Women are increasingly using botanical dietary supplements (BDS) to reduce menopausal hot flashes. Although licorice (Glycyrrhiza sp.) is one of the frequently used ingredients in BDS, the exact plant species is often not identified. We previously showed that in breast epithelial cells (MCF-10A), Glycyrrhiza glabra (GG) and G. inflata (GI), and their compounds differentially modulated P450 1A1 and P450 1B1 gene expression, which are responsible for estrogen detoxification and genotoxicity, respectively. GG and isoliquiritigenin (LigC) increased CYP1A1, whereas GI and its marker compound, licochalcone A (LicA), decreased CYP1A1 and CYP1B1. The objective of this study was to determine the distribution of the bioactive licorice compounds, the metabolism of LicA, and whether GG, GI, and/or pure LicA modulate NAD(P)H quinone oxidoreductase (NQO1) in an ACI rat model. In addition, the effect of licorice extracts and compounds on biomarkers of estrogen chemoprevention (CYP1A1) as well as carcinogenesis (CYP1B1) was studied. LicA was extensively glucuronidated and formed GSH adducts; however, free LicA as well as LigC were bioavailable in target tissues after oral intake of licorice extracts. GG, GI, and LicA caused induction of NQO1 activity in the liver. In mammary tissue, GI increased CYP1A1 and decreased CYP1B1, whereas GG only increased CYP1A1. LigC may have contributed to the upregulation of CYP1A1 after GG and GI administration. In contrast, LicA was responsible for GI-mediated downregulation of CYP1B1. These studies highlight the polypharmacologic nature of botanicals and the importance of standardization of licorice BDS to specific Glycyrrhiza species and to multiple constituents.

In 2018, breast cancer will account for nearly one-third of new cancer cases in women (1). A decline in hormone therapy (HT) usage was observed after the Women's Health Initiative study in 2002 due to the increased breast cancer risk caused by the estrogen + progestin HT regimen (2). Besides the well-known hormonal estrogen carcinogenesis pathway, breast cancer risk is also influenced by changes in estrogen oxidative metabolism (3). In the breast, P450 1A1 and P450 1B1 catalyze the metabolism of estrogens to 2- and 4-hydroxylated catechols, 2-OHE1/E2 and 4-OHE1/E2, respectively (Fig. 1; ref. 4). The 2-hydroxylated metabolites are strongly associated with reduced breast cancer risk (5) because they inhibit E2-induced proliferation (6) and are converted to nontoxic quinones (Fig. 1). On the other hand, P450 1B1 is linked to carcinogenesis because it is overexpressed in malignant tissues (7). P450 1B1 produces 4-OHE1/E2 which are oxidized by peroxidases/P450s to genotoxic quinones (4-OHE1/E2-Q) that alkylate DNA and generate depurinating adducts (estrogen chemical carcinogenesis, Fig. 1; refs. 4, 8). Hence, upregulation of the 2-hydroxylation (P450 1A1) and downregulation of the 4-hydroxylation pathway (P450 1B1) may significantly inhibit estrogen chemical carcinogenesis in the breast. In addition, NAD(P)H quinone oxidoreductase 1 (NQO1) decreases depurinating estrogen-DNA adducts (4) due to reduction of the reactive 4-OHE1/E2-Q to its catechol. Catechol-O-methyltransferase (COMT) also prevents quinone formation through methylation of estrogen catechols to produce stable metabolites, 2-MeOE1/E2 and 4-MeOE1/E2 (Fig. 1; ref. 9).

Figure 1.

Biological targets and phase II metabolism of chemopreventive licorice compounds. A, The GI-specific compound, LicA, is a Michael acceptor that can covalently modify Keap1 to upregulate the detoxification enzyme, NQO1, in MCF-10A and liver cells (21) as well as in liver tissue. LicA is also an AhR antagonist that can downregulate P450 1A1/1B1-mediated estrogen oxidative metabolism (11). In this animal model (ACI rats), GG and GI increased P450 1A1 gene expression (CYP1A1) in mammary tissue (Fig. 5B). In addition, GI also decreased P450 1B1 gene expression (CYP1B1) in the mammary gland as indicated with arrows. B, LicA is mainly metabolized to various glucuronides by UDP-glucuronosyltransferases (UGT; Supplementary Fig. S2). As a Michael acceptor, LicA also forms GSH conjugates. In the liver and serum, LicA sulfate conjugates catalyzed by sulfotransferases (SULT) were detected as minor metabolites (Supplementary Fig. S2).

Figure 1.

Biological targets and phase II metabolism of chemopreventive licorice compounds. A, The GI-specific compound, LicA, is a Michael acceptor that can covalently modify Keap1 to upregulate the detoxification enzyme, NQO1, in MCF-10A and liver cells (21) as well as in liver tissue. LicA is also an AhR antagonist that can downregulate P450 1A1/1B1-mediated estrogen oxidative metabolism (11). In this animal model (ACI rats), GG and GI increased P450 1A1 gene expression (CYP1A1) in mammary tissue (Fig. 5B). In addition, GI also decreased P450 1B1 gene expression (CYP1B1) in the mammary gland as indicated with arrows. B, LicA is mainly metabolized to various glucuronides by UDP-glucuronosyltransferases (UGT; Supplementary Fig. S2). As a Michael acceptor, LicA also forms GSH conjugates. In the liver and serum, LicA sulfate conjugates catalyzed by sulfotransferases (SULT) were detected as minor metabolites (Supplementary Fig. S2).

Close modal

Due to fear of increased breast cancer risk with HT, many women have turned to botanical dietary supplements (BDS) as a complementary approach therapy for menopausal symptom relief. Female consumers generally perceive BDS as safer modalities because BDS often contain constituents that are reported to be anti-inflammatory and antioxidant, and induce detoxification enzymes (10). However, the effects of most BDS on estrogen oxidative metabolism are unknown. Although licorice belongs to one of the most popular botanicals contained in BDS used for women's health issues, several licorice species (Glycyrrhiza sp., Fabaceae) are used to source these BDS without discrimination (11). Also, clinical evidence for efficacy is generally lacking (10).

Botanically, over 30 species of licorice exist (12, 13). From a pharmacopoeial perspective, three species are currently used in BDS interchangeably: Glycyrrhiza glabra (GG), G. inflata (GI), and G. uralensis. Notably, these three species have distinctive and very different chemical profiles, as already demonstrated in previous studies (Table 1; refs. 12, 14). Cultivated in China, GI is naturally more popular in Asia. The roots from this species contain an abundant species-specific chemopreventive Michael acceptor, licochalcone A (LicA; ref. Fig. 2). GG is the most popular licorice species in the United States and Europe, and its species-specific compound is glabridin (Table 1; Fig. 2; refs. 15, 16). Both licorice species contain isoliquiritigenin (LigC, C for chalcone), also a chemopreventive Michael acceptor, and liquiritigenin (LigF, F for flavanone), a phytoestrogen with estrogen receptor (ER) β preferential properties (refs. 13, 17; Fig. 2). Both LigC and LigF are spontaneously interconvertible isomers (Fig. 2; ref. 18), found mainly as glycosides (e.g., liquiritin and isoliquiritin) in the roots (Table 1). Due to the species-specific compound profile, each licorice species has a unique bioactivity that could lead to differential clinical activity.

