanisms
Recycling prolongs the exposure of drugs to the systemic circulation due to repeated hydrolysis of glucuronides by (microbial) β-glucuronidase in gut, followed by reabsorption of the parent compound. Endogenous and exogenous substances such as xenobiotics and environmental pollutants can participate in one or more of the three types of recycling processes; enterohepatic, enteric and local recycling. Many compounds undergo only enterohepatic recycling (Gao et al., 2014) but some compounds such as polyphenols undergo duo recycling scheme involving both enteric and enterohepatic recycling (Roberts et al., 2002; Chen et al., 2003a; Liu et al., 2003; Jia et al., 2004; Silberberg et al., 2006; Liu and Hu, 2007). Similarly, local recycling could also occur independently or along with enteric and enterohepatic recycling, depending on the drug disposition characteristics. It is suggested that the relative contribution of enterohepatic, enteric and local recycling in the overall disposition of drug depends on the efficiency of enzyme-transporter coupling (see Section 8) by controlling the amounts of metabolites excreted by the intestine and liver (Jia et al., 2004; Jeong et al., 2005b; Liu and Hu, 2007).
Enterohepatic Recycling.
Enterohepatic recycling also known as enterohepatic circulation is a process where certain drug absorbed by enterocytes reenters intestine via bile excretion as glucuronides, which upon hydrolyzing back to aglycone, are reabsorbed. The drug once absorbed by intestinal cells enters the portal vein by the passive diffusion, whereas its glucuronides formed in the intestinal cell excrete on the basolateral side (gut lumen) by the efflux transporters. The drug from portal vein then enters hepatocytes by passive diffusion, where it can get metabolized again. On the other hand, glucuronides in portal vein may be taken up by the hepatocytes with the aid of hepatic sinusoidal uptake transporters. Hepatic efflux transporters on the apical side (MRP2 and BCRP) then can return the glucuronide to the intestine via the bile duct (Roberts et al., 2002; Gao et al., 2014) (Fig 2).
The bile contains glucuronides that are emptied into the gut after a meal, where glucuronides are hydrolyzed into parent drug by gut microflora followed by reabsorption in colon, thereby entering enterohepatic recycling (Roberts et al., 2002; Chen et al., 2003a; Liu et al., 2003; Jia et al., 2004; Silberberg et al., 2006; Liu and Hu, 2007). This process repeats itself until the drug is eliminated from the body. Enterohepatic recycling increases the half-life and the residence time for the species being recycled, and thus increase systemic exposure and delay drug clearance, as evident by the prolonged terminal elimination phase (Ouellet and Pollack, 1995; Schaiquevich et al., 2002). A drug undergoing enterohepatic recycling usually shows the multiple-peak phenomenon in its plasma-concentration–time profile and the prolonged elimination half-life (Gao et al., 2014). Ezetimibe (Ezzet et al., 2001; Yamamoto et al., 2007), sorafenib (Vasilyeva et al., 2015), diclofenac (Fukuyama et al., 1994), irinotecan (and SN-38) (Younis et al., 2009) and morphine (Ouellet and Pollack, 1995) are some of the drugs, which have shown to undergo biliary excretion and enterohepatic recycling as glucuronides in various animal models.
The enterohepatic recycling of glucuronides can have either beneficial or harmful effects on the body. In certain cases, such as flavonoids with lower bioavailability due to high metabolic clearance, the recycling increases their systemic exposure, half-life and the residence time in body, which is a favorable outcome (Hu, 2007; Thilakarathna and Rupasinghe, 2013; Dai et al., 2015). However, in other cases, this process is an unwanted outcome. For example, the enterohepatic recycling of diclofenac acyl glucuronides could increase the potential of gut toxicity by the repeated exposure to NSAID (Seitz and Boelsterli, 1998). Similarly, deconjugation of SN-38-glucuronide by gut microflora, results in high concentrations of SN-38 locally, thereby causing severe delayed diarrhea (Kaneda et al., 1990).
Enteric Recycling.
Certain percentages of parent drug that enter intestinal cells after oral administration are metabolized into glucuronides(s). These glucuronides can either enter the mesenteric blood system by the basolateral efflux transporters, or effluxed back into intestinal lumen by the apical efflux transporter(s). In the gut lumen, glucuronides are not absorbed and they will travel down the intestine until they reach terminal ileum or colon (bacteria-rich regions of the gut), where they are converted back into the parent drug by the gut microflora, and reabsorbed, thereby completing the enteric recycling scheme. A compound may be recycled repeatedly until it is completely eliminated from the system (Fig 2).
Multiple studies have been published to delineate the role of various components of enteric recycling scheme of glucuronides (UGT and efflux transporters) and understand how these components can be modulated to affect the local and systemic bioavailability of compounds/aglycones undergoing glucuronidation. A combination of in situ rat/mouse intestinal perfusion model along with in vitro intestinal/hepatic microsomes and Caco-2 cell transport studies demonstrated that enteric recycling plays an important role in disposition of various flavonoids, owing to their extensive glucuronidation in intestine and excretion of these glucuronides in gut lumen (Chen et al., 2003a; Hu et al., 2003; Liu et al., 2003; Jia et al., 2004; Chen et al., 2005a; Jeong et al., 2005b; Wang et al., 2006). MK-571 (Mrp2 inhibitor) and dipyridamole (BCRP inhibitor) when used together were able to significantly decrease the intestinal and biliary excretion of naringenin glucuronides in Wistar rats. These findings strongly suggested the involvement of MRP2 and BCRP efflux transporters in the enterohepatic and enteric recycling, by controlling the biliary and luminal efflux of glucuronides in liver and intestine, respectively (Xu et al., 2009).
Contrary to enterohepatic recycling, excretion of conjugates by enterocytes in enteric recycling do not usually cause double peak phenomenon, mainly because metabolites are gradually and continuously excreted into large intestine to be hydrolyzed and reabsorbed (Roberts et al., 2002; Chen et al., 2003a; Liu et al., 2003; Jia et al., 2004; Silberberg et al., 2006; Liu and Hu, 2007). In the enteric recycling of glucuronides, action of both, UGT enzyme (present at higher levels in the small intestine) and microbial β-glucuronidases (present at higher levels in the large intestine), are required and recycling is completed over the entire intestine. Moreover, enteric recycling does not require hepatic enzymes and efflux transporters observed for enterohepatic recycling, but rely only on the intestinal enzymes and efflux transporters.
Local Recycling.
More recently, a novel recycling system called local recycling of glucuronide has been reported, where drug enters the recycling mechanism without the intervention of bacterial β-glucuronidases. In local recycling, the deconjugation of glucuronides into parent drug is carried out by β-glucuronidases of enterocytes in upper small intestine, followed by reabsorption of drug in lower part of gut (Fig 2) (Xia et al., 2012; Dai et al., 2015). Wogonoside was rapidly hydrolyzed into wogonin by the β-glucuronidase present in the enterocytes rather than that of gut lumen (Xia et al., 2012). Dai et al. showed that tilianin could enter enteric, enterohepatic and local recycling scheme, called the triple-recycling mechanisms, after metabolizing into three metabolites; tilianin glucuronide, acacetin, and acacetin glucuronide (Dai et al., 2015).
Local recycling prolongs the residence time and increase local exposure of flavonoids in the gut and thus, it is assumed that flavonoids may have more biological activities in the gut than predicted based on their poor systemic bioavailability (Jeong et al., 2005a; Hu, 2007; Zhang et al., 2007; Xia et al., 2012). Though the local recycling has been reported in only two instances so far (Xia et al., 2012; Dai et al., 2015), based on the proposed mechanism of action, it is very much possible that local recycling significantly affects the biological activities of other drugs, which are undergoing extensive glucuronidation in gut. Local recycling of glucuronides, along with their enteric and enterohepatic recycling, can lead to prolonged and higher systemic exposure of poorly bioavailable phenolics, both locally and systemically. This prospect is equally important in the case of locally active drugs such as ezetimibe and ezetimibe glucuronide (more active than the parent compound), which exerts their cholesterol-lowering action by reducing the uptake and absorption of cholesterol by enterocytes. Possible triple recycling of ezetimibe glucuronide leads to prolonged local exposure in gut, resulting in more bioactivity at the site of action (Kosoglou et al., 2005). Similarly, SN-38 toxicity in large intestine is attributed to deconjugation of SN-38 glucuronide to toxic aglycone by bacteria β-glucuronidase in colon (Wallace et al., 2010). However, upper small intestine toxicity may be due to the local recycling, but this has not been investigated fully yet.
Like enteric recycling, local recycling requires only intestinal enzymes and transporters, and does not exhibit any double peak phenomenon, as the excretion of glucuronide is continuous. On the other hand, unlike the enteric recycling, local recycling could complete in the small intestine alone, without involving the whole intestine, as the action of microbial β-glucuronidases is not needed. Local recycling can occur either independently or in conjugation with other recycling processes. For drugs that are extensively metabolized in the gut, glucuronides can be excreted in gut lumen from enterocytes where they can re-enter the intestine after hydrolyzing back to aglycone by β-glucuronidases of enterocytes, thereby completing the cycle without the involvement of the other recycling mechanisms. However, rapidly absorbing drugs can saturate the gut UGT at high concentrations, so that they bypass intestinal metabolism and are predominantly glucuronidated in the liver. For these compounds, local recycling does not play significant role in first pass metabolism, but the glucuronides can later participate in local recycling followed by excretion in lumen through bile (Xia et al., 2012).
7. Driving Forces for the Elimination of Glucuronides
Routes of Elimination.
The elimination of glucuronides includes biliary, urinary, and intestinal excretion. For example, it has been shown that the biliary, urinary, and intestinal excretion of acetaminophen glucuronide accounts for 13, 9, and 1% of the orally administered acetaminophen in rats (Villanueva et al., 2008). Because of their involvement in the excretion of hydrophilic glucuronides, various membrane transporters (efflux and uptake) together determine the preferential elimination pathway for the disposition of glucuronides.
Intestinal Excretion.
In enterocytes, MRP2 and BCRP are the apical transporters, which have been shown to mediate the efflux of intracellular-formed glucuronide metabolites into the lumen (Fig 1a). In the lumen, the glucuronides can either be excreted in the feces or hydrolyzed back to the aglycone. For example, in Caco-2 cells, there was about one-fold decrease in the apical efflux of hesperetin glucuronide when Ko143 (5 μM) was used as BCRP inhibitor (Brand et al., 2008). Similarly, the use of LTC4 (an inhibitor of MRP2) decreased the efflux of emodin glucuronide from basolateral to apical side significantly (Liu et al., 2012a). The efflux of luteolin glucuronides from HeLa cells overexpressing UGT1A9 was inhibited by Ko143 in a dose-dependent manner (Tang et al., 2014). In rat perfusion model, Mrp2 and Bcrp1 were shown to efflux naringenin glucuronide to intestine and compensate for each other (Xu et al., 2009). As reported by our group, in Bcrp1 knockout mice, the excretion rate of genistein glucuronide in the small intestine decreased significantly (78%) (Zhu et al., 2010), whereas in Bcrp1-defecient mice, a substantial increase (>10 folds) in plasma AUC of genistein glucuronide after oral dose of genistein was observed (Yang et al., 2012). Similarly, in bioavailability and tissue distribution studies of resveratrol in Bcrp1 knockout mice, it was shown that Bcrp1 mediated the efflux of resveratrol glucuronides to the intestinal lumen and the AUC of resveratrol glucuronides increased in Bcrp1-deficient mice (Alfaras et al., 2010).
Hepatocyte Excretion.
In hepatocytes, MRP2 and BCRP are located on the canalicular membrane, where they function as efflux pumps to move intracellular glucuronides into bile (Fig 1b). The presence of these transporters provides a pathway for hepatic excretion and facilitates enterohepatic recycling. In Mrp2 knockout mice, a 56% decrease in biliary excretion of ezetimibe glucuronide was observed (de Waart et al., 2009), while the serum ezetimibe glucuronide levels in Mrp2-deficient rat increased by 10 folds compared to that in wild-type rats (de Waart et al., 2009; Oswald et al., 2010). Moreover, in the intestinal and liver perfusion studies in Mrp2-deficient rats, the excretion of ethinylestradiol glucuronide into intestine and bile was significantly deceased, and the systemic exposure of ethinylestradiol glucuronide was 46-fold higher in Mrp-2 knockout mice due to its decreased excretion into lumen and bile (Zamek-Gliszczynski et al., 2011). Similarly, Mrp2 has also been reported to mediate the biliary excretion of mycophenolic acid-7-O-glucuronide, 4-methylumbelliferyl glucuronide, flavopiridol glucuronide, grepafloxacin glucuronide, 17β-estradiol-17-β-D-glucuronide, and resveratrol glucuronide (Sasabe et al., 1998; Morikawa et al., 2000; Jager et al., 2003b; Jager et al., 2003a; Westley et al., 2006; Zamek-Gliszczynski et al., 2006a; Maier-Salamon et al., 2008). In in situ perfusion studies in Bcrp1(−/−) and Mrp2 (−/−) mouse livers, Bcrp was demonstrated to play a major role in biliary excretion of glucuronide metabolites of acetaminophen, 4-methylumbelliferone, and harmol, whereas Mrp2 played only a minor role (Zamek-Gliszczynski et al., 2006c). In efflux transporter-deficient mice, it was shown that both MRP2 and BCRP mediate the biliary excretion of droxydiclofenac acyl glucuronide (Lagas et al., 2010). Since, MRP2 and BCRP have overlapping substrate specificities, it is challenging to predict the impact of altered function of one or more transporter on the biliary excretion and systemic exposure of glucuronides (Yang et al., 2014).
Kidney Excretion.
In hepatocytes, glucuronides are either excreted by apical efflux transporters such as MRP2 and BCRP into bile, or they enter the systemic circulation by basolateral efflux transporters such as MRP3 (Fig 1c). From systemic circulation, glucuronides can be taken up into the kidney proximal tubular cells via organic anion transporters to be excreted via MRP2 or MRP4 at the apical side of these cells. Several transporters haven been shown to mediate the renal handling of glucuronides on the apical and basolateral sides of renal epithelial cells. Glomerular Filtration Rate (GRF) is also an important factor for the urinary excretion of glucuronides. For example, it had been shown that mycophenolic acid glucuronide (MPAG) and morphine glucuronides levels were higher in patients with poor renal function because of the decreased renal clearance of these glucuronide metabolites (Osborne et al., 1993; Naesens et al., 2007).
Organic anion transporters (OATs) expressed on the basolateral membrane of the proximal tubules in kidney are known to be involved in the transport of glucuronides. Studies in human embryonic kidney 293 (HEK 293) cells overexpressing hOAT3 showed that there was an enhanced uptake of MPAG (Uwai et al., 2007), morinidazole glucuronides (Zhong et al., 2014), daidzein-7-O-glucuronide, genistein-7-O-glucuronide, glycitein-7-O-glucuronide and quercetin-3-O-glucuronide (Wong et al., 2011), which suggested that OAT3 contributes to the renal tubular secretion of these glucuronide conjugates.
