US20100210869A1 - Ethanol extract of antrodia camphorata for inducing apoptosis and preparation method thereof - Google Patents

Ethanol extract of antrodia camphorata for inducing apoptosis and preparation method thereof Download PDF

Info

Publication number
US20100210869A1
US20100210869A1 US12/538,549 US53854909A US2010210869A1 US 20100210869 A1 US20100210869 A1 US 20100210869A1 US 53854909 A US53854909 A US 53854909A US 2010210869 A1 US2010210869 A1 US 2010210869A1
Authority
US
United States
Prior art keywords
ethyl acetate
extract
ethanol
extracting
fruiting body
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US12/538,549
Inventor
Yang-Chang Wu
Mei-Chin Lu
Fang-Rong Chang
Ying-Chi Du
Tung-Ying Wu
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Kaohsiung Medical University
Original Assignee
Kaohsiung Medical University
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 Kaohsiung Medical University filed Critical Kaohsiung Medical University
Assigned to KAOHSIUNG MEDICAL UNIVERSITY reassignment KAOHSIUNG MEDICAL UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHANG, FANG-RONG, DU, YING-CHI, LU, MEI-CHIN, WU, TUNG-YING, WU, YANG-CHANG
Publication of US20100210869A1 publication Critical patent/US20100210869A1/en
Priority to US13/398,368 priority Critical patent/US8883167B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/06Fungi, e.g. yeasts
    • A61K36/07Basidiomycota, e.g. Cryptococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/21Hydrocarbon
    • Y10T436/214Acyclic [e.g., methane, octane, isoparaffin, etc.]
    • Y10T436/216Unsaturated [e.g., ethylene, diene, etc.]

