WO2001058410A2 - A method of treating colon cancer by administering apigenin, luteolin, diosmetin and crysin - Google Patents

A method of treating colon cancer by administering apigenin, luteolin, diosmetin and crysin Download PDF

Info

Publication number
WO2001058410A2
WO2001058410A2 PCT/US2001/004656 US0104656W WO0158410A2 WO 2001058410 A2 WO2001058410 A2 WO 2001058410A2 US 0104656 W US0104656 W US 0104656W WO 0158410 A2 WO0158410 A2 WO 0158410A2
Authority
WO
WIPO (PCT)
Prior art keywords
chrysin
cells
udp
glucuronosyltransferase
glucuronidation
Prior art date
Application number
PCT/US2001/004656
Other languages
French (fr)
Other versions
WO2001058410A3 (en
Inventor
Thomas Walle
Perry V. Halushka
Original Assignee
Musc Foundation For Research Development
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Musc Foundation For Research Development filed Critical Musc Foundation For Research Development
Priority to AU2001236982A priority Critical patent/AU2001236982A1/en
Publication of WO2001058410A2 publication Critical patent/WO2001058410A2/en
Publication of WO2001058410A3 publication Critical patent/WO2001058410A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 

Definitions

  • Caco-2 cell line As emphasized during the continuing development of the Caco-2 cell line as a model for human intestinal abso ⁇ tion, these cells in culture morphologically resemble small intestine abso ⁇ tive cells with many of its typical enzymes and transporters (1-4).
  • Caco-2 cells in the past have not been considered to express high levels of drug metabolizing enzymes (5, 6), conjugation reactions have previously been described. This includes sulfation of dopamine and »-nitrophenol (7) and L-a- methyldopa (8).
  • the model substrate >-nitrophenol has also been shown to be glucuroni dated by Caco-2 cells (5, 9), as has L-a-methyldopa (8).
  • UDP-Glucuronosyltransferase is a superfamily of enzymes that catalyze the glucuronidation of both endogenous toxins, e.g. bilirubin, as well as xenobiotics such as carcinogens, in general rendering them biologically inactive (Burchell and Coughtrie, 1989; Miners and Mackenzie, 1991; Mackenzie et al., 1997; de Wildt et al., 1999; Radominska-Pandya et al., 1999).
  • endogenous toxins e.g. bilirubin
  • xenobiotics such as carcinogens
  • the invention provides a method of treating any condition of elevated levels of unconjugated bilirubin in adults or children, such as Gilbert's syndrome or liver cirrhosis.
  • the method calls for the administration of chrysin or chrysin analogs via dietary or other means.
  • a method of increasing the expression of UDP-glucuronosyltransferase in a subject comprising administering to the subject an amount of chrysin effective to increase the UDP-glucuronosyltransferase.
  • Upregulation of the expression of UDP-glucuronosyltransferase levels is expected to be beneficial in a number of areas: 1) increased detoxification of ingested carcinogens, such as the cooked- food mutagens (chrysin is an aromatase inhibitor in cells); 2) decreasing the conversion of androgens to estrogens, potentially building muscle mass or for prevention of breast cancer in women.
  • UDP-glucuronosyltransferase in the method of upregulating UGT, can be a UGT1 A isoform.
  • the UGT1 A isoform can be UGT1 Al .
  • the UGT can be UGT1A9.
  • a method of treating colon cancer comprising administering an effective amount of a flavone, whereby the administration of the flavone treats the colon cancer.
  • the flavone can be a 5,7-dihydroxyflavone, for example one selected from the group consisting of chrysin, apigenin, luteolin and diosmetin.
  • Chrysin analogs can be used in the methods and compositions of the invention.
  • a chrysin analog having a flavonoid structure is contemplated.
  • a chrysin analog having a flavone structure is contemplated.
  • a chrysin analog having a 5,7-substituted flavone structure is further contemplated.
  • Chrysin and its analogs can be administered as a dietary supplement or in a food. Examples of chrysin analogs having a 5,7- substituted flavone
  • Fig. 1 Chemical structures of chrysin and quercetin.
  • Fig. 3 Chrysin glucuronidation by Caco-2 cell homogenates prepared from untreated cells (u) and cells pretreated with 50 ⁇ M chrysin for 4 days (s). Mean values ⁇ SEM from 3 experiments are shown. The error bars for the untreated cells are contained within the symbols. Data were fit to the Michaelis-Menten equation.
  • Fig. 4 Immunoblot analysis of Caco-2 cell microsomes from cells pretreated with 50 mM chrysin for 4 days (lanes 4 and 6, 50 and 100 mg loaded) and from untreated Control cells (lanes 3 and 5, 50 and 100 mg loaded). UGT1A6 microsomal protein (50 mg loaded) was used as a positive control in lanes 1 and 2.
  • Fig. 7 Chrysin glucuronidation by Hep G2 cell homogenates prepared from untreated cells (m) and cells pretreated with 25 ⁇ M chrysin for 3 days (1). Mean values from 2 experiments are shown. The data were fitted to the Michaelis-Menten equation using Microsoft Excel.
  • Fig. 8 Western blot analysis of recombinant UGT isoforms (2B7, 1 Al and 1 A6; lanes 1, 2 and 3, respectively) and microsomes from untreated (Control) Hep G2 cells (lane 4) and cells pretreated with 25 ⁇ M chrysin for 3 days (lane 5).
  • Four different immunoblots were done with antibodies specific for UGT 1A, 1A6, 2B7 and 1A1, respectively.
  • the arrows designate the UGT bands.
  • Fig. 9 Northern blot analysis of UGTl Al mRNA expression in Hep G2 cells.
  • the blots were then stripped and hybridized to a GAPDH cDNA probe (lower panel) to confirm equal sample loading for control and chrysin-treated samples on each blot.
  • Fig. 10 Bilirubin glucuronidation by Hep G2 homogenates prepared from untreated cells (m) and cells pretreated with 25 ⁇ M chrysin for 3 days (1). Mean values from 2 experiments are shown. The data were fitted to the Michaelis-Menten equation using Microsoft Excel. Significantly higher than Control, p ⁇ 0.05.
  • a or “an” can mean one or more than one.
  • a cell can mean a single cell or multiple cells.
  • the invention provides a method of treating any condition of elevated levels of unconjugated bilirubin in adults or children, such as Gilbert's syndrome or liver cirrhosis.
  • the method calls for the administration of chrysin or chrysin analogs via dietary or other means.
  • a method of treating jaundice in a neonate comprising administering to the neonate an amount of chrysin effective to treat jaundice.
  • a method of treating hyperbilirubinemia in a neonate comprising administering to the neonate an amount of chrysin effective to reduce the amount of bilirubin in the neonate.
  • a method of increasing the expression of UDP-glucuronosyltransferase in a subject comprising administering to the subject an amount of chrysin effective to increase the UDP-glucuronosyltransferase.
  • Upregulation of the expression of UDP-glucuronosyltransferase levels is expected to be beneficial in a number of areas: 1) increased detoxification of ingested carcinogens, such as the cooked-food mutagens (chrysin is an aromatase inhibitor in cells); 2) decreasing the conversion of androgens to estrogens, potentially building muscle mass or for prevention of breast cancer in women.
  • upregulation of the expression of UDP- glucuronosyltransferase levels is associated with neutralization of colon carcinogens which include, but are not limited to, N-OH-PhlP.
  • UDP-glucuronosyltransferase in the method of upregulating UGT, can be a UGTl A isoform.
  • the UGTl A isoform can be UGTl Al .
  • the UGT can be UGTl A9.
  • a method of treating colon cancer comprising administering an effective amount of a flavone, whereby the administration of the flavone treats the colon cancer.
  • the flavone can be a 5,7-dihydroxyflavone, for example one selected from the group consisting of chrysin, apigenin, luteolin and diosmetin.
  • Chrysin analogs can be used in the methods and compositions of the invention.
  • a chrysin analog having a flavonoid structure is contemplated.
  • a chrysin analog having a flavone structure is contemplated.
  • a chrysin analog having a 5,7-substituted flavone structure is further contemplated.
  • Chrysin and its analogs can be administered as a dietary supplement or in a food.
  • Examples of chrysin analogs having a 5,7- substituted flavone structure include, but are not limited to, quercetin, apigenin, luteolin and diosmetin.
  • compositions of the present invention are dietary supplements and can thus be dietary supplements.
  • fine powders or granules may contain
  • diluting, dispersing, and/or surface active agents may be presented in water or in a syrup, in capsules or sachets in the dry state, or in a nonaqueous solution or suspension
  • suspending agents may be included, in tablets wherein binders and lubricants
  • flavoring, preserving, suspending, thickening, or emulsifying agents may be included.
  • Tablets and granules are preferred oral administration forms, and these may be coated.
  • intestinal UGT may be important for the bioavailability of carcinogens and other toxic
  • protease inhibitors were purchased from Sigma Chemical Co. (St. Louis, MO).
  • Trifluoroacetic acid was of spectrophotometric grade from Aldrich Chemical Co.
  • HBSS Hanks' balanced salts solution
  • HRP-conjugated goat anti-rabbit IgG was purchased from HRP-conjugated goat anti-rabbit IgG.
  • Substrate was from Pierce (Rockford, IL). Electrophoresis and blotting supplies and prestained molecular weight markers were purchased from Bio-Rad Laboratories
  • the human colon adenocarcinoma cell line Caco-2 was obtained from
  • the cells were used at passage 35-75.
  • Control cells were treated with the same
  • the cells were digested with 0.5 M NaOH and analyzed for protein content (15).
  • Caco-2 cells were plated in 100-mm petri dishes. On day 6 the cells were
  • the cell monolayers were washed twice with HBSS, scraped off the
  • chrysin in the chrysin-pretreated cell homogenate were ⁇ 0.5 mM, as determined by HPLC.
  • the S 10 fraction was further centrifuged at 100,000g for 60 min at 4°C.
  • microsomal pellets were resuspended in 300 ⁇ l of homogenization buffer with protease
  • microsomes were stored in aliquots at -80 °C.
  • Microsomal samples were heated at 90 °C for 5 min with an equal volume of
  • reaction mixtures 500 ⁇ l were incubated at 37°C for 60 min.
  • Caco-2 cells were cultured as above and subcultured at a seeding density of
  • quercetin or DMSO was added on days 2, 4 and 6 after each subculturing. After 5
  • homogenate was estimated to be about 0.8 mM.
  • microsomes of pretreated and untreated cells were prepared and an antibody, under
  • flavonoids for a period of 5 weeks.
  • quercetin the most common
  • Flavonoid in our diet (18) was also studied. Flavonoid concentrations of only 10 mM
  • quercetin the most abundant flavonoid in our diet (18),
  • UGT1A1 UGT1A3 (27), UGT1A8
  • polyaromatic hydrocarbons i.e. may not involve the Ah receptor.
  • isoflavone phytoestrogens a site-directed mutagenesis study. Environ. Health Perspect.
  • Flavonoids, potent inhibitors of the human P-form phenolsulfotransferase potential
  • human intestinal cell line Induction of UDP-glucuronosyltransferase 1*6. Biochem.
  • UDP-glucuronosyltransferase (UGT2B15) with activity toward steroid and xenobiotic
  • UDP-glucuronosyltransferases have long been known to be
  • UGTIAI was confirmed by Northern analyses of total RNA as well as mRNA, using a
  • chrysin was found to be a high affinity substrate for UGTIAI (K m 0.35 ⁇ M).
  • contraceptive drug ethinylestradiol two of the best known and specific UGTIAI
  • Example 1 demonstrated efficient induction of one or several isoforms of the
  • induction response is different in a hepatic as compared to an intestinal cell line.
  • Hep G2 cells were obtained from American Type Culture Collection (Rockville, 1995).
  • the cells were maintained in Williams' Medium E with 10% fetal bovine serum,
  • chrysin or solvent for 3 days were rinsed with phosphate-buffered saline and harvested
  • the cells were sonified for 3 x 5 sec. The homogenates were stored in aliquots at -
  • Bilirubin sample HPLC analyses also used a Symmetry C18
  • microsomes were prepared by centrifugation of the cell
  • liver as well as recombinant standard proteins were heated at 90°C for 5 min with an
  • anti-UGTl Al was prepared and used as recently described (Ritter et al., 1999) with the
  • RNAzol B Tel-Test, Inc., Friendswood, TX
  • RNA was precipitated from the aqueous phase with isopropanol and washed with
  • RNAzol 6 ml
  • RNA sample loading buffer 20 ⁇ g RNA was loaded on a 1%
  • mm dishes of confluent cells were used for each preparation.
  • the mRNA was
  • Chrysin and bilirubin (0.1-100 ⁇ M) were used as potential substrates for
  • UGT isoforms i.e., 1 Al, 1 A4, 1 A6 and 1A9, supplied as microsomes of
  • liver microsomes prepared by standard differential centrifugation (La Du et al., 1972).
  • the flavonoid chrysin is efficiently metabolized by human Hep G2 cells both by
  • V max value high 360 pmol/min/mg protein.
  • Fig. 10 Total glucuronidation of bilirubin by the cell homogenates is shown in Fig. 10.
  • the oral contraceptive drug ethinylestradiol has also been shown to be a
  • Example 1 demonstrated induction of glucuronidation when the human
  • intestinal cell line Caco-2 was treated with chrysin, suggesting the possibility that the
  • molecular weight was also induced, which may be a splice variant of UGTIAI.
  • chrysin may also be induced by chrysin.
  • chrysin or other flavonoids as substrates. These include UGTl A3 (Green et al., 1998),
  • homogenate is about 5 times higher than by recombinant UGTIAI may potentially be
  • UGTIAI The best known substrate for UGTIAI is the endogenous toxin bilirubin (Chowdhury
  • V max were very similar for bilirubin and chrysin glucuronidation.
  • Phenobarbital is suggested to be effective (Chowdhury et al., 1995; Ritter et al., 1999) and so is 3-methylcholanthrene
  • Chrysin is not one of the major flavonoids in our diet, but its ability to inhibit
  • Example 1 already shows that quercetin can produce the same
  • Caco-2 human intestinal cell line Induction of UDP-glucuronosyltransferase 1*6.
  • UDP-glucuronosyltransferase 1A8 Arch. Biochem. Biophys.,
  • glucuronosyltransferase catalyzes the glucuronidation of ethinylestradiol. Mol.
  • UDP-glucuronosyltransferase (UGT2B15) with activity toward steroid and
  • glucuronosyltransferase UGTIAI in liver and cultured primary hepatocytes glucuronosyltransferase UGTIAI in liver and cultured primary hepatocytes:
  • Chrysin a potent inhibitor of the enzyme aromatase, is currently available as a dietary supplement in health food stores.
  • low bioavailability of chrysin was found, mainly due to extensive glucuronic acid and sulfate conjugation by the enterocytes (Biochem Pharmacol 58:431-8, 1999).
  • enterocytes Biochem Pharmacol 58:431-8, 1999.
  • these findings directly in normal volunteers after oral doses (400 mg) of chrysin. Only trace levels of chrysin ( ⁇ 25 ng/ml) could be detected in plasma together with its sulfate conjugate ( ⁇ 250 ng/ml).
  • the bioflavonoid chrysin, induced UDP- glucuronosyltransferase (UGT) activity and expression in the human intestinal cell line Caco-2 (Galijatovic et al, Pharm. Res. 17:21, 2000).
  • UGT induced UDP- glucuronosyltransferase
  • Caco-2 human intestinal cell line Caco-2
  • the specific UGT isoform(s) that chrysin induces and whether induction of a specific isoform facilitates increased glucuronidation of the dietary colon carcinogen N-OH- PhlP were determined.
  • Human intestinal Caco-2 cells were exposed to 50 ⁇ M chrysin or vehicle (control) for 3 days. The cells were harvested and the microsomal fraction, containing UGT enzymes was prepared. Control and chrysin-treated microsomes were then incubated with 25 ⁇ M N-hydroxy-PhlP. The reaction mixtures were analyzed by HPLC. This is similar methodology that was used to show the increased bilirubin glucuronidation in chrysin-treated Hep G2 and Caco-2 cells.
  • N-hydroxy-PhIP N2- glucuronide was 6.5 times higher in the chrysin-treated compared with the control cell preparation.
  • the formation of this glucuronide has previously clearly been linked to detoxification of N-hydroxy-PhIP. Similar detoxification may occur for other human colon carcinogens.