Table 1.

Concentration of bioactive compounds in licorice extracts determined by UHPLC-UV

Compounds (% w/w crude extract)
SpeciesGlabridinLicALigCaLigC equivalentsbLigFLigF equivalents
GG 1.34 ± 0.02 – – 3.61 ± 0.07 0.19 ± 0.01 8.55 ± 0.06 
GI – 7.07 ± 0.61 0.10 ± 0.07 2.32 ± 0.04 0.24 ± 0.05 3.67 ± 0.31 
Compounds (% w/w crude extract)
SpeciesGlabridinLicALigCaLigC equivalentsbLigFLigF equivalents
GG 1.34 ± 0.02 – – 3.61 ± 0.07 0.19 ± 0.01 8.55 ± 0.06 
GI – 7.07 ± 0.61 0.10 ± 0.07 2.32 ± 0.04 0.24 ± 0.05 3.67 ± 0.31 

aLigC was below the limit of detection in GG.

bThe term equivalents is used to represent the total amount of aglycone plus glycosides of LigC (i.e., isoliquiritigenin, isoliquiritin, isoliquiritin apioside, and licuraside) or LigF (liquiritigenin, liquiritin, liquiritin apioside, and liquiritigenin-7-O-apiosylglucoside) in each crude extract. The values are expressed as mean ± SD of independent measures.

Figure 2.

Key compounds of GG and GI including LigF and LigC equivalents.

Figure 2.

Key compounds of GG and GI including LigF and LigC equivalents.

Close modal

Previously, we showed that GG and LigC increased estrogen oxidative metabolism (2- and 4-hydroxylation), whereas GI and LicA decreased metabolism in MCF-10A “normal” breast epithelial cells (11). The purpose of the current study was to determine the chemopreventive effects of GI and GG on estrogen oxidative metabolism in the ACI rat model, which is frequently used for in vivo estrogen carcinogenesis studies (19). LicA was administered in parallel to determine its role in GI's bioactivity and its metabolic profile. LicA's distribution in liver and mammary tissues was determined and compared with LigC and LigF. Levels of 2-MeOE1 in serum were quantified by LC-MS/MS, as a biomarker for overall estrogen oxidative metabolism, because rat P450 1B1 predominantly performs estrogen 2-hydroxylation (20). CYP1A1 and CYP1B1 expressions were determined in mammary tissues, and NQO1 activity was measured in liver and mammary tissues. Studies in MCF-10A cells with GI revealed different results between in vitro and in vivo studies and identified potential bioactive compounds. The current experiments carried out in ACI rats provide crucial information to continue studying these licorice species and their effects on estrogen carcinogenesis in long-term studies with ACI rats and eventually estrogen metabolism in women. The data herein explore additional chemopreventive modes to help develop more beneficial and safer standardized licorice dietary supplements for women's health.

Materials, chemicals, and reagents

All chemicals and reagents were purchased from Fisher Scientific or Sigma except for the following: 2-methoxyestrone (MeOE1)-1,4,16,16-d4 was obtained from CDN isotope, E2 and 2-/4-MeOE1/E2 reference compounds were acquired from Steraloids Inc., LigF and LigC were obtained from ChromaDex, and rat serum for initial standardization studies was purchased from BioreclamationIVT.

Plant material, extraction, and characterization

GI was provided as a gift to SNC by Dr. Liang Zhao (Lanzhou Institute of Chemical Physics CAS), and GG was purchased from Mountain Rose Herbs. Raw materials were identified by macroscopic/microscopic analyses and DNA barcoding, as previously described (14). Ground roots of GG and GI were extracted by maceration and percolation at room temperature with a solvent mixture [ethanol (200 USP proof), isopropanol, and water (90:5:5, v/v)] at the ratio of botanical to solvent as 1:15 (g/mL). The extracts were concentrated and freeze dried to yield 12% w/w of the initial ground roots. Extracts were analyzed by UHPLC-UV to quantify major chalcone and flavanone constituents, as previously reported (21, 22). Briefly, a standard curve containing the following 11 reference standards was used for their quantitation in both licorice extracts. The AUC was taken at 360 nm for all chalcones (isoliquiritin, isoliquiritin apioside, licuraside, LigC, and LicA), and at 275 nm for all flavanones (liquiritin, liquiritin apioside, liquiritigenin 7-O-apiosylglucoside, and LigF) and for glabridin. Quantitative results obtained for each LigF glycoside or LigC glycoside were converted by their molecular weight, thereby leading to their concentration as LigF or LigC equivalents, respectively (Table 1).

Preparation and characterization of LicA

The crude LicA sample, enriched from GI (eLicA, purity of LicA, ∼ 50%), was a gift to SNC from Qinghai Lake Medicinal CO., Ltd. A loss-free countercurrent separation was implemented for the purification of LicA from the eLicA, as follows: TLC-based solvent system strategy (23) was performed for screening a proper solvent system. Among screened solvent systems (Supplementary Table S1), LicA was eluted by the organic phase of n-hexane-ethyl acetate-methanol-water (HEMWat, 4:6:5:5, v/v) to Rf = 0.43 on a precoating normal-phase Si TLC plate (MACHEREY-NAGEL). Considering the main principles of the GUESS method (Generally Useful Estimate of Solvent Systems; ref. 24), these data suggested that the corresponding solvent system may elute the target analyte into a high-resolution separation range (the sweet spot, partition coefficient, K value from 0.25 to 4 of a countercurrent separation method; ref. 25). Regarding verification of the selected solvent system, an analytical scale of high-speed countercurrent chromatography (HSCCC; 16 mL, Tauto Biotech) was applied, which achieved a higher purity of LicA than that from the literature reported solvent system on the same instrument (Supplementary Fig. S1; ref. 26). The scaled-up separation is described in the Supplementary Information. The purity of LicA used in this animal study was determined as 95% (w/w) by qHNMR.

Animal experiment

Female August-Copenhagen Irish (ACI) rats were purchased from Harlan Laboratories at 5 weeks of age, acclimated for 1 week, fed a phytoestrogen-free diet (AIN-76A), and randomly divided into five groups with 6 rats each: vehicle, estradiol benzoate (EB, 1 mg/kg/day), LicA (80 mg/kg/day) + EB, GG (2 g/kg/day, gavage) + EB, and GI (2 g/kg/day, containing 141 mg LicA) + EB. At 6 weeks of age, one vehicle was applied subcutaneously (s.c., sesame oil) and one by gavage (50% corn oil with 50% PEG/H2O), EB and LicA were given s.c., and the licorice extracts were administered by gavage for 4 days. The rats were sacrificed by CO2 asphyxiation on day 5. Blood was collected, and serum prepared immediately after collection; mammary tissues, uterus, and liver were collected and snap frozen in liquid nitrogen and stored at −80°C until analysis. The animal protocol complied with the Guide for the Care and Use of Laboratory Animals, and all procedures were approved by UIC's Institutional Animal Care and Use Committee (Protocol No. 16-033).