For renally excreted drugs, MRP2 and MRP4 plays a significantly role in the excretion of glucuronide in a substrate-dependent manner. On the apical side of renal cells, it was demonstrated that MRP4 was involved in the urinary excretion of glucuronide metabolite of edaravone, and the renal excretion of edaravone glucuronide was 2-fold lower in Mrp4-deficient mice (Mizuno et al., 2007). The role of MRP2 and MRP4 in renal excretion of MPAG was studied in HEK293 cells overexpressing human transporters and in isolated perfused kidneys. It was found that MPAG was a substrate of MRP2 but not MRP4, and the urinary excretion of MPAG was significantly greater in wild-type rat kidneys than in Mrp2 (−/−) rat kidneys (El-Sheikh et al., 2014). It was also reported that the renal excretion of acetaminophen glucuronide increased by 200% in bile duct-ligated rats, and this alteration was attributed to the up-regulation of renal Mrp2 (Villanueva et al., 2008). The glucuronide of 7-hydroxycoumarin (7-HC-G) was shown to be a substrate of MRP4 in studies using membrane vesicles overexpressing MRP transporters, and MRP4 was suggested to play a role in the excretion of 7-HC-G in kidney (Wittgen et al., 2012).
8. Interplay of UGT Enzymes, Efflux and Uptake Transporters
The disposition of drugs undergoing glucuronidation is usually controlled by multiple serial and/or parallel processes. First the drug is absorbed into enterocytes via passive diffusion, where it converts to glucuronides by intestinal UGTs. The gut-generated glucuronides are then excreted into gut lumen and portal vein by apical and basolateral intestinal efflux transporters, respectively. From portal vein, drug enter hepatocytes by passive diffusion and undergo glucuronidation by hepatic UGTs, whereas, glucuronides are taken up by the hepatic uptake transporters. From hepatocytes, both intestinal- and hepatic-generated glucuronides are then excreted into bile and systemic circulation by the apical and basolateral hepatic efflux transporters, respectively. These steps are further inter-linked with the three recycling mechanism mentioned in section 6.
In such a multi-component system, interplay between two or more components and processes is highly probable. The interplay could be explained by a combination of variety of coupling mechanisms for the disposition of glucuronides: UGT enzyme-efflux transporter coupling; UGT-uptake transporter coupling; efflux-uptake transporters coupling; and UGT enzyme-efflux transporter-uptake transporters coupling. These coupling mechanisms with the participation of multiple enzyme isoforms, efflux transporters and uptake transporters create a multi-component Enzyme–Transporter coupling network. Despite its complexity, this coupling network is highly capable of protecting human body against exogenous toxins; maintaining homeostasis of endogenous chemicals; and acting as bioavailability barrier to many xenobiotics. Importantly, failure in any of the individual components of the network is highly unlikely to cause the failure of the entire network.
UGT Enzyme–Efflux Transporter Coupling.
In the disposition of the drugs undergoing glucuronidation, the process of glucuronidation by UGTs couples with the process of excretion of glucuronides by efflux transporters. This phenomenon can be explained by the “Revolving door theory”, where the efflux transporters act as “revolving door” to facilitate and/or control the excretion of hydrophilic glucuronides out of the liver and intestinal cells (Liu and Hu, 2007; Singh and Hu, 2011). The UGT-efflux transporter coupling can leads to an imbalance between formation and excretion of glucuronides if one of the above processes act as the rate-limiting step. As a result, the actual rate of glucuronide excretion could either be lower (in efflux rate –limiting) or higher (in UGT rate-limiting) than the estimated rate of glucuronide excretion based on the cellular UGT activity (as measured from subcellular fraction) (Chen et al., 2003a; Jia et al., 2004; Jeong et al., 2005b; Wang et al., 2006).
Multiple UGT isoforms and efflux transporters (such as MRP2, MRP3 and BCRP) are shown to participates in the coupling process (Zamek-Gliszczynski et al., 2006b; Zhou et al., 2010; Jiang et al., 2012; Yang et al., 2012; Wei et al., 2013; Tang et al., 2014; Zhang et al., 2015; Wang et al., 2016). For example, estradiol-17-beta-d-glucuronide, which is formed by UGTs 1A1, 1A3, 1A4, 1A8, 1A9, and 1A10, was shown to interact with MRP2 and MRP3, expressed in isolated Sf9 membrane vesicles (Bodo et al., 2003). Also, enzyme-transporter interplay was observed in genistein glucuronide excretion in Hela cell overexpressed with UGT1A9 and BCRP. Ko143, a potent BCRP inhibitor was able to reduce the clearance of genistein glucuronide by about 75–94% in a dose-dependent manner (Jiang et al., 2012). Similarly, MK-571, a non-specific chemical inhibitor of MRP2, MRP3, and MRP4, significantly reduced the efflux of emodine glucuronide in the apical-to-basolateral (A-B) and B-A directions in Caco-2 cell lines in a dose-dependent manner (Liu et al., 2012a). Emodine is majorly glucuronidated by UGT1A1, 1A9, 1A10 and 2B7 (Wu et al., 2014). Due to overlapping substrate-specificity (Tian et al., 2008; Zhou et al., 2010; Singh et al., 2011a; Keppler, 2014; Yang et al., 2014) among UGT isoforms and efflux transporters, in an event of inhibition or deficiency of a UGT isoform or efflux transporters, other isoforms and transporters from same family and/or sub-family compensate for them (Wang et al., 2009; Xu et al., 2009). This mechanism makes it highly difficult to delineate individual UGT Enzyme-Efflux Transporter coupling pairs.
Specific inhibitors for various UGT isoforms and efflux transporter participating in the network are not readily available, so that the importance of individual enzyme isoform and transporter in the network has not been determined so far. Therefore, it has not yet been possible to overcome the bioavailability barrier by targeting a specific UGT enzyme(s) and/or an efflux transporter(s) of glucuronides. Furthermore, it is challenging to study this compensation mechanism, as when gene(s) of interest is silenced or induced for prolonged period of time, the expression of other enzymes and transporters of same family or subfamily are up- or down-regulated, respectively (Johnson et al., 2006; Hoffmann and Loscher, 2007; Kubo et al., 2009; Miyawaki et al., 2012).
UGT Enzyme-efflux transporter-uptake transporters coupling.
Once a glucuronide is excreted into gut lumen or the portal vein by efflux transporters in enterocytes, the uptake transporters in hepatocytes and enterocytes could take it up from portal vein and intestinal lumen, respectively. Due to overlapping substrate-specificity between various efflux and uptake transporters of glucuronides (Liu et al., 2006; Kalliokoski and Niemi, 2009; Kindla et al., 2009; Fahrmayr et al., 2010; Keppler, 2014), it could be hypothesized that the net excretion of glucuronides from enterocyte and hepatocyte could be dependent both on uptake of glucuronides into the cell by uptake transporters (such as OATPs and OATs) and efflux of glucuronides out of the cell by efflux transporters (such as MRPs and BCRP). The glucuronide taken up in the process can further be excreted into bile by hepatocytes or portal vein by enterocytes. The glucuronide excreted into the intestinal lumen and bile (emptying into gut) from apical efflux transporters of enterocytes and hepatocytes, respectively, can be reabsorbed after getting deconjugated in intestine thereby entering the recycling mechanisms (see Section 6).
UGT Enzyme-efflux transporter interplay has been investigated extensively in last decade, however, other coupling mechanisms with respect to xenobiotic glucuronidation are yet to be explored in depth. Though such couplings have been successfully shown for CYP substrates and endogenous glucuronides (Nies et al., 2004; van de Steeg et al., 2010; Iusuf et al., 2012; van de Steeg et al., 2012; Daali et al., 2013; Neve et al., 2013; Li et al., 2014; Shi and Li, 2014; Vasilyeva et al., 2015), very little has been reported for UGT substrates. OATP1B1 has been shown to play crucial role in the hepatic transport of glucuronides. Gemfibrozil glucuronide was able to inhibit the OATP1B1- and OATP1B3-mediated hepatic uptake of pravastatin (Nakagomi-Hagihara et al., 2007a; Nakagomi-Hagihara et al., 2007b). Uptake of ezetimibe glucuronide in cell expressing OATP1B1*1b was reduced as compared to the uptake in cell expressing wild-type protein (Oswald et al., 2008). Similarly, pharmacokinetics of ezetimibe glucuronide in human subjects with OATP1B1 polymorphism was affected. Fecal ezetimibe glucuronide excretion was significantly decreased whereas renal glucuronide excretion was increased in carriers of *1b/*1b. Polymorphism of OAT1B1 affect the uptake of ezetimibe glucuronide from portal vein into hepatocyte, such that reduced levels of ezetimibe glucuronides are available in hepatocytes for biliary excretion. However, this did not cause the expected increase in systemic concentration of ezetimibe glucuronides, probably due to increased renal clearance of glucuronides (Oswald et al., 2008). Very recently, sorafenib-glucuronide has been shown to display in UGT enzyme - efflux transporter (MRP2/MRP3) - uptake transporter (OAPT1B1/1B3) coupling, hepatocyte shuttling/hopping, as well as enterohepatic recycling (Vasilyeva et al., 2015). However, further mechanistic studies are required to understand these mechanisms and their implications in clinical drug-drug interactions.
9. New Directions
An important area, which is gaining significant attention and requires further exploration is the effect of gut microbiome on the disposition of drugs undergoing glucuronidation through enteric and enterohepatic recycling mechanism. Diet and antibiotics drugs can significant alter the microbial population in gut, thereby influencing the drug systemic exposure and disposition by affecting its re-entry into systemic circulation. Moreover, gut microbiome can also be used as therapeutic target to reduce drug related gut and liver toxicity owing to enterohepatic recycling. SN-38-glucuronide (phase-II metabolite of CPT-11) is converted into SN-38 (phase-I metabolite of CPT-11) by β-glucuronidase present in gut microflora, resulting in high luminal concentrations of SN-38, thereby causing severe delayed diarrhea (Kaneda et al., 1990). Potent bacterial β-glucuronidase inhibitors (1, 2, 3, and 4) (Fig 3) with submicromolar IC50 and Ki values have been identified recently that can block the conversion of glucuronides to aglycone, thereby blocking the enterohepatic recirculation of CPT-11 and NSAIDs. Crystal structures of E Coli β-glucuronidase complexes with inhibitors showed that inhibitors were bound at the “bacterial loops” at the entrance to the active-site cavity. Inhibitor 1 was shown to significantly reduce diarrhea and lower GI damage in 6- to 8-week-old Balb/cJ mouse models of CPT-11-induced toxicity (Wallace et al., 2010; LoGuidice et al., 2012).
10. Summary
The presence of multiple driving forces makes the disposition of drugs via glucuronidation process very complex in nature, when comparing to drug disposition via CYPs. Atypical behavior in glucuronidation is often observed when two drugs interact via a phase II disposition mechanism. For example, for drug interaction via phase I enzymatic inhibition, substrate levels in plasma increases but metabolite levels decreases (Dresser et al., 2000; Stearns et al., 2003; Laugesen et al., 2005). For a drug that is a substrate of both CYP and p-glycoprotein, inhibition of the efflux transporter led to higher concentration of both the metabolite and the corresponding aglycone (Pang et al., 2009; Li et al., 2014). In contrast, for drugs undergoing phase II metabolism, a drug interaction via efflux transporter inhibition could lead to higher plasma levels of metabolite with or without a corresponding increase in the aglycone levels, even though the enzyme activities were not altered (Yang et al., 2012; Wei et al., 2013; Ge et al., 2016). This complex process is also the reason why in vitro glucuronidation obtained from organ microsomes often cannot predict in vivo glucuronide production or levels of glucuronides in plasma (Wang et al., 2006; Wu et al., 2013).
The complex process involves the interplay of various enzyme and transporter systems including recycling mechanisms to control the system exposure and clearance of these drugs. The interplay can happen between enzyme system (UGT1A and UGT2B) and efflux transporters (both apical and basolateral) in enterocytes and hepatocytes as well as between the efflux and uptake transporters of hepatocytes (Liu and Hu, 2007; Jiang and Hu, 2012; Kock and Brouwer, 2012; Wu, 2012; Pfeifer et al., 2014; Zamek-Gliszczynski et al., 2014). Such a complex interplay of various components is possibly essential for the body to maintain a tight control over the disposition of various endogenous compounds such as bile acid, bilirubin and steroids in order to maintain their homeostasis, as well as the detoxification of environmental and dietary toxins.
The absence or inhibition of a particular component in this complex equation can cause compensation by one or more components to avoid cell toxicity, thereby resulting in increased or decreased concentration of parent compound or the glucuronide in the systemic circulation. For e.g., MRP3 up-regulation in event of MRP2-deficiency can cause increased bilirubin glucuronide excretion in blood (Kamisako et al., 2000). Similarly, comparable or even higher level of glucuronidation by Ugt1a-deficient Gunn rats as compared to the control Wistar rats, were ascribed to the compensatory up-regulation of intestinal Ugt2bs and hepatic anion efflux transporters (Wang et al., 2009).
However, the downside of this complex system is a very difficult-to-overcome oral bioavailability barrier for xenobiotics using glucuronidation as major elimination pathway (Hu, 2007; Gao and Hu, 2010). Many recent studies have been published showing how these individual components contribute to the overall disposition mechanism, as well as how the modulation of one or more these components can alter the systemic exposure of glucuronidated compounds (Wei et al., 2013; Tang et al., 2014; Dai et al., 2015; Zhang et al., 2015; Wang et al., 2016; Zeng et al., 2016). The published research with UGT- or transporter- deficient or over-expressed animal or cell models indicates that it is very difficult to improve bioavailability of the drug by interfering with one or the other components. However, the systemic exposure can be modulated by altering the excretion of glucuronide in blood by inhibiting the one or more efflux and/or uptake transporters. Further detailed studies to understand the interplay of various components will be needed in order to improve the systemic and/or local exposure of beneficial UGT substrates.
In conclusion, systemic glucuronide levels are often not determined by the UGT enzyme activities alone but also by the action of efflux transporters that mediate the distribution of glucuronides into the systemic circulation. This mechanism means that any drug interaction involving an efflux transporter of glucuronides can have a direct impact on the systemic levels of the glucuronides, which in turn could change the levels of their corresponding aglycone due to the presence of glucuronidases. On the other hand, the systemic clearance of glucuronides is also affected by the recycling of substrates, which undergo glucuronidation via local, enteric and enterohepatic recycling (i.e., so called triple-recycling mechanisms). Hence, predicting systemic glucuronide levels requires the consideration of the structure and function of intestinal microbiome. Taken together, recent advances in understanding the glucuronidation process will help us improve the systemic and local bioavailability of drugs that undergo phase II glucuronidation.
