검색
검색 팝업 닫기

Ex) Article Title, Author, Keywords

Article

Split Viewer

Original Research Article

DTT 2024; 3(1): 39-50

Published online March 31, 2024

https://doi.org/10.58502/DTT.23.0029

Copyright © The Pharmaceutical Society of Korea.

Beneficial Interaction between Isoliquiritigenin and Liquiritigenin in their Pharmacokinetics in Rats Following Oral Intake of Glycyrrhizae Radix Extract Compared with Oral Intake of an Individual Component

You Jin Han, Kyung-Sik Song, Im-Sook Song

Department of Pharmacy, Kyungpook National University, Daegu, Korea

Correspondence to:Im-Sook Song, isssong@knu.ac.kr

Received: October 4, 2023; Revised: January 25, 2024; Accepted: January 26, 2024

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Glycyrrhizae Radix (GR) is a widely used herbal medicine. Its pharmacological efficacy depends largely on the composition of bioactive saponins and flavonoids, including glycyrrhizin, liquiritin, isoliquiritin, isoliquiritigenin, and liquiritigenin. This study aimed to compare the pharmacokinetics of glycyrrhizin, liquiritin, isoliquiritin, isoliquiritigenin, and liquiritigenin between orally administered GR extract and an equal amount of a single component. This study also aimed to investigate the intestinal absorption and metabolism of these major pharmacological components in rats. Plasma concentrations of liquiritigenin and isoliquiritigenin from the administered GR extract rapidly decreased for 4 h, increased for 4-10 h, and subsequently stabilized. However, a different profile was observed following a single administration of liquiritigenin or isoliquiritigenin despite a similar dose (0.5 mg/kg or 0.2 mg/kg, respectively). In contrast, the plasma concentrations and pharmacokinetic parameters of glycyrrhizin, liquiritin, and isoliquiritin differed insignificantly from the corresponding results of equivalent doses in rats. Consistent with the pharmacokinetic results, the apparent permeability of liquiritigenin and isoliquiritigenin from the administered GR extract increased by 2.2- and 4.8-fold, respectively, compared with that of individual components. Additionally, isoliquiritigenin was formed from isoliquiritin at the highest rate, converted from liquiritigenin at the lower rate, and converted from liquiritin at the lowest rate in intestinal segments of the rats. Liquiritigenin had a similar process. Therefore, isoliquiritin, liquiritin, and liquiritigenin could be biosources for isoliquiritigenin and liquiritigenin, which could occur in intestinal enterocytes, and the converted metabolites are absorbed into the plasma. Conclusively, the beneficial interaction between liquiritigenin and isoliquiritigenin in orally administered GR extract via metabolic conversion in intestinal enterocytes and enhanced absorption could provide a basis for treatment with GR extract rather than with the individual components.

KeywordsGlycyrrhizae Radix (GR) extract, isoliquiritigenin, liquiritigenin, pharmacokinetics, intestinal permeability, biotransformation

Glycyrrhizae Radix (GR, root of licorice) has several pharmacological activities, including chemopreventive, hypoglycemic, anti-inflammatory, and antioxidative activities (Xie et al. 2014; Ng et al. 2021). It has been used as an adjuvant to increase the therapeutic efficacy of other drugs (Qiao et al. 2014) and as a sweetener in traditional medicines, chewing gums, and chocolates (Ishida et al. 1992; Cantelli-Forti et al. 1994; Kobayashi et al. 1995; Lin et al. 2005). Moreover, orally administered GR extract (0.1-0.5 g/kg) in mice for nine days effectively ameliorated interferon-γ-related autoimmune responses and demonstrated neuroprotective efficacy (Yang et al. 2013; Yang et al. 2019). To understand the relationship between the response elicited by GR extract and its pharmacokinetics, bioanalysis of the predominant or pharmacological components of GR extract in biological samples following supplementation and investigation of their pharmacokinetics are necessary.

Glycyrrhizin (Fig. 1A), a major marker component of GR (Kobayashi et al. 1995; Qiao et al. 2014; Peng et al. 2015; Dong et al. 2016), exerts strong neuroprotective effects on experimental autoimmune encephalomyelitis and glutamate-induced apoptosis in neuronal cells and shows therapeutic effects against arthritis, hepatotoxicity, leukemia, allergies, stomach ulcers, and inflammation (Maggiolini et al. 2002). The major active flavonoids of GR, including isoliquiritin, liquiritin, isoliquiritigenin, and liquiritigenin (Gao et al. 2009) (Fig. 1B-1E), are frequently used as antidepressant, anticancer, cardioprotective, antimicrobial, and neuroprotective agents (Cuendet et al. 2006; Cuendet et al. 2010). The pharmacological activities of the major active flavonoids can overlap; however, the main activities could be differentiated. For instance, liquiritin, isoliquiritin, and glycyrrhizic acid mainly and positively influence the anti-inflammatory, anticancer, cardioprotective, and antioxidant activities with different potencies, whereas their deglycosylated metabolites, liquiritigenin, isoliquiritigenin, and glycyrrhetinic acid, mainly and positively influence the immunoregulatory and neuroprotective activities (Qin et al. 2022; Wu et al. 2022; He et al. 2023). Additionally, chemically interconnected liquiritigenin and isoliquiritigenin showed significant efficacy in Alzheimer’s disease, Parkinson’s disease, and other neurodegenerative diseases and disorders in numerous in vitro and in vivo experimental studies (Ramalingam et al. 2018).

Figure 1.Structure of (A) glycyrrhizin, (B) liquritin, (C) isoliquiritin, (D) liquiritigenin, and (E) isoliquiritigenin.

Based on literature search, glycyrrhizin, liquiritin, isoliquiritigenin, and liquiritigenin were selected as the predominant or pharmacological components of the GR extract. We previously developed an analytical method for glycyrrhizin, liquiritin, isoliquiritigenin, and liquiritigenin analysis using a liquid chromatography with tandem mass spectrometry (LC-MS/MS) system with a relatively higher sensitivity (e.g., lower limit of quantification: glycyrrhizin [2 ng/mL] and isoliquiritigenin, liquiritigenin, and liquiritin [0.2 ng/mL each]) with a small volume (50 μL) of plasma samples (Han et al. 2019). Using these analytical methods, glycyrrhizin, isoliquiritigenin, liquiritin, and liquiritigenin plasma concentrations in rats were determined following a single oral GR extract administration at a dose of 1 g/kg (Han et al. 2019). In this study, glycyrrhizin concentration in plasma showed the highest and most stable profile for 12 h. Isoliquiritigenin and liquiritigenin concentrations in plasma were similar and eliminated rapidly after 4 h; however, they rebounded to the initial plasma concentration for 10 h, which could be explained from the biotransformation of liquiritin to isoliquiritigenin (Han et al. 2019). Similarly, co-administration with Jiegeng changed glycyrrhizin and liquiritigenin metabolism in rat fecal lysate, consequently altering their pharmacokinetic profiles (Mao et al. 2017). Besides this biotransformation, compared with the addition of an equal amount of liquiritin alone, liquiritin permeability was much greater when added as a part of the GR extract (Mao et al. 2017), consistent with our case.

Therefore, this study aimed to pharmacokinetically compare GR extract administration with that of an equal amount of a single component contained in the GR extract, including glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin, in rats (Wang et al. 2016). This study also aimed to investigate and compare the intestinal absorption and metabolism of these major pharmacological components in coexisting rats. Isoliquiritin was included in the pharmacokinetic and permeability study to better understand the in vivo biotransformation process.

Materials

Glycyrrhizin (purity > 95.0%), isoliquiritigenin (purity > 99.0%), isoliquiritin (purity > 90.0%), liquiritin (purity > 98.0%), liquiritigenin (purity > 97.0%), berberine (purity ≥ 98.0%), dimethyl sulfoxide (DMSO), and Hank’s balanced salt solution (HBSS) were purchased from Sigma-Aldrich (St. Louis, MO, USA). Water, methanol, and other solvents were obtained from J.T. Baker Korea (Seoul, Korea) and TEDIA (Fairfield, OH, USA). All other chemicals were of reagent and analytical grade.

Plant material and extraction

Dried GR (1 kg), obtained from CK Pharm Co. (Seoul, Korea), was extracted with 12 L of 94% ethanol for 3 h and filtered through Whatman Qualitative filter paper grade 1. The filtrate was concentrated using a rotary evaporator (EYELA, Tokyo, Japan) to obtain the GR extract. The voucher specimen (#KNUNPM GR-2015-002) was deposited at the laboratory of Natural Products Medicine at Kyungpook National University (Daegu, Korea).

Animals

Male Sprague–Dawley rats (7 weeks old, 249 ± 6 g) were obtained from Samtako (Osan, Korea) and were acclimated for one week at the animal facility of the College of Pharmacy, Kyungpook National University. Rats were maintained in an environmentally clean room at 21℃-27℃ with 12-h light (09:00-21:00) and relative humidity of 60% ± 5%. Furthermore, they were fasted for 12 h with water ad libitum. The Kyungpook National University Animal Committee approved the protocols and procedures for the animal studies (IACUC No.: KNU-2017-0126, approval date: September 23, 2017).

Pharmacokinetic study

Rats were fasted for 16 h with water ad libitum before the pharmacokinetic experiments. Their femoral arteries were cannulated with a polyethylene tube (PE50, Jungdo, Seoul, Korea) under anesthesia with isoflurane (isoflurane vaporizer set to 2%, with 0.8 L/min oxygen flow).

The GR extract (1 g/kg/2 mL suspended in distilled water containing 10% DMSO) was orally administered to four rats through gavage. The corresponding doses of glycyrrhizin (20 mg/kg/2 mL suspended in distilled water containing 10% DMSO), liquiritin (5.8 mg/kg/2 mL), isoliquiritin (1.5 mg/kg/2 mL), liquiritigenin (0.5 mg/kg/2 mL), or isoliquiritigenin (0.2 mg/kg/2 mL) were orally administered to rats (four rats per component) through gavage. Blood samples (approximately 120 μL) were taken via the femoral artery at 0, 0.25, 0.5, 0.75, 1, 2, 4, 6, 8, 10, and 12 h and centrifuged at 10,000 × g for 1 min at 4℃. Supernatant plasma samples (50 μL each) were stored at −80℃ until analysis.

Intestinal permeability test

Rats were fasted for 16 h. However, they had nonrestricted access to water before the study commenced. They were anesthetized using isoflurane (isoflurane vaporizer set to 2%, with 0.8 L/min oxygen flow), and the proximal part of the ileal segments (approximately 20 cm) were excised, opened along the mesenteric border, and washed in prewarmed HBSS (pH 7.4). Segments were mounted in a tissue holder of a Navicyte Easy Mount Ussing Chamber (Warner Instruments, Holliston, MA, USA) with a 0.76 cm2 surface area. They were acclimated in HBSS for 15 min with continuous oxygenation (95% O2/5% CO2 gas). Intestinal permeability studies were conducted by adding HBSS to both sides of the intestinal segments, which included 1 mL of prewarmed HBSS containing the GR extract or equivalent single component (e.g., GR extract [100 mg/6 mL], glycyrrhizin [2 mg/6 mL], liquiritin [0.6 mg/6 mL], isoliquiritin [0.15 mg/6 mL], liquiritigenin [0.05 mg/6 mL], and isoliquiritigenin [0.02 mg/6 mL]) on the apical side of the ileal segments and 1 mL of prewarmed fresh HBSS on the basal side. Sample aliquots (400 μL) were withdrawn at 30-min intervals for 2 h from the basal side, and an equal volume of prewarmed fresh HBSS was subsequently replenished. During the experiment, carbogen gas (5% CO2/95% O2) was bubbled into the Ussing chambers at 150 drops/min. Samples were stored at −80℃ until analysis.

Next, we investigated the permeability of isoliquiritigenin (0.02 mg/6 mL) in the absence or presence of a single component of the GR extract using the same experimental procedures described above. Briefly, its permeability was assessed by adding 1 mL of prewarmed HBSS containing isoliquiritigenin (0.02 mg/6 mL) in the absence or presence of a single component of the GR extract (e.g., glycyrrhizin [2 mg/6 mL], liquiritin [0.6 mg/6 mL], isoliquiritin [0.15 mg/6 mL], and liquiritigenin [0.05 mg/6 mL]) on the apical side of the ileal segments and 1 mL of prewarmed fresh HBSS on the basal side. Sample aliquots (400 μL) were withdrawn at 30-min intervals for 2 h from the basal side, and an equal volume of prewarmed fresh HBSS was subsequently replenished. Samples were stored at −80℃ until analysis.

Biotransformation of liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin

Rats were fasted for 16 h with water ad libitum before ileal dissection. They were anesthetized using isoflurane (isoflurane vaporizer set to 2%, with 0.8 L/min oxygen flow), and the proximal part of the ileal segments (approximately 20 cm) were excised. The dissected ileal segments were washed using a 10 mL syringe filled with prewarmed HBSS (pH 7.4), and the eluent was vortexed for 1 min followed by centrifugation at 1,000 × g for 5 min at 4℃. The supernatant of the intestinal eluent was used for incubation with isoliquiritigenin, liquiritigenin, and liquiritin.

The ileal segments were mounted onto a Navicyte Easy Mount Ussing Chamber and acclimatized with HBSS for 30 min. Experiments started by changing HBSS with a prewarmed intestinal eluent (1 mL) containing liquiritin, isoliquiritin, liquiritigenin, or isoliquiritigenin (100 μg/mL each) to the apical side of the ileal segments and incubated for 2 h. During the experiment, carbogen gas (5% CO2/95% O2) was bubbled into the Ussing chambers at 150 drops/min. Sample aliquots (50 μL) were mixed with 300 µL methanol containing berberine (0.1 ng/mL), vortexed for 10 min, and centrifuged at 10,000 × g for 10 min at 4℃. An aliquot (10 µL) of the supernatant was injected into the LC-MS/MS system.