Preparation of serum and tissue samples for LicA metabolism profile analysis and for LicA quantitation in vivo

LicA was quantified in serum, liver, and mammary gland samples from the LicA + EB and GI + EB treatment groups (Fig. 3C; Supplementary Table S2). After thawing at room temperature, serum (50 μL) was transferred to a 1.5 mL Eppendorf tube and mixed with 10 μL of ACN containing naringenin (500 nmol/L) as the internal standard (IS). Liver and mammary tissues (500–800 mg for liver and 50–100 mg for mammary gland) were weighed and homogenized in 70% aqueous methanol (containing 0.1% formic acid) at 5 mL for liver and 1 mL for mammary tissues. The homogenate (200 μL) was taken and spiked with 20 μL naringenin (500 nmol/L). Ice-cold ACN (600 μL) was added, and the mixture was centrifuged for 15 minutes at 13,000 x g at 4°C for protein precipitation. The supernatant (400 μL) was transferred to a new Eppendorf tube and evaporated to dryness under a stream of nitrogen. The residue was reconstituted in 100 μL of 20% ACN, and 5 μL was injected into LC-MS/MS for analysis. The same rat samples were analyzed for LicA metabolites.

Figure 3.

UHPLC-MS/MS analysis of key licorice compounds. Licorice compounds, LicA, LigC, LigF, and glabridin (GB), were detected by UHPLC-MS/MS in the crude extracts and serum, liver, and mammary gland after administration of (A) GG (2 g/kg/day) and EB and (B) GI (2 g/kg/day) and EB to ACI rats for 4 days. C, Free LicA was quantified by UHPLC-MS/MS in rat serum, liver, and mammary gland after GI and LicA administration.

Figure 3.

UHPLC-MS/MS analysis of key licorice compounds. Licorice compounds, LicA, LigC, LigF, and glabridin (GB), were detected by UHPLC-MS/MS in the crude extracts and serum, liver, and mammary gland after administration of (A) GG (2 g/kg/day) and EB and (B) GI (2 g/kg/day) and EB to ACI rats for 4 days. C, Free LicA was quantified by UHPLC-MS/MS in rat serum, liver, and mammary gland after GI and LicA administration.

Close modal

UHPLC-MS/MS analyses

UHPLC-MS/MS analysis was performed as described previously (27, 28). Briefly, a Shimadzu LCMS-8060 triple quadrupole mass spectrometer equipped with a Shimadzu Nexera UHPLC system was used for analysis. For quantitative analysis, analytes were separated on a Waters Acquity UPLC BEH C18 2.1 × 50 mm column (1.7 μm particle size). Mass-spectrometer parameters were as follows: nebulizing gas flow: 2.5 L/min; heating gas flow: 10 L/min; interface temperature: 300°C; DL temperature: 250°C; heating block temperature: 400°C; drying gas flow: 10 L/min. The data were acquired using selected reaction monitoring (SRM) with positive ion electrospray as follows: naringenin ([M+H]+m/z 273 to 153 and m/z 273 to 147, IS); and LicA ([M+H]+m/z 339 to 121 and m/z 339 to 297).

For determination of LicA metabolites, the positive ion electrospray SRM transitions for each analyte were established as m/z 515 to 339 for LicA monoglucuronides, m/z 646 to 339 for the glutathione conjugate of LicA, m/z 419 to 339 for LicA sulfate, m/z 531 to 339 for monooxygenated LicA glucuronides, and m/z 545 to 369 for catechol-O-methylated LicA glucuronides.

For qualitative determination of LigC, LigF, LicA, and glabridin in the GI and GG extracts, serum, and tissues, the licorice extracts (GG and GI) were dissolved in 50% aqueous methanol at 10 μg/mL. Rat serum and tissue samples were prepared as described above. Samples were analyzed using UHPLC-MS/MS with negative ion electrospray and SRM (28) as follows: liquiritin and isoliquiritin, m/z 417 to 255; liquiritin apioside, isoliquiritin apioside, and licuraside, m/z 549 to 255; LigF and LigC, m/z 255 to 119; glabridin, m/z 323 to 201; and glycyrrhetinic acid, m/z 469 to 425.

Analysis of estrogen oxidative metabolism (2-MeOE1) and E1/E2 in serum

LC-MS/MS analysis was performed as previously described with minor modifications (29). Serum samples (150 μL) were incubated at 37°C for 4 hours after adding glucuronidase and sulfatase hydrolysis buffer (300 μL) and the IS, 2-methoxyestrone-d4. After the incubation, sample preparation and analysis were conducted as previously described (29). The SRM transitions were as follows: m/z 534.3 to 171.2, m/z 504.3 to 171.0, and m/z 506.3 to 171.0, for 2-MeOE1, E1, and E2, respectively. Results are expressed as fold change from average amount of analytes of rats treated with EB alone.

Analysis of NQO1 activity in liver tissue and mammary gland

The NQO1 activity in frozen liver and mammary tissue was determined as described previously (30). The NQO1 activity was determined in a clear supernatant solution of the tissue homogenate (5 μg of liver protein and 30 μg of mammary gland protein) as previously described (31). The absorbance was measured at 610 nm, and the results were expressed as fold induction of NQO1 activity relative to the control group.

Quantification of P450 1A1/1B1 mRNA expression via RT-qPCR

Mammary tissues (100 mg) were homogenized in TRIzol reagent. The total RNA was extracted using the RNeasy lipid tissue Kit (Qiagen), and RNA (5 μg) was reverse transcribed with Invitrogen's SuperScript III First-Strand Synthesis System. RT-qPCR was performed using TaqMan rat CYP1A1, CYP1B1, and ACTB primers with FAM/MGB probe (Applied Biosystems) as described previously (29). MCF-10A cells were obtained from the ATCC and authenticated via short tandem repeat profiling (Promega). MCF-10A cells were cultured as described previously (29). For the in vitro CYP1A1/CYP1B1 induction experiments, cells having approximately 15 through 20 passages were plated in 96-well plates and treated with vehicle (DMSO), GG, GI, and licorice compounds for 24 hours. RT-qPCR was performed as previously described (29) using TaqMan 1-Step RT-PCR Master Mix, and CYP1A1 and CYP1B1 primer with FAM-MGB probe, and GAPDH primer with VIC-MGB probe. Data were analyzed with the comparative CT (ΔΔCT) method and expressed as fold induction relative to the vehicle control group.

Statistical analysis

The data were expressed as mean ± SEM from 6 animals per group or ± SEM for three independent experiments in MCF-10A cells. Significance was determined using the Student t test to compare two samples or one-way ANOVA with Dunnett posttest to compare multiple samples with the control (*, P < 0.05).

