Skip to main content
  • Review
  • Published:

Metabolic inactivation of estrogens in breast tissue by UDP-glucuronosyltransferase enzymes: an overview

Abstract

The breast tissue is the site of major metabolic conversions of estradiol (E2) mediated by specific cytochromes P450 hydroxylations and methylation by catechol-O-methytransferase. In addition to E2 itself, recent findings highlight the significance of 4-hydroxylated estrogen metabolites as chemical mediators and their link to breast cancer development and progression, whereas, in opposition, 2-methoxylated estrogens appear to be protective. Recent data also indicate that breast tissue possesses enzymatic machinery to inactivate and eliminate E2 and its oxidized and methoxylated metabolites through conjugation catalyzed by UDP-glucuronosyltransferases (UGTs), which involves the covalent addition of glucuronic acid. In opposition to other metabolic pathways of estrogen, the UGT-mediated process leads to the formation of glucuronides that are devoid of biologic activity and are readily excreted from the tissue into the circulation. This review addresses the most recent findings on the identification of UGT enzymes that are responsible for the glucuronidation of E2 and its metabolites, and evidence regarding their potential role in breast cancer.

Introduction

Estrogens are essential for development of the reproductive system in women, in whom they exert beneficial effects in a large number of tissues, including breast, bone, brain, and cardiovascular system. In contrast, the proliferation and genetic instability induced by estrogens in breast and uterus have been considered to increase further the likelihood that normal cells will transform into a malignant type. Over the past 30 years a large number of case–control and cohort studies have been conducted that examined circulating levels of estrogens and/or their urinary excretion to detect any differences in estrogen concentrations that may contribute to cancer. Based on their findings it has been suggested that increased estrogen exposure for lengthy periods of time may induce breast cancer [1–3].

Several observations have also associated in situ production of hydroxylated estrogen metabolites, namely catecholestrogens, with the development of estrogen-sensitive cancers. Oxidative reactions are also important because 2- and 4-hydroxylated metabolites possess distinctive biologic properties as compared with estradiol (E2), the 4-hydroxylated metabolites being particularly important to the carcinogenic effects of estrogen [4–6]. It is also important to emphasize that these metabolic reactions not only take place in the liver but also in estrogen target tissues such as breast, ovary and uterus, and thus they play a critical role in the local homeostasis of E2 and its metabolites.

In addition to the hydroxylation of estrogen, it has been known for many years that estrogen can be conjugated in breast tissue. Conjugative estrogen metabolism includes sulfonation [7, 8], glucuronidation [9, 10] and O-methylation [11], which are catalyzed by sulfotransferases, UDP-glucuronosyltransferases (UGTs) and catechol-O-methyltransferase (COMT), respectively. Sulfates and glucuronides are the most abundant circulating estrogen conjugates [12]. Several recent observations support a role for UGT enzymes in relation to estrogen metabolism and cancer risk in estrogen target tissues [13–19]. This review addresses the most recent findings regarding the identification of UGT enzymes responsible for the glucuronidation of E2 and its metabolites, and their potential role in breast cancer susceptibility.

Site of production and secretion of estrogens in women during aging

In comparison with men, who exhibit relatively constant blood levels of androgen after puberty, circulating levels of sex steroid hormones in women vary tremendously throughout life. During ovarian cycles the follicles and the corpus luteum are important sources of E2 and estrone (E1), the serum concentrations of which ranging from 50 to 300 pg/ml and from 30 to 250 pg/ml, respectively [20–23]. E1 sulfate (E1S) is also found in the circulation (concentration approximately 1 ng/ml), but it is probably formed by estrogen sulfotransferase, which is present in several tissues (Fig. 1). In adult women the term 'circulating estrogen' is generally used to refer to E2, E1, and E1S; the latter two represent a form of estrogen storage and are precursors of E2 [20, 22]. In addition to the direct secretion of estrogens by the ovaries into the bloodstream, a second source of estrogen in adult women is through adrenal steroids such as dehydroepiandrosterone, its sulfate, and androstenedione (referred as C19 steroids); these are converted to estrogen by the aromatase enzyme (CYP19), which is expressed in several extra-ovarian tissues, particularly adipose tissue [22, 24, 25].

Figure 1
figure 1

Estrogen biosynthesis and metabolism in breast tissue. In breast tissue, estrogens come directly from ovaries or they are synthesized from androgens secreted by the adrenals. (1) The first metabolic pathway of estrogen involves the transformation of estradiol (E2) to estrone (E1) by 17β-hydroxysteroid dehydrogenase (17βHSD) and its subsequent conjugation to estrone sulfate (E1S) by estrogen sulfotransferase. (2) Both E2 and E1 can be oxidized by the action of different cytochromes P450 (CYPs) to generate 2- and 4-hydroxy-catecholestrogen (OHCE). (3) These metabolites can be further methoxylated by catechol-O-methyltransferase (COMT). (4) UDP-glucuronosyltransferases (UGTs) are able to conjugate parent estrogens E2 and E1, as well as there derivatives 2/4-OHCE and 2/4-methoxy-catecholestrogen (MeOCE). The resulting estrogen glucuronides are devoid of biologic activities. AR, androgen receptor; 3β-HSD, 3β-hydroxysteroid dehydrogenase; CYP19, aromatase; Δ5-diol, androstenediol; DHT, dihydrotestosterone; ER, estrogen receptor; MeO, methoxy; OH, hydroxy; testo, testosterone.

At menopause, however, production of estrogen by the ovaries is abolished. Despite the marked decline in adrenal C19 steroid production observed in humans after age 30 years, these steroid precursors become the only source of estrogen as substrates of CYP19 (Fig. 1) [25–27]. Thus, the profile in the circulation is markedly modified in postmenopausal women as compared with premenopausal women; serum concentrations of E2 are relatively low (about 2–10 pg/ml), whereas E1 (about 10–30 pg/ml) and E1S (about 150 pg/ml) exceed E2 concentrations [28]. By contrast, the concentrations of E2 in estrogen sensitive tissues are not significantly decreased, despite the low serum E2 levels in menopausal women [29, 30]. In addition, several observations indicate that, in the absence of ovarian production of estrogen, the low concentrations of E2 in the circulation are not representative of E2 content in tissue [23, 29–33]. Indeed, only a small fraction of E2 would be released from the various tissues because of further local conversion of E2 into E1 and E1S, in addition to a series of polar metabolites (Fig. 1). Thus, despite marked changes in blood estrogen levels, the tissue content of estrogens and polar metabolites appears to remain relatively constant during aging. That a significant proportion of breast cancers occur in postmenopausal women, with relatively low circulating levels of E2 and its precursors E1 and E1S, further supports this concept.

It has been hypothesized that homeostasis of tissue concentrations of E2 and active metabolites may be influenced significantly by the balance between the activity of local estrogen producing enzymes and those that involve conjugation and the activity of inactivation enzymes. In the following section we address the various metabolic transformations of E2 that take place in the breast tissue and that lead to production of estrogen metabolites that are substrates of UGT-inactivating enzymes.