References
- Adlercreutz H, Markkanen H, and Watanabe S (1993) Plasma concentrations of phyto-oestrogens in Japanese men. Lancet 342:1209–1210. [DOI] [PubMed] [Google Scholar]
- Alfaras I, Perez M, Juan ME, Merino G, Prieto JG, Planas JM, and Alvarez AI (2010) Involvement of breast cancer resistance protein (BCRP1/ABCG2) in the bioavailability and tissue distribution of trans-resveratrol in knockout mice. J Agric Food Chem 58:4523–4528. [DOI] [PubMed] [Google Scholar]
- Alvarez AI, Vallejo F, Barrera B, Merino G, Prieto JG, Tomas-Barberan F, and Espin JC (2011) Bioavailability of the glucuronide and sulfate conjugates of genistein and daidzein in breast cancer resistance protein 1 knockout mice. Drug Metab Dispos 39:2008–2012. [DOI] [PubMed] [Google Scholar]
- An G and Morris ME (2011) The sulfated conjugate of biochanin A is a substrate of breast cancer resistant protein (ABCG2). Biopharm Drug Dispos 32:446–457. [DOI] [PubMed] [Google Scholar]
- Andersen G, Christrup L, and Sjogren P (2003) Relationships among morphine metabolism, pain and side effects during long-term treatment: an update. J Pain Symptom Manage 25:74–91. [DOI] [PubMed] [Google Scholar]
- Andersen G, Christrup LL, and Sjogren P (1997) [Morphine metabolism--pharmacokinetics and pharmacodynamics]. Ugeskr Laeger 159:3383–3386. [PubMed] [Google Scholar]
- Bachmann M and Schlatter C (1981) Metabolism of [14C]emodin in the rat. Xenobiotica 11:217–225. [DOI] [PubMed] [Google Scholar]
- Badee J, Achour B, Rostami-Hodjegan A, and Galetin A (2015) Meta-analysis of expression of hepatic organic anion-transporting polypeptide (OATP) transporters in cellular systems relative to human liver tissue. Drug Metab Dispos 43:424–432. [DOI] [PubMed] [Google Scholar]
- Balistreri WF, Bezerra JA, Jansen P, Karpen SJ, Shneider BL, and Suchy FJ (2005) Intrahepatic cholestasis: summary of an American Association for the Study of Liver Diseases single-topic conference. Hepatology 42:222–235. [DOI] [PubMed] [Google Scholar]
- Belinsky MG, Chen ZS, Shchaveleva I, Zeng H, and Kruh GD (2002) Characterization of the drug resistance and transport properties of multidrug resistance protein 6 (MRP6, ABCC6). Cancer Res 62:6172–6177. [PubMed] [Google Scholar]
- Bera TK, Iavarone C, Kumar V, Lee S, Lee B, and Pastan I (2002) MRP9, an unusual truncated member of the ABC transporter superfamily, is highly expressed in breast cancer. Proc Natl Acad Sci U S A 99:6997–7002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beyerle J, Frei E, Stiborova M, Habermann N, and Ulrich CM (2015) Biotransformation of xenobiotics in the human colon and rectum and its association with colorectal cancer. Drug Metab Rev 47:199–221. [DOI] [PubMed] [Google Scholar]
- Biemans-Oldehinkel E, Doeven MK, and Poolman B (2006) ABC transporter architecture and regulatory roles of accessory domains. FEBS Lett 580:1023–1035. [DOI] [PubMed] [Google Scholar]
- Bloedon LT, Jeffcoat AR, Lopaczynski W, Schell MJ, Black TM, Dix KJ, Thomas BF, Albright C, Busby MG, Crowell JA, and Zeisel SH (2002) Safety and pharmacokinetics of purified soy isoflavones: single-dose administration to postmenopausal women. Am J Clin Nutr 76:1126–1137. [DOI] [PubMed] [Google Scholar]
- Bodo A, Bakos E, Szeri F, Varadi A, and Sarkadi B (2003) Differential modulation of the human liver conjugate transporters MRP2 and MRP3 by bile acids and organic anions. J Biol Chem 278:23529–23537. [DOI] [PubMed] [Google Scholar]
- Borst P and Elferink RO (2002) Mammalian ABC transporters in health and disease. Annu Rev Biochem 71:537–592. [DOI] [PubMed] [Google Scholar]
- Bortfeld M, Rius M, Konig J, Herold-Mende C, Nies AT, and Keppler D (2006) Human multidrug resistance protein 8 (MRP8/ABCC11), an apical efflux pump for steroid sulfates, is an axonal protein of the CNS and peripheral nervous system. Neuroscience 137:1247–1257. [DOI] [PubMed] [Google Scholar]
- Bosch TM (2008) Pharmacogenomics of drug-metabolizing enzymes and drug transporters in chemotherapy. Methods Mol Biol 448:63–76. [DOI] [PubMed] [Google Scholar]
- Brand W, van der Wel PA, Rein MJ, Barron D, Williamson G, van Bladeren PJ, and Rietjens IM (2008) Metabolism and transport of the citrus flavonoid hesperetin in Caco-2 cell monolayers. Drug Metab Dispos 36:1794–1802. [DOI] [PubMed] [Google Scholar]
- Buckley DB and Klaassen CD (2007) Tissue- and gender-specific mRNA expression of UDP-glucuronosyltransferases (UGTs) in mice. Drug Metab Dispos 35:121–127. [DOI] [PubMed] [Google Scholar]
- Busby MG, Jeffcoat AR, Bloedon LT, Koch MA, Black T, Dix KJ, Heizer WD, Thomas BF, Hill JM, Crowell JA, and Zeisel SH (2002) Clinical characteristics and pharmacokinetics of purified soy isoflavones: single-dose administration to healthy men. Am J Clin Nutr 75:126–136. [DOI] [PubMed] [Google Scholar]
- Cai J and Gros P (2003) Overexpression, purification, and functional characterization of ATP-binding cassette transporters in the yeast, Pichia pastoris. Biochim Biophys Acta 1610:63–76. [DOI] [PubMed] [Google Scholar]
- Chang JH, Uchizono JA, and Park MS (2011) Efflux of Drugs via Transporters—The Antiabsorption Pathway, in: Oral Bioavailability: Basic Principles, Advanced Concepts, and Applications (Hu M and Li X eds), pp 111–126, Wiley, New Jersey. [Google Scholar]
- Chen J, Lin H, and Hu M (2003a) Metabolism of flavonoids via enteric recycling: role of intestinal disposition. J Pharmacol Exp Ther 304:1228–1235. [DOI] [PubMed] [Google Scholar]
- Chen J, Wang S, Jia X, Bajimaya S, Tam V, and Hu M (2005a) Disposition of Flavonoids via Recycling: Comparison of Intestinal versus Hepatic Disposition. Drug Metab Dispos 33:1777–1784. [DOI] [PubMed] [Google Scholar]
- Chen T, Li LP, Lu XY, Jiang HD, and Zeng S (2007) Absorption and excretion of luteolin and apigenin in rats after oral administration of Chrysanthemum morifolium extract. J Agric Food Chem 55:273–277. [DOI] [PubMed] [Google Scholar]
- Chen ZS, Guo Y, Belinsky MG, Kotova E, and Kruh GD (2005b) Transport of bile acids, sulfated steroids, estradiol 17-beta-D-glucuronide, and leukotriene C4 by human multidrug resistance protein 8 (ABCC11). Mol Pharmacol 67:545–557. [DOI] [PubMed] [Google Scholar]
- Chen ZS, Hopper-Borge E, Belinsky MG, Shchaveleva I, Kotova E, and Kruh GD (2003b) Characterization of the transport properties of human multidrug resistance protein 7 (MRP7, ABCC10). Mol Pharmacol 63:351–358. [DOI] [PubMed] [Google Scholar]
- Chen ZS and Tiwari AK (2011) Multidrug resistance proteins (MRPs/ABCCs) in cancer chemotherapy and genetic diseases. FEBS J 278:3226–3245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cherrington NJ, Hartley DP, Li N, Johnson DR, and Klaassen CD (2002) Organ distribution of multidrug resistance proteins 1, 2, and 3 (Mrp1, 2, and 3) mRNA and hepatic induction of Mrp3 by constitutive androstane receptor activators in rats. J Pharmacol Exp Ther 300:97–104. [DOI] [PubMed] [Google Scholar]
- Christrup LL (1997) Morphine metabolites. Acta Anaesthesiol Scand 41:116–122. [DOI] [PubMed] [Google Scholar]
- Collins DC, Balikian HM, and Preedy JR (1976) Splanchnic and intestinal uptake and formation of estriol and estriol conjugates in the dog in vivo. Steroids 28:597–612. [DOI] [PubMed] [Google Scholar]
- Court MH and Greenblatt DJ (2000) Molecular genetic basis for deficient acetaminophen glucuronidation by cats: UGT1A6 is a pseudogene, and evidence for reduced diversity of expressed hepatic UGT1A isoforms. Pharmacogenetics 10:355–369. [DOI] [PubMed] [Google Scholar]
- Crespy V, Nancoz N, Oliveira M, Hau J, Courtet-Compondu MC, and Williamson G (2004) Glucuronidation of the green tea catechins, (−)-epigallocatechin-3-gallate and (−)-epicatechin-3-gallate, by rat hepatic and intestinal microsomes. Free Radic Res 38:1025–1031. [DOI] [PubMed] [Google Scholar]
- Daali Y, Millet P, Dayer P, and Pastor CM (2013) Evidence of drug-drug interactions through uptake and efflux transport systems in rat hepatocytes: implications for cellular concentrations of competing drugs. Drug Metab Dispos 41:1548–1556. [DOI] [PubMed] [Google Scholar]
- Dai P, Zhu L, Luo F, Lu L, Li Q, Wang L, Wang Y, Wang X, Hu M, and Liu Z (2015) Triple Recycling Processes Impact Systemic and Local Bioavailability of Orally Administered Flavonoids. AAPS J 17:723–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Waart DR, Vlaming ML, Kunne C, Schinkel AH, and Oude Elferink RP (2009) Complex pharmacokinetic behavior of ezetimibe depends on abcc2, abcc3, and abcg2. Drug Metab Dispos 37:1698–1702. [DOI] [PubMed] [Google Scholar]
- Dietrich CG, de Waart DR, Ottenhoff R, Bootsma AH, van Gennip AH, and Elferink RP (2001) Mrp2-deficiency in the rat impairs biliary and intestinal excretion and influences metabolism and disposition of the food-derived carcinogen 2-amino-1-methyl-6-phenylimidazo. Carcinogenesis 22:805–811. [DOI] [PubMed] [Google Scholar]
- Ding J, Chen X, Gao Z, Dai X, Li L, Xie C, Jiang H, Zhang L, and Zhong D (2013) Metabolism and pharmacokinetics of novel selective vascular endothelial growth factor receptor-2 inhibitor apatinib in humans. Drug Metab Dispos 41:1195–1210. [DOI] [PubMed] [Google Scholar]
- Dowty ME, Lin J, Ryder TF, Wang W, Walker GS, Vaz A, Chan GL, Krishnaswami S, and Prakash C (2014) The pharmacokinetics, metabolism, and clearance mechanisms of tofacitinib, a janus kinase inhibitor, in humans. Drug Metab Dispos 42:759–773. [DOI] [PubMed] [Google Scholar]
- Doyle L and Ross DD (2003) Multidrug resistance mediated by the breast cancer resistance protein BCRP (ABCG2). Oncogene 22:7340–7358. [DOI] [PubMed] [Google Scholar]
- Dresser GK, Spence JD, and Bailey DG (2000) Pharmacokinetic-pharmacodynamic consequences and clinical relevance of cytochrome P450 3A4 inhibition. Clin Pharmacokinet 38:41–57. [DOI] [PubMed] [Google Scholar]
- Du J, Ma Z, Zhang Y, Wang T, Chen X, and Zhong D (2014) Simultaneous determination of ornidazole and its main metabolites in human plasma by LC-MS/MS: application to a pharmacokinetic study. Bioanalysis 6:2343–2356. [DOI] [PubMed] [Google Scholar]
- Du J, You T, Chen X, and Zhong D (2013) Stereoselective glucuronidation of ornidazole in humans: predominant contribution of UDP-glucuronosyltransferases 1A9 and 2B7. Drug Metab Dispos 41:1306–1318. [DOI] [PubMed] [Google Scholar]
- El-Sheikh AA, Koenderink JB, Wouterse AC, van den Broek PH, Verweij VG, Masereeuw R, and Russel FG (2014) Renal glucuronidation and multidrug resistance protein 2-/ multidrug resistance protein 4-mediated efflux of mycophenolic acid: interaction with cyclosporine and tacrolimus. Transl Res 164:46–56. [DOI] [PubMed] [Google Scholar]
- Emami Riedmaier A, Nies AT, Schaeffeler E, and Schwab M (2012) Organic anion transporters and their implications in pharmacotherapy. Pharmacol Rev 64:421–449. [DOI] [PubMed] [Google Scholar]
- Estudante M, Morais JG, Soveral G, and Benet LZ (2013) Intestinal drug transporters: an overview. Adv Drug Deliv Rev 65:1340–1356. [DOI] [PubMed] [Google Scholar]
- Ezzet F, Krishna G, Wexler DB, Statkevich P, Kosoglou T, and Batra VK (2001) A population pharmacokinetic model that describes multiple peaks due to enterohepatic recirculation of ezetimibe. Clinical therapeutics 23:871–885. [DOI] [PubMed] [Google Scholar]
- Fahrmayr C, Fromm MF, and Konig J (2010) Hepatic OATP and OCT uptake transporters: their role for drug-drug interactions and pharmacogenetic aspects. Drug Metab Rev 42:380–401. [DOI] [PubMed] [Google Scholar]
- Fedeniuk RW, Mizuno M, Neiser C, and O’Byrne C (2015) Development of LC-MS/MS methodology for the detection/determination and confirmation of chloramphenicol, chloramphenicol 3-O-beta-d-glucuronide, florfenicol, florfenicol amine and thiamphenicol residues in bovine, equine and porcine liver. J Chromatogr B Analyt Technol Biomed Life Sci 991:68–78. [DOI] [PubMed] [Google Scholar]
- Fong SY, Wong YC, and Zuo Z (2014) Development of a SPE-LC/MS/MS method for simultaneous quantification of baicalein, wogonin, oroxylin A and their glucuronides baicalin, wogonoside and oroxyloside in rats and its application to brain uptake and plasma pharmacokinetic studies. J Pharm Biomed Anal 97:9–23. [DOI] [PubMed] [Google Scholar]
- Frances B, Gout R, Campistron G, Panconi E, and Cros J (1990) Morphine-6-glucuronide is more mu-selective and potent in analgesic tests than morphine. Prog Clin Biol Res 328:477–480. [PubMed] [Google Scholar]
- Frost J, Lokken TN, Brede WR, Hegstad S, Nordrum IS, and Slordal L (2015) A validated method for simultaneous determination of codeine, codeine-6-glucuronide, norcodeine, morphine, morphine-3-glucuronide and morphine-6-glucuronide in post-mortem blood, vitreous fluid, muscle, fat and brain tissue by LC-MS. J Anal Toxicol 39:203–212. [DOI] [PubMed] [Google Scholar]