Measurement of bioactive compounds in Glycyrrhiza species

Extracts were analyzed by UHPLC-UV, and all compounds were expressed as % w/w of each extract (Table 1). Glabridin (Fig. 2) was present at 1.34% of total GG extract, whereas LicA represents one of the major compounds of GI crude extract (7.07%). Glabridin and LicA exist as aglycones, but LigC and LigF occur primarily as glycosylated compounds, which exceeded the corresponding aglycones by 15-fold. LigC glycosides (isoliquiritin, isoliquiritin apioside, and licuraside) and LigF glycosides (liquiritin, liquiritin apioside, and liquiritigenin-7-O-apiosylglucoside; Fig. 2) together with the corresponding aglycones are represented as total LigC and LigF equivalents, as the glycosides are deglycosylated in vivo (Fig. 3A and B). GG extract contained 1.5-fold more LigC equivalents and over 2-fold more LigF equivalents than GI. The most abundant LigF glycoside (>70%) in GG extract was liquiritin apioside (Table 1).

Distribution of LicA, LigF, LigC, and glabridin in serum, liver, and mammary tissue after GG and GI administration

LigC/LigF, LigC/LigF glycosides, glabridin, and LicA were qualitatively determined in crude extracts, serum, liver, and mammary tissues by UHPLC-MS/MS analysis. The concentration of glabridin was low in the serum, and it was not detected in liver and mammary tissues (Fig. 3A). The three other licorice compounds, LicA, LigF, and LigC, were all available in the liver and mammary gland. The most striking observation was that most of the LigC/LigF glycosides were hydrolyzed, which increased LigC/LigF aglycone concentrations in the serum and both tissue samples. LigC and LigF concentrations were similar in mammary tissues, although LigC was notably higher in serum but significantly lower than LigF in liver tissues (Fig. 3A and B). In the GI crude extract, LicA levels were much higher than free LigC; however, because LigC equivalents were hydrolyzed in vivo, the concentration of free LigC exceeded LicA concentrations in serum and mammary tissues (Fig. 3B). After LicA s.c. and GI gavage administration for 4 days, LicA was also quantified by UHPLC-MS/MS in serum, liver tissue, and mammary gland. Interestingly, after both applications, free LicA was available in serum, liver, and mammary gland 24 hours after the last dose (day 5; Fig. 3C). Although LicA is highly glucuronidated in vivo (Fig. 1B; Supplementary Fig. S2), considerable amounts of free LicA were observed in the target tissues after oral administration of the extract (Supplementary Table S2; Fig. 3C).

Metabolism of LicA in serum, liver tissue, and mammary gland

LicA metabolites were analyzed in serum, liver, and mammary gland samples by UHPLC-MS/MS after s.c. administration of LicA. In serum, liver, and mammary tissues, glucuronidation reactions dominated LicA metabolism and resulted in two major (MG1 and MG2) and one minor (MG3) LicA glucuronide metabolites (Fig. 1B; Supplementary Fig. S2A–S2D). LicA also formed a GSH conjugate, which was a major metabolite in the liver (Supplementary Fig. S2; Fig. 1B) and a minor metabolite in the serum and mammary tissues (Supplementary Fig. S2A and S2C). Sulfation was minor in both serum and liver (Supplementary Fig. S2A and S2B; Fig. 1B) and not detectable in mammary tissue (Supplementary Fig. S2C). LicA also formed phase I metabolites (M1, M2, and M3); however, these metabolites were only detectable in vivo after their glucuronides were hydrolyzed with β-glucuronidase/sulfatase (Supplementary Fig. S2D). These LicA phase I and II metabolites have been previously determined from incubations of LicA in liver microsomes (27).

Licorice species and LicA increase NQO1 activity in the liver

NQO1 activity was measured in liver and mammary tissues. EB treatment alone did not affect the NQO1 activity significantly in liver tissue (Fig. 4). EB treatment slightly reduced NQO1 activity in the mammary gland which is consistent with previous reports (Supplementary Fig. S3; ref. 32).

Figure 4.

NQO1 induction by GG, GI, and LicA in the liver. NQO1 activity was measured in the liver after administration of vehicle, EB (1 mg/kg/day), and EB (1 mg/kg/day) with GG (2 g/kg/day), GI (2 g/kg/day), or LicA (80 mg/kg/day) to ACI rats for 4 days. Results are normalized to EB control.

Figure 4.

NQO1 induction by GG, GI, and LicA in the liver. NQO1 activity was measured in the liver after administration of vehicle, EB (1 mg/kg/day), and EB (1 mg/kg/day) with GG (2 g/kg/day), GI (2 g/kg/day), or LicA (80 mg/kg/day) to ACI rats for 4 days. Results are normalized to EB control.

Close modal

Upon cotreatment of EB with botanicals, GG and GI significantly induced NQO1 activity to 2-fold of the EB control groups in liver tissue. LicA significantly increased NQO1 activity to 1.5-fold (Fig. 4). No induction in NQO1 activity by the licorice extracts or LicA was observed in mammary tissue (Supplementary Fig. S3).

GG, GI, and LicA significantly downregulate overall estrogen oxidative metabolism in serum

As rat P450 1B1 preferentially catalyzes estrogen 2-hydroxylation (20), the levels of 2-MeOE1 were measured in serum samples as a biomarker for overall estrogen oxidative metabolism. The results showed that all three groups, GG, GI, and LicA, significantly reduced 2-MeOE1 by almost 60%, 70%, and 70% (Fig. 5A; Supplementary Fig. S4) of the EB treatment group, respectively. To analyze the influence of the botanicals on the overall amount of E2/E1 levels, the E2/E1 concentrations were also determined in serum. GG and GI slightly reduced E2/E1 levels; however, the difference did not reach significance (Supplementary Fig. S5).

Figure 5.

Influence of GG, GI, and LicA on estrogen oxidative metabolism in serum and modulation of CYP1A1 and CYP1B1 expression in mammary tissue. A, Serum analyzed for 2-MeOE1 after ACI rats were dosed for 4 days with vehicle, EB (1 mg/kg/day), and EB (1 mg/kg/day) coadministered with GG (2 g/kg/day), GI (2 g/kg/day), or LicA (80 mg/kg/day). The vehicle control group did not contain quantifiable amounts of 2-MeOE1 (Supplementary Fig. S4). Significance was calculated by comparing EB treatment with the treatment groups (EB plus GG, or GI, or LicA). B, Mammary tissues were collected from these ACI rats, and CYP1A1/CYP1B1 expression was analyzed with RT-qPCR. Significance was determined by comparing EB treatment with vehicle control and by comparing the treatment groups (EB plus GG, or GI, or LicA) with EB treatment.

Figure 5.

Influence of GG, GI, and LicA on estrogen oxidative metabolism in serum and modulation of CYP1A1 and CYP1B1 expression in mammary tissue. A, Serum analyzed for 2-MeOE1 after ACI rats were dosed for 4 days with vehicle, EB (1 mg/kg/day), and EB (1 mg/kg/day) coadministered with GG (2 g/kg/day), GI (2 g/kg/day), or LicA (80 mg/kg/day). The vehicle control group did not contain quantifiable amounts of 2-MeOE1 (Supplementary Fig. S4). Significance was calculated by comparing EB treatment with the treatment groups (EB plus GG, or GI, or LicA). B, Mammary tissues were collected from these ACI rats, and CYP1A1/CYP1B1 expression was analyzed with RT-qPCR. Significance was determined by comparing EB treatment with vehicle control and by comparing the treatment groups (EB plus GG, or GI, or LicA) with EB treatment.