Estrogen biosynthesis and metabolism in breast tissue

There are essentially four major pathways of E2 metabolism in estrogen-sensitive tissues (Fig. 1). The first involves transformation of E2 to E1 and its subsequent conjugation to E1S by 17β-hydroxysteroid dehydrogenase and estrogen sulfotransferase, respectively. This pathway is considered to enhance further the half-life of E2 because these enzymes and those responsible for the back transformation of E1S to E1 to E2 are present in several tissues of the body, including the breast [34]. In contrast, the other three pathways, which are active within target cells, generate estrogen metabolites with distinctive biologic properties as compared with E2, with only one metabolic process leading to complete inactivation of estrogen (i.e. glucuronidation catalyzed by UGT enzymes; Fig. 1).

The second pathway employs several cytochrome P450 (CYP) enzymes that use E2 and E1 as substrates for extensive oxidative metabolism at various positions in the estrogen molecule [35]. Recently, the specificity of 15 human CYPs to estrogen was thoroughly characterized by Lee and coworkers [36], and they concluded that the major oxidative products formed from E2 by CYPs are the 2-hydroxy-catecholestrogens (OHCEs) and 4-OHCEs (Fig. 1). As indicated above, these catechol estrogens possess distinct biologic properties as compared with E2. For instance, several groups have postulated that the depurinating adducts formed from 4-OHCEs may generate mutations that initiate cancer, whereas 2-OHCE metabolites may protect against cancer [4, 6, 35, 37–40].

The formation of 2-OHCEs and 4-OHCEs in breast tissue was recently studied by Rogan and coworkers [41]. Those investigators quantified the total pool of conjugated and nonconjugated E1, E2, their hydroxylated metabolites at positions 2 and 4, and methylated derivatives of catecholestrogens in breast tissue samples, after treatment of tissue homogenates with glucuronidase/sulfatase to release the corresponding unconjugated steroids. In tissues obtained from healthy women, they found that levels of the 2- and 4-hydroxy and/or methoxy metabolites were within the same ranges as the parent estrogens E1 and E2 (about 1–2 ng/g tissue). In tumor tissues a significant increase in 4-OHCE concentrations was noted, which is in agreement with several previous reports that indicated changes in estrogen metabolism in cancer tissue that favour the 4-hydroxy pathway and formation of the genotoxic catechol estrogen quinones [4, 42]. As a subsequent step to production of 2-OHCEs and 4-OHCEs in mammary tissue, COMT adds a methyl group to 2-OHCEs and, to a lesser degree, 4-OHCEs (Fig. 1) [35, 43, 44]. O-methylation of 2-OHCE into 2-methoxy-catecholestrogen results in the formation of a protective metabolite with very potent inhibitory action on cell proliferation, tubulin activity, and angiogenesis [45–47].

Overall, these data are in agreement with the concept that E1 and E2 are converted by CYP and COMT in estrogen sensitive tissues before their release into the circulation; this would account for the difficulties in relating serum E2 concentrations to exposure to estrogen in target tissues, particularly in postmenopausal women. It is also clear that the hydroxylated estrogens are subsequently conjugated locally by UGT enzymes, as suggested by previous observations [9, 41]. In the following section we address the extensive knowledge on UGT enzymes and recent findings that support a role for these enzymes in estrogen inactivation in the breast tissue.

The glucuronidation process leads to biologically inactive estrogens

Conjugation by glucuronidation, which is catalyzed by UGT enzymes, is a pathway found in all vertebrates and has primarily been studied because of its role in the detoxification of exogenous compounds, mostly drugs [48–51]. UGTs are membrane-bound enzymes that are present in the endoplasmic reticulum, which mediate transfer of the ubiquitous co-substrate glucuronic acid group of uridine diphospho-glucuronic acid to the functional group (e.g. hydroxyl, carboxyl, amino, sulphur) of a specific substrate. This biochemical reaction increases the polarity of the target compound, and the resulting glucuronide product is generally water soluble, less toxic, and more easily excreted from the body than is the parent compound. The glucuronide is subsequently recognized by the biliary and renal organic anion transport systems, which enable secretion into urine and bile. Because addition of glucuronic acid to compounds changes their structure, glucuronidation modifies the biological activity of the parent molecule and therefore prevents it binding to receptors. As a result, it is believed that most glucuronides, including estrogen glucuronides, correspond to inactive end-products of the parent estrogen and are devoid of biologic activity.

Human UDP-glucuronosyltransferase enzymes

Based on homology of primary structures, the UGT proteins have been categorized into two families, UGT1 and UGT2, in which the latter is further subdivided into two subfamilies, UGT2A and UGT2B. In humans, there are sixteen functional UGT proteins, of which nine are of the UGT1 family (UGT1A1, A3–A10) and seven are of the UGT2 family (UGT2B4, B7, B10, B11, B15, B17 and B28) [48–50, 52].

In contrast to the UGT2B subfamily, which comprises several independent genes that are located on chromosome 4q13 [53–55], members of the UGT1 family are derived from a single gene locus (UGT1) that spans about 210 kilobases on chromosome 2 (2q37) and is composed of 17 exons [52, 56, 57]. To synthesize the final protein, only one of 13 different exon-1 sequences on the locus is associated with four downstream exons, which are common to all UGT1 isoforms. Of the 13 exon-1 sequences, nine encode functional proteins (UGT1A1, UGT1A3–1A10) and four correspond to pseudogenes (p; UGT1A2p, UGT1A11p, UGT1A12p and UGT1A13p) [52, 56, 57]. The regulatory sequences flanking each of the exon 1 regions are thought to dictate the individual expression profile of the UGT1 isoforms [52, 58].

Two members of the UGT2A subfamily localized to chromosome 4q13 have also been characterized and share approximately 70% identity with the UGT2Bs. The biologic function of UGT2As was proposed to be the termination of odorant signals, although it is not limited to this function because their transcripts have been detected in the liver and several extrahepatic tissues [49, 59, 60].

UGT1 and UGT2 proteins are composed of 527–530 amino acid residues, for a molecular weight of 50–57 kDa. The exon-1 sequence of UGTs encodes the substrate-binding domain (amino-terminal half of the protein), whereas the four common exons of the UGT1 gene and exons 2–6 of the UGT2B genes encode the cosubstrate-binding domain (carboxyl-terminal half of the protein). The presence of different possible substrate-binding domains confers the great substrate specificity and selectivity of UGT proteins, together with their broad tissue expression profiles. UGTs are localized in the liver and in all ports of entry of chemicals, including the epithelial surfaces of the nasal mucosa, the gut, skin, white blood cells and lung [58], and they probably play a pivotal role in eliminating synthetic substances and pollutants.

Although it was believed for many years that glucuronidation is a process limited to elimination of exogenous compounds and synthetic drugs, recent data indicate that these enzymes represent key elements in the homeostasis of a number of endobiotics, including steroid and thyroid hormones, bilirubin, fatty acids, and biliary acids [10, 61–64]. For instance, in a steroid target tissue such as the prostate, high concentrations of glucuronides from dihydrotestosterone metabolites have been measured, and expression of highly specific androgen-conjugating UGT enzymes UGT2B15 and UGT2B17 has been detected [65]. It is believed that these polar metabolites represent the final step of androgen metabolism in this tissue.