- Fukuyama T, Yamaoka K, Ohata Y, and Nakagawa T (1994) A new analysis method for disposition kinetics of enterohepatic circulation of diclofenac in rats. Drug Metab Dispos 22:479–485. [PubMed] [Google Scholar]
- Gao C, Zhang H, Guo Z, You T, Chen X, and Zhong D (2012) Mechanistic studies on the absorption and disposition of scutellarin in humans: selective OATP2B1-mediated hepatic uptake is a likely key determinant for its unique pharmacokinetic characteristics. Drug Metab Dispos 40:2009–2020. [DOI] [PubMed] [Google Scholar]
- Gao S and Hu M (2010) Bioavailability challenges associated with development of anti-cancer phenolics. Mini Rev Med Chem 10:550–567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao S, Yang Z, Yin T, You M, and Hu M (2011) Validated LC-MS/MS method for the determination of maackiain and its sulfate and glucuronide in blood: application to pharmacokinetic and disposition studies. J Pharm Biomed Anal 55:288–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao Y, Shao J, Jiang Z, Chen J, Gu S, Yu S, Zheng K, and Jia L (2014) Drug enterohepatic circulation and disposition: constituents of systems pharmacokinetics. Drug Discov Today 19:326–340. [DOI] [PubMed] [Google Scholar]
- Ge S, Gao S, Yin T, and Hu M (2015) Determination of Pharmacokinetics of Chrysin and Its Conjugates in Wild-Type FVB and Bcrp1 Knockout Mice Using a Validated LC-MS/MS Method. J Agric Food Chem 63:2902–2910. [DOI] [PubMed] [Google Scholar]
- Ge S, Yin T, Xu B, Gao S, and Hu M (2016) Curcumin Affects Phase II Disposition of Resveratrol Through Inhibiting Efflux Transporters MRP2 and BCRP. Pharm Res 33:590–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ghosal A, Hapangama N, Yuan Y, Achanfuo-Yeboah J, Iannucci R, Chowdhury S, Alton K, Patrick JE, and Zbaida S (2004) Identification of human UDP-glucuronosyltransferase enzyme(s) responsible for the glucuronidation of ezetimibe (Zetia). Drug Metab Dispos 32:314–320. [DOI] [PubMed] [Google Scholar]
- Ginsberg G, Guyton K, Johns D, Schimek J, Angle K, and Sonawane B (2010) Genetic polymorphism in metabolism and host defense enzymes: implications for human health risk assessment. Crit Rev Toxicol 40:575–619. [DOI] [PubMed] [Google Scholar]
- Glare PA and Walsh TD (1991) Clinical pharmacokinetics of morphine. Ther Drug Monit 13:1–23. [DOI] [PubMed] [Google Scholar]
- Gong A, Chen X, Deng P, and Zhong D (2010) Metabolism of flumatinib, a novel antineoplastic tyrosine kinase inhibitor, in chronic myelogenous leukemia patients. Drug Metab Dispos 38:1328–1340. [DOI] [PubMed] [Google Scholar]
- Gradolatto A, Basly JP, Berges R, Teyssier C, Chagnon MC, Siess MH, and Canivenc-Lavier MC (2005) Pharmacokinetics and metabolism of apigenin in female and male rats after a single oral administration. Drug Metab Dispos 33:49–54. [DOI] [PubMed] [Google Scholar]
- Grosser G, Doring B, Ugele B, Geyer J, Kulling SE, and Soukup ST (2015) Transport of the soy isoflavone daidzein and its conjugative metabolites by the carriers SOAT, NTCP, OAT4, and OATP2B1. Arch Toxicol 89:2253–2263. [DOI] [PubMed] [Google Scholar]
- Gu D, Yang Y, Chen Q, Habasi M, Zhao J, and Aisa HA (2015) Identification of metabolites of rupestonic acid in rat urine by liquid chromatography combined with electrospray ionization quadrupole time-of-flight tandem mass spectrometry. Biomed Chromatogr 29:595–603. [DOI] [PubMed] [Google Scholar]
- Guillemette C (2003) Pharmacogenomics of human UDP-glucuronosyltransferase enzymes. Pharmacogenomics J 3:136–158. [DOI] [PubMed] [Google Scholar]
- Guo L, Duan L, Dong X, Dou LL, Zhou P, Li P, and Liu EH (2015) Metabolic profile of miltirone in rats by high performance liquid chromatography/quadrupole time-of-flight mass spectrometry. J Pharm Biomed Anal 107:473–479. [DOI] [PubMed] [Google Scholar]
- Hande K, Anthony L, Hamilton R, Bennett R, Sweetman B, and Branch R (1988a) Identification of etoposide glucuronide as a major metabolite of etoposide in the rat and rabbit. Cancer Res 48:1829–1834. [PubMed] [Google Scholar]
- Hande K, Bennett R, Hamilton R, Grote T, and Branch R (1988b) Metabolism and excretion of etoposide in isolated, perfused rat liver models. Cancer Res 48:5692–5695. [PubMed] [Google Scholar]
- Harding D, Fournel-Gigleux S, Jackson MR, and Burchell B (1988) Cloning and substrate specificity of a human phenol UDP-glucuronosyltransferase expressed in COS-7 cells. Proc Natl Acad Sci U S A 85:8381–8385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hattori M, Endo Y, Takebe S, Kobashi K, Fukasaku N, and Namba T (1986) Metabolism of magnolol from Magnoliae cortex. II. Absorption, metabolism and excretion of [ring-14C]magnolol in rats. Chem Pharm Bull (Tokyo) 34:158–167. [DOI] [PubMed] [Google Scholar]
- Hirano H, Kurata A, Onishi Y, Sakurai A, Saito H, Nakagawa H, Nagakura M, Tarui S, Kanamori Y, Kitajima M, and Ishikawa T (2006) High-speed screening and QSAR analysis of human ATP-binding cassette transporter ABCB11 (bile salt export pump) to predict drug-induced intrahepatic cholestasis. Mol Pharm 3:252–265. [DOI] [PubMed] [Google Scholar]
- Hirano M, Maeda K, Hayashi H, Kusuhara H, and Sugiyama Y (2005) Bile salt export pump (BSEP/ABCB11) can transport a nonbile acid substrate, pravastatin. J Pharmacol Exp Ther 314:876–882. [DOI] [PubMed] [Google Scholar]
- Hoffmann K and Loscher W (2007) Upregulation of brain expression of P-glycoprotein in MRP2-deficient TR(−) rats resembles seizure-induced up-regulation of this drug efflux transporter in normal rats. Epilepsia 48:631–645. [DOI] [PubMed] [Google Scholar]
- Hollman PC, van Trijp JM, Mengelers MJ, de Vries JH, and Katan MB (1997) Bioavailability of the dietary antioxidant flavonol quercetin in man. Cancer Lett 114:139–140. [DOI] [PubMed] [Google Scholar]
- Hu M (2007) Commentary: bioavailability of flavonoids and polyphenols: call to arms. Mol Pharm 4:803–806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu M, Chen J, and Lin H (2003) Metabolism of flavonoids via enteric recycling: mechanistic studies of disposition of apigenin in the Caco-2 cell culture model. J Pharmacol Exp Ther 307:314–321. [DOI] [PubMed] [Google Scholar]
- Itoh T, Takemoto I, Itagaki S, Sasaki K, Hirano T, and Iseki K (2004) Biliary excretion of irinotecan and its metabolites. J Pharm Pharm Sci 7:13–18. [PubMed] [Google Scholar]
- Iusuf D, van de Steeg E, and Schinkel AH (2012) Hepatocyte hopping of OATP1B substrates contributes to efficient hepatic detoxification. Clin Pharmacol Ther 92:559–562. [DOI] [PubMed] [Google Scholar]
- Izukawa T, Nakajima M, Fujiwara R, Yamanaka H, Fukami T, Takamiya M, Aoki Y, Ikushiro S, Sakaki T, and Yokoi T (2009) Quantitative analysis of UDP-glucuronosyltransferase (UGT) 1A and UGT2B expression levels in human livers. Drug Metab Dispos 37:1759–1768. [DOI] [PubMed] [Google Scholar]
- Jaganath IB, Mullen W, Edwards CA, and Crozier A (2006) The relative contribution of the small and large intestine to the absorption and metabolism of rutin in man. Free Radic Res 40:1035–1046. [DOI] [PubMed] [Google Scholar]
- Jager W, Gehring E, Hagenauer B, Aust S, Senderowicz A, and Thalhammer T (2003a) Biliary excretion of flavopiridol and its glucuronides in the isolated perfused rat liver: role of multidrug resistance protein 2 (Mrp2). Life Sci 73:2841–2854. [DOI] [PubMed] [Google Scholar]
- Jager W, Gehring E, Hagenauer B, Aust S, Senderowicz A, and Thalhammer T (2003b) The role of hepatic Mrp2 in the interaction of flavopiridol and bilirubin: impact on therapy. Int J Clin Pharmacol Ther 41:610–611. [DOI] [PubMed] [Google Scholar]
- James A, Blumenstein L, Glaenzel U, Jin Y, Demailly A, Jakab A, Hansen R, Hazell K, Mehta A, Trandafir L, and Swart P (2015) Absorption, distribution, metabolism, and excretion of [(14)C]BYL719 (alpelisib) in healthy male volunteers. Cancer Chemother Pharmacol 76:751–760. [DOI] [PubMed] [Google Scholar]
- Jansen PL, Peters WH, and Lamers WH (1985) Hereditary chronic conjugated hyperbilirubinemia in mutant rats caused by defective hepatic anion transport. Hepatology 5:573–579. [DOI] [PubMed] [Google Scholar]
- Jemnitz K, Heredi-Szabo K, Janossy J, Ioja E, Vereczkey L, and Krajcsi P (2010) ABCC2/Abcc2: a multispecific transporter with dominant excretory functions. Drug Metab Rev 42:402–436. [DOI] [PubMed] [Google Scholar]
- Jeong EJ, Jia X, and Hu M (2005a) Disposition of formononetin via enteric recycling: metabolism and excretion in mouse intestinal perfusion and Caco-2 cell models. Mol Pharm 2:319–328. [DOI] [PubMed] [Google Scholar]
- Jeong EJ, Liu X, Jia X, Chen J, and Hu M (2005b) Coupling of conjugating enzymes and efflux transporters: impact on bioavailability and drug interactions. Curr Drug Metab 6:455–468. [DOI] [PubMed] [Google Scholar]
- Jia X, Chen J, Lin H, and Hu M (2004) Disposition of flavonoids via enteric recycling: enzyme-transporter coupling affects metabolism of biochanin A and formononetin and excretion of their phase II conjugates. J Pharmacol Exp Ther 310:1103–1113. [DOI] [PubMed] [Google Scholar]
- Jiang W and Hu M (2012) Mutual interactions between flavonoids and enzymatic and transporter elements responsible for flavonoid disposition via phase II metabolic pathways. RSC Adv 2:7948–7963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jiang W, Xu B, Wu B, Yu R, and Hu M (2012) UDP-glucuronosyltransferase (UGT) 1A9-overexpressing HeLa cells is an appropriate tool to delineate the kinetic interplay between breast cancer resistance protein (BRCP) and UGT and to rapidly identify the glucuronide substrates of BCRP. Drug Metab Dispos 40:336–345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jirasko R, Holcapek M, and Nobilis M (2011) Identification of phase I and phase II metabolites of benfluron and dimefluron in rat urine using high-performance liquid chromatography/tandem mass spectrometry. Rapid Commun Mass Spectrom 25:2153–2162. [DOI] [PubMed] [Google Scholar]
- Johnson BM, Zhang P, Schuetz JD, and Brouwer KL (2006) Characterization of transport protein expression in multidrug resistance-associated protein (Mrp) 2-deficient rats. Drug Metab Dispos 34:556–562. [DOI] [PubMed] [Google Scholar]
- Jonker JW and Schinkel AH (2004) Pharmacological and physiological functions of the polyspecific organic cation transporters: OCT1, 2, and 3 (SLC22A1–3). J Pharmacol Exp Ther 308:2–9. [DOI] [PubMed] [Google Scholar]
- Joy MS, Hilliard T, Hu Y, Hogan SL, Dooley MA, Falk RJ, and Smith PC (2009) Pharmacokinetics of mycophenolic acid in patients with lupus nephritis. Pharmacotherapy 29:7–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaivosaari S, Toivonen P, Hesse LM, Koskinen M, Court MH, and Finel M (2007) Nicotine glucuronidation and the human UDP-glucuronosyltransferase UGT2B10. Mol Pharmacol 72:761–768. [DOI] [PubMed] [Google Scholar]
- Kalliokoski A and Niemi M (2009) Impact of OATP transporters on pharmacokinetics. Br J Pharmacol 158:693–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kamdem LK, Liu Y, Stearns V, Kadlubar SA, Ramirez J, Jeter S, Shahverdi K, Ward BA, Ogburn E, Ratain MJ, Flockhart DA, and Desta Z (2010) In vitro and in vivo oxidative metabolism and glucuronidation of anastrozole. Br J Clin Pharmacol 70:854–869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kamisako T, Kobayashi Y, Takeuchi K, Ishihara T, Higuchi K, Tanaka Y, Gabazza EC, and Adachi Y (2000) Recent advances in bilirubin metabolism research: the molecular mechanism of hepatocyte bilirubin transport and its clinical relevance. J Gastroenterol 35:659–664. [DOI] [PubMed] [Google Scholar]
- Kamisako T, Leier I, Cui Y, Konig J, Buchholz U, Hummel-Eisenbeiss J, and Keppler D (1999) Transport of monoglucuronosyl and bisglucuronosyl bilirubin by recombinant human and rat multidrug resistance protein 2. Hepatology 30:485–490. [DOI] [PubMed] [Google Scholar]
- Kaneda N, Nagata H, Furuta T, and Yokokura T (1990) Metabolism and pharmacokinetics of the camptothecin analogue CPT-11 in the mouse. Cancer Res 50:1715–1720. [PubMed] [Google Scholar]
- Kato A, Ueyama J, Abe F, Hotta K, Tsukiyama I, Oshima T, Kondo F, Saito H, and Hasegawa T (2011) Panipenem does not alter the pharmacokinetics of the active metabolite of irinotecan SN-38 and inactive metabolite SN-38 glucuronide (SN-38G) in rats. Anticancer Res 31:2915–2922. [PubMed] [Google Scholar]