Close modal

CYP1A1 and CYP1B1 expression in mammary tissues

To assess the effect on P450 1A1 and 1B1 expression in the mammary gland, CYP1A1 and CYP1B1 mRNA expression levels were measured. Treatment with EB significantly suppressed the expression of both, CYP1A1 and CYP1B1, to 3- and 5-fold lower than the vehicle control group, respectively (Fig. 5B). GG, GI, and LicA were administered with EB; therefore, their effects were compared with the EB treatment group to determine statistical significance. Interestingly, both GG, but also GI, upregulated CYP1A1 to 7- and 2-fold, respectively. Only GI caused significant downregulation of CYP1B1 expression, reducing it 3-fold (Fig. 5B); however, LicA-dosed rats showed no statistical difference from the EB-dosed rats.

CYP1A1 and CYP1B1 expression after treatment with GG, GI, and licorice compounds in MCF-10A cells

To identify potential compounds that might be responsible for the observed upregulation of CYP1A1 in mammary tissue by GG and especially GI, CYP1A1 and CYP1B1 expressions were determined in MCF-10A cells after treatment with GI, GG, LicA, LigC, and LigF. As expected from previous results (11), GG caused an increase in both genes, CYP1A1 (3-fold) and CYP1B1 (1.5-fold); however, after GI treatment, CYP1A1 and CYP1B1 expressions were reduced to nearly 13-fold and 6.5-fold less than basal levels, respectively (Fig. 6A). LicA (20 μmol/L) was responsible for decreases in both CYP1A1 and CYP1B1 expressions seen by GI (Fig. 6B), which were 34-fold and 10-fold lower than basal levels, respectively. In contrast, LigC (20 μmol/L) caused an increase in CYP1A1 and CYP1B1 expression. Interestingly, LigC preferentially increased CYP1A1 to 4-fold compared with a 2.5-fold induction of CYP1B1, and LigF significantly increased CYP1B1 to 2-fold of control (Fig. 6B).

Figure 6.

Modulation of CYP1A1 and CYP1B1 expression by GG, GI, and licorice compounds in MCF-10A cells. MCF-10A cells were treated with (A) licorice extracts, GG and GI (5 μg/mL), or (B) licorice compounds, LicA, LigC, and LigF (20 μmol/L), for 24 hours before analysis of CYP1A1/CYP1B1 expression by qPCR.

Figure 6.

Modulation of CYP1A1 and CYP1B1 expression by GG, GI, and licorice compounds in MCF-10A cells. MCF-10A cells were treated with (A) licorice extracts, GG and GI (5 μg/mL), or (B) licorice compounds, LicA, LigC, and LigF (20 μmol/L), for 24 hours before analysis of CYP1A1/CYP1B1 expression by qPCR.

Close modal

Licorice is contained in very popular BDS utilized for women's health and regarded as a potential chemopreventive agent (10, 33). It is also one of the prevalent plants in Traditional Chinese Medicine (TCM) as about 1/3 of TCM formulae contain licorice (Gan Cao; ref. 34). In the United States, GG is the most frequently utilized species. Besides GG, GI and also G. uralensis can be found in European dietary supplements. In China, all three species are cultivated and utilized without discrimination as Gan Cao. GG, GI, and their compounds demonstrated differential effects on estrogen oxidative metabolism in MCF-10A cells due to their distinct chemical profiles (11). In our previous studies, LigF had no effect, and GG and LigC treatments led to an increase in estrogen oxidative metabolism, whereas GI and LicA inhibited estrogen metabolism (11). In the ACI rat model, we explored GI's effect on detoxification (CYP1A1) and genotoxic (CYP1B1) pathways involved in estrogen chemical carcinogenesis and compared it with GG's effect. In addition, the contribution of LicA to GI's bioactivity and LicA's metabolism and distribution were analyzed. Although LicA was significantly conjugated with glucuronic acid (Fig. 1B; Supplementary Fig. S2A–S2D), free LicA was still detected in serum, liver, and mammary tissues 24 hours after the last LicA injection and oral administration of the GI extract (Supplementary Table S2; Supplementary Fig. S2; Fig. 3). This demonstrated that LicA was bioavailable in its free, bioactive form in target tissues. The present study corroborated previous reports (30, 35) that LigC and LigF were bioavailable (Fig. 3A and B).

GG, GI, and LicA induced NQO1 activity in the liver, but not in mammary tissue (Fig. 4; Supplementary Fig. S3). The lack of NQO1 activity in the mammary gland is consistent with lower LicA levels in mammary tissue compared with the liver (Supplementary Table S2; Fig. 3C). In addition, the inducibility of NQO1 in mammary epithelial cells has been demonstrated to be much lower than in liver cells (36). In support of this, other known NQO1 inducers, 4′bromoflavone, hops, and xanthohumol, caused only low (4′bromoflavone) or no NQO1 induction in the mammary gland compared with significant NQO1 induction in liver tissues of Sprague–Dawley rats (37). In comparison with vehicle, EB moderately reduced NQO1 activity in the mammary gland in this study (Supplementary Fig. S3). Estradiol has been shown before to reduce NQO1 in ER-positive cells and estrogen-sensitive tissue (32). However, EB did not change NQO1 activity in the liver in the present and previous investigations (Fig. 4; ref. 32).

The increase in NQO1 activity observed in liver tissues from GG, GI, and LicA groups confirmed the observed in vitro NQO1-inducing properties in liver cells (21). LicA (Fig. 2) contains a Michael acceptor group that reacts with sulfhydryl groups, such as in Keap1. LicA GSH conjugates were detected in liver tissues which show reaction with sulfur nucleophiles (Fig. 1B; Supplementary Fig. S2B and S2D). Activation of the Keap1/Nrf2 and ARE pathway by LicA and also LigC has been demonstrated previously in in vitro studies (21, 38) leading to reduction of oxidative stress (39, 40). However, although LigC formed GSH conjugates in vivo (Sprague–Dawley rats) similar to LicA, LigC did not induce NQO1 activity in in vivo models (21, 30). In previous in vitro studies, LigF had no effect on NQO1 activity (21). In the case of GG, these data suggest that other phytoconstituents other than LigC or LigF must contribute to the observed NQO1-inducing activity (Fig. 4).