Definitions

  • the present invention relates to an extract of the fruiting body of Antrodia camphorata (AC) and the preparation method thereof.
  • the present invention related to an ethanol extract of the fruiting body of A. camphorata (abbreviated as EEAC) and the preparation method thereof.
  • EEAC can effectively induce cancer apoptosis.
  • the organic solvent extracts are obtained after EEAC is extracted with organic solvents of increasing polarities and can effectively inhibit proliferation, induce cancer cells to enter cell death and apoptosis program.
  • Antrodia camphorata by name niu-chang-chih or jang-jy is an endemic fungus in Taiwan and grows in the internal heartwood (or the dark/humid wood surface) of the particular Cinnamomum kanehirai in 400 to 2000 meters altitude. Therefore, it is uneasily to find out the wide fruiting body of AC or identify the morphological appearance of this Aphyllophorales fungus. In addition, the price of AC is still high due to their biologically active components having potential pharmaceutical value.
  • mycelia products of AC are popular in the market and announce to own anticancer activity, reduced treatment-related symptoms and other side effects.
  • mycelia products of AC have recently been reported to have anti-oxidant, antihypersensitive and immunostimulatory effects (Liu et al., 2007). It has been claimed of these mycelia products that they contain active components similar to the wild fruiting bodies with cytotoxic triterpenes, steroids, as well as immunostimulatory polysaccharides reported previously (Chen et al., 2005; Yang et al., 1996).
  • AC has been used as health food to prevent inflammation, hypertension, itchy skin and liver cancer. Therefore, extracts of mycelia and fruiting body of AC are deemed as a potential chemotherapeutic agent against hepatoma, as well as prostate, bladder, lung cancer cells and so on (Chen et al., 2007; Hsu et al., 2007; Peng et al., 2007; Song et al., 2005; Wu et al., 2006). However, the chemical distribution and pharmacological research of niu-chang-chih products are not clarified up to now.
  • Taiwan Patent No. 1299665 discloses the extract of AC and the preparation thereof, in which the mycelia of AC is extracted with ethanol to obtain polysaccharides for inhibiting matrix metalloproteinase activities.
  • the extract is not extracted with the fruiting body of AC, and the mycelia product thereof cannot inhibit cancer cell growth.
  • Taiwan Patent No. 1279439 discloses that the mycelia of AC is cultured to obtain the cultured products by adjusting pH value of medium.
  • Taiwan Patent No. 591110 discloses that ⁇ -aminobutyric acid is extracted from the lyophilized mycelia of AC with water or organic solvents.
  • the above-mentioned inventions did not disclose any product of the fruiting body of AC extracted with water or organic solvent, and there is no component for cancer therapy or growth inhibition of cancer cell is identified.
  • an ethyl acetate extract of the fruiting body of AC includes at least one characteristic of: (a) causing a plurality of methyl signals generated from sterol or triterpene at ⁇ H 0.5 to 2 and terminal olefinic protons at ⁇ H 4.8 to 5.0 on a first 1 H NM spectrum when the ethyl acetate extract is solved in a deuterium chloroform (CDCl 3 ); and (b) causing signals of terminal olefinic protons at ⁇ H 5.0 to 5.4 on a second 1 H NM spectrum when the ethyl acetate extract is solved in pyridine-D5 (C 5 D 5 N).
  • the ethyl acetate extract further includes at least one compound being one selected from a group consisting of zhankuic acid A, dehydroeburicoic acid, actcin C and zhankuic acid C.
  • zhankuic acid A has characteristic 1 H NMR signals of two tertiary methyls (CH 3 -18 and CH 3 -19) and terminal olefinic protons (H 2 -28) in 24-exo-methylene-26-oic acid side-chain, and the ethyl acetate extract further has characteristic signals of triterpenoid the same with those of the zhankuic acid A.
  • the ethyl acetate extract is fractionized from an ethanol extract of the fruiting body of AC.
  • a method for measuring an existence and an amount of triterpenoid of a target object includes steps of: (a) extracting the target object with a first ethanol solution to obtain an ethanol extract; (b) sequentially extracting the ethanol extract with n-hexane and ethyl acetate to obtain an ethyl acetate extract; and (c) measuring the existence and the amount of triterpenoid in the ethyl acetate extract.
  • the existence of the triterpenoid is performed is performed by a nuclear magnetic resonance at a resolution of at least 200 MHz
  • the ethyl acetate extract is solved in a pyridine-D5 (C 5 D 5 N)
  • the amount of the triterpenoid is performed by a high performance liquid chromatography (HPLC).
  • the target object is the fruiting body of AC.
  • the step (a) further includes a step (a1) of grinding the fruiting body.
  • the step (c) further includes a step (c1) of extracting the ethanol extract with at least one organic solvent to obtain an organic solvent extract.
  • the at least one organic solvent has a total number of species more than one, each of the at least one organic solvent has a polarity, and the step (c1) has a extracting sequence that the polarity of a specific one of the at least one organic solvent is higher than that of a preceding one of the specific organic solvent.
  • the at least one organic solvent is one selected from a group consisting of n-hexane, ethyl acetate, a second ethanol solution and a combination thereof.
  • the step (c1) further includes steps of: (c11) extracting the ethanol extract with n-hexane to obtain an n-hexane extract and a first residue; (c12) extracting the first residue with ethyl acetate to obtain an ethyl acetate extract and a second residue; and (c13) extracting the second residue with a second ethanol solution to obtain a second ethanol extract.
  • the step (c12) further includes a step (c121) of isolating the ethyl acetate extract with a plurality of trichloromethane-methanols to obtain zhankuic acid A, wherein the plurality of trichloromethane-methanols have respective ratios of trichloromethane to methanol.
  • the step (c12) further includes a step (c121) of isolating the ethyl acetate extract with a plurality of n-hexane-ethyl acetate-methanols to obtain a plurality of fractions of the ethyl acetate extract, wherein the plurality of n-hexane-ethyl acetate-methanols have respective ratios of n-hexane-ethyl acetate to methanol.
  • the step (c121) further includes at least one step of: (c121-1) sequentially isolating a first one of the plurality of fractions with ethyl acetate-dichloromethane-methanol, trichloromethane-methanol and methanol-water to obtain dehydroeburicoic acid; (c121-2) sequentially isolating a second one of the plurality of fractions with ethyl acetate-dichloromethane-methanol and acetonitrile-water having a first ratio of acetonitrile to water to obtain zhankuic acid A; (c121-3) sequentially isolating a third one of the plurality of fractions with dichloromethane-methanol and acetonitrile-water having a second ratio of acetonitrile to water to obtain actcin C; and (c121-4) sequentially isolating a fourth one of the plurality of fractions with dichloromethane-methanol and
  • the step (b) further generates a third residue
  • the method further includes a step (c1) of extracting the third residue with water to obtain a water extract.
  • the ethanol extract is used to treat a cell for regulating expression of a specific protein of the cell.
  • the cell is a leukemia cell
  • the specific protein is one selected from a group consisting of histone, histone deacetylase, histone acetylase, a p21, a poly (ADP-ribose) polymerase (PARP), a Bax, a Bcl-2, a death receptor (DR5), a tumor necrosis factor receptor-associated death domain (TRADD), a p50 and a p65 and a combination thereof.
  • the ethanol extract and 7-[4-(dimethylamino)phenyl]-N-hydroxy-4,6-dimethyl-7-oxohepta-2,4-dienamide have a synergistic effect of causing a cell to go into apoptosis.
  • FIG. 1 illustrates the flowchart for preparing EEAC and its organic solvent extracts in the present invention
  • FIG. 2(A) illustrates the growth inhibition of the respective extracts of the fruiting body of AC on HL 60 cells
  • FIG. 2(B) illustrates the apoptosis of HL 60 cells induced by respective extracts of the fruiting body of AC;
  • FIG. 3(A) illustrates the 1 H NMR pattern of the ethyl acetate extract of the fruiting body of AC solved in CDCl 3 ;
  • FIG. 3(B) illustrates the 1 H NMR pattern of zhankuic acid A solved in CDCl 3 ;
  • FIG. 4 illustrates the effect of EEAC on DNA fragmentation in HL 60 cells
  • FIG. 5(A) illustrates the immunoblotting patterns showing the effect of EEAC on histone acetylation in HL 60 cells
  • FIG. 5(B) illustrates the immunoblotting patterns showing the activity of the histone acetylation-related enzymes regulated by EEAC in HL 60 cells;
  • FIG. 6 illustrates the synergistic effect of EEAC and TSA on the survival in HL 60 cells
  • FIGS. 7(A) and 7(B) respectively are the immunoblotting patterns showing various protein expressions of TSA, EEAC and the combination thereof in HL 60 cells, wherein FIG. 7(A) shows expressions of p21, PARP, Bcl-2 and Bax, and FIG. 7(B) shows expressions of p50, p65, TRADD and DR5;
  • FIG. 8 illustrates the NFKB p65 activity of TSA, EEAC and the combination thereof in HL 60 cells
  • FIGS. 9(A) to 9(E) respectively show 1 H NMR spectra of (A) EEAC, (B) FC, (C) FA, (D) FB and (E) zhankuic acid A solved in C 5 D 5 N at 400 MHz; and
  • FIGS. 10(A) and 10(B) respectively show HPLC profiles of FA at (A) 254 nm and (B) 270 nm.
  • the supernatants were filtered and then centrifuged at 3,000 rpm for 30 minutes to remove the precipitate.
  • the product named as “the water extract of the fruiting body of A. camphorata (WEAC)”, was lyophilized and stored at ⁇ 70° C.
  • EEAC biologically active components of EEAC
  • 1 g of the dried fruiting body of AC was ground and extracted according to the method in Experiment 1.
  • Fifty-percent inhibition concentration (IC 50 ) of EEAC was determined as 104.82 ⁇ g/ml by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay; however, WEAC was inactive (IC 50 >200 ⁇ g/ml).
  • EEAC (203.1 mg) was extracted with solvents of increasing polarities (n-hexane, ethyl acetate and ethanol sequentially), and three different extracts, 12.8% (26.0 mg) of n-hexane extract (FC), 61.6% (125.1 mg) of ethyl acetate extract (FA) and 8.4% (17.0 mg) of ethanol extract (FB), and 10.0% (20.2 mg) of the insoluble residue were yielded. Percentage is referred to yields in weight percentage of EEAC, milligram is referred to the corresponding dried weight after fractionation.
  • EEAC fraction of 95.4 mg was chromatographed using Sephadex LH-20 with trichloromethane-methanol (CHCl 3 -MeOH) (1:3) to obtain three fractions, wherein Fraction 2 was further separated by preparative thin layer chromatography (TLC) with CHCl 3 -MeOH (25:1) to obtain 8 sub-fractions.
  • Sub-fraction 2-1 of 10.3 mg was purified using ODS HPLC column (250 ⁇ 10 mm, Hypersil®, MeOH—H 2 O (85:15)) to obtain 4.3 mg of zhankuic acid A (retention time (R t ) 10 min, flow rate 2 ml/min).
  • Human leukemia HL 60 cell line was purchased from American Type Culture Collection (ATCC, Manassas, Va.) and incubated in RPMI 1640 medium supplemented with 10% fetal calf serum, 2 mM glutamine, 100 units/ml penicillin and 100 ⁇ g/ml streptomycin at 37° C. in a humidified atmosphere of 5% carbon dioxide.
  • MTT assay Cell viability was determined by the trypan blue dye exclusion and cytotoxicity was assessed by MTT assay.
  • 1 ⁇ 10 5 HL 60 cells were plated in 96-well plates and treated with different concentrations of EEAC dissolved in dimethyl sulfoxide (DMSO) at the concentrations of 0 to 150 ⁇ g/ml for 24 hours. Death cells then were stained with trypan blue, and the survival cells were calculated with hemocytometer.
  • DMSO dimethyl sulfoxide
  • MTT assay 1.2 mg/ml MTT was added in each well, which was incubated for 2 hours to form crystal violet, formazan. Formazan was dissolved in DMSO, and absorbance was detected at a wavelength of 570 nm.
  • HL 60 cells (1 ⁇ 10 6 ) were incubated in the 10-cm dish and treated with known concentrations of EEAC for 24 hours.
  • the treated cells were washed with ice-cold phosphate-buffered saline (PBS) twice and harvested by centrifuging at 37° C. at 200 ⁇ g for 5 minutes.
  • Cells were fixed with 70% (v/v) ethanol at 37° C. for 30 minutes, and then were resuspended in 1 ml propidium iodium (PI) reagent (containing 0.1% Triton X-100, 100 ⁇ g/ml ribonuclease (RNase) A and 500 ⁇ g/ml PI) at 37° C. for 30 minutes.
  • PI propidium iodium
  • the cells were detected with the flow cytometer, and data were analyzed with FACScan and Cell Quest Program (Becton Dickinson).
  • DNA fragmentation in the apoptotic cells were determined by gel electrophoresis.
  • HL 60 Cells (1 ⁇ 10 6 ) were incubated in the 10-cm dish for 24 hours, then were treated with known concentrations of EEAC (0, 50, 100, 150 and 200 ⁇ g/ml respectively) for another 24 hours.
  • Control group was the cells with DMSO treatment.
  • the cells were washed with PBS twice and resuspended in the solution A (containing 10 ⁇ M EDTA (2-[2-(bis(carboxymethyl)amino)ethyl-(carboxymethyl)amino]acetic acid), 50 mM Tris-HCl (pH 8.0), 0.5% (v/v) sodium dodecyl sulfate (SDS) and 1 mg/ml proteinase K) at 56° C. for 3 hours. Subsequently, RNase at a final concentration of 50 ⁇ g/ml was added therein for reacting 1 hour. Cellular DNA was extracted with phenol/chloroform/isopropanol (25:24:1, v/v), and cellular DNA fragmentation was determined by gel electrophoresis so as to evaluate apoptosis.
  • solution A containing 10 ⁇ M EDTA (2-[2-(bis(carboxymethyl)amino)ethyl-(carboxymethyl)amino
  • the cells were resuspended in cold sterile water, and sulfuric acid was added therein to 0.4 N.
  • the cells were centrifuged at 10,000 ⁇ g for 5 minutes after incubation on ice for 1 hour. Total histones in the supernatant were precipitated overnight with 10 ⁇ volumes of acetone at ⁇ 20° C.
  • the precipitated histones were collected by centrifugation, dried and resuspended in distilled water, and the protein content was determined by the Bio-Rad protein assay kit (BioRad Laboratories GmbH, Munchen, Germany).