- Kawai K, Kawasaki-Tokui Y, Odaka T, Tsuruta F, Kazui M, Iwabuchi H, Nakamura T, Kinoshita T, Ikeda T, Yoshioka T, Komai T, and Nakamura K (1997) Disposition and metabolism of the new oral antidiabetic drug troglitazone in rats, mice and dogs. Arzneimittelforschung 47:356–368. [PubMed] [Google Scholar]
- Keppler D (2011) Multidrug resistance proteins (MRPs, ABCCs): importance for pathophysiology and drug therapy. Handb Exp Pharmacol:299–323. [DOI] [PubMed] [Google Scholar]
- Keppler D (2014) The roles of MRP2, MRP3, OATP1B1, and OATP1B3 in conjugated hyperbilirubinemia. Drug Metab Dispos 42:561–565. [DOI] [PubMed] [Google Scholar]
- Keppler D and Konig J (1997) Hepatic canalicular membrane 5: Expression and localization of the conjugate export pump encoded by the MRP2 (cMRP/cMOAT) gene in liver. FASEB J 11:509–516. [DOI] [PubMed] [Google Scholar]
- Keppler D and Konig J (2000) Hepatic secretion of conjugated drugs and endogenous substances. Semin Liver Dis 20:265–272. [DOI] [PubMed] [Google Scholar]
- Kiang TK, Ensom MH, and Chang TK (2005) UDP-glucuronosyltransferases and clinical drug-drug interactions. Pharmacol Ther 106:97–132. [DOI] [PubMed] [Google Scholar]
- Kindla J, Fromm MF, and Konig J (2009) In vitro evidence for the role of OATP and OCT uptake transporters in drug-drug interactions. Expert Opin Drug Metab Toxicol 5:489–500. [DOI] [PubMed] [Google Scholar]
- King RA and Bursill DB (1998) Plasma and urinary kinetics of the isoflavones daidzein and genistein after a single soy meal in humans. Am J Clin Nutr 67:867–872. [DOI] [PubMed] [Google Scholar]
- Kitamura Y, Kusuhara H, and Sugiyama Y (2010) Functional characterization of multidrug resistance-associated protein 3 (mrp3/abcc3) in the basolateral efflux of glucuronide conjugates in the mouse small intestine. J Pharmacol Exp Ther 332:659–666. [DOI] [PubMed] [Google Scholar]
- Klaassen CD and Aleksunes LM (2010) Xenobiotic, bile acid, and cholesterol transporters: function and regulation. Pharmacol Rev 62:1–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kock K and Brouwer KL (2012) A perspective on efflux transport proteins in the liver. Clin Pharmacol Ther 92:599–612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kool M, van der Linden M, de Haas M, Baas F, and Borst P (1999) Expression of human MRP6, a homologue of the multidrug resistance protein gene MRP1, in tissues and cancer cells. Cancer Res 59:175–182. [PubMed] [Google Scholar]
- Kosaka K, Sakai N, Endo Y, Fukuhara Y, Tsuda-Tsukimoto M, Ohtsuka T, Kino I, Tanimoto T, Takeba N, Takahashi M, and Kume T (2011) Impact of intestinal glucuronidation on the pharmacokinetics of raloxifene. Drug Metab Dispos 39:1495–1502. [DOI] [PubMed] [Google Scholar]
- Kosoglou T, Statkevich P, Johnson-Levonas AO, Paolini JF, Bergman AJ, and Alton KB (2005) Ezetimibe: a review of its metabolism, pharmacokinetics and drug interactions. Clin Pharmacokinet 44:467–494. [DOI] [PubMed] [Google Scholar]
- Kouzuki H, Suzuki H, Ito K, Ohashi R, and Sugiyama Y (1999) Contribution of organic anion transporting polypeptide to uptake of its possible substrates into rat hepatocytes. J Pharmacol Exp Ther 288:627–634. [PubMed] [Google Scholar]
- Kubo K, Sekine S, and Saito M (2009) Compensatory expression of MRP3 in the livers of MRP2-deficient EHBRs is promoted by DHA intake. Biosci Biotechnol Biochem 73:2432–2438. [DOI] [PubMed] [Google Scholar]
- Kuhnle G, Spencer JP, Schroeter H, Shenoy B, Debnam ES, Srai SK, Rice-Evans C, and Hahn U (2000) Epicatechin and catechin are O-methylated and glucuronidated in the small intestine. Biochem Biophys Res Commun 277:507–512. [DOI] [PubMed] [Google Scholar]
- Lagas JS, Sparidans RW, Wagenaar E, Beijnen JH, and Schinkel AH (2010) Hepatic clearance of reactive glucuronide metabolites of diclofenac in the mouse is dependent on multiple ATP-binding cassette efflux transporters. Mol Pharmacol 77:687–694. [DOI] [PubMed] [Google Scholar]
- Lai MY, Hsiu SL, Chen CC, Hou YC, and Chao PD (2003a) Urinary pharmacokinetics of baicalein, wogonin and their glycosides after oral administration of Scutellariae Radix in humans. Biol Pharm Bull 26:79–83. [DOI] [PubMed] [Google Scholar]
- Lai MY, Hsiu SL, Tsai SY, Hou YC, and Chao PD (2003b) Comparison of metabolic pharmacokinetics of baicalin and baicalein in rats. J Pharm Pharmacol 55:205–209. [DOI] [PubMed] [Google Scholar]
- Lai YC, Kuo TF, Chen CK, Tsai HJ, and Lee SS (2010) Metabolism of dicentrine: identification of the phase I and phase II metabolites in miniature pig urine. Drug Metab Dispos 38:1714–1722. [DOI] [PubMed] [Google Scholar]
- Lakshmi VM, Hsu FF, and Zenser TV (2009) Identification of new 2-amino-3-methylimidazo[4,5-f]quinoline urinary metabolites from beta-naphthoflavone-treated mice. Drug Metab Dispos 37:1690–1697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Laugesen S, Enggaard TP, Pedersen RS, Sindrup SH, and Brosen K (2005) Paroxetine, a cytochrome P450 2D6 inhibitor, diminishes the stereoselective O-demethylation and reduces the hypoalgesic effect of tramadol. Clin Pharmacol Ther 77:312–323. [DOI] [PubMed] [Google Scholar]
- Li C, Homma M, and Oka K (1998) Characteristics of delayed excretion of flavonoids in human urine after administration of Shosaiko-to, a herbal medicine. Biol Pharm Bull 21:1251–1257. [DOI] [PubMed] [Google Scholar]
- Li Y, Zhou J, Ramsden D, Taub ME, O’Brien D, Xu J, Busacca CA, Gonnella N, and Tweedie DJ (2014) Enzyme-transporter interplay in the formation and clearance of abundant metabolites of faldaprevir found in excreta but not in circulation. Drug Metab Dispos 42:384–393. [DOI] [PubMed] [Google Scholar]
- Lin LC, Pai YF, and Tsai TH (2015) Isolation of Luteolin and Luteolin-7-O-glucoside from Dendranthema morifolium Ramat Tzvel and Their Pharmacokinetics in Rats. J Agric Food Chem 63:7700–7706. [DOI] [PubMed] [Google Scholar]
- Liu GY, Wang W, Jia WD, Xu GL, Ma JL, Ge YS, Yu JH, Sun QK, and Meng FL (2014) Protective effect of S-adenosylmethionine on hepatic ischemia-reperfusion injury during hepatectomy in HCC patients with chronic HBV infection. World J Surg Oncol 12:27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu HX, Liu Y, Zhang JW, Li W, Liu HT, and Yang L (2008) UDP-glucuronosyltransferase 1A6 is the major isozyme responsible for protocatechuic aldehyde glucuronidation in human liver microsomes. Drug Metab Dispos 36:1562–1569. [DOI] [PubMed] [Google Scholar]
- Liu L, Cui Y, Chung AY, Shitara Y, Sugiyama Y, Keppler D, and Pang KS (2006) Vectorial transport of enalapril by Oatp1a1/Mrp2 and OATP1B1 and OATP1B3/MRP2 in rat and human livers. J Pharmacol Exp Ther 318:395–402. [DOI] [PubMed] [Google Scholar]
- Liu W, Feng Q, Li Y, Ye L, Hu M, and Liu Z (2012a) Coupling of UDP-glucuronosyltransferases and multidrug resistance-associated proteins is responsible for the intestinal disposition and poor bioavailability of emodin. Toxicol Appl Pharmacol 265:316–324. [DOI] [PubMed] [Google Scholar]
- Liu X, Li H, Bi KS, Chen XH, Cai H, and Cai BC (2012b) [Identification of metabolites of arbidol by ultra-high performance liquid chromatography tandem mass spectrometry]. Yao Xue Xue Bao 47:1521–1526. [PubMed] [Google Scholar]
- Liu Y, Hao H, Xie H, Lv H, Liu C, and Wang G (2009) Oxidative demethylenation and subsequent glucuronidation are the major metabolic pathways of berberine in rats. J Pharm Sci 98:4391–4401. [DOI] [PubMed] [Google Scholar]
- Liu Y, Liu Y, Dai Y, Xun L, and Hu M (2003) Enteric disposition and recycling of flavonoids and ginkgo flavonoids. J Altern Complement Med 9:631–640. [DOI] [PubMed] [Google Scholar]
- Liu Z and Hu M (2007) Natural polyphenol disposition via coupled metabolic pathways. Expert Opin Drug Metab Toxicol 3:389–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lo MW, Pond SM, Effeney DJ, Silber BM, Riegelman S, and Tozer TN (1984) Nonlinear formation of propranolol metabolites in dogs after portacaval transpositions. J Pharmacokinet Biopharm 12:401–412. [DOI] [PubMed] [Google Scholar]
- LoGuidice A, Wallace BD, Bendel L, Redinbo MR, and Boelsterli UA (2012) Pharmacologic targeting of bacterial beta-glucuronidase alleviates nonsteroidal anti-inflammatory drug-induced enteropathy in mice. J Pharmacol Exp Ther 341:447–454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Loi CM, Young M, Randinitis E, Vassos A, and Koup JR (1999) Clinical pharmacokinetics of troglitazone. Clin Pharmacokinet 37:91–104. [DOI] [PubMed] [Google Scholar]
- Lokiec F, Canal P, Gay C, Chatelut E, Armand JP, Roche H, Bugat R, Goncalves E, and Mathieu-Boue A (1995) Pharmacokinetics of irinotecan and its metabolites in human blood, bile, and urine. Cancer Chemother Pharmacol 36:79–82. [DOI] [PubMed] [Google Scholar]
- Loureiro AI, Rocha JF, Fernandes-Lopes C, Nunes T, Wright LC, Almeida L, and Soares-da-Silva P (2014) Human disposition, metabolism and excretion of etamicastat, a reversible, peripherally selective dopamine beta-hydroxylase inhibitor. Br J Clin Pharmacol 77:1017–1026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Luo CF, Cai B, Hou N, Yuan M, Liu SM, Ji H, Xiong LG, Xiong W, Luo JD, and Chen MS (2012) UDP-glucuronosyltransferase 1A1 is the principal enzyme responsible for puerarin metabolism in human liver microsomes. Arch Toxicol 86:1681–1690. [DOI] [PubMed] [Google Scholar]
- Luukkanen L, Taskinen J, Kurkela M, Kostiainen R, Hirvonen J, and Finel M (2005) Kinetic characterization of the 1A subfamily of recombinant human UDP-glucuronosyltransferases. Drug Metab Dispos 33:1017–1026. [DOI] [PubMed] [Google Scholar]
- Ma G, Lin J, Cai W, Tan B, Xiang X, Zhang Y, and Zhang P (2014) Simultaneous determination of bilirubin and its glucuronides in liver microsomes and recombinant UGT1A1 enzyme incubation systems by HPLC method and its application to bilirubin glucuronidation studies. J Pharm Biomed Anal 92:149–159. [DOI] [PubMed] [Google Scholar]
- Ma L, Sun J, Peng Y, Zhang R, Shao F, Hu X, Zhu J, Wang X, Cheng X, Zhu Y, Wan P, Feng D, Wu H, and Wang G (2012) Glucuronidation of edaravone by human liver and kidney microsomes: biphasic kinetics and identification of UGT1A9 as the major UDP-glucuronosyltransferase isoform. Drug Metab Dispos 40:734–741. [DOI] [PubMed] [Google Scholar]
- Mackenzie PI, Bock KW, Burchell B, Guillemette C, Ikushiro S, Iyanagi T, Miners JO, Owens IS, and Nebert DW (2005) Nomenclature update for the mammalian UDP glycosyltransferase (UGT) gene superfamily. Pharmacogenet Genomics 15:677–685. [DOI] [PubMed] [Google Scholar]
- Mackenzie PI, Gregory PA, Gardner-Stephen DA, Lewinsky RH, Jorgensen BR, Nishiyama T, Xie W, and Radominska-Pandya A (2003) Regulation of UDP glucuronosyltransferase genes. Curr Drug Metab 4:249–257. [DOI] [PubMed] [Google Scholar]
- Madon J, Hagenbuch B, Landmann L, Meier PJ, and Stieger B (2000) Transport function and hepatocellular localization of mrp6 in rat liver. Mol Pharmacol 57:634–641. [DOI] [PubMed] [Google Scholar]
- Maier-Salamon A, Hagenauer B, Reznicek G, Szekeres T, Thalhammer T, and Jager W (2008) Metabolism and disposition of resveratrol in the isolated perfused rat liver: role of Mrp2 in the biliary excretion of glucuronides. J Pharm Sci 97:1615–1628. [DOI] [PubMed] [Google Scholar]
- Manach C, Morand C, Texier O, Favier ML, Agullo G, Demigne C, Regerat F, and Remesy C (1995) Quercetin metabolites in plasma of rats fed diets containing rutin or quercetin. J Nutr 125:1911–1922. [DOI] [PubMed] [Google Scholar]
- Marin JJ (2012) Plasma membrane transporters in modern liver pharmacology. Scientifica (Cairo) 2012:428139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marquez B, Caceres NE, Mingeot-Leclercq MP, Tulkens PM, and Van Bambeke F (2009) Identification of the efflux transporter of the fluoroquinolone antibiotic ciprofloxacin in murine macrophages: studies with ciprofloxacin-resistant cells. Antimicrob Agents Chemother 53:2410–2416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Marshall AW, Mihaly GW, Smallwood RA, Morgan DJ, and Hardy KJ (1981) Fetal hepatic function: the disposition of propranolol in the pregnant sheep. Res Commun Chem Pathol Pharmacol 32:3–25. [PubMed] [Google Scholar]
- Matsunaga N, Wada S, Nakanishi T, Ikenaga M, Ogawa M, and Tamai I (2014) Mathematical modeling of the in vitro hepatic disposition of mycophenolic acid and its glucuronide in sandwich-cultured human hepatocytes. Mol Pharm 11:568–579. [DOI] [PubMed] [Google Scholar]
- Mazur CS, Marchitti SA, Dimova M, Kenneke JF, Lumen A, and Fisher J (2012) Human and rat ABC transporter efflux of bisphenol a and bisphenol a glucuronide: interspecies comparison and implications for pharmacokinetic assessment. Toxicol Sci 128:317–325. [DOI] [PubMed] [Google Scholar]