LC-MS/MS analysis of glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin

An Agilent 6470 triple quadrupole mass spectrometer equipped with an Agilent Infinity 1260 Infinite II HPLC system (Agilent Technologies, Santa Clara, CA, USA) was used to analyze glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin. The method developed by Han et al. (2019) was used with slight modifications. Table 1 summarizes the MS/MS conditions for the detection of these analytes. The mobile phase consisted of methanol:water (65:35, v/v) with 0.1% formic acid and was eluted in the isocratic mode. A Synergi Polar-RP column (4 µm, 150 × 2 mm; Phenomenex, Torrance, CA, USA) equipped with a polar-RP guard column (4 × 2 mm; Phenomenex) was used for chromatographic separation.

Table 1 MS/MS parameters for the detection of the analytes and IS

CompoundsMRM transitions (m/z)Ion modeTR (min)CE (eV)
Precursor ionProduct ion
Liquiritin417.2254.9Negative2.020
Isoliquritin417.2254.9Negative2.420
Liquiritigenin255.1135.0Negative3.215
Isoliquiritigenin255.1135.0Negative4.215
Glycyrrhizin845.5669.4Positive8.735
Berberine (IS)336.1320.0Positive4.930

For the analysis of the glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin contents in the GR extract, 100 mg of the GR extract was diluted 200 times with methanol. The diluted samples (50 μL) were added to 300 µL methanol containing berberine (0.1 ng/mL), vortexed for 10 min, and centrifuged at 10,000 × g for 10 min at 4℃. A supernatant aliquot (10 µL) was injected into the LC-MS/MS system.

For the analysis of glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin plasma concentrations, plasma samples (50 µL) were added to 300 µL methanol containing berberine (0.1 ng/mL), vortexed for 10 min, and centrifuged at 10,000 × g for 10 min at 4℃. A supernatant aliquot (10 µL) was injected into the LC-MS/MS system. For the analysis of glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin in HBSS, HBSS samples (50 µL) were added to 300 µL methanol containing berberine (0.1 ng/mL), vortexed for 10 min, and centrifuged at 10,000 × g for 10 min at 4℃. A supernatant aliquot (10 µL) was injected into the LC-MS/MS system.

Data and statistical analysis

Using noncompartmental analysis (WinNonlin 6.3; Pharsight Corp., Mountain View, CA, USA), the following pharmacokinetic parameters of glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin were calculated: terminal half-life (t1/2), area under the plasma concentration–time curve during the observation period (AUC12h), AUC to infinite time (AUCinf), and mean residence time (MRT). The maximum concentration (Cmax) and the time to reach Cmax (Tmax) were obtained from the experimental data.

To calculate permeability, the transport rate of the analytes (i.e., glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin) was calculated from the slope of the regression line from the mean permeated amounts versus the incubation time plot. The apparent permeability (Papp) was calculated using the following equation (Jeon et al. 2021; Lim et al. 2022):

Papp(cm/s)=transport rate (nmol/s)Initial concentration of analyte (μM)×surface area of membrane (cm2)

The concentrations of the analytes (i.e., liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin) were quantitated, and their formation rates (pmol/mg protein/min) were calculated by dividing the amounts of metabolites formed from the incubation with enterocytes in the rats’ ileal segments by the incubation time.

Student’s t-test (IBM SPSS Statistics, version 26; Armonk, NY, USA) was used to determine the statistical significance of the pharmacokinetic parameters and treatment groups. p-values of < 0.05 were considered statistically significant.

Content of the GR extract

Table 2 summarizes the glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin concentrations in the GR extract. Glycyrrhizin had the highest concentration (2.0%), consistent with that of previous studies (Kobayashi et al. 1995; Lin et al. 2005; Wang et al. 2009; Xie et al. 2014). Liquritin, isoliquiritin, liquiritigenin, and isoliquiritigenin had lower concentrations (0.6%, 0.1%, 0.05%, and 0.03%, respectively).

Table 2 The contents of glycyrrhizin and 4 flavonoids in the GR extract

CompoundsContent (mg/g extract)Content (%)
Glycyrrhizin20.2 ± 3.22.0 ± 0.3
Liquiritin5.8 ± 1.10.6 ± 0.1
Isoliquiritin1.5 ± 0.30.1 ± 0.0
Liquiritigenin0.49 ± 0.060.05 ± 0.01
Isoliquiritigenin0.29 ± 0.060.03 ± 0.01

For the pharmacokinetic comparison, we administered 1 g of GR extract/kg to the rats, and the corresponding doses of saponin and flavonoids, 20 mg/kg of glycyrrhizin, 5.8 mg/kg of liquiritin, 1.5 mg/kg of isoliquiritin, 0.5 mg/kg of liquiritigenin, and 0.2 mg/kg of isoliquiritigenin were determined based on the content in the GR extract (Table 2). Additionally, to mimic the intestinal situation that occurred when the GR extract was orally administered to the rats, isoliquiritigenin, liquiritigenin, and liquiritin concentrations were determined based on the oral dose of the GR extract, their content in the GR extract, and the fluid volume of the small intestine. For example, the GR extract was administered at a dose of 1 g/kg/2 mL to rats with stomach and intestinal fluid volumes of 3.38-6.63 and 11.1-16.5 mL, respectively (McConnell et al. 2008), resulting in a 15-30-fold dilution. Therefore, for the intestinal permeability study, the GR extract (100 mg/6 mL), glycyrrhizin (2 mg/6 mL), liquiritin (0.6 mg/6 mL), isoliquiritin (0.15 mg/6 mL), liquiritigenin (0.05 mg/6 mL), and isoliquiritigenin (0.02 mg/6 mL) were used (30-fold dilution).

Pharmacokinetic study of the GR extract

Next, we investigated the glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin plasma concentrations following a single oral administration of the GR extract at a 1 g/kg dose. Considering the glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin contents in the GR extract, equal amounts of glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin were also orally administered at a single dose of 20, 5.8, 1.5, 0.5, and 0.2 mg/kg, respectively, and their plasma concentrations were compared with the corresponding profile obtained from the oral administration of the GR extract.

Glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin plasma concentrations over time and their pharmacokinetic parameters are shown in Fig. 2 and Table 3, respectively. Of the major components in the rats’ plasma, glycyrrhizin was maintained at the highest concentration for 12 h, and the glycyrrhizin plasma concentrations from the GR extract administration were similar for 2 h but lower than that following orally administered glycyrrhizin (20 mg/kg) during the 4-12 h period. Liquiritin and isoliquiritin plasma concentrations gradually decreased at elimination half-lives (T1/2) of 2.13 ± 0.83 h and 4.68 ± 3.09 h, respectively. Moreover, liquiritin and isoliquiritin plasma concentrations from the GR extract administration (1 g/kg) were similar to those following the oral administration of liquiritin and isoliquiritin at the corresponding doses (5.8 and 1.5 mg/kg, respectively). In contrast, liquiritigenin and isoliquiritigenin plasma concentrations from the GR extract administration were similar, which increased for 10 h and maintained. However, liquiritigenin and isoliquiritigenin plasma concentration profiles following a single administration of liquiritigenin (0.5 mg/kg) or isoliquiritigenin (0.2 mg/kg), respectively, differed from the corresponding liquiritigenin and isoliquiritigenin concentrations of the GR extract. Therefore, Cmax, AUC, Tmax, and MRT values of liquiritigenin and isoliquiritigenin from the GR extract differed significantly from those of liquiritigenin and isoliquiritigenin following the administration of a single component (Table 3). These results suggested that isoliquiritigenin and liquiritigenin levels increased during the late intestinal absorption.

Figure 2.Plasma concentration–time profiles of (A) glycyrrhizin, (B) liquiritin, (C) isoliquiritin, (D) liquiritigenin, and (E) isoliquiritigenin following the oral administration of the GR extract (1 g/kg) or an equivalent single component to rats. The following are oral doses of individual component: (A) glycyrrhizin 20 mg/kg, (B) liquiritin 5.8 mg/kg, (C) isoliquiritin 1.5 mg/kg, (D) liquiritigenin 0.5 mg/kg, and (E) isoliquiritigenin 0.2 mg/kg. Data are expressed as mean ± SD (n = 4).

Table 3 Pharmacokinetic parameters of glycyrrhizin and 4 flavonoids in rats

ComponentParametersSingle componentGR extract
GglycyrrhizinCmax (ng/mL)260.4 ± 117.3311.5 ± 154.3
Tmax (h)2.44 ± 1.810.50 ± 0.35
AUC12h (ng·h/mL)1653 ± 368.11191 ± 808.8
T1/2 (h)4.95 ± 1.948.63 ± 8.95
MRT (h)9.08 ± 2.7912.8 ± 11.3
LiquiritinCmax (ng/mL)18.47 ± 2.1926.82 ± 8.50
Tmax (h)0.31 ± 0.130.38 ± 0.14
AUC12h (ng·h/mL)26.71 ± 9.5235.01 ± 5.05
T1/2 (h)1.67 ± 0.652.13 ± 0.83
MRT (h)1.85 ± 0.542.22 ± 0.39
IsoliquiritinCmax (ng/mL)23.00 ± 11.912.29 ± 4.72
Tmax (h)0.31 ± 0.130.31 ± 0.13
AUC12h (ng·h/mL)26.99 ± 10.920.85 ± 7.66
T1/2 (h)4.26 ± 3.944.68 ± 3.09
MRT (h)2.89 ± 0.733.07 ± 0.61
LiquiritigeninCmax (ng/mL)3.07 ± 0.4810.1 ± 3.83*
Tmax (h)0.25 ± 0.011.0 ± 1.15***
AUC12h (ng·h/mL)8.66 ± 0.6340.4 ± 11.7**
T1/2 (h)7.29 ± 3.68NC
MRT (h)4.74 ± 0.808.64 ± 0.53***
IsoliquiritigeninCmax (ng/mL)7.84 ± 5.0116.52 ± 2.66*
Tmax (h)0.31 ± 0.138.13 ± 5.17*
AUC12h (ng·h/mL)8.09 ± 3.1368.70 ± 12.49***
T1/2 (h)2.02 ± 0.26NC
MRT (h)2.07 ± 0.517.91 ± 1.12 ***

Cmax, maximum plasma concentration; Tmax, time to reach Cmax; AUC12h, area under plasma concentration-time curve from zero to 12 h; T1/2, elimination half-life; MRT, mean residence time; NC, not calculated. *p < 0.05, **p < 0.01, ***p < 0.001; statistically significant compared with equivalent single component. Data were expressed as mean ± SD (n = 4).


Intestinal permeability of the GR extract

The apparent permeability (Papp) of glycyrrhizin, liquiritin, and isoliquiritin differed insignificantly when added as a part of the GR extract or an equivalent amount of a single component. However, the Papp of liquiritigenin and isoliquiritigenin was significantly higher when added as part of the GR extract than when added individually. The fold increase in the Papp of liquiritigenin and isoliquiritigenin from the GR extract was 2.2- and 4.8-fold, respectively, compared with that of a single component (Fig. 3).

Figure 3.Apparent permeability (Papp) of glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin from the GR extract (100 mg/6 mL) or an equivalent single component in the rat ileum. The following are the initial concentration of individual component: glycyrrhizin 2 mg/6 mL, liquiritin 0.6 mg/6 mL, isoliquiritin 0.15 mg/6 mL, liquiritigenin 0.05 mg/6 mL, and isoliquiritigenin 0.02 mg/6 mL. *p < 0.05, **p < 0.01, and ***p < 0.001; statistically significant compared with an equivalent single component. Data are expressed as mean ± SD (n = 3).

To investigate the mechanisms underlying the increased Papp of liquiritigenin and isoliquiritigenin from the GR extract, the Papp of isoliquiritigenin in the presence of a major GR extract component was measured. Isoliquiritigenin was selected given that it showed the highest increase in Papp among the tested flavonoids in the GR extract. Fig. 4A shows that adding glycyrrhizin did not change the Papp of isoliquiritigenin. Coexistence of liquiritin, isoliquiritin, and liquiritigenin significantly increased it with fold-changes of 2.2, 5.4, and 2.3, respectively (Fig. 4A), but not vice versa (Fig. 4B). This suggested that liquiritin, isoliquiritin, and liquiritigenin coexistence increased the Papp of isoliquiritigenin, thereby enhancing its absorption and plasma exposure.

Figure 4.(A) Apparent permeability (Papp) of isoliquiritigenin in the absence or presence of glycyrrhizin, liquiritin, isoliquiritin, and liquiritigenin. (B) Papp of liquiritin, isoliquiritin, and liquiritigenin in the absence or presence of isoliquiritigenin in the rat ileum. The following are the initial concentrations of individual component: liquiritin 0.6 mg/6 mL, isoliquiritin 0.15 mg/6 mL, liquiritigenin 0.05 mg/6 mL, and isoliquiritigenin 0.02 mg/6 mL. *p < 0.05, **p < 0.01, and ***p < 0.001; statistically significant compared with an equivalent single component. Data are expressed as mean ± SD (n = 3).