  • the harvested cells were treated with RIPA lysogenic buffer (containing 1 ⁇ PBS, 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% SDS, 1 mM sodium orthovanadate, 100 ⁇ g/ml PMSF and 30 mg/ml aprotinin) for 30 minutes (Lu et al., 2006), and then was centrifuged at 20,000 ⁇ g for 30 minutes.
  • the protein concentration was determined by BCA protein assay kit (Pierce, Rockford, Ill.). Protein was resolved by 7.5%, 10% or 12% SDS-polyacrylamide gel electrophoresis (PAGE) respectively and transferred onto the PVDF nitrocellulose membrane. Protein expressions were sequentially monitored with primary antibody and horse-radish peroxidase (HRP)-conjugated secondary antibody, and then was developed with enhanced chemical luminescence (ECL). Protein expressions were exposed on X-ray film.
  • RIPA lysogenic buffer containing 1 ⁇ PBS, 1% Nonidet P-40, 0.5% sodium
  • This experiment is to determine the effect of EEAC treatment on NFKB activation by TranAM® transcription factor assay (Active Motif, Tokyo, Japan) (Schwab et al., 2007).
  • 7 ⁇ g of nuclear extract was added to each well of a 96-well culture plate which was pre-coated with NF ⁇ B consensus binding oligonucleotide (SEQ ID NO. 1). After 1 hour of incubation with smooth agitation, wells were washed three times with washes buffer and then incubated with p50 or p65 antibody (dilution 1:1,000) for 1 hour at room temperature.
  • the wells were incubated with HRP-conjugated secondary antibody (dilution 1:1,000) for 1 hour at room temperature followed by the addition of developing solution.
  • the optical density was determined at 405 nm by keeping the reference wavelength at 655 nm.
  • FIG. 1 illustrates the flowchart for preparing EEAC and its organic-solvent extracts in the present invention.
  • the ground fruiting body of AC was extracted with ethanol (Steps 101 , 102 ), and then was filtered and centrifuged to obtain EEAC (Step 103 ) and the residue of the fruiting body of AC (Step 110 ).
  • EEAC was further extracted with the organic solvents of increasing polarity, which were ranked: n-hexane, ethyl acetate and ethanol.
  • the preparation method of the organic solvent extracts of the fruiting body of AC was described as follows.
  • EEAC was extracted with n-hexane to obtain the n-hexane extract (fraction C, FC) (Step 104 ) and the first residue (Step 105 ).
  • the first residue was further extracted with ethyl acetate to obtain the ethyl acetate extract (fraction A, FA) (Step 106 ) and the second residue (step 107 ).
  • the second residue was further extracted with ethanol solution to obtain the another ethanol extract (fraction B, FB) (step 108 ) and the third residue (step 109 ).
  • the residue (step 110 ) was further boiled and extracted with water (step 111 ) to obtain the water extract of the fruiting body of AC (WEAC) (step 112 ).
  • FIG. 2(A) illustrates the growth inhibition of the respective extracts of fruiting body of AC on HL 60 cells.
  • growth inhibition on HL 60 cells was increased with the increasing concentrations of the respective extracts.
  • FIG. 2(B) which illustrates the apoptosis of HL 60 cells induced by 100 ⁇ g/ml of respective extracts.
  • FIG. 2(B) comparing with other extracts, FA showed the significant apoptotic ability on HL 60 cells. From the above-mentioned results, it can be known that FA contains the biologically active component for cytotoxicity on HL 60 cells.
  • the 1 H NMR spectrum of FA was further analyzed.
  • the methyl signals (CH 3 -18 and CH 3 -19) of sterol or triterpene was shown at 6 0.5 to 2 ( FIG. 3(A) ) when the NMR profile of FA was 3.0 mg/ml in deuterium chloroform (CDCl 3 ), and the major component of FA was predicted as zhankuic acid A from the spectrum of FIG. 3(A) .
  • FIG. 3 (B) comparing 1 H NMR spectrum of FA with that of zhankuic acid A at the same conditions (FIG. 3 (B)), it can be found that FA has the similar signals of zhankuic acid A.
  • the major component, zhankuic acid A, of FA was a pale-yellow amorphous solid, and its molecular formula was established as C 29 H 40 O 5 by electrospray ionization mass spectra (ESIMS, m/z 469 [M+H] + ) and NMR spectra.
  • EIMS electrospray ionization mass spectra
  • NMR spectra NMR spectra.
  • FIG. 4 illustrates the effect of EEAV on DNA fragmentation in HL 60 cells. It can be known from FIG. 4 that the concentration of EEAC is proportional to the DNA fragmentation of HL 60 cells, which means EEAC has significant cytotoxicity to HL 60 cells.
  • FIG. 5(A) illustrates the immunoblotting patterns showing the effect of EEAC on histone acetylation in HL 60 cells.
  • FIG. 5(A) illustrates the immunoblotting patterns showing the effect of EEAC on histone acetylation in HL 60 cells.
  • EEAC showed the dose-dependent relationship to the low level acetylation of histones (H3, H3K18 and H3K9) in HL 60 cells.
  • FIG. 5(B) illustrates the immunoblotting patterns showing the activity of the histone acetylation-related enzymes regulated by EEAC in HL 60 cells.
  • deacetylations of histone H3, including inhibition of GCN5, CBP, SRC-1 and PCAF and promotion of HDAC1 were regulated by EEAC with HAT and HDAC.
  • Trichostatin A is a microbial metabolite, which can promote acetylation of histone H3. Since H3 acetylation is accumulated (hyper-acetylation), trichostatin A can effectively inhibit leukemia proliferation.
  • FIG. 6 illustrates the synergistic effect of EEAC and TSA on the survival in HL 60 cells.
  • EEAC had better cytotoxicity effect on HL 60 cells than TSA.
  • the combined treatment of 100 g/ml EEAC and different dosages of TSA had synergistic effect on HL 60 cells, and the cytotoxicity effect was increased with the increasing TSA dosages.
  • FIGS. 7(A) and 7(B) respectively are the immunoblotting patterns showing various protein expressions of TSA, EEAC and the combination thereof in HL 60 cells.
  • FIG. 7(A) was subjected to p21, PARP, Bcl-2 and Bax which related to growth inhibition and apoptotic induction. It is found that p21 expression of HL 60 cells is increased and Bcl-2 and cleaved PARP expressions thereof are inhibited by cooperatively treating TSA with EEAC.
  • A. camphorata is potent FAS (TRAIL)-sensitizing agent in a variety of different TRAIL-resistant human cancer cells (Hsu et al., 2005), DR5 and TRADD interact specifically with death domain of DR4. DR4 activation promotes the binding of TRADD and the subsequent recruitment of DR5 to promote apoptosis (Swarup et al., 2007).
  • TRAIL TRAIL-sensitizing agent in a variety of different TRAIL-resistant human cancer cells
  • DR4 activation promotes the binding of TRADD and the subsequent recruitment of DR5 to promote apoptosis (Swarup et al., 2007).
  • TSA or EEAC When HL 60 cells were individually treated TSA or EEAC or cooperatively treated with TSA and EEAC, it can be found that the immunoblotting analysis for TRADD and DR5 revealed 2 and 4 fold increases over the corresponding control and TSA treatment alone ( FIG. 7(B) ). It can be known
  • TRAIL is one of the promising new candidates for cancer therapeutics.
  • NF ⁇ B activation can be triggered by direct stimulation of CD95 and the TRAIL death receptors as well as by overexpression of FLIP protein, and NF ⁇ B transcription factor differentially regulated the DR5 expression involving HDAC1 (Shetty et al., 2005).
  • NF ⁇ B activation on cellular apoptosis in response to combined treatment, it is found that the combined treatment significantly increases the translocation of NF ⁇ B p65, but does not affect the expression with total extracts of cells.
  • combination-induced p65 translocation may be mediated by EEAC treatment alone.
  • the obtained EEAC can effectively inhibit growth of HL 60 cells and induce apoptosis mediated histone hypoacetylation and suppression of acetyltransferase activity (including GCN5, CBP and PCAF) in HL 60 cells.
  • apoptosis mediated histone hypoacetylation and suppression of acetyltransferase activity including GCN5, CBP and PCAF
  • combined treatment of EEAC and TSA on HL 60 cells can synergistically inhibit cellular growth and induce apoptosis.
  • EEAC Zhankuic acid A was identified as the major active component in EEAC after the respective organic solvent extracts obtained by further extracting EEAC with organic solvents and NMR analysis. Accordingly, EEAC can be the potential chemotherapeutic agent.
  • Embodiment used another fruiting body of AC to prepare 11 g of lyophilized EEAC according to the preparation method in Experiment 1 of Embodiment 1. Subsequently, EEAC was sequentially extracted with n-hexane, ethyl acetate and ethanol according to the method in Experiment 2 of Embodiment 1 to obtain three different extracts, 12.7% (1.4 g) FC, 61.8% (6.8 g) FA and 10.0% (1.1 g) FB, and 5.5% (0.6 g) insoluble residue. Percentage is referred to the weight percentage in EEAC, gram is referred to the dry weight after extraction.
  • the respective lyophilized EEAC (100 mg) in Embodiment 2 was extracted with three partition extraction methods, and anti-leukemia activity between the respective obtained extracts and the extracts in Embodiment 2 was compared.
  • 101.9 mg lyophilized EEAC was fractionation with ethyl acetate and water (100 ml each, 1:1 (v/v)) by liquid-liquid partition extraction according to the literatures of Chen et al., 1995 and Hsu et al., 2007.
  • the ethyl acetate extract of the fruiting body of AC FEA-EA, 78.1%, 79.6 mg
  • the water extract of the fruiting body of AC FW-EA, 19.6%, 20.0 mg
  • Table 1 illustrates the comparison of anti-leukemia activity of the above-mentioned extracts.
  • FEA exhibited the best activity that demonstrated the repeatability in production for concentrating cytotoxic components in EEAC. Comparing antiproliferative activity of FA with those in Control 1, FA (104.10 ⁇ g/ml) were more potent than FEA-EA (156.47 ⁇ g/ml) which prepared by the acceptable partition method in food science. Although the fractions (FC13-C13 and FC12-C12) had shown competitive antiproliferative activity referring to FEA, usage of those chlorine-containing organic solvents were limited or forbidden. Therefore, the methods in the prior art are not ideal preparation procedures.
  • Embodiment 2 TABLE 1 Antiproliferative activity of extracts in Embodiment 2 and the respective controls Product Anti-leukemia activity (IC 50 ) ( ⁇ g/ml) Embodiment 2 EEAC 182.94 FC 156.36 FA 104.10 FB >200 Control 1 FEA-EA 156.47 FW-EA >200 Control 2 FCl3-Cl3 126.04 FW-Cl3 >200 Control 3 FCl2-Cl2 115.03 FW-Cl2 >200
  • FIGS. 9(A) to 9(E) respectively show 1 H NMR spectra of (A) EEAC, (B) FC, (C) FA, (D) FB and (E) zhankuic acid A solved in C 5 D 5 N at 400 MHz.
  • the first sub-fraction (Fraction 4-1, 568.6 mg) was further chromatographed with CHCl 3 -MeOH (35:1) in Silica gel 60 to yield 3 another sub-fractions.
  • Fifty milligrams (50 mg) from 240.3 mg of the second sub-fraction (Fraction 4-1-2) was purified with ODS HPLC column (250 ⁇ 10 mm, Hypersil®, methanol-water (90:10)) to obtain 10.3 mg dehydroeburicoic acid (R t 27 min, flow rate 2 ml/min).
  • HPLC spectra of four obtained major active components and FA were compared.
  • the HPLC conditions were provided as follows.
  • HPLC was Shimadzu LC-10AT, the detector was Shimadzu SPD-M10A photodiode array detector, the auto sampler was Shimadzu SIL-20A prominence auto sampler;
  • the HPLC column was Cosmosil 5C-18-MS-II (250 ⁇ 4.6 mm), solvent A in mobile phase was acetonitrile and solvent B was water (in 0.1% acetic acid), flow rate was 1 ml/min, column temperature was room temperature, and detection wavelength was 254 nm and 270 nm.
  • zhankuic acid A, antcin C and zhankuic acid C are stereo isomers having the chiral center at C-25 position, but they cannot be purified at the isolation procedures.
  • two stereo isomers can be separated and observed, i.e. two peaks represented on the chromatographic spectrum, at the above-mentioned HPLC conditions.
  • FIG. 10(A) and Table 2 show comparisons of peak area and height for the respective components in FA after with HPLC at 254 nm wavelength.
  • zhankuic acid A represented two peaks at 254 nm wavelength and retention time of 46.739 and 47.347 min, and the determined area occupied 16.26% and the determined height occupied 17.27% of the total active layer.
  • Zhankuic acid A had the highest area and height in four major active components.
  • FIG. 10(B) and Table 3 shows comparisons of peak area and height of the respective components in FA with HPLC at 270 nm wavelength.
  • zhankuic acid A represented two peaks at 270 nm wavelength and retention time of 46.736 and 47.352 min, and the determined area occupied 26.53% and the determined height occupied 27.86% of the total active layer.
  • Zhankuic acid A also had the highest peak height in four major active components.
  • zhankuic acid A is the major active component in EEAC
  • antcin C, zhankuic acid C and dehydroeburicoic acid are another ones.
  • FA is defined as the triterpenoids-rich fraction and shows antiproliferative activity in HL 60 cells. Accordingly, the above-mentioned experimental method can be the power tool for detecting the components of triterpenoids of A. camphorata in the industry.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Natural Medicines & Medicinal Plants (AREA)
  • Mycology (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medical Informatics (AREA)
  • Microbiology (AREA)
  • Epidemiology (AREA)
  • Alternative & Traditional Medicine (AREA)
  • Botany (AREA)
  • Biotechnology (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines Containing Plant Substances (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A preparation method for an ethanol extract of the fruiting body of Antrodia camphorata (EEAC) is provided. The preparation method includes steps of: (a) providing the fruiting body of A. camphorata (AC); (b) extracting the fruiting bodies with a first ethanol solution; and (c) obtaining EEAC. EEAC further can be sequentially extracted or fractioned by n-hexane, ethyl acetate and ethanol, and an n-hexane fraction (FC), an ethyl acetate fraction (FA) and an ethanol fraction (FB) respectively are generated. The growth inhibition and apoptosis induction of leukemia cell line HL 60 are effectively mediated by FA product, in which zhankuic acid A is the bioactive marker. The amount of triterpenoid in the fruiting body of AC can be determined by NMR and HPLC analysis.