- Meerman JH, Nijland C, and Mulder GJ (1987) Sex differences in sulfation and glucuronidation of phenol, 4-nitrophenol and N-hydroxy-2-acetylaminofluorene in the rat in vivo. Biochem Pharmacol 36:2605–2608. [DOI] [PubMed] [Google Scholar]
- Meng X, Maliakal P, Lu H, Lee MJ, and Yang CS (2004) Urinary and plasma levels of resveratrol and quercetin in humans, mice, and rats after ingestion of pure compounds and grape juice. J Agric Food Chem 52:935–942. [DOI] [PubMed] [Google Scholar]
- Meyer GM, Meyer MR, Wissenbach DK, and Maurer HH (2013) Studies on the metabolism and toxicological detection of glaucine, an isoquinoline alkaloid from Glaucium flavum (Papaveraceae), in rat urine using GC-MS, LC-MS(n) and LC-high-resolution MS(n). J Mass Spectrom 48:24–41. [DOI] [PubMed] [Google Scholar]
- Meyer MR, Holderbaum A, Kavanagh P, and Maurer HH (2015) Low resolution and high resolution MS for studies on the metabolism and toxicological detection of the new psychoactive substance methoxypiperamide (MeOP). J Mass Spectrom 50:1163–1174. [DOI] [PubMed] [Google Scholar]
- Meyer zu Schwabedissen HE and Kroemer HK (2011) In vitro and in vivo evidence for the importance of breast cancer resistance protein transporters (BCRP/MXR/ABCP/ABCG2). Handb Exp Pharmacol:325–371. [DOI] [PubMed] [Google Scholar]
- Michels GM, Boudinot FD, Ferguson DC, and Hoenig M (2000) Pharmacokinetics of the insulin-sensitizing agent troglitazone in cats. Am J Vet Res 61:775–778. [DOI] [PubMed] [Google Scholar]
- Michely JA, Helfer AG, Brandt SD, Meyer MR, and Maurer HH (2015) Metabolism of the new psychoactive substances N,N-diallyltryptamine (DALT) and 5-methoxy-DALT and their detectability in urine by GC-MS, LC-MSn, and LC-HR-MS-MS. Anal Bioanal Chem 407:7831–7842. [DOI] [PubMed] [Google Scholar]
- Ming X (2008) Role of Basolateral Efflux Transporters in Intestinal Absorption of Drugs and Prodrugs in: School of Pharmacy, pp 216, University of North Carolina at Chapel Hill Chapel Hill, North Carolina. [Google Scholar]
- Miyawaki I, Tamura A, Matsumoto I, Inada H, Kunimatsu T, Kimura J, and Funabashi H (2012) The effects of clobazam treatment in rats on the expression of genes and proteins encoding glucronosyltransferase 1A/2B (UGT1A/2B) and multidrug resistance-associated protein-2 (MRP2), and development of thyroid follicular cell hypertrophy. Toxicol Appl Pharmacol 265:351–359. [DOI] [PubMed] [Google Scholar]
- Miyazaki T, Mizukoshi H, Araki Y, and Shimizu N (1980) The metabolism of estriol-3-glucosiduronate and estriol in the rabbit. Endocrinol Jpn 27:175–182. [DOI] [PubMed] [Google Scholar]
- Miyazaki T, Peric-Golia L, Slaunwhite WR Jr., and Sandberg AA (1972) Estriol metabolism in sheep: excretion of biliary and urinary conjugates. Endocrinology 90:516–524. [DOI] [PubMed] [Google Scholar]
- Mizuno N, Takahashi T, Kusuhara H, Schuetz JD, Niwa T, and Sugiyama Y (2007) Evaluation of the role of breast cancer resistance protein (BCRP/ABCG2) and multidrug resistance-associated protein 4 (MRP4/ABCC4) in the urinary excretion of sulfate and glucuronide metabolites of edaravone (MCI-186; 3-methyl-1-phenyl-2-pyrazolin-5-one). Drug Metab Dispos 35:2045–2052. [DOI] [PubMed] [Google Scholar]
- Moon YJ and Morris ME (2007) Pharmacokinetics and bioavailability of the bioflavonoid biochanin A: effects of quercetin and EGCG on biochanin A disposition in rats. Mol Pharm 4:865–872. [DOI] [PubMed] [Google Scholar]
- Morand C, Crespy V, Manach C, Besson C, Demigne C, and Remesy C (1998) Plasma metabolites of quercetin and their antioxidant properties. Am J Physiol 275:R212–219. [DOI] [PubMed] [Google Scholar]
- Morikawa A, Goto Y, Suzuki H, Hirohashi T, and Sugiyama Y (2000) Biliary excretion of 17beta-estradiol 17beta-D-glucuronide is predominantly mediated by cMOAT/MRP2. Pharm Res 17:546–552. [DOI] [PubMed] [Google Scholar]
- Motheova O, Bezek S, Durisova M, Faberova V, Zemanek M, Misanikova K, and Trnovec T (1986) The pharmacokinetics of exaprolol and propranolol in rats with interrupted enterohepatic circulation. Biopharm Drug Dispos 7:151–162. [DOI] [PubMed] [Google Scholar]
- Musey PI, Kirdani RY, Bhanalaph T, and Sandberg AA (1973) Estriol metabolism in the baboon: analysis of urinary and biliary metabolites. Steroids 22:795–817. [DOI] [PubMed] [Google Scholar]
- Nadal T, Ortuno J, and Pascual JA (1996) Rapid and sensitive determination of zidovudine and zidovudine glucuronide in human plasma by ion-pair high-performance liquid chromatography. J Chromatogr A 721:127–137. [DOI] [PubMed] [Google Scholar]
- Naesens M, de Loor H, Vanrenterghem Y, and Kuypers DR (2007) The impact of renal allograft function on exposure and elimination of mycophenolic acid (MPA) and its metabolite MPA 7-O-glucuronide. Transplantation 84:362–373. [DOI] [PubMed] [Google Scholar]
- Nagar S and Blanchard RL (2006) Pharmacogenetics of uridine diphosphoglucuronosyltransferase (UGT) 1A family members and its role in patient response to irinotecan. Drug Metab Rev 38:393–409. [DOI] [PubMed] [Google Scholar]
- Nagar S and Remmel RP (2006) Uridine diphosphoglucuronosyltransferase pharmacogenetics and cancer. Oncogene 25:1659–1672. [DOI] [PubMed] [Google Scholar]
- Nakagomi-Hagihara R, Nakai D, and Tokui T (2007a) Inhibition of human organic anion transporter 3 mediated pravastatin transport by gemfibrozil and the metabolites in humans. Xenobiotica 37:416–426. [DOI] [PubMed] [Google Scholar]
- Nakagomi-Hagihara R, Nakai D, Tokui T, Abe T, and Ikeda T (2007b) Gemfibrozil and its glucuronide inhibit the hepatic uptake of pravastatin mediated by OATP1B1. Xenobiotica 37:474–486. [DOI] [PubMed] [Google Scholar]
- Nakanishi T and Ross DD (2012) Breast cancer resistance protein (BCRP/ABCG2): its role in multidrug resistance and regulation of its gene expression. Chin J Cancer 31:73–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nakazawa T, Yasuda T, and Ohsawa K (2003) Metabolites of orally administered Magnolia officinalis extract in rats and man and its antidepressant-like effects in mice. J Pharm Pharmacol 55:1583–1591. [DOI] [PubMed] [Google Scholar]
- Nambara T and Kawarada Y (1977) Biliary conjugated metabolites of estriol in the rat. Chem Pharm Bull (Tokyo) 25:942–948. [DOI] [PubMed] [Google Scholar]
- Neve EP, Artursson P, Ingelman-Sundberg M, and Karlgren M (2013) An integrated in vitro model for simultaneous assessment of drug uptake, metabolism, and efflux. Mol Pharm 10:3152–3163. [DOI] [PubMed] [Google Scholar]
- Ni ZL, Bikadi Z, Rosenberg MF, and Mao QC (2010) Structure and Function of the Human Breast Cancer Resistance Protein (BCRP/ABCG2). Curr Drug Metab 11:603–617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nies AT, Jedlitschky G, Konig J, Herold-Mende C, Steiner HH, Schmitt HP, and Keppler D (2004) Expression and immunolocalization of the multidrug resistance proteins, MRP1-MRP6 (ABCC1-ABCC6), in human brain. Neuroscience 129:349–360. [DOI] [PubMed] [Google Scholar]
- Nies AT, Schwab M, and Keppler D (2008) Interplay of conjugating enzymes with OATP uptake transporters and ABCC/MRP efflux pumps in the elimination of drugs. Expert Opin Drug Metab Toxicol 4:545–568. [DOI] [PubMed] [Google Scholar]
- Ohno S and Nakajin S (2009) Determination of mRNA expression of human UDP-glucuronosyltransferases and application for localization in various human tissues by real-time reverse transcriptase-polymerase chain reaction. Drug Metab Dispos 37:32–40. [DOI] [PubMed] [Google Scholar]
- Ohtsuki S, Schaefer O, Kawakami H, Inoue T, Liehner S, Saito A, Ishiguro N, Kishimoto W, Ludwig-Schwellinger E, Ebner T, and Terasaki T (2012) Simultaneous absolute protein quantification of transporters, cytochromes P450, and UDP-glucuronosyltransferases as a novel approach for the characterization of individual human liver: comparison with mRNA levels and activities. Drug Metab Dispos 40:83–92. [DOI] [PubMed] [Google Scholar]
- Osborne R, Joel S, Grebenik K, Trew D, and Slevin M (1993) The pharmacokinetics of morphine and morphine glucuronides in kidney failure. Clin Pharmacol Ther 54:158–167. [DOI] [PubMed] [Google Scholar]
- Oswald S, Koll C, and Siegmund W (2007) Disposition of the cholesterol absorption inhibitor ezetimibe in mdr1a/b (−/−) mice. J Pharm Sci 96:3478–3484. [DOI] [PubMed] [Google Scholar]
- Oswald S, Konig J, Lutjohann D, Giessmann T, Kroemer HK, Rimmbach C, Rosskopf D, Fromm MF, and Siegmund W (2008) Disposition of ezetimibe is influenced by polymorphisms of the hepatic uptake carrier OATP1B1. Pharmacogenet Genomics 18:559–568. [DOI] [PubMed] [Google Scholar]
- Oswald S, May K, Rosin J, Lutjohann D, and Siegmund W (2010) Synergistic influence of Abcb1 and Abcc2 on disposition and sterol lowering effects of ezetimibe in rats. J Pharm Sci 99:422–429. [DOI] [PubMed] [Google Scholar]
- Ouellet DM and Pollack GM (1995) Biliary excretion and enterohepatic recirculation of morphine-3-glucuronide in rats. Drug Metab Dispos 23:478–484. [PubMed] [Google Scholar]
- Owens IS, Basu NK, and Banerjee R (2005) UDP-glucuronosyltransferases: gene structures of UGT1 and UGT2 families. Methods Enzymol 400:1–22. [DOI] [PubMed] [Google Scholar]
- Pan S, Neeraj A, Srivastava KS, Kishore P, Danquah MK, and Sarethy IP (2013) A proposal for a quality system for herbal products. J Pharm Sci 102:4230–4241. [DOI] [PubMed] [Google Scholar]
- Pang KS, Maeng HJ, and Fan J (2009) Interplay of transporters and enzymes in drug and metabolite processing. Mol Pharm 6:1734–1755. [DOI] [PubMed] [Google Scholar]
- Peng HW, Huang YT, Chen CF, and Tsai TH (1998) Glucuronidation of naringenin in rats. Planta Med 64:779. [DOI] [PubMed] [Google Scholar]
- Peng KW, Bacon J, Zheng M, Guo Y, and Wang MZ (2015) Ethnic variability in the expression of hepatic drug transporters: absolute quantification by an optimized targeted quantitative proteomic approach. Drug Metab Dispos 43:1045–1055. [DOI] [PubMed] [Google Scholar]
- Perera MA, Innocenti F, and Ratain MJ (2008) Pharmacogenetic testing for uridine diphosphate glucuronosyltransferase 1A1 polymorphisms: are we there yet? Pharmacotherapy 28:755–768. [DOI] [PubMed] [Google Scholar]
- Pfeifer ND, Hardwick RN, and Brouwer KL (2014) Role of hepatic efflux transporters in regulating systemic and hepatocyte exposure to xenobiotics. Annu Rev Pharmacol Toxicol 54:509–535. [DOI] [PubMed] [Google Scholar]
- Phillips IR, Shephard EA, Povey S, Davis MB, Kelsey G, Monteiro M, West LF, and Cowell J (1985) A cytochrome P-450 gene family mapped to human chromosome 19. Ann Hum Genet 49:267–274. [DOI] [PubMed] [Google Scholar]
- Pogell BM and Leloir LF (1961) Nucleotide activation of liver microsomal glucuronidation. J Biol Chem 236:293–298. [PubMed] [Google Scholar]
- Qian MR and Zeng S (2006) Biosynthesis of imipramine glucuronide and characterization of imipramine glucuronidation catalyzed by recombinant UGT1A4. Acta Pharmacol Sin 27:623–628. [DOI] [PubMed] [Google Scholar]
- Radominska-Pandya A, Bratton SM, Redinbo MR, and Miley MJ (2010) The crystal structure of human UDP-glucuronosyltransferase 2B7 C-terminal end is the first mammalian UGT target to be revealed: the significance for human UGTs from both the 1A and 2B families. Drug Metab Rev 42:133–144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Radominska-Pandya A, Little JM, Pandya JT, Tephly TR, King CD, Barone GW, and Raufman JP (1998) UDP-glucuronosyltransferases in human intestinal mucosa. Biochim Biophys Acta 1394:199–208. [DOI] [PubMed] [Google Scholar]
- Ramanathan R, Reyderman L, Kulmatycki K, Su AD, Alvarez N, Chowdhury SK, Alton KB, Wirth MA, Clement RP, Statkevich P, and Patrick JE (2007) Disposition of loratadine in healthy volunteers. Xenobiotica 37:753–769. [DOI] [PubMed] [Google Scholar]
- Redmon JM, Shrestha B, Cerundolo R, and Court MH (2016) Soy isoflavone metabolism in cats compared with other species: urinary metabolite concentrations and glucuronidation by liver microsomes. Xenobiotica 46:406–415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reynolds JW (1966) 16-alpha-hydroxylation of pregnenolone by human liver. Steroids 7:261–271. [DOI] [PubMed] [Google Scholar]
- Roberts MS, Magnusson BM, Burczynski FJ, and Weiss M (2002) Enterohepatic circulation: physiological, pharmacokinetic and clinical implications. Clin Pharmacokinet 41:751–790. [DOI] [PubMed] [Google Scholar]
- Roth M, Obaidat A, and Hagenbuch B (2012) OATPs, OATs and OCTs: the organic anion and cation transporters of the SLCO and SLC22A gene superfamilies. Br J Pharmacol 165:1260–1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Routledge PA and Shand DG (1979) Clinical pharmacokinetics of propranolol. Clin Pharmacokinet 4:73–90. [DOI] [PubMed] [Google Scholar]