Biotransformation of the GR extract in the rat intestines

Next, we investigated the biotransformation of liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin based on their structural features and previous studies (Ramalingam et al. 2018; Han et al. 2019). After a 2 h incubation of a single component of the GR extract, including liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin, with enterocytes from the rats’ ileal segments, the components’ formation rates were calculated (Fig. 5). Liquiritin transformed into isoliquiritigenin and liquiritigenin, and the liquiritigenin formation rate was 4-fold greater than that of isoliquiritigenin. Isoliquiritin incubation resulted in isoliquiritigenin and liquiritigenin formation with isoliquiritigenin having a 12.2-fold higher formation rate than that of liquiritigenin (Fig. 5A and 5B). Moreover, liquiritigenin or isoliquiritigenin incubation with rat enterocytes resulted in isoliquiritigenin or liquiritigenin formation with a much lower formation rate than that of liquiritin or isoliquiritin (Fig. 5C and 5D). These results suggested that liquiritigenin and isoliquiritigenin were interchangeable with lower formation rates; however, they did not transform into liquiritin or isoliquiritin (Fig. 5E). The major biotransformation processes were from liquiritin to liquiritigenin and from isoliquiritin to isoliquiritigenin. They had similar formation rates. However, the biotransformation process from liquiritin to isoliquiritigenin and from isoliquiritin to liquiritigenin also occurred with lower formation rates (Fig. 5E).

Figure 5.Formation rates of liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin for a 2 h incubation of (A) liquiritin (0.2 mg/mL), (B) isoliquiritin (0.2 mg/mL), (C) liquiritigenin (0.2 mg/mL), and (D) isoliquiritigenin (0.2 mg/mL) with enterocytes from ileal segments. The arrow indicates the transformation process, and the thickness represents the formation rate. Data are expressed as mean ± SD (n = 3).

GR is a widely used herbal medicine for relieving indigestion and promoting detoxification (Gao et al. 2009; Xie et al. 2014). Glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin have been reported as major pharmacological components of GR (Kobayashi et al. 1995; Xie et al. 2014). Liquiritigenin and isoliquiritigenin are particularly known for their chemopreventive, anti-inflammatory, and antioxidative effects in various carcinomas and disease models (Ramalingam et al. 2018).

Previously, Kim et al. (2020) reported that 2 g/kg of GR extract was orally administered to mice without an acute toxicity profile. In the subchronic study, the GR extract (0.05-1 g/kg) was orally administered to mice for four months with insignificant differences in blood pressure, hematological, biochemical, and histological parameters (Kim et al. 2020). For the pharmacological effect, the GR extract (0.1-0.5 g/kg) was orally administered to mice for nine days to investigate the neuroprotective efficacy (Yang et al. 2019). Repeated oral doses of the GR extract (0.075-0.3 g/kg) for seven days showed dose-dependent antioxidative effects in triptolide-induced liver injury in mice (Cao et al. 2017). As a single-dose therapy, the oral administration of the GR extract (1-10 g/kg) attenuates neuronal apoptosis by inhibiting oxidative stress in D-galactose-induced aging rats (Zhou et al. 2017). Based on the above safety and efficacy profile of the orally administered GR extract, we chose 1 g/kg of single oral administration of the GR extract for the pharmacokinetic study.

GR or its extract is considered generally safe for use in food and approved for use in some over-the-counter drugs by the United States Food and Drug Administration (US FDA) (Isbrucker and Burdock 2006). Traditional Chinese Medicine recommends a decoction of 8-15 g of GR for health protection and up to 100 g for diseases. Intake of the GR extract in the range of 150-300 mg/day appear to be commonly used with supplementation, and 1.8 g/day for four weeks was not associated with any toxicity in humans (Omar et al. 2012). Because of the glycyrrhizin-related mineralocorticoid effect, the daily consumption of glycyrrhizin for therapeutic effects should not exceed 500 mg (Omar et al. 2012). The estimated consumption of glycyrrhizin in the GR extract or powder as a food ingredient is 1.62-216 mg/day in the US, which can be used without concern for safety issues (Isbrucker and Burdock 2006). In our case, the dose of 1 g/kg of the GR extract in rats can be converted to a human dose of 164 mg/kg (9.83 g in 60 kg human) according to the simple practice guide for dose conversion from animal to human (Nair and Jacob 2016). The glycyrrhizin content in 9.83 g of the GR extract was calculated to be 196 mg, which was in the acceptable range for human use.

From the pharmacokinetic comparison of pharmacologically active components from the GR extract and individual components, glycyrrhizin, liquiritin, and isoliquiritin had similar pharmacokinetic profiles regardless of the administration formulation. Glycyrrhizin plasma concentrations after single-component administration were higher than those after the GR extract administration at 4-12 h (Fig. 2A). AUC of glycyrrhizin after single-component administration (1653 ± 368.1 ng·h/mL) decreased to 1191 ± 808.8 ng·h/mL after the GR extract administration but was statistically insignificant. This phenomenon was consistent with a previous report (Cantelli-Forti et al. 1994). AUC and renal excretion of glycyrrhizin in rats after oral administration (480 mg/kg) were significantly smaller than those after the GR extract administration (6.3 g/kg), which could be attributed to the interaction between glycyrrhizin and unknown components in the RG extract during intestinal absorption. A similar interaction was also evidenced in human studies (Cantelli-Forti et al. 1994). High-dose administration of the GR extract (6.3 g/kg in rats and 21 g in humans) may produce a higher interaction than that in our case (1 g/kg in rats).

However, when administered in the form of GR extract, liquiritigenin and isoliquiritigenin plasma concentrations increased significantly compared with those of a single component (Fig. 2D and 2E). These results suggested the beneficial interaction of liquiritigenin and isoliquiritigenin with other components by the orally administered GR extract. Because increased liquiritigenin and isoliquiritigenin plasma concentrations distinctively occurred for 6-12 h, we hypothesized that the interaction could occur in the late phase of the intestinal absorption process. Therefore, we investigated glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin permeability from the GR extract, and the equivalent amount of a single component was measured using the rats’ ileum segments. Consistent with the pharmacokinetic results, the Papp of liquiritigenin and isoliquiritigenin from the GR extract increased significantly compared with that of liquiritigenin and isoliquiritigenin individually (Fig. 3). The increased Papp of isoliquiritigenin was exhibited by the coexistence of liquiritin, isoliquiritin, and liquiritigenin, but not vice versa (Fig. 4).

Literature on the biotransformation of isoliquiritigenin and liquiritigenin have been reported previously. Briefly, β-glucosidase hydrolyzes liquiritin in the intestinal flora, and liquiritigenin is absorbed from the jejunum to the colon and conjugated with UDPGA and sulfate or methylated to produce liquiritigenin-glucuronide and sulfoconjugates or methylconjugates in the gut wall enzymes and in the liver (Németh et al. 2003; Zhang et al. 2016; Ma et al. 2018). Additionally, isoliquiritigenin is biosynthetically and structurally interrelated to liquiritigenin (Simmler et al. 2013; Choi et al. 2015). Chalcone isomerase stereochemically converts isoliquiritigenin to liquiritigenin (Jez and Noel 2002). Liquiritigenin and isoliquiritigenin are interchangeable based on pH and temperature, i.e., isoliquiritigenin undergoes rapid cyclization into liquiritigenin at neutral and lower acidic pH, whereas liquiritigenin isomers undergo a reversible reaction to isoliquiritigenin at basic pH (Jez and Noel 2002). These findings suggest that there is a higher presence of isoliquiritigenin at the lower part compared with liquiritigenin with the help of β-glucosidase and chalcone isomerase, and consequently, intestinal absorption of isoliquiritigenin could be increased at the lower part of intestine. Therefore, in this study, the biotransformation process among liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin was investigated using the Ussing chamber system-mounted rat ileum segments based on the increased plasma exposure at 6 h following oral administration (Fig. 2) and the gastrointestinal transit time (Davies and Morris 1993).

The results showed that isoliquiritigenin was formed from isoliquiritin at the highest rate, converted from liquiritigenin at the lower rate, and from liquiritin at the lowest rate (Fig. 5). Therefore, isoliquiritin, liquiritin, and liquiritigenin could be biosources for isoliquiritigenin, consistent with the increase in the Papp of isoliquiritigenin following the coexistence of isoliquiritin, liquiritin, and liquiritigenin. Similarly, isoliquiritin, liquiritin, and isoliquiritigenin could be biosources for liquiritigenin. Furthermore, this process could occur in intestinal enterocytes, and the converted metabolites are absorbed into the plasma. This could be the underlying mechanism for the increased absorption and pharmacokinetic features of liquiritigenin and isoliquiritigenin after the oral administration of the GR extract.

GR contains at least 400 different chemical constituents, including triterpenoid saponins, flavanones, coumarins, and their glycosides (Fujii et al. 2014; Kao et al. 2014). Furthermore, over 25% of the GR components have been identified as active constituents through oral bioavailability, virtual screening, and drug-likeness (Liu et al. 2013). Therefore, the GR extract has been widely investigated for its pharmacological efficacy in human and animal disease models. Additionally, the beneficial interaction of liquiritigenin and isoliquiritigenin from the oral administration of the GR extract via metabolic conversion in intestinal enterocytes and facilitated absorption could provide a rationale for treatment with GR extract rather than with an individual component. However, the beneficial pharmacological efficacy of these interactions needs further investigation since the pharmacological efficacy of the major flavonoids significantly overlapped in their anti-inflammatory, anticancer, cardioprotective, antioxidant, immunoregulatory, and neuroprotective activities (Ramalingam et al. 2018; Qin et al. 2022; Wu et al. 2022; He et al. 2023).

The authors declare that they have no conflict of interest.

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (No. NRF-2020R1I1A3074384 and NRF-2020R1A5A2017323) and by the Korea Institute of Planning and Evaluation for Technology in Food, Agriculture, Forestry and Fisheries (IPET) through Export Promotion Technology Development Program, funded by Ministry of Agriculture, Food and Rural Affairs (MAFRA) (No 316017-3), Republic of Korea.