Description

    FIELD OF THE INVENTION
  • The present invention relates to an extract of the fruiting body of Antrodia camphorata (AC) and the preparation method thereof. In particular, the present invention related to an ethanol extract of the fruiting body of A. camphorata (abbreviated as EEAC) and the preparation method thereof. EEAC can effectively induce cancer apoptosis. Further, the organic solvent extracts are obtained after EEAC is extracted with organic solvents of increasing polarities and can effectively inhibit proliferation, induce cancer cells to enter cell death and apoptosis program.
  • BACKGROUND OF THE INVENTION
  • Antrodia camphorata (AC), by name niu-chang-chih or jang-jy is an endemic fungus in Taiwan and grows in the internal heartwood (or the dark/humid wood surface) of the particular Cinnamomum kanehirai in 400 to 2000 meters altitude. Therefore, it is uneasily to find out the wide fruiting body of AC or identify the morphological appearance of this Aphyllophorales fungus. In addition, the price of AC is still high due to their biologically active components having potential pharmaceutical value.
  • Since the fruiting body of AC cannot be easily found and be artificially cultured, mycelia products of AC are popular in the market and announce to own anticancer activity, reduced treatment-related symptoms and other side effects. In addition, mycelia products of AC have recently been reported to have anti-oxidant, antihypersensitive and immunostimulatory effects (Liu et al., 2007). It has been claimed of these mycelia products that they contain active components similar to the wild fruiting bodies with cytotoxic triterpenes, steroids, as well as immunostimulatory polysaccharides reported previously (Chen et al., 2005; Yang et al., 1996).
  • Traditionally AC has been used as health food to prevent inflammation, hypertension, itchy skin and liver cancer. Therefore, extracts of mycelia and fruiting body of AC are deemed as a potential chemotherapeutic agent against hepatoma, as well as prostate, bladder, lung cancer cells and so on (Chen et al., 2007; Hsu et al., 2007; Peng et al., 2007; Song et al., 2005; Wu et al., 2006). However, the chemical distribution and pharmacological research of niu-chang-chih products are not clarified up to now.
  • In addition, Taiwan Patent No. 1299665 discloses the extract of AC and the preparation thereof, in which the mycelia of AC is extracted with ethanol to obtain polysaccharides for inhibiting matrix metalloproteinase activities. However, the extract is not extracted with the fruiting body of AC, and the mycelia product thereof cannot inhibit cancer cell growth. Taiwan Patent No. 1279439 discloses that the mycelia of AC is cultured to obtain the cultured products by adjusting pH value of medium. However, there is no extraction method disclosed. Taiwan Patent No. 591110 discloses that γ-aminobutyric acid is extracted from the lyophilized mycelia of AC with water or organic solvents. However, the above-mentioned inventions did not disclose any product of the fruiting body of AC extracted with water or organic solvent, and there is no component for cancer therapy or growth inhibition of cancer cell is identified.
  • It is therefore attempted by the applicant to deal with the above situation encountered in the prior art.
  • SUMMARY OF THE INVENTION
  • To overcome the defect that the effective/uniform anticancer components of AC products cannot be obtained from extraction of the mycelia of AC since the anti-cancer triterpenoid does not exist in the mycelia of AC, in the present invention, the wild fruiting body of AC is extracted with ethanol to obtain the ethanol extract of the fruiting body of A. camphorata (EEAC) so as to effectively inhibit growth of leukemia and induce apoptosis. Next, the organic solvent extracts of the fruiting body of AC are obtained by extracting EEAC with the organic solvents of increasing polarities, and zhankuic acid A is identified as the effective anticancer component in the organic solvent extract by high performance liquid chromatography (HPLC) and nuclear magnetic resonance (NMR).
  • In accordance with the first aspect of the present invention, an ethyl acetate extract of the fruiting body of AC is provided. The ethyl acetate extract includes at least one characteristic of: (a) causing a plurality of methyl signals generated from sterol or triterpene at δH 0.5 to 2 and terminal olefinic protons at δH 4.8 to 5.0 on a first 1H NM spectrum when the ethyl acetate extract is solved in a deuterium chloroform (CDCl3); and (b) causing signals of terminal olefinic protons at δH 5.0 to 5.4 on a second 1H NM spectrum when the ethyl acetate extract is solved in pyridine-D5 (C5D5N).
  • Preferably, the ethyl acetate extract further includes at least one compound being one selected from a group consisting of zhankuic acid A, dehydroeburicoic acid, actcin C and zhankuic acid C.
  • Preferably, zhankuic acid A has characteristic 1H NMR signals of two tertiary methyls (CH3-18 and CH3-19) and terminal olefinic protons (H2-28) in 24-exo-methylene-26-oic acid side-chain, and the ethyl acetate extract further has characteristic signals of triterpenoid the same with those of the zhankuic acid A.
  • Preferably, the ethyl acetate extract is fractionized from an ethanol extract of the fruiting body of AC.
  • In accordance with the second aspect of the present invention, a method for measuring an existence and an amount of triterpenoid of a target object is provided. The method includes steps of: (a) extracting the target object with a first ethanol solution to obtain an ethanol extract; (b) sequentially extracting the ethanol extract with n-hexane and ethyl acetate to obtain an ethyl acetate extract; and (c) measuring the existence and the amount of triterpenoid in the ethyl acetate extract.
  • Preferably, in the step (c), the existence of the triterpenoid is performed is performed by a nuclear magnetic resonance at a resolution of at least 200 MHz, the ethyl acetate extract is solved in a pyridine-D5 (C5D5N), and the amount of the triterpenoid is performed by a high performance liquid chromatography (HPLC).
  • Preferably, the target object is the fruiting body of AC.
  • In accordance with the third aspect of the present invention, an extracting method is provided. The method includes steps of: (a) providing the fruiting body of AC; (b) extracting the fruiting body with a first ethanol solution; and (c) obtaining an ethanol extract of the fruiting body.
  • Preferably, the step (a) further includes a step (a1) of grinding the fruiting body.
  • Preferably, the step (c) further includes a step (c1) of extracting the ethanol extract with at least one organic solvent to obtain an organic solvent extract.
  • Preferably, the at least one organic solvent has a total number of species more than one, each of the at least one organic solvent has a polarity, and the step (c1) has a extracting sequence that the polarity of a specific one of the at least one organic solvent is higher than that of a preceding one of the specific organic solvent.
  • Preferably, the at least one organic solvent is one selected from a group consisting of n-hexane, ethyl acetate, a second ethanol solution and a combination thereof.
  • Preferably, the step (c1) further includes steps of: (c11) extracting the ethanol extract with n-hexane to obtain an n-hexane extract and a first residue; (c12) extracting the first residue with ethyl acetate to obtain an ethyl acetate extract and a second residue; and (c13) extracting the second residue with a second ethanol solution to obtain a second ethanol extract.
  • Preferably, the step (c12) further includes a step (c121) of isolating the ethyl acetate extract with a plurality of trichloromethane-methanols to obtain zhankuic acid A, wherein the plurality of trichloromethane-methanols have respective ratios of trichloromethane to methanol.
  • Preferably, the step (c12) further includes a step (c121) of isolating the ethyl acetate extract with a plurality of n-hexane-ethyl acetate-methanols to obtain a plurality of fractions of the ethyl acetate extract, wherein the plurality of n-hexane-ethyl acetate-methanols have respective ratios of n-hexane-ethyl acetate to methanol.
  • Preferably, the step (c121) further includes at least one step of: (c121-1) sequentially isolating a first one of the plurality of fractions with ethyl acetate-dichloromethane-methanol, trichloromethane-methanol and methanol-water to obtain dehydroeburicoic acid; (c121-2) sequentially isolating a second one of the plurality of fractions with ethyl acetate-dichloromethane-methanol and acetonitrile-water having a first ratio of acetonitrile to water to obtain zhankuic acid A; (c121-3) sequentially isolating a third one of the plurality of fractions with dichloromethane-methanol and acetonitrile-water having a second ratio of acetonitrile to water to obtain actcin C; and (c121-4) sequentially isolating a fourth one of the plurality of fractions with dichloromethane-methanol and acetonitrile-water having a third ratio of acetonitrile to water to obtain zhankuic acid C.
  • Preferably, the step (b) further generates a third residue, and the method further includes a step (c1) of extracting the third residue with water to obtain a water extract.
  • Preferably, the ethanol extract is used to treat a cell for regulating expression of a specific protein of the cell.
  • Preferably, the cell is a leukemia cell, and the specific protein is one selected from a group consisting of histone, histone deacetylase, histone acetylase, a p21, a poly (ADP-ribose) polymerase (PARP), a Bax, a Bcl-2, a death receptor (DR5), a tumor necrosis factor receptor-associated death domain (TRADD), a p50 and a p65 and a combination thereof.
  • Preferably, the ethanol extract and 7-[4-(dimethylamino)phenyl]-N-hydroxy-4,6-dimethyl-7-oxohepta-2,4-dienamide (trichostatin A) have a synergistic effect of causing a cell to go into apoptosis.
  • The above objectives and advantages of the present invention will become more readily apparent to those ordinarily skilled in the art after reviewing the following detailed descriptions and accompanying drawings, in which:
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 illustrates the flowchart for preparing EEAC and its organic solvent extracts in the present invention;
  • FIG. 2(A) illustrates the growth inhibition of the respective extracts of the fruiting body of AC on HL 60 cells;
  • FIG. 2(B) illustrates the apoptosis of HL 60 cells induced by respective extracts of the fruiting body of AC;
  • FIG. 3(A) illustrates the 1H NMR pattern of the ethyl acetate extract of the fruiting body of AC solved in CDCl3;
  • FIG. 3(B) illustrates the 1H NMR pattern of zhankuic acid A solved in CDCl3;
  • FIG. 4 illustrates the effect of EEAC on DNA fragmentation in HL 60 cells;
  • FIG. 5(A) illustrates the immunoblotting patterns showing the effect of EEAC on histone acetylation in HL 60 cells;
  • FIG. 5(B) illustrates the immunoblotting patterns showing the activity of the histone acetylation-related enzymes regulated by EEAC in HL 60 cells;
  • FIG. 6 illustrates the synergistic effect of EEAC and TSA on the survival in HL 60 cells;
  • FIGS. 7(A) and 7(B) respectively are the immunoblotting patterns showing various protein expressions of TSA, EEAC and the combination thereof in HL 60 cells, wherein FIG. 7(A) shows expressions of p21, PARP, Bcl-2 and Bax, and FIG. 7(B) shows expressions of p50, p65, TRADD and DR5;
  • FIG. 8 illustrates the NFKB p65 activity of TSA, EEAC and the combination thereof in HL 60 cells;
  • FIGS. 9(A) to 9(E) respectively show 1H NMR spectra of (A) EEAC, (B) FC, (C) FA, (D) FB and (E) zhankuic acid A solved in C5D5N at 400 MHz; and
  • FIGS. 10(A) and 10(B) respectively show HPLC profiles of FA at (A) 254 nm and (B) 270 nm.
  • DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENT
  • The present invention will now be described more specifically with reference to the following Embodiments. It is to be noted that the following descriptions of preferred Embodiments of this invention are presented herein for purpose of illustration and description only; it is not intended to be exhaustive or to be limited to the precise form disclosed.
  • Embodiment 1 Biological Experiments
  • Experiment 1: Ethanol Extract and Water Extract Prepared from Fruiting Body of Wild Antrodia campharata
  • Fifty-two grams (52 g) of the dried fruiting body of AC was ground as fine powder, which was boiled to flux with ethanol at 75° C. at a ratio of 1:10 (w/v) for 2 hours. The extract was cooled and then was allowed to precipitate overnight at 4° C. The supernatant of extract was further filtered by a filter paper, centrifuged at 3,000 rpm for 30 minutes to remove the precipitate, and then the extract was lyophilized and stored at −70° C. This extract is named as “the ethanol extract of the fruiting body of A. camphorata (EEAC)”. Further, the residue was further boiled to flux at a ratio of 1:10 (w/v) for 6 to 8 hours. The supernatants were filtered and then centrifuged at 3,000 rpm for 30 minutes to remove the precipitate. The product, named as “the water extract of the fruiting body of A. camphorata (WEAC)”, was lyophilized and stored at −70° C.
  • Experiment 2: Further Extraction and Fractionation of EEAC
  • To identify the biologically active components of EEAC, 1 g of the dried fruiting body of AC was ground and extracted according to the method in Experiment 1. Fifty-percent inhibition concentration (IC50) of EEAC was determined as 104.82 μg/ml by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay; however, WEAC was inactive (IC50>200 μg/ml). EEAC (203.1 mg) was extracted with solvents of increasing polarities (n-hexane, ethyl acetate and ethanol sequentially), and three different extracts, 12.8% (26.0 mg) of n-hexane extract (FC), 61.6% (125.1 mg) of ethyl acetate extract (FA) and 8.4% (17.0 mg) of ethanol extract (FB), and 10.0% (20.2 mg) of the insoluble residue were yielded. Percentage is referred to yields in weight percentage of EEAC, milligram is referred to the corresponding dried weight after fractionation.