- Rowland A, Mackenzie PI, and Miners JO (2015) Transporter-mediated uptake of UDP-glucuronic acid by human liver microsomes: assay conditions, kinetics, and inhibition. Drug Metab Dispos 43:147–153. [DOI] [PubMed] [Google Scholar]
- Russel FG, Koenderink JB, and Masereeuw R (2008) Multidrug resistance protein 4 (MRP4/ABCC4): a versatile efflux transporter for drugs and signalling molecules. Trends Pharmacol Sci 29:200–207. [DOI] [PubMed] [Google Scholar]
- Sasabe H, Tsuji A, and Sugiyama Y (1998) Carrier-mediated mechanism for the biliary excretion of the quinolone antibiotic grepafloxacin and its glucuronide in rats. J Pharmacol Exp Ther 284:1033–1039. [PubMed] [Google Scholar]
- Schaiquevich P, Niselman A, and Rubio M (2002) Comparison of two compartmental models for describing ranitidine’s plasmatic profiles. Pharmacol Res 45:399–405. [DOI] [PubMed] [Google Scholar]
- Schwartz DE, Jordan JC, Vetter W, and Oesterhelt G (1979) Metabolic studies of ornidazole in the rat, in the dog and in man. Xenobiotica 9:571–581. [DOI] [PubMed] [Google Scholar]
- Seitz S and Boelsterli UA (1998) Diclofenac acyl glucuronide, a major biliary metabolite, is directly involved in small intestinal injury in rats. Gastroenterology 115:1476–1482. [DOI] [PubMed] [Google Scholar]
- Shelby MK, Cherrington NJ, Vansell NR, and Klaassen CD (2003) Tissue mRNA expression of the rat UDP-glucuronosyltransferase gene family. Drug Metab Dispos 31:326–333. [DOI] [PubMed] [Google Scholar]
- Shi J, Zheng L, Lin Z, Hou C, Liu W, Yan T, Zhu L, Wang Y, Lu L, and Liu Z (2015) Study of pharmacokinetic profiles and characteristics of active components and their metabolites in rat plasma following oral administration of the water extract of Astragali radix using UPLC-MS/MS. J Ethnopharmacol 169:183–194. [DOI] [PubMed] [Google Scholar]
- Shi S and Li Y (2014) Interplay of Drug-Metabolizing Enzymes and Transporters in Drug Absorption and Disposition. Curr Drug Metab 15:915–941. [DOI] [PubMed] [Google Scholar]
- Shia CS, Hou YC, Tsai SY, Huieh PH, Leu YL, and Chao PD (2010) Differences in pharmacokinetics and ex vivo antioxidant activity following intravenous and oral administrations of emodin to rats. J Pharm Sci 99:2185–2195. [DOI] [PubMed] [Google Scholar]
- Shimoi K, Okada H, Furugori M, Goda T, Takase S, Suzuki M, Hara Y, Yamamoto H, and Kinae N (1998) Intestinal absorption of luteolin and luteolin 7-O-beta-glucoside in rats and humans. FEBS Lett 438:220–224. [DOI] [PubMed] [Google Scholar]
- Shipkova M, Armstrong VW, Oellerich M, and Wieland E (2003) Acyl glucuronide drug metabolites: toxicological and analytical implications. Ther Drug Monit 25:1–16. [DOI] [PubMed] [Google Scholar]
- Silberberg M, Morand C, Mathevon T, Besson C, Manach C, Scalbert A, and Remesy C (2006) The bioavailability of polyphenols is highly governed by the capacity of the intestine and of the liver to secrete conjugated metabolites. Eur J Nutr 45:88–96. [DOI] [PubMed] [Google Scholar]
- Singh R and Hu M (2011) Drug Metabolism in Gastrointestinal Tract in: Oral Bioavailability: Basic Principles, Advanced Concepts, and Applications (Hu M and Li X eds), pp 91–109, Wiley, New Jersey. [Google Scholar]
- Singh R, Wu B, Tang L, and Hu M (2011a) Uridine diphosphate glucuronosyltransferase isoform-dependent regiospecificity of glucuronidation of flavonoids. J Agric Food Chem 59:7452–7464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Singh SP, Wahajuddin, Tewari D, Pradhan T, and Jain GK (2011b) PAMPA permeability, plasma protein binding, blood partition, pharmacokinetics and metabolism of formononetin, a methoxylated isoflavone. Food Chem Toxicol 49:1056–1062. [DOI] [PubMed] [Google Scholar]
- Song YL, Jing WH, Yan R, and Wang YT (2014) Metabolic characterization of (+/−)-praeruptorin A in vitro and in vivo by high performance liquid chromatography coupled with hybrid triple quadrupole-linear ion trap mass spectrometry and time-of-flight mass spectrometry. J Pharm Biomed Anal 90:98–110. [DOI] [PubMed] [Google Scholar]
- Stearns V, Johnson MD, Rae JM, Morocho A, Novielli A, Bhargava P, Hayes DF, Desta Z, and Flockhart DA (2003) Active tamoxifen metabolite plasma concentrations after coadministration of tamoxifen and the selective serotonin reuptake inhibitor paroxetine. J Natl Cancer Inst 95:1758–1764. [DOI] [PubMed] [Google Scholar]
- Stephanson N, Dahl H, Helander A, and Beck O (2005) Determination of urinary 5-hydroxytryptophol glucuronide by liquid chromatography-mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 816:107–112. [DOI] [PubMed] [Google Scholar]
- Stone AN, Mackenzie PI, Galetin A, Houston JB, and Miners JO (2003) Isoform selectivity and kinetics of morphine 3- and 6-glucuronidation by human udp-glucuronosyltransferases: evidence for atypical glucuronidation kinetics by UGT2B7. Drug Metab Dispos 31:1086–1089. [DOI] [PubMed] [Google Scholar]
- Stopfer P, Rathgen K, Bischoff D, Ludtke S, Marzin K, Kaiser R, Wagner K, and Ebner T (2011) Pharmacokinetics and metabolism of BIBF 1120 after oral dosing to healthy male volunteers. Xenobiotica 41:297–311. [DOI] [PubMed] [Google Scholar]
- Talbi A, Zhao D, Liu Q, Li J, Fan A, Yang W, Han X, and Chen X (2014) Pharmacokinetics, tissue distribution, excretion and plasma protein binding studies of wogonin in rats. Molecules 19:5538–5549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tan EY, Hartmann G, Chen Q, Pereira A, Bradley S, Doss G, Zhang AS, Ho JZ, Braun MP, Dean DC, Tang W, and Kumar S (2010) Pharmacokinetics, metabolism, and excretion of anacetrapib, a novel inhibitor of the cholesteryl ester transfer protein, in rats and rhesus monkeys. Drug Metab Dispos 38:459–473. [DOI] [PubMed] [Google Scholar]
- Tang L, Li Y, Chen WY, Zeng S, Dong LN, Peng XJ, Jiang W, Hu M, and Liu ZQ (2014) Breast cancer resistance protein-mediated efflux of luteolin glucuronides in HeLa cells overexpressing UDP-glucuronosyltransferase 1A9. Pharm Res 31:847–860. [DOI] [PubMed] [Google Scholar]
- Thilakarathna SH and Rupasinghe HP (2013) Flavonoid bioavailability and attempts for bioavailability enhancement. Nutrients 5:3367–3387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tian JX, Wang M, Xu L, Tian Y, Song R, Xu FG, and Zhang ZJ (2014) Metabolism of brucine: the important metabolic pathways of dihydroindole-type alkaloid for excretion in rats. Bioanalysis 6:137–149. [DOI] [PubMed] [Google Scholar]
- Tian S, He G, Song J, Wang S, Xin W, Zhang D, and Du G (2012) Pharmacokinetic study of baicalein after oral administration in monkeys. Fitoterapia 83:532–540. [DOI] [PubMed] [Google Scholar]
- Tian X, Swift B, Zamek-Gliszczynski MJ, Belinsky MG, Kruh GD, and Brouwer KL (2008) Impact of basolateral multidrug resistance-associated protein (Mrp) 3 and Mrp4 on the hepatobiliary disposition of fexofenadine in perfused mouse livers. Drug Metab Dispos 36:911–915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tong Z, Chandrasekaran A, DeMaio W, Espina R, Lu W, Jordan R, and Scatina J (2010) Metabolism of vabicaserin in mice, rats, dogs, monkeys, and humans. Drug Metab Dispos 38:2266–2277. [DOI] [PubMed] [Google Scholar]
- Town C, Henderson L, Chang D, Mortillo M, and Garland W (1993) Distribution of 1-aminobenzotriazole in male rats after administration of an oral dose. Xenobiotica 23:383–390. [DOI] [PubMed] [Google Scholar]
- Trdan T, Roskar R, Trontelj J, Ravnikar M, and Mrhar A (2011) Determination of raloxifene and its glucuronides in human urine by liquid chromatography-tandem mass spectrometry assay. J Chromatogr B Analyt Technol Biomed Life Sci 879:2323–2331. [DOI] [PubMed] [Google Scholar]
- Tukey RH and Strassburg CP (2000) Human UDP-glucuronosyltransferases: metabolism, expression, and disease. Annu Rev Pharmacol Toxicol 40:581–616. [DOI] [PubMed] [Google Scholar]
- Uchaipichat V, Mackenzie PI, Elliot DJ, and Miners JO (2006) Selectivity of substrate (trifluoperazine) and inhibitor (amitriptyline, androsterone, canrenoic acid, hecogenin, phenylbutazone, quinidine, quinine, and sulfinpyrazone) “probes” for human udp-glucuronosyltransferases. Drug Metab Dispos 34:449–456. [DOI] [PubMed] [Google Scholar]
- Uutela P, Karhu L, Piepponen P, Kaenmaki M, Ketola RA, and Kostiainen R (2009) Discovery of dopamine glucuronide in rat and mouse brain microdialysis samples using liquid chromatography tandem mass spectrometry. Anal Chem 81:427–434. [DOI] [PubMed] [Google Scholar]
- Uwai Y, Motohashi H, Tsuji Y, Ueo H, Katsura T, and Inui K (2007) Interaction and transport characteristics of mycophenolic acid and its glucuronide via human organic anion transporters hOAT1 and hOAT3. Biochem Pharmacol 74:161–168. [DOI] [PubMed] [Google Scholar]
- Vaidyanathan JB and Walle T (2002) Glucuronidation and sulfation of the tea flavonoid (−)-epicatechin by the human and rat enzymes. Drug Metab Dispos 30:897–903. [DOI] [PubMed] [Google Scholar]
- van de Steeg E, Stranecky V, Hartmannova H, Noskova L, Hrebicek M, Wagenaar E, van Esch A, de Waart DR, Oude Elferink RP, Kenworthy KE, Sticova E, al-Edreesi M, Knisely AS, Kmoch S, Jirsa M, and Schinkel AH (2012) Complete OATP1B1 and OATP1B3 deficiency causes human Rotor syndrome by interrupting conjugated bilirubin reuptake into the liver. J Clin Invest 122:519–528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van de Steeg E, Wagenaar E, van der Kruijssen CM, Burggraaff JE, de Waart DR, Elferink RP, Kenworthy KE, and Schinkel AH (2010) Organic anion transporting polypeptide 1a/1b-knockout mice provide insights into hepatic handling of bilirubin, bile acids, and drugs. J Clin Invest 120:2942–2952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van de Wetering K, Burkon A, Feddema W, Bot A, de Jonge H, Somoza V, and Borst P (2009) Intestinal breast cancer resistance protein (BCRP)/Bcrp1 and multidrug resistance protein 3 (MRP3)/Mrp3 are involved in the pharmacokinetics of resveratrol. Mol Pharmacol 75:876–885. [DOI] [PubMed] [Google Scholar]
- van Den Elsen JM, Kuntz DA, Hoedemaeker FJ, and Rose DR (1999) Antibody C219 recognizes an alpha-helical epitope on P-glycoprotein. Proc Natl Acad Sci U S A 96:13679–13684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van der Deen M, de Vries EG, Timens W, Scheper RJ, Timmer-Bosscha H, and Postma DS (2005) ATP-binding cassette (ABC) transporters in normal and pathological lung. Respir Res 6:59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vasilyeva A, Durmus S, Li L, Wagenaar E, Hu S, Gibson AA, Panetta JC, Mani S, Sparreboom A, Baker SD, and Schinkel AH (2015) Hepatocellular Shuttling and Recirculation of Sorafenib-Glucuronide Is Dependent on Abcc2, Abcc3, and Oatp1a/1b. Cancer Res 75:2729–2736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Villanueva SS, Ruiz ML, Ghanem CI, Luquita MG, Catania VA, and Mottino AD (2008) Hepatic synthesis and urinary elimination of acetaminophen glucuronide are exacerbated in bile duct-ligated rats. Drug Metab Dispos 36:475–480. [DOI] [PubMed] [Google Scholar]
- Wakabayashi Y, Lippincott-Schwartz J, and Arias IM (2004) Intracellular trafficking of bile salt export pump (ABCB11) in polarized hepatic cells: constitutive cycling between the canalicular membrane and rab11-positive endosomes. Mol Biol Cell 15:3485–3496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wallace BD, Wang H, Lane KT, Scott JE, Orans J, Koo JS, Venkatesh M, Jobin C, Yeh LA, Mani S, and Redinbo MR (2010) Alleviating cancer drug toxicity by inhibiting a bacterial enzyme. Science 330:831–835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walle T, Hsieh F, DeLegge MH, Oatis JE Jr., and Walle UK (2004) High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab Dispos 32:1377–1382. [DOI] [PubMed] [Google Scholar]
- Walle T, Otake Y, Brubaker JA, Walle UK, and Halushka PV (2001) Disposition and metabolism of the flavonoid chrysin in normal volunteers. Br J Clin Pharmacol 51:143–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walle T, Walle UK, and Olanoff LS (1985) Quantitative account of propranolol metabolism in urine of normal man. Drug Metab Dispos 13:204–209. [PubMed] [Google Scholar]
- Wang H, Fang ZZ, Zheng Y, Zhou K, Hu C, Krausz KW, Sun D, Idle JR, and Gonzalez FJ (2014) Metabolic profiling of praziquantel enantiomers. Biochem Pharmacol 90:166–178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang M, Yang G, He Y, Xu B, Zeng M, Ge S, Yin T, Gao S, and Hu M (2016) Establishment and use of new MDCK II cells overexpressing both UGT1A1 and MRP2 to characterize flavonoid metabolism via the glucuronidation pathway. Mol Nutr Food Res 60:1967–1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang SW, Chen J, Jia X, Tam VH, and Hu M (2006) Disposition of flavonoids via enteric recycling: structural effects and lack of correlations between in vitro and in situ metabolic properties. Drug Metab Dispos 34:1837–1848. [DOI] [PubMed] [Google Scholar]