  1. Cantelli-Forti G, Maffei F, Hrelia P, Bugamelli F, Bernardi M, D'Intino P, Maranesi M, Raggi MA (1994) Interaction of licorice on glycyrrhizin pharmacokinetics. Environ Health Perspect 102(Suppl 9):65-68. doi: 10.1289/ehp.94102s965
    Pubmed KoreaMed CrossRef
  2. Cao LJ, Hou ZY, Li HD, Zhang BK, Fang PF, Xiang DX, Li ZH, Gong H, Deng Y, Ma YX, Tang HB, Yan M (2017) The ethanol extract of licorice (Glycyrrhiza uralensis) protects against triptolide-induced oxidative stress through activation of Nrf2. Evid Based Complement Alternat Med 2017:2752389. doi: 10.1155/2017/2752389
    Pubmed KoreaMed CrossRef
  3. Choi YH, Kim YJ, Chae HS, Chin YW (2015) In vivo gastroprotective effect along with pharmacokinetics, tissue distribution and metabolism of isoliquiritigenin in mice. Planta Med 81:586-593. doi: 10.1055/s-0035-1545914
    Pubmed CrossRef
  4. Cuendet M, Guo J, Luo Y, Chen S, Oteham CP, Moon RC, van Breemen RB, Marler LE, Pezzuto JM (2010) Cancer chemopreventive activity and metabolism of isoliquiritigenin, a compound found in licorice. Cancer Prev Res (Phila) 3:221-232. doi: 10.1158/1940-6207.CAPR-09-0049
    Pubmed KoreaMed CrossRef
  5. Cuendet M, Oteham CP, Moon RC, Pezzuto JM (2006) Quinone reductase induction as a biomarker for cancer chemoprevention. J Nat Prod 69:460-463. doi: 10.1021/np050362q
    Pubmed KoreaMed CrossRef
  6. Davies B, Morris T (1993) Physiological parameters in laboratory animals and humans. Pharm Res 10:1093-1095. doi: 10.1023/a:1018943613122
    Pubmed CrossRef
  7. Dong S, Fan H, Li Q, Wei G, Li Y, Liu C, Si D (2016) LC-MS/MS method for quantification of liquiritigenin in rat plasma: application to pharmacokinetic study of liquiritin. Chin Herb Med 8:53-60. doi: 10.1016/S1674-6384(16)60008-4
    CrossRef
  8. Fujii S, Morinaga O, Uto T, Nomura S, Shoyama Y (2014) Development of a monoclonal antibody-based immunochemical assay for liquiritin and its application to the quality control of licorice products. J Agric Food Chem 62:3377-3383. doi: 10.1021/jf404731z
    Pubmed CrossRef
  9. Gao X, Wang W, Wei S, Li W (2009) [Review of pharmacological effects of Glycyrrhiza radix and its bioactive compounds]. Zhongguo Zhong Yao Za Zhi 34:2695-2700. Chinese
  10. Han YJ, Kang B, Yang EJ, Choi MK, Song IS (2019) Simultaneous determination and pharmacokinetic characterization of glycyrrhizin, isoliquiritigenin, liquiritigenin, and liquiritin in rat plasma following oral administration of Glycyrrhizae Radix extract. Molecules 24:1816. doi: 10.3390/molecules24091816
    Pubmed KoreaMed CrossRef
  11. He R, Ma TT, Gong MX, Xie KL, Wang ZM, Li J (2023) The correlation between pharmacological activity and contents of eight constituents of Glycyrrhiza uralensis Fisch. Heliyon 9:e14570. doi: 10.1016/j.heliyon.2023.e14570
    Pubmed KoreaMed CrossRef
  12. Isbrucker RA, Burdock GA (2006) Risk and safety assessment on the consumption of Licorice root (Glycyrrhiza sp.), its extract and powder as a food ingredient, with emphasis on the pharmacology and toxicology of glycyrrhizin. Regul Toxicol Pharmacol 46:167-192. doi: 10.1016/j.yrtph.2006.06.002
    Pubmed CrossRef
  13. Ishida S, Sakiya Y, Ichikawa T, Taira Z (1992) Dose-dependent pharmacokinetics of glycyrrhizin in rats. Chem Pharm Bull (Tokyo) 40:1917-1920. doi: 10.1248/cpb.40.1917
    Pubmed CrossRef
  14. Jeon JH, Lee J, Park JH, Lee CH, Choi MK, Song IS (2021) Effect of lactic acid bacteria on the pharmacokinetics and metabolism of ginsenosides in mice. Pharmaceutics 13:1496. doi: 10.3390/pharmaceutics13091496
    Pubmed KoreaMed CrossRef
  15. Jez JM, Noel JP (2002) Reaction mechanism of chalcone isomerase. pH dependence, diffusion control, and product binding differences. J Biol Chem 277:1361-1369. doi: 10.1074/jbc.M109224200
    Pubmed CrossRef
  16. Kao TC, Wu CH, Yen GC (2014) Bioactivity and potential health benefits of licorice. J Agric Food Chem 62:542-553. doi: 10.1021/jf404939f
    Pubmed CrossRef
  17. Kim HY, Zuo G, Lee SK, Lim SS (2020) Acute and subchronic toxicity study of nonpolar extract of licorice roots in mice. Food Sci Nutr 8:2242-2250. doi: 10.1002/fsn3.1465
    Pubmed KoreaMed CrossRef
  18. Kobayashi S, Miyamoto T, Kimura I, Kimura M (1995) Inhibitory effect of isoliquiritin, a compound in licorice root, on angiogenesis in vivo and tube formation in vitro. Biol Pharm Bull 18:1382-1386. doi: 10.1248/bpb.18.1382
    Pubmed CrossRef
  19. Lim DY, Pang M, Lee J, Lee J, Jeon JH, Park JH, Choi MK, Song IS (2022) Enhanced bioavailability and hepatoprotective effect of silymarin by preparing silymarin-loaded solid dispersion formulation using freeze-drying method. Arch Pharm Res 45:743-760. doi: 10.1007/s12272-022-01407-0
    Pubmed CrossRef
  20. Lin ZJ, Qiu SX, Wufuer A, Shum L (2005) Simultaneous determination of glycyrrhizin, a marker component in radix Glycyrrhizae, and its major metabolite glycyrrhetic acid in human plasma by LC-MS/MS. J Chromatogr B Analyt Technol Biomed Life Sci 814:201-207. doi: 10.1016/j.jchromb.2004.10.026
    Pubmed CrossRef
  21. Liu H, Wang J, Zhou W, Wang Y, Yang L (2013) Systems approaches and polypharmacology for drug discovery from herbal medicines: an example using licorice. J Ethnopharmacol 146:773-793. doi: 10.1016/j.jep.2013.02.004
    Pubmed CrossRef
  22. Ma L, Zhao Y, Zhang X, Liu T, Han F, Yin R (2018) Characterization of the global metabolic profile of liquiritin in rat plasma, urine, bile and feces based on UHPLC-FT-ICR MS. RSC Adv 8:5945-5952. doi: 10.1039/c7ra12529a
    Pubmed KoreaMed CrossRef
  23. Maggiolini M, Statti G, Vivacqua A, Gabriele S, Rago V, Loizzo M, Menichini F, Amdò S (2002) Estrogenic and antiproliferative activities of isoliquiritigenin in MCF7 breast cancer cells. J Steroid Biochem Mol Biol 82:315-322. doi: 10.1016/s0960-0760(02)00230-3
    Pubmed CrossRef
  24. Mao Y, Peng L, Kang A, Xie T, Xu J, Shen C, Ji J, Di L, Wu H, Shan J (2017) Influence of Jiegeng on pharmacokinetic properties of flavonoids and saponins in Gancao. Molecules 22:1587. doi: 10.3390/molecules22101587
    Pubmed KoreaMed CrossRef
  25. McConnell EL, Basit AW, Murdan S (2008) Measurements of rat and mouse gastrointestinal pH, fluid and lymphoid tissue, and implications for in-vivo experiments. J Pharm Pharmacol 60:63-70. doi: 10.1211/jpp.60.1.0008
    Pubmed CrossRef
  26. Nair AB, Jacob S (2016) A simple practice guide for dose conversion between animals and human. J Basic Clin Pharm 7:27-31. doi: 10.4103/0976-0105.177703
    Pubmed KoreaMed CrossRef
  27. Németh K, Plumb GW, Berrin JG, Juge N, Jacob R, Naim HY, Williamson G, Swallow DM, Kroon PA (2003) Deglycosylation by small intestinal epithelial cell beta-glucosidases is a critical step in the absorption and metabolism of dietary flavonoid glycosides in humans. Eur J Nutr 42:29-42. doi: 10.1007/s00394-003-0397-3
    Pubmed CrossRef
  28. Ng SL, Khaw KY, Ong YS, Goh HP, Kifli N, Teh SP, Ming LC, Kotra V, Goh BH (2021) Licorice: a potential herb in overcoming SARS-CoV-2 infections. J Evid Based Integr Med 26:2515690X21996662. doi: 10.1177/2515690X21996662
    Pubmed KoreaMed CrossRef
  29. Omar HR, Komarova I, El-Ghonemi M, Fathy A, Rashad R, Abdelmalak HD, Yerramadha MR, Ali Y, Helal E, Camporesi EM (2012) Licorice abuse: time to send a warning message. Ther Adv Endocrinol Metab 3:125-138. doi: 10.1177/2042018812454322
    Pubmed KoreaMed CrossRef
  30. Peng F, Du Q, Peng C, Wang N, Tang H, Xie X, Shen J, Chen J (2015) A review: the pharmacology of isoliquiritigenin. Phytother Res 29:969-977. doi: 10.1002/ptr.5348
    Pubmed CrossRef
  31. Qiao X, Ji S, Yu SW, Lin XH, Jin HW, Duan YK, Zhang LR, Guo DA, Ye M (2014) Identification of key licorice constituents which interact with cytochrome P450: evaluation by LC/MS/MS cocktail assay and metabolic profiling. AAPS J 16:101-113. doi: 10.1208/s12248-013-9544-9
    Pubmed KoreaMed CrossRef
  32. Qin J, Chen J, Peng F, Sun C, Lei Y, Chen G, Li G, Yin Y, Lin Z, Wu L, Li J, Liu W, Peng C, Xie X (2022) Pharmacological activities and pharmacokinetics of liquiritin: a review. J Ethnopharmacol 293:115257. doi: 10.1016/j.jep.2022.115257
    Pubmed CrossRef
  33. Ramalingam M, Kim H, Lee Y, Lee YI (2018) Phytochemical and pharmacological role of liquiritigenin and isoliquiritigenin from Radix Glycyrrhizae in human health and disease models. Front Aging Neurosci 10:348. doi: 10.3389/fnagi.2018.00348
    Pubmed KoreaMed CrossRef
  34. Simmler C, Hajirahimkhan A, Lankin DC, Bolton JL, Jones T, Soejarto DD, Chen SN, Pauli GF (2013) Dynamic residual complexity of the isoliquiritigenin-liquiritigenin interconversion during bioassay. J Agric Food Chem 61:2146-2157. doi: 10.1021/jf304445p
    Pubmed KoreaMed CrossRef
  35. Wang T, Ding L, Jin H, Shi R, Li Y, Wu J, Li Y, Zhu L, Ma Y (2016) Simultaneous quantification of catechin, epicatechin, liquiritin, isoliquiritin, liquiritigenin, isoliquiritigenin, piperine and glycyrrhetinic acid in rat plasma by HPLC-MS/MS: application to a pharmacokinetic study of Longhu Rendan pills. Biomed Chromatogr 30:1166-1174. doi: 10.1002/bmc.3662
    Pubmed CrossRef
  36. Wang Y, Yao Y, An R, You L, Wang X (2009) Simultaneous determination of puerarin, daidzein, baicalin, wogonoside and liquiritin of GegenQinlian decoction in rat plasma by ultra-performance liquid chromatography-mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 877:1820-1826. doi: 10.1016/j.jchromb.2009.05.035
    Pubmed CrossRef
  37. Wu Y, Wang Z, Du Q, Zhu Z, Chen T, Xue Y, Wang Y, Zeng Q, Shen C, Jiang C, Liu L, Zhu H, Liu Q (2022) Pharmacological effects and underlying mechanisms of licorice-derived flavonoids. Evid Based Complement Alternat Med 2022:9523071. doi: 10.1155/2022/9523071
    Pubmed KoreaMed CrossRef
  38. Xie J, Zhang Y, Wang W, Hou J (2014) Identification and simultaneous determination of glycyrrhizin, formononetin, glycyrrhetinic acid, liquiritin, isoliquiritigenin, and licochalcone A in licorice by LC-MS/MS. Acta Chromatogr 26:507-516. doi: 10.1556/achrom.26.2014.3.9
    CrossRef
  39. Yang EJ, Park GH, Song KS (2013) Neuroprotective effects of liquiritigenin isolated from licorice roots on glutamate-induced apoptosis in hippocampal neuronal cells. Neurotoxicology 39:114-123. doi: 10.1016/j.neuro.2013.08.012
    Pubmed CrossRef
  40. Yang EJ, Song IS, Song KS (2019) Ethanol extract of Glycyrrhizae Radix modulates the responses of antigen-specific splenocytes in experimental autoimmune encephalomyelitis. Phytomedicine 54:56-65. doi: 10.1016/j.phymed.2018.09.189
    Pubmed CrossRef
  41. Zhang L, Zhao H, Liu Y, Dong H, Lv B, Fang M, Zhao H (2016) Metabolic routes along digestive system of licorice: multicomponent sequential metabolism method in rat. Biomed Chromatogr 30:902-912. doi: 10.1002/bmc.3626
    Pubmed CrossRef
  42. Zhou YZ, Zhao FF, Gao L, Du GH, Zhang X, Qin XM (2017) Licorice extract attenuates brain aging of d-galactose induced rats through inhibition of oxidative stress and attenuation of neuronal apoptosis. RSC Adv 7:47758-47766. doi: 10.1039/C7RA07110H
    CrossRef

Article

Original Research Article

DTT 2024; 3(1): 39-50

Published online March 31, 2024 https://doi.org/10.58502/DTT.23.0029

Copyright © The Pharmaceutical Society of Korea.

Beneficial Interaction between Isoliquiritigenin and Liquiritigenin in their Pharmacokinetics in Rats Following Oral Intake of Glycyrrhizae Radix Extract Compared with Oral Intake of an Individual Component

You Jin Han, Kyung-Sik Song, Im-Sook Song

Department of Pharmacy, Kyungpook National University, Daegu, Korea

Correspondence to:Im-Sook Song, isssong@knu.ac.kr

Received: October 4, 2023; Revised: January 25, 2024; Accepted: January 26, 2024

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Glycyrrhizae Radix (GR) is a widely used herbal medicine. Its pharmacological efficacy depends largely on the composition of bioactive saponins and flavonoids, including glycyrrhizin, liquiritin, isoliquiritin, isoliquiritigenin, and liquiritigenin. This study aimed to compare the pharmacokinetics of glycyrrhizin, liquiritin, isoliquiritin, isoliquiritigenin, and liquiritigenin between orally administered GR extract and an equal amount of a single component. This study also aimed to investigate the intestinal absorption and metabolism of these major pharmacological components in rats. Plasma concentrations of liquiritigenin and isoliquiritigenin from the administered GR extract rapidly decreased for 4 h, increased for 4-10 h, and subsequently stabilized. However, a different profile was observed following a single administration of liquiritigenin or isoliquiritigenin despite a similar dose (0.5 mg/kg or 0.2 mg/kg, respectively). In contrast, the plasma concentrations and pharmacokinetic parameters of glycyrrhizin, liquiritin, and isoliquiritin differed insignificantly from the corresponding results of equivalent doses in rats. Consistent with the pharmacokinetic results, the apparent permeability of liquiritigenin and isoliquiritigenin from the administered GR extract increased by 2.2- and 4.8-fold, respectively, compared with that of individual components. Additionally, isoliquiritigenin was formed from isoliquiritin at the highest rate, converted from liquiritigenin at the lower rate, and converted from liquiritin at the lowest rate in intestinal segments of the rats. Liquiritigenin had a similar process. Therefore, isoliquiritin, liquiritin, and liquiritigenin could be biosources for isoliquiritigenin and liquiritigenin, which could occur in intestinal enterocytes, and the converted metabolites are absorbed into the plasma. Conclusively, the beneficial interaction between liquiritigenin and isoliquiritigenin in orally administered GR extract via metabolic conversion in intestinal enterocytes and enhanced absorption could provide a basis for treatment with GR extract rather than with the individual components.

Keywords: Glycyrrhizae Radix (GR) extract, isoliquiritigenin, liquiritigenin, pharmacokinetics, intestinal permeability, biotransformation

Introduction

Glycyrrhizae Radix (GR, root of licorice) has several pharmacological activities, including chemopreventive, hypoglycemic, anti-inflammatory, and antioxidative activities (Xie et al. 2014; Ng et al. 2021). It has been used as an adjuvant to increase the therapeutic efficacy of other drugs (Qiao et al. 2014) and as a sweetener in traditional medicines, chewing gums, and chocolates (Ishida et al. 1992; Cantelli-Forti et al. 1994; Kobayashi et al. 1995; Lin et al. 2005). Moreover, orally administered GR extract (0.1-0.5 g/kg) in mice for nine days effectively ameliorated interferon-γ-related autoimmune responses and demonstrated neuroprotective efficacy (Yang et al. 2013; Yang et al. 2019). To understand the relationship between the response elicited by GR extract and its pharmacokinetics, bioanalysis of the predominant or pharmacological components of GR extract in biological samples following supplementation and investigation of their pharmacokinetics are necessary.