Abstract

The invention provides a method of treating any condition of elevated levels of unconjugated bilirubin in adults or children, such as Gilbert's syndrome or liver cirrhosis. The method calls for the administration of chrysin or chrysin analogs via dietary or other means. Also provided is a method of increasing glucuronidation of bilirubin in a subject, comprising administering to the subject an amount of chrysin effective to increase the glucuronidation of bilirubin. A method of increasing the expression of UDP-glucuronosyltransferase in a subject is provided, comprising administering to the subject an amount of chrysin effective to increase the UDP-glucuronosyltransferase. Upregulation of the expression of UDP-glucuronosyltransferase levels is expected to be beneficial in a number of areas: 1) increased detoxification of ingested carcinogens, such as the cooked-food mutagens (chrysin is an aromatase inhibitor in cells); 2) decreasing the conversion of androgens to estrogens, potentially building muscle mass or for prevention of breast cancer in women. In the method of upregulating UGT, UDP-glucuronosyltransferase can be a UGT1A isoform. A method of treating colon cancer is provided, comprising administering an effective amount of a flavone, whereby the administration of the flavone treats the colon cancer.

Description

A METHOD OF TREATING COLON CANCER BY ADMINISTERING APIGENIN, LUTEOLIN, DIOSMETIN AND CHRYSIN
This application claims priority to U.S. No. 60/182,072, filed February 11, 2000, and U.S. No. 60/232,771, filed September 15, 2000, and which applications are incorporated by reference herein.
The work described herein was carried out, in part, under National Institutes of Health grants CA69138 and GM55561. The government therefore may have certain rights in the invention.
BACKGROUND
As emphasized during the continuing development of the Caco-2 cell line as a model for human intestinal absoφtion, these cells in culture morphologically resemble small intestine absoφtive cells with many of its typical enzymes and transporters (1-4). Although Caco-2 cells in the past have not been considered to express high levels of drug metabolizing enzymes (5, 6), conjugation reactions have previously been described. This includes sulfation of dopamine and »-nitrophenol (7) and L-a- methyldopa (8). The model substrate >-nitrophenol has also been shown to be glucuroni dated by Caco-2 cells (5, 9), as has L-a-methyldopa (8).
This report describes for the first time the induction of UGT by chrysin and the ubiquitous dietary flavonoid quercetin (Fig. 1) in Caco-2 cells.
UDP-Glucuronosyltransferase (UGT) is a superfamily of enzymes that catalyze the glucuronidation of both endogenous toxins, e.g. bilirubin, as well as xenobiotics such as carcinogens, in general rendering them biologically inactive (Burchell and Coughtrie, 1989; Miners and Mackenzie, 1991; Mackenzie et al., 1997; de Wildt et al., 1999; Radominska-Pandya et al., 1999). Induction of these enzymes, which has long been known to occur (Burchell and Coughtrie, 1989; Miners and Mackenzie, 1991; Bock et al., 1999), could therefore be considered beneficial. On the other hand, increased glucuronidation of drugs would be expected to result in diminished pharmacologic response. Most inducers of the UGTs are potentially harmful chemicals or drugs (Burchell and Coughtrie, 1989; Miners and Mackenzie, 1991; Bock et al., 1999).
SUMMARY OF THE INVENTION
The invention provides a method of treating any condition of elevated levels of unconjugated bilirubin in adults or children, such as Gilbert's syndrome or liver cirrhosis. The method calls for the administration of chrysin or chrysin analogs via dietary or other means.
Also provided is a method of increasing glucuronidation of bilirubin in a subject, comprising administering to the subject an amount of chrysin effective to increase the glucuronidation of bilirubin.
A method of increasing the expression of UDP-glucuronosyltransferase in a subject is provided, comprising administering to the subject an amount of chrysin effective to increase the UDP-glucuronosyltransferase. Upregulation of the expression of UDP-glucuronosyltransferase levels is expected to be beneficial in a number of areas: 1) increased detoxification of ingested carcinogens, such as the cooked- food mutagens (chrysin is an aromatase inhibitor in cells); 2) decreasing the conversion of androgens to estrogens, potentially building muscle mass or for prevention of breast cancer in women. In the method of upregulating UGT, UDP-glucuronosyltransferase can be a UGT1 A isoform. The UGT1 A isoform can be UGT1 Al . The UGT can be UGT1A9. A method of treating colon cancer is provided, comprising administering an effective amount of a flavone, whereby the administration of the flavone treats the colon cancer. The flavone can be a 5,7-dihydroxyflavone, for example one selected from the group consisting of chrysin, apigenin, luteolin and diosmetin.
Chrysin analogs can be used in the methods and compositions of the invention. A chrysin analog having a flavonoid structure is contemplated. A chrysin analog having a flavone structure is contemplated. A chrysin analog having a 5,7-substituted flavone structure is further contemplated. Chrysin and its analogs can be administered as a dietary supplement or in a food. Examples of chrysin analogs having a 5,7- substituted flavone
BRIEF DESCRIPTION OF DRAWINGS
Fig. 1. Chemical structures of chrysin and quercetin.
Fig. 2. Chrysin metabolism by Caco-2 cells after pretreatment with 50 μM chrysin for 4 days. Open bars designate chrysin glucuronide and hatched bars chrysin sulfate. Mean values ± SEM are shown (N = 9). * Significantly higher than Control, p < 0.0001. ** Significantly lower than Control, p < 0.01.
Fig. 3. Chrysin glucuronidation by Caco-2 cell homogenates prepared from untreated cells (u) and cells pretreated with 50 μM chrysin for 4 days (s). Mean values ± SEM from 3 experiments are shown. The error bars for the untreated cells are contained within the symbols. Data were fit to the Michaelis-Menten equation.
Fig. 4. Immunoblot analysis of Caco-2 cell microsomes from cells pretreated with 50 mM chrysin for 4 days (lanes 4 and 6, 50 and 100 mg loaded) and from untreated Control cells (lanes 3 and 5, 50 and 100 mg loaded). UGT1A6 microsomal protein (50 mg loaded) was used as a positive control in lanes 1 and 2.
Fig. 5. Chrysin glucuronidation by intact Caco-2 cells after pretreatment of the cells with 10 μM quercetin or chrysin for 5 weeks. Medium containing solvent (Control), quercetin or chrysin was replaced every other day. After the last treatment, all cells were incubated with 50 μM chrysin for 24 hr for metabolite determinations. Mean values ± SEM are shown (N = 3-6).
Fig. 6. Chrysin metabolism by Hep G2 cells after pretreatment with 25 μM chrysin for 3 days. Open bars denote chrysin glucuronide and hatched bars chrysin sulfate. Mean values ± SEM are shown (N = 11). * Significantly higher than Control (P < 0.01).
Fig. 7. Chrysin glucuronidation by Hep G2 cell homogenates prepared from untreated cells (m) and cells pretreated with 25 μM chrysin for 3 days (1). Mean values from 2 experiments are shown. The data were fitted to the Michaelis-Menten equation using Microsoft Excel.
Fig. 8. Western blot analysis of recombinant UGT isoforms (2B7, 1 Al and 1 A6; lanes 1, 2 and 3, respectively) and microsomes from untreated (Control) Hep G2 cells (lane 4) and cells pretreated with 25 μM chrysin for 3 days (lane 5). Four different immunoblots were done with antibodies specific for UGT 1A, 1A6, 2B7 and 1A1, respectively. The arrows designate the UGT bands.
Fig. 9. Northern blot analysis of UGTl Al mRNA expression in Hep G2 cells. Total RNA (20 μg) from human liver (positive control) (lane 1), control and chrysin-treated cells (lanes 2 and 3, respectively) and poly(A)+ RNA (= 5 μg) isolated from control and chrysin-treated cells (lanes 4 and 5, respectively) were subjected to agarose- formaldehyde electrophoresis, hybridization with a [32P]-dCTP-labeled EcoRI/XhoI cDNA fragment of UGTl Al (Ritter et al., 1999) and autoradiography. The blots were then stripped and hybridized to a GAPDH cDNA probe (lower panel) to confirm equal sample loading for control and chrysin-treated samples on each blot.
Fig. 10. Bilirubin glucuronidation by Hep G2 homogenates prepared from untreated cells (m) and cells pretreated with 25 μM chrysin for 3 days (1). Mean values from 2 experiments are shown. The data were fitted to the Michaelis-Menten equation using Microsoft Excel. Significantly higher than Control, p < 0.05.
DETAILED DESCRIPTION OF THE INVENTION
As used herein, "a" or "an" can mean one or more than one. For example "a cell" can mean a single cell or multiple cells.
The invention provides a method of treating any condition of elevated levels of unconjugated bilirubin in adults or children, such as Gilbert's syndrome or liver cirrhosis. The method calls for the administration of chrysin or chrysin analogs via dietary or other means.
For example, a method of treating jaundice in a neonate is provided, comprising administering to the neonate an amount of chrysin effective to treat jaundice. A method of treating hyperbilirubinemia in a neonate is thus provided, comprising administering to the neonate an amount of chrysin effective to reduce the amount of bilirubin in the neonate.
Also provided is a method of increasing glucuronidation of bilirubin in a subject, comprising administering to the subject an amount of chrysin effective to increase the glucuronidation of bilirubin. A method of increasing the expression of UDP-glucuronosyltransferase in a subject is provided, comprising administering to the subject an amount of chrysin effective to increase the UDP-glucuronosyltransferase. Upregulation of the expression of UDP-glucuronosyltransferase levels is expected to be beneficial in a number of areas: 1) increased detoxification of ingested carcinogens, such as the cooked-food mutagens (chrysin is an aromatase inhibitor in cells); 2) decreasing the conversion of androgens to estrogens, potentially building muscle mass or for prevention of breast cancer in women. For example, upregulation of the expression of UDP- glucuronosyltransferase levels is associated with neutralization of colon carcinogens which include, but are not limited to, N-OH-PhlP.
In the method of upregulating UGT, UDP-glucuronosyltransferase can be a UGTl A isoform. The UGTl A isoform can be UGTl Al . The UGT can be UGTl A9.
A method of treating colon cancer is provided, comprising administering an effective amount of a flavone, whereby the administration of the flavone treats the colon cancer. The flavone can be a 5,7-dihydroxyflavone, for example one selected from the group consisting of chrysin, apigenin, luteolin and diosmetin.
Chrysin analogs can be used in the methods and compositions of the invention.
A chrysin analog having a flavonoid structure is contemplated. A chrysin analog having a flavone structure is contemplated. A chrysin analog having a 5,7-substituted flavone structure is further contemplated. Chrysin and its analogs can be administered as a dietary supplement or in a food. Examples of chrysin analogs having a 5,7- substituted flavone structure include, but are not limited to, quercetin, apigenin, luteolin and diosmetin.
The compositions of the present invention are dietary supplements and can thus
be administered orally. For oral administration, fine powders or granules may contain
diluting, dispersing, and/or surface active agents, and may be presented in water or in a syrup, in capsules or sachets in the dry state, or in a nonaqueous solution or suspension
wherein suspending agents may be included, in tablets wherein binders and lubricants
may be included, or in a suspension in water or a syrup. Where desirable or necessary,
flavoring, preserving, suspending, thickening, or emulsifying agents may be included.