  • Experiment 3: Isolation and Purification of Zhankuic Acid A
  • EEAC fraction of 95.4 mg was chromatographed using Sephadex LH-20 with trichloromethane-methanol (CHCl3-MeOH) (1:3) to obtain three fractions, wherein Fraction 2 was further separated by preparative thin layer chromatography (TLC) with CHCl3-MeOH (25:1) to obtain 8 sub-fractions. Sub-fraction 2-1 of 10.3 mg was purified using ODS HPLC column (250×10 mm, Hypersil®, MeOH—H2O (85:15)) to obtain 4.3 mg of zhankuic acid A (retention time (Rt) 10 min, flow rate 2 ml/min).
  • The equipments for analyzing other physical and chemical characters of zhankuic acid A were used as follows. 1H and 13C NMR spectra were recorded on Varian Unity Plus 400, or Varian Gemini 200 NMR spectrometers. Chemical shifts and coupling constants respectively were represented as parts per million (ppm, 6) and Hertz (Hz). LRESIMS (low-resolution electrospray ionization mass spectra) was measured on a VG Biotech Quatro 5022 mass spectrometer. Silicon gel 60 (Merck, 230 to 400 mesh) and Sephadex LH-20 were used for column chromatography, while TLC was carried out on Silica gel GF254 pre-coated plates with detection using 50% sulfuric acid followed by heating on the hot plate. HPLC was performed with a Hitachi L-7100 pump and D-7000 interface equipped with a Bischoff RI detector using ODS columns.
  • Experiment 4: Cell Culture, Cell Viability and Cytotoxicity
  • Human leukemia HL 60 cell line was purchased from American Type Culture Collection (ATCC, Manassas, Va.) and incubated in RPMI 1640 medium supplemented with 10% fetal calf serum, 2 mM glutamine, 100 units/ml penicillin and 100 μg/ml streptomycin at 37° C. in a humidified atmosphere of 5% carbon dioxide.
  • Cell viability was determined by the trypan blue dye exclusion and cytotoxicity was assessed by MTT assay. First, 1×105 HL 60 cells were plated in 96-well plates and treated with different concentrations of EEAC dissolved in dimethyl sulfoxide (DMSO) at the concentrations of 0 to 150 μg/ml for 24 hours. Death cells then were stained with trypan blue, and the survival cells were calculated with hemocytometer. In MTT assay, 1.2 mg/ml MTT was added in each well, which was incubated for 2 hours to form crystal violet, formazan. Formazan was dissolved in DMSO, and absorbance was detected at a wavelength of 570 nm.
  • Experiment 5: Flow Cytometry
  • HL 60 cells (1×106) were incubated in the 10-cm dish and treated with known concentrations of EEAC for 24 hours. The treated cells were washed with ice-cold phosphate-buffered saline (PBS) twice and harvested by centrifuging at 37° C. at 200×g for 5 minutes. Cells were fixed with 70% (v/v) ethanol at 37° C. for 30 minutes, and then were resuspended in 1 ml propidium iodium (PI) reagent (containing 0.1% Triton X-100, 100 μg/ml ribonuclease (RNase) A and 500 μg/ml PI) at 37° C. for 30 minutes. The cells were detected with the flow cytometer, and data were analyzed with FACScan and Cell Quest Program (Becton Dickinson).
  • Experiment 6: Evaluation of DNA Fragmentation
  • In this experiment, DNA fragmentation in the apoptotic cells were determined by gel electrophoresis. HL 60 Cells (1×106) were incubated in the 10-cm dish for 24 hours, then were treated with known concentrations of EEAC (0, 50, 100, 150 and 200 μg/ml respectively) for another 24 hours. Control group was the cells with DMSO treatment. Next, the cells were washed with PBS twice and resuspended in the solution A (containing 10 μM EDTA (2-[2-(bis(carboxymethyl)amino)ethyl-(carboxymethyl)amino]acetic acid), 50 mM Tris-HCl (pH 8.0), 0.5% (v/v) sodium dodecyl sulfate (SDS) and 1 mg/ml proteinase K) at 56° C. for 3 hours. Subsequently, RNase at a final concentration of 50 μg/ml was added therein for reacting 1 hour. Cellular DNA was extracted with phenol/chloroform/isopropanol (25:24:1, v/v), and cellular DNA fragmentation was determined by gel electrophoresis so as to evaluate apoptosis.
  • Experiment 7: Histone Isolated by Acid Extraction
  • This experiment was performed in accordance with the literature of Taplick et al. (1998). First, cells were harvested at 700×g, washed with ice-cold PBS once, resuspended in 1 ml lysis buffer (containing 10 mM Tris-HCl (pH 6.5), 50 mM sodium sulfate, 10 mM magnesium chloride, 10 mM sodium butyrate, 8.6% sucrose and 1% Triton X-100) and centrifuged at 1,000×g. After three washes in lysis buffer, pellet was resuspended in a buffer containing 10 mM Tris-HCl (pH 7.4) and 13 mM EDTA. After centrifugation, the cells were resuspended in cold sterile water, and sulfuric acid was added therein to 0.4 N. The cells were centrifuged at 10,000×g for 5 minutes after incubation on ice for 1 hour. Total histones in the supernatant were precipitated overnight with 10× volumes of acetone at −20° C. The precipitated histones were collected by centrifugation, dried and resuspended in distilled water, and the protein content was determined by the Bio-Rad protein assay kit (BioRad Laboratories GmbH, Munchen, Germany).
  • Experiment 8: Nucleoprotein and Cytoplasmic Protein Extraction
  • This experiment was performed in accordance with the literature of Vandergeeten et al. (2007). Cells were washed with cold PBS and resuspended in the lysis buffer (containing 10 mM HEPES (4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid)-KOH (pH 7.9), 2 mM magnesium chloride, 0.1 mM EDTA, 10 mM potassium chloride, 0.5% IGEPAL (Sigma, I3021), 1 mM phenylmethanesulfonyl fluoride (PMSF), 1 mM DTT and proteinase inhibitors) on ice for 30 minutes. After centrifugation at 10,000×g for 15 minutes, the supernatant containing nucleoprotein was harvested and stored at −80° C, and the protein concentration of the supernatant was determined.
  • Experiment 9: Western Immunoblotting
  • The harvested cells were treated with RIPA lysogenic buffer (containing 1× PBS, 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% SDS, 1 mM sodium orthovanadate, 100 μg/ml PMSF and 30 mg/ml aprotinin) for 30 minutes (Lu et al., 2006), and then was centrifuged at 20,000×g for 30 minutes. The protein concentration was determined by BCA protein assay kit (Pierce, Rockford, Ill.). Protein was resolved by 7.5%, 10% or 12% SDS-polyacrylamide gel electrophoresis (PAGE) respectively and transferred onto the PVDF nitrocellulose membrane. Protein expressions were sequentially monitored with primary antibody and horse-radish peroxidase (HRP)-conjugated secondary antibody, and then was developed with enhanced chemical luminescence (ECL). Protein expressions were exposed on X-ray film.
  • Experiment 10: Nuclear Factor Kappa B (NFκB) Transcriptional Factor Assay
  • This experiment is to determine the effect of EEAC treatment on NFKB activation by TranAM® transcription factor assay (Active Motif, Tokyo, Japan) (Schwab et al., 2007). First, 7 μg of nuclear extract was added to each well of a 96-well culture plate which was pre-coated with NFκB consensus binding oligonucleotide (SEQ ID NO. 1). After 1 hour of incubation with smooth agitation, wells were washed three times with washes buffer and then incubated with p50 or p65 antibody (dilution 1:1,000) for 1 hour at room temperature. After three successive washings, the wells were incubated with HRP-conjugated secondary antibody (dilution 1:1,000) for 1 hour at room temperature followed by the addition of developing solution. The optical density was determined at 405 nm by keeping the reference wavelength at 655 nm.
  • Experiment 11: Statistic Analysis
  • All data were represented as “mean±standard deviation (SD)”. The difference between the experiment and the control was analyzed by t-test, and a probability of p<0.05 was considered significant.
  • Experimental Results
  • Please refer to FIG. 1, which illustrates the flowchart for preparing EEAC and its organic-solvent extracts in the present invention. In the preparation method of FIG. 1, the ground fruiting body of AC was extracted with ethanol (Steps 101, 102), and then was filtered and centrifuged to obtain EEAC (Step 103) and the residue of the fruiting body of AC (Step 110). To identify the biologically active components for cytotoxicity in the fruiting body of AC, EEAC was further extracted with the organic solvents of increasing polarity, which were ranked: n-hexane, ethyl acetate and ethanol. The preparation method of the organic solvent extracts of the fruiting body of AC was described as follows. EEAC was extracted with n-hexane to obtain the n-hexane extract (fraction C, FC) (Step 104) and the first residue (Step 105). Next, the first residue was further extracted with ethyl acetate to obtain the ethyl acetate extract (fraction A, FA) (Step 106) and the second residue (step 107). The second residue was further extracted with ethanol solution to obtain the another ethanol extract (fraction B, FB) (step 108) and the third residue (step 109). In addition, the residue (step 110) was further boiled and extracted with water (step 111) to obtain the water extract of the fruiting body of AC (WEAC) (step 112).
  • To analyze the cytotoxicity of these organic solvents on HL 60 cells, 50% inhibition concentrations (IC50) of EEAC and FA respectively on HL 60 cells were determined as 104.82 μg/ml and 80.53 μg/ml by MTT assay, but IC50 of FB and FC respectively on HL 60 cells were more than 200 μg/ml. Therefore, FA has better growth inhibition effect on HL 60 cells. Please refer to FIG. 2(A), which illustrates the growth inhibition of the respective extracts of fruiting body of AC on HL 60 cells. In FIG. 2(A), growth inhibition on HL 60 cells was increased with the increasing concentrations of the respective extracts. Please refer to FIG. 2(B), which illustrates the apoptosis of HL 60 cells induced by 100 μg/ml of respective extracts. In FIG. 2(B), comparing with other extracts, FA showed the significant apoptotic ability on HL 60 cells. From the above-mentioned results, it can be known that FA contains the biologically active component for cytotoxicity on HL 60 cells.
  • The 1H NMR spectrum of FA was further analyzed. The methyl signals (CH3-18 and CH3-19) of sterol or triterpene was shown at 6 0.5 to 2 (FIG. 3(A)) when the NMR profile of FA was 3.0 mg/ml in deuterium chloroform (CDCl3), and the major component of FA was predicted as zhankuic acid A from the spectrum of FIG. 3(A). Further, comparing 1H NMR spectrum of FA with that of zhankuic acid A at the same conditions (FIG. 3(B)), it can be found that FA has the similar signals of zhankuic acid A. Characteristic 1H NMR signals of zhankuic acid A showed two tertiary methyls (CH3-18 and CH3-19) and terminal olefinic protons (H2-28) in 24-exo-methylene-26-oic acid side-chain, and 1H NMR spectrum of the ethyl acetate extract exhibited clearly these distinguishable signals. Therefore, FA indeed is mainly composed of zhankuic acid A which has a chemical formula I as follows.
  • Figure US20100210869A1-20100819-C00001
  • The major component, zhankuic acid A, of FA was a pale-yellow amorphous solid, and its molecular formula was established as C29H40O5 by electrospray ionization mass spectra (ESIMS, m/z 469 [M+H]+) and NMR spectra. The most significant signals of its 1H NMR spectrum were those corresponding to five methyls (three secondary at δ=0.95, 1.05, 1.30, and two tertiary at δ=0.70, 1.53) and two terminal olefinic methylene protons at δ=4.92 and 4.98. The 13C NMR spectrum displayed three carbonyl groups (δC=200.8, 202.6, and 210.8), four olefinic carbons (δC=111.4, 145.5, 148.0, 151.9) and one carboxylic acid (δC=178.8). According to the above-mentioned 1H NMR analysis, our results suggested that zhankuic acid A was the major component of FA. Moreover, zhankuic acid A exhibited significant cytotoxicity to HL 60 cells (IC50 5.45 μg/ml), and it could serve as a biological marker.
  • Please refer to FIG. 4, which illustrates the effect of EEAV on DNA fragmentation in HL 60 cells. It can be known from FIG. 4 that the concentration of EEAC is proportional to the DNA fragmentation of HL 60 cells, which means EEAC has significant cytotoxicity to HL 60 cells.
  • Since chromatin structure can be regulated by histone acetylation and histone deacetylation, histone plays an important role on regulating gene expression, which is accomplished by acetylating specific lysine of histone with histone acetyltransferase (HAT) and deacetylating lysine thereof with histone deacetylase (HDAC). Transcription only can be processed on acetylated chromatin, otherwise, transcription would be ceased. Please refer to FIG. 5(A), which illustrates the immunoblotting patterns showing the effect of EEAC on histone acetylation in HL 60 cells. In FIG. 5(A), EEAC showed the dose-dependent relationship to the low level acetylation of histones (H3, H3K18 and H3K9) in HL 60 cells. Please refer to FIG. 5(B), which illustrates the immunoblotting patterns showing the activity of the histone acetylation-related enzymes regulated by EEAC in HL 60 cells. In FIG. 5(B), deacetylations of histone H3, including inhibition of GCN5, CBP, SRC-1 and PCAF and promotion of HDAC1, were regulated by EEAC with HAT and HDAC.
  • Trichostatin A (TSA) is a microbial metabolite, which can promote acetylation of histone H3. Since H3 acetylation is accumulated (hyper-acetylation), trichostatin A can effectively inhibit leukemia proliferation. Please refer to FIG. 6, which illustrates the synergistic effect of EEAC and TSA on the survival in HL 60 cells. In FIG. 6, comparing 100 μg/ml EEAC treatment alone with TSA treatment alone, EEAC had better cytotoxicity effect on HL 60 cells than TSA. Further, the combined treatment of 100 g/ml EEAC and different dosages of TSA had synergistic effect on HL 60 cells, and the cytotoxicity effect was increased with the increasing TSA dosages.
  • Please refer to FIGS. 7(A) and 7(B), which respectively are the immunoblotting patterns showing various protein expressions of TSA, EEAC and the combination thereof in HL 60 cells. FIG. 7(A) was subjected to p21, PARP, Bcl-2 and Bax which related to growth inhibition and apoptotic induction. It is found that p21 expression of HL 60 cells is increased and Bcl-2 and cleaved PARP expressions thereof are inhibited by cooperatively treating TSA with EEAC.