Glycyrrhizin (Fig. 1A), a major marker component of GR (Kobayashi et al. 1995; Qiao et al. 2014; Peng et al. 2015; Dong et al. 2016), exerts strong neuroprotective effects on experimental autoimmune encephalomyelitis and glutamate-induced apoptosis in neuronal cells and shows therapeutic effects against arthritis, hepatotoxicity, leukemia, allergies, stomach ulcers, and inflammation (Maggiolini et al. 2002). The major active flavonoids of GR, including isoliquiritin, liquiritin, isoliquiritigenin, and liquiritigenin (Gao et al. 2009) (Fig. 1B-1E), are frequently used as antidepressant, anticancer, cardioprotective, antimicrobial, and neuroprotective agents (Cuendet et al. 2006; Cuendet et al. 2010). The pharmacological activities of the major active flavonoids can overlap; however, the main activities could be differentiated. For instance, liquiritin, isoliquiritin, and glycyrrhizic acid mainly and positively influence the anti-inflammatory, anticancer, cardioprotective, and antioxidant activities with different potencies, whereas their deglycosylated metabolites, liquiritigenin, isoliquiritigenin, and glycyrrhetinic acid, mainly and positively influence the immunoregulatory and neuroprotective activities (Qin et al. 2022; Wu et al. 2022; He et al. 2023). Additionally, chemically interconnected liquiritigenin and isoliquiritigenin showed significant efficacy in Alzheimer’s disease, Parkinson’s disease, and other neurodegenerative diseases and disorders in numerous in vitro and in vivo experimental studies (Ramalingam et al. 2018).

Figure 1. Structure of (A) glycyrrhizin, (B) liquritin, (C) isoliquiritin, (D) liquiritigenin, and (E) isoliquiritigenin.

Based on literature search, glycyrrhizin, liquiritin, isoliquiritigenin, and liquiritigenin were selected as the predominant or pharmacological components of the GR extract. We previously developed an analytical method for glycyrrhizin, liquiritin, isoliquiritigenin, and liquiritigenin analysis using a liquid chromatography with tandem mass spectrometry (LC-MS/MS) system with a relatively higher sensitivity (e.g., lower limit of quantification: glycyrrhizin [2 ng/mL] and isoliquiritigenin, liquiritigenin, and liquiritin [0.2 ng/mL each]) with a small volume (50 μL) of plasma samples (Han et al. 2019). Using these analytical methods, glycyrrhizin, isoliquiritigenin, liquiritin, and liquiritigenin plasma concentrations in rats were determined following a single oral GR extract administration at a dose of 1 g/kg (Han et al. 2019). In this study, glycyrrhizin concentration in plasma showed the highest and most stable profile for 12 h. Isoliquiritigenin and liquiritigenin concentrations in plasma were similar and eliminated rapidly after 4 h; however, they rebounded to the initial plasma concentration for 10 h, which could be explained from the biotransformation of liquiritin to isoliquiritigenin (Han et al. 2019). Similarly, co-administration with Jiegeng changed glycyrrhizin and liquiritigenin metabolism in rat fecal lysate, consequently altering their pharmacokinetic profiles (Mao et al. 2017). Besides this biotransformation, compared with the addition of an equal amount of liquiritin alone, liquiritin permeability was much greater when added as a part of the GR extract (Mao et al. 2017), consistent with our case.

Therefore, this study aimed to pharmacokinetically compare GR extract administration with that of an equal amount of a single component contained in the GR extract, including glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin, in rats (Wang et al. 2016). This study also aimed to investigate and compare the intestinal absorption and metabolism of these major pharmacological components in coexisting rats. Isoliquiritin was included in the pharmacokinetic and permeability study to better understand the in vivo biotransformation process.

Materials and Methods

Materials

Glycyrrhizin (purity > 95.0%), isoliquiritigenin (purity > 99.0%), isoliquiritin (purity > 90.0%), liquiritin (purity > 98.0%), liquiritigenin (purity > 97.0%), berberine (purity ≥ 98.0%), dimethyl sulfoxide (DMSO), and Hank’s balanced salt solution (HBSS) were purchased from Sigma-Aldrich (St. Louis, MO, USA). Water, methanol, and other solvents were obtained from J.T. Baker Korea (Seoul, Korea) and TEDIA (Fairfield, OH, USA). All other chemicals were of reagent and analytical grade.

Plant material and extraction

Dried GR (1 kg), obtained from CK Pharm Co. (Seoul, Korea), was extracted with 12 L of 94% ethanol for 3 h and filtered through Whatman Qualitative filter paper grade 1. The filtrate was concentrated using a rotary evaporator (EYELA, Tokyo, Japan) to obtain the GR extract. The voucher specimen (#KNUNPM GR-2015-002) was deposited at the laboratory of Natural Products Medicine at Kyungpook National University (Daegu, Korea).

Animals

Male Sprague–Dawley rats (7 weeks old, 249 ± 6 g) were obtained from Samtako (Osan, Korea) and were acclimated for one week at the animal facility of the College of Pharmacy, Kyungpook National University. Rats were maintained in an environmentally clean room at 21℃-27℃ with 12-h light (09:00-21:00) and relative humidity of 60% ± 5%. Furthermore, they were fasted for 12 h with water ad libitum. The Kyungpook National University Animal Committee approved the protocols and procedures for the animal studies (IACUC No.: KNU-2017-0126, approval date: September 23, 2017).

Pharmacokinetic study

Rats were fasted for 16 h with water ad libitum before the pharmacokinetic experiments. Their femoral arteries were cannulated with a polyethylene tube (PE50, Jungdo, Seoul, Korea) under anesthesia with isoflurane (isoflurane vaporizer set to 2%, with 0.8 L/min oxygen flow).

The GR extract (1 g/kg/2 mL suspended in distilled water containing 10% DMSO) was orally administered to four rats through gavage. The corresponding doses of glycyrrhizin (20 mg/kg/2 mL suspended in distilled water containing 10% DMSO), liquiritin (5.8 mg/kg/2 mL), isoliquiritin (1.5 mg/kg/2 mL), liquiritigenin (0.5 mg/kg/2 mL), or isoliquiritigenin (0.2 mg/kg/2 mL) were orally administered to rats (four rats per component) through gavage. Blood samples (approximately 120 μL) were taken via the femoral artery at 0, 0.25, 0.5, 0.75, 1, 2, 4, 6, 8, 10, and 12 h and centrifuged at 10,000 × g for 1 min at 4℃. Supernatant plasma samples (50 μL each) were stored at −80℃ until analysis.

Intestinal permeability test

Rats were fasted for 16 h. However, they had nonrestricted access to water before the study commenced. They were anesthetized using isoflurane (isoflurane vaporizer set to 2%, with 0.8 L/min oxygen flow), and the proximal part of the ileal segments (approximately 20 cm) were excised, opened along the mesenteric border, and washed in prewarmed HBSS (pH 7.4). Segments were mounted in a tissue holder of a Navicyte Easy Mount Ussing Chamber (Warner Instruments, Holliston, MA, USA) with a 0.76 cm2 surface area. They were acclimated in HBSS for 15 min with continuous oxygenation (95% O2/5% CO2 gas). Intestinal permeability studies were conducted by adding HBSS to both sides of the intestinal segments, which included 1 mL of prewarmed HBSS containing the GR extract or equivalent single component (e.g., GR extract [100 mg/6 mL], glycyrrhizin [2 mg/6 mL], liquiritin [0.6 mg/6 mL], isoliquiritin [0.15 mg/6 mL], liquiritigenin [0.05 mg/6 mL], and isoliquiritigenin [0.02 mg/6 mL]) on the apical side of the ileal segments and 1 mL of prewarmed fresh HBSS on the basal side. Sample aliquots (400 μL) were withdrawn at 30-min intervals for 2 h from the basal side, and an equal volume of prewarmed fresh HBSS was subsequently replenished. During the experiment, carbogen gas (5% CO2/95% O2) was bubbled into the Ussing chambers at 150 drops/min. Samples were stored at −80℃ until analysis.

Next, we investigated the permeability of isoliquiritigenin (0.02 mg/6 mL) in the absence or presence of a single component of the GR extract using the same experimental procedures described above. Briefly, its permeability was assessed by adding 1 mL of prewarmed HBSS containing isoliquiritigenin (0.02 mg/6 mL) in the absence or presence of a single component of the GR extract (e.g., glycyrrhizin [2 mg/6 mL], liquiritin [0.6 mg/6 mL], isoliquiritin [0.15 mg/6 mL], and liquiritigenin [0.05 mg/6 mL]) on the apical side of the ileal segments and 1 mL of prewarmed fresh HBSS on the basal side. Sample aliquots (400 μL) were withdrawn at 30-min intervals for 2 h from the basal side, and an equal volume of prewarmed fresh HBSS was subsequently replenished. Samples were stored at −80℃ until analysis.

Biotransformation of liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin

Rats were fasted for 16 h with water ad libitum before ileal dissection. They were anesthetized using isoflurane (isoflurane vaporizer set to 2%, with 0.8 L/min oxygen flow), and the proximal part of the ileal segments (approximately 20 cm) were excised. The dissected ileal segments were washed using a 10 mL syringe filled with prewarmed HBSS (pH 7.4), and the eluent was vortexed for 1 min followed by centrifugation at 1,000 × g for 5 min at 4℃. The supernatant of the intestinal eluent was used for incubation with isoliquiritigenin, liquiritigenin, and liquiritin.

The ileal segments were mounted onto a Navicyte Easy Mount Ussing Chamber and acclimatized with HBSS for 30 min. Experiments started by changing HBSS with a prewarmed intestinal eluent (1 mL) containing liquiritin, isoliquiritin, liquiritigenin, or isoliquiritigenin (100 μg/mL each) to the apical side of the ileal segments and incubated for 2 h. During the experiment, carbogen gas (5% CO2/95% O2) was bubbled into the Ussing chambers at 150 drops/min. Sample aliquots (50 μL) were mixed with 300 µL methanol containing berberine (0.1 ng/mL), vortexed for 10 min, and centrifuged at 10,000 × g for 10 min at 4℃. An aliquot (10 µL) of the supernatant was injected into the LC-MS/MS system.

LC-MS/MS analysis of glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin

An Agilent 6470 triple quadrupole mass spectrometer equipped with an Agilent Infinity 1260 Infinite II HPLC system (Agilent Technologies, Santa Clara, CA, USA) was used to analyze glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin. The method developed by Han et al. (2019) was used with slight modifications. Table 1 summarizes the MS/MS conditions for the detection of these analytes. The mobile phase consisted of methanol:water (65:35, v/v) with 0.1% formic acid and was eluted in the isocratic mode. A Synergi Polar-RP column (4 µm, 150 × 2 mm; Phenomenex, Torrance, CA, USA) equipped with a polar-RP guard column (4 × 2 mm; Phenomenex) was used for chromatographic separation.

Table 1 . MS/MS parameters for the detection of the analytes and IS.

CompoundsMRM transitions (m/z)Ion modeTR (min)CE (eV)
Precursor ionProduct ion
Liquiritin417.2254.9Negative2.020
Isoliquritin417.2254.9Negative2.420
Liquiritigenin255.1135.0Negative3.215
Isoliquiritigenin255.1135.0Negative4.215
Glycyrrhizin845.5669.4Positive8.735
Berberine (IS)336.1320.0Positive4.930


For the analysis of the glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin contents in the GR extract, 100 mg of the GR extract was diluted 200 times with methanol. The diluted samples (50 μL) were added to 300 µL methanol containing berberine (0.1 ng/mL), vortexed for 10 min, and centrifuged at 10,000 × g for 10 min at 4℃. A supernatant aliquot (10 µL) was injected into the LC-MS/MS system.

For the analysis of glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin plasma concentrations, plasma samples (50 µL) were added to 300 µL methanol containing berberine (0.1 ng/mL), vortexed for 10 min, and centrifuged at 10,000 × g for 10 min at 4℃. A supernatant aliquot (10 µL) was injected into the LC-MS/MS system. For the analysis of glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin in HBSS, HBSS samples (50 µL) were added to 300 µL methanol containing berberine (0.1 ng/mL), vortexed for 10 min, and centrifuged at 10,000 × g for 10 min at 4℃. A supernatant aliquot (10 µL) was injected into the LC-MS/MS system.

Data and statistical analysis

Using noncompartmental analysis (WinNonlin 6.3; Pharsight Corp., Mountain View, CA, USA), the following pharmacokinetic parameters of glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin were calculated: terminal half-life (t1/2), area under the plasma concentration–time curve during the observation period (AUC12h), AUC to infinite time (AUCinf), and mean residence time (MRT). The maximum concentration (Cmax) and the time to reach Cmax (Tmax) were obtained from the experimental data.

To calculate permeability, the transport rate of the analytes (i.e., glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin) was calculated from the slope of the regression line from the mean permeated amounts versus the incubation time plot. The apparent permeability (Papp) was calculated using the following equation (Jeon et al. 2021; Lim et al. 2022):

Papp(cm/s)=transport rate (nmol/s)Initial concentration of analyte (μM)×surface area of membrane (cm2)

The concentrations of the analytes (i.e., liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin) were quantitated, and their formation rates (pmol/mg protein/min) were calculated by dividing the amounts of metabolites formed from the incubation with enterocytes in the rats’ ileal segments by the incubation time.

Student’s t-test (IBM SPSS Statistics, version 26; Armonk, NY, USA) was used to determine the statistical significance of the pharmacokinetic parameters and treatment groups. p-values of < 0.05 were considered statistically significant.

Results

Content of the GR extract

Table 2 summarizes the glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin concentrations in the GR extract. Glycyrrhizin had the highest concentration (2.0%), consistent with that of previous studies (Kobayashi et al. 1995; Lin et al. 2005; Wang et al. 2009; Xie et al. 2014). Liquritin, isoliquiritin, liquiritigenin, and isoliquiritigenin had lower concentrations (0.6%, 0.1%, 0.05%, and 0.03%, respectively).

Table 2 . The contents of glycyrrhizin and 4 flavonoids in the GR extract.

CompoundsContent (mg/g extract)Content (%)
Glycyrrhizin20.2 ± 3.22.0 ± 0.3
Liquiritin5.8 ± 1.10.6 ± 0.1
Isoliquiritin1.5 ± 0.30.1 ± 0.0
Liquiritigenin0.49 ± 0.060.05 ± 0.01
Isoliquiritigenin0.29 ± 0.060.03 ± 0.01


For the pharmacokinetic comparison, we administered 1 g of GR extract/kg to the rats, and the corresponding doses of saponin and flavonoids, 20 mg/kg of glycyrrhizin, 5.8 mg/kg of liquiritin, 1.5 mg/kg of isoliquiritin, 0.5 mg/kg of liquiritigenin, and 0.2 mg/kg of isoliquiritigenin were determined based on the content in the GR extract (Table 2). Additionally, to mimic the intestinal situation that occurred when the GR extract was orally administered to the rats, isoliquiritigenin, liquiritigenin, and liquiritin concentrations were determined based on the oral dose of the GR extract, their content in the GR extract, and the fluid volume of the small intestine. For example, the GR extract was administered at a dose of 1 g/kg/2 mL to rats with stomach and intestinal fluid volumes of 3.38-6.63 and 11.1-16.5 mL, respectively (McConnell et al. 2008), resulting in a 15-30-fold dilution. Therefore, for the intestinal permeability study, the GR extract (100 mg/6 mL), glycyrrhizin (2 mg/6 mL), liquiritin (0.6 mg/6 mL), isoliquiritin (0.15 mg/6 mL), liquiritigenin (0.05 mg/6 mL), and isoliquiritigenin (0.02 mg/6 mL) were used (30-fold dilution).

Pharmacokinetic study of the GR extract

Next, we investigated the glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin plasma concentrations following a single oral administration of the GR extract at a 1 g/kg dose. Considering the glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin contents in the GR extract, equal amounts of glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin were also orally administered at a single dose of 20, 5.8, 1.5, 0.5, and 0.2 mg/kg, respectively, and their plasma concentrations were compared with the corresponding profile obtained from the oral administration of the GR extract.

Glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin plasma concentrations over time and their pharmacokinetic parameters are shown in Fig. 2 and Table 3, respectively. Of the major components in the rats’ plasma, glycyrrhizin was maintained at the highest concentration for 12 h, and the glycyrrhizin plasma concentrations from the GR extract administration were similar for 2 h but lower than that following orally administered glycyrrhizin (20 mg/kg) during the 4-12 h period. Liquiritin and isoliquiritin plasma concentrations gradually decreased at elimination half-lives (T1/2) of 2.13 ± 0.83 h and 4.68 ± 3.09 h, respectively. Moreover, liquiritin and isoliquiritin plasma concentrations from the GR extract administration (1 g/kg) were similar to those following the oral administration of liquiritin and isoliquiritin at the corresponding doses (5.8 and 1.5 mg/kg, respectively). In contrast, liquiritigenin and isoliquiritigenin plasma concentrations from the GR extract administration were similar, which increased for 10 h and maintained. However, liquiritigenin and isoliquiritigenin plasma concentration profiles following a single administration of liquiritigenin (0.5 mg/kg) or isoliquiritigenin (0.2 mg/kg), respectively, differed from the corresponding liquiritigenin and isoliquiritigenin concentrations of the GR extract. Therefore, Cmax, AUC, Tmax, and MRT values of liquiritigenin and isoliquiritigenin from the GR extract differed significantly from those of liquiritigenin and isoliquiritigenin following the administration of a single component (Table 3). These results suggested that isoliquiritigenin and liquiritigenin levels increased during the late intestinal absorption.

Figure 2. Plasma concentration–time profiles of (A) glycyrrhizin, (B) liquiritin, (C) isoliquiritin, (D) liquiritigenin, and (E) isoliquiritigenin following the oral administration of the GR extract (1 g/kg) or an equivalent single component to rats. The following are oral doses of individual component: (A) glycyrrhizin 20 mg/kg, (B) liquiritin 5.8 mg/kg, (C) isoliquiritin 1.5 mg/kg, (D) liquiritigenin 0.5 mg/kg, and (E) isoliquiritigenin 0.2 mg/kg. Data are expressed as mean ± SD (n = 4).

Table 3 . Pharmacokinetic parameters of glycyrrhizin and 4 flavonoids in rats.

ComponentParametersSingle componentGR extract
GglycyrrhizinCmax (ng/mL)260.4 ± 117.3311.5 ± 154.3
Tmax (h)2.44 ± 1.810.50 ± 0.35
AUC12h (ng·h/mL)1653 ± 368.11191 ± 808.8
T1/2 (h)4.95 ± 1.948.63 ± 8.95
MRT (h)9.08 ± 2.7912.8 ± 11.3
LiquiritinCmax (ng/mL)18.47 ± 2.1926.82 ± 8.50
Tmax (h)0.31 ± 0.130.38 ± 0.14
AUC12h (ng·h/mL)26.71 ± 9.5235.01 ± 5.05
T1/2 (h)1.67 ± 0.652.13 ± 0.83
MRT (h)1.85 ± 0.542.22 ± 0.39
IsoliquiritinCmax (ng/mL)23.00 ± 11.912.29 ± 4.72
Tmax (h)0.31 ± 0.130.31 ± 0.13
AUC12h (ng·h/mL)26.99 ± 10.920.85 ± 7.66
T1/2 (h)4.26 ± 3.944.68 ± 3.09
MRT (h)2.89 ± 0.733.07 ± 0.61
LiquiritigeninCmax (ng/mL)3.07 ± 0.4810.1 ± 3.83*
Tmax (h)0.25 ± 0.011.0 ± 1.15***
AUC12h (ng·h/mL)8.66 ± 0.6340.4 ± 11.7**
T1/2 (h)7.29 ± 3.68NC
MRT (h)4.74 ± 0.808.64 ± 0.53***
IsoliquiritigeninCmax (ng/mL)7.84 ± 5.0116.52 ± 2.66*
Tmax (h)0.31 ± 0.138.13 ± 5.17*
AUC12h (ng·h/mL)8.09 ± 3.1368.70 ± 12.49***
T1/2 (h)2.02 ± 0.26NC
MRT (h)2.07 ± 0.517.91 ± 1.12 ***

Cmax, maximum plasma concentration; Tmax, time to reach Cmax; AUC12h, area under plasma concentration-time curve from zero to 12 h; T1/2, elimination half-life; MRT, mean residence time; NC, not calculated. *p < 0.05, **p < 0.01, ***p < 0.001; statistically significant compared with equivalent single component. Data were expressed as mean ± SD (n = 4)..



Intestinal permeability of the GR extract

The apparent permeability (Papp) of glycyrrhizin, liquiritin, and isoliquiritin differed insignificantly when added as a part of the GR extract or an equivalent amount of a single component. However, the Papp of liquiritigenin and isoliquiritigenin was significantly higher when added as part of the GR extract than when added individually. The fold increase in the Papp of liquiritigenin and isoliquiritigenin from the GR extract was 2.2- and 4.8-fold, respectively, compared with that of a single component (Fig. 3).

Figure 3. Apparent permeability (Papp) of glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin from the GR extract (100 mg/6 mL) or an equivalent single component in the rat ileum. The following are the initial concentration of individual component: glycyrrhizin 2 mg/6 mL, liquiritin 0.6 mg/6 mL, isoliquiritin 0.15 mg/6 mL, liquiritigenin 0.05 mg/6 mL, and isoliquiritigenin 0.02 mg/6 mL. *p < 0.05, **p < 0.01, and ***p < 0.001; statistically significant compared with an equivalent single component. Data are expressed as mean ± SD (n = 3).

To investigate the mechanisms underlying the increased Papp of liquiritigenin and isoliquiritigenin from the GR extract, the Papp of isoliquiritigenin in the presence of a major GR extract component was measured. Isoliquiritigenin was selected given that it showed the highest increase in Papp among the tested flavonoids in the GR extract. Fig. 4A shows that adding glycyrrhizin did not change the Papp of isoliquiritigenin. Coexistence of liquiritin, isoliquiritin, and liquiritigenin significantly increased it with fold-changes of 2.2, 5.4, and 2.3, respectively (Fig. 4A), but not vice versa (Fig. 4B). This suggested that liquiritin, isoliquiritin, and liquiritigenin coexistence increased the Papp of isoliquiritigenin, thereby enhancing its absorption and plasma exposure.

Figure 4. (A) Apparent permeability (Papp) of isoliquiritigenin in the absence or presence of glycyrrhizin, liquiritin, isoliquiritin, and liquiritigenin. (B) Papp of liquiritin, isoliquiritin, and liquiritigenin in the absence or presence of isoliquiritigenin in the rat ileum. The following are the initial concentrations of individual component: liquiritin 0.6 mg/6 mL, isoliquiritin 0.15 mg/6 mL, liquiritigenin 0.05 mg/6 mL, and isoliquiritigenin 0.02 mg/6 mL. *p < 0.05, **p < 0.01, and ***p < 0.001; statistically significant compared with an equivalent single component. Data are expressed as mean ± SD (n = 3).

Biotransformation of the GR extract in the rat intestines

Next, we investigated the biotransformation of liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin based on their structural features and previous studies (Ramalingam et al. 2018; Han et al. 2019). After a 2 h incubation of a single component of the GR extract, including liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin, with enterocytes from the rats’ ileal segments, the components’ formation rates were calculated (Fig. 5). Liquiritin transformed into isoliquiritigenin and liquiritigenin, and the liquiritigenin formation rate was 4-fold greater than that of isoliquiritigenin. Isoliquiritin incubation resulted in isoliquiritigenin and liquiritigenin formation with isoliquiritigenin having a 12.2-fold higher formation rate than that of liquiritigenin (Fig. 5A and 5B). Moreover, liquiritigenin or isoliquiritigenin incubation with rat enterocytes resulted in isoliquiritigenin or liquiritigenin formation with a much lower formation rate than that of liquiritin or isoliquiritin (Fig. 5C and 5D). These results suggested that liquiritigenin and isoliquiritigenin were interchangeable with lower formation rates; however, they did not transform into liquiritin or isoliquiritin (Fig. 5E). The major biotransformation processes were from liquiritin to liquiritigenin and from isoliquiritin to isoliquiritigenin. They had similar formation rates. However, the biotransformation process from liquiritin to isoliquiritigenin and from isoliquiritin to liquiritigenin also occurred with lower formation rates (Fig. 5E).

Figure 5. Formation rates of liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin for a 2 h incubation of (A) liquiritin (0.2 mg/mL), (B) isoliquiritin (0.2 mg/mL), (C) liquiritigenin (0.2 mg/mL), and (D) isoliquiritigenin (0.2 mg/mL) with enterocytes from ileal segments. The arrow indicates the transformation process, and the thickness represents the formation rate. Data are expressed as mean ± SD (n = 3).

Discussion

GR is a widely used herbal medicine for relieving indigestion and promoting detoxification (Gao et al. 2009; Xie et al. 2014). Glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin have been reported as major pharmacological components of GR (Kobayashi et al. 1995; Xie et al. 2014). Liquiritigenin and isoliquiritigenin are particularly known for their chemopreventive, anti-inflammatory, and antioxidative effects in various carcinomas and disease models (Ramalingam et al. 2018).

Previously, Kim et al. (2020) reported that 2 g/kg of GR extract was orally administered to mice without an acute toxicity profile. In the subchronic study, the GR extract (0.05-1 g/kg) was orally administered to mice for four months with insignificant differences in blood pressure, hematological, biochemical, and histological parameters (Kim et al. 2020). For the pharmacological effect, the GR extract (0.1-0.5 g/kg) was orally administered to mice for nine days to investigate the neuroprotective efficacy (Yang et al. 2019). Repeated oral doses of the GR extract (0.075-0.3 g/kg) for seven days showed dose-dependent antioxidative effects in triptolide-induced liver injury in mice (Cao et al. 2017). As a single-dose therapy, the oral administration of the GR extract (1-10 g/kg) attenuates neuronal apoptosis by inhibiting oxidative stress in D-galactose-induced aging rats (Zhou et al. 2017). Based on the above safety and efficacy profile of the orally administered GR extract, we chose 1 g/kg of single oral administration of the GR extract for the pharmacokinetic study.