GG, GI, and LicA caused a significant decrease in 2-MeOE1 levels (Fig. 5A) in serum. P450 1B1 in rats primarily catalyzes estrogen 2-hydroxylation; therefore, 2-MeOE1 was used as a general biomarker for estrogen oxidative metabolism (20). Estrogen hydroxylation in the liver is higher than in any other tissue and is predominantly performed by P450 3A4 and P450 1A2 (41). P450 1A1/2 and P450 1B1 are regulated by the aryl hydrocarbon receptor (AhR) and the xenobiotic response element (XRE; Fig. 1). LicA is an AhR antagonist (Fig. 1; ref. 11), and because P450 1A2 is regulated by AhR (42), it can be downregulated by LicA similarly to P450 1A1 (Fig. 6B) leading to reduced formation of 2-MeOE1 levels in serum. It is interesting that GG, which led to an induction of 2-MeOE1 in MCF-10A cells (Fig. 6), downregulated 2-MeOE1 levels in serum in this study (Fig. 5A). Literature data demonstrate that GG as well as GI, and licorice compounds inhibit P450 3A4 and P450 1A2 activity leading to reduction of estrogen oxidative metabolism as demonstrated by reduced 2-MeOE1 levels (Fig. 5A; refs. 43, 44). Specifically, P450 1A2 and P450 3A4 activity was moderately inhibited by GI and LicA and only weakly by GG, glabridin, and LigC (43, 44). LicA inhibited P450 3A4 irreversibly as a mechanism-based inhibitor (44) and decreased P450 3A4 gene expression in HepG2 cells (45). These results suggest that GG, GI, and LicA might interfere with general P450 metabolism (44); however, it is unclear at this point if these two licorice species lead to clinically relevant drug–botanical interactions. Clinical studies to analyze the influence of GG and GI on P450 enzymes and its drug interaction potential are warranted. In contrast to the 2-MeOE1 levels, E1/E2 levels were only slightly influenced by GG, GI, and LicA, suggesting that the decrease in estrogen oxidative metabolism did not lead to an increase in E1/E2 levels (Supplementary Fig. S5).

To gauge the effects of GG, GI, and LicA on estrogen metabolism in mammary tissues, CYP1A1 and CYP1B1 expressions were analyzed. These data closely correlated with 2-MeOE1 and 4-MeOE1 levels in our previous estrogen metabolism studies in MCF-10A and MCF-7 cells (11, 29, 46). A significant reduction in CYP1A1 and CYP1B1 by EB was observed in mammary tissues (Fig. 5B). This estrogen effect on CYP1A1 has previously been described in MCF-7 cells (46). As GG and GI caused upregulation of CYP1A1 and GI additionally caused CYP1B1 downregulation, GG and GI promote estrogen detoxification and GI also reduces the estrogen genotoxic pathway in mammary tissue (Fig. 5B). Our current study and literature reports indicated that LigC preferentially increased biomarkers of the 2-hydroxylation pathway, 2-MeOE1 and CYP1A1, in MCF-10A cells (Fig. 6B; ref. 11) and increased CYP1A1 levels (6.84-fold) in mammary tissues from female Sprague–Dawley rats (30). Thus, LigC is suggested to significantly contribute to the reversal of E2-mediated CYP1A1 downregulation as seen after GG and GI administration (Fig. 5B). Conversely, LicA, an AhR antagonist that inhibited CYP1A1/CYP1B1 expression in MCF-10A cells (Fig. 6B) and XRE-luciferase reporter activity in HepG2 cells (11), primarily downregulated CYP1B1 expression in vivo after GI administration (Fig. 5B). Surprisingly, GI did not decrease CYP1A1 expression in ACI rats (Fig. 5B), although GI and LicA treatments caused a significant reduction in CYP1A1 well below basal levels in MCF-10A cells (Fig. 6A). The GI crude extract used in MCF-10A cells and ACI rats (Table 1; Fig. 3B) contained LigC glycosides that were hydrolyzed extensively in the gut/intestine, increasing the concentration of LigC aglycone and consequently the LigC:LicA ratio and CYP1A1 induction in mammary tissues (Figs. 3B and 5B). LigF did not affect CYP1A1 expression in MCF-10A cells in these and previous studies (Fig. 6B; ref. 11). Considering that GG exhibits a very complex phytochemical profile, other constituents of GG may add to the observed CYP1A1 induction (16).

In summary, this study investigated the effects of two popular licorice species on biomarkers of detoxification and genotoxic estrogen metabolism pathways in ACI rats and breast epithelial cells. GG and GI increased NQO1 activity in the liver (Fig. 4) and the detoxification estrogen 2-hydroxyation pathway in the mammary tissues of ACI rats (Fig. 5B). In addition, GI also decreased CYP1B1 expression (genotoxic pathway), because it contains the chemopreventive AhR antagonist, LicA (Fig. 5B). Other studies suggest that GI has superior chemopreventive properties to GG (11, 47). GI had the greatest anti-inflammatory activity among medicinal licorice species in macrophage cells because it contains two major anti-inflammatory compounds, LicA and LigC (11). Also, GI contains the ERβ preferential agonist 8-prenylapigenin (47), which may have a better safety profile. Long-term studies are planned to compare the effect of GG and GI on E2-induced mammary carcinogenesis in ACI rats and ultimately estrogen metabolism in women. This study highlights the fact that the chemopreventive bioactivity of licorice species cannot be reduced to the activity of single bioactive compounds, but is rather a result of multiple constituents leading to polypharmacologic actions. Furthermore, knowing that licorice species have very different chemical profiles (14) that profoundly affect their bioactivity in vitro as well as in vivo should help design safer and more efficacious botanicals with the greatest chemopreventive potential in women. These data suggest that GI containing the chemopreventive compounds, LicA and LigC, might be the optimal licorice species used for women's health.

No potential conflicts of interest were disclosed.

Conception and design: S. Wang, L. Huang, G.F. Pauli, B.M. Dietz, J.L. Bolton

Development of methodology: S. Wang, L. Huang, Y. Liu, J.L. Bolton

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S. Wang, T.L. Dunlap, L. Huang, C. Simmler, D.D. Lantvit, J. Crosby, H. Dong, R.B. van Breemen

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S. Wang, T.L. Dunlap, L. Huang, C.E. Howell, H. Dong, B.M. Dietz

Writing, review, and/or revision of the manuscript: S. Wang, T.L. Dunlap, L. Huang, C. Simmler, C.E. Howell, S-N. Chen, G.F. Pauli, R.B. van Breemen, B.M. Dietz, J.L. Bolton, B.M. Dietz

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): S. Wang, T.L. Dunlap, L. Huang, R.B. van Breemen, B.M. Dietz, J.L. Bolton

Study supervision: S. Wang, G.F. Pauli, B.M. Dietz, J.L. Bolton

Other (prepared LicoA for in vivo assays): Y. Liu

Other (botanical integrity to ensure consistency and reproducibility): S.-N. Chen, C. Simmler, G.F. Pauli