Glucuronidation of estrogens by UDP-glucuronosyltransferases expressed in the breast tissue

Knowledge on the substrate specificity of human UGT enzymes for estrogens has progressed significantly in recent years. Data indicate that members of the UGT1 family have preferential recognition for estrogens, whereas only one member of the UGT2B subfamily, namely UGT2B7, has been found to be active on these molecules [66–71]. The bilirubin-conjugating enzyme UGT1A1 was initially reported to possess very high specificity for E2 and to be responsible for formation of E2-3-glucuronide (E2-3G) in human liver [71, 72]. Until recently little information was available regarding the conjugation of catecholestrogens and their methoxylated metabolites.

Glucuronidation of catecholestrogens may occur at three sites of hydroxylation, namely 3, 17, and 2 or 4 (Fig. 2), and localization of the glucuronide group on the steroid molecule was only possible using high performance liquid chromatography mass spectrometry comparison with authentic standards [66]. Six UGTs – 1A1, 1A3, 1A8, 1A9, 1A10, and 2B7 – were found to conjugate E2 and E1, their hydroxyls and their methoxy derivatives. The regioselectivity of the glucuronidation reaction has been demonstrated for E2, which is conjugated at positions 3 and 17 by at least two different UGTs, namely UGT1A1 and UGT2B7, respectively [67, 73, 74]. In the case of catecholestrogens, glucuronidation occurs predominantly at the 3-hydroxyl position of 2-OHCEs and the 4-hydroxyl position of 4-OHCEs and is performed by a limited number of UGT enzymes (Fig. 2) [66]. These reactions are mostly mediated by UGT1A1 and UGT1A8, which have a preference for 2-hydroxy E1 and 2-hydroxy E2, whereas UGT2B7 has greater specificity for 4-hydroxy E1 and 4-hydroxy E2 (Fig. 2) [13, 68, 75, 76].

Figure 2
figure 2

Regioselectivity of human UDP-glucuronosyltransferases (UGTs) for parent estrogens estrone (E1) and estradiol (E2), their catechol estrogens metabolites 2-hydroxy (OH)E1/E2 and 4-OHE1/E2, and their methoxy metabolites 2-methoxy (MeO)E1/E2 and 4-MeOE1/E2. Red is used for E2 and its metabolites, whereas blue refers to E1 and its metabolites.

These data were obtained using UGT proteins that are over-expressed in eukaryotic cells, but little is known regarding the actual level of their formation in breast tissue. On the other hand, the presence of high levels of estrogen glucuronides in breast cyst fluid strongly suggests that estrogen glucuronides are formed in situ. This observation was recently confirmed in studies that demonstrated expression of several UGTs in estrogen-sensitive tissues, including breast tissue [13, 14, 16, 66, 77]. In addition, the capacity of the breast tissue to form estrogen glucuronides in vitro was demonstrated by enzymatic assays [13]. Accordingly, given the substrate specificity of UGTs for E2 and its various metabolites, and their presence in the breast, it is believed that this pathway may contribute to in situ metabolic transformation of estrogen and participate in the maintenance of their homeostasis [13–16, 78].

Recent data on the potential role played by UDP-glucuronosyltransferases in breast cancer risk

UGT enzymes that metabolize estrogen to inactive compounds are highly polymorphic in humans (for review [50]). The presence of genetic variations associated with altered enzymatic activity or expression of this metabolic pathway may result in significant changes in breast estrogen levels and subsequent modification of cancer risk. To date, few investigations have tested this hypothesis. Studies of the associations between genetic polymorphisms, hormone circulating levels, and breast cancer risk may yield important insights into the physiologic roles played by these enzymes.

Four population-based studies of breast cancer patients have assessed the association between genetic variability in the E2 conjugating UGT1A1 enzyme and risk for breast cancer [14, 15, 17, 19]. UGT1A1 is expressed in human breast parenchyma and involved in the formation of the major glucuronide of E2, E2-3G [14, 73]. UGT1A1 status is genetically determined by the presence of a common polymorphism in its promoter region. This polymorphism is characterized by a variation in the number of TA repeats in the TATA box region of the gene. Six TA repeats characterizes the common allele (UGT1A1*1) whereas the most common variant allele consists of seven TA repeats in the A(TA)nTAA motif (UGT1A1*28). Two less frequent alleles, A(TA)5TAA and A(TA)8TAA, are referred as UGT1A1*36 and UGT1A1*37 [79]. In in vitro investigations it has been demonstrated that the presence of seven and eight repeats leads a decrease in UGT1A1 gene expression [14, 80, 81]. Based on these findings, the four UGT1A1 promoter alleles were divided into two phenotypic classes [15, 80–83]. The first category of alleles are alleles with high transcriptional activity (UGT1A1*1 and *36), and their presence results in high levels of UGT1A1 protein. The second group of alleles – the low transcriptional activity alleles (UGT1A1*28 and *37) – lead to lower levels of expressed UGT1A1 protein.

In epidemiologic studies it was hypothesized that the presence of lower levels of UGT1A1 protein (individuals with UGT1A1*28 and *37) would result in decreased production of estrogen glucuronides and consequently potential higher exposure to E2 as compared with patients who carry the UGT1A1*1 and/or *36 alleles. This hypothesis was first tested in an analysis of 200 African-American women with invasive breast cancer and 200 matched control individuals [14], which revealed that the low activity alleles (UGT1A1*28 [A(TA)7TAA] and UGT1A1*37 [A(TA)8TAA]) were slightly positively associated with breast cancer (odds ratio [OR] 1.8, 95% confidence interval [CI] 1.0–3.1; P = 0.06). Among premenopausal women the association was stronger for estrogen receptor (ER)-negative breast cancer (OR 2.1, 95% CI 1.0–4.2; P = 0.04) than for ER-positive breast cancer (OR 1.3, 95% CI 0.6–3.0; P = 0.5). A second study [19] suggested that the risk for ER-negative tumors is increased in the presence of polymorphism in the UGT1A1 gene. This second, recent study was conducted in 163 (mostly postmenopausal) women, diagnosed with stage I breast cancer and enrolled in the HEAL (Health, Eating, Activity and Lifestyle) study [19]. Contrary to the reported association of UGT1A1 genotype with an elevated risk for breast cancer in premenopausal African-American women, this second study found a reduced risk for ER-negative breast tumor (OR 0.0, 95% CI 0.0–0.5; P for trend = 0.03) in breast cancer women with the UGT1A1 variant *28/*28 genotype as compared with UGT1A1*1/*1.

The mechanism by which estrogen leads to the development of ER-negative tumors is not well understood, but it is clear that hormones are essential to the genesis of both hormone dependent and independent mammary tumors [84–86]. Hormones can trigger the development of ER-negative tumors via hormone-induced stimulation and secretion of growth factors produced by hormone responsive ER-positive cells or other mammary cell types. Furthermore, ER-negative tumors may arise following prolonged exposure and inadequate elimination of catechol estrogen reactive metabolites formed locally in the mammary gland.