GR or its extract is considered generally safe for use in food and approved for use in some over-the-counter drugs by the United States Food and Drug Administration (US FDA) (Isbrucker and Burdock 2006). Traditional Chinese Medicine recommends a decoction of 8-15 g of GR for health protection and up to 100 g for diseases. Intake of the GR extract in the range of 150-300 mg/day appear to be commonly used with supplementation, and 1.8 g/day for four weeks was not associated with any toxicity in humans (Omar et al. 2012). Because of the glycyrrhizin-related mineralocorticoid effect, the daily consumption of glycyrrhizin for therapeutic effects should not exceed 500 mg (Omar et al. 2012). The estimated consumption of glycyrrhizin in the GR extract or powder as a food ingredient is 1.62-216 mg/day in the US, which can be used without concern for safety issues (Isbrucker and Burdock 2006). In our case, the dose of 1 g/kg of the GR extract in rats can be converted to a human dose of 164 mg/kg (9.83 g in 60 kg human) according to the simple practice guide for dose conversion from animal to human (Nair and Jacob 2016). The glycyrrhizin content in 9.83 g of the GR extract was calculated to be 196 mg, which was in the acceptable range for human use.

From the pharmacokinetic comparison of pharmacologically active components from the GR extract and individual components, glycyrrhizin, liquiritin, and isoliquiritin had similar pharmacokinetic profiles regardless of the administration formulation. Glycyrrhizin plasma concentrations after single-component administration were higher than those after the GR extract administration at 4-12 h (Fig. 2A). AUC of glycyrrhizin after single-component administration (1653 ± 368.1 ng·h/mL) decreased to 1191 ± 808.8 ng·h/mL after the GR extract administration but was statistically insignificant. This phenomenon was consistent with a previous report (Cantelli-Forti et al. 1994). AUC and renal excretion of glycyrrhizin in rats after oral administration (480 mg/kg) were significantly smaller than those after the GR extract administration (6.3 g/kg), which could be attributed to the interaction between glycyrrhizin and unknown components in the RG extract during intestinal absorption. A similar interaction was also evidenced in human studies (Cantelli-Forti et al. 1994). High-dose administration of the GR extract (6.3 g/kg in rats and 21 g in humans) may produce a higher interaction than that in our case (1 g/kg in rats).

However, when administered in the form of GR extract, liquiritigenin and isoliquiritigenin plasma concentrations increased significantly compared with those of a single component (Fig. 2D and 2E). These results suggested the beneficial interaction of liquiritigenin and isoliquiritigenin with other components by the orally administered GR extract. Because increased liquiritigenin and isoliquiritigenin plasma concentrations distinctively occurred for 6-12 h, we hypothesized that the interaction could occur in the late phase of the intestinal absorption process. Therefore, we investigated glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin permeability from the GR extract, and the equivalent amount of a single component was measured using the rats’ ileum segments. Consistent with the pharmacokinetic results, the Papp of liquiritigenin and isoliquiritigenin from the GR extract increased significantly compared with that of liquiritigenin and isoliquiritigenin individually (Fig. 3). The increased Papp of isoliquiritigenin was exhibited by the coexistence of liquiritin, isoliquiritin, and liquiritigenin, but not vice versa (Fig. 4).

Literature on the biotransformation of isoliquiritigenin and liquiritigenin have been reported previously. Briefly, β-glucosidase hydrolyzes liquiritin in the intestinal flora, and liquiritigenin is absorbed from the jejunum to the colon and conjugated with UDPGA and sulfate or methylated to produce liquiritigenin-glucuronide and sulfoconjugates or methylconjugates in the gut wall enzymes and in the liver (Németh et al. 2003; Zhang et al. 2016; Ma et al. 2018). Additionally, isoliquiritigenin is biosynthetically and structurally interrelated to liquiritigenin (Simmler et al. 2013; Choi et al. 2015). Chalcone isomerase stereochemically converts isoliquiritigenin to liquiritigenin (Jez and Noel 2002). Liquiritigenin and isoliquiritigenin are interchangeable based on pH and temperature, i.e., isoliquiritigenin undergoes rapid cyclization into liquiritigenin at neutral and lower acidic pH, whereas liquiritigenin isomers undergo a reversible reaction to isoliquiritigenin at basic pH (Jez and Noel 2002). These findings suggest that there is a higher presence of isoliquiritigenin at the lower part compared with liquiritigenin with the help of β-glucosidase and chalcone isomerase, and consequently, intestinal absorption of isoliquiritigenin could be increased at the lower part of intestine. Therefore, in this study, the biotransformation process among liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin was investigated using the Ussing chamber system-mounted rat ileum segments based on the increased plasma exposure at 6 h following oral administration (Fig. 2) and the gastrointestinal transit time (Davies and Morris 1993).

The results showed that isoliquiritigenin was formed from isoliquiritin at the highest rate, converted from liquiritigenin at the lower rate, and from liquiritin at the lowest rate (Fig. 5). Therefore, isoliquiritin, liquiritin, and liquiritigenin could be biosources for isoliquiritigenin, consistent with the increase in the Papp of isoliquiritigenin following the coexistence of isoliquiritin, liquiritin, and liquiritigenin. Similarly, isoliquiritin, liquiritin, and isoliquiritigenin could be biosources for liquiritigenin. Furthermore, this process could occur in intestinal enterocytes, and the converted metabolites are absorbed into the plasma. This could be the underlying mechanism for the increased absorption and pharmacokinetic features of liquiritigenin and isoliquiritigenin after the oral administration of the GR extract.

GR contains at least 400 different chemical constituents, including triterpenoid saponins, flavanones, coumarins, and their glycosides (Fujii et al. 2014; Kao et al. 2014). Furthermore, over 25% of the GR components have been identified as active constituents through oral bioavailability, virtual screening, and drug-likeness (Liu et al. 2013). Therefore, the GR extract has been widely investigated for its pharmacological efficacy in human and animal disease models. Additionally, the beneficial interaction of liquiritigenin and isoliquiritigenin from the oral administration of the GR extract via metabolic conversion in intestinal enterocytes and facilitated absorption could provide a rationale for treatment with GR extract rather than with an individual component. However, the beneficial pharmacological efficacy of these interactions needs further investigation since the pharmacological efficacy of the major flavonoids significantly overlapped in their anti-inflammatory, anticancer, cardioprotective, antioxidant, immunoregulatory, and neuroprotective activities (Ramalingam et al. 2018; Qin et al. 2022; Wu et al. 2022; He et al. 2023).

Conflict of interest

The authors declare that they have no conflict of interest.

Acknowledgements

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (No. NRF-2020R1I1A3074384 and NRF-2020R1A5A2017323) and by the Korea Institute of Planning and Evaluation for Technology in Food, Agriculture, Forestry and Fisheries (IPET) through Export Promotion Technology Development Program, funded by Ministry of Agriculture, Food and Rural Affairs (MAFRA) (No 316017-3), Republic of Korea.

Fig 1.

Figure 1.Structure of (A) glycyrrhizin, (B) liquritin, (C) isoliquiritin, (D) liquiritigenin, and (E) isoliquiritigenin.
Drug Targets and Therapeutics 2024; 3: 39-50https://doi.org/10.58502/DTT.23.0029

Fig 2.

Figure 2.Plasma concentration–time profiles of (A) glycyrrhizin, (B) liquiritin, (C) isoliquiritin, (D) liquiritigenin, and (E) isoliquiritigenin following the oral administration of the GR extract (1 g/kg) or an equivalent single component to rats. The following are oral doses of individual component: (A) glycyrrhizin 20 mg/kg, (B) liquiritin 5.8 mg/kg, (C) isoliquiritin 1.5 mg/kg, (D) liquiritigenin 0.5 mg/kg, and (E) isoliquiritigenin 0.2 mg/kg. Data are expressed as mean ± SD (n = 4).
Drug Targets and Therapeutics 2024; 3: 39-50https://doi.org/10.58502/DTT.23.0029

Fig 3.

Figure 3.Apparent permeability (Papp) of glycyrrhizin, liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin from the GR extract (100 mg/6 mL) or an equivalent single component in the rat ileum. The following are the initial concentration of individual component: glycyrrhizin 2 mg/6 mL, liquiritin 0.6 mg/6 mL, isoliquiritin 0.15 mg/6 mL, liquiritigenin 0.05 mg/6 mL, and isoliquiritigenin 0.02 mg/6 mL. *p < 0.05, **p < 0.01, and ***p < 0.001; statistically significant compared with an equivalent single component. Data are expressed as mean ± SD (n = 3).
Drug Targets and Therapeutics 2024; 3: 39-50https://doi.org/10.58502/DTT.23.0029

Fig 4.

Figure 4.(A) Apparent permeability (Papp) of isoliquiritigenin in the absence or presence of glycyrrhizin, liquiritin, isoliquiritin, and liquiritigenin. (B) Papp of liquiritin, isoliquiritin, and liquiritigenin in the absence or presence of isoliquiritigenin in the rat ileum. The following are the initial concentrations of individual component: liquiritin 0.6 mg/6 mL, isoliquiritin 0.15 mg/6 mL, liquiritigenin 0.05 mg/6 mL, and isoliquiritigenin 0.02 mg/6 mL. *p < 0.05, **p < 0.01, and ***p < 0.001; statistically significant compared with an equivalent single component. Data are expressed as mean ± SD (n = 3).
Drug Targets and Therapeutics 2024; 3: 39-50https://doi.org/10.58502/DTT.23.0029

Fig 5.

Figure 5.Formation rates of liquiritin, isoliquiritin, liquiritigenin, and isoliquiritigenin for a 2 h incubation of (A) liquiritin (0.2 mg/mL), (B) isoliquiritin (0.2 mg/mL), (C) liquiritigenin (0.2 mg/mL), and (D) isoliquiritigenin (0.2 mg/mL) with enterocytes from ileal segments. The arrow indicates the transformation process, and the thickness represents the formation rate. Data are expressed as mean ± SD (n = 3).
Drug Targets and Therapeutics 2024; 3: 39-50https://doi.org/10.58502/DTT.23.0029

Table 1 MS/MS parameters for the detection of the analytes and IS

CompoundsMRM transitions (m/z)Ion modeTR (min)CE (eV)
Precursor ionProduct ion
Liquiritin417.2254.9Negative2.020
Isoliquritin417.2254.9Negative2.420
Liquiritigenin255.1135.0Negative3.215
Isoliquiritigenin255.1135.0Negative4.215
Glycyrrhizin845.5669.4Positive8.735
Berberine (IS)336.1320.0Positive4.930

Table 2 The contents of glycyrrhizin and 4 flavonoids in the GR extract

CompoundsContent (mg/g extract)Content (%)
Glycyrrhizin20.2 ± 3.22.0 ± 0.3
Liquiritin5.8 ± 1.10.6 ± 0.1
Isoliquiritin1.5 ± 0.30.1 ± 0.0
Liquiritigenin0.49 ± 0.060.05 ± 0.01
Isoliquiritigenin0.29 ± 0.060.03 ± 0.01

Table 3 Pharmacokinetic parameters of glycyrrhizin and 4 flavonoids in rats

ComponentParametersSingle componentGR extract
GglycyrrhizinCmax (ng/mL)260.4 ± 117.3311.5 ± 154.3
Tmax (h)2.44 ± 1.810.50 ± 0.35
AUC12h (ng·h/mL)1653 ± 368.11191 ± 808.8
T1/2 (h)4.95 ± 1.948.63 ± 8.95
MRT (h)9.08 ± 2.7912.8 ± 11.3
LiquiritinCmax (ng/mL)18.47 ± 2.1926.82 ± 8.50
Tmax (h)0.31 ± 0.130.38 ± 0.14
AUC12h (ng·h/mL)26.71 ± 9.5235.01 ± 5.05
T1/2 (h)1.67 ± 0.652.13 ± 0.83
MRT (h)1.85 ± 0.542.22 ± 0.39
IsoliquiritinCmax (ng/mL)23.00 ± 11.912.29 ± 4.72
Tmax (h)0.31 ± 0.130.31 ± 0.13
AUC12h (ng·h/mL)26.99 ± 10.920.85 ± 7.66
T1/2 (h)4.26 ± 3.944.68 ± 3.09
MRT (h)2.89 ± 0.733.07 ± 0.61
LiquiritigeninCmax (ng/mL)3.07 ± 0.4810.1 ± 3.83*
Tmax (h)0.25 ± 0.011.0 ± 1.15***
AUC12h (ng·h/mL)8.66 ± 0.6340.4 ± 11.7**
T1/2 (h)7.29 ± 3.68NC
MRT (h)4.74 ± 0.808.64 ± 0.53***
IsoliquiritigeninCmax (ng/mL)7.84 ± 5.0116.52 ± 2.66*
Tmax (h)0.31 ± 0.138.13 ± 5.17*
AUC12h (ng·h/mL)8.09 ± 3.1368.70 ± 12.49***
T1/2 (h)2.02 ± 0.26NC
MRT (h)2.07 ± 0.517.91 ± 1.12 ***

Cmax, maximum plasma concentration; Tmax, time to reach Cmax; AUC12h, area under plasma concentration-time curve from zero to 12 h; T1/2, elimination half-life; MRT, mean residence time; NC, not calculated. *p < 0.05, **p < 0.01, ***p < 0.001; statistically significant compared with equivalent single component. Data were expressed as mean ± SD (n = 4).


References

  1. Cantelli-Forti G, Maffei F, Hrelia P, Bugamelli F, Bernardi M, D'Intino P, Maranesi M, Raggi MA (1994) Interaction of licorice on glycyrrhizin pharmacokinetics. Environ Health Perspect 102(Suppl 9):65-68. doi: 10.1289/ehp.94102s965
    Pubmed KoreaMed CrossRef
  2. Cao LJ, Hou ZY, Li HD, Zhang BK, Fang PF, Xiang DX, Li ZH, Gong H, Deng Y, Ma YX, Tang HB, Yan M (2017) The ethanol extract of licorice (Glycyrrhiza uralensis) protects against triptolide-induced oxidative stress through activation of Nrf2. Evid Based Complement Alternat Med 2017:2752389. doi: 10.1155/2017/2752389
    Pubmed KoreaMed CrossRef
  3. Choi YH, Kim YJ, Chae HS, Chin YW (2015) In vivo gastroprotective effect along with pharmacokinetics, tissue distribution and metabolism of isoliquiritigenin in mice. Planta Med 81:586-593. doi: 10.1055/s-0035-1545914
    Pubmed CrossRef
  4. Cuendet M, Guo J, Luo Y, Chen S, Oteham CP, Moon RC, van Breemen RB, Marler LE, Pezzuto JM (2010) Cancer chemopreventive activity and metabolism of isoliquiritigenin, a compound found in licorice. Cancer Prev Res (Phila) 3:221-232. doi: 10.1158/1940-6207.CAPR-09-0049
    Pubmed KoreaMed CrossRef
  5. Cuendet M, Oteham CP, Moon RC, Pezzuto JM (2006) Quinone reductase induction as a biomarker for cancer chemoprevention. J Nat Prod 69:460-463. doi: 10.1021/np050362q
    Pubmed KoreaMed CrossRef
  6. Davies B, Morris T (1993) Physiological parameters in laboratory animals and humans. Pharm Res 10:1093-1095. doi: 10.1023/a:1018943613122
    Pubmed CrossRef
  7. Dong S, Fan H, Li Q, Wei G, Li Y, Liu C, Si D (2016) LC-MS/MS method for quantification of liquiritigenin in rat plasma: application to pharmacokinetic study of liquiritin. Chin Herb Med 8:53-60. doi: 10.1016/S1674-6384(16)60008-4
    CrossRef
  8. Fujii S, Morinaga O, Uto T, Nomura S, Shoyama Y (2014) Development of a monoclonal antibody-based immunochemical assay for liquiritin and its application to the quality control of licorice products. J Agric Food Chem 62:3377-3383. doi: 10.1021/jf404731z
    Pubmed CrossRef
  9. Gao X, Wang W, Wei S, Li W (2009) [Review of pharmacological effects of Glycyrrhiza radix and its bioactive compounds]. Zhongguo Zhong Yao Za Zhi 34:2695-2700. Chinese
  10. Han YJ, Kang B, Yang EJ, Choi MK, Song IS (2019) Simultaneous determination and pharmacokinetic characterization of glycyrrhizin, isoliquiritigenin, liquiritigenin, and liquiritin in rat plasma following oral administration of Glycyrrhizae Radix extract. Molecules 24:1816. doi: 10.3390/molecules24091816
    Pubmed KoreaMed CrossRef
  11. He R, Ma TT, Gong MX, Xie KL, Wang ZM, Li J (2023) The correlation between pharmacological activity and contents of eight constituents of Glycyrrhiza uralensis Fisch. Heliyon 9:e14570. doi: 10.1016/j.heliyon.2023.e14570
    Pubmed KoreaMed CrossRef
  12. Isbrucker RA, Burdock GA (2006) Risk and safety assessment on the consumption of Licorice root (Glycyrrhiza sp.), its extract and powder as a food ingredient, with emphasis on the pharmacology and toxicology of glycyrrhizin. Regul Toxicol Pharmacol 46:167-192. doi: 10.1016/j.yrtph.2006.06.002
    Pubmed CrossRef
  13. Ishida S, Sakiya Y, Ichikawa T, Taira Z (1992) Dose-dependent pharmacokinetics of glycyrrhizin in rats. Chem Pharm Bull (Tokyo) 40:1917-1920. doi: 10.1248/cpb.40.1917
    Pubmed CrossRef
  14. Jeon JH, Lee J, Park JH, Lee CH, Choi MK, Song IS (2021) Effect of lactic acid bacteria on the pharmacokinetics and metabolism of ginsenosides in mice. Pharmaceutics 13:1496. doi: 10.3390/pharmaceutics13091496
    Pubmed KoreaMed CrossRef
  15. Jez JM, Noel JP (2002) Reaction mechanism of chalcone isomerase. pH dependence, diffusion control, and product binding differences. J Biol Chem 277:1361-1369. doi: 10.1074/jbc.M109224200
    Pubmed CrossRef
  16. Kao TC, Wu CH, Yen GC (2014) Bioactivity and potential health benefits of licorice. J Agric Food Chem 62:542-553. doi: 10.1021/jf404939f
    Pubmed CrossRef
  17. Kim HY, Zuo G, Lee SK, Lim SS (2020) Acute and subchronic toxicity study of nonpolar extract of licorice roots in mice. Food Sci Nutr 8:2242-2250. doi: 10.1002/fsn3.1465
    Pubmed KoreaMed CrossRef
  18. Kobayashi S, Miyamoto T, Kimura I, Kimura M (1995) Inhibitory effect of isoliquiritin, a compound in licorice root, on angiogenesis in vivo and tube formation in vitro. Biol Pharm Bull 18:1382-1386. doi: 10.1248/bpb.18.1382
    Pubmed CrossRef
  19. Lim DY, Pang M, Lee J, Lee J, Jeon JH, Park JH, Choi MK, Song IS (2022) Enhanced bioavailability and hepatoprotective effect of silymarin by preparing silymarin-loaded solid dispersion formulation using freeze-drying method. Arch Pharm Res 45:743-760. doi: 10.1007/s12272-022-01407-0
    Pubmed CrossRef
  20. Lin ZJ, Qiu SX, Wufuer A, Shum L (2005) Simultaneous determination of glycyrrhizin, a marker component in radix Glycyrrhizae, and its major metabolite glycyrrhetic acid in human plasma by LC-MS/MS. J Chromatogr B Analyt Technol Biomed Life Sci 814:201-207. doi: 10.1016/j.jchromb.2004.10.026
    Pubmed CrossRef
  21. Liu H, Wang J, Zhou W, Wang Y, Yang L (2013) Systems approaches and polypharmacology for drug discovery from herbal medicines: an example using licorice. J Ethnopharmacol 146:773-793. doi: 10.1016/j.jep.2013.02.004
    Pubmed CrossRef
  22. Ma L, Zhao Y, Zhang X, Liu T, Han F, Yin R (2018) Characterization of the global metabolic profile of liquiritin in rat plasma, urine, bile and feces based on UHPLC-FT-ICR MS. RSC Adv 8:5945-5952. doi: 10.1039/c7ra12529a
    Pubmed KoreaMed CrossRef
  23. Maggiolini M, Statti G, Vivacqua A, Gabriele S, Rago V, Loizzo M, Menichini F, Amdò S (2002) Estrogenic and antiproliferative activities of isoliquiritigenin in MCF7 breast cancer cells. J Steroid Biochem Mol Biol 82:315-322. doi: 10.1016/s0960-0760(02)00230-3
    Pubmed CrossRef
  24. Mao Y, Peng L, Kang A, Xie T, Xu J, Shen C, Ji J, Di L, Wu H, Shan J (2017) Influence of Jiegeng on pharmacokinetic properties of flavonoids and saponins in Gancao. Molecules 22:1587. doi: 10.3390/molecules22101587
    Pubmed KoreaMed CrossRef
  25. McConnell EL, Basit AW, Murdan S (2008) Measurements of rat and mouse gastrointestinal pH, fluid and lymphoid tissue, and implications for in-vivo experiments. J Pharm Pharmacol 60:63-70. doi: 10.1211/jpp.60.1.0008
    Pubmed CrossRef
  26. Nair AB, Jacob S (2016) A simple practice guide for dose conversion between animals and human. J Basic Clin Pharm 7:27-31. doi: 10.4103/0976-0105.177703
    Pubmed KoreaMed CrossRef
  27. Németh K, Plumb GW, Berrin JG, Juge N, Jacob R, Naim HY, Williamson G, Swallow DM, Kroon PA (2003) Deglycosylation by small intestinal epithelial cell beta-glucosidases is a critical step in the absorption and metabolism of dietary flavonoid glycosides in humans. Eur J Nutr 42:29-42. doi: 10.1007/s00394-003-0397-3
    Pubmed CrossRef
  28. Ng SL, Khaw KY, Ong YS, Goh HP, Kifli N, Teh SP, Ming LC, Kotra V, Goh BH (2021) Licorice: a potential herb in overcoming SARS-CoV-2 infections. J Evid Based Integr Med 26:2515690X21996662. doi: 10.1177/2515690X21996662
    Pubmed KoreaMed CrossRef
  29. Omar HR, Komarova I, El-Ghonemi M, Fathy A, Rashad R, Abdelmalak HD, Yerramadha MR, Ali Y, Helal E, Camporesi EM (2012) Licorice abuse: time to send a warning message. Ther Adv Endocrinol Metab 3:125-138. doi: 10.1177/2042018812454322
    Pubmed KoreaMed CrossRef
  30. Peng F, Du Q, Peng C, Wang N, Tang H, Xie X, Shen J, Chen J (2015) A review: the pharmacology of isoliquiritigenin. Phytother Res 29:969-977. doi: 10.1002/ptr.5348
    Pubmed CrossRef
  31. Qiao X, Ji S, Yu SW, Lin XH, Jin HW, Duan YK, Zhang LR, Guo DA, Ye M (2014) Identification of key licorice constituents which interact with cytochrome P450: evaluation by LC/MS/MS cocktail assay and metabolic profiling. AAPS J 16:101-113. doi: 10.1208/s12248-013-9544-9
    Pubmed KoreaMed CrossRef
  32. Qin J, Chen J, Peng F, Sun C, Lei Y, Chen G, Li G, Yin Y, Lin Z, Wu L, Li J, Liu W, Peng C, Xie X (2022) Pharmacological activities and pharmacokinetics of liquiritin: a review. J Ethnopharmacol 293:115257. doi: 10.1016/j.jep.2022.115257
    Pubmed CrossRef
  33. Ramalingam M, Kim H, Lee Y, Lee YI (2018) Phytochemical and pharmacological role of liquiritigenin and isoliquiritigenin from Radix Glycyrrhizae in human health and disease models. Front Aging Neurosci 10:348. doi: 10.3389/fnagi.2018.00348
    Pubmed KoreaMed CrossRef
  34. Simmler C, Hajirahimkhan A, Lankin DC, Bolton JL, Jones T, Soejarto DD, Chen SN, Pauli GF (2013) Dynamic residual complexity of the isoliquiritigenin-liquiritigenin interconversion during bioassay. J Agric Food Chem 61:2146-2157. doi: 10.1021/jf304445p
    Pubmed KoreaMed CrossRef
  35. Wang T, Ding L, Jin H, Shi R, Li Y, Wu J, Li Y, Zhu L, Ma Y (2016) Simultaneous quantification of catechin, epicatechin, liquiritin, isoliquiritin, liquiritigenin, isoliquiritigenin, piperine and glycyrrhetinic acid in rat plasma by HPLC-MS/MS: application to a pharmacokinetic study of Longhu Rendan pills. Biomed Chromatogr 30:1166-1174. doi: 10.1002/bmc.3662
    Pubmed CrossRef
  36. Wang Y, Yao Y, An R, You L, Wang X (2009) Simultaneous determination of puerarin, daidzein, baicalin, wogonoside and liquiritin of GegenQinlian decoction in rat plasma by ultra-performance liquid chromatography-mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 877:1820-1826. doi: 10.1016/j.jchromb.2009.05.035
    Pubmed CrossRef
  37. Wu Y, Wang Z, Du Q, Zhu Z, Chen T, Xue Y, Wang Y, Zeng Q, Shen C, Jiang C, Liu L, Zhu H, Liu Q (2022) Pharmacological effects and underlying mechanisms of licorice-derived flavonoids. Evid Based Complement Alternat Med 2022:9523071. doi: 10.1155/2022/9523071
    Pubmed KoreaMed CrossRef
  38. Xie J, Zhang Y, Wang W, Hou J (2014) Identification and simultaneous determination of glycyrrhizin, formononetin, glycyrrhetinic acid, liquiritin, isoliquiritigenin, and licochalcone A in licorice by LC-MS/MS. Acta Chromatogr 26:507-516. doi: 10.1556/achrom.26.2014.3.9
    CrossRef
  39. Yang EJ, Park GH, Song KS (2013) Neuroprotective effects of liquiritigenin isolated from licorice roots on glutamate-induced apoptosis in hippocampal neuronal cells. Neurotoxicology 39:114-123. doi: 10.1016/j.neuro.2013.08.012
    Pubmed CrossRef
  40. Yang EJ, Song IS, Song KS (2019) Ethanol extract of Glycyrrhizae Radix modulates the responses of antigen-specific splenocytes in experimental autoimmune encephalomyelitis. Phytomedicine 54:56-65. doi: 10.1016/j.phymed.2018.09.189
    Pubmed CrossRef
  41. Zhang L, Zhao H, Liu Y, Dong H, Lv B, Fang M, Zhao H (2016) Metabolic routes along digestive system of licorice: multicomponent sequential metabolism method in rat. Biomed Chromatogr 30:902-912. doi: 10.1002/bmc.3626
    Pubmed CrossRef
  42. Zhou YZ, Zhao FF, Gao L, Du GH, Zhang X, Qin XM (2017) Licorice extract attenuates brain aging of d-galactose induced rats through inhibition of oxidative stress and attenuation of neuronal apoptosis. RSC Adv 7:47758-47766. doi: 10.1039/C7RA07110H
    CrossRef

Stats or Metrics

Share this article on :

  • line