This work was supported by NIH grant P50 AT000155 from the NIH Office of Dietary Supplements (ODS) and the National Center for Complementary and Integrative Health (NCCIH) to the UIC/NIH Center for Botanical Dietary Supplements Research and by NIH grant U41 AT008706 from NCCIH/ODS to the Center for Natural Product Technologies. The construction of the UIC CSB NMR facility and instrumentation was funded by NIGMS grant P41 GM068944 to Dr. Peter Gettins. We thank Shimadzu for providing the LCMS-8060 mass spectrometer used in this investigation. We also thank Dr. Liang Zhao at Lanzhou Institute of Chemical Physics, CAS, and Qinghai Lake Medicinal CO., Ltd. for their generous gifts.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer statistics, 2018
.
CA Cancer J Clin
2018
;
68
:
7
30
.
2.
Manson
JE
,
Chlebowski
RT
,
Stefanick
ML
,
Aragaki
AK
,
Rossouw
JE
,
Prentice
RL
, et al
Menopausal hormone therapy and health outcomes during the intervention and extended poststopping phases of the Women's Health Initiative randomized trials
.
JAMA
2013
;
310
:
1353
68
.
3.
Russo
J
,
Russo
IH
. 
The role of estrogen in the initiation of breast cancer
.
J Steroid Biochem Mol Biol
2006
;
102
:
89
96
.
4.
Yang
L
,
Zahid
M
,
Liao
Y
,
Rogan
EG
,
Cavalieri
EL
,
Davidson
NE
, et al
Reduced formation of depurinating estrogen-DNA adducts by sulforaphane or KEAP1 disruption in human mammary epithelial MCF-10A cells
.
Carcinogenesis
2013
;
34
:
2587
92
.
5.
Sampson
JN
,
Falk
RT
,
Schairer
C
,
Moore
SC
,
Fuhrman
BJ
,
Dallal
CM
, et al
Association of estrogen metabolism with breast cancer risk in different cohorts of postmenopausal women
.
Cancer Res
2017
;
77
:
918
25
.
6.
Gupta
M
,
McDougal
A
,
Safe
S
. 
Estrogenic and antiestrogenic activities of 16alpha- and 2-hydroxy metabolites of 17beta-estradiol in MCF-7 and T47D human breast cancer cells
.
J Steroid Biochem Mol Biol
1998
;
67
:
413
9
.
7.
Wen
W
,
Ren
Z
,
Shu
XO
,
Cai
Q
,
Ye
C
,
Gao
Y-T
, et al
Expression of cytochrome P450 1B1 and catechol-O-methyltransferase in breast tissue and their associations with breast cancer risk
.
Cancer Epidemiol Biomarkers Prev
2007
;
16
:
917
20
.
8.
Bolton
JL
,
Dunlap
T
. 
Formation and biological targets of quinones: cytotoxic versus cytoprotective effects
.
Chem Res Toxicol
2017
;
30
:
13
37
.
9.
Yager
JD
. 
Mechanisms of estrogen carcinogenesis: The role of E2/E1–quinone metabolites suggests new approaches to preventive intervention – A review
.
Steroids
2015
;
99
:
56
60
.
10.
Dietz
BM
,
Hajirahimkhan
A
,
Dunlap
TL
,
Bolton
JL
. 
Botanicals and their bioactive phytochemicals for women's health
.
Pharmacol Rev
2016
;
68
:
1026
73
.
11.
Dunlap
TL
,
Wang
S
,
Simmler
C
,
Chen
S-N
,
Pauli
GF
,
Dietz
BM
, et al
Differential effects of Glycyrrhiza species on genotoxic estrogen metabolism: licochalcone A downregulates P450 1B1, whereas isoliquiritigenin stimulates it
.
Chem Res Toxicol
2015
;
28
:
1584
94
.
12.
Kondo
K
,
Shiba
M
,
Yamaji
H
,
Morota
T
,
Zhengmin
C
,
Huixia
P
, et al
Species identification of licorice using nrDNA and cpDNA genetic markers
.
Biol Pharm Bull
2007
;
30
:
1497
502
.
13.
Hajirahimkhan
A
,
Simmler
C
,
Yuan
Y
,
Anderson
JR
,
Chen
S-N
,
Nikolić
D
, et al
Evaluation of estrogenic activity of licorice species in comparison with hops used in botanicals for menopausal symptoms
.
PLoS One
2013
;
8
:
e67947
.
14.
Simmler
C
,
Anderson
JR
,
Gauthier
L
,
Lankin
DC
,
McAlpine
JB
,
Chen
SN
, et al
Metabolite profiling and classification of DNA-authenticated licorice botanicals
.
J Nat Prod
2015
;
78
:
2007
22
.
15.
Tao
W
,
Duan
J
,
Zhao
R
,
Li
X
,
Yan
H
,
Li
J
, et al
Comparison of three officinal Chinese pharmacopoeia species of Glycyrrhiza based on separation and quantification of triterpene saponins and chemometrics analysis
.
Food Chem
2013
;
141
:
1681
9
.
16.
Farag
MA
,
Porzel
A
,
Wessjohann
LA
. 
Comparative metabolite profiling and fingerprinting of medicinal licorice roots using a multiplex approach of GC-MS, LC-MS and 1D NMR techniques
.
Phytochemistry
2012
;
76
:
60
72
.
17.
Mersereau
JE
,
Levy
N
,
Staub
RE
,
Baggett
S
,
Zogovic
T
,
Chow
S
, et al
Liquiritigenin is a plant-derived highly selective estrogen receptor beta agonist
.
Mol Cell Endocrinol
2008
;
283
:
49
57
.
18.
Simmler
C
,
Hajirahimkhan
A
,
Lankin
DC
,
Bolton
JL
,
Jones
T
,
Soejarto
DD
, et al
Dynamic residual complexity of the isoliquiritigenin-liquiritigenin interconversion during bioassay
.
J Agric Food Chem
2013
;
61
:
2146
57
.
19.
Shull
JD
,
Spady
TJ
,
Snyder
MC
,
Johansson
SL
,
Pennington
KL
. 
Ovary-intact, but not ovariectomized female ACI rats treated with 17β-estradiol rapidly develop mammary carcinoma
.
Carcinogenesis
1997
;
18
:
1595
601
.
20.
Nishida
CR
,
Everett
S
,
Ortiz de Montellano
PR
. 
Specificity determinants of CYP1B1 estradiol hydroxylation
.
Mol Pharmacol
2013
;
84
:
451
8
.
21.
Hajirahimkhan
A
,
Simmler
C
,
Dong
H
,
Lantvit
DD
,
Li
G
,
Chen
SN
, et al
Induction of NAD(P)H:quinone oxidoreductase 1 (NQO1) by Glycyrrhiza species used for women's health: differential effects of the Michael acceptors isoliquiritigenin and licochalcone A
.
Chem Res Toxicol
2015
;
28
:
2130
41
.
22.
Simmler
C
,
Jones
T
,
Anderson
JR
,
Nikolić
DC
,
van Breemen
RB
,
Soejarto
DD
, et al
Species-specific standardisation of licorice by metabolomic profiling of flavanones and chalcones
.
Phytochem Anal
2014
;
25
:
378
88
.
23.
Liu
Y
,
Friesen
JB
,
Grzelak
EM
,
Fan
Q