  • Since A. camphorata is potent FAS (TRAIL)-sensitizing agent in a variety of different TRAIL-resistant human cancer cells (Hsu et al., 2005), DR5 and TRADD interact specifically with death domain of DR4. DR4 activation promotes the binding of TRADD and the subsequent recruitment of DR5 to promote apoptosis (Swarup et al., 2007). When HL 60 cells were individually treated TSA or EEAC or cooperatively treated with TSA and EEAC, it can be found that the immunoblotting analysis for TRADD and DR5 revealed 2 and 4 fold increases over the corresponding control and TSA treatment alone (FIG. 7(B)). It can be known from FIG. 7(B) that the combination-induced upregulation of DR5 is mediated by EEAC, but upregulation of TRADD is mediated synergistically by TSA and EEAC. Therefore, the combined treatment with TSA and EEAC cooperatively targeted the TRAIL pathway.
  • Certain cancer therapeutic agents induce the expression of DR5 in cancer cells and are thereby able to augment TRAIL-induced apoptosis or initiate apoptosis. Therefore, TRAIL is one of the promising new candidates for cancer therapeutics. In addition, NFκB activation can be triggered by direct stimulation of CD95 and the TRAIL death receptors as well as by overexpression of FLIP protein, and NFκB transcription factor differentially regulated the DR5 expression involving HDAC1 (Shetty et al., 2005). To examine the effect of NFκB activation on cellular apoptosis in response to combined treatment, it is found that the combined treatment significantly increases the translocation of NFκB p65, but does not affect the expression with total extracts of cells. Obviously, combination-induced p65 translocation may be mediated by EEAC treatment alone.
  • Treatment of HL 60 cells with TSA and EEAC resulted in an increase of basal NFκB p65 DNA binding activity in the nucleus (+78% vs. control, p<0.05). In contrast, basal p50 NFκB DNA binding activity was not affected in response to different treatments (FIG. 8), further supporting the suggestion that nuclear translocation of p65 and activation are responsible for the combined treatment-induced NDκB p65 activation.
  • In the present invention, the obtained EEAC can effectively inhibit growth of HL 60 cells and induce apoptosis mediated histone hypoacetylation and suppression of acetyltransferase activity (including GCN5, CBP and PCAF) in HL 60 cells. In addition, combined treatment of EEAC and TSA on HL 60 cells can synergistically inhibit cellular growth and induce apoptosis.
  • Zhankuic acid A was identified as the major active component in EEAC after the respective organic solvent extracts obtained by further extracting EEAC with organic solvents and NMR analysis. Accordingly, EEAC can be the potential chemotherapeutic agent.
  • Embodiment 2
  • This Embodiment used another fruiting body of AC to prepare 11 g of lyophilized EEAC according to the preparation method in Experiment 1 of Embodiment 1. Subsequently, EEAC was sequentially extracted with n-hexane, ethyl acetate and ethanol according to the method in Experiment 2 of Embodiment 1 to obtain three different extracts, 12.7% (1.4 g) FC, 61.8% (6.8 g) FA and 10.0% (1.1 g) FB, and 5.5% (0.6 g) insoluble residue. Percentage is referred to the weight percentage in EEAC, gram is referred to the dry weight after extraction.
  • Experiment 1: Anti-Leukemia Activity
  • To compare with the controls in the prior art, the respective lyophilized EEAC (100 mg) in Embodiment 2 was extracted with three partition extraction methods, and anti-leukemia activity between the respective obtained extracts and the extracts in Embodiment 2 was compared. In Control 1, 101.9 mg lyophilized EEAC was fractionation with ethyl acetate and water (100 ml each, 1:1 (v/v)) by liquid-liquid partition extraction according to the literatures of Chen et al., 1995 and Hsu et al., 2007. The ethyl acetate extract of the fruiting body of AC (FEA-EA, 78.1%, 79.6 mg) and the water extract of the fruiting body of AC (FW-EA, 19.6%, 20.0 mg) were obtained.
  • In Control 2, 100.3 mg lyophilized EEAC was fractionation with chloroform and water (100 ml each, 1:1 (v/v)) by liquid-liquid partition extraction according to the literature of Chemg et al., 1995. The chloroform extract of the fruiting body of AC (FC13-C13, 77.4%, 77.6 mg) and the water extract of the fruiting body of AC (FW-C13, 23.5%, 23.6 mg) were obtained.
  • In Control 3, 100.9 mg lyophilized EEAC was fractionation with dichloromethane and water (100 ml each, 1:1 (v/v)) by liquid-liquid partition extraction according to the literature of Shen et al., 2003. The dichrolomethane extract of the fruiting body of AC (FC12-C12, 76.2%, 76.9 mg) and the water extract of the fruiting body of AC (FW-C12, 23.3%, 23.5 mg) were obtained.
  • Please refer to Table 1, which illustrates the comparison of anti-leukemia activity of the above-mentioned extracts. In the present Embodiment 2, FEA exhibited the best activity that demonstrated the repeatability in production for concentrating cytotoxic components in EEAC. Comparing antiproliferative activity of FA with those in Control 1, FA (104.10 μg/ml) were more potent than FEA-EA (156.47 μg/ml) which prepared by the acceptable partition method in food science. Although the fractions (FC13-C13 and FC12-C12) had shown competitive antiproliferative activity referring to FEA, usage of those chlorine-containing organic solvents were limited or forbidden. Therefore, the methods in the prior art are not ideal preparation procedures.
  • TABLE 1
    Antiproliferative activity of extracts in Embodiment 2 and the
    respective controls
    Product Anti-leukemia activity (IC50) (μg/ml)
    Embodiment 2 EEAC 182.94
    FC 156.36
    FA 104.10
    FB >200
    Control 1 FEA-EA 156.47
    FW-EA >200
    Control 2 FCl3-Cl3 126.04
    FW-Cl3 >200
    Control 3 FCl2-Cl2 115.03
    FW-Cl2 >200
  • Experiment 2: NMR Analysis
  • The experimental conditions of 1H NMR spectrum in Experiment 2 were: the equipment with 400 MHz resolution; the concentration of 10.0 ml; and EEAC, FC, EA, FB and zhankuic acid A were solved in C5D5N solution.
  • Please refer to FIGS. 9(A) to 9(E), which respectively show 1H NMR spectra of (A) EEAC, (B) FC, (C) FA, (D) FB and (E) zhankuic acid A solved in C5D5N at 400 MHz. The characteristic 1H NMR signals of zhankuic acid A showed two tertiary methyls [δH 0.70 (H3-18) and 1.61 (H3-19) (each 3H, s)], three secondary methyls [δH 0.89 (3H-21, d, J=5.6 Hz), 1.03 (3H-29, d, J=6.4 Hz), and 1.52 (3H-27, d, J=7.2 Hz)], and terminal olefinic protons (H2-28) in 24-exo-methylene-26-oic acid side-chain at δH 5.08 (1H, br s) and 5.24 (1H, br s). When 1H NMR spectra in FIGS. 9(A) to 9(E) were partially aligned and compared with each other, 1H NMR spectra of EEAC and FA exhibited clearly these distinguishable signals in FIG. 9(A) and 9(C), and this result was identical with that in FIGS. 3(A) and 3(B). Moreover, the signals of the methine protons [δH 3.01 (1H-12, d, J=13.6 Hz), 3.17 (1H-1, ddd, J=13.3, 6.8, 2.4 Hz), and 3.48 (1H-25, q, J=7.2 Hz)] of zhankuic acid A were also found circumspectly in 1H-NMR spectra of EEAC and FA. However, the above-mentioned specific signals were not observed at all in the 1H NMR spectra of FC and FB in FIGS. 9(B) and 9(D). Therefore, it can be identified that the major active component, zhankuic acid A, of the fruiting body of AC can be concentrated in ethyl acetate layer by the preparation method of the present invention.
  • Standard was isolated from 6.8 g FA. In addition to zhankuic acid A, another three major components, antcin C (formula II), zhankuic acid C (formula III) and dehydroeburicoic acid (formula IV), could be obtained and identified by NMR and ESIMS. The structural formulas, isolations and purifications of these three major active components were illustrated as follows.
  • Figure US20100210869A1-20100819-C00002
  • (1) Isolation and purification of dehydroeburicoic acid: FA (6.8 g) was chromatographed with n-hexane-ethyl acetate-methanol (10:1:0, 5:1:0, 1:1:0, 0:1:0, 0:40:1, 0:30:1, 0:20:1 and 0:10:1 respectively) in silica gel 60 (Merck, 230-400 mesh) to obtain 17 fractions, wherein the fourth fraction (Fraction 4, 588.4 mg) was chromatographed with ethyl acetate-dichloromethane-methanol (EA-CH2Cl2-MeOH, 1:1:6) in Sephadex LH-20 to yield 3 sub-fractions. The first sub-fraction (Fraction 4-1, 568.6 mg) was further chromatographed with CHCl3-MeOH (35:1) in Silica gel 60 to yield 3 another sub-fractions. Fifty milligrams (50 mg) from 240.3 mg of the second sub-fraction (Fraction 4-1-2) was purified with ODS HPLC column (250×10 mm, Hypersil®, methanol-water (90:10)) to obtain 10.3 mg dehydroeburicoic acid (R t 27 min, flow rate 2 ml/min).
  • (2) Isolation and purification of zhankuic acid A: The sixth fraction (Fraction 6, 970.9 mg) was chromatographed with EA-CH2Cl2-MeOH (1:1:6) in Sephadex LH-20 to yield sub-fractions. One hundred milligrams (100 mg) from 901.0 mg of the second sub-fraction (Fraction 6-2) was further chromatographed with ODS HPLC column (250×10 mm, Hypersil®, acetonitrile-water (75:25)) to obtain 77.5 mg of zhankuic acid A (R t 12 min, flow rate 2 ml/min).
  • (3) Isolation and purification of antcin C: The tenth fraction (Fraction 10, 132.6 mg) was chromatographed with CH2Cl2-MeOH (15:1) by preparative TLC to yield 5 sub-fractions. The second sub-fraction (Fraction 10-2, 85.0 mg) was purified with ODS HPLC column (250×10 mm, Hypersil®, acetonitrile-water (70:30)) to obtain 40.9 mg of antcin C (R t 10 min, flow speed 2 mil/min).
  • (4) Isolation and purification of zhankuic acid C: The thirteenth fraction (Fraction 13, 1.4 g) was chromatographed with CH2Cl2-MeOH (15:1) in Silica gel 60 to yield 7 sub-fractions. The fifth sub-fraction (Fraction 13-5, 107.5 mg) was purified with ODS HPLC column (250×10 mm, Hypersil®, acetonitrile-water (70:30)) to obtain 26.0 mg of zhankuic acid C (R t 10 min, flow rate 2 ml/min).
  • Experiment 3: HPLC Analysis
  • HPLC spectra of four obtained major active components and FA were compared. The HPLC conditions were provided as follows. HPLC was Shimadzu LC-10AT, the detector was Shimadzu SPD-M10A photodiode array detector, the auto sampler was Shimadzu SIL-20A prominence auto sampler; the HPLC column was Cosmosil 5C-18-MS-II (250×4.6 mm), solvent A in mobile phase was acetonitrile and solvent B was water (in 0.1% acetic acid), flow rate was 1 ml/min, column temperature was room temperature, and detection wavelength was 254 nm and 270 nm.
  • The parameters of solvent system were illustrated as follows. Mobile phase included solvent A and B, linear gratitude was operated at 0 to 30 minutes (45% A to 50% A), 30 to 35 minutes (50% A to 55% A), 35 to 45 minutes (55% A to 60% A), 45 to 55 minutes (60% A to 70% A), 55 to 60 minutes (70% A to 85% A) and 60 to 100 minutes (85% A to 100% A). Flow rate and column temperature were illustrated as above.
  • In accordance with the published literatures (Chen et al., 1995; Cherng et al., 1995; Hsu et al., 2007; Shen et al., 2003), zhankuic acid A, antcin C and zhankuic acid C are stereo isomers having the chiral center at C-25 position, but they cannot be purified at the isolation procedures. However, two stereo isomers can be separated and observed, i.e. two peaks represented on the chromatographic spectrum, at the above-mentioned HPLC conditions.
  • Please refer to FIG. 10(A) and Table 2, which show comparisons of peak area and height for the respective components in FA after with HPLC at 254 nm wavelength. In Table 2, zhankuic acid A represented two peaks at 254 nm wavelength and retention time of 46.739 and 47.347 min, and the determined area occupied 16.26% and the determined height occupied 17.27% of the total active layer. Zhankuic acid A had the highest area and height in four major active components.
  • TABLE 2
    Comparisons of peak area and height for the respective components
    in FA after with HPLC at 254 nm wavelength
    Retention
    Product Peak number time (min) Area (%) Height (%)
    Antcin C 1a 24.779 6.27 4.84
    Antcin C 1b 26.734 5.70 4.90
    Zhankuic acid C 2a 27.933 6.48 5.87
    Zhankuic acid C 2b 29.140 8.37 6.96
    Zhankuic acid A 3a 46.739 8.97 9.51
    Zhankuic acid A 3b 47.347 7.29 7.76
    Dehydroeburicoic 4   77.531 6.24 9.22
    acid
    Others 50.68 50.94
    Total 100.00 100.00
  • Please refer to FIG. 10(B) and Table 3, which shows comparisons of peak area and height of the respective components in FA with HPLC at 270 nm wavelength. In Table 3, zhankuic acid A represented two peaks at 270 nm wavelength and retention time of 46.736 and 47.352 min, and the determined area occupied 26.53% and the determined height occupied 27.86% of the total active layer. Zhankuic acid A also had the highest peak height in four major active components.
  • TABLE 3
    Comparisons of peak area and height of the respective components in
    FA with HPLC at 270 nm wavelength
    Retention
    Product Peak number time (min) Area (%) Height (%)
    Antcin C 1a 24.761 5.42 3.49
    Antcin C 1b 26.719 4.07 3.27
    Zhankuic acid C 2a 27.931 12.86 11.47
    Zhankuic acid C 2b 29.147 16.38 12.80
    Zhankuic acid A 3a 46.736 14.53 15.43
    Zhankuic acid A 3b 47.352 12.00 12.43
    Others 34.74 41.11
    Total 100.00 100.00
  • From the results of NMR and HPLC analysis, it can be known that zhankuic acid A is the major active component in EEAC, and antcin C, zhankuic acid C and dehydroeburicoic acid are another ones. FA is defined as the triterpenoids-rich fraction and shows antiproliferative activity in HL 60 cells. Accordingly, the above-mentioned experimental method can be the power tool for detecting the components of triterpenoids of A. camphorata in the industry.
  • While the invention has been described in terms of what is presently considered to be the most practical and preferred Embodiments, it is to be understood that the invention needs not be limited to the disclosed Embodiments. On the contrary, it is intended to cover various modifications and similar arrangements included within the spirit and scope of the appended claims, which are to be accorded with the broadest interpretation so as to encompass all such modifications and similar structures.