- Wang SW, Kulkarni KH, Tang L, Wang JR, Yin T, Daidoji T, Yokota H, and Hu M (2009) Disposition of flavonoids via enteric recycling: UDP-glucuronosyltransferase (UGT) 1As deficiency in Gunn rats is compensated by increases in UGT2Bs activities. J Pharmacol Exp Ther 329:1023–1031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wei Y, Wu B, Jiang W, Yin T, Jia X, Basu S, Yang G, and Hu M (2013) Revolving door action of breast cancer resistance protein (BCRP) facilitates or controls the efflux of flavone glucuronides from UGT1A9-overexpressing HeLa cells. Mol Pharm 10:1736–1750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Welter J, Meyer MR, Kavanagh P, and Maurer HH (2014) Studies on the metabolism and the detectability of 4-methyl-amphetamine and its isomers 2-methyl-amphetamine and 3-methyl-amphetamine in rat urine using GC-MS and LC-(high-resolution)-MSn. Anal Bioanal Chem 406:1957–1974. [DOI] [PubMed] [Google Scholar]
- Wen Z, Tallman MN, Ali SY, and Smith PC (2007) UDP-glucuronosyltransferase 1A1 is the principal enzyme responsible for etoposide glucuronidation in human liver and intestinal microsomes: structural characterization of phenolic and alcoholic glucuronides of etoposide and estimation of enzyme kinetics. Drug Metab Dispos 35:371–380. [DOI] [PubMed] [Google Scholar]
- Westley IS, Brogan LR, Morris RG, Evans AM, and Sallustio BC (2006) Role of Mrp2 in the hepatic disposition of mycophenolic acid and its glucuronide metabolites: effect of cyclosporine. Drug Metab Dispos 34:261–266. [DOI] [PubMed] [Google Scholar]
- Williams JA, Hyland R, Jones BC, Smith DA, Hurst S, Goosen TC, Peterkin V, Koup JR, and Ball SE (2004) Drug-drug interactions for UDP-glucuronosyltransferase substrates: a pharmacokinetic explanation for typically observed low exposure (AUCi/AUC) ratios. Drug Metab Dispos 32:1201–1208. [DOI] [PubMed] [Google Scholar]
- Wink CS, Meyer MR, Braun T, Turcant A, and Maurer HH (2015) Biotransformation and detectability of the designer drug 2,5-dimethoxy-4-propylphenethylamine (2C-P) studied in urine by GC-MS, LC-MS(n), and LC-high-resolution-MS(n). Anal Bioanal Chem 407:831–843. [DOI] [PubMed] [Google Scholar]
- Wittgen HG, van den Heuvel JJ, van den Broek PH, Siissalo S, Groothuis GM, de Graaf IA, Koenderink JB, and Russel FG (2012) Transport of the coumarin metabolite 7-hydroxycoumarin glucuronide is mediated via multidrug resistance-associated proteins 3 and 4. Drug Metab Dispos 40:1076–1079. [DOI] [PubMed] [Google Scholar]
- Wolff T, Samuelsson H, and Hedner T (1995) Morphine and morphine metabolite concentrations in cerebrospinal fluid and plasma in cancer pain patients after slow-release oral morphine administration. Pain 62:147–154. [DOI] [PubMed] [Google Scholar]
- Wong CC, Barron D, Orfila C, Dionisi F, Krajcsi P, and Williamson G (2011) Interaction of hydroxycinnamic acids and their conjugates with organic anion transporters and ATP-binding cassette transporters. Mol Nutr Food Res 55:979–988. [DOI] [PubMed] [Google Scholar]
- Wu B (2012) Pharmacokinetic interplay of phase II metabolism and transport: a theoretical study. J Pharm Sci 101:381–393. [DOI] [PubMed] [Google Scholar]
- Wu B, Dong D, Hu M, and Zhang S (2013) Quantitative prediction of glucuronidation in humans using the in vitro- in vivo extrapolation approach. Curr Top Med Chem 13:1343–1352. [DOI] [PubMed] [Google Scholar]
- Wu W, Hu N, Zhang Q, Li Y, Li P, Yan R, and Wang Y (2014) In vitro glucuronidation of five rhubarb anthraquinones by intestinal and liver microsomes from humans and rats. Chem Biol Interact 219:18–27. [DOI] [PubMed] [Google Scholar]
- Wu WN, McKown LA, and Reitz AB (2007) Metabolic fate of the antipsychotic agent, mazapertine, in man--API-MS and MS/MS identification of urinary metabolites. Eur J Drug Metab Pharmacokinet 32:171–176. [DOI] [PubMed] [Google Scholar]
- Xia B, Zhou Q, Zheng Z, Ye L, Hu M, and Liu Z (2012) A novel local recycling mechanism that enhances enteric bioavailability of flavonoids and prolongs their residence time in the gut. Mol Pharm 9:3246–3258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu B, Yang G, Ge S, Yin T, Hu M, and Gao S (2013) Validated LC-MS/MS method for the determination of 3-hydroxflavone and its glucuronide in blood and bioequivalent buffers: application to pharmacokinetic, absorption, and metabolism studies. J Pharm Biomed Anal 85:245–252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu H, Kulkarni KH, Singh R, Yang Z, Wang SW, Tam VH, and Hu M (2009) Disposition of naringenin via glucuronidation pathway is affected by compensating efflux transporters of hydrophilic glucuronides. Mol Pharm 6:1703–1715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yabuuchi H, Tanaka K, Maeda M, Takemura M, Oka M, Ohashi R, and Tamai I (2008) Cloning of the dog bile salt export pump (BSEP; ABCB11) and functional comparison with the human and rat proteins. Biopharm Drug Dispos 29:441–448. [DOI] [PubMed] [Google Scholar]
- Yamamoto T, Ito K, Honma M, Takada T, and Suzuki H (2007) Cholesterol-lowering effect of ezetimibe in uridine diphosphate glucuronosyltransferase 1A-deficient (Gunn) rats. Drug Metab Dispos 35:1455–1458. [DOI] [PubMed] [Google Scholar]
- Yang K, Pfeifer ND, Hardwick RN, Yue W, Stewart PW, and Brouwer KL (2014) An experimental approach to evaluate the impact of impaired transport function on hepatobiliary drug disposition using Mrp2-deficient TR- rat sandwich-cultured hepatocytes in combination with Bcrp knockdown. Mol Pharm 11:766–775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang Z, Zhu W, Gao S, Yin T, Jiang W, and Hu M (2012) Breast cancer resistance protein (ABCG2) determines distribution of genistein phase II metabolites: reevaluation of the roles of ABCG2 in the disposition of genistein. Drug Metab Dispos 40:1883–1893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yeh SL, Lin YC, Lin YL, Li CC, and Chuang CH (2016) Comparing the metabolism of quercetin in rats, mice and gerbils. Eur J Nutr 55:413–422. [DOI] [PubMed] [Google Scholar]
- Yin H, Bennett G, and Jones JP (1994) Mechanistic studies of uridine diphosphate glucuronosyltransferase. Chem Biol Interact 90:47–58. [DOI] [PubMed] [Google Scholar]
- Younis IR, Malone S, Friedman HS, Schaaf LJ, and Petros WP (2009) Enterohepatic recirculation model of irinotecan (CPT-11) and metabolite pharmacokinetics in patients with glioma. Cancer Chemother Pharmacol 63:517–524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yue Q, Chen YH, Mulder T, Deese A, Takahashi R, Rudewicz PJ, Reynolds M, Solon E, Hop CE, Wong H, and Khojasteh SC (2011) Absorption, distribution, metabolism, and excretion of [(1)(4)C]GDC-0449 (vismodegib), an orally active hedgehog pathway inhibitor, in rats and dogs: a unique metabolic pathway via pyridine ring opening. Drug Metab Dispos 39:952–965. [DOI] [PubMed] [Google Scholar]
- Yue Q, Mulder T, Rudewicz PJ, Solon E, Budha N, Ware JA, Lyssikatos J, Hop CE, Wong H, and Khojasteh SC (2013) Evaluation of metabolism and disposition of GDC-0152 in rats using 14C labeling strategy at two different positions: a novel formation of hippuric acid from 4-phenyl-5-amino-1,2,3-thiadiazole. Drug Metab Dispos 41:508–517. [DOI] [PubMed] [Google Scholar]
- Zamek-Gliszczynski MJ, Chu X, Polli JW, Paine MF, and Galetin A (2014) Understanding the transport properties of metabolites: case studies and considerations for drug development. Drug Metab Dispos 42:650–664. [DOI] [PubMed] [Google Scholar]
- Zamek-Gliszczynski MJ, Day JS, Hillgren KM, and Phillips DL (2011) Efflux transport is an important determinant of ethinylestradiol glucuronide and ethinylestradiol sulfate pharmacokinetics. Drug Metab Dispos 39:1794–1800. [DOI] [PubMed] [Google Scholar]
- Zamek-Gliszczynski MJ, Hoffmaster KA, Humphreys JE, Tian X, Nezasa KI, and Brouwer KL (2006a) Differential involvement of Mrp2 (Abcc2) and Bcrp (Abcg2) in biliary excretion of 4-methylumbelliferyl glucuronide and sulfate in the rat. J Pharmacol Exp Ther 319:459–467. [DOI] [PubMed] [Google Scholar]
- Zamek-Gliszczynski MJ, Hoffmaster KA, Nezasa K, and Brouwer KL (2008) Apparent differences in mechanisms of harmol sulfate biliary excretion in mice and rats. Drug Metab Dispos 36:2156–2158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zamek-Gliszczynski MJ, Hoffmaster KA, Nezasa K, Tallman MN, and Brouwer KL (2006b) Integration of hepatic drug transporters and phase II metabolizing enzymes: mechanisms of hepatic excretion of sulfate, glucuronide, and glutathione metabolites. Eur J Pharm Sci 27:447–486. [DOI] [PubMed] [Google Scholar]
- Zamek-Gliszczynski MJ, Nezasa K, Tian X, Kalvass JC, Patel NJ, Raub TJ, and Brouwer KL (2006c) The important role of Bcrp (Abcg2) in the biliary excretion of sulfate and glucuronide metabolites of acetaminophen, 4-methylumbelliferone, and harmol in mice. Mol Pharmacol 70:2127–2133. [DOI] [PubMed] [Google Scholar]
- Zelcer N, Saeki T, Reid G, Beijnen JH, and Borst P (2001) Characterization of drug transport by the human multidrug resistance protein 3 (ABCC3). J Biol Chem 276:46400–46407. [DOI] [PubMed] [Google Scholar]
- Zelcer N, van de Wetering K, Hillebrand M, Sarton E, Kuil A, Wielinga PR, Tephly T, Dahan A, Beijnen JH, and Borst P (2005) Mice lacking multidrug resistance protein 3 show altered morphine pharmacokinetics and morphine-6-glucuronide antinociception. Proc Natl Acad Sci U S A 102:7274–7279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zeng M, Sun R, Basu S, Ma Y, Ge S, Yin T, Gao S, Zhang J, and Hu M (2016) Disposition of flavonoids via recycling: Direct biliary excretion of enterically or extrahepatically derived flavonoid glucuronides. Mol Nutr Food Res 60:1006–1019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang L, Lin G, Kovacs B, Jani M, Krajcsi P, and Zuo Z (2007) Mechanistic study on the intestinal absorption and disposition of baicalein. Eur J Pharm Sci 31:221–231. [DOI] [PubMed] [Google Scholar]
- Zhang X, Dong D, Wang H, Ma Z, Wang Y, and Wu B (2015) Stable knock-down of efflux transporters leads to reduced glucuronidation in UGT1A1-overexpressing HeLa cells: the evidence for glucuronidation-transport interplay. Mol Pharm 12:1268–1278. [DOI] [PubMed] [Google Scholar]
- Zhao M, Ding W, Wang S, Wang C, Du Y, Xu H, Wang Q, and Jin S (2016) Simultaneous determination of nine coumarins in rat plasma by HPLC-MS/MS for pharmacokinetics studies following oral administration of Fraxini Cortex extract. J Chromatogr B Analyt Technol Biomed Life Sci 1025:25–32. [DOI] [PubMed] [Google Scholar]
- Zhao X, Yang DH, Zhou QL, Xu F, Zhang L, Liang J, Liu GX, Cai SQ, and Yang XW (2013) Identification of metabolites in WZS-miniature pig urine after oral administration of Danshen decoction by HPLC coupled with diode array detection with electrospray ionization tandem ion trap and time-of-flight mass spectrometry. Biomed Chromatogr 27:720–735. [DOI] [PubMed] [Google Scholar]
- Zhong K, Li X, Xie C, Zhang Y, Zhong D, and Chen X (2014) Effects of renal impairment on the pharmacokinetics of morinidazole: uptake transporter-mediated renal clearance of the conjugated metabolites. Antimicrob Agents Chemother 58:4153–4161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou Q, Zheng Z, Xia B, Tang L, Lv C, Liu W, Liu Z, and Hu M (2010) Use of isoform-specific UGT metabolism to determine and describe rates and profiles of glucuronidation of wogonin and oroxylin A by human liver and intestinal microsomes. Pharm Res 27:1568–1583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou X, Li L, Deng P, Chen X, and Zhong D (2013) Characterization of metabolites of GLS4 in humans using ultrahigh-performance liquid chromatography/quadrupole time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 27:2483–2492. [DOI] [PubMed] [Google Scholar]
- Zhu W, Xu H, Wang SW, and Hu M (2010) Breast cancer resistance protein (BCRP) and sulfotransferases contribute significantly to the disposition of genistein in mouse intestine. Aaps J 12:525–536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu Y, Li L, Deng P, Chen X, and Zhong D (2016) Characterization of TPN729 metabolites in humans using ultra-performance liquid chromatography/quadrupole time-of-flight mass spectrometry. J Pharm Biomed Anal 117:217–226. [DOI] [PubMed] [Google Scholar]