,
Tang
T
,
Durić
K
, et al
Sweet spot matching: a thin-layer chromatography-based countercurrent solvent system selection strategy
.
J Chromatogr A
2017
;
1504
:
46
54
.
24.
Liu
Y
,
Friesen
JB
,
Klein
LL
,
McAlpine
JB
,
Lankin
DC
,
Pauli
GF
, et al
The generally useful estimate of solvent systems (GUESS) method enables the rapid purification of methylpyridoxine regioisomers by countercurrent chromatography
.
J Chromatogr A
2015
;
1426
:
248
51
.
25.
Liu
Y
,
Friesen
JB
,
McAlpine
JB
,
Pauli
GF
. 
Solvent system selection strategies in countercurrent separation
.
Planta Med
2015
;
81
:
1582
91
.
26.
Wang
QE
,
Lee
FSC
,
Wang
X
. 
Isolation and purification of inflacoumarin A and licochalcone A from licorice by high-speed counter-current chromatography
.
J Chromatogr A
2004
;
1048
:
51
7
.
27.
Huang
L
,
Nikolic
D
,
van Breemen
RB
. 
Hepatic metabolism of licochalcone A, a potential chemopreventive chalcone from licorice (Glycyrrhiza inflata), determined using liquid chromatography-tandem mass spectrometry
.
Anal Bioanal Chem
2017
;
409
:
6937
48
.
28.
Li
G
,
Nikolic
D
,
van Breemen
RB
. 
Identification and chemical standardization of licorice raw materials and dietary supplements using UHPLC-MS/MS
.
J Agric Food Chem
2016
;
64
:
8062
70
.
29.
Wang
S
,
Dunlap
TL
,
Howell
CE
,
Mbachu
OC
,
Rue
EA
,
Phansalkar
R
, et al
Hop (Humulus lupulus L.) extract and 6-prenylnaringenin induce P450 1A1 catalyzed estrogen 2-hydroxylation
.
Chem Res Toxicol
2016
;
29
:
1142
50
.
30.
Cuendet
M
,
Guo
J
,
Luo
Y
,
Chen
S
,
Oteham
CP
,
Moon
RC
, et al
Cancer chemopreventive activity and metabolism of isoliquiritigenin, a compound found in licorice
.
Cancer Prev Res
2010
;
3
:
221
32
.
31.
Prochaska
HJ
,
Santamaria
AB
. 
Direct measurement of NAD(P)H:quinone reductase from cells cultured in microtiter wells: a screening assay for anticarcinogenic enzyme inducers
.
Anal Biochem
1988
;
169
:
328
36
.
32.
Ansell
PJ
,
Espinosa-Nicholas
C
,
Curran
EM
,
Judy
BM
,
Philips
BJ
,
Hannink
M
, et al
In vitro and in vivo regulation of antioxidant response element-dependent gene expression by estrogens
.
Endocrinology
2004
;
145
:
311
7
.
33.
Wang
R
,
Zhang
CY
,
Bai
LP
,
Pan
HD
,
Shu
LM
,
Kong
A-NT
, et al
Flavonoids derived from liquorice suppress murine macrophage activation by up-regulating heme oxygenase-1 independent of Nrf2 activation
.
Int Immunopharmacol
2015
;
28
:
917
24
.
34.
Wang
X
,
Zhang
H
,
Chen
L
,
Shan
L
,
Fan
G
,
Gao
X
. 
Liquorice, a unique “guide drug” of traditional Chinese medicine: a review of its role in drug interactions
.
J Ethnopharmacol
2013
;
150
:
781
90
.
35.
Madak-Erdogan
Z
,
Gong
P
,
Zhao
YC
,
Xu
L
,
Wrobel
KU
,
Hartman
JA
, et al
Dietary licorice root supplementation reduces diet-induced weight gain, lipid deposition, and hepatic steatosis in ovariectomized mice without stimulating reproductive tissues and mammary gland
.
Mol Nutr Food Res
2016
;
60
:
369
80
.
36.
Jiang
Z-Q
,
Chen
C
,
Yang
B
,
Hebbar
V
,
Kong
A-NT
. 
Differential responses from seven mammalian cell lines to the treatments of detoxifying enzyme inducers
.
Life Sci
2003
;
72
:
2243
53
.
37.
Dietz
BM
,
Hagos
GK
,
Eskra
JN
,
Wijewickrama
GT
,
Anderson
JR
,
Nikolic
D
, et al
Differential regulation of detoxification enzymes in hepatic and mammary tissue by hops (Humulus lupulus) in vitro and in vivo
.
Mol Nutr Food Res
2013
;
57
:
1055
66
.
38.
Luo
Y
,
Eggler
AL
,
Liu
D
,
Liu
G
,
Mesecar
AD
,
van Breemen
RB
. 
Sites of alkylation of human keap1 by natural chemoprevention agents
.
J Am Soc Mass Spectrom
2007
;
18
:
2226
32
.
39.
Dinkova-Kostova
AT
,
Kostov
RV
,
Canning
P
. 
Keap1, the cysteine-based mammalian intracellular sensor for electrophiles and oxidants
.
Arch Biochem Biophys
2017
;
617
:
84
93
.
40.
Kühnl
J
,
Roggenkamp
D
,
Gehrke
SA
,
Stäb
F
,
Wenck
H
,
Kolbe
L
, et al
Licochalcone A activates Nrf2 in vitro and contributes to licorice extract-induced lowered cutaneous oxidative stress in vivo
.
Exp Dermatol
2015
;
24
:
42
7
.
41.
Tsuchiya
Y
,
Nakajima
M
,
Yokoi
T
. 
Cytochrome P450-mediated metabolism of estrogens and its regulation in human
.
Cancer Lett
2005
;
227
:
115
24
.
42.
Hankinson
O
. 
The role of AHR-inducible cytochrome P450s in metabolism of polyunsaturated fatty acids
.
Drug Metab Rev
2016
;
48
:
342
50
.
43.
He
W
,
Wu
J-J
,
Ning
J
,
Hou
J
,
Xin
H
,
He
Y-Q
, et al
Inhibition of human cytochrome P450 enzymes by licochalcone A, a naturally occurring constituent of licorice
.
Toxicol Vitr
2015
;
29
:
1569
76
.
44.
Li
G
,
Simmler
C
,
Chen
L
,
Nikolic
D
,
Chen
SN
,
Pauli
GF
, et al
Cytochrome P450 inhibition by three licorice species and fourteen licorice constituents
.
Eur J Pharm Sci
2017
;
109
:
182
90
.
45.
Chen
H
,
Zhang
X
,
Feng
Y
,
Rui
W
,
Shi
Z
,
Wu
L
. 
Bioactive components of Glycyrrhiza uralensis mediate drug functions and properties through regulation of CYP450 enzymes
.
Mol Med Rep
2014
;
10
:
1355
62
.
46.
Dunlap
TL
,
Howell
CE
,
Mukand
N
,
Chen
S-N
,
Pauli
GF
,
Dietz
BM
, et al
Red clover aryl hydrocarbon receptor (AhR) and estrogen receptor (ER) agonists enhance genotoxic estrogen metabolism
.
Chem Res Toxicol
2017
;
30
:
2084
92
.
47.
Hajirahimkhan
A
,
Mbachu
O
,
Simmler
C
,
Ellis
S
,
Dong
H
,
Nikolic
D
, et al
Estrogen receptor (ER) subtype selectivity identifies 8-prenylapigenin as an ERβ agonist from Glycyrrhiza inflata and highlights the importance of chemical and biological authentication
.
J Nat Prod
2018;
81:966–75.