Claims (20)

1. An ethyl acetate extract of a fruiting body of an Antrodia camphorata, comprising at least one characteristic of:
(a) causing a plurality of methyl signals generated from one of a sterol and a triterpene at δH 0.5 to 2 and terminal olefinic protons at δH 4.8 to 5.0 on a first 1H nuclear magnetic resonance (1H NMR) spectrum when the ethyl acetate extract is solved in a deuterium chloroform (CDCl3); and
(b) causing the signals of terminal olefinic protons at δH 5.0 to 5.4 on a second 1H NMR spectrum when the ethyl acetate extract is solved in a pyridine-D5 (C5D5N).
2. The ethyl acetate extract according to claim 1 further comprising at least one compound being one selected from a group consisting of a zhankuic acid A, a dehydroeburicoic acid, an actcin C and a zhankuic acid C.
3. The ethyl acetate extract according to claim 2, wherein the zhankuic acid A has characteristic 1H NMR signals of two tertiary methyls (CH3-18 and CH3-19) and terminal olefinic protons (H2-28) in 24-exo-methylene-26-oic acid side-chain, and the ethyl acetate extract further has characteristic signals of triterpenoid the same with those of the zhankuic acid A.
4. The ethyl acetate extract according to claim 1 being fractionized from an ethanol extract of the fruiting body of the Antrodia camphorata.
5. A method for measuring an existence and an amount of triterpenoid of a target object, comprising steps of:
(a) extracting the target object with a first ethanol solution to obtain an ethanol extract;
(b) sequentially extracting the ethanol extract with an n-hexane and an ethyl acetate to obtain an ethyl acetate extract; and
(c) measuring the existence and the amount of the triterpenoid in the ethyl acetate extract.
6. The method according to claim 5, wherein in the step (c), the existence of the triterpenoid is performed by a nuclear magnetic resonance at a resolution of at least 200 MHz, the ethyl acetate extract is solved in a pyridine-D5 (C5D5N), and the amount of the triterpenoid is performed by a high performance liquid chromatography (HPLC).
7. The method according to claim 5, wherein the target object is a fruiting body of an Antrodia camphorata.
8. An extracting method, comprising steps of:
(a) providing a fruiting body of an Antrodia camphorata;
(b) extracting the fruiting body with a first ethanol solution; and
(c) obtaining an ethanol extract of the fruiting body.
9. The method according to claim 8, wherein the step (a) further comprises a step (a1) of grinding the fruiting body.
10. The method according to claim 8, wherein the step (c) further comprises a step (c1) of extracting the ethanol extract with at least one organic solvent to obtain an organic solvent extract.
11. The method according to claim 10, wherein the at least one organic solvent has a total number of species more than one, each of the at least one organic solvent has a polarity, and the step (c1) has a extracting sequence that the polarity of a specific one of the at least one organic solvent is higher than that of a preceding one of the specific organic solvent.
12. The method according to claim 10, wherein the at least one organic solvent is one selected from a group consisting of an n-hexane, an ethyl acetate, a second ethanol solution and a combination thereof.
13. The method according to claim 10, wherein the step (c1) further comprises steps of:
(c11) extracting the ethanol extract with an n-hexane to obtain an n-hexane extract and a first residue;
(c12) extracting the first residue with an ethyl acetate to obtain an ethyl acetate extract and a second residue; and
(c13) extracting the second residue with a second ethanol solution to obtain a second ethanol extract.
14. The method according to claim 13, wherein the step (c12) further comprises a step (c121) of isolating the ethyl acetate extract with a plurality of trichloromethane-methanols to obtain a zhankuic acid A, wherein the plurality of trichloromethane-methanols have respective ratios of a trichloromethane to a methanol.
15. The method according to claim 13, wherein the step (c12) further comprises a step (c121) of isolating the ethyl acetate extract with a plurality of n-hexane-ethyl acetate-methanols to obtain a plurality of fractions of the ethyl acetate extract, wherein the plurality of n-hexane-ethyl acetate-methanols have respective ratios of an n-hexane-ethyl acetate to a methanol.
16. The method according to claim 15, wherein the step (c121) further comprises at least one step of:
(c121-1) sequentially isolating a first one of the plurality of fractions with an ethyl acetate-dichloromethane-methanol, a trichloromethane-methanol and a methanol-water to obtain a dehydroeburicoic acid;
(c121-2) sequentially isolating a second one of the plurality of fractions with the ethyl acetate-dichloromethane-methanol and an acetonitrile-water having a first ratio of an acetonitrile to a water to obtain a zhankuic acid A;
(c121-3) sequentially isolating a third one of the plurality of fractions with a dichloromethane-methanol and the acetonitrile-water having a second ratio of an acetonitrile to a water to obtain an actcin C; and
(c121-4) sequentially isolating a fourth one of the plurality of fractions with the dichloromethane-methanol and the acetonitrile-water having a third ratio of an acetonitrile to a water to obtain a zhankuic acid C.
17. The method according to claim 8, wherein the step (b) further generates a third residue, and the method further comprises a step (c1) of extracting the third residue with a water to obtain a water extract.
18. The method according to claim 8, wherein the ethanol extract is used to treat a cell for regulating an expression of a specific protein of the cell.
19. The method according to claim 18, wherein the cell is a leukemia cell, and the specific protein is one selected from a group consisting of a histone, a histone deacetylase, a histone acetylase, a p21, a poly (ADP-ribose) polymerase (PARP), a Bax, a Bcl-2, a death receptor (DR5), a tumor necrosis factor receptor-associated death domain (TRADD), a p50 and a p65 and a combination thereof.
20. The method according to claim 8, wherein the ethanol extract and a 7-[4-(dimethylamino)phenyl]-N-hydroxy-4,6-dimethyl-7-oxohepta-2,4-dienamide (trichostatin A) have a synergistic effect of causing a cell to go into an apoptosis.
US12/538,549 2009-02-13 2009-08-10 Ethanol extract of antrodia camphorata for inducing apoptosis and preparation method thereof Abandoned US20100210869A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/398,368 US8883167B2 (en) 2009-02-13 2012-02-16 Ethanol extract of Antrodia camphorata for inducing apoptosis and preparation method thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
TW098104772A TWI389699B (en) 2009-02-13 2009-02-13 Ethanol extract of antrodia camphorata for inducing apoptosis and preparation method thereof
TW098104772 2009-02-13

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/398,368 Division US8883167B2 (en) 2009-02-13 2012-02-16 Ethanol extract of Antrodia camphorata for inducing apoptosis and preparation method thereof

Publications (1)

Publication Number Publication Date
US20100210869A1 true US20100210869A1 (en) 2010-08-19

Family

ID=42560514

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/538,549 Abandoned US20100210869A1 (en) 2009-02-13 2009-08-10 Ethanol extract of antrodia camphorata for inducing apoptosis and preparation method thereof
US13/398,368 Active US8883167B2 (en) 2009-02-13 2012-02-16 Ethanol extract of Antrodia camphorata for inducing apoptosis and preparation method thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/398,368 Active US8883167B2 (en) 2009-02-13 2012-02-16 Ethanol extract of Antrodia camphorata for inducing apoptosis and preparation method thereof

Country Status (2)