Tablets and granules are preferred oral administration forms, and these may be coated.
The present invention is more particularly described in the following examples
which are intended as illustrative only since numerous modifications and variations
therein will be apparent to those skilled in the art.
EXAMPLE 1
Flavonoid Induction of Glucuronidation IN Caco-2 Cells
Dietary flavonoids have been reported to be potent inhibitors of drug
metabolizing enzymes. In the present study the inducing effect of three of these
compounds, chrysin, quercetin and genistein, on UDP-glucuronosyltransferase (UGT)
in the human intestinal cell line Caco-2 were examined. The induction of UGT by
flavonoid pretreatment was studied both in the intact cells and cell homogenates,
measured as the glucuronidation of chrysin, and by immunoblot analysis of the UGT
1A protein. Exposure of Caco-2 cells to 50 μM chrysin resulted in a 3.8-fold increase
in chrysin glucuronidation in intact cells (p < 0.0001) with a 38% decrease in sulfation (p < 0.01). In the cell homogenate the induction was much larger, 14-fold. The
induction was slow to develop with maximum induction after 3-4 days. Interestingly,
the isoflavonoid genistein was without effect. Immunoblot analysis of Caco-2 cell
microsomes with a UGTl A subfamily-selective antibody showed a markedly increased
band at about 59 kDa, consistent with induction of one or more UGTl A isoforms. A 5-
week exposure of Caco-2 cells to low concentrations (10 μM) of chrysin or quercetin
also showed markedly increased glucuronidation activity. Diet-mediated induction of
intestinal UGT may be important for the bioavailability of carcinogens and other toxic
chemicals as well as therapeutic drugs.
MATERIALS AND METHODS
Materials
Chrysin, quercetin, genistein, uridine 5'-diphosphoglucuronic acid (UDPGA)
and protease inhibitors were purchased from Sigma Chemical Co. (St. Louis, MO).
Trifluoroacetic acid was of spectrophotometric grade from Aldrich Chemical Co.
(Milwaukee, WI). Hanks' balanced salts solution (HBSS) was obtained from Cellgro,
Mediatech, Fisher Scientific (Pittsburgh, PA). Recombinant human UGTl A6 and the
corresponding Western blotting kit were purchased from Gentest Coφ. (Woburn, MA).
Polyclonal anti-human UGTl A antibodies, raised against 15 amino acids in the
common C-terminal end of the UGTl A proteins, were a generous gift from Gentest.
Secondary antibody, HRP-conjugated goat anti-rabbit IgG, was purchased from
Kirkegaard & Perry Laboratories (Gaithersburg, MD). SuperSignal Chemiluminescent
Substrate was from Pierce (Rockford, IL). Electrophoresis and blotting supplies and prestained molecular weight markers were purchased from Bio-Rad Laboratories
(Hercules, CA).
Caco-2 Cell Culture
The human colon adenocarcinoma cell line Caco-2 was obtained from
American Type Culture Collection (Rockville, MD). The Caco-2 cells were cultured in
Eagle's Minimum Essential Medium with Earle's salts and L-glutamine (Cellgro,
Mediatech, Fisher Scientific, Pittsburgh, PA), supplemented with 1% non-essential
amino acids (Cellgro), 10% fetal bovine serum (Summit Biotechnology, Ft. Collins,
CO) and penicillin/streptomycin (Sigma Chemical Co.). Cells were grown in
humidified air with 5% CO2 and subcultured in 6-well plates for incubation
experiments and 100 mm petri dishes for homogenate and microsomal preparation.
The cells were used at passage 35-75.
Chrysin Metabolism by Pretreated and Control Caco-2 cells
Four days after seeding Caco-2 cells in 6-wells, pretreatment with 50 mM
chrysin or 50 mM genistein in complete cell culture medium was started. The medium
was changed every 24 hr. Chrysin was dissolved in ethanokDMSO (80:20, v/v) at a
final concentration not exceeding 0.5%). Control cells were treated with the same
volume of solvent. After 4 days of pretreatment, 50 μM chrysin was added to both
chrysin-treated, genistein-treated and control cells. The medium was harvested 24 hr
later for analysis of chrysin and metabolites. Conjugated metabolites and unmetabolized chrysin were separated from the cellular medium using Oasis HLB 3cc
extraction cartridges (Waters, Milford, MA). The methanol eluates were taken to
dryness and the samples were reconstituted in 250 ml mobile phase and analyzed by
reversed phase HPLC, using a Symmetry C18 column (Waters) with 55% methanol and
0.3% trifluoroacetic acid as the mobile phase at 0.9 ml/min with 268 nm UV detection
(11). The cells were digested with 0.5 M NaOH and analyzed for protein content (15).
In another set of experiments, Caco-2 cells in 6-wells were pretreated with 50
μM chrysin for 1, 2, 3 or 4 days. The medium was analyzed for chrysin metabolites 24
hr after the last addition of chrysin (8 days after plating). The effect of flavonoid
pretreatment concentration on the glucuronidation was investigated using 0, 5, 10, 25
and 50 μM chrysin for 4 days.
Preparation of Caco-2 cell Homogenate and Microsomes
Caco-2 cells were plated in 100-mm petri dishes. On day 6 the cells were
incubated with 50 mM chrysin or solvent. The treatment with chrysin or solvent
continued for 4 days every 24 hr, as described above. Twenty-four hr after the last
medium change, the cell monolayers were washed twice with HBSS, scraped off the
dishes into 0.15 M KC1 in 10 mM sodium phosphate buffer (pH 7.4) (1 ml/dish). The
cells were disrupted by sonication on ice (5 x 5 sec). The cell homogenate was either
used for the catalytic assays or centrifuged at 9000 for 20 min at 4°C to obtain the
supernatant fraction (S10). The residual concentrations of chrysin glucuronide and
chrysin in the chrysin-pretreated cell homogenate were < 0.5 mM, as determined by HPLC. The S 10 fraction was further centrifuged at 100,000g for 60 min at 4°C. The
microsomal pellets were resuspended in 300 μl of homogenization buffer with protease
inhibitors (2 mM PMSF, 50 mg/ml antipain, 2 mg/ml aprotinin, 0.2 mg/ml
benzamidine, 0.5 mg/ml leupeptin and 1 mg/ml pepstatin). Homogenates and
microsomes were stored in aliquots at -80 °C.
Determination of UGT Protein Levels by Immunoblotting
Microsomal samples were heated at 90 °C for 5 min with an equal volume of
sample buffer and loaded on a 12% SDS-polyacrylamide minigel together with
molecular weight markers and positive and negative controls. After electrophoresis
(16), the proteins were transferred to a nitrocellulose membrane (17). The membrane
was blocked with 5% nonfat milk in 10 mM Tris/150 mM NaCl/0.05% Tween 20
(TBST) for 1 hr and incubated overnight at 4°C with anti-human UGTl A primary
antibody in 5% nonfat milk in TBST at a 1:100 dilution. The blot was washed with 5%
nonfat milk in TBST for 3 times 20 min and incubated with the secondary antibody,
HRP-conjugated goat anti-rabbit IgG, at a 1:5000 dilution in 5% milk in TBST for 1 hr.
After washing for 3 times 20 min with TBST, SuperSignal chemiluminescent substrate
was added and the membrane was exposed to Hyperfilm ECL (Amersham, Piscataway,
NJ) for 5 min. Chrysin Glucuronidation by Caco-2 Cell Homogenates
Chrysin (1 - 20 mM) and 100 ml of pretreated or control Caco-2 cell
homogenate (1.0 mg of protein) in 100 mM Tris.HCl buffer (pH 7.4) with 5 mM MgCl2
were prewarmed for 10 min at 37 °C. The reactions were initiated by the addition of
0.5 mM UDPGA. The reaction mixtures (500 μl) were incubated at 37°C for 60 min.
The samples were cooled on ice and centrifuged for 2 min at 10,000g. The supernatant
was subjected to solid-phase extraction and HPLC analysis, as described above.
Kinetic parameters were obtained by fitting the data to the Michaelis-Menten equation
in Microsoft Excel.
Chronic Caco-2 Cell Flavonoid Treatment
Caco-2 cells were cultured as above and subcultured at a seeding density of
80,000 cells/cm2 every 7 days. Fresh medium containing 10 μM chrysin, 10 μM
quercetin or DMSO was added on days 2, 4 and 6 after each subculturing. After 5
weeks of pretreatment, chrysin metabolism experiments were done in 6-wells at 24 hr
after the last flavonoid addition. The growth rate and viability were identical for
flavonoid-treated and control cells.
Statistical analysis used student's unpaired t-test with a significance level of p <
0.05. RESULTS
When the metabolism of chrysin by Caco-2 cells was analyzed by reversed-
phase HPLC, a glucuronic acid and a sulfate conjugate were identified by MS and
enzymatic techniques (11). After pretreatment of the cells for 4 days with 50 mM
chrysin, the concentration of unchanged chrysin was reduced, whereas that of the
glucuronic acid conjugate was markedly increased, with minimal change in the sulfate
conjugate concentration. The results from 9 experiments, with and without
pretreatment of Caco-2 cells with chrysin for 4 days are summarized in Fig. 2.
Whereas the glucuronic acid conjugate increased 3.8-fold (p < 0.0001), the sulfate
conjugate decreased 38% (p < 0.01). Although an increase in glucuronidation occurred
as early as after a 1-day pretreatment, the maximum response to chrysin was reached
after a 3- to 4-day pretreatment (Table I). The 30-50 % reduction in the formation of
the sulfate conjugate did not appear to be related to extensive consumption of the
substrate by the glucuronidation pathway. When examining the influence of chrysin
concentration on glucuronic acid conjugation, there appeared to be a linear increase
with concentration up to 50 mM (Table II). Whereas low chrysin concentrations did
not influence sulfate conjugation, 50 mM chrysin reduced this pathway similar to that
in Table I. In separate experiments also examined was the effect of pretreatment with
50 mM genistein, an isoflavonoid, on chrysin metabolism. There was no effect on
glucuronidation (126 ± 12 vs. 113 ± 12 pmol/mg protein/hr; treated vs. control), whereas sulfation was slightly decreased (226 ± 25 vs. 150 ± 19 pmol/mg protein/hr; p
< 0.05)(n=3).
We next examined the induction response in the Caco-2 cell homogenate after
careful washing of the cells prior to the homogenization. The increase in
glucuronidation after chrysin pretreatment was now as high as 14-fold compared to
control (Fig. 3). Because of the relatively low level of chrysin glucuronidation in the
homogenate of uninduced cells, accurate kinetics could not be determined. However,
the apparent K-,, value for glucuronidation of chrysin in the induced Caco-2 cell
homogenate was estimated to be about 0.8 mM.
In the next series of experiments, whether chrysin pretreatment of Caco-2 cells
produced an increase in UGT protein was determined. For these experiments,
microsomes of pretreated and untreated cells were prepared and an antibody, under
development by Gentest (Woburn, MA), which detects all proteins in the UGTl A
subfamily (Fig. 4) was used . There was a substantial increase in the intensity of a ~ 59
kDa band in the induced vs. uninduced control cells. Recombinant UGTl A6 protein
served as a positive control, producing a band with a migration identical to that of the
Caco-2 cell microsomes. Thus, chrysin pretreatment of Caco-2 cells resulted in
markedly increased expression of one or more UGTl A isoforms. In contrast,
experiments using an antibody specific for UGTl A6 failed to demonstrate induction of
this isoform. Finally, experiments were designed in which Caco-2 cells were exposed to
flavonoids for a period of 5 weeks. In addition to chrysin, quercetin, the most common
flavonoid in our diet (18) was also studied. Flavonoid concentrations of only 10 mM
were used in these experiments (Fig. 5). There was a more than 3-fold increase in the
glucuronidation of chrysin in the intact Caco-2 cells after chrysin pretreatment, similar
to the effect of 50 mM pretreatment in the acute experiments (Fig. 2). After quercetin
pretreatment, there was an about 2-fold increase in the glucuronidation activity.
This is the first report on induction of members of the UGT family by dietary
flavonoids in the human intestinal Caco-2 cells. This might have implications with
respect to the oral bioavailability of carcinogens as well as other toxicological agents
and commonly used drugs.
Pretreatment of human intestinal cells with chrysin markedly induces the
metabolism of this flavonoid through the glucuronidation but not the sulfation pathway.
The maximum induction response in the intact Caco-2 cells was a 3.8-fold increase in
chrysin glucuronidation (Fig. 2) which was magnified to a 14-fold increase in the Caco-
2 cell homogenate (Fig. 3). The lower apparent response in the intact cells may be due
to high accumulation of substrate and product, as demonstrated in a previous study for
quercetin (21), resulting in substrate/product inhibition. In contrast, both substrate and
product in the homogenate were efficiently removed, as evidenced by HPLC.
Maximum induction appeared to occur after 3-4 days of pretreatment and was linear with chrysin concentration in these short-term experiments. The inhibitory effect of
chrysin on the sulfation pathway at 50 μM concentration (Tables I and II) appears
similar to a previous observation for quercetin (22).
In order to test the effect of a chronic treatment schedule, the Caco-2 cells were
exposed to either chrysin or quercetin for up to 5 weeks, using only 10 μM
concentrations of each flavonoid. The induction response to chrysin was identical to
that of the short-term exposure; quercetin, the most abundant flavonoid in our diet (18),
was somewhat less effective. In terms of the flavonoid concentrations needed to
produce a significant induction response, the 10-50 μM concentrations used in the
present study are very likely to be achieved in the intestinal lumen after normal dietary
exposure to these compounds (10).
A different measure of the induction response was obtained by SDS-PAGE of
the microsomal proteins and immunoblotting. The use of a UGTl A subfamily-
selective antibody clearly demonstrated increased levels of one or several isoforms of
this group of enzymes (Fig. 4). Interestingly, experiments with a UGTlA6-specific
antibody showed no evidence of induction. UGTl A6 (23-25) as well as UGT1A9 and
UGT2B7 (25) have so far been identified in the Caco-2 cells.
There are a number of UGT isoforms, which could potentially use chrysin and
other flavonoids as substrates; these include UGT1A1 (26), UGT1A3 (27), UGT1A8
(28), UGTl A9 (29) and UGT2B15 (30, 31). As recently shown by Strassburg et al. (32), UGT1A1, UGTl A3, UGT1A8 and UGT1A9 are all expressed in the normal
human colon in addition to UGTl A4, UGTl A6 and UGTl A10.
Whereas UGTl Al, the isoform that metabolizes bilirubin, may be induced by
phenobarbital (33, 34), another UGT induction response is triggered by dioxin and
polyaromatic hydrocarbons via an effect on the Ah-receptor. This has been shown to
involve mainly UGT1A6 (24, 25) but also UGTl A9 (25). A recent study indicated that
the antioxidant t-butylhydroquinone induced UGT1A6, 1A9 and 2B7 (25). These
observations suggesting that UGT1A6 may not be involved (see above) also suggest
that the induction response produced by chrysin is different from that by dioxin and the
polyaromatic hydrocarbons, i.e. may not involve the Ah receptor.
These studies have suggested that the intestinal UGTs play an important role in
the detoxification of carcinogens and cytotoxic agents in general.
Throughout this application, various publications are referenced. The
disclosures of these publications in their entireties, as well as the references cited in
these publications, are hereby incoφorated by reference into this application in order to
more fully describe the state of the art to which this invention pertains. References
1. P. Artursson. Epithelial transport of drugs in cell culture. I: a model for studying
the passive diffusion of drugs over intestinal absorbtive (Caco-2) cells. J. Pharm. Sci.
79:476-482 (1990).
2. J. Hunter, M. A. Jepson, T. Tsuruo, N. L. Simmons, and B. H. Hirst. Functional
expression of P-glycoprotein in apical membranes of human intestinal Caco-2 cells -
kinetics of vinblastine secretion and interaction with modulators. J. Biol. Chem.
268:14991-14997 (1993).
3. V. Meunier, M. Bourne, Y. Berger, and G. Fabre. The human intestinal
epithelial cell line Caco-2; pharmacological and pharmacokinetic applications. Cell
Biol. Toxicol. 11:187-194 (1995).
4. U. K. Walle, A. Galijatovic, and T. Walle. Transport of the flavonoid chrysin
and its conjugated metabolites by the human intestinal cell line Caco-2. Biochem.
Pharmacol. 58:431-438 (1999).
5. T. Prueksaritanont, L. M. Gorham, j. H. Hochman, L. O. Tran, and K. P. Vyas.
Comparative studies of drug-metabolizing enzymes in dog, monkey, and human small
intestines, and in Caco-2 cells. Drug Metab. Dispos. 24:634-642 (1996). 6. L.-S. L. Gan and D. R. Thakker. Applications of the Caco-2 model in the design
and development of orally active drugs: elucidation of biochemical and physical
barriers posed by the intestinal epithelium. Adv. Drug Delivery Rev. 23:77-98 (1997).
7. A. Baranczyk-Kuzma, J. A. Garren, I. J. Hidalgo, and R. T. Borchardt. Substrate
specificity and some properties of phenol sulfotransferase from human intestinal Caco-
2 cells. Life Sci. 49:1197-1206 (1991).
8. P. J. Chikhale and R. T. Borchardt. Metabolism of L-a-methyldopa in cultured
human intestinal epithelial (Caco-2) cell monolayers: Comparison with metabolism in
vivo. Drug Metab. Dispos. 22:592-600 (1994).
9. S. Bjorge, K. L. Hamelehle, R. Homan, S. E. Rose, D. A. Turluck, and D. S.
Wright. Evidence for glucuronide conjugation of p-nitrophenol in the Caco-2 cell
model. Pharm. Res. 8:1441-1443 (1991).
10. R. A. Walgren, U. K. Walle, and T. Walle. Transport of quercetin and its
glucosides across human intestinal epithelial Caco-2 cells. Biochem. Pharmacol.
55:1721-1727 (1998).
11. A. Galijatovic, Y. Otake, U. K. Walle, and T. Walle. Extensive metabolism of
the flavonoid chrysin by human Caco-2 and Hep G2 cells. Xenobiotica in press:(1999). 12. X.-Y. Sun, C. A. Plouzek, J. P. Henry, T. T. Y. Wang, and J. M. Phang.
Increased UDP-glucuronosyltransferase activity and decreased prostate specific antigen
production by biochanin A in prostate cancer cell. Cancer Res. 58:2379-2384 (1998).
13. M.-H. Siess, J.-P. Mas, M.-C. Canivenc-Lavier, and M. Suschetet. Time course
of induction of rat hepatic drug-metabolizing enzyme activities following dietary
administration of flavonoids. J. Toxicol. Envir. Health 49:481-496 (1996).
14. M.-C. Canivenc-Lavier, M.-F. Vernevaut, M. Totis, M.-H. Siess, J. Magdalou,
and M. Suschetet. Comparative effects of flavonoids and model inducers on drug-
metabolizing enzymes in rat liver. Toxicology 114:19-27 (1996).
15. O. H. Lowry, N. J. Rosebrough, A. L. Farr, and R. J. Randall. Protein
measurement with the Folin phenol reagent. J. Biol. Chem. 193:265-275 (1951).
16. U. K. Laemmli. Cleavage of structural proteins during the assembly of the head
of bacteriophage T4. Nature 227:680-685 (1970).
17. H. Towbin, T. Staehelin, and J. Gordon. Electrophoretic transfer of proteins
from polyacrylamide gels to nitrocellulose sheets: procedure and some applications.
Proc. Natl. Acad. Sci. USA 76:4350-4354 (1979). 18. M. G. L. Hertog, P. C. H. Hollman, and M. B. Katan. Content of potentially
anticarcinogenic flavonoids of 28 vegetables and 9 fruits commonly consumed in the
Netherlands. J. Agric. Food Chem. 40:2379-2383 (1992).
19. Y.-C. Kao, C. Zhou, M. Sherman, C. A. Laughton, and S. Chen. Molecular
basis of the inhibition of human aromatase (estrogen synthetase) by flavone and
isoflavone phytoestrogens: a site-directed mutagenesis study. Environ. Health Perspect.
106:85-92 (1998).
20. J. W. Critchfield, J. E. Coligan, T. M. Folks, and S. T. Butera. Casein kinase II
is a selective target of HTV-1 transcriptional inhibitors. Proc. Natl. Acad. Sci USA
94:6110-6115 (1997).
21. D. W. Boulton, U. K. Walle, and T. Walle. Fate of the flavonoid quercetin in
human cell lines: Chemical instability and metabolism. J. Pharm. Pharmacol. 51:353-
359 (1999).
22. E. A. Eaton, U. K. Walle, A. j. Lewis, T. Hudson, A. A. Wilson, and T. Walle.
Flavonoids, potent inhibitors of the human P-form phenolsulfotransferase: potential
role in drug metabolism and chemoprevention. Drug Metab. Dispos. 24:232-237
(1996). 23. A. Abid, I. Bouchon, G. Siest, and N. Sabolovic. Glucuronidation in the Caco-2
human intestinal cell line: Induction of UDP-glucuronosyltransferase 1*6. Biochem.
Pharmacol. 50:557-561 (1995).
24. P. A. Mϋnzel, G. Bookjans, G. Mehner, T. Lehmkδster, and K. W. Bock.
Tissue-specific 2,3,7,8-tetrachlorodibenzo-/?-dioxin-inducible expression of human
UDP-glucuronosyltransferase UGT 1A6. Arch. Biochem. Biophys. 335:205-210 (1996).
25. P. A. Mϋnzel, S. Schmohl, H. Heel, K. Kalberer, B. S. Bock-Hennig, and K. W.
Bock. Induction of human UDP glucuronosyltransferases (UGT1A6, UGTl A9, and
UGT2B7) by t-butylhydroquinone and 2,3,7,8-tetrachlorodibenzo-p-dioxin in Caco-2
cells. Drug Metab. Dispos. 27:569-573 (1999).
26. C. D. King, M. D. Green, G. R. Rios, B. L. Coffman, I. S. Owens, W. P.
Bishop, and T. R. Tephly. The glucuronidation of exogenous and endogenous
compounds by stably expressed rat and human UDP-glucuronosyltransferase 1.1. Arch.
Biochem. Biophys. 332:92-100 (1996).
27. M. D. Green, C. D. King, B. Mojarrabi, P. I. Mackenzie, and T. R. Tephly.
Glucuronidation of amines and other xenobiotics catalyzed by expressed human UDP-
glucuronosyltransferase 1A3. Drug Metab. Dispos. 26:507-512 (1998). 28. Z. Cheng, A. Radominska-Pandya, and T. R. Tephly. Cloning and expression of
human UDP-glucuronosyltransferase (UGT) 1A8. Arch. Biochem. Biophys. 356:301-
305 (1998).
29. T. Ebner and B. Burchell. Substrate specificities of two stably expressed human
liver UDP-glucuronosyltransferases of the UGTl gene family. Drug Metab. Dispos.
21:50-56 (1993).
30. M. D. Green, E. M. Oturo, and T. R. Tephly. Stable expression of a human liver
UDP-glucuronosyltransferase (UGT2B15) with activity toward steroid and xenobiotic
substrates. Drug Metab. Dispos. 22:799-805 (1994).
31. E. Levesque, M. Beaulieu, M. D. Green, T. R. Tephly, A. Belanger, and D. W.
Hum. Isolation and characterization of UGT2B15 (Y85): a UDP-
glucuronosyltransferase encoded by a polymoφhic gene. Pharmacogenetics 7:317-325
(1997).
32. C. P. Strassburg, N. Nguyen, M. P. Manns, and R. H. Tukey. UDP-
Glucuronosyltransferase activity in human liver and colon. Gastroenterology 116:149-
160 (1999).
33. Y. Ishii, A. Takami, K. Tsuruda, A. Kurogi, H. Yamada, and K. Oguri.
Induction of two UDP-glucuronosyltransferase isoforms sensitive to phenobarbital that are involved in moφhine glucuronidation: production of isoform-selective antipeptide
antibodies toward UGTl.lr and UGT2B1. Drug Metab. Dispos. 25:163-167 (1997).
34. T. Iyanagi, Y. Emi, and S. Ikushiro. Biochemical and molecular aspects of
genetic disorders of bilirubin metabolism. Biochim. Biophys. Acta 1407:173-184
(1998).
Table 1. Effect of Pretreatment Time with Chrysin (50 mM) on the Formation of
Chrysin Glucuronide and Sulfate
Pretreatment Time Metabolite Formation0
(days) (pmol/mg protein/hr)
Glucuronide Sulfate
0 115 ± 11 109 ± 8
1 182 ± 15* 72 ± 8*
2 2 2 27711 ±± 4499** 58 ± 16*
3 427 ± 66* 75 ± lb
4 450 ± 20* 73 ± 6b
a Mean values ± SD (N=3)
* Significantly different from 0 days (p < 0.05)
Table 2. Effect of Pretreatment Chrysin Concentration on the Formation of Chrysin
Glucuronide and Sulfate
Pretreatment Concentration Metabolite Formation"
(mM) (pmol/mg protein/hr)
Glucuronide Sulfate
0 115 ± 11 109 ± 8
5 135 ±7 115 ± 5
10 187 ±13* 108 ± 10
25 330 ±46* 94 ± 15
50 445 ± 29* 60 ± 5*
Incubations were done for four days
a Mean values ± SD (N=3)
* Significantly different from 0 days (p < 0.05)
EXAMPLE 2
Chrysin Induction of UDP-Glucuronosyltransferase 1 Al in Hep G2 Cells
The UDP-glucuronosyltransferases (UGTs) have long been known to be
inducible by various chemicals, including drugs, although the extent of induction in general has been modest. In the present study the ability of the dietary flavonoid
chrysin to induce UGT activity, protein and mRNA were determined. When
pretreating human hepatoma Hep G2 cells with 25 μM of chrysin, the glucuronidation
of chrysin itself increased 4.2-fold when measured in the intact cell and 14-fold in the
cell homogenate, i.e. autoinduction. Microsomes from chrysin-treated cells probed
with specific antibodies in Western analyses showed marked induction of the UGTl A
family of proteins. Iso form-specific induction of the important hepatic UGTIAI
protein was observed, but not of UGTl A6 or UGT2B7. The strong induction of
UGTIAI was confirmed by Northern analyses of total RNA as well as mRNA, using a
specific probe. UGTIAI message as well as protein were detectable also in untreated
Hep G2 cells. In catalytic activity assays with recombinant UGTIAI, 1 A4, 1A6 and
1A9, chrysin was found to be a high affinity substrate for UGTIAI (Km 0.35 μM).
Catalytic activity was also found for UGTl A9 and 1 A6 but not for 1 A4. Further
studies demonstrated a 20-fold induction of the glucuronidation of bilirubin by the
chrysin-treated cells and an 7.9-fold induction of the glucuronidation of the oral
contraceptive drug ethinylestradiol, two of the best known and specific UGTIAI
substrates, demonstrating the potential importance of this induction. In view of these
findings, it is expected that these results will apply to other dietary flavonoids.
Example 1 demonstrated efficient induction of one or several isoforms of the
UGTl A subfamily by chrysin in the human intestinal cell line Caco-2. The induction
resulted in as much as a 14-fold increase in the glucuronidation of chrysin by the cell homogenate. The response was linear with concentration of flavonoid over the range of
5-50 μM and was maximal after pretreatment for 3-4 days. As recently shown by
Strassburg et al. (Strassburg et al., 1999), the UGTl A isoforms are differentially
expressed in human hepatic and intestinal tissue, raising the question whether the
induction response is different in a hepatic as compared to an intestinal cell line.
In the present Example, the effect of chrysin on glucuronic acid conjugation in
the human hepatic cell line Hep G2 was examined. Studies with recombinant enzymes
and isoform-specific antibodies were performed to establish which UGTl A isoform(s)
were induced. The finding of induction of UGTl Al was confirmed by Northern
analysis and its functional significance explored for several substrates, such as bilirubin
and the oral contraceptive drug ethinylestradiol.
Materials and Methods
Chemicals
Chrysin (5,7-dihydroxyflavone) (Fig. 1), bilirubin (mixed isomers),
ethinylestradiol, uridine 5'-diphosphoglucuronic acid (UDPGA) and Williams'
Medium E were purchased from Sigma Chemical Co (St. Louis, MO). Fetal bovine
serum was obtained from Summit Biotechnology (Fort Collins, CO). Other cell culture
supplies were prepared by Mediatech (Herndon, VA) and obtained from Fisher
Scientific (Pittsburgh, PA). [3H]17a-Ethinylestradiol (50 Ci/mmol) and [a32P]dCTP
(3000 Ci/mmol) were purchased from NEN Life Science Products (Boston, MA). Cell Culture
Hep G2 cells were obtained from American Type Culture Collection (Rockville,
MD). The cells were maintained in Williams' Medium E with 10% fetal bovine serum,
L-glutamine and antibiotic/antimycotic solution in a humidified 37°C incubator with
5% carbon dioxide. When cells in 100-mm dishes or 6-wells were just confluent (4-6
days after seeding), they were treated with 25 μM chrysin in dimethylsulfoxide
(DMSO; 0.3%) of final volume) or the same volume of DMSO for 3 days. The medium
was changed every 24 hr and the cells were used for in situ metabolism assays,
homogenate or microsomal preparation or RNA isolation at 24 hr after the last medium
change. For in situ evaluation of the conjugation activity of induced and control Hep
G2, the cells grown in 6-wells were incubated for 6 hr with 3 ml of medium containing
25 μM chrysin. The medium was then collected, subjected to solid phase extraction
with Oasis cartridges (Waters, Milford, MA), as previously described (Galijatovic et
al., 1999), and subjected to HPLC analysis.
Glucuronidation Assays with Cell Homogenates and Microsomes
Confluent Hep G2 cells grown in 100-mm dishes and treated as above with
chrysin or solvent for 3 days were rinsed with phosphate-buffered saline and harvested
by scraping the cells into 0.15 M KC1 in 10 mM phosphate buffer (pH 7.4, 1 ml/dish).
The cells were sonified for 3 x 5 sec. The homogenates were stored in aliquots at -
80°C and used for catalytic assays of glucuronidation of chrysin and bilirubin. These
incubations were carried out for 1 hr at 37°C with 0.1-100 μM substrate concentrations, 1 mM UDPGA and cell homogenate (0.7-2 mg protein) in 0.5 ml 100 mM Tris.HCl
buffer, pH 7.4, with 5 mM MgCl2. Control samples were incubated without UDPGA.
The glucuronidation reactions for chrysin were terminated by putting the samples on
ice and subjecting them to solid phase extraction, as described above. Bilirubin
incubations were done under argon and the samples were protected from light, using
modifications of a method suggested by Chris Patten at Gentest. The reactions were
terminated by putting the samples on ice and adding ascorbic acid (10 mg/ml) and an
equal volume of acetonitrile to precipitate proteins. The supernatant, after vortex
mixing and centrifugation at 14,000g, was used for HPLC analysis.
HPLC analyses of chrysin incubates were done with 55% methanol and 0.3%
trifluoroacetic acid on a Symmetry C18 column (Waters), as previously described
(Galijatovic et al., 1999). Bilirubin sample HPLC analyses also used a Symmetry C18
column with a linear solvent gradient from 0.1% trifluoroacetic acid in water to 100%
acetonitrile and detection at 450 nm.
Assays of the glucuronidation of ethinylestradiol used microsomes of cells
treated as above. The microsomes were prepared by centrifugation of the cell
homogenates above for 1 hr at 100,000 g and 4°C and resuspension of the pellets in
homogenization buffer. A substrate concentration of 100 μM with 0.5 μCi of
[ H] ethinylestradiol per 0.5-ml sample was used. The incubations were carried out as
for chrysin above and terminated with an equal volume of 0.25 M Tris buffer, pH 8.7.
The samples were extracted twice with toluene to remove unreacted substrate (Zhu et al., 1998) and aliquots of the aqueous phase were subjected to liquid scintillation
spectrometry after the addition of ScintiSafe Econo2 (Fisher) scintillant.
Immunoblot Analyses of UGT Proteins
Microsomes prepared from chrysin-treated and control Hep G2 cells and human
liver as well as recombinant standard proteins were heated at 90°C for 5 min with an
equal volume of sample buffer and loaded on 12% SDS polyacrylamide gels. After
electrophoresis the proteins were transferred to nitrocellulose membranes, blocked and
incubated with primary and secondary antibodies as previously described in Example 1.
The following primary polyclonal antibodies against human UGT proteins were
obtained from Gentest: anti-UGTl A, anti-UGTlA6 and anti-UGT2B7. In addition,
anti-UGTl Al was prepared and used as recently described (Ritter et al., 1999) with the
exception that incubations with primary and secondary antibodies were done in the
presence of 5% nonfat milk.
Northern Blot Analyses of UGT mRNAs
Total RNA was isolated from chrysin-treated and control Hep G2 cells (5 100-
mm dishes each) and 1 g human liver. The cell monolayers were washed with PBS and
the cells were digested with RNAzol B (Tel-Test, Inc., Friendswood, TX) lysis buffer
(1 ml/plate). The digests were treated with chloroform to remove protein and DNA and
the RNA was precipitated from the aqueous phase with isopropanol and washed with
70% ethanol according to the manufacturer's protocol. The liver tissue was treated identically after homogenization with RNAzol (6 ml) in a Potter-Elvehjem
homogenizer. All samples were reconstituted in 500 μl 0.1% SDS (3-4 μg/μl,
determined by optical density at 260 nm) and, after denaturation at 65°C for 15 min
with 3 volumes of RNA sample loading buffer, 20 μg RNA was loaded on a 1%
agarose gel with 3% formaldehyde. After electrophoresis in MOPS/EDTA buffer, the
RNA was transferred to nylon filters (Hybond-N, Amersham Pharmacia Biotech,
Piscataway, NJ). The filters were hybridized overnight at 65°C to a [32P] -labeled probe
prepared from a 0.7-kbp Xhol-EcoRI restriction fragment pSK-UGTl Al containing the
5' end of UGTIAI (Ritter et al., 1999). The probe was labeled by random primed
synthesis, using [a32P]dCTP and a kit from Amersham Pharmacia Biotech. After
repeated washes with SDS/EDTA in phosphate buffer, the blots were subjected to
autoradiography, using Hyperfilm MP (Amersham). The same membranes were then
stripped and reprobed with a 1 kbp glyceraldehyde-3-phosphate dehydrogenase
(GAPDH) cDNA (Ambion, Austin, TX) to confirm equal loading beween control and
chrysin-treated cell samples.
In order to improve sensitivity in detecting UGTIAI mRNA, poly(A)+ RNA
was also isolated from chrysin-treated and control Hep G2 cells, using a FastTrack 2.0
kit (Invitrogen, Carlsbad, CA) according to the manufacturer's instructions. Five 100-
mm dishes of confluent cells were used for each preparation. The mRNA was
dissolved in 25 μl buffer (~ 0.7 μg/μl) and 5 μg was loaded on a gel. Electrophoresis
and hybridization with the UGTIAI and GAPDH probes were done as for the total
RNA samples. Glucuronidation Assays with Recombinant Enzymes and Human Liver
Microsomes
Chrysin and bilirubin (0.1-100 μM) were used as potential substrates for
recombinant UGT isoforms, i.e., 1 Al, 1 A4, 1 A6 and 1A9, supplied as microsomes of
human lymphoblast-expressed enzymes (Gentest, Wobum, MA), and also with human
liver microsomes prepared by standard differential centrifugation (La Du et al., 1972).
Human livers were obtained from Liver Tissue Procurement and Distribution System
(University of Minnesota, Minneapolis, MN) kept frozen at -80°C. The incubations
and analyses were done exactly as with the Hep G2 cell homogenates above.
Results
The flavonoid chrysin is efficiently metabolized by human Hep G2 cells both by
glucuronidation and sulfation. When the Hep G2 cells were pretreated with 25 μM
chrysin for 3 days, the glucuronidation of chrysin in the intact cell increased 4.2-fold (p
< 0.01; N = 11) while the sulfation remained unaffected, Fig. 6. The next experiments
focused on glucuronidation only, using the cell homogenate, rather than the intact cell,
to monitor chrysin glucuronidation, after addition of the cofactor UDPGA. In these
experiments chrysin pretreatment resulted in a 14-fold increase in activity compared to
untreated cells, Fig. 7. The apparent enzyme kinetic parameters determined in these
experiments demonstrated that the increase in chrysin glucuronidation efficiency
(Nmax/Km) was mainly due to an increase in the Vmax value, although the Km value of 4.9 μM was reduced after induction, 1.9 μM. In the next series of experiments four antibodies were used in Western analyses of
microsomes isolated from chrysin-treated and untreated Hep G2 cells, Fig. 8. An
antibody recognizing all of the UGTl A isoforms demonstrated a low expression in
untreated cells but a marked induction in the chrysin-pretreated Hep G2 cells (Fig. 8,
top panel). Antibodies recogmzing the common hepatic UGTl A6 and 2B7 isoforms
showed that these isoforms were expressed in Hep G2 cells but were not induced by
chrysin (middle panels). In contrast, an antibody recently shown to specifically
recognize UGTIAI (Ritter et al., 1999) detected a high level of induction of this
isoform by chrysin in the Hep G2 cells with virtually no staining in the control cells
(Fig. 8, bottom panel).
Total RNA isolated from control and chrysin-induced Hep G2 cells, subjected
to agarose-formaldehyde electrophoresis and hybridized to a [32P]-labeled cDNA
fragment of pBluescript SK/UGT1A1, clearly showed the presence of UGTIAI mRNA
with the expected size (2.3 kb) in the chrysin-treated cells but only trace amounts in the
control cells (Fig. 9, lanes 3 and 2, respectively). As expected, total RNA isolated from
human liver gave a positive signal (lane 1). The membrane was then stripped and
hybridized with a GAPDH cDNA probe (lower panel), showing similar loading of
control and induced cells. When poly(A)+ RNA was isolated from control and chrysin-
treated Hep G2 cells, allowing the loading of about 20-30-fold more mRNA on the gel,
UGTIAI mRNA could be clearly detected also in the control cells and was strongly
induced in the chrysin-treated cells (Fig. 9, lanes 4 and 5, respectively). Again,
reprobing with GAPDH cDNA showed similar loading. When comparing the catalytic activities towards chrysin of recombinant UGT
isoforms 1A1, 1A6, 1A9 and 1A4, all of which are hepatic isoforms, the isoform that
demonstrated the highest activity (V--,- /!^), interestingly, was the inducible form,
UGTIAI, Table 3. Its Km value for chrysin glucuronidation was low, 0.35 μM, and its
Vmax value high, 360 pmol/min/mg protein. A high catalytic efficiency for UGTl A9,
about 70%) of that for UGTIAI was also found, but relatively low efficiency for
UGTl A6, the isoform that is known to use simple planar phenols as substrates was
found, Table 3. Chrysin was not a substrate for UGTl A4.
In view of the findings of the induction of the UGTIAI isoform, the
glucuronidation of compounds which have been shown to be specific substrates for this
isoform was examined, using the homogenates of chrysin-induced and non-induced
Hep G2 cells. The best recognized substrate, bilirubin, was glucuronidated both by the
non-induced and the chrysin-treated cells, producing both isomeric monoglucuronides
and the diglucuronide in a ratio of about 6:1, using a high resolution HPLC approach.
Total glucuronidation of bilirubin by the cell homogenates is shown in Fig. 10.
Although activity was clearly measurable at all bilirubin concentrations, 0.1 - 5 μM,
accurate enzyme kinetic parameters could not be determined for the non-induced cells.
After pretreatment of Hep G2 cells with 25 μM chrysin, total bilirubin glucuronidation
increased about 20-fold with an apparent Km value of 1.4 μM and a Vmax value of 3.7
pmol/min/mg protein. For comparison, total bilirubin glucuronidation by recombinant UGTIAI yielded a K--, value of 0.23 μM and by liver microsomes from two human
donors K-,, values of 0.78 and 0.79 μM.
The oral contraceptive drug ethinylestradiol has also been shown to be a
substrate for UGTIAI (Ebner and Burchell, 1993). When using this drug to assay
glucuronidation activity of the non-induced Hep G2 cell homogenate, the activity was
barely above background, 1.4 pmol/min mg protein. In the homogenate from the
chrysin-induced cells, the activity increased 7-fold to 9.7 ± 2.8 (SEM; N = 4)
pmol/min/mg protein (P < 0.05).
Example 1 demonstrated induction of glucuronidation when the human
intestinal cell line Caco-2 was treated with chrysin, suggesting the possibility that the
flavonoids might induce their own elimination. In the present Example these
observations were extend to include also the human hepatic cell line Hep G2. Thus,
pretreatment of these cells for 3 days with 25 μM chrysin resulted in a 4.2-fold increase
in the glucuronidation of chrysin by the intact cells with no change in sulfonation.
When chrysin glucuronidation was examined in the cell homogenate after
washing of the cells to remove chrysin used in the pretreatment of the cells, the
induction was as high as 14-fold. The lower level of induction in the intact cells may
be attributed to substrate/product inhibition, although the possibility of UDPGA
cofactor depletion in the intact cell cannot be excluded, as opposed to the homogenate
incubations where UDPGA was added in excess. Another possible reason for this phenomenon is that latent enzyme is activated by sonification in the cell
homogenization procedure. The apparent enzyme kinetic parameters for the
homogenate experiments indicated that the Km value remained virtually the same after
induction, whereas the Vmax value increased dramatically. As shown for Caco-2 cells,
maximal induction of glucuronidation in the Hep G2 cells occurred after pretreatment
with chrysin for 3-4 days and the minimum concentration for induction was about 10
μM.
In experiments using UGT isoform-specific antibodies and Western analyses,
microsomes from chrysin-treated vs. control Hep G2 cells were probed to identify
which isoforms may have been induced. An antibody selective for all UGTl A, as
opposed to UGT2B, isoforms demonstrated a high degree of induction. Interestingly, a
major hepatic isoform, UGTl A6, was not induced and this was also the case with
UGT2B7. In contrast, a band with migration identical to that of UGTIAI was
markedly induced. This was detected using a newly developed antibody highly specific
for this isoform (Ritter et al., 1999). The finding of UGTl Al induction points to a
highly selective induction response of the UGTs to chrysin.
Strongly supporting the UGTIAI protein induction were the Northern analyses
using a probe highly specific for the mRNA for this isoform (Ritter et al., 1999). In the
analysis of total RNA, while there was a strong signal in the chrysin-treated cells, there
was no detectable message for UGTIAI in the control cells, consistent with previous observations (Batt et al., 1995; Ritter et al., 1999). However, in the Northern analysis
of purified mRNA, the message was clearly present also in the uninduced cells and,
again, strongly induced in the chrysin-treated cells. An additional band with lower
molecular weight was also induced, which may be a splice variant of UGTIAI.
Consistent with induction of chrysin glucuronidation, chrysin was a substrate
for UGTIAI, in fact was a highly efficient substrate for this isoform. In the present
study, the enzyme kinetic properties, demonstrating a very low K--, value of 0.35 μM
for the recombinant protein were also established. Chrysin was also an efficient
substrate for UGTl A9, with a low K--, value of 1.7 μM, but a poor substrate for
UGT1A6 (K-π 12.8 μM) and not a substrate for UGT1A4. This suggests that UGTl A9
may also be induced by chrysin. The finding that chrysin is a substrate for UGTl A6,
suggests that this isoform might be responsible for chrysin glucuronidation in the
uninduced Hep G2 cells. This is supported by the Western analysis, showing equal
presence of UGT1A6 in induced and control cells. The induction of chrysin
glucuronidation in the Hep G2 cells resulted in a reduction in the Km value, although it
was not as low as the K,-, value for chrysin glucuronidation by recombinant UGTIAI.
This observation could suggest that additional UGT isoforms may be induced.
Previous work has identified several other UGT isoforms which could potentially use
chrysin or other flavonoids as substrates. These include UGTl A3 (Green et al., 1998),
1 A8 (Cheng et al., 1998), 1A9 (Ebner and Burchell, 1993) and 2B15 (Green et al.,
1994; Levesque et al., 1997). As recently shown by Strassburg et al. (Strassburg et al., 1999), UGTIAI, 1 A3, 1 A4, 1A6 and 1 A9 are all present in the liver, whereas the
intestine also contains UGT1A8 and 1A10.
The finding that the Km value for chrysin glucuronidation by the Hep G2 cell
homogenate is about 5 times higher than by recombinant UGTIAI may potentially be
due to an endogenous inhibitor in the cell homogenate. This hypothesis is supported by
the very similar finding with bilirubin as substrate, i.e. 6 times higher K--, for the Hep
G2 cell homogenate compared to recombinant UGTIAI.
The induction of UGTl Al by chrysin has important biological implications.
The best known substrate for UGTIAI is the endogenous toxin bilirubin (Chowdhury
et al., 1995; Ritter et al., 1999). Induction of UGTl Al therefore suggests that bilirubin
glucuronidation is increased, as clearly shown in the present example, with the level of
induction being as high as with chrysin glucuronidation, i.e. more than 10- fold. Also,
the values for Vmax were very similar for bilirubin and chrysin glucuronidation.
Interestingly, the Km value for bilirubin glucuronidation both by induced Hep G2 cells,
recombinant UGTIAI and human liver microsomes of 0.23-1.4 μM is considerably
lower than in general reported (Radominska-Pandya et al., 1999). This may be the
result of a more efficient and sensitive separation/detection system for the bilirubin
glucuronides in the present study. Because of the importance of glucuronidation in the
removal of the endogenous toxin bilirubin, the induction of UGTIAI, the isoform
responsible for this reaction, has been of interest. Phenobarbital is suggested to be effective (Chowdhury et al., 1995; Ritter et al., 1999) and so is 3-methylcholanthrene
(Ritter et al., 1999), although the inducibility appears to be considerably less than
observed with chrysin in the present Example. The induction by 3-methylcholanthrene
suggests the involvement of the aryl hydrocarbon receptor (Ritter et al., 1999).
However, UGTl A6, inducible by arylhydrocarbon receptor agonists in Caco-2 cells
(Abid et al., 1995; Bock et al, 1999; Mϋnzel et al., 1999), was not affected by chrysin.
The mechanism of chrysin induction thus needs to be investigated.
Finally, the induction of UGTIAI may affect metabolism of some drugs. Thus,
the glucuronidation of the synthetic estrogen ethinylestradiol, which has been shown to
be a substrate for UGTIAI (Ebner et al., 1993), was induced about 7-fold by chrysin.
Such increased glucuronidation may lead to diminished contraceptive efficacy of this
drug, of obvious importance.
Chrysin is not one of the major flavonoids in our diet, but its ability to inhibit
the enzyme aromatase (Kao et al., 1998), catalyzing the conversion of testosterone to
17b-estradiol, suggests its use in lowering breast cancer risk. These findings are likely
to apply to other flavonoids, especially those present at higher levels in various fruits
and vegetables. Example 1 already shows that quercetin can produce the same
response when cells are exposed for several weeks to this flavonoid (Galijatovic et al.,
2000). Throughout this application, various publications are referenced. The
disclosures of these publications in their entireties, as well as the references cited in
these publications, are hereby incoφorated by reference into this application in order to
more fully describe the state of the art to which this invention pertains.
References
1. Abe KI, Inoue O and Yumioka E (1990). Biliary excretion of metabolites of
baicalin and baicalein in rats. Chem. Pharm. Bull., 38: 208-211.
2. Abid A, Bouchon I, Siest G and Sabolovic N (1995). Glucuronidation in the
Caco-2 human intestinal cell line: Induction of UDP-glucuronosyltransferase 1*6.
Biochem. Pharmacol., 50: 557-561.
3. Batt AM, Ferrari L, Abid A and Sabolovic N (1995). Human cell lines in
pharmaco-toxicology: an introduction to a panel discussion. Cell Biol. Toxicol,
11: 179-185.
4. Bock KW, Gschaidmeier H, Heel H, Lehmkόster T, Mϋnzel PA and Bock-Hennig
BS (1999). Functions and transcriptional regulation of PAH-inducible human
UDP-glucuronosyltransferases. Drug Metab. Rev., 31: 411-422. 5. Boutin JA, Meunier F, Lambert PH, Hennig P, Bertin D, Serkiz B and Volland JP
(1993). In vivo and in vitro glucuronidation of the flavonoid diosmetin in rats.
Drug Metab. Dispos., 21: 1157-1166.
6. Burchell B and Coughtrie MWH ( 1989). UDP-Glucuronosyltransferases.
Pharmac. Ther., 43: 261-289.
7. Canivenc-Lavier M-C, Vernevaut M-F, Totis M, Siess M-H, Magdalou J and
Suschetet M (1996). Comparative effects of flavonoids and model inducers on
drug-metabolizing enzymes in rat liver. Toxicology, 114: 19-27.
8. Cheng Z, Radominska-Pandya A and Tephly TR (1998). Cloning and expression
of human UDP-glucuronosyltransferase (UGT) 1A8. Arch. Biochem. Biophys.,
356: 301-305.
9. Chowdhury JR, Wolkoff AW, Chowdhury NR and Arias IM ( 1995). Hereditary
jaundice and disorders of bilirubin metabolism. In The Metabolic and Molecular
Basis of Inherited Disease, ed. Scriver, C.R., Beaudet, A.L., Sly, W.S. & Valle, D.
pp. 2161-2208. New York: McGraw-Hill.
10. Clapper ML (1998). Chemopreventive activity of oltipraz. Pharmacol. Ther., 78:
17-27. 11. de Wildt SN, Kearns GL, Leeder JS and van den Anker JN (1999).
Glucuronidation in humans: pharmacogenetic and developmental aspects. Clin.
Pharmacokinet, 36: 439-452.
12. Ebner T and Burchell B (1993). Substrate specificities of two stably expressed
human liver UDP-glucuronosyltransferases of the UGTl gene family. Drug
Metab. Dispos., 21: 50-56.
13. Ebner T, Remmel RP and Burchell B (1993). Human bilirubin UDP-
glucuronosyltransferase catalyzes the glucuronidation of ethinylestradiol. Mol.
Pharmacol, 43: 649-654.
14. Galijatovic A, Otake Y, Walle UK and Walle T (1999). Extensive metabolism of
the flavonoid chrysin by human Caco-2 and Hep G2 cells. Xenobiotica, 29: 1241-
1256.
15. Galijatovic A, Walle UK and Walle T (2000). Induction of UDP-
glucuronosyltransferase by the flavonoids chrysin and quercetin in Caco-2 cells.
Pharm. Res., 17: in press. 16. Green MD, King CD, Mojarrabi B, Mackenzie PI and Tephly TR (1998).
Glucuronidation of amines and other xenobiotics catalyzed by expressed human
UDP-glucuronosyltransferase 1A3. Drug Metab. Dispos., 26: 507-512.
17. Green MD, Oturo EM and Tephly TR (1994). Stable expression of a human liver
UDP-glucuronosyltransferase (UGT2B15) with activity toward steroid and
xenobiotic substrates. Drug Metab. Dispos., 22: 799-805.
18. Grove AD, Kessler FK, Metz RP and Ritter JK (1997). Identification of a rat
oltipraz-inducible UDP-glucuronosyltransferase (UGT1A7) with activity towards
benzo(α)pyrene-7,8-dihydrodiol. J. Biol. Chem., 272: 1621-1627.
19. Hecht SS, Carmella SG and Muφhy SE (1999). Effects of watercress
consumption on urinary metabolites of nicotine in smokers. Cancer Epidemiol.
Biomarkers Prev., 8: 907-913.
20. Jager W, Zembsch B, Wolschann P, Pittenauer E, Senderowicz AM, Sausville
EA, Sedlacek HH, Graf J and Thalhammer T (1998). Metabolism of the anticancer
drug flavopiridol, a new inhibitor of cyclin dependent kinases, in rat liver. Life
Sci., 62: 1861-1873.
21. Kao Y-C, Zhou C, Sherman M, Laughton CA and Chen S (1998). Molecular
basis of the inhibition of human aromatase (estrogen synthetase) by flavone and isoflavone phytoestrogens: a site-directed mutagenesis study. Environ. Health
Perspect., 106: 85-92.
22. King CD, Green MD, Rios GR, Coffman BL, Owens IS, Bishop WP and Tephly
TR (1996). The glucuronidation of exogenous and endogenous compounds by
stably expressed rat and human UDP-glucuronosyltransferase 1.1. Arch. Biochem.
Biophys., 332: 92-100.
23. La Du BN, Mandel HG and Way EL (1972). Fundamentals of Drug Metabolism
and Drug Disposition. Baltimore: Williams & Wilkins.
24. Levesque E, Beaulieu M, Green MD, Tephly TR, Belanger A and Hum DW
(1997). Isolation and characterization of UGT2B15 (Y85): a UDP-
glucuronosyltransferase encoded by a polymoφhic gene. Pharmacogenetics, 7:
317-325.
25. Mackenzie PI, Owens IS, Burchell B, Bock KW, Bairoch A, Belanger A,
Fournel-Gigleux S, Green M, Hum DW, Iyanagi T, Lancet D, Louisot P,
Magdalou J, Chowdhury JR, Ritter JK, Schachter H, Tephly TR, Tipton KF and
Nebert DW (1997). The UDP glycosyltransferase gene superfamily:
recommended nomenclature update based on evolutionary divergence.
Pharmacogenetics, 7: 255-269. 26. Manach C, Morand C, Crespy V, Demigne C, Texier O, Regerat F and Remesy C
(1998). Quercetin is recovered in human plasma as conjugated derivatives which
retain antioxidant properties. FEBSLett., 426: 331-336.
27. Miners JO and Mackenzie PI (1991). Drug glucuronidation in humans. Pharmac.
Ther., 51: 347 '-369.
28. Mϋnzel PA, Schmohl S, Heel H, Kalberer K, Bock-Hennig BS and Bock KW
(1999). Induction of human UDP glucuronosyltransferases (UGT1A6, UGT1A9,
and UGT2B7) by t-butylhydroquinone and 2,3,7,8-tetrachlorodibenzo-j->-dioxin in
Caco-2 cells. Drug Metab. Dispos., 27: 569-573.
29. Radominska-Pandya A, Czernik PJ, Little JM, Battaglia E and Mackenzie PI
(1999). Structural and functional studies of UDP-glucuronosyltransferases. Drug
Metab. Reviews, 31: 817-899.
30. Ritter JK, Kessler FK, Thompson MT, Grove AD, Auyeung DJ and Fisher RA
(1999). Expression and inducibility of the human bilirubin UDP-
glucuronosyltransferase UGTIAI in liver and cultured primary hepatocytes:
evidence for both genetic and environmental influences. Hepatology, 30: 476-484.
31. Shimamura H, Suzuki H, Hanano M, Suzuki A and Sugiyama Y (1993).
Identification of tissues responsible for the conjugative metaoblism of liquiritigenin in rats: an analysis based on metabolite kinetics. Biol. Pharm. Bull,
16: 899-907.
32. Siess M-H, Mas J-P, Canivenc-Lavier M-C and Suschetet M (1996). Time course
of induction of rat hepatic drug-metabolizing enzyme activities following dietary
administration of flavonoids. J. Toxicol. Envir. Health, 49: 481-496.
33. Strassburg CP, Nguyen N, Manns MP and Tukey RH (1999). UDP-
Glucuronosyltransferase activity in human liver and colon. Gastroenterology,
116: 149-160.
34. Sun X-Y, Plouzek CA, Henry JP, Wang TTY and Phang JM (1998). Increased
UDP-glucuronosyltransferase activity and decreased prostate specific antigen
production by biochanin A in prostate cancer cells. Cancer Res., 58: 2379-2384.
35. Zhu BT, Ezell EL and Liehr JG (1994). Catechol-O-methyltransferase-catalyzed
rapid O-methylation of mutagenic flavonoids - metabolic inactivation as a
possible reason for their lack of carcinogenicity in vivo. J. Biol. Chem., 269: 292-
299. 6. Zhu BT, Taneja N, Loder DP, Balentine DA and Conney AH (1998). Effects of
tea polyphenols and flavonoids on liver microsomal glucuronidation of estradiol
and estrone. J. Steroid Biolchem. Molec. Biol, 64: 207-215.
Table 3. Chrysin glucuronidation by recombinant UGTl A isoforms. Apparent
enzyme kinetic parameters were obtained by fitting the data, as in Fig. 3, to the
Michaelis-Menten equation. Mean values from 2 experiments are shown.
UGT isoform Km V τ max /'¥ -"__T.I
(μM) (pmol/min/mg protein) (rrtl/rnin/mg protein)
UGTIAI 0.35 360 1.02
UGT1A6 12.8 157 0.012
UGT1A9 1.7 1210 0.71
UGT1A4 n.d. n.d. n.d.
n.d., Activity not detectable
EXAMPLE 3
Low Oral Bioavailability Of The Flavonoid Chrysin, An Aromatase Inhibitor.
Chrysin, a potent inhibitor of the enzyme aromatase, is currently available as a dietary supplement in health food stores. In a previous study using the human intestinal Caco-2 cells as a model, low bioavailability of chrysin was found, mainly due to extensive glucuronic acid and sulfate conjugation by the enterocytes (Biochem Pharmacol 58:431-8, 1999). In the present study we tested these findings directly in normal volunteers after oral doses (400 mg) of chrysin. Only trace levels of chrysin (<25 ng/ml) could be detected in plasma together with its sulfate conjugate (<250 ng/ml). Low levels of chrysin and the sulfate conjugate were also found in urine, accounting for <2% of the dose. The fecal excretion of unchanged chrysin, however, was very high. To be able to rationalize these observations, parallel studies were performed in bile-duct cannulated rats. Based on these studies, it is suggested that chrysin is absorbed in humans and efficiently conjugated, with the conjugates secreted both in intestine and liver by MRP2 and subsequently hydrolyzed to chrysin in the feces.
EXAMPLE 4
As shown herein, the bioflavonoid chrysin, induced UDP- glucuronosyltransferase (UGT) activity and expression in the human intestinal cell line Caco-2 (Galijatovic et al, Pharm. Res. 17:21, 2000). In the present example, the specific UGT isoform(s) that chrysin induces and whether induction of a specific isoform facilitates increased glucuronidation of the dietary colon carcinogen N-OH- PhlP were determined.
Western blot analysis using isoform specific antibodies showed that pretreatment of Caco-2 cells with 50 μM chrysin results in substantial induction of the UGT 1 Al isoform. Chrysin pretreatment had no effect on the expression of UGT 1 A6, UGT 1 A9 and UGT 2B7.
Northern blot analysis using UGT 1 Al specific probe showed markedly increased expression of mRNA after chrysin treatment. Incubation of 25 μM N-OH- PhlP with control and chrysin treated Caco-2 microsomes resulted in a 6.5 fold increase in glucuronidation of N-OH-PhlP. Diet-mediated induction of intestinal UGT can be an important protective mechanism in deactivating carcinogens and other toxic chemicals.
Human intestinal Caco-2 cells were exposed to 50 μM chrysin or vehicle (control) for 3 days. The cells were harvested and the microsomal fraction, containing UGT enzymes was prepared. Control and chrysin-treated microsomes were then incubated with 25 μM N-hydroxy-PhlP. The reaction mixtures were analyzed by HPLC. This is similar methodology that was used to show the increased bilirubin glucuronidation in chrysin-treated Hep G2 and Caco-2 cells.
In the presence of the cofactor UDPGA, the formation of N-hydroxy-PhIP N2- glucuronide was 6.5 times higher in the chrysin-treated compared with the control cell preparation. The formation of this glucuronide has previously clearly been linked to detoxification of N-hydroxy-PhIP. Similar detoxification may occur for other human colon carcinogens.
EXAMPLE 5
The examples above, using the human hepatic cell line Hep G2 and the human intestinal cell line Caco-2 have demonstrated induction of UGTl Al by the flavonoid chrysin (5,7-dihydroxyflavone), using catalytic activity assays as well as Western and Northern blotting (1,2). What features of the flavonoid structures were associated with the greatest induction as well as whether common drug metabolizing enzyme inducers also produce this induction were studied. The model used for these studies was intact Hep G2 cells with chrysin as the model substrate. Both glucuronidation and sulfation were measured. Hep G2 cells were pretreated for 3 days with 25 μM concentrations of each of 18 flavonoids (N = 4). Only 3 flavonoids demonstrated induction of glucuronidation similar to that of chrysin, i.e. 3-5-fold in the intact cells. These were apigenin, luteolin and diosmetin, all of which, like chrysin, are 5,7-dihydroxyflavones with varying substituents in the B-ring. 5- and 7-hydroxy-flavone produced a modest 1.5-2-fold induction, whereas all other flavonoids examined were without effect. In addition, the induction by 7 common drug metabolizing enzyme inducers at their optimal enzyme inducing concentrations was determined. Only 3-methylcholanthrene and oltipraz showed modest induction of chrysin glucuronidation, but not e.g., TCDD or phenobarbital. (Galijatovic et al., Pharmaceut Res 17:21,2000; 2. Walle et al., Drug Metab Disp, subm.)
Cultured human liver-derived Hep G2 cells were exposed to 25 μM of each of 18 flavonoids for three days. The culture medium was then removed. The UGT enzyme activity was measured as the amount of chrysin glucuronide formed after a 6- hour incubation of the intact cells with culture medium containing 25 μM chrysin.
Only 3 flavonoids demonstrated induction of glucuronidation similar to that of chrysin, i.e. 3-5-fold in the intact cells. These were apigenin, luteolin and diosmetin, all of which, like chrysin, are 5,7-dihydroxyflavones with varying substituents in the B-ring. Other flavonoids with this common structural feature can therefore be predicted to be inducers as well.
It will be apparent to those skilled in the art that various modifications and variations can be made in the present invention without departing from the scope or spirit of the invention. Other embodiments of the invention will be apparent to those skilled in the art from consideration of the specification and practice of the invention disclosed herein. It is intended that the specification and examples be considered as exemplary only, with a true scope and spirit of the invention being indicated by the following claims.

Claims

What is claimed is:
1. A method of treating jaundice in a neonate, comprising administering to the neonate an amount of chrysin effective to treat jaundice.
2. A method of treating hyperbilirubinemia in a neonate, comprising administering to the neonate an amount of chrysin effective to reduce the amount of billirubin in the neonate.
3. A method of increasing glucuronidation of bilirubin in a subject, comprising administering to the subject an amount of chrysin effective to increase the glucuronidation of bilirubin.
4. A method of increasing the expression of UDP-glucuronosyltransferase in a subject, comprising administering to the subject an amount of chrysin effective to increase the UDP-glucuronosyltransferase.
5. The method of claim 4, wherein the UDP-glucuronosyltransferase is a UGTl A isoform.
6. The method of claim 5, wherein the UGTl A isoform is UGTIAI .
7. The method of claim 5, wherein the UGT1A isoform isUGTlA9.
8. A method of increasing the expression of UDP-glucuronosyltransferase in a subject, comprising administering to the subject an amount of apigenin effective to increase the UDP-glucuronosyltransferase.
9. The method of claim 8, wherein the UDP-glucuronosyltransferase is a UGT1A isoform.
10. The method of claim 9, wherein the UGTl A isoform is UGTIAI .
11. The method of claim 9, wherein the UGT 1 A isoform is UGT 1 A9.
12. A method of increasing the expression of UDP-glucuronosyltransferase in a subject, comprising administering to the subject an amount of luteolin effective to increase the UDP-glucuronosyltransferase.
13. The method of claim 12, wherein the UDP-glucuronosyltransferase is a UGTl A isoform.
14. The method of claim 13 , wherein the UGT 1 A isoform is UGT 1 A 1.
15. The method of claim 13, wherein the UGT1A isoform is UGT1A9.
16. A method of increasing the expression of UDP-glucuronosyltransferase in a subject, comprising administering to the subject an amount of diosmetin effective to increase the UDP-glucuronosyltransferase.
17. The method of claim 16, wherein the UDP-glucuronosyltransferase is a UGT 1 A isoform.
18. The method of claim 17, wherein the UGTl A isoform is UGTIAI .
19. The method of claim 17, wherein the UGTl A isoform is UGT1A9.
20. A method of treating colon cancer, comprising administering an effective amount of a flavone, whereby the administration of the flavone treats the colon cancer.
PCT/US2001/004656 2000-02-11 2001-02-12 A method of treating colon cancer by administering apigenin, luteolin, diosmetin and crysin WO2001058410A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2001236982A AU2001236982A1 (en) 2000-02-11 2001-02-12 A method of treating colon cancer by administering apigenin, luteolin, diosmetinand crysin

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US18207200P 2000-02-11 2000-02-11
US60/182,072 2000-02-11
US23277100P 2000-09-15 2000-09-15
US60/232,771 2000-09-15

Publications (2)

Publication Number Publication Date
WO2001058410A2 true WO2001058410A2 (en) 2001-08-16
WO2001058410A3 WO2001058410A3 (en) 2002-01-24

Family

ID=26877757

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2001/004656 WO2001058410A2 (en) 2000-02-11 2001-02-12 A method of treating colon cancer by administering apigenin, luteolin, diosmetin and crysin

Country Status (2)

Country Link
AU (1) AU2001236982A1 (en)
WO (1) WO2001058410A2 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006105725A1 (en) * 2005-04-05 2006-10-12 Shanghai Gloriayx Biopharmaceuticals Co., Ltd A combination of luteolin and one of pt chemotherapeutics
CN104208598A (en) * 2014-09-26 2014-12-17 禤燕华 Chinese medicinal composition for treating chronic jaundice
WO2015166041A1 (en) * 2014-05-01 2015-11-05 De Montfort University Flavones as cyp1a1 inhibitors for the treatment of cancer
CN106389405A (en) * 2015-07-30 2017-02-15 中国科学院大连化学物理研究所 A class of glucuronosyltransferase UGT1A1 inducers and applications thereof
CN114053414A (en) * 2021-12-31 2022-02-18 大连医科大学 Application of flavonoid compound USP22 inhibitor in preparation of anti-tumor immunity medicine

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5126129A (en) * 1988-05-23 1992-06-30 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health & Human Services Cancer therapy using interleukin-2 and flavone compounds
US5858371A (en) * 1997-02-05 1999-01-12 Panacea Biotech Limited Pharmaceutical composition for the control and treatment of anorectal and colonic diseases
WO1999021550A1 (en) * 1997-10-24 1999-05-06 Bio-Monde Preparations Limited Use of coumarin derivatives for the treatment of digestive tract disorders
US6087385A (en) * 1998-10-30 2000-07-11 University Of Mississippi Flavonoid derivatives

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5126129A (en) * 1988-05-23 1992-06-30 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health & Human Services Cancer therapy using interleukin-2 and flavone compounds
US5858371A (en) * 1997-02-05 1999-01-12 Panacea Biotech Limited Pharmaceutical composition for the control and treatment of anorectal and colonic diseases
WO1999021550A1 (en) * 1997-10-24 1999-05-06 Bio-Monde Preparations Limited Use of coumarin derivatives for the treatment of digestive tract disorders
US6087385A (en) * 1998-10-30 2000-07-11 University Of Mississippi Flavonoid derivatives

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
KLOSS J.: 'Back to Eden', March 1972, BEREFICIAL BOOKS, NEW YORK, US XP002943606 2nd ed. * page 505 - page 506 * *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006105725A1 (en) * 2005-04-05 2006-10-12 Shanghai Gloriayx Biopharmaceuticals Co., Ltd A combination of luteolin and one of pt chemotherapeutics
WO2015166041A1 (en) * 2014-05-01 2015-11-05 De Montfort University Flavones as cyp1a1 inhibitors for the treatment of cancer
CN104208598A (en) * 2014-09-26 2014-12-17 禤燕华 Chinese medicinal composition for treating chronic jaundice
CN106389405A (en) * 2015-07-30 2017-02-15 中国科学院大连化学物理研究所 A class of glucuronosyltransferase UGT1A1 inducers and applications thereof
CN114053414A (en) * 2021-12-31 2022-02-18 大连医科大学 Application of flavonoid compound USP22 inhibitor in preparation of anti-tumor immunity medicine

Also Published As

Publication number Publication date
AU2001236982A1 (en) 2001-08-20
WO2001058410A3 (en) 2002-01-24

Similar Documents

Publication Publication Date Title
Galijatovic et al. Induction of UDP-glucuronosyl-transferase by the flavonoids chrysin and quercetin in Caco-2 cells
Walle et al. Induction of UDP-glucuronosyltransferase UGT1A1 by the flavonoid chrysin in the human hepatoma cell line hep G2
Galijatovic et al. Extensive metabolism of the flavonoid chrysin by human Caco-2 and Hep G2 cells
Sha et al. Stimulation of free radical formation by aminoglycoside antibiotics
Chen et al. Metabolism of flavonoids in human: a comprehensive review
Chen et al. Metabolism of flavonoids via enteric recycling: role of intestinal disposition
Walle et al. Induction of human UDP-glucuronosyltransferase UGT1A1 by flavonoids—structural requirements
Reen et al. Impairment of UDP-glucose dehydrogenase and glucuronidation activities in liver and small intestine of rat and guinea pig in vitro by piperine
O’Leary et al. Metabolism of quercetin-7-and quercetin-3-glucuronides by an in vitro hepatic model: the role of human β-glucuronidase, sulfotransferase, catechol-O-methyltransferase and multi-resistant protein 2 (MRP2) in flavonoid metabolism
Williams et al. Comparative studies on the effects of green tea extracts and individual tea catechins on human CYP1A gene expression
Debiton et al. Sanguinarine-induced apoptosis is associated with an early and severe cellular glutathione depletion
Lotito et al. Metabolic conversion of dietary flavonoids alters their anti-inflammatory and antioxidant properties
Jeong et al. Coupling of conjugating enzymes and efflux transporters: impact on bioavailability and drug interactions
Chatterjee et al. Inhibitors of poly (ADP-ribose) synthetase protect rat proximal tubular cells against oxidant stress
Trottier et al. Human UDP‐glucuronosyltransferase (UGT) 1A3 enzyme conjugates chenodeoxycholic acid in the liver
Pias et al. Apoptosis in mitotic competent undifferentiated cells is induced by cellular redox imbalance independent of reactive oxygen species production
Ma et al. Disposition of flavonoids impacts their efficacy and safety
Zhu et al. Effects of tea polyphenols and flavonoids on liver microsomal glucuronidation of estradiol and estrone
Totta et al. Daidzein-sulfate metabolites affect transcriptional and antiproliferative activities of estrogen receptor-β in cultured human cancer cells
EP1836320A2 (en) Compositions for modulation of parp and methods for screening for same
Waldron et al. Increased AICD generation does not result in increased nuclear translocation or activation of target gene transcription
Marc et al. Regulation of phenobarbital induction of the cytochrome P450 2b9/10 genes in primary mouse hepatocyte culture: Involvement of calcium‐and cAMP‐dependent pathways
Vahrmeijer et al. Effect of glutathione depletion on inhibition of cell cycle progression and induction of apoptosis by melphalan (L-phenylalanine mustard) in human colorectal cancer cells
Barrington et al. MK571 inhibits phase-2 conjugation of flavonols by Caco-2/TC7 cells, but does not specifically inhibit their apical efflux
WO2001058410A2 (en) A method of treating colon cancer by administering apigenin, luteolin, diosmetin and crysin

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase in:

Ref country code: JP