The relationship between UGT1A1 variant alleles and risk for breast cancer was not observed in a larger study of 455 Caucasian women with breast cancer and 603 women without breast cancer within the Nurses' Health Study [15]. A fourth study investigated 1047 breast cancer cases and 1083 community-based control individuals as part of the Shanghai Breast Cancer Study [17] – a population-based case–control study. Consistent with the increased risk observed among premenopausal African-American women [14], a higher risk for breast cancer was evident only in those Chinese women who were younger than 40 years (OR 1.7, 95% CI 1.0–2.7), and not among women who were 40 years old or older (OR 0.8, 95% CI 0.7–1.1) [17].

Analysis of the relationship of this UGT1A1 polymorphism and levels of circulating estrogens revealed that healthy postmenopausal women who were not using hormone replacement therapy tended to have higher E2 and E1 levels associated with the low activity allele [15]. Consistent with these findings, Sparks and coworkers [19] recently showed that breast cancer patients with the UGT1A1*28 homozygous variant genotype tended to have increased concentrations of E2 as compared with cancer patients with the UGT1A1*1/*1 genotype. However, in that study of breast cancer patients, the majority of women were overweight and on tamoxifen therapy, which may alter estrogen levels independent of UGT1A1 genotype.

These findings support a role of the glucuronidation pathway in estrogen metabolism and its potential contribution to the regulation of the estrogenic environment in target cells. Additional studies are needed to assess clearly the relationship between UGT genotypes and hormonal status in healthy women and women with breast cancer. In support of the potential role of UGTs in modulating exposure of breast cells to hormones, a recent report suggests that inter-individual differences in estrogen glucuronidation influence breast density. Compared with women with the UGT1A1*1/*1 genotype, premenopausal women with the UGT1A1*28 allele had a lower breast density (-43.1% difference; P = 0.04) whereas postmenopausal women presented with greater breast density (+32.0% difference; P = 0.05) [78]. However, the effect of aging on the relationship between UGT1A1 genotype and breast density remains unclear. Based on the direction of the changes in breast density observed among women with different UGT1A1 genotypes, we would expect premenopausal women with the UGT1A1*28 allele to have decreased risk for breast cancer, and postmenopausal women with the UGT1A1*28 allele to have increased risk. However, as stated above, the UGT1A1 genotype did not alter risk for breast cancer in the large, nested, case–control study from the Nurses' Health Study [15], in which the Caucasian women studied in relation to breast density were a subset. It seems that the effect on breast density may be insufficient to modify breast cancer risk via the UGT1A1-mediated pathway.

These preliminary findings raise a number of questions. It is not known whether this polymorphism in the UGT1A1 gene has a direct effect on altering estrogen glucuronidation, and indeed whether it would affect solely E2 glucuronidation or the conjugation of its hydroxylated and methoxylated metabolites, as predicted by the in vitro metabolic profile for estrogens [66]. It is likely that polymorphisms in other UGTs that are active toward various estrogens may also play a role. In addition, inter-individual differences in estrogen glucuronidation due to other environmental and dietary factors may also influence breast density or breast cancer risk. In fact, it is still largely unknown how breast tissues metabolize via the glucuronidation pathway under physiologic and pathologic conditions. The available data concerning the difference in expression of UGT in normal versus cancer cells suggest that this metabolic pathway is downregulated during breast carcinogenesis, and as a consequence this would allow greater exposure to estrogen. More specifically, expression of UGT2B7, a 4-OHCE metabolizing UGT [66, 87], was shown to be decreased in cancerous mammary gland tissue as compared with normal tissue, and was over-expressed in in situ lesions [13]. Furthermore, the presence of the UGT2B7 protein was shown to be highly variable between individuals in the epithelium lining the mammary gland ductal system. A previous study reported that glucuronidation activity was lower in breast cancer specimens as compared with normal tissues [88]. Because UGT2B7 is highly reactive UGT for 4-OHCEs, it was suggested that UGT2B7 would protect the mammary gland from genotoxic 4-OHCEs (bearing in mind that mammary gland is a tissue that also expresses CYP1B1, which is involved in the local formation of 4-OHCE) [89, 90]. In support of this hypothesis, Gestl and coworkers [13] measured glucuronidation rates of 4-hydroxy E1, and activities were significantly lower in neoplastic tissues than in normal tissues, which is consistent with the hypothesis of a locally protective role of the UGT2B7 enzyme. To date, no expression data are available for other UGTs in cancer versus normal breast tissues, and it remains to be clearly demonstrated whether alterations in glucuronidation rates or expression levels result in increased estrogen bioavailability within target cells and consequent impact on cancer risk. Analytical methods for the analysis of estrogen glucuronides would need to be developed to address these issues carefully.

To conclude, recent findings support possible influences of the UGT1A1 genotype and UGT2B7 expression on metabolism and excretion of estrogen, which might affect the hormonal status of the breast and therefore modulate the risk for developing cancer. However, conflicting findings raised questions, particularly regarding the role played by UGT1A1 genotypes. Overall, the findings have revealed differences in associations of UGT1A1 genotypes with ER-negative breast cancer and breast density by menopausal status, and this needs additional corroboration. Racial differences in susceptibility to breast cancer associated with genetic factors such as polymorphisms in the UGT1A1 promoter were also evident. Larger studies are needed to uncover its exact role as well as the involvement of other estrogen-metabolizing UGTs.

Conclusion

Little is known on the biologic role of UGTs in estrogen target cells, but recent data support a key inactivation role of this biochemical pathway in breast tissue. This function is distinctive of those of other phase I and II metabolizing enzymes such as CYPs and COMT, which lead respectively to the generation of reactive metabolites, some with carcinogenic effects, and estrogen methoxy metabolites that demonstrate protective properties. It is now clear that UGTs confer polarity to a wide range of estrogens and that this reaction not only takes place in the liver but also in estrogen target tissues, producing glucuronides of estrogens that are readily excreted from the tissues and released into the circulation. Nevertheless, the UGT-mediated metabolic pathway remains a largely unexplored area in the field of estrogen metabolism, but recent data point toward the concept that this pathway may participate in the homeostasis of tissue concentrations of E1, E2, catechol estrogens, and methoxy derivatives, in coordination with P450-mediated hydroxylations and COMT-mediated methylation.

Abbreviations

CI:

confidence interval

COMT:

catechol-O-methyltransferase

CYP:

cytochromes P450

E1 :

estrone

E1S:

estrone sulfate

E2 :

estradiol

ER:

estrogen receptor

MeOCE:

methoxy-catecholestrogen

OHCE:

hydroxy-catecholestrogen

OR:

odds ratio

UGT:

UDP-glucuronosyltransferase.

References

  1. Key TJ: Serum oestradiol and breast cancer risk. Endocr Relat Cancer. 1999, 6: 175-180.

    Article  CAS  PubMed  Google Scholar 

  2. Thomas HV, Reeves GK, Key TJ: Endogenous estrogen and postmenopausal breast cancer: a quantitative review. Cancer Causes Control. 1997, 8: 922-928. 10.1023/A:1018476631561.

    Article  CAS  PubMed  Google Scholar 

  3. Endogenous Hormones and Breast Cancer Collaborative Group: Endogenous sex hormones and breast cancer in postmenopausal women: reanalysis of nine prospective studies. J Natl Cancer Inst. 2002, 94: 606-616.

    Article  Google Scholar 

  4. Liehr JG, Ricci MJ: 4-Hydroxylation of estrogens as marker of human mammary tumors. Proc Natl Acad Sci USA. 1996, 93: 3294-3296. 10.1073/pnas.93.8.3294.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Castagnetta LA, Granata OM, Traina A, Ravazzolo B, Amoroso M, Miele M, Bellavia V, Agostara B, Carruba G: Tissue content of hydroxyestrogens in relation to survival of breast cancer patients. Clin Cancer Res. 2002, 8: 3146-3155.

    CAS  PubMed  Google Scholar 

  6. Jefcoate CR, Liehr JG, Santen RJ, Sutter TR, Yager JD, Yue W, Santner SJ, Tekmal R, Demers L, Pauley R, et al: Tissue-specific synthesis and oxidative metabolism of estrogens. J Natl Cancer Inst Monogr. 2000, 27: 95-112.

    Article  CAS  Google Scholar 

  7. Brooks SC, Horn L: Hepatic sulfation of estrogen metabolites. Biochim Biophys Acta. 1971, 231: 233-241. 10.1016/0005-2760(71)90272-4.

    Article  CAS  PubMed  Google Scholar 

  8. Hobkirk R: Steroid sulfotransferases and steroid sulfate sulfatases: characteristics and biological roles. Can J Biochem Cell Biol. 1985, 63: 1127-1144.

    Article  CAS  PubMed  Google Scholar 

  9. Longcope C, Gorbach S, Goldin B, Woods M, Dwyer J, Warram J: The metabolism of estradiol; oral compared to intravenous administration. J Steroid Biochem. 1985, 23: 1065-1070. 10.1016/0022-4731(85)90068-8.

    Article  CAS  PubMed  Google Scholar 

  10. Belanger A, Hum DW, Beaulieu M, Levesque E, Guillemette C, Tchernof A, Belanger G, Turgeon D, Dubois S: Characterization and regulation of UDP-glucuronosyltransferases in steroid target tissues. J Steroid Biochem Mol Biol. 1998, 65: 301-310. 10.1016/S0960-0760(97)00183-0.

    Article  CAS  PubMed  Google Scholar 

  11. Guldberg HC, Marsden CA: Catechol-O-methyl transferase: pharmacological aspects and physiological role. Pharmacol Rev. 1975, 27: 135-206.

    CAS  PubMed  Google Scholar 

  12. Raftogianis R, Creveling C, Weinshilboum R, Weisz J: Estrogen metabolism by conjugation. J Natl Cancer Inst Monogr. 2000, 27: 113-124.

    Article  CAS  Google Scholar 

  13. Gestl SA, Green MD, Shearer DA, Frauenhoffer E, Tephly TR, Weisz J: Expression of UGT2B7, a UDP-glucuronosyltransferase implicated in the metabolism of 4-hydroxyestrone and all-trans retinoic acid, in normal human breast parenchyma and in invasive and in situ breast cancers. Am J Pathol. 2002, 160: 1467-1479.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Guillemette C, Millikan RC, Newman B, Housman DE: Genetic polymorphisms in uridine diphospho-glucuronosyltransferase 1A1 and association with breast cancer among African Americans. Cancer Res. 2000, 60: 950-956.

    CAS  PubMed  Google Scholar 

  15. Guillemette C, De Vivo I, Hankinson SE, Haiman CA, Spiegelman D, Housman DE, Hunter DJ: Association of genetic polymorphisms in UGT1A1 with breast cancer and plasma hormone levels. Cancer Epidemiol Biomarkers Prev. 2001, 10: 711-714.

    CAS  PubMed  Google Scholar 

  16. Duguay Y, McGrath M, Lepine J, Gagne JF, Hankinson SE, Colditz GA, Hunter DJ, Plante M, Tetu B, Belanger A, et al: The functional UGT1A1 promoter polymorphism decreases endometrial cancer risk. Cancer Res. 2004, 64: 1202-1207.

    Article  CAS  PubMed  Google Scholar 

  17. Adegoke OJ, Shu XO, Gao YT, Cai Q, Breyer J, Smith J, Zheng W: Genetic polymorphisms in uridine diphospho-glucuronosyltransferase 1A1 (UGT1A1) and risk of breast cancer. Breast Cancer Res Treat. 2004, 85: 239-245. 10.1023/B:BREA.0000025419.26423.b8.

    Article  CAS  PubMed  Google Scholar 

  18. Cecchin E, Russo A, Corona G, Campagnutta E, Martella L, Boiocchi M, Toffoli G: UGT1A1*28 polymorphism in ovarian cancer patients. Oncol Rep. 2004, 12: 457-462.

    CAS  PubMed  Google Scholar 

  19. Sparks R, Ulrich C, Bigler J, Tworoger S, Yasui Y, Rajan K, Porter P, Stanczyk F, Ballard-Barbash R, Yuan X, et al: UDP-glucuronosyltransferase and sulfotransferase polymorphisms, sex hormone concentrations, and tumor receptor status in breast cancer patients. Breast Cancer Res. 2004, 6: R488-R498. 10.1186/bcr818.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Labrie F: Intracrinology. Mol Cell Endocrinol. 1991, 78: C113-C118. 10.1016/0303-7207(91)90116-A.

    Article  CAS  PubMed  Google Scholar 

  21. Labrie F, Belanger A, Luu-The V, Labrie C, Simard J, Cusan L, Gomez JL, Candas B: DHEA and the intracrine formation of androgens and estrogens in peripheral target tissues: its role during aging. Steroids. 1998, 63: 322-328. 10.1016/S0039-128X(98)00007-5.

    Article  CAS  PubMed  Google Scholar 

  22. Labrie F: Extragonadal synthesis of sex steroids: intracrinology. Ann Endocrinol (Paris). 2003, 64: 95-107.

    CAS  Google Scholar 

  23. Szymczak J, Milewicz A, Thijssen JH, Blankenstein MA, Daroszewski J: Concentration of sex steroids in adipose tissue after menopause. Steroids. 1998, 63: 319-321. 10.1016/S0039-128X(98)00019-1.

    Article  CAS  PubMed  Google Scholar 

  24. Simpson ER, Clyne C, Speed C, Rubin G, Bulun S: Tissue-specific estrogen biosynthesis and metabolism. Ann N Y Acad Sci. 2001, 949: 58-67.

    Article  CAS  PubMed  Google Scholar 

  25. Simpson ER: Aromatization of androgens in women: current concepts and findings. Fertil Steril. 2002, 77 (suppl 4): S6-S10. 10.1016/S0015-0282(02)02984-9.

    Article  PubMed  Google Scholar 

  26. Simpson ER: Role of aromatase in sex steroid action. J Mol Endocrinol. 2000, 25: 149-156.

    Article  CAS  PubMed  Google Scholar 

  27. Purohit A, Reed MJ: Regulation of estrogen synthesis in postmenopausal women. Steroids. 2002, 67: 979-983. 10.1016/S0039-128X(02)00046-6.

    Article  CAS  PubMed  Google Scholar 

  28. Vermeulen A: The hormonal activity of the postmenopausal ovary. J Clin Endocrinol Metab. 1976, 42: 247-253.

    Article  CAS  PubMed  Google Scholar 

  29. Cauley JA, Gutai JP, Sandler RB, LaPorte RE, Kuller LH, Sashin D: The relationship of endogenous estrogen to bone density and bone area in normal postmenopausal women. Am J Epidemiol. 1986, 124: 752-761.

    CAS  PubMed  Google Scholar 

  30. Cauley JA, Lucas FL, Kuller LH, Stone K, Browner W, Cummings SR: Elevated serum estradiol and testosterone concentrations are associated with a high risk for breast cancer. Study of Osteoporotic Fractures Research Group. Ann Intern Med. 1999, 130: 270-277.

    Article  CAS  PubMed  Google Scholar 

  31. Thijssen JH, Blankenstein MA: Endogenous oestrogens and androgens in normal and malignant endometrial and mammary tissues. Eur J Cancer Clin Oncol. 1989, 25: 1953-1959. 10.1016/0277-5379(89)90377-5.

    Article  CAS  PubMed  Google Scholar 

  32. Mady EA, Ramadan EE, Ossman AA: Sex steroid hormones in serum and tissue of benign and malignant breast tumor patients. Dis Markers. 2000, 16: 151-157.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Blankenstein MA, van de Ven J, Maitimu-Smeele I, Donker GH, de Jong PC, Daroszewski J, Szymczak J, Milewicz A, Thijssen JH: Intratumoral levels of estrogens in breast cancer. J Steroid Biochem Mol Biol. 1999, 69: 293-297. 10.1016/S0960-0760(99)00048-5.

    Article  CAS  PubMed  Google Scholar 

  34. Chatterton RT, Geiger AS, Gann PH, Khan SA: Formation of estrone and estradiol from estrone sulfate by normal breast parenchymal tissue. J Steroid Biochem Mol Biol. 2003, 86: 159-166. 10.1016/S0960-0760(03)00266-8.

    Article  CAS  PubMed  Google Scholar 

  35. Zhu BT, Conney AH: Functional role of estrogen metabolism in target cells: review and perspectives. Carcinogenesis. 1998, 19: 1-27. 10.1093/carcin/19.1.1.

    Article  PubMed  Google Scholar 

  36. Lee AJ, Cai MX, Thomas PE, Conney AH, Zhu BT: Characterization of the oxidative metabolites of 17beta-estradiol and estrone formed by 15 selectively expressed human cytochrome p450 isoforms. Endocrinology. 2003, 144: 3382-3398. 10.1210/en.2003-0192.

    Article  CAS  PubMed  Google Scholar 

  37. Cavalieri EL, Rogan EG: A unified mechanism in the initiation of cancer. Ann N Y Acad Sci. 2002, 959: 341-354.

    Article  CAS  PubMed  Google Scholar 

  38. Cavalieri E, Frenkel K, Liehr JG, Rogan E, Roy D: Estrogens as endogenous genotoxic agents: DNA adducts and mutations. J Natl Cancer Inst Monogr. 2000, 27: 75-93.

    Article  CAS  Google Scholar 

  39. Cavalieri EL, Kumar S, Todorovic R, Higginbotham S, Badawi AF, Rogan EG: Imbalance of estrogen homeostasis in kidney and liver of hamsters treated with estradiol: implications for estrogen-induced initiation of renal tumors. Chem Res Toxicol. 2001, 14: 1041-1050. 10.1021/tx010042g.

    Article  CAS  PubMed  Google Scholar 

  40. Cavalieri EL, Li KM, Balu N, Saeed M, Devanesan P, Higginbotham S, Zhao J, Gross ML, Rogan EG: Catechol orthoquinones: the electrophilic compounds that form depurinating DNA adducts and could initiate cancer and other diseases. Carcinogenesis. 2002, 23: 1071-1077. 10.1093/carcin/23.6.1071.

    Article  CAS  PubMed  Google Scholar 

  41. Rogan EG, Badawi AF, Devanesan PD, Meza JL, Edney JA, West WW, Higginbotham SM, Cavalieri EL: Relative imbalances in estrogen metabolism and conjugation in breast tissue of women with carcinoma: potential biomarkers of susceptibility to cancer. Carcinogenesis. 2003, 24: 697-702. 10.1093/carcin/bgg004.

    Article  CAS  PubMed  Google Scholar 

  42. Liehr JG: Is estradiol a genotoxic mutagenic carcinogen?. Endocr Rev. 2000, 21: 40-54. 10.1210/er.21.1.40.

    CAS  PubMed  Google Scholar 

  43. Dawling S, Roodi N, Mernaugh RL, Wang X, Parl FF: Catechol-O-methyltransferase (COMT)-mediated metabolism of catechol estrogens: comparison of wild-type and variant COMT isoforms. Cancer Res. 2001, 61: 6716-6722.

    CAS  PubMed  Google Scholar 

  44. Goodman JE, Jensen LT, He P, Yager JD: Characterization of human soluble high and low activity catechol-O-methyltransferase catalyzed catechol estrogen methylation. Pharmacogenetics. 2002, 12: 517-528. 10.1097/00008571-200210000-00003.

    Article  CAS  PubMed  Google Scholar 

  45. D'Amato RJ, Lin CM, Flynn E, Folkman J, Hamel E: 2-Methoxyestradiol, an endogenous mammalian metabolite, inhibits tubulin polymerization by interacting at the colchicine site. Proc Natl Acad Sci USA. 1994, 91: 3964-3968.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Fotsis T, Zhang Y, Pepper MS, Adlercreutz H, Montesano R, Nawroth PP, Schweigerer L: The endogenous oestrogen metabolite 2-methoxyoestradiol inhibits angiogenesis and suppresses tumour growth. Nature. 1994, 368: 237-239. 10.1038/368237a0.

    Article  CAS  PubMed  Google Scholar 

  47. Lakhani NJ, Sarkar MA, Venitz J, Figg WD: 2-Methoxyestradiol, a promising anticancer agent. Pharmacotherapy. 2003, 23: 165-172. 10.1592/phco.23.2.165.32088.

    Article  CAS  PubMed  Google Scholar 

  48. Mackenzie PI, Owens IS, Burchell B, Bock KW, Bairoch A, Belanger A, Fournel-Gigleux S, Green M, Hum DW, Iyanagi T, et al: The UDP glycosyltransferase gene superfamily: recommended nomenclature update based on evolutionary divergence. Pharmacogenetics. 1997, 7: 255-269.

    Article  CAS  PubMed  Google Scholar 

  49. Tukey RH, Strassburg CP: Genetic multiplicity of the human UDP-glucuronosyltransferases and regulation in the gastrointestinal tract. Mol Pharmacol. 2001, 59: 405-414.

    CAS  PubMed  Google Scholar 

  50. Guillemette C: Pharmacogenomics of human UDP-glucuronosyltransferase enzymes. Pharmacogenomics J. 2003, 3: 136-158. 10.1038/sj.tpj.6500171.

    Article  CAS  PubMed  Google Scholar 

  51. Fisher MB, Paine MF, Strelevitz TJ, Wrighton SA: The role of hepatic and extrahepatic UDP-glucuronosyltransferases in human drug metabolism. Drug Metab Rev. 2001, 33: 273-297. 10.1081/DMR-120000653.

    Article  CAS  PubMed  Google Scholar 

  52. Gong QH, Cho JW, Huang T, Potter C, Gholami N, Basu NK, Kubota S, Carvalho S, Pennington MW, Owens IS, et al: Thirteen UDPglucuronosyltransferase genes are encoded at the human UGT1 gene complex locus. Pharmacogenetics. 2001, 11: 357-368. 10.1097/00008571-200106000-00011.

    Article  CAS  PubMed  Google Scholar 

  53. Monaghan G, Clarke DJ, Povey S, See CG, Boxer M, Burchell B: Isolation of a human YAC contig encompassing a cluster of UGT2 genes and its regional localization to chromosome 4q13. Genomics. 1994, 23: 496-499. 10.1006/geno.1994.1531.

    Article  CAS  PubMed  Google Scholar 

  54. Turgeon D, Carrier JS, Levesque E, Beatty BG, Belanger A, Hum DW: Isolation and characterization of the human UGT2B15 gene, localized within a cluster of UGT2B genes and pseudogenes on chromosome 4. J Mol Biol. 2000, 295: 489-504. 10.1006/jmbi.1999.3374.

    Article  CAS  PubMed  Google Scholar 

  55. Beaulieu M, Levesque E, Tchernof A, Beatty BG, Belanger A, Hum DW: Chromosomal localization, structure, and regulation of the UGT2B17 gene, encoding a C19 steroid metabolizing enzyme. DNA Cell Biol. 1997, 16: 1143-1154.

    Article  CAS  PubMed  Google Scholar 

  56. Owens IS, Ritter JK: Gene structure at the human UGT1 locus creates diversity in isozyme structure, substrate specificity, and regulation. Prog Nucleic Acid Res Mol Biol. 1995, 51: 305-338.

    Article  CAS  PubMed  Google Scholar 

  57. Ritter JK, Chen F, Sheen YY, Tran HM, Kimura S, Yeatman MT, Owens IS: A novel complex locus UGT1 encodes human bilirubin, phenol, and other UDP-glucuronosyltransferase isozymes with identical carboxyl termini. J Biol Chem. 1992, 267: 3257-3261.

    CAS  PubMed  Google Scholar 

  58. Tukey RH, Strassburg CP: Human UDP-glucuronosyltransferases: metabolism, expression, and disease. Annu Rev Pharmacol Toxicol. 2000, 40: 581-616. 10.1146/annurev.pharmtox.40.1.581.

    Article  CAS  PubMed  Google Scholar 

  59. Jedlitschky G, Cassidy AJ, Sales M, Pratt N, Burchell B: Cloning and characterization of a novel human olfactory UDP-glucuronosyltransferase. Biochem J. 1999, 340: 837-843. 10.1042/0264-6021:3400837.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Lazard D, Zupko K, Poria Y, Nef P, Lazarovits J, Horn S, Khen M, Lancet D: Odorant signal termination by olfactory UDP glucuronosyl transferase. Nature. 1991, 349: 790-793. 10.1038/349790a0.

    Article  CAS  PubMed  Google Scholar 

  61. Hum DW, Belanger A, Levesque E, Barbier O, Beaulieu M, Albert C, Vallee M, Guillemette C, Tchernof A, Turgeon D, et al: Characterization of UDP-glucuronosyltransferases active on steroid hormones. J Steroid Biochem Mol Biol. 1999, 69: 413-423. 10.1016/S0960-0760(99)00061-8.

    Article  CAS  PubMed  Google Scholar 

  62. Liu J, Liu Y, Barter RA, Klaassen CD: Alteration of thyroid homeostasis by UDP-glucuronosyltransferase inducers in rats: a dose-response study. J Pharmacol Exp Ther. 1995, 273: 977-985.

    CAS  PubMed  Google Scholar 

  63. Dutton GJ: Glucuronidation of Drugs and Other Compounds. 1980, Boca Raton, FL: CRC Press

    Google Scholar 

  64. Jude AR, Little JM, Bull AW, Podgorski I, Radominska-Pandya A: 13-hydroxy- and 13-oxooctadecadienoic acids: novel substrates for human UDP-glucuronosyltransferases. Drug Metab Dispos. 2001, 29: 652-655.

    CAS  PubMed  Google Scholar 

  65. Belanger A, Pelletier G, Labrie F, Barbier O, Chouinard S: Inactivation of androgens by UDP-glucuronosyltransferase enzymes in humans. Trends Endocrinol Metab. 2003, 14: 473-479. 10.1016/j.tem.2003.10.005.

    Article  CAS  PubMed  Google Scholar 

  66. Lepine J, Bernard O, Plante M, Têtu B, Labrie F, Pelletier G, Bélanger A, Guillemette C: Specificity and regioselectivity of the conjugation of estradiol, estrone and their catechol estrogen and methoxyestrogen metabolites by human uridine diphosphoglucuronosyltransferases expressed in endometrium. J Clin Endocrinol Metab. 2004: in press.

    Google Scholar 

  67. Gall WE, Zawada G, Mojarrabi B, Tephly TR, Green MD, Coffman BL, Mackenzie PI, Radominska-Pandya A: Differential glucuronidation of bile acids, androgens and estrogens by human UGT1A3 and 2B7. J Steroid Biochem Mol Biol. 1999, 70: 101-108. 10.1016/S0960-0760(99)00088-6.

    Article  CAS  PubMed  Google Scholar 

  68. Cheng Z, Rios GR, King CD, Coffman BL, Green MD, Mojarrabi B, Mackenzie PI, Tephly TR: Glucuronidation of catechol estrogens by expressed human UDP-glucuronosyltransferases (UGTs) 1A1, 1A3, and 2B7. Toxicol Sci. 1998, 45: 52-57. 10.1006/toxs.1998.2494.

    CAS  PubMed  Google Scholar 

  69. Wells PG, Mackenzie PI, Chowdhury JR, Guillemette C, Gregory PA, Ishii Y, Hansen AJ, Kessler FK, Kim PM, Chowdhury NR, et al: Glucuronidation and the UDP-glucuronosyltransferases in health and disease. Drug Metab Dispos. 2004, 32: 281-290. 10.1124/dmd.32.3.281.

    Article  CAS  PubMed  Google Scholar 

  70. King CD, Green MD, Rios GR, Coffman BL, Owens IS, Bishop WP, Tephly TR: The glucuronidation of exogenous and endogenous compounds by stably expressed rat and human UDP-glucuronosyltransferase 1.1. Arch Biochem Biophys. 1996, 332: 92-100. 10.1006/abbi.1996.0320.

    Article  CAS  PubMed  Google Scholar 

  71. Williams JA, Ring BJ, Cantrell VE, Campanale K, Jones DR, Hall SD, Wrighton SA: Differential modulation of UDP-glucuronosyltransferase 1A1 (UGT1A1)-catalyzed estradiol-3-glucuronidation by the addition of UGT1A1 substrates and other compounds to human liver microsomes. Drug Metab Dispos. 2002, 30: 1266-1273. 10.1124/dmd.30.11.1266.

    Article  CAS  PubMed  Google Scholar 

  72. Soars MG, Ring BJ, Wrighton SA: The effect of incubation conditions on the enzyme kinetics of udp-glucuronosyltransferases. Drug Metab Dispos. 2003, 31: 762-767. 10.1124/dmd.31.6.762.

    Article  CAS  PubMed  Google Scholar 

  73. Senafi SB, Clarke DJ, Burchell B: Investigation of the substrate specificity of a cloned expressed human bilirubin UDP-glucuronosyltransferase: UDP-sugar specificity and involvement in steroid and xenobiotic glucuronidation. Biochem J. 1994, 303: 233-240.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Fisher MB, Vandenbranden M, Findlay K, Burchell B, Thummel KE, Hall SD, Wrighton SA: Tissue distribution and interindividual variation in human UDP-glucuronosyltransferase activity: relationship between UGT1A1 promoter genotype and variability in a liver bank. Pharmacogenetics. 2000, 10: 727-739. 10.1097/00008571-200011000-00007.

    Article  CAS  PubMed  Google Scholar 

  75. Albert C, Vallee M, Beaudry G, Belanger A, Hum DW: The monkey and human uridine diphosphate-glucuronosyltransferase UGT1A9, expressed in steroid target tissues, are estrogen-conjugating enzymes. Endocrinology. 1999, 140: 3292-3302. 10.1210/en.140.7.3292.

    CAS  PubMed  Google Scholar 

  76. Mojarrabi B, Butler R, Mackenzie PI: cDNA cloning and characterization of the human UDP glucuronosyltransferase, UGT1A3. Biochem Biophys Res Commun. 1996, 225: 785-790. 10.1006/bbrc.1996.1251.

    Article  CAS  PubMed  Google Scholar 

  77. Belanger A, Caron S, Labrie F, Naldoni C, Dogliotti L, Angeli A: Levels of eighteen non-conjugated and conjugated steroids in human breast cyst fluid: relationships with cyst type. Eur J Cancer. 1990, 26: 277-281.

    Article  CAS  PubMed  Google Scholar 

  78. Haiman CA, Hankinson SE, De Vivo I, Guillemette C, Ishibe N, Hunter DJ, Byrne C: Polymorphisms in steroid hormone pathway genes and mammographic density. Breast Cancer Res Treat. 2003, 77: 27-36. 10.1023/A:1021112121782.

    Article  CAS  PubMed  Google Scholar 

  79. UDP Glucuronosyltransferase Nomenclature Committee: 1A1 alleles. [http://som.flinders.edu.au/FUSA/ClinPharm/UGT/1A1alleles.html]

  80. Beutler E, Gelbart T, Demina A: Racial variability in the UDP-glucuronosyltransferase 1 (UGT1A1) promoter: a balanced polymorphism for regulation of bilirubin metabolism?. Proc Natl Acad Sci USA. 1998, 95: 8170-8174. 10.1073/pnas.95.14.8170.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Bosma PJ, Chowdhury JR, Bakker C, Gantla S, de Boer A, Oostra BA, Lindhout D, Tytgat GN, Jansen PL, Oude Elferink RP, et al: The genetic basis of the reduced expression of bilirubin UDP-glucuronosyltransferase 1 in Gilbert's syndrome. N Engl J Med. 1995, 333: 1171-1175. 10.1056/NEJM199511023331802.

    Article  CAS  PubMed  Google Scholar 

  82. Monaghan G, Ryan M, Seddon R, Hume R, Burchell B: Genetic variation in bilirubin UPD-glucuronosyltransferase gene promoter and Gilbert's syndrome. Lancet. 1996, 347: 578-581. 10.1016/S0140-6736(96)91273-8.

    Article  CAS  PubMed  Google Scholar 

  83. Iolascon A, Faienza MF, Centra M, Storelli S, Zelante L, Savoia A: (TA)8 allele in the UGT1A1 gene promoter of a Caucasian with Gilbert's syndrome. Haematologica. 1999, 84: 106-109.

    CAS  PubMed  Google Scholar 

  84. Nandi S, Guzman RC, Yang J: Hormones and mammary carcinogenesis in mice, rats, and humans: a unifying hypothesis. Proc Natl Acad Sci USA. 1995, 92: 3650-3657.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Lopez-Otin C, Diamandis EP: Breast and prostate cancer: an analysis of common epidemiological, genetic, and biochemical features. Endocr Rev. 1998, 19: 365-396. 10.1210/er.19.4.365.

    CAS  PubMed  Google Scholar 

  86. Lower EE, Blau R, Gazder P, Stahl DL: The effect of estrogen usage on the subsequent hormone receptor status of primary breast cancer. Breast Cancer Res Treat. 1999, 58: 205-211. 10.1023/A:1006315607241.

    Article  CAS  PubMed  Google Scholar 

  87. Coffman BL, Rios GR, King CD, Tephly TR: Human UGT2B7 catalyzes morphine glucuronidation. Drug Metab Dispos. 1997, 25: 1-4.

    CAS  PubMed  Google Scholar 

  88. Albin N, Massaad L, Toussaint C, Mathieu MC, Morizet J, Parise O, Gouyette A, Chabot GG: Main drug-metabolizing enzyme systems in human breast tumors and peritumoral tissues. Cancer Res. 1993, 53: 3541-3546.

    CAS  PubMed  Google Scholar 

  89. Iscan M, Klaavuniemi T, Coban T, Kapucuoglu N, Pelkonen O, Raunio H: The expression of cytochrome P450 enzymes in human breast tumours and normal breast tissue. Breast Cancer Res Treat. 2001, 70: 47-54. 10.1023/A:1012526406741.

    Article  CAS  PubMed  Google Scholar 

  90. Muskhelishvili L, Thompson PA, Kusewitt DF, Wang C, Kadlubar FF: In situ hybridization and immunohistochemical analysis of cytochrome P450 1B1 expression in human normal tissues. J Histochem Cytochem. 2001, 49: 229-236.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work is supported by Canadian Institutes of Health Research (CIHR; to CG and AB #129805) and the Canada Research Chair Program (CG). CG is chairholder of the Canada Research Chair in Pharmacogenomics. JL is a recipient of a CIHR studentship

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Chantal Guillemette.

Additional information

Competing interests

The author(s) declare that they have no competing interests.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Authors’ original file for figure 2

Rights and permissions

Reprints and permissions

About this article

Cite this article

Guillemette, C., Bélanger, A. & Lépine, J. Metabolic inactivation of estrogens in breast tissue by UDP-glucuronosyltransferase enzymes: an overview. Breast Cancer Res 6, 246 (2004). https://doi.org/10.1186/bcr936

Download citation

  • Published:

  • DOI: https://doi.org/10.1186/bcr936

Keywords