Country Link
US (2) US20100210869A1 (en)
TW (1) TWI389699B (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110123562A1 (en) * 2009-11-26 2011-05-26 Chiang Been-Huang Anti-cancer active substance from antrodia camphorata, method for preparing the same and use thereof
WO2014130618A1 (en) * 2013-02-20 2014-08-28 Golden Biotechnology Corporation Cyclohexenone compositions and process for making thereof
WO2014130619A3 (en) * 2013-02-20 2014-10-16 Golden Biotechnology Corporation Methods and compositions for treating leukemia
US8865181B1 (en) 2013-04-26 2014-10-21 Kang Li Biotech Co., Ltd. Extracting method of Antrodia cinnamomea
US8883167B2 (en) 2009-02-13 2014-11-11 Kaohsiung Medical University Ethanol extract of Antrodia camphorata for inducing apoptosis and preparation method thereof
JP2014214113A (en) * 2013-04-25 2014-11-17 康力生技股▲分▼有限公司 Extraction method of antrodia camphorate extract
CN105237365A (en) * 2014-07-09 2016-01-13 台湾利得生物科技股份有限公司 Extract of solid-state cultured mycelium of antrodia camphorata and application thereof
US9751956B2 (en) 2015-03-06 2017-09-05 Ultra-Microrigin Biomedical Technology Co., Ltd. Method of using supercritical fluid technology to separate and purify functional components of Antrodia camphorata
US10806762B2 (en) 2016-05-20 2020-10-20 Taiwan Indigena Botanica Co., Ltd. Method of treatment of cancer cells by using pharmaceutical composition
WO2021222534A1 (en) * 2020-05-01 2021-11-04 Arjil Biotech Holding Company Limited Compounds and methods for prevention and treatment of coronavirus infections

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI454301B (en) * 2011-01-10 2014-10-01 Univ Kaohsiung Medical Benzenoid compounds of antrodia cinnamomea, preparation and analysis method thereof
TW201231474A (en) * 2011-01-26 2012-08-01 Univ Kaohsiung Medical Triterpenoid composition of Antrodia cinnamomea, preparation and analysis method thereof
JP2014139153A (en) * 2012-08-24 2014-07-31 Taiwan Antitumer Biotech Co Ltd Antcin derivatives in combination with anti-cancer drugs in the treatment and/or prevention of tumors
CN102863496A (en) * 2012-09-28 2013-01-09 南京泽朗农业发展有限公司 Extraction method of Zhankuic acid A
US20170172922A1 (en) * 2013-05-15 2017-06-22 Hsu-Shen WU Micron taiwanofungus camphoratus liposome structure
CN104111302A (en) * 2014-08-13 2014-10-22 中山安荞生物科技有限公司 Method for measuring adenosine content of health-protection antrodia food
TWI527520B (en) * 2015-03-02 2016-04-01 美和學校財團法人美和科技大學 A use of antcin compound for improving algal bloom
TWM513697U (en) * 2015-06-26 2015-12-11 Ma Shen Kai Ruei Co Ltd Carrier structure of biomimetic interstitial system
CN106279332B (en) * 2016-07-29 2019-08-30 北京大学 Triterpene compound Antcin K and its liver-protecting activity and application
CN110749661B (en) * 2018-07-24 2020-08-25 厦门大学 Quality control method for culturing antrodia camphorata in dish culture mode
TWI832481B (en) * 2020-05-19 2024-02-11 吉亞生物科技股份有限公司 Use of terpenoids in the treatment or prevention of fibrotic diseases
KR20230094886A (en) * 2021-12-21 2023-06-28 한국 한의학 연구원 Anticancer composition comprising Asaroidoxazine B
KR20230094887A (en) * 2021-12-21 2023-06-28 한국 한의학 연구원 Asaroidoxazine C and anticancer composition comprising the same
CN114487252B (en) * 2021-12-28 2024-03-26 陕西嘉禾生物科技股份有限公司 Thin-layer chromatography identification method for distinguishing Antrodia camphorata from Antrodia camphorata

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5475120A (en) * 1990-11-02 1995-12-12 University Of Florida Method for the isolation and purification of taxol and its natural analogues
AUPO864097A0 (en) * 1997-08-19 1997-09-11 Peplin Pty Ltd Anti-cancer compounds
IT1317033B1 (en) * 2000-05-30 2003-05-26 Istituto Di Medicina Speriment METHOD OF EXTRACTION OF PHARMACEUTICAL ACTIVITY PRODUCTS FROM PLANTS SPERMATOPHYTES, PRODUCTS SO OBTAINED AND THEIR USE IN MEDICINE, IN
WO2003041834A1 (en) * 2001-11-13 2003-05-22 Metanomics Gmbh & Co. Kgaa Method for the extraction of components made from organic material
TW591110B (en) 2001-12-03 2004-06-11 Grape King Entpr Inc A method for producing gamma-aminobutyric acid with Antrodia camphorate mycelium and composition thereof
TWI241344B (en) 2002-03-29 2005-10-11 Food Industry Res & Dev Inst Processes for producing an antrodia camphorata culture having pharmacological activity, processes for obtaining a pharmacologically active composition from a culture of A camphorata, products produced thereby and pharmaceutical compositions of cancer...
WO2005053812A1 (en) * 2003-12-08 2005-06-16 Xylon Biotechnologies Ltd. Plant materials extraction method
TWI299665B (en) 2006-05-09 2008-08-11 Univ Chia Nan Pharm & Sciency A method for preparing the extracts from taiwanofungus camphoratus with a capacity for inhibiting the activity of matrix metalloproteinases
TWI389699B (en) 2009-02-13 2013-03-21 Univ Kaohsiung Medical Ethanol extract of antrodia camphorata for inducing apoptosis and preparation method thereof

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8883167B2 (en) 2009-02-13 2014-11-11 Kaohsiung Medical University Ethanol extract of Antrodia camphorata for inducing apoptosis and preparation method thereof
US9950021B2 (en) 2009-11-26 2018-04-24 National Taiwan University Anti-cancer active substance from Antrodia camphorata, method for preparing the same and use thereof
US20110123562A1 (en) * 2009-11-26 2011-05-26 Chiang Been-Huang Anti-cancer active substance from antrodia camphorata, method for preparing the same and use thereof
CN105263893A (en) * 2013-02-20 2016-01-20 国鼎生物科技股份有限公司 Cyclohexenone compositions and process for making thereof
KR20150120424A (en) * 2013-02-20 2015-10-27 골든 바이오테크놀러지 코포레이션 Methods and Compositions For Treating Leukemia
WO2014130619A3 (en) * 2013-02-20 2014-10-16 Golden Biotechnology Corporation Methods and compositions for treating leukemia
WO2014130618A1 (en) * 2013-02-20 2014-08-28 Golden Biotechnology Corporation Cyclohexenone compositions and process for making thereof
CN108309961A (en) * 2013-02-20 2018-07-24 国鼎生物科技股份有限公司 method and composition for treating leukaemia
AU2014218938B2 (en) * 2013-02-20 2018-12-06 Golden Biotechnology Corporation Methods and compositions for treating leukemia
KR102054038B1 (en) 2013-02-20 2019-12-09 골든 바이오테크놀러지 코포레이션 Methods and Compositions For Treating Leukemia
JP2014214113A (en) * 2013-04-25 2014-11-17 康力生技股▲分▼有限公司 Extraction method of antrodia camphorate extract
US8865181B1 (en) 2013-04-26 2014-10-21 Kang Li Biotech Co., Ltd. Extracting method of Antrodia cinnamomea
CN105237365A (en) * 2014-07-09 2016-01-13 台湾利得生物科技股份有限公司 Extract of solid-state cultured mycelium of antrodia camphorata and application thereof
US9751956B2 (en) 2015-03-06 2017-09-05 Ultra-Microrigin Biomedical Technology Co., Ltd. Method of using supercritical fluid technology to separate and purify functional components of Antrodia camphorata
US10806762B2 (en) 2016-05-20 2020-10-20 Taiwan Indigena Botanica Co., Ltd. Method of treatment of cancer cells by using pharmaceutical composition
WO2021222534A1 (en) * 2020-05-01 2021-11-04 Arjil Biotech Holding Company Limited Compounds and methods for prevention and treatment of coronavirus infections

Also Published As

Publication number Publication date
US8883167B2 (en) 2014-11-11
US20120141524A1 (en) 2012-06-07
TW201029658A (en) 2010-08-16
TWI389699B (en) 2013-03-21

Similar Documents

Publication Publication Date Title
US8883167B2 (en) Ethanol extract of Antrodia camphorata for inducing apoptosis and preparation method thereof
Su et al. Fucoxanthin, a marine xanthophyll isolated from Conticribra weissflogii ND-8: Preventive anti-inflammatory effect in a mouse model of sepsis
Liu et al. Ganoderic acid Mf and S induce mitochondria mediated apoptosis in human cervical carcinoma HeLa cells
Chiu et al. Anti-inflammatory cerebrosides from cultivated Cordyceps militaris
Rao et al. Constituents isolated from Cordyceps militaris suppress enhanced inflammatory mediator's production and human cancer cell proliferation
De Silva et al. Bioactive metabolites from macrofungi: ethnopharmacology, biological activities and chemistry
Zha et al. Anti-inflammatory effect of soyasaponins through suppressing nitric oxide production in LPS-stimulated RAW 264.7 cells by attenuation of NF-κB-mediated nitric oxide synthase expression
Kuo et al. Ganoderma lucidum mycelia enhance innate immunity by activating NF-κB
Chen et al. Cytotoxic activities of 9, 11-dehydroergosterol peroxide and ergosterol peroxide from the fermentation mycelia of Ganoderma lucidum cultivated in the medium containing leguminous plants on Hep 3B cells
Van Kiem et al. Chemical constituents of the rhizomes of Hedychium coronarium and their inhibitory effect on the pro-inflammatory cytokines production LPS-stimulated in bone marrow-derived dendritic cells
Xiao et al. Secondary metabolites from Cordyceps species and their antitumor activity studies
JP5395260B2 (en) Anti-inflammatory composition containing macrolactin A and its derivatives as active ingredients
Hseu et al. Antrodia camphorata suppresses lipopolysaccharide-induced nuclear factor-κB activation in transgenic mice evaluated by bioluminescence imaging
CN112159376B (en) Sesterterpene compound and application thereof in preparing anti-inflammatory drugs
Lu et al. Active extracts of wild fruiting bodies of Antrodia camphorata (EEAC) induce leukemia HL 60 cells apoptosis partially through histone hypoacetylation and synergistically promote anticancer effect of trichostatin A
Yang et al. Berberine and limonin suppress IgE production by human B cells and peripheral blood mononuclear cells from food-allergic patients
Bhandari et al. Synthesis of new heterocyclic lupeol derivatives as nitric oxide and pro-inflammatory cytokine inhibitors
Lee et al. IL-17A inhibitions of indole alkaloids from traditional Chinese medicine Qing Dai
Le et al. Chemical constituents of the rhizome of Eleutherine bulbosa and their inhibitory effect on the pro-inflammatory cytokines production in lipopolysaccharide-stimulated bone marrow-derived dendritic cells
WO2011081488A2 (en) Composition for treatment of obesity using wheat bran extract or active ingredient isolated therefrom
Chen et al. A mycochemical investigation of the black Lingzhi mushroom, Amauroderma rugosum (Agaricomycetes), reveals several lipidic compounds with anti-inflammatory and antiproliferative activities
Zhang et al. Molecular mechanisms of the chemical constituents from anti-inflammatory and antioxidant active fractions of Ganoderma neo-japonicum Imazeki
KR20140100117A (en) Compositions comprising the extract of Alnus japonica or the compounds derived therefrom for inhibiting adipogenesis
US20130005826A1 (en) Inhibition of the Survival of Gastric Cancer by Cyclohexenone Compounds from Antrodia Camphorata
US20110059123A1 (en) Inhibition of the Survival of Colorectal Cancer by Cyclohexenone Compounds from Antrodia Camphorata

Legal Events

Date Code Title Description
AS Assignment

Owner name: KAOHSIUNG MEDICAL UNIVERSITY, TAIWAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:WU, YANG-CHANG;LU, MEI-CHIN;CHANG, FANG-RONG;AND OTHERS;REEL/FRAME:023091/0914

Effective date: 20090804

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION