WO2014051359A1 - Pharmaceutical composition comprising neferine as active ingredient for preventing or treating hepatoma - Google Patents

Pharmaceutical composition comprising neferine as active ingredient for preventing or treating hepatoma Download PDF

Info

Publication number
WO2014051359A1
WO2014051359A1 PCT/KR2013/008639 KR2013008639W WO2014051359A1 WO 2014051359 A1 WO2014051359 A1 WO 2014051359A1 KR 2013008639 W KR2013008639 W KR 2013008639W WO 2014051359 A1 WO2014051359 A1 WO 2014051359A1
Authority
WO
WIPO (PCT)
Prior art keywords
neferine
cells
present
liver cancer
neferrin
Prior art date
Application number
PCT/KR2013/008639
Other languages
French (fr)
Korean (ko)
Inventor
윤진수
김군도
최재수
최영현
Original Assignee
부경대학교 산학협력단
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 부경대학교 산학협력단 filed Critical 부경대학교 산학협력단
Publication of WO2014051359A1 publication Critical patent/WO2014051359A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • 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
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/62Nymphaeaceae (Water-lily family)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention relates to a novel use for the treatment of liver cancer of neferine (neferine), and more particularly to a pharmaceutical composition, health functional food for preventing or treating liver cancer containing neferin as an active ingredient.
  • the liver is the largest organ in the body, 800-1,200g in adults, where various types of malignant tumors occur. Hepatocellular carcinoma and cholangiocarcinoma account for 95%. The remaining 5% are very rare, such as hepatoblastoma (hepatoblastoma), hepatocellular carcinoma in children, hepatocellular carcinoma, mixed cholangiocarcinoma, undifferentiated cancer, cholangiocystic adenocarcinoma, carcinoid tumor.
  • HCC hepatocellular carcinoma
  • Hepatocellular carcinoma is the fifth most common tumor in the world, with half a million deaths each year (Okuda 2000). Survival rates of hepatocellular carcinoma patients have not improved over the last 20 years and have almost the same incidence of mortality (Marrero, Fontana et al. 2005).
  • the average age of onset of hepatocellular carcinoma (HCC) is approximately 55 years, most of which were found to be between 40-60 years.
  • Hepatitis viruses include A, B, C, D, E, and F.
  • hepatitis G has been discovered.
  • viruses that are a problem in our country, A, B, and C. Of these, two are related to liver cancer: B and C.
  • B and C According to statistics since 1990, when hepatitis B and C viruses became available, 85% of those who received liver cancer were infected with hepatitis B or C virus. It is estimated that hepatitis B and C viruses act on normal hepatocytes, causing mutations to increase the chance of developing cancer. Therefore, in Korea, people infected with hepatitis virus are classified as a high risk group of liver cancer.
  • liver cancer treatment agent having high sensitivity without side effects.
  • Neferine is one of the major bisbenzylisoquinline alkaloids from the lotus ( Nelumbo nucifera Gaerth.) Embryo. Alkaloids abundant in these green plant embryos have been reported to have a variety of biological activities, including anti-arrhythmia, anti-hypertension, relaxants, anti-diabetic, cholinesterase inhibition, sedatives and anti-multi-drug resistance.
  • the lotus embryo has been widely used as a traditional herbal medicine for neurological diseases, insomnia, continuous fever and cardiovascular diseases such as hypertension and arrhythmia.
  • nephrine can stop the cell cycle progression of hepatocellular carcinoma caused by viral infection, induce apoptosis, autophagy.
  • the present invention was completed by confirming that it is useful for preventing, improving or treating liver cancer as it has anticancer effects through autophagy induction, angiogenesis inhibition, and cell migration inhibitory activity.
  • Still another object of the present invention is to provide a health functional food that is effective in preventing or improving liver cancer by having excellent cytotoxicity to liver cancer cells caused by viral infection.
  • the present invention provides a pharmaceutical composition for preventing or treating liver cancer comprising Neferine (Neferine) as an active ingredient.
  • Neferine Neferine
  • the neferine may be derived from the lotus embryo.
  • the liver cancer may be hepatocellular carcinoma caused by viral infection.
  • the pharmaceutical composition is through the cell cycle arrest (cell cycle arrest), induction of apoptosis (apoptosis), autophagy induction, angiogenesis inhibition and cell migration inhibitory activity of cancer cells It can have an anticancer effect.
  • the present invention also provides a health functional food for the prevention or improvement of liver cancer comprising Neferine (Neferine) as an active ingredient.
  • Neferine Neferine
  • the neferine may be derived from the lotus embryo.
  • the liver cancer may be hepatocellular carcinoma caused by viral infection.
  • the food is selected from the group consisting of beverages, meat, chocolate, foods, confectionery, pizza, ramen, other noodles, gums, candy, ice cream, alcoholic beverages, vitamin complexes and health supplements Can be.
  • Neferine of the present invention can stop the cell cycle progression of liver cancer cells, and have anticancer effects through induction of apoptosis, induction of autophagy, inhibition of angiogenesis and cell migration.
  • neferrin has a therapeutic effect optimized for hepatocellular carcinoma induced by viral infection, as it has cytotoxicity specifically to hepatocellular carcinoma cells caused by viral infection among hepatic cancer cells.
  • 1 is a graph showing the survival rate of cells after treatment with neprene for each concentration of Hep3B, Sk-Hep1 and THLE-3 cancer cell lines for 24 hours.
  • Figure 2 shows the change in nuclear condensation of cells through DAPI staining after treatment with Hep3B cells neferin (20, 25 ⁇ M) at 6 hour intervals.
  • Figure 4 shows the expression level of proteins related to the cell cycle after treatment with Heper3B cells for 24 hours at different concentrations (15, 20, 25 ⁇ M) by Western blot.
  • FIG. 5 shows that p21 Waf1 / Cip1 protein expression levels detected by immunofluorescence analysis after treatment of Hep3B cells with 15 ⁇ M concentration of nephrin for 12 hours using a fluorescence microscope ( ⁇ 1000) (p21 Waf1 / Cip1). : green, nucleic: blue, cytoskeletal actin: red).
  • FIG. 6A shows that hep3B cells were treated with nephrine at different concentrations (15, 20, 25 ⁇ M) for 24 hours, and then mitochondrial membrane-related proteins (Bim, BID, Bax, Bak, Puma) were measured by Western blot.
  • Figure 6B shows the amount of apoptotic protein (cleaved caspase-8, -3, -6, -7 and PARP) involved in apoptosis after treatment with Hep3B cells for 24 hours at different concentrations (15, 20, 25 ⁇ M) was measured via Western blot.
  • FIG. 7A shows the cleaved caspase-3 protein expression level detected by immunofluorescence analysis after treatment with Hep3B cells for 15 hours at 15 ⁇ M concentration of neferin (cleaved caspase-3). : green, nucleic: blue, cytoskeletal actin: red).
  • FIG. 7B shows the cleaved caspase-8 protein expression level detected by immunofluorescence assay after treatment with Hep3B cells for 15 hours at 15 ⁇ M concentration of neferin (cleaved caspase-8). : green, nucleic: blue, cytoskeletal actin: red).
  • vesicle stress-related chaperon proteins (Bip, calnexin, PDI, calpain2 and caspase-12) over time after treatment with 20 ⁇ M concentration of neprine in Hep3B cells.
  • FIG. 9 is a caspase-12 protein expression level detected by immunofluorescence analysis after treatment with Hep3B cells for 15 hours at 15 ⁇ M concentration of nephrin using a fluorescent microscope ( ⁇ 1000) (caspase-12: green , nucleic: blue, cytoskeletal actin: red).
  • FIG. 10 shows the degree of autophagosomes formation in the cytoplasm with fluorescence microscopy ( ⁇ 1000) after treatment with 20 ⁇ M of nephrine in Hep3B cells transformed with GFP-LC3B plasmid. .
  • FIG. 12A is a measurement of the degree of tube formation using an inverted microscope ( ⁇ 40) after 18 hours of treatment with concentrations (10, 20, 30 ⁇ M) of HUVECs cells, and FIG. 12B shows the total tube based on the results. This is a graph of lengths converted into percentages using Wimasis Image Aalysis software.
  • the present invention is characterized by providing a pharmaceutical composition for preventing or treating liver cancer comprising Neferine (Neferine) represented by the following formula (1) as an active ingredient.
  • Neferine represented by the following formula (1) as an active ingredient.
  • Alkaloids contained in green plant embryos have been reported to have a variety of biological activities including anti-arrhythmia, anti-hypertension, relaxants, anti-diabetic, cholinesterase inhibition, sedatives and anti-multi-drug resistance.
  • the lotus embryo has been widely used as a traditional herbal medicine for neurological diseases, insomnia, continuous fever and cardiovascular diseases such as hypertension and arrhythmia.
  • the present inventors can stop the cell cycle progression of hepatocellular carcinoma caused by nephrine-induced viral infection, and anti-cancer effect through induction of apoptosis, induction of autophagy, inhibition of angiogenesis and cell migration.
  • a substance useful for the prevention, improvement or treatment of liver cancer, as well as pharmaceuticals such as pharmacological composition, as well as health functional food, it was first identified the first.
  • Experimental Example 2 of the present invention in order to examine whether apoptosis of hep3B, a hepatocellular carcinoma cell line of neferrin, was performed, a DAPI assay was performed. 6 hours after the treatment, it was confirmed that nuclear membrane changes such as condensation and blebbing of cells appeared in the experimental group. In particular, apoptosome formation, which is typical of apoptotic cells, was observed in the experimental group 24 hours after neferrin treatment (see FIG. 2).
  • the cell cycle is performed in a certain order by a mechanism defined in the cell.
  • a regulator that acts to restore it acts, such as cyclin (cyclin), cyclin- Cyclin-dependent kinase (hereinafter abbreviated 'CDK') and cyclin-dependent kinase inhibitor (hereinafter abbreviated 'CDKI').
  • CDK4, 6, 8, etc. are activated by the type of cells in the early stage of G1 phase, CDK2 in the late stage of G1 and early stage of S1, and the progression from G2 to M CDK1 is known to play an important role. Binding to cyclins is essential for the activity of CDK.
  • CDK4, 6 and 8 are activated by binding to cyclin D, and CDK2 binds to cyclins A and E.
  • CDK1 binds to cyclins B and A, in addition to cyclins G, F and the like.
  • the specific cyclin-CDK complex is activated and the proteins phosphorylated specifically for CDK are responsible for the progression of the cell cycle.
  • CDK acts as an essential factor for cyclin activity.
  • Activated CDK-cyclin is divided into cyclin regulatory unit and CDK active unit, and cyclin CDK regulation can be seen in two ways.
  • the combination of cyclin and CDK induces structural changes in the protein, making the arrangement of the ATP phosphate group easy to transfer to the substrate protein.
  • the position of the T loop that prevents the protein substrate from accessing the CDK is changed, thereby facilitating access to the substrate.
  • the activity of CDK is activated only at certain times of the cell cycle because of cyclin synthesis that occurs specifically in the cell cycle.
  • cyclin D is most likely synthesized in the middle of G1, and is mainly induced by mitogens such as cell growth factors.
  • Cyclin D has three types of subtypes (D1, 2, and 3), and the degree of expression varies depending on the type of cell. For example, inhibiting the synthesis of cyclin D stops the cell cycle G1, and overexpressing cyclin D shortens the G1 group and starts the cell cycle without mitogen.
  • the p15 INK4B, p16 INK4A, p18 INK4C , p19 INK4D, p27 Kip1 and p21 WAF1 / Cip1 and cell cycle regulation such as characters are known as core control to inhibit cyclin D1 / CDK4,6 or Cyclin E / CDK2 complexes. Therefore, such cell cycle regulators are called negative cell cycle regulators, and when they are expressed, cell cycle progression can be inhibited.
  • cleaved caspase-8 and -3 were found to be prominently expressed in neprine-treated Hep3B cells compared to the untreated group, which was not treated with neferin, and apoptosomes induced by neferin were caspase in the cytoplasm. It could be deduced that activating -3 and caspase-8 causes apoptosis (see FIG. 7).
  • nepherin has cytotoxicity to liver cancer cells induced by virus infection among hepatic cancer cells, cell cycle arrest, induction of apoptosis and autophagy of liver cancer cells. It has been experimentally demonstrated to have anticancer effects through induction, angiogenesis inhibition and cell migration inhibitory activity.
  • composition of the present invention comprising nephrine as an active ingredient can effectively prevent or treat liver cancer, and in particular, as it has cytotoxicity specifically to liver cancer cells induced by viral infection, liver cancer caused by viral infection It has a therapeutic effect optimized for cells.
  • the neferrin according to the present invention can be used in the form of salts, preferably pharmaceutically acceptable salts.
  • the salt is preferably an acid addition salt formed by a pharmaceutically acceptable free acid, and an organic acid and an inorganic acid may be used as the free acid.
  • the organic acid is not limited thereto, citric acid, acetic acid, lactic acid, tartaric acid, maleic acid, fumaric acid, formic acid, propionic acid, oxalic acid, trifluoroacetic acid, benzoic acid, gluconic acid, metasulfonic acid, glycolic acid, succinic acid, 4-toluenesulfonic acid, Glutamic acid and aspartic acid.
  • the inorganic acid includes, but is not limited to, hydrochloric acid, bromic acid, sulfuric acid and phosphoric acid.
  • neferrin according to the present invention may be commercially available, or may be one that is separated from nature or manufactured by chemical synthesis known in the art.
  • the separation method When separating the neferrin of the present invention from natural products, there is no limitation to the separation method, any known method can be used.
  • the N. nucifera embryo was extracted under reflux with methanol, and concentrated by vacuum drying to obtain a lotus embryo methanol extract.
  • the extract thus obtained was suspended in distilled water, followed by dichloromethane and ethyl acetate. And sequentially fractionated with n-butanol, and the dichloromethane fraction was subjected to silica gel column chromatography again to purify the final neferrin.
  • composition of the present invention may be prepared using a pharmaceutically acceptable and physiologically acceptable adjuvant in addition to such an active ingredient as a pharmaceutical composition comprising the neferin as an active ingredient, the adjuvant, an disintegrant, Sweetening agents, binders, coatings, expanding agents, lubricants, lubricants, or flavoring agents may be used.
  • the pharmaceutical composition may be preferably formulated into a pharmaceutical composition including one or more pharmaceutically acceptable carriers in addition to the above-described active ingredient for administration.
  • Formulation forms of the pharmaceutical composition may be granules, powders, tablets, coated tablets, capsules, suppositories, solutions, syrups, juices, suspensions, emulsions, drops or injectable solutions.
  • the active ingredient may be combined with an oral, nontoxic, pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like.
  • suitable binders, lubricants, disintegrants and coloring agents may also be included in the mixture.
  • Suitable binders include but are not limited to natural and synthetic gums such as starch, gelatin, glucose or beta-lactose, corn sweeteners, acacia, trackercance or sodium oleate, sodium stearate, magnesium stearate, sodium Benzoate, sodium acetate, sodium chloride and the like.
  • Disintegrants include, but are not limited to, starch, methyl cellulose, agar, bentonite, xanthan gum, and the like.
  • Acceptable pharmaceutical carriers in compositions formulated in liquid solutions are sterile and physiologically compatible, including saline, sterile water, Ringer's solution, buffered saline, albumin injectable solutions, dextrose solution, maltodextrin solution, glycerol, ethanol and One or more of these components may be mixed and used, and other conventional additives such as antioxidants, buffers and bacteriostatic agents may be added as necessary. Diluents, dispersants, surfactants, binders and lubricants may also be added in addition to formulate into injectable formulations, pills, capsules, granules or tablets such as aqueous solutions, suspensions, emulsions and the like. Furthermore, the method disclosed in Remington's Pharmaceutical Science, Mack Publishing Company, Easton PA can be formulated according to each disease or component, as appropriate in the art.
  • the neferrin of the present invention may be included in the composition in a concentration of 1 to 300 ⁇ M, and the neferrin compound of the present invention may be included in 0.1 to 95% by weight relative to the total weight of the composition.
  • the present invention also provides the use of a composition comprising neferine (neferine) as an active ingredient for the manufacture of a medicament for preventing or treating liver cancer.
  • a composition comprising neferine (neferine) as an active ingredient for the manufacture of a medicament for preventing or treating liver cancer.
  • the composition of the present invention comprising the neferrin as an active ingredient can be used for the manufacture of a medicament for the prevention or treatment of liver cancer.
  • the present invention also provides a method for preventing or treating liver cancer comprising administering neferine to a mammal.
  • mammal refers to a mammal that is the subject of treatment, observation or experimentation, preferably human.
  • the term “therapeutically effective amount” means an amount of an active ingredient or pharmaceutical composition that induces a biological or medical response in a tissue system, animal or human, as thought by a researcher, veterinarian, doctor or other clinician, which Amounts that induce alleviation of the symptoms of the disease or disorder being treated. It will be apparent to those skilled in the art that the therapeutically effective dosages and frequency of administrations for the active ingredients of the present invention will vary depending on the desired effect. Therefore, the optimal dosage to be administered can be easily determined by one skilled in the art and includes the type of disease, the severity of the disease, the amount of active ingredients and other ingredients contained in the composition, the type of formulation, and the age, weight, general health of the patient.
  • the neferine of the present invention is preferably administered at a dose of 0.01 mg / kg to 250 mg / kg once or several times a day.
  • the composition comprising the neferine of the present invention as an active ingredient is an oral, rectal, intravenous, intraarterial, intraperitoneal, intramuscular, intrasternal, transdermal, topical, intraocular or intradermal route. Can be administered in a conventional manner.
  • the present invention also provides a health functional food for preventing or improving liver cancer comprising neferine as an active ingredient.
  • the health functional food of the present invention can be prepared and processed in the form of tablets, capsules, powders, granules, liquids, pills and the like for the purpose of preventing and improving liver cancer.
  • health functional food refers to a food manufactured and processed using raw materials or ingredients having functional properties useful for the human body according to the Health Functional Food Act No. 6767, and nutrients for the structure and function of the human body. It is meant to be consumed for the purpose of regulating or obtaining a useful effect for health use such as physiological action.
  • the health functional food of the present invention may include a conventional food additive, and the suitability as a food additive, unless otherwise specified, in accordance with the General Regulations of the Food Additives and General Test Methods approved by the Food and Drug Administration, etc. Judging by the standards and standards.
  • Food Additive Reduction examples include chemical compounds such as ketones, glycine, calcium citrate, nicotinic acid, and cinnamic acid; Natural additives such as dark blue pigment, licorice extract, crystalline cellulose, high color pigment and guar gum; And mixed preparations such as sodium L-glutamate, algae additives, preservatives and tar dyes.
  • the health functional food in the form of tablets is a mixture of neferine, an active ingredient of the present invention, with an excipient, a binder, a disintegrant, and other additives, followed by granulation in a conventional manner, and then a lubricant and the like. Compression molding, or the mixture may be directly compression molded.
  • the health functional food in the form of tablets may contain a mating agent or the like as necessary.
  • Hard capsules among the health functional foods in the form of capsules may be prepared by filling a mixture of additives such as excipients with neferine, which is the active ingredient of the present invention, in a conventional hard capsule, and the soft capsules are neferine )
  • the soft capsule agent may contain a plasticizer such as glycerin or sorbitol, a colorant, a preservative, and the like, as necessary.
  • the cyclic health functional food can be prepared by molding a mixture of neferine, an active ingredient of the present invention, an excipient, a binder, a disintegrant, and the like by using a conventionally known method. It may be encapsulated with a skin coating, or the surface may be coated with a material such as starch, talc.
  • the health functional food in the form of granules can be prepared by granulation of a mixture of neferine, an excipient, an excipient, a binder, a disintegrant, and the like, which is an active ingredient of the present invention.
  • a mating agent and the like are examples of neferine, an excipient, an excipient, a binder, a disintegrant, and the like.
  • the health functional food may be beverages, meat, chocolate, foods, confectionery, pizza, ramen, other noodles, gum, candy, ice cream, alcoholic beverages, vitamin complexes and health supplements.
  • DMSO, BSA and propidium iodide were purchased from Sigma Aldrich (St. Louis, MO, USA); EZ-Cytox Cell Viability Assay Solution WST-1 was purchased from Daeil Lab Service (Jong-No, Seoul, Korea); DAPI was purchased from Roche (Pleasanton, CA, USA); Lysis buffer [50 mM Tris-Cl (pH 7.5), 150 mM NaCl, 1 mM DTT, 0.5% NP-40, 1% Triton X-100, 1% deoxycholate, 0.1% SDS] and proteinase inhibitors (PMSF, EDTA, Aprotinin, Leupeptin, and Prostatin A) were purchased from Intron biotechnology (Gyeonggi, Korea); The Protein Quantification Kit (CBB solution) was purchased from Dojindo Molecular Technologies (Rockville, MD, USA); Nitrocellulose membranes were purchased from PALL Life Sciences (Ann Arbor, MI, USA); Enhanced chemiluminescent (ECL
  • the dichloromethane fraction (13.8 g) in the fraction was again subjected to chromatography on a silica gel column (120 mm id), whereby finally, 920 mg of neferine was obtained.
  • a mixed solvent of benzene-ethyl acetate-diethylamine (7: 2: 1, isocratic) was used as the elution solvent.
  • HCC Hep3B, Sk-Hep1, THLE-3 (cells immortalized by infecting normal hepatocytes with SV40 large T antigen) and HUVEC (human venous venous endothelial cells) cells were obtained from the American Tissue Culture Collection (Manassas, VA, USA) .
  • Hep3B and Sk-Hep1 cells were cultured using minimum essential medium (MEM / EBSS) (HyClone, Logan, UT, USA) containing Earle's Balanced Salts, and THLE-3 cells were gentamicin / Empotericin and epinephrine were removed and cultured using Bronchial Epithelia Cell Growth Medium (Clonetics BEGM bullet kit; CC3170, Lonza, Walkersville, MD, USA) medium containing 500 ml basal medium, 5 ng / ml EGF, 70 ng. Culture was added by adding / ml phosphoethanolamine.
  • MEM / EBSS minimum essential medium
  • THLE-3 cells were gentamicin / Empotericin and epinephrine were removed and cultured using Bronchial Epithelia Cell Growth Medium (Clonetics BEGM bullet kit; CC3170, Lonza, Walkersville, MD, USA) medium containing 500 ml basal medium
  • HAVECs Human Umbilical Vein Endothelial Cells
  • EBM-2 endothelial basal medium-2
  • EGM-2 kit Lignogen kit
  • All cell lines in this experiment were 37 ° C., 5% CO in media containing 10% heat inactivated FBS (HyClone), 100 U / ml penicillin, and 100 ⁇ g / ml streptomycin (PAA Laboratories GmbH, PA, Austria). It was incubated in the conditions of 2 .
  • THLE-3 cells were coated with 0.01 mg / ml BSA, 0.01 mg / ml fibronectin and 0.03 mg / ml bovine collagen type ⁇ (BD biosciences, San Jose, Calif., USA) for 2 hours at 37 ° C and then incubated.
  • Example 2 the cell viability assay was performed using Hep3B, Sk-Hep1, and THLE-3 cell lines cultured in Example 2 to confirm the cytotoxicity of the cancer cell line of nephrin obtained through Example 1. .
  • Hep3B, Sk-Hep1 and THLE-3 cells were each resuspended in 100 ⁇ l medium at 1 ⁇ 10 4 cells density and dispensed into three 96-well plates and incubated for 24 hours. Then, neferrin at various concentrations (5, 10, 15, 20, 25, 30 ⁇ M) was treated with the cells, incubated for 24 hours, exchanged with fresh medium, and then added with 10 ⁇ l of WST-1 solution for 5 hours. Reacted for a while. Reaction absorbance was measured at 460 nm using an ELISA reader (Molecular Devices, Sunnyvale, Calif., USA). The results were expressed as mean ⁇ standard deviation through three independent experiments, and the inhibition graph was derived using the mean values obtained from each concentration associated with the control values.
  • neferrin has a specific cytotoxic sensitivity to Hep3B cells.
  • neferin did not show cytotoxicity to both Hep3B and Sk-Hep1 liver cancer cell lines, and showed specific cytotoxicity to Hep3B cell line, suggesting that hepatic cancer cells may have anticancer activity in different types.
  • the nephrine of the present invention confirmed specific cytotoxicity to Hep3B cell line even in hepatocellular carcinoma cell line.
  • experiments were conducted on various mechanisms related to anticancer activity in Hep3B cell line.
  • DAPI is an abbreviation of 4, 6-diamidino-2-phenylindole dihydrochloride hydrate and is a probe that can be permeated to cells and used to stain living cell nuclei in a fixed state. It forms a bond with DNA and a minor groove, and prefers to bind with AT rich DNA, and becomes stable through hydrogen bonding.
  • DAPI bound to DNA is about 20 times brighter than non-DNA bound and emits light by ultraviolet light under a fluorescence microscope. When combined with DNA, it exhibits an absorption maximum at 358 nm and an emission maximum at 461 nm. In this experiment, hepatic cancer cells were stained with DAPI for detection of nuclear condensation and apoptosome formation following neferrin treatment and analyzed under fluorescence microscope ( ⁇ 1000).
  • Hep3B cells were first incubated in a cover glass bottom culture dish for 24 hours, treated with neferin (20, 25 ⁇ M) at 6 hour intervals, and washed once with PBS buffer, followed by DAPI solution (1 ⁇ g / ml). Staining was added. Incubated in the dark at 37 ° C. for 20 minutes, and the cells were washed once again with methanol. Nuclear changes were observed using an ECLIPSE 50i fluorescence microscope (Nikon, Tokyo, Japan).
  • nuclear membrane changes such as shrinkage and blebbing of cells were observed in the experimental group 6 hours after the neferrin treatment compared to the non-nephrrine-treated group. It was confirmed that it appeared.
  • a typical characteristic of apoptotic cells nuclear condensation (apoptotic formation) was observed in the experimental group 24 hours after neferrin treatment.
  • Hep3B cells were treated with nephrine at different concentrations (15, 20, 25 ⁇ M) for 24 hours, and then treated with trypsin, and then fixed overnight using 70% ethanol at 4 ° C. The cells were then resuspended in PBS buffer containing 0.2 mg / ml RNase A and then further incubated at 37 ° C. for 1 hour. The cells were stained with 40 ⁇ g / ml propidium iodide (PI) for 30 minutes in the dark. Dispersion of subgenomic DNA content was analyzed using flow cytometer (BD biosciences).
  • PI propidium iodide
  • the sub-G1 cell population increases in proportion to the neferrin treatment concentration (15, 20, 25 ⁇ M).
  • the sub-G1 cell population showed 1.35% in the untreated group without neferrin, while 4.78% in the experimental group treated with 15 ⁇ M concentration of neferrin, and 16.01% in the experimental group treated with 20 ⁇ M concentration of neferin. Increased.
  • the G1 / S phase was increased approximately 9% in the experimental group treated with 20 ⁇ M concentration of neferrin compared to the untreated group.
  • Hep3B cells were treated with neferin by concentration (15, 20, 25 ⁇ M) for 24 hours, washed twice with cold PBS, and then lysed with cold lysis buffer. Then, the reaction was carried out on ice for 30 minutes, followed by centrifugation at 14,000 rpm for 20 minutes to remove insoluble matters. Protein content of the cell lysate was measured using a Protein Quantification Kit (CBB solution). Western blots were performed using 30 ⁇ g of protein isolated via electrophoresis on a 12% polyacrylamide gel, transferred to nitrocellulose membrane and subjected to immune reaction with indicator antibody. Chemiluminescence was detected using chemiluminescent (ECL) reagent.
  • ECL chemiluminescent
  • FIG. 6A shows Western blot results of Bcl-2 family proteins (Bim, Bid, Bax, Bak and Puma) known to modulate mitochondrial-mediated apoptosis pathways, whereby Bak and Puma protein expression was treated with nephrin. It was shown that the concentration was significantly increased, and Bim, Bid and Bax were found to be slightly increased in expression.
  • Bcl-2 family proteins Bim, Bid, Bax, Bak and Puma
  • Figure 6b shows the Western blot results of apoptotic proteins (cleaved caspase-8, -3, -6, -7 and PARP) associated with apoptosis due to endoplasmic reticulum stress, through which caspase-3 protein expression is neferin It was found that the expression of cleaved caspase-6 and cleaved PARP protein was markedly increased while it was shown to decrease depending on the treatment concentration.
  • apoptotic proteins cleaved caspase-8, -3, -6, -7 and PARP
  • Autophagy has mechanisms by which eukaryotic cells, old organelles, damaged organelles, and cytoplasm are phagocytized, digested, and reused, which are involved in the turnover of basic cellular components or changes in the external environment, ie nutrients. It may also be induced by depletion, pathogen infiltration and oxygen starvation. Recently published studies have reported that cancer cells may have anti-cancer effects by inducing autophagy. Autophagy has been reported to be activated by tumor therapies in many tumor cell lines, but the association between cancer formation and inhibition has not been elucidated.
  • the neferin-treated experimental group was found to significantly up-regulate Bip, calnexin, PDI, calpain2 and caspase-12 protein expression levels depending on time.
  • These results mean that the expression of chaperon protein is increased by intracellular mechanisms for endoplasmic reticulum stress relief because nephrin of the present invention causes endoplasmic reticulum stress in liver cancer cell line.
  • LC3 is an important component in the development of autophagy and is used as a specific marker for autophagy by participating in the formation of autophagosome double membranes.
  • LC3 exists in two forms in cells.
  • LC3- ⁇ is pre-existing in the cytoplasm and is transformed into LC3-II when stressed inside or outside the cell, which is involved in the formation of autophagosome bilayers. Therefore, when much LC3-II is expressed, autolysates are also in a correlation.
  • GFP-LC3B green fluorescent protein-light chain 3B plasmids were prepared, and the present plasmid was transformed by introducing Hep3B cells.
  • PCR was used to amplify LC3B full-length sequences from human liver cDNA libraries (Clontech, Mountain View, CA, USA). Primer sequences used for PCR amplification are shown in Table 1 below. Purified PCR products were introduced into the pGEM-T Easy vector (Promega, Madison, Wis., USA) and then cleaved back with HindIII and Bam Hl and fused with N-terminal GFP in the pEGFP-N2 vector (Clontech).
  • GFP-LC3B plasmid The structure thus prepared was confirmed through DNA sequencing and named GFP-LC3B plasmid. Meanwhile, Hep3B cells were cultured in sterile 6-well plates for 24 hours, and then transformed into GFP-LC3B plasmid using Fugene 6 reagent (Roche Diagnostics GmbH, Mannheim, Germany) mixture in FBS-free medium. Transformed cells were incubated in regular complete medium for one day, then treated with 20 ⁇ M neferrin at regular intervals (6, 12, 18, 24), washed with cold PBS buffer and fixed in cold methanol for 5 minutes at room temperature. I was. The cells were washed again with PBS buffer twice and placed on coverslips with Gold Antifade Reagent. The cells were observed using an ECLIPSE 50i fluorescence microscope.
  • wound-healing assay was performed after treating neferin to Hep3B, a hepatic cancer cell line. ) was performed.
  • Hep3B (3.5 ⁇ 10 4 cells per well) cells were dispensed into an IBIDI culture insert (Ibidi GmbH, M, Germany) chamber consisting of two reservoirs separated by a 50 ⁇ m septum, followed by 5% CO 2 at 24 ° C. Incubate for hours. After incubation, the contents were removed and the cells harvested and re-cultured in culture medium. Cell migration was measured at 12 hour intervals after neferrin treatment using an inverted microscope. Images were calculated as percentage of cell-covered area using Wimasis Image Analysis software.
  • the cell migration was suppressed depending on the neferrin treatment concentration.
  • the cell coverage area was reduced by about 20% in the experimental group treated with neferrin at a concentration of 15 ⁇ M after 72 hours of treatment, compared to the increase in cell migration in the untreated group without nephrin treatment. 13B).
  • HCC hepatocellular carcinoma
  • CDKs Cyclin-dependent kinases
  • ER endoplasmic reticulum
  • HBV hepatitis B virus
  • BSA bovine serum albumin
  • HUVECs human umbilical vein endothelial cells
  • WST-1 2- (4-iodophenyl) -3- (4-nitrophenyl) -5- (2,4-disulfophenyl) -2H-tetrazolium, monosodium salt
  • DAPI 4, 6-diamidino-2-phenylindole dihydrochloride hydrate
  • GFP Green fluorescent protein
  • PBS phosphate-buffered saline
  • PARP poly (ADP-ribose) polymerase
  • PCD programmed cell death
  • MOMP mitochondrial outer membrane permeabilization
  • Neferin of the present invention is a substance capable of preventing or treating liver cancer, as well as pharmaceuticals such as pharmacological compositions, and can be used as a health functional food, etc. It is expected that its value will be very high when applied to food industry and pharmaceutical industry . In addition, since the neferine of the present invention does not show cytotoxicity except certain liver cancer cells, the composition of the present invention comprising the same as an active ingredient has a safe advantage even for long-term use.

Landscapes

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

Abstract

The present invention relates to a novel use of neferine for treating hepatoma, and more particularly, to a pharmaceutical composition and to a health functional food comprising neferine as an active ingredient for preventing or treating hepatoma. The neferine of the present invention can have an anticancer effect through the activities of stopping a cell cycle, inducing apoptosis and autophagy, and inhibiting angiogenesis and cell migration of hepatocarcinoma cells. In particular, since neferine has cytotoxicity specificity for hepatocarcinoma cells caused by viral infection even among hepatocarcinoma cells, the present invention has an effect optimized for treating hepatocarcinoma cells caused by viral infection. Thus, the neferine of the present invention can be used as a health functional food, as well as a medicine such as a pharmacological composition used as a material which can prevent or treat hepatoma. Therefore, it is expected that the value of the present invention is very high when the present invention is applied to the food industry and the pharmaceutical industry. Furthermore, since the neferine of the present invention does not show cytotoxicity for cells other than certain hepatocarcinoma cells, the composition of the present invention, which comprises neferine as an active ingredient, is beneficial in consideration of the safety of long-term use.

Description

네퍼린을 유효성분으로 포함하는 간암 예방 또는 치료용 약제학적 조성물Pharmaceutical composition for preventing or treating liver cancer, including neferin as an active ingredient
본 발명은 네퍼린(neferine)의 간암 치료를 위한 신규 용도에 관한 것으로, 더욱 상세하게는 네퍼린을 유효성분으로 포함하는 간암 예방 또는 치료용 약제학적 조성물, 건강기능식품에 관한 것이다.The present invention relates to a novel use for the treatment of liver cancer of neferine (neferine), and more particularly to a pharmaceutical composition, health functional food for preventing or treating liver cancer containing neferin as an active ingredient.
간장은 성인의 경우 800-1,200g로 체내 최대의 장기이며 이곳에서는 여러 종류의 악성종양이 발생하는데, 간세포암과 담관세포암이 95%를 차지한다. 나머지 5%에는 소아의 간암인 간모세포종(간세포아종), 성인에서의 간세포, 담관세포혼합암, 미분화암, 담관낭포선암, 카르시노이드종양 등 극히 드문 것들이 있다.The liver is the largest organ in the body, 800-1,200g in adults, where various types of malignant tumors occur. Hepatocellular carcinoma and cholangiocarcinoma account for 95%. The remaining 5% are very rare, such as hepatoblastoma (hepatoblastoma), hepatocellular carcinoma in children, hepatocellular carcinoma, mixed cholangiocarcinoma, undifferentiated cancer, cholangiocystic adenocarcinoma, carcinoid tumor.
현재 성인 간암의 대부분(90% 이상)은 간세포암(hepatocellular carcinoma: HCC)으로 집계되고 있다. 간세포암은 해마다 50 만명이 사망하는 세계에서 5번째로 일반적인 종양이다(Okuda 2000). 간세포암 환자의 생존률은 지난 20년에 걸쳐 개선되지 않고 있으며 사망률과 거의 동일한 발병률을 지니고 있다 (Marrero, Fontana et al. 2005). 간세포암(HCC)의 평균 발증연령은 대략 55세이며, 대부분은 40-60세 사이에 상기 질환에 걸리는 것으로 조사되었다.Currently, the majority of adult liver cancer (90% or more) is hepatocellular carcinoma (HCC). Hepatocellular carcinoma is the fifth most common tumor in the world, with half a million deaths each year (Okuda 2000). Survival rates of hepatocellular carcinoma patients have not improved over the last 20 years and have almost the same incidence of mortality (Marrero, Fontana et al. 2005). The average age of onset of hepatocellular carcinoma (HCC) is approximately 55 years, most of which were found to be between 40-60 years.
간세포암의 원인은 전세계적 규모로 보았을 때 몇 가지가 있지만, 국내의 경우 대부분이 간염바이러스에 의한 감염이 원인으로 추측되고 있다. 간염바이러스는 A, B, C, D, E, F 등의 종류가 있으며, 아주 최근에 G형이 발견되었다. 우리나라에서 문제가 되고 있는 바이러스는 A, B, C의 세 종류인데, 이중 간암과 관련되는 것은 B, C의 두 종류이다. B형, C형 간염바이러스의 검사가 가능하게 된 1990년 이후의 통계에 의하면, 간암의 수수를 받은 사람들 중 85%는 B형 또는 C형 간염바이러스에 감염되어 있는 것으로 조사되었다. 이는 B형, C형 간염바이러스가 정상 간세포에 작용하여 돌연변이를 일으켜 간암으로 발전 가능성을 높이는 것으로 추정되고 있다. 따라서 우리나라의 경우에는 간염바이러스에 감염된 사람이 간염에 걸리기 쉬운 간암의 고위험군으로 분류되고 있다.There are several causes of hepatocellular carcinoma on a global scale, but most cases in Korea are suspected to be caused by hepatitis virus. Hepatitis viruses include A, B, C, D, E, and F. Very recently, hepatitis G has been discovered. There are three types of viruses that are a problem in our country, A, B, and C. Of these, two are related to liver cancer: B and C. According to statistics since 1990, when hepatitis B and C viruses became available, 85% of those who received liver cancer were infected with hepatitis B or C virus. It is estimated that hepatitis B and C viruses act on normal hepatocytes, causing mutations to increase the chance of developing cancer. Therefore, in Korea, people infected with hepatitis virus are classified as a high risk group of liver cancer.
지난 수십년 동안 간암을 치료하는 방법으로 수술, 방사선요법, 화학요법 등이 이루어져 왔고, 동시에 간암 치료제, 간염 치료제, 간염 백신 등의 연구 개발과 상품화가 진행되어 왔으나, 간암의 발생률 및 사망률은 크게 줄어들지 않고 있다. 간세포암의 치료와 관련하여, 현재까지 알려진 치료제로는 epirubicin, cisplatin, 5-fluorouracil, etoposide 등이 있으나 낮은 치료 효율을 보이며, 또한 이러한 화학치료제들의 장기사용은 화학치료적 내성을 야기하는 문제점이 있다. 따라서 부작용 없이 높은 민감도를 가진 간암 치료제의 개발이 절실한 실정이다.Surgery, radiotherapy, and chemotherapy have been used to treat liver cancer for the past several decades. At the same time, research and development and commercialization of liver cancer, hepatitis, and hepatitis vaccines have progressed. It is not. Regarding the treatment of hepatocellular carcinoma, the therapeutic agents known to date are epirubicin, cisplatin, 5-fluorouracil, etoposide, etc., but have low therapeutic efficiency, and long-term use of these chemotherapeutic agents may cause chemotherapy resistance. . Therefore, there is an urgent need to develop a liver cancer treatment agent having high sensitivity without side effects.
한편, 네퍼린(neferine)은 연꽃(Nelumbo nucifera Gaerth.) 배아 유래의 주요 bisbenzylisoquinline 알카로이드 중 하나이다. 이러한 녹색 식물 배아 내에 풍부한 알칼로이드는 항-부정맥, 항-고혈압, 이완제, 항-당뇨, 콜린에스테라아제 억제, 진정제 및 항-다중약물내성을 포함하는 다양한 생물적 활성을 갖는 것이 보고되고 있다. 특히, 연꽃 배아는 신경질환, 불면증, 계속적인 발열 및 고혈압과 부정맥과 같은 심혈관질환을 위한 전통적 한약제로 널리 사용되어져 왔다.Neferine, on the other hand, is one of the major bisbenzylisoquinline alkaloids from the lotus ( Nelumbo nucifera Gaerth.) Embryo. Alkaloids abundant in these green plant embryos have been reported to have a variety of biological activities, including anti-arrhythmia, anti-hypertension, relaxants, anti-diabetic, cholinesterase inhibition, sedatives and anti-multi-drug resistance. In particular, the lotus embryo has been widely used as a traditional herbal medicine for neurological diseases, insomnia, continuous fever and cardiovascular diseases such as hypertension and arrhythmia.
그러나 이러한 연꽃 배아 유래의 네퍼린(neferine)이라는 화합물의 간암 예방 또는 치료 효과에 관한 연구된 바 없다.However, there has been no study on the effect of preventing or treating liver cancer of a compound called neferine derived from lotus embryos.
이에 본 발명자들은 연꽃 배아에서 추출·정제한 네퍼린의 약리효과를 연구하던 중, 네퍼린이 바이러스감염에 의해 유발되는 간세포암의 세포주기 진행을 중지시킬 수 있으며, 세포사멸(apoptosis) 유도, 자가포식(autophagy) 유도, 신생혈관형성 억제 및 세포이동 억제 활성을 통해 항암 효과를 가짐에 따라 간암의 예방, 개선 또는 치료에 유용함을 확인함으로써 본 발명을 완성하였다.Therefore, while the present inventors are studying the pharmacological effects of nephrine extracted and purified from lotus embryos, nephrine can stop the cell cycle progression of hepatocellular carcinoma caused by viral infection, induce apoptosis, autophagy. The present invention was completed by confirming that it is useful for preventing, improving or treating liver cancer as it has anticancer effects through autophagy induction, angiogenesis inhibition, and cell migration inhibitory activity.
따라서 본 발명의 목적은 바이러스감염에 의해 유발되는 간암세포에 대해 특이적으로 세포독성이 우수하여 간암 세포 사멸에 의한 간암 예방 또는 치료에 효과적인 약제학적 조성물을 제공하는 것이다.Accordingly, it is an object of the present invention to provide a pharmaceutical composition which is particularly effective in preventing or treating liver cancer by liver cancer cell death due to excellent cytotoxicity against liver cancer cells caused by viral infection.
본 발명의 또 다른 목적은 바이러스감염에 의해 유발되는 간암세포에 대해 특이적으로 세포독성이 우수하여 간암 예방 또는 개선에 효과적인 건강기능식품을 제공하는 것이다.Still another object of the present invention is to provide a health functional food that is effective in preventing or improving liver cancer by having excellent cytotoxicity to liver cancer cells caused by viral infection.
상기와 같은 본 발명의 목적을 달성하기 위해서, 본 발명은 네퍼린(Neferine)을 유효성분으로 포함하는 간암 예방 또는 치료용 약제학적 조성물을 제공한다.In order to achieve the object of the present invention as described above, the present invention provides a pharmaceutical composition for preventing or treating liver cancer comprising Neferine (Neferine) as an active ingredient.
본 발명의 일실시예에 있어서, 상기 네퍼린(Neferine)은 연꽃 배아로부터 유래될 수 있다.In one embodiment of the invention, the neferine (Neferine) may be derived from the lotus embryo.
본 발명의 일실시예에 있어서, 상기 간암은 바이러스감염에 의해 유발되는 간세포암(hepatocellular carcinoma)일 수 있다.In one embodiment of the present invention, the liver cancer may be hepatocellular carcinoma caused by viral infection.
본 발명의 일실시예에 있어서, 상기 약제학적 조성물은 암세포의 세포주기중지(cell cycle arrest), 세포사멸(apoptosis) 유도, 자가포식(autophagy) 유도, 신생혈관형성 억제 및 세포이동 억제 활성을 통해 항암 효과를 가질 수 있다.In one embodiment of the invention, the pharmaceutical composition is through the cell cycle arrest (cell cycle arrest), induction of apoptosis (apoptosis), autophagy induction, angiogenesis inhibition and cell migration inhibitory activity of cancer cells It can have an anticancer effect.
또한, 본 발명은 네퍼린(Neferine)을 유효성분으로 포함하는 간암의 예방 또는 개선용 건강기능식품을 제공한다.The present invention also provides a health functional food for the prevention or improvement of liver cancer comprising Neferine (Neferine) as an active ingredient.
본 발명의 일실시예에 있어서, 상기 네퍼린(Neferine)은 연꽃 배아로부터 유래될 수 있다.In one embodiment of the invention, the neferine (Neferine) may be derived from the lotus embryo.
본 발명의 일실시예에 있어서, 상기 간암은 바이러스감염에 의해 유발되는 간세포암(hepatocellular carcinoma)일 수 있다.In one embodiment of the present invention, the liver cancer may be hepatocellular carcinoma caused by viral infection.
본 발명의 일실시예에 있어서, 상기 식품은 음료류, 육류, 초코렛, 식품류, 과자류, 피자, 라면, 기타 면류, 껌류, 사탕류, 아이스크림류, 알코올 음료류, 비타민 복합제 및 건강보조식품류로 이루어진 군으로부터 선택될 수 있다.In one embodiment of the invention, the food is selected from the group consisting of beverages, meat, chocolate, foods, confectionery, pizza, ramen, other noodles, gums, candy, ice cream, alcoholic beverages, vitamin complexes and health supplements Can be.
본 발명의 네퍼린(Neferine)은 간암세포의 세포주기 진행을 중지시킬 수 있으며, 세포사멸(apoptosis) 유도, 자가포식(autophagy) 유도, 신생혈관형성 억제 및 세포이동 억제 활성을 통해 항암 효과를 가질 수 있다. 특히, 네퍼린은 간암세포 중에서도 바이러스감염에 의해 유발된 간암세포에 특이적으로 세포독성을 가짐에 따라, 바이러스감염에 의해 유발된 간암세포에 최적화된 치료효과를 가진다. Neferine of the present invention can stop the cell cycle progression of liver cancer cells, and have anticancer effects through induction of apoptosis, induction of autophagy, inhibition of angiogenesis and cell migration. Can be. In particular, neferrin has a therapeutic effect optimized for hepatocellular carcinoma induced by viral infection, as it has cytotoxicity specifically to hepatocellular carcinoma cells caused by viral infection among hepatic cancer cells.
도 1은 Hep3B, Sk-Hep1 및 THLE-3 암세포주 각각에 네퍼린을 농도별로 24시간 동안 처리한 후 세포 생존률을 측정하여 그래프로 나타낸 것이다.1 is a graph showing the survival rate of cells after treatment with neprene for each concentration of Hep3B, Sk-Hep1 and THLE-3 cancer cell lines for 24 hours.
도 2는 Hep3B 세포에 네퍼린(20, 25μM)을 6시간 간격으로 처리한 후 DAPI 염색을 통해 세포의 핵 응축 변화를 확인한 것이다.Figure 2 shows the change in nuclear condensation of cells through DAPI staining after treatment with Hep3B cells neferin (20, 25μM) at 6 hour intervals.
도 3은 Hep3B 세포에 네퍼린(15, 20, 25μM)을 24시간 동안 처리한 후 유세포분석기를 이용하여 세포주기를 분석한 것이다.Figure 3 after treatment with Hep3B cells neferin (15, 20, 25μM) for 24 hours is analyzed the cell cycle using a flow cytometer.
도 4는 Hep3B 세포에 네퍼린를 농도별(15, 20, 25μM)로 24시간 동안 처리한 후 세포주기와 관련된 단백질들의 발현량을 웨스턴 블랏을 통해 측정한 것이다.Figure 4 shows the expression level of proteins related to the cell cycle after treatment with Heper3B cells for 24 hours at different concentrations (15, 20, 25μM) by Western blot.
도 5는 Hep3B 세포에 15μM 농도의 네퍼린을 12시간 동안 처리한 후 면역형광분석법을 통해 검출한 p21Waf1/Cip1 단백질 발현 수준을 형광현미경(×1000)을 이용하여 측정한 것이다 (p21Waf1/Cip1: green, nucleic: blue, cytoskeletal actin: red).FIG. 5 shows that p21 Waf1 / Cip1 protein expression levels detected by immunofluorescence analysis after treatment of Hep3B cells with 15 μM concentration of nephrin for 12 hours using a fluorescence microscope (× 1000) (p21 Waf1 / Cip1). : green, nucleic: blue, cytoskeletal actin: red).
도 6A는 Hep3B 세포에 네퍼린를 농도별(15, 20, 25μM)로 24시간 동안 처리한 후 미토콘드리아 막 관련 단백질(Bim, BID, Bax, Bak, Puma) 발현량을 웨스턴 블랏을 통해 측정한 것이다.FIG. 6A shows that hep3B cells were treated with nephrine at different concentrations (15, 20, 25 μM) for 24 hours, and then mitochondrial membrane-related proteins (Bim, BID, Bax, Bak, Puma) were measured by Western blot.
도 6B는 Hep3B 세포에 네퍼린를 농도별(15, 20, 25μM)로 24시간 동안 처리한 후 세포사멸에 수반되는 apoptotic 단백질(cleaved caspase-8, -3, -6, -7 및 PARP) 발현량을 웨스턴 블랏을 통해 측정한 것이다.Figure 6B shows the amount of apoptotic protein (cleaved caspase-8, -3, -6, -7 and PARP) involved in apoptosis after treatment with Hep3B cells for 24 hours at different concentrations (15, 20, 25 μM) Was measured via Western blot.
도 7A는 Hep3B 세포에 15μM 농도의 네퍼린을 12시간 동안 처리한 후 면역형광분석법을 통해 검출한 cleaved caspase-3 단백질 발현 수준을 형광현미경(×1000)을 이용하여 측정한 것이다 (cleaved caspase-3: green, nucleic: blue, cytoskeletal actin: red).FIG. 7A shows the cleaved caspase-3 protein expression level detected by immunofluorescence analysis after treatment with Hep3B cells for 15 hours at 15 μM concentration of neferin (cleaved caspase-3). : green, nucleic: blue, cytoskeletal actin: red).
도 7B는 Hep3B 세포에 15μM 농도의 네퍼린을 12시간 동안 처리한 후 면역형광분석법을 통해 검출한 cleaved caspase-8 단백질 발현 수준을 형광현미경(×1000)을 이용하여 측정한 것이다 (cleaved caspase-8: green, nucleic: blue, cytoskeletal actin: red).FIG. 7B shows the cleaved caspase-8 protein expression level detected by immunofluorescence assay after treatment with Hep3B cells for 15 hours at 15 μM concentration of neferin (cleaved caspase-8). : green, nucleic: blue, cytoskeletal actin: red).
도 8은 Hep3B 세포에 20μM 농도의 네퍼린를 처리한 후 시간 경과에 따라 소포체 스트레스 관련 샤페론 단백질(Bip, calnexin, PDI, calpain2 및 caspase-12)의 발현량을 웨스턴 블랏을 통해 측정한 것이다.8 is a Western blot expression of vesicle stress-related chaperon proteins (Bip, calnexin, PDI, calpain2 and caspase-12) over time after treatment with 20 μM concentration of neprine in Hep3B cells.
도 9는 Hep3B 세포에 15μM 농도의 네퍼린을 12시간 동안 처리한 후 면역형광분석법을 통해 검출한 caspase-12 단백질 발현 수준을 형광현미경(×1000)을 이용하여 측정한 것이다 (caspase-12: green, nucleic: blue, cytoskeletal actin: red).FIG. 9 is a caspase-12 protein expression level detected by immunofluorescence analysis after treatment with Hep3B cells for 15 hours at 15 μM concentration of nephrin using a fluorescent microscope (× 1000) (caspase-12: green , nucleic: blue, cytoskeletal actin: red).
도 10은 GFP-LC3B 플라스미드로 형질전환된 Hep3B 세포에 20μM 농도의 네퍼린를 처리한 후, 시간 경과에 따라 세포질 내 자가포식소체(autophagosomes) 형성 정도를 형광현미경(×1000)을 이용하여 측정한 것이다.FIG. 10 shows the degree of autophagosomes formation in the cytoplasm with fluorescence microscopy (× 1000) after treatment with 20 μM of nephrine in Hep3B cells transformed with GFP-LC3B plasmid. .
도 11은 Hep3B 세포에 네퍼린를 농도별(15, 20, 25μM)로 12시간 처리한 후 LC3B 단백질의 발현량을 웨스턴 블랏을 통해 측정한 것이다.FIG. 11 shows the expression of LC3B protein after Western blot treatment of Hep3B cells with nephrine at different concentrations (15, 20, 25 μM) for 12 hours.
도 12A는 HUVECs 세포에 네퍼린을 농도별(10, 20, 30μM)로 18시간 처리한 후 튜브 형성 정도를 도립현미경(×40)을 이용하여 측정한 것이며, 도 12B는 상기 결과를 토대로 총 튜브 길이를 Wimasis Image Aalysis 소프트웨어를 이용하여 퍼센트로 변환한 그래프이다.FIG. 12A is a measurement of the degree of tube formation using an inverted microscope (× 40) after 18 hours of treatment with concentrations (10, 20, 30 μM) of HUVECs cells, and FIG. 12B shows the total tube based on the results. This is a graph of lengths converted into percentages using Wimasis Image Aalysis software.
도 13A는 Hep3B 세포에 네퍼린를 15μM로 처리한 후 시간 경과에 따른 세포 이동 정도를 도립현미경(×40)을 이용하여 측정한 것이며, 도 13B는 상기 결과를 토대로 세포 이동을 Wimasis Image Aalysis 소프트웨어를 이용하여 cell-covered area퍼센트로 변환한 그래프이다.FIG. 13A is a measure of cell migration over time after treatment with Hep3B cells at 15 μM using an inverted microscope (× 40), and FIG. 13B is based on the results of cell migration using Wimasis Image Aalysis software. This graph is then converted to cell-covered area percent.
본 발명은 하기 화학식 1로 표시되는 네퍼린(Neferine)을 유효성분으로 포함하는 간암 예방 또는 치료용 약제학적 조성물을 제공함에 그 특징이 있다.The present invention is characterized by providing a pharmaceutical composition for preventing or treating liver cancer comprising Neferine (Neferine) represented by the following formula (1) as an active ingredient.
<화학식 1><Formula 1>
Figure PCTKR2013008639-appb-I000001
Figure PCTKR2013008639-appb-I000001
본 발명의‘네퍼린(neferine)’은 연꽃(Nelumbo nucifera Gaerth.) 배아 유래의 주요 bisbenzylisoquinline 알카로이드 중 하나이다. 네퍼린은 4-[[(1R)-6,7-Dimethoxy-2-methyl-3,4-dihydro-1H-isoquinolin-1-yl]methyl]-2-[[(1R)-6-methoxy-1-[(4-methoxyphenyl)methyl]-2-methyl-3,4-dihydro-1H-isoquinolin-7-yl]oxy]phenol로도 불리며, 이 물질의 분자식은 C38H44N2O6이고 분자량은 624.77이다.The 'neferine' of the present invention is one of the main bisbenzylisoquinline alkaloids from the lotus (Nelumbo nucifera Gaerth.) Embryo. Nefferin is 4-[[(1R) -6,7-Dimethoxy-2-methyl-3,4-dihydro-1H-isoquinolin-1-yl] methyl] -2-[[(1R) -6-methoxy- Also called 1-[(4-methoxyphenyl) methyl] -2-methyl-3,4-dihydro-1H-isoquinolin-7-yl] oxy] phenol, the molecular formula of this substance is C 38 H 44 N 2 O 6 and has a molecular weight Is 624.77.
녹색 식물 배아 내에 함유된 알칼로이드는 항-부정맥, 항-고혈압, 이완제, 항-당뇨, 콜린에스테라아제 억제, 진정제 및 항-다중약물내성을 포함하는 다양한 생물적 활성을 갖는 것이 보고되고 있다. 특히, 연꽃 배아는 신경질환, 불면증, 계속적인 발열 및 고혈압과 부정맥과 같은 심혈관질환을 위한 전통적 한약제로 널리 사용되어져 왔다.Alkaloids contained in green plant embryos have been reported to have a variety of biological activities including anti-arrhythmia, anti-hypertension, relaxants, anti-diabetic, cholinesterase inhibition, sedatives and anti-multi-drug resistance. In particular, the lotus embryo has been widely used as a traditional herbal medicine for neurological diseases, insomnia, continuous fever and cardiovascular diseases such as hypertension and arrhythmia.
그러나 현재까지 네퍼린(neferine)이라는 화합물의 간암 예방 또는 치료 효과에 관해서는 연구된 바 없다.However, there has been no study on the effect of preventing or treating liver cancer of the compound called neferine.
본 발명자들은 네퍼린이 바이러스감염에 의해 유발되는 간세포암의 세포주기 진행을 중지시킬 수 있으며, 세포사멸(apoptosis) 유도, 자가포식(autophagy) 유도, 신생혈관형성 억제 및 세포이동 억제 활성을 통해 항암 효과를 가짐에 따라 간암의 예방, 개선 또는 치료에 유용한 물질로서 약리학적 조성물과 같은 의약품은 물론이고, 건강기능식품 등으로 활용할 수 있다는 사실을 최초로 규명하였다.The present inventors can stop the cell cycle progression of hepatocellular carcinoma caused by nephrine-induced viral infection, and anti-cancer effect through induction of apoptosis, induction of autophagy, inhibition of angiogenesis and cell migration. As a substance useful for the prevention, improvement or treatment of liver cancer, as well as pharmaceuticals such as pharmacological composition, as well as health functional food, it was first identified the first.
본 발명의 하기 실험예 1에서는, 네퍼린의 암세포주에 대한 세포 독성을 확인하기 위해 Hep3B, Sk-Hep1 및 THLE-3 세포주를 이용하여 세포 생존율을 측정한 결과, Hep3B 세포주에서만 네퍼린 처리 농도에 의존적으로 세포 생존율이 두드러지게 감소되는 것으로 나타났다. 이러한 결과를 통해 네퍼린은 Hep3B 세포에 특이적인 세포독성 민감도를 가지는 것을 확인할 수 있었다(도 1 참조).In Experimental Example 1 of the present invention, in order to confirm the cytotoxicity of the nephrine against cancer cell lines, the cell viability was measured using Hep3B, Sk-Hep1 and THLE-3 cell lines, and only the Hep3B cell line was treated with the neferrin treatment concentration. Independently, cell viability has been shown to be markedly reduced. These results confirm that neferrin has a specific cytotoxic sensitivity to Hep3B cells (see Fig. 1).
또한 본 발명의 하기 실험예 2에서는, 네퍼린의 간암세포주인 Hep3B에 대한 세포사멸(apoptosis)를 유도하는지를 살펴보기 위하여, DAPI 에세이를 실시한 결과, 네퍼린를 처리하지 않은 무처리군과 비교하여 네퍼린을 처리한 후 6시간이 경과한 실험군에서 세포들의 응축(shrinkage) 및 파열(blebbing)과 같은 핵 막 변화가 나타나는 것을 확인할 수 있었다. 특히 네퍼린 처리 후 24시간이 경과한 실험군에서 사멸세포(apoptotic cells)의 전형적인 특징인 핵 응축(apoptosome formation)이 관찰되었다(도 2 참조).In addition, in Experimental Example 2 of the present invention, in order to examine whether apoptosis of hep3B, a hepatocellular carcinoma cell line of neferrin, was performed, a DAPI assay was performed. 6 hours after the treatment, it was confirmed that nuclear membrane changes such as condensation and blebbing of cells appeared in the experimental group. In particular, apoptosome formation, which is typical of apoptotic cells, was observed in the experimental group 24 hours after neferrin treatment (see FIG. 2).
또한 본 발명의 하기 실험예 3에서는, 네퍼린의 간암세포주인 Hep3B에 대한 세포주기 중지(cell cycle arrest)를 유도하는지를 살펴본 결과, 네퍼린 처리 농도(15, 20, 25μM)에 비례하여 sub-G1 세포 집단수가 증대되었으며(도 3 참조); 세포주기 진행의 중지를 나타내는 증거로써 세포주기와 관련된 단백질들의 발현이 조절되어지는 것이 관찰되었으며(도 4 참조); 네퍼린을 처리한 Hep3B 세포들의 세포질에서 핵 주변의 p21 Waf1/Cip1 단백질(세포주기를 억제시킬 수 있는 조절자) 발현 수준이 두드러지게 증대되는 것을 확인할 수 있었다(도 5 참조).In addition, in Experimental Example 3 of the present invention, as a result of examining the cell cycle arrest of Hep3B, a hepatocellular carcinoma cell line of neferrin, sub-G1 was proportional to the neferrin treatment concentration (15, 20, 25 μM). Cell population increased (see FIG. 3); It was observed that the expression of proteins associated with the cell cycle is regulated as evidence of the suspension of cell cycle progression (see FIG. 4); In the cytoplasm of neprene-treated Hep3B cells, it was confirmed that the expression level of p21 Waf1 / Cip1 protein (regulator capable of inhibiting the cell cycle) of the nucleus was significantly increased (see FIG. 5).
일반적으로 세포주기는 세포내에서 정해진 메카니즘에 의해 일정순서 대로 이루어지고 있다. 그러나 이렇게 정해진 순서에 이상이 초래되면 세포주기의 유지는 어려워지게 되며, 세포주기의 이상이 초래되었을 때 이를 복구하는 역할을 하는 조절인자가 작용을 하게 되는데, 이러한 인자로서 사이클린(cyclin), 사이클린-의존성 키나제(cyclin-dependent kinase: 이하 간략하게 ‘CDK'라 표기함) 및 사이클린-의존성 키나제 억제제(cyclin-dependent kinase inhibitor: 이하 간략하게 ‘CDKI'라 표기함)가 있다. In general, the cell cycle is performed in a certain order by a mechanism defined in the cell. However, if the abnormality occurs in this order, the maintenance of the cell cycle becomes difficult, and when the abnormality of the cell cycle is induced, a regulator that acts to restore it acts, such as cyclin (cyclin), cyclin- Cyclin-dependent kinase (hereinafter abbreviated 'CDK') and cyclin-dependent kinase inhibitor (hereinafter abbreviated 'CDKI').
세포주기 과정에서, G1기(G1 phase) 초기에는 세포의 종류에 따라서 CDK4, 6, 8등이 활성화되어 작용하고, G1기 후기와 S기 초기에서는 CDK2가 작용하며, G2에서 M으로의 진행에는 CDK1이 중요한 역할을 하는 것으로 알려져 있다. CDK의 활성에는 사이클린과의 결합이 필수적인데 CDK4, 6, 8은 사이클린 D와의 결합에 의하여 활성화되며, CDK2는 사이클린 A 및 E와 결합한다. CDK1은 사이클린 B 및 A와 결합하며, 이외에 사이클린 G, F 등이 알려져 있다. 이러한 세포주기의 각 시기에서 특이적인 cyclin-CDK 복합체가 각각 활성화되고 CDK에 특이적으로 인산화되는 단백질들이 세포주기의 진행을 담당하게 되므로 세포주기를 CDK 주기로 명명할 수도 있다.In the cell cycle, CDK4, 6, 8, etc. are activated by the type of cells in the early stage of G1 phase, CDK2 in the late stage of G1 and early stage of S1, and the progression from G2 to M CDK1 is known to play an important role. Binding to cyclins is essential for the activity of CDK. CDK4, 6 and 8 are activated by binding to cyclin D, and CDK2 binds to cyclins A and E. CDK1 binds to cyclins B and A, in addition to cyclins G, F and the like. At each period of the cell cycle, the specific cyclin-CDK complex is activated and the proteins phosphorylated specifically for CDK are responsible for the progression of the cell cycle.
또한, CDK는 사이클린의 활성에 필수적인 인자로 작용하는데, 활성화된 CDK-사이클린은 사이클린의 조절단위와 CDK의 활성단위로 구분되어 있으며, 사이클린의 CDK 조절방법은 두 가지로 볼 수 있는데, 첫 번째가 사이클린과 CDK가 결합하여 단백질의 구조변화를 유도하여 ATP phosphate group의 배치가 기질 단백질에 전달하기 쉽게 변한다. 또한, 단백질의 기질이 CDK에 접근할 수 없도록 막고 있는 T loop의 위치가 변하여 기질의 접근이 용이해진다. CDK의 활성이 세포주기의 특정 시기에만 활성화되는 것은 세포주기 특이적으로 일어나는 사이클린 합성 때문이다. 또한, 사이클린 D는 주로 G1 중기에 합성이 최고조에 달하며 주로 세포성장인자 등의 마이토젠(mitogen)에 의하여 유도된다. 사이클린 D는 세 종류의 서브타입(D1, 2, 3)이 있는데, 세포의 종류에 따라서 발현되는 정도가 다르다. 예컨대, 사이클린 D의 합성을 저해하면 세포주기 G1이 정지되고, 사이클린 D를 과발현하면 G1기가 짧아지고 마이토젠 없이도 세포주기가 시작된다.In addition, CDK acts as an essential factor for cyclin activity. Activated CDK-cyclin is divided into cyclin regulatory unit and CDK active unit, and cyclin CDK regulation can be seen in two ways. The combination of cyclin and CDK induces structural changes in the protein, making the arrangement of the ATP phosphate group easy to transfer to the substrate protein. In addition, the position of the T loop that prevents the protein substrate from accessing the CDK is changed, thereby facilitating access to the substrate. The activity of CDK is activated only at certain times of the cell cycle because of cyclin synthesis that occurs specifically in the cell cycle. In addition, cyclin D is most likely synthesized in the middle of G1, and is mainly induced by mitogens such as cell growth factors. Cyclin D has three types of subtypes (D1, 2, and 3), and the degree of expression varies depending on the type of cell. For example, inhibiting the synthesis of cyclin D stops the cell cycle G1, and overexpressing cyclin D shortens the G1 group and starts the cell cycle without mitogen.
한편 p15INK4B, p16INK4A, p18INK4C, p19INK4D, p27Kip1 및 p21WAF1/Cip1과 같은 세포주기 조절자들은 사이클린 D1/CDK4,6 또는 사이클린 E/CDK2 복합체를 억제하는 핵심 조절자로 알려져 있다. 따라서 상기와 같은 세포주기 조절자들을 negative cell cycle regulator라고 하며, 이들이 발현되는 경우 세포주기 진행이 억제될 수 있다.The p15 INK4B, p16 INK4A, p18 INK4C , p19 INK4D, p27 Kip1 and p21 WAF1 / Cip1 and cell cycle regulation such as characters are known as core control to inhibit cyclin D1 / CDK4,6 or Cyclin E / CDK2 complexes. Therefore, such cell cycle regulators are called negative cell cycle regulators, and when they are expressed, cell cycle progression can be inhibited.
본 발명의 하기 실험예 4에서는, 네퍼린의 간암세포주인 Hep3B에 대한 세포사멸(apoptosis) 유도를 살펴본 결과, 미토콘드리아-매개된 세포사멸 경로를 조절하는 것으로 알려진 Bcl-2 패밀리 단백질(Bim, Bid, Bax, Bak 및 Puma) 발현이 증대되는 것을 확인할 수 있었으며, 소포체 스트레스에 따른 세포사멸에 수반되는 apoptotic 단백질(cleaved caspase-8, -3, -6, -7 및 PARP)들의 변화가 관찰되었다(도 6 참조). 뿐만 아니라, 네퍼린을 처리하지 않은 무처리군과 비교하여 네퍼린을 처리한 Hep3B 세포에서 cleaved caspase-8, 및 -3이 두드러지게 발현되는 것으로 나타나, 네퍼린에 의해 유도된 apoptosomes이 세포질에서 caspase-3 및 caspase-8을 활성화시켜 세포사멸을 야기하는 것이라 유추할 수 있었다(도 7 참조).In Experimental Example 4 of the present invention, as a result of examining the induction of apoptosis of Hep3B, a hepatocellular carcinoma cell line of nephrine, Bcl-2 family proteins (Bim, Bid, known to regulate the mitochondrial-mediated apoptosis pathway) It was confirmed that the expression of Bax, Bak and Puma was increased, and changes of apoptotic proteins (cleaved caspase-8, -3, -6, -7 and PARP) accompanying apoptosis due to vesicle stress were observed (FIG. 6). In addition, cleaved caspase-8 and -3 were found to be prominently expressed in neprine-treated Hep3B cells compared to the untreated group, which was not treated with neferin, and apoptosomes induced by neferin were caspase in the cytoplasm. It could be deduced that activating -3 and caspase-8 causes apoptosis (see FIG. 7).
또한 본 발명의 하기 실험예 5에서는, 네퍼린의 간암세포에 대한 항암 효과로서 자가포식을 유도할 수 있는지를 확인한 결과, 네퍼린을 처리한 실험군에서 시간 경과에 따라 자가포식의 형태학적 특징 중 하나인 세포질에서의 소체(vacuole) 형성이 발생되었다. 또한 자가포식은 자가포식소체(autophagosomes)라 불리는 이중막으로 둘러싸인 소포를 형성하는데, 네퍼린을 처리한 실험군에서 시간 경과에 따라 강력한 형광 스팟(GFP-LC3B "dots")이 나타남으로써 자가포식소체 수가 세포질 내에서 증가함을 확인할 수 있었다(도 10 참조).In addition, in Experimental Example 5 of the present invention, as a result of confirming that nephrine can induce autophagy as an anticancer effect on liver cancer cells, one of the morphological characteristics of autophagy over time in the experimental group treated with neferrin Vacuole formation occurred in the phosphorus cytoplasm. Autophagy also forms vesicles enclosed in a double membrane called autophagosomes, which show strong fluorescence spots (GFP-LC3B "dots") over time in the nephrine treated group. It was confirmed that the increase in the cytoplasm (see Figure 10).
또한 본 발명의 하기 실험예 6에서는, 네퍼린의 HUVECs 세포에서 신생혈관형성 억제 효과를 실험한 결과, 네퍼린 처리 농도에 의존하여 HUVECs의 튜브 길이(px)가 감소되는 것을 확인할 수 있었다.In addition, in Experimental Example 6 of the present invention, as a result of testing the neovascularization inhibitory effect of neferrin in HUVECs cells, it was confirmed that the tube length (px) of HUVECs is reduced depending on the concentration of neferrin treatment.
또한 본 발명의 하기 실험예 7에서는, 네퍼린의 간암세포에 대한 항암 효과로서 세포이동 억제를 유도할 수 있는지를 확인한 결과, 네퍼린 처리 농도에 의존하여 세포이동이 억제되는 것을 확인할 수 있었다(도 13 참조).In addition, in Experimental Example 7 of the present invention, it was confirmed that the cell migration is suppressed depending on the neferoline treatment concentration as a result of confirming that nephrine can induce cell migration inhibition as an anticancer effect on liver cancer cells (FIG. 13).
이와 같은 결과를 통해, 본 발명자들은 네퍼린이 간암세포 중에서도 바이러스감염에 의해 유발된 간암세포에 특이적으로 세포독성을 가지며, 간암세포의 세포주기중지, 세포사멸(apoptosis) 유도, 자가포식(autophagy) 유도, 신생혈관형성 억제 및 세포이동 억제 활성을 통해 항암 효과를 가진다는 사실을 실험적으로 입증하였다.Through these results, the present inventors have found that nepherin has cytotoxicity to liver cancer cells induced by virus infection among hepatic cancer cells, cell cycle arrest, induction of apoptosis and autophagy of liver cancer cells. It has been experimentally demonstrated to have anticancer effects through induction, angiogenesis inhibition and cell migration inhibitory activity.
그러므로 네퍼린을 유효성분으로 포함하는 본 발명의 조성물은 간암을 효과적으로 예방 또는 치료할 수 있으며, 특히 바이러스감염에 의해 유발된 간암세포에 특이적으로 세포독성을 가짐에 따라, 바이러스감염에 의해 유발된 간암세포에 최적화된 치료효과를 가진다.Therefore, the composition of the present invention comprising nephrine as an active ingredient can effectively prevent or treat liver cancer, and in particular, as it has cytotoxicity specifically to liver cancer cells induced by viral infection, liver cancer caused by viral infection It has a therapeutic effect optimized for cells.
본 발명에 따른 상기 네퍼린은 염, 바람직하게는 약학적으로 허용 가능한 염의 형태로 사용될 수 있다. 상기 염으로는 약학적으로 허용 가능한 유리산(free acid)에 의하여 형성된 산 부가염이 바람직하며, 상기 유리산으로는 유기산과 무기산을 사용할 수 있다. 상기 유기산은 이에 제한되는 것은 아니나, 구연산, 초산, 젖산, 주석산, 말레인산, 푸마르산, 포름산, 프로피온산, 옥살산, 트리플로오로아세트산, 벤조산, 글루콘산, 메타술폰산, 글리콜산, 숙신산, 4-톨루엔술폰산, 글루탐산 및 아스파르트산을 포함한다. 또한 상기 무기산은 이에 제한되는 것은 아니나, 염산, 브롬산, 황산 및 인산을 포함한다.The neferrin according to the present invention can be used in the form of salts, preferably pharmaceutically acceptable salts. The salt is preferably an acid addition salt formed by a pharmaceutically acceptable free acid, and an organic acid and an inorganic acid may be used as the free acid. The organic acid is not limited thereto, citric acid, acetic acid, lactic acid, tartaric acid, maleic acid, fumaric acid, formic acid, propionic acid, oxalic acid, trifluoroacetic acid, benzoic acid, gluconic acid, metasulfonic acid, glycolic acid, succinic acid, 4-toluenesulfonic acid, Glutamic acid and aspartic acid. In addition, the inorganic acid includes, but is not limited to, hydrochloric acid, bromic acid, sulfuric acid and phosphoric acid.
본 발명에 따른 네퍼린은 시중에서 판매되는 것을 사용할 수도 있으며, 또는 천연으로부터 분리되거나 당업계에 공지된 화학적 합성법으로 제조된 것을 사용할 수 있다.The neferrin according to the present invention may be commercially available, or may be one that is separated from nature or manufactured by chemical synthesis known in the art.
본 발명의 네퍼린을 천연물로부터 분리하고 하는 경우, 분리방법에는 제한이 없으며, 공지되어 있는 어떠한 방법도 이용될 수 있다. 예를 들어, 본 발명의 하기 실시예에서는 N. nucifera의 배아를 메탄올 환류 추출한 후 진공 건조를 통해 농축하여 연꽃 배아 메탄올 추출물을 수득하였으며, 이렇게 수득한 추출물을 증류수에 현탁시킨 후 디클로로메탄, 에틸아세테이트 및 n-부탄올로 순차적으로 분획한 다음, 상기 디클로로메탄 분획물을 다시 실리카겔 컬럼 크로마토그래피를 수행하여 최종적인 네퍼린을 정제하였다.When separating the neferrin of the present invention from natural products, there is no limitation to the separation method, any known method can be used. For example, in the following examples of the present invention, the N. nucifera embryo was extracted under reflux with methanol, and concentrated by vacuum drying to obtain a lotus embryo methanol extract. The extract thus obtained was suspended in distilled water, followed by dichloromethane and ethyl acetate. And sequentially fractionated with n-butanol, and the dichloromethane fraction was subjected to silica gel column chromatography again to purify the final neferrin.
본 발명의 조성물은 상기 네퍼린을 유효성분으로 포함하는 약제학적 조성물로서 이러한 유효성분 이외에 약제학적으로 적합하고 생리학적으로 허용되는 보조제를 사용하여 제조될 수 있으며, 상기 보조제로는 부형제, 붕해제, 감미제, 결합제, 피복제, 팽창제, 윤활제, 활택제 또는 향미제 등을 사용할 수 있다.The composition of the present invention may be prepared using a pharmaceutically acceptable and physiologically acceptable adjuvant in addition to such an active ingredient as a pharmaceutical composition comprising the neferin as an active ingredient, the adjuvant, an disintegrant, Sweetening agents, binders, coatings, expanding agents, lubricants, lubricants, or flavoring agents may be used.
상기 약제학적 조성물은 투여를 위해서 상기 기재한 유효성분 이외에 추가로 약제학적으로 허용 가능한 담체를 1종 이상 포함하여 약제학적 조성물로 바람직하게 제제화할 수 있다.The pharmaceutical composition may be preferably formulated into a pharmaceutical composition including one or more pharmaceutically acceptable carriers in addition to the above-described active ingredient for administration.
상기 약제학적 조성물의 제제 형태는 과립제, 산제, 정제, 피복정, 캡슐제, 좌제, 액제, 시럽, 즙, 현탁제, 유제, 점적제 또는 주사 가능한 액제 등이 될 수 있다. 예를 들어, 정제 또는 캡슐제의 형태로의 제제화를 위해, 유효 성분은 에탄올, 글리세롤, 물 등과 같은 경구, 무독성의 약제학적으로 허용 가능한 불활성 담체와 결합될 수 있다. 또한, 원하거나 필요한 경우, 적합한 결합제, 윤활제, 붕해제 및 발색제 또한 혼합물로 포함될 수 있다. 적합한 결합제는 이에 제한되는 것은 아니나, 녹말, 젤라틴, 글루코스 또는 베타-락토오스와 같은 천연 당, 옥수수 감미제, 아카시아, 트래커캔스 또는 소듐올레이트와 같은 천연 및 합성 검, 소듐 스테아레이트, 마그네슘 스테아레이트, 소듐 벤조에이트, 소듐 아세테이트, 소듐 클로라이드 등을 포함한다. 붕해제는 이에 제한되는 것은 아니나, 녹말, 메틸 셀룰로스, 아가, 벤토니트, 잔탄 검 등을 포함한다. 액상 용액으로 제제화되는 조성물에 있어서 허용 가능한 약제학적 담체로는, 멸균 및 생체에 적합한 것으로서, 식염수, 멸균수, 링거액, 완충 식염수, 알부민 주사용액, 덱스트로즈 용액, 말토 덱스트린 용액, 글리세롤, 에탄올 및 이들 성분 중 1 성분 이상을 혼합하여 사용할 수 있으며, 필요에 따라 항산화제, 완충액, 정균제 등 다른 통상의 첨가제를 첨가할 수 있다. 또한 희석제, 분산제, 계면활성제, 결합제 및 윤활제를 부가적으로 첨가하여 수용액, 현탁액, 유탁액 등과 같은 주사용 제형, 환약, 캡슐, 과립 또는 정제로 제제화할 수 있다. 더 나아가 해당분야의 적절한 방법으로 Remington's Pharmaceutical Science, Mack Publishing Company, Easton PA에 개시되어 있는 방법을 이용하여 각 질환에 따라 또는 성분에 따라 바람직하게 제제화 할 수 있다.Formulation forms of the pharmaceutical composition may be granules, powders, tablets, coated tablets, capsules, suppositories, solutions, syrups, juices, suspensions, emulsions, drops or injectable solutions. For example, for formulation in the form of tablets or capsules, the active ingredient may be combined with an oral, nontoxic, pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like. In addition, if desired or necessary, suitable binders, lubricants, disintegrants and coloring agents may also be included in the mixture. Suitable binders include but are not limited to natural and synthetic gums such as starch, gelatin, glucose or beta-lactose, corn sweeteners, acacia, trackercance or sodium oleate, sodium stearate, magnesium stearate, sodium Benzoate, sodium acetate, sodium chloride and the like. Disintegrants include, but are not limited to, starch, methyl cellulose, agar, bentonite, xanthan gum, and the like. Acceptable pharmaceutical carriers in compositions formulated in liquid solutions are sterile and physiologically compatible, including saline, sterile water, Ringer's solution, buffered saline, albumin injectable solutions, dextrose solution, maltodextrin solution, glycerol, ethanol and One or more of these components may be mixed and used, and other conventional additives such as antioxidants, buffers and bacteriostatic agents may be added as necessary. Diluents, dispersants, surfactants, binders and lubricants may also be added in addition to formulate into injectable formulations, pills, capsules, granules or tablets such as aqueous solutions, suspensions, emulsions and the like. Furthermore, the method disclosed in Remington's Pharmaceutical Science, Mack Publishing Company, Easton PA can be formulated according to each disease or component, as appropriate in the art.
본 발명의 일실시예에 있어서, 본 발명의 네퍼린은 조성물에 1 내지 300μM의 농도로 포함될 수 있으며, 또한 본 발명의 네퍼린 화합물은 조성물 총 중량에 대하여 0.1 ~ 95중량%로 포함될 수 있다. In one embodiment of the present invention, the neferrin of the present invention may be included in the composition in a concentration of 1 to 300μM, and the neferrin compound of the present invention may be included in 0.1 to 95% by weight relative to the total weight of the composition.
또한, 본 발명은 간암 예방 또는 치료용 의약의 제조를 위한 네퍼린(neferine)을 유효성분으로 포함하는 조성물의 용도를 제공한다. 상기한 네퍼린을 유효성분으로 포함하는 본 발명의 조성물은 간암의 예방 또는 치료용 의약의 제조를 위한 용도로 이용될 수 있다.The present invention also provides the use of a composition comprising neferine (neferine) as an active ingredient for the manufacture of a medicament for preventing or treating liver cancer. The composition of the present invention comprising the neferrin as an active ingredient can be used for the manufacture of a medicament for the prevention or treatment of liver cancer.
또한, 본 발명은 포유동물에게 네퍼린(neferine)을 투여하는 것을 포함하는 간암의 예방 또는 치료방법을 제공한다.The present invention also provides a method for preventing or treating liver cancer comprising administering neferine to a mammal.
여기에서 사용된 용어 "포유동물"은 치료, 관찰 또는 실험의 대상인 포유동물을 말하며, 바람직하게는 인간을 말한다.As used herein, the term "mammal" refers to a mammal that is the subject of treatment, observation or experimentation, preferably human.
여기에서 사용된 용어 "치료상 유효량"은 연구자, 수의사, 의사 또는 기타 임상에 의해 생각되는 조직계, 동물 또는 인간에서 생물학적 또는 의학적 반응을 유도하는 유효 성분 또는 약제학적 조성물의 양을 의미하는 것으로, 이는 치료되는 질환 또는 장애의 증상의 완화를 유도하는 양을 포함한다. 본 발명의 유효 성분에 대한 치료상 유효 투여량 및 투여횟수는 원하는 효과에 따라 변화될 것임은 당업자에게 자명하다. 그러므로 투여될 최적의 투여량은 당업자에 의해 쉽게 결정될 수 있으며, 질환의 종류, 질환의 중증도, 조성물에 함유된 유효성분 및 다른 성분의 함량, 제형의 종류, 및 환자의 연령, 체중, 일반 건강 상태, 성별 및 식이, 투여 시간, 투여 경로 및 조성물의 분비율, 치료기간, 동시 사용되는 약물을 비롯한 다양한 인자에 따라 조절될 수 있다. 본 발명의 치료방법에 있어서, 성인의 경우, 본 발명의 네퍼린(neferine)을 1일 1회 내지 수회 투여시, 0.01㎎/kg~250㎎/kg의 용량으로 투여하는 것이 바람직하다.As used herein, the term “therapeutically effective amount” means an amount of an active ingredient or pharmaceutical composition that induces a biological or medical response in a tissue system, animal or human, as thought by a researcher, veterinarian, doctor or other clinician, which Amounts that induce alleviation of the symptoms of the disease or disorder being treated. It will be apparent to those skilled in the art that the therapeutically effective dosages and frequency of administrations for the active ingredients of the present invention will vary depending on the desired effect. Therefore, the optimal dosage to be administered can be easily determined by one skilled in the art and includes the type of disease, the severity of the disease, the amount of active ingredients and other ingredients contained in the composition, the type of formulation, and the age, weight, general health of the patient. , Sex and diet, time of administration, route of administration and rate of composition, duration of treatment, and drugs used concurrently. In the treatment method of the present invention, in the case of an adult, the neferine of the present invention is preferably administered at a dose of 0.01 mg / kg to 250 mg / kg once or several times a day.
본 발명의 치료방법에서 본 발명의 네퍼린(neferine)을 유효성분으로 포함하는 조성물은 경구, 직장, 정맥내, 동맥내, 복강내, 근육내, 흉골내, 경피, 국소, 안구내 또는 피내 경로를 통해 통상적인 방식으로 투여할 수 있다.In the treatment method of the present invention, the composition comprising the neferine of the present invention as an active ingredient is an oral, rectal, intravenous, intraarterial, intraperitoneal, intramuscular, intrasternal, transdermal, topical, intraocular or intradermal route. Can be administered in a conventional manner.
또한, 본 발명은 네퍼린(neferine)을 유효성분으로 포함하는 간암의 예방 또는 개선용 건강기능식품을 제공한다.The present invention also provides a health functional food for preventing or improving liver cancer comprising neferine as an active ingredient.
본 발명의 건강기능식품은 간암의 예방 및 개선을 목적으로, 정제, 캅셀, 분말, 과립, 액상, 환 등의 형태로 제조 및 가공할 수 있다.The health functional food of the present invention can be prepared and processed in the form of tablets, capsules, powders, granules, liquids, pills and the like for the purpose of preventing and improving liver cancer.
본 발명에서 “건강기능식품”이라 함은 건강기능식품에 관한 법률 제6727호에 따른 인체에 유용한 기능성을 가진 원료나 성분을 사용하여 제조 및 가공한 식품을 말하며, 인체의 구조 및 기능에 대하여 영양소를 조절하거나 생리학적 작용 등과 같은 보건 용도에 유용한 효과를 얻을 목적으로 섭취하는 것을 의미한다.In the present invention, "health functional food" refers to a food manufactured and processed using raw materials or ingredients having functional properties useful for the human body according to the Health Functional Food Act No. 6767, and nutrients for the structure and function of the human body. It is meant to be consumed for the purpose of regulating or obtaining a useful effect for health use such as physiological action.
본 발명의 건강기능식품은 통상의 식품 첨가물을 포함할 수 있으며, 식품 첨가물로서의 적합 여부는 다른 규정이 없는 한, 식품의약품안전청에 승인된 식품 첨가물 공전의 총칙 및 일반시험법 등에 따라 해당 품목에 관한 규격 및 기준에 의하여 판정한다.The health functional food of the present invention may include a conventional food additive, and the suitability as a food additive, unless otherwise specified, in accordance with the General Regulations of the Food Additives and General Test Methods approved by the Food and Drug Administration, etc. Judging by the standards and standards.
상기 “식품 첨가물 공전”에 수재된 품목으로는 예를 들어, 케톤류, 글리신, 구연산칼슘, 니코틴산, 계피산 등의 화학적 합성물; 감색소, 감초추출물, 결정셀룰로오스, 고량색소, 구아검 등의 천연첨가물; L-글루타민산나트륨제제, 면류첨가알칼리제, 보존료제제, 타르색소제제 등의 혼합제제류 등을 들 수 있다.Examples of the items listed in the "Food Additive Reduction" include chemical compounds such as ketones, glycine, calcium citrate, nicotinic acid, and cinnamic acid; Natural additives such as dark blue pigment, licorice extract, crystalline cellulose, high color pigment and guar gum; And mixed preparations such as sodium L-glutamate, algae additives, preservatives and tar dyes.
예를 들어, 정제 형태의 건강기능식품은 본 발명의 유효성분인 네퍼린(neferine)을 부형제, 결합제, 붕해제 및 다른 첨가제와 혼합한 혼합물을 통상의 방법으로 과립화한 다음, 활택제 등을 넣어 압축성형하거나, 상기 혼합물을 직접 압축 성형할 수 있다. 또한 상기 정제 형태의 건강기능식품은 필요에 따라 교미제 등을 함유할 수도 있다.For example, the health functional food in the form of tablets is a mixture of neferine, an active ingredient of the present invention, with an excipient, a binder, a disintegrant, and other additives, followed by granulation in a conventional manner, and then a lubricant and the like. Compression molding, or the mixture may be directly compression molded. In addition, the health functional food in the form of tablets may contain a mating agent or the like as necessary.
캅셀 형태의 건강기능식품 중 경질 캅셀제는 통상의 경질 캅셀에 본 발명의 유효성분인 네퍼린(neferine)을 부형제 등의 첨가제와 혼합한 혼합물을 충진하여 제조할 수 있으며, 연질 캅셀제는 네퍼린(neferine)을 부형제 등의 첨가제와 혼합한 혼합물을 젤라틴과 같은 캅셀기제에 충진하여 제조할 수 있다. 상기 연질 캅셀제는 필요에 따라 글리세린 또는 소르비톨 등의 가소제, 착색제, 보존제 등을 함유할 수 있다.Hard capsules among the health functional foods in the form of capsules may be prepared by filling a mixture of additives such as excipients with neferine, which is the active ingredient of the present invention, in a conventional hard capsule, and the soft capsules are neferine ) Can be prepared by filling a capsule base such as gelatin with a mixture of additives such as excipients. The soft capsule agent may contain a plasticizer such as glycerin or sorbitol, a colorant, a preservative, and the like, as necessary.
환 형태의 건강기능식품은 본 발명의 유효성분인 네퍼린(neferine)과 부형제, 결합제, 붕해제 등을 혼합한 혼합물을 기존에 공지된 방법으로 성형하여 조제할 수 있으며, 필요에 따라 백당이나 다른 제피제로 제피할 수 있으며, 또는 전분, 탈크와 같은 물질로 표면을 코팅할 수도 있다.The cyclic health functional food can be prepared by molding a mixture of neferine, an active ingredient of the present invention, an excipient, a binder, a disintegrant, and the like by using a conventionally known method. It may be encapsulated with a skin coating, or the surface may be coated with a material such as starch, talc.
과립 형태의 건강기능식품은 본 발명의 유효성분인 네퍼린(neferine)과 부형제, 결합제, 붕해제 등을 혼합한 혼합물을 기존에 공지된 방법으로 입상으로 제조할 수 있으며, 필요에 따라 착향제, 교미제 등을 함유할 수 있다.The health functional food in the form of granules can be prepared by granulation of a mixture of neferine, an excipient, an excipient, a binder, a disintegrant, and the like, which is an active ingredient of the present invention. A mating agent and the like.
상기 건강기능식품은 음료류, 육류, 초코렛, 식품류, 과자류, 피자, 라면, 기타 면류, 껌류, 사탕류, 아이스크림류, 알코올 음료류, 비타민 복합제 및 건강보조식품류 등일 수 있다.The health functional food may be beverages, meat, chocolate, foods, confectionery, pizza, ramen, other noodles, gum, candy, ice cream, alcoholic beverages, vitamin complexes and health supplements.
이하, 실시예를 통하여 본 발명을 보다 상세히 설명하고자 한다. 이들 실시예는 본 발명을 보다 구체적으로 설명하기 위한 것으로, 본 발명의 범위가 이들 실시예에 한정되는 것은 아니다.Hereinafter, the present invention will be described in more detail with reference to Examples. These examples are intended to illustrate the present invention more specifically, but the scope of the present invention is not limited to these examples.
<실시예><Example>
시약reagent
DMSO, BSA 및 propidium iodide은 Sigma Aldrich (St. Louis, MO, USA)로부터 구입하였으며; EZ-Cytox Cell Viability Assay Solution WST-1은 Daeil Lab Service (Jong-No, Seoul, Korea)로부터 구입하였고; DAPI은 Roche (Pleasanton, CA, USA)로부터 구입하였으며; Lysis buffer[50 mM Tris-Cl (pH 7.5), 150 mM NaCl, 1 mM DTT, 0.5% NP-40, 1% Triton X-100, 1% deoxycholate, 0.1% SDS] 및 proteinase inhibitors (PMSF, EDTA, Aprotinin, Leupeptin, and Prostatin A)는 Intron biotechnology (Gyeonggi, Korea)에서 구입하였고; The Protein Quantification Kit (CBB solution)는 Dojindo Molecular Technologies (Rockville, MD, USA)에서 구입하였으며; Nitrocellulose membranes는 PALL Life Sciences (Ann Arbor, MI, USA)에서 구입하였고; Enhanced chemiluminescent (ECL) detection solutions은 Pierce (Rockford, IL, USA)로부터 구입하였으며; caspase-12 및 사이클린 D1에 대한 항체는 Abcam (Cambridge, MA, USA)으로부터 구입하였고; 및 c-Myc, Cyclin D3, CDK4, E2F-1, p21Waf1/Cip1, phospho-cdc2(Tyr15), GAPDH, Bim, Bid, Bax, Bak, Puma, β-actin, cleaved caspase-8, caspase-3, cleaved caspase-3, cleaved caspase-6, cleaved caspase-7, cleaved PARP, Bip, calnexin, PDI, Calpain 2, LC3B, SirT6, DFF45/DFF35, Lamin A/C, HRP conjugated anti-rabbit with anti-mouse, anti-mouse IgG (H+L), F(ab’)2 fragment (Alexa Fluor 555 Conjugate) and anti-rabbit IgG (H+L), F(ab’)2 fragment (Alexa Fluor 488 Conjugate)에 대한 항체들은 Cell Signaling Biotechnology (Beverly, MA, USA)로부터 구입하였다.DMSO, BSA and propidium iodide were purchased from Sigma Aldrich (St. Louis, MO, USA); EZ-Cytox Cell Viability Assay Solution WST-1 was purchased from Daeil Lab Service (Jong-No, Seoul, Korea); DAPI was purchased from Roche (Pleasanton, CA, USA); Lysis buffer [50 mM Tris-Cl (pH 7.5), 150 mM NaCl, 1 mM DTT, 0.5% NP-40, 1% Triton X-100, 1% deoxycholate, 0.1% SDS] and proteinase inhibitors (PMSF, EDTA, Aprotinin, Leupeptin, and Prostatin A) were purchased from Intron biotechnology (Gyeonggi, Korea); The Protein Quantification Kit (CBB solution) was purchased from Dojindo Molecular Technologies (Rockville, MD, USA); Nitrocellulose membranes were purchased from PALL Life Sciences (Ann Arbor, MI, USA); Enhanced chemiluminescent (ECL) detection solutions were purchased from Pierce (Rockford, IL, USA); antibodies against caspase-12 and cyclin D1 were purchased from Abcam (Cambridge, MA, USA); And c-Myc, Cyclin D3, CDK4, E2F-1, p21Waf1 / Cip1, phospho-cdc2 (Tyr15), GAPDH, Bim, Bid, Bax, Bak, Puma, β-actin, cleaved caspase-8, caspase-3, cleaved caspase-3, cleaved caspase-6, cleaved caspase-7, cleaved PARP, Bip, calnexin, PDI, Calpain 2, LC3B, SirT6, DFF45 / DFF35, Lamin A / C, HRP conjugated anti-rabbit with anti-mouse, Antibodies to anti-mouse IgG (H + L), F (ab ') 2 fragment (Alexa Fluor 555 Conjugate) and anti-rabbit IgG (H + L), F (ab') 2 fragment (Alexa Fluor 488 Conjugate) Were purchased from Cell Signaling Biotechnology (Beverly, MA, USA).
통계statistics
모든 실험결과는 평균±표준편차로 표기하였고, 그 결과들의 통계 분석하기 위하여 통계 소프트웨어 시그마 플랏 v.12.3 (Systat Software, Inc., San Jose, CA, USA)을 이용 Tukey’s test에 따른 one-way ANOVA를 수행하였다. P = <0.001 가 통계적으로 유의하다고 판단하였다.All experimental results were expressed as mean ± standard deviation and one-way ANOVA according to Tukey's test using statistical software Sigma plot v.12.3 (Systat Software, Inc., San Jose, CA, USA) for statistical analysis of the results. Was performed. P = <0.001 was determined to be statistically significant.
<< 실시예Example 1>  1> 네퍼린Neferrin (( neferineneferine )의 정제Tablets
연꽃 (N. nucifera) 배아 2.0kg을 3.0L 메탄올에서 3시간 동안 환류 추출한 다음 40℃에서 진공 건조를 통해 농축시켜 연꽃 배아 메탄올 추출물을 543.5g를 수득하였다. 상기 과정을 통해 수득한 추출물은 증류수에 현탁시킨 후 디클로로메탄, 에틸아세테이트 및 n-부탄올로 순차적으로 분획하여 디클로로메탄 분획물 13.8g, 에틸아세테이트 분획물 2.2g 및 부탄올 분획물 58.3g을 각각 수득하였으며, 물 잔여물은 469.0g을 얻었다. 상기 분획물 중 디클로로메탄 분획물(13.8g)은 다시 실리카겔 컬럼(120mm i.d.) 에 걸어 크로마토그래피를 수행하였으며, 그 결과 최종적으로 네퍼린(neferine) 920mg을 수득하였다. 이때 용출용매로는 벤젠-에틸아세테이트-디에틸아민(7:2:1, isocratic)의 혼합용매를 이용하였다.2.0 kg of lotus ( N. nucifera ) embryos were extracted under reflux for 3 hours in 3.0 L methanol and concentrated through vacuum drying at 40 ° C. to obtain 543.5 g of lotus embryo methanol extract. The extract obtained through the above process was suspended in distilled water and sequentially partitioned into dichloromethane, ethyl acetate and n-butanol to obtain 13.8 g of dichloromethane fraction, 2.2 g of ethyl acetate fraction and 58.3 g of butanol fraction, respectively. Water obtained 469.0 g. The dichloromethane fraction (13.8 g) in the fraction was again subjected to chromatography on a silica gel column (120 mm id), whereby finally, 920 mg of neferine was obtained. At this time, a mixed solvent of benzene-ethyl acetate-diethylamine (7: 2: 1, isocratic) was used as the elution solvent.
상기 과정을 통해 최종적으로 수득한 물질이 네퍼린인 것을 확인하기 위하여, 1H- 및 13C-NMR을 포함하는 스펙트럼을 분석하였으며, 이러한 분석 데이터를 종래 공개된 데이터와 비교하여 상기 물질이 네퍼린이 것을 검증하였다.In order to confirm that the material finally obtained through the above process was neferrin, spectra including 1 H- and 13 C-NMR were analyzed, and the analytical data was compared with the previously published data. Was verified.
1H-NMR (400 MHz, CDCl3): δ 6.88 (2H, d, J = 8.4 Hz, H-11/H-15), 6.81 (1H, d, J = 8.1 Hz, H-14′), 6.66 (3H, d, J = 8.2 Hz, H-15′/H-12/H-14), 6.62 (1H, s, H-5), 6.51 (1H, m, H-11′), 6.49 (1H, s, H-5′), 6.35 (1H, s, H-8), 5.98 (1H, s, H-8′), 3.79 (3H, s, 6-OCH3), 3.76 (3H, s, 7′-OCH3), 3.70 (3H, s, 13-OCH3), 3.60 (2H, m, H-1/H-1′), 3.52 (3H, s, 6′-OCH3), 3.15~2.95 (4H, m, H-3/H-3′/H-9/H-9′), 3.04 (1H, dd, J = 12.0, 5.0 Hz, H-9), 2.98 (1H, dd, J = 12.0, 5.3 Hz, H-9′), 2.832.53 (8H, m, H-3/H-3′/H-4/H-4′/H-9/H-9′), 2.47 (3H, H-2), 2.43 (3H, s, H-2′); 13C-NMR (100 MHz, CDCl3): δ 157.7 (C-13), 148.8 (C-6), 147.1 (C-7′), 146.2 (C-6′), 145.5 (C-13′), 144.7 (C-12′), 142.7 (C-7), 131.6 (C-10′), 131.3 (C-10), 130.8 (C-8a), 130.3 (C-4a), 130.3 (C-11), 130.3 (C-15), 129.0 (C-4a′), 125.5 (C-8a′), 125.1 (C-11′), 119.8 (C-8), 119.1 (C-15′), 115.6 (C-14′), 113.3 (C-12), 113.3 (C-14), 112.2 (C-5), 111.0 (C-5′), 110.9 (C-8′), 64.6 (C-1′), 64.3 (C-1), 55.7 (7′-OCH3), 55.6 (6′-OCH3), 55.3 (6-OCH3), 55.0 (13-OCH3), 47.0 (C-3), 46.5 (C-3′), 42.5 (C-2), 42.3 (C-2′), 40.5 (C-9), 39.7 (C-9′), 26.0 (C-4), 25.0 (C-4′). 1 H-NMR (400 MHz, CDCl 3): δ 6.88 (2H, d, J = 8.4 Hz, H-11 / H-15), 6.81 (1H, d, J = 8.1 Hz, H-14 ′), 6.66 (3H, d, J = 8.2 Hz, H-15 ′ / H-12 / H-14), 6.62 (1H, s, H-5), 6.51 (1H, m, H-11 ′), 6.49 (1H , s, H-5 ′), 6.35 (1H, s, H-8), 5.98 (1H, s, H-8 ′), 3.79 (3H, s, 6-OCH3), 3.76 (3H, s, 7 ′ -OCH3), 3.70 (3H, s, 13-OCH3), 3.60 (2H, m, H-1 / H-1 ′), 3.52 (3H, s, 6′-OCH3), 3.15-2.95 (4H, m, H-3 / H-3 ′ / H-9 / H-9 ′), 3.04 (1H, dd, J = 12.0, 5.0 Hz, H-9), 2.98 (1H, dd, J = 12.0, 5.3 Hz, H-9 ′), 2.832.53 (8H, m, H-3 / H-3 ′ / H-4 / H-4 ′ / H-9 / H-9 ′), 2.47 (3H, H- 2), 2.43 (3H, s, H-2 '); 13 C-NMR (100 MHz, CDCl 3): δ 157.7 (C-13), 148.8 (C-6), 147.1 (C-7 ′), 146.2 (C-6 ′), 145.5 (C-13 ′), 144.7 (C-12 '), 142.7 (C-7), 131.6 (C-10'), 131.3 (C-10), 130.8 (C-8a), 130.3 (C-4a), 130.3 (C-11) , 130.3 (C-15), 129.0 (C-4a ′), 125.5 (C-8a ′), 125.1 (C-11 ′), 119.8 (C-8), 119.1 (C-15 ′), 115.6 (C -14 '), 113.3 (C-12), 113.3 (C-14), 112.2 (C-5), 111.0 (C-5'), 110.9 (C-8 '), 64.6 (C-1'), 64.3 (C-1), 55.7 (7'-OCH3), 55.6 (6'-OCH3), 55.3 (6-OCH3), 55.0 (13-OCH3), 47.0 (C-3), 46.5 (C-3 ' ), 42.5 (C-2), 42.3 (C-2 '), 40.5 (C-9), 39.7 (C-9'), 26.0 (C-4), 25.0 (C-4 ').
<< 실시예Example 2>  2> 세포주 및 배양Cell line and culture
HCC Hep3B, Sk-Hep1, THLE-3(정상 간세포를 SV40 large T antigen으로 감염시켜 불멸화한 세포) 및 HUVEC(인간제대정맥내피세포) 세포들은 the American Tissue Culture Collection (Manassas, VA, USA)로부터 얻었다.HCC Hep3B, Sk-Hep1, THLE-3 (cells immortalized by infecting normal hepatocytes with SV40 large T antigen) and HUVEC (human venous venous endothelial cells) cells were obtained from the American Tissue Culture Collection (Manassas, VA, USA) .
Hep3B 및 Sk-Hep1 세포는 Earle's Balanced Salts가 포함된 최소필수배지(MEM/EBSS) (HyClone, Logan, UT, USA)를 이용하여 배양하였으며, THLE-3세포는 상기 세포 배지에 함유된 젠타마이신/엠포테리신 및 에피네프린을 제거하고 500ml 기초 배지가 함유된 Bronchial Epithelia Cell Growth Medium (Clonetics BEGM bullet kit; CC3170, Lonza, Walkersville, MD, USA) 배지를 이용하여 배양하였으며, 여기에 5ng/ml EGF, 70ng/ml 포스포에탄올아민을 첨가하여 배양하였다. 인간제대정맥내피세포(Human Umbilical Vein Endothelial Cell, HUVECs)는 EGM-2 kit (Lonza)가 보충된 endothelial basal medium-2 (EBM-2)를 이용하여 배양하였다. 본 실험의 모든 세포주들은 10% 열 불활성화된 FBS (HyClone), 100 U/ml 페니실린, 및 100 μg/ml 스트렙토마이신 (PAA Laboratories GmbH, PA, Austria)을 포함하는 배지에서 37℃, 5% CO2의 조건에서 배양되었다. THLE-3 세포들은 0.01 mg/ml BSA, 0.01 mg/ml 피브로넥틴 및 0.03 mg/ml bovine collagen type Ι (BD biosciences, San Jose, CA, USA)으로 37℃에서 2시간 동안 코팅시킨 후 배양되었다.Hep3B and Sk-Hep1 cells were cultured using minimum essential medium (MEM / EBSS) (HyClone, Logan, UT, USA) containing Earle's Balanced Salts, and THLE-3 cells were gentamicin / Empotericin and epinephrine were removed and cultured using Bronchial Epithelia Cell Growth Medium (Clonetics BEGM bullet kit; CC3170, Lonza, Walkersville, MD, USA) medium containing 500 ml basal medium, 5 ng / ml EGF, 70 ng. Culture was added by adding / ml phosphoethanolamine. Human Umbilical Vein Endothelial Cells (HUVECs) were cultured using endothelial basal medium-2 (EBM-2) supplemented with EGM-2 kit (Lonza). All cell lines in this experiment were 37 ° C., 5% CO in media containing 10% heat inactivated FBS (HyClone), 100 U / ml penicillin, and 100 μg / ml streptomycin (PAA Laboratories GmbH, PA, Austria). It was incubated in the conditions of 2 . THLE-3 cells were coated with 0.01 mg / ml BSA, 0.01 mg / ml fibronectin and 0.03 mg / ml bovine collagen type Ι (BD biosciences, San Jose, Calif., USA) for 2 hours at 37 ° C and then incubated.
<< 실험예Experimental Example 1> 1>
본 실험에서는 상기 실시예 1을 통해 수득한 네퍼린의 암세포주에 대한 세포 독성을 확인하기 위해 실시예 2를 통해 배양된 Hep3B, Sk-Hep1 및 THLE-3 세포주를 이용하여 Cell viability assay를 실시하였다. In this experiment, the cell viability assay was performed using Hep3B, Sk-Hep1, and THLE-3 cell lines cultured in Example 2 to confirm the cytotoxicity of the cancer cell line of nephrin obtained through Example 1. .
먼저 Hep3B, Sk-Hep1 및 THLE-3 세포 각각을 1×104 cells 밀도로 100μl 배지에 재현탁시켜 96-웰 플레이트 3개에 분주한 후 24시간 인큐베이션시켰다. 그런 다음 다양한 농도(5, 10, 15, 20, 25, 30μM)의 네퍼린을 상기 세포에 처리하여 24시간 동안 배양한 후 새 배지로 교환 한 뒤, 10μl의 WST-1 용액을 첨가하여 5시간 동안 반응시켰다. 반응 흡광도는 ELISA reader(Molecular Devices, Sunnyvale, CA, USA)를 이용하여 460nm에서 측정하였다. 결과는 3번의 독립적인 실험을 통해 평균±표준편차로 나타냈으며, 억제 그래프는 대조군 값과 관련된 각각의 농도로부터 얻어진 평균값을 이용해 도출하였다.First, Hep3B, Sk-Hep1 and THLE-3 cells were each resuspended in 100 μl medium at 1 × 10 4 cells density and dispensed into three 96-well plates and incubated for 24 hours. Then, neferrin at various concentrations (5, 10, 15, 20, 25, 30 μM) was treated with the cells, incubated for 24 hours, exchanged with fresh medium, and then added with 10 μl of WST-1 solution for 5 hours. Reacted for a while. Reaction absorbance was measured at 460 nm using an ELISA reader (Molecular Devices, Sunnyvale, Calif., USA). The results were expressed as mean ± standard deviation through three independent experiments, and the inhibition graph was derived using the mean values obtained from each concentration associated with the control values.
그 결과 도 1에서 나타낸 바와 같이, Hep3B, Sk-Hep1 및 THLE-3 세포주 중에서 단지 Hep3B 세포주에서만 네퍼린 처리 농도에 의존적으로 세포 생존율이 두드러지게 감소되는 것을 확인할 수 있었다. 특히 10μM 농도 이상의 네퍼린이 처리된 실험군에서 두드러진 세포 억제가 나타났다. 한편, Sk-Hep1 및 THLE-3 세포들은 네퍼린 처리에 따른 세포 독성이 나타나지 않았으며, Hep3B 세포주에서 10% 이하의 세포생존율을 보인 30μM 농도의 네퍼린 처리군에서, Sk-Hep1 및 THLE-3 세포들의 생존율은 약 90%에 달하는 것으로 측정되었다.As a result, as shown in FIG. 1, only Hep3B cell lines among Hep3B, Sk-Hep1 and THLE-3 cell lines showed a significant decrease in cell viability depending on the neferrin treatment concentration. In particular, there was a marked cell suppression in the experimental group treated with neferrin concentration of 10 μM or more. On the other hand, Sk-Hep1 and THLE-3 cells did not show cytotoxicity according to the neferrin treatment, and in the 30 μM concentration of the nephrine treated group showing less than 10% cell viability in the Hep3B cell line, the Sk-Hep1 and THLE-3 cells. The survival rate of the cells was measured to reach about 90%.
이러한 결과를 통해 네퍼린은 Hep3B 세포에 특이적인 세포독성 민감도를 가지는 것을 확인할 수 있었다. 즉 네퍼린은 간암세포주인 Hep3B 및 Sk-Hep1에 모두 세포독성을 보이는 것이 아니며, Hep3B 세포주에 특이적인 세포독성을 보임으로써, 간암세포에서도 종류를 달리하여 항암활성을 가질 수 있음을 시사하였다.These results confirm that neferrin has a specific cytotoxic sensitivity to Hep3B cells. In other words, neferin did not show cytotoxicity to both Hep3B and Sk-Hep1 liver cancer cell lines, and showed specific cytotoxicity to Hep3B cell line, suggesting that hepatic cancer cells may have anticancer activity in different types.
상기 실험을 통해 본 발명의 네퍼린이 간암세포주에서도 Hep3B 세포주에 대한 특이적인 세포독성을 확인하였는바, 하기 실험에서는 Hep3B 세포주를 대상으로 하여 항암활성과 관련된 다양한 기작에 대한 실험을 실시하였다.Through the above experiment, the nephrine of the present invention confirmed specific cytotoxicity to Hep3B cell line even in hepatocellular carcinoma cell line. In the following experiment, experiments were conducted on various mechanisms related to anticancer activity in Hep3B cell line.
<< 실험예Experimental Example 2>  2> 네퍼린의Neferrin 간암세포에 대한 세포사멸 유도 Induction of Apoptosis on Liver Cancer Cells
본 실험에서는 네퍼린의 간암세포주인 Hep3B에 대한 세포사멸(apoptosis) 유도능을 살펴보기 위하여, DAPI 에세이를 실시하였다.In this experiment, we conducted a DAPI assay to examine the ability of nephrine to induce apoptosis on Hep3B, a liver cancer cell line.
DAPI는 4, 6-diamidino-2-phenylindole dihydrochloride hydrate의 약칭으로, 세포에 투과될 수 있고 고정된 상태의 살아있는 세포핵들을 염색하는데 사용될 수 있는 프로브(probe)이다. DNA와 마이너 그루브(minor groove)와 결합을 이루며 AT rich DNA와 결합하는 것을 선호하며, 수소결합을 통해 안정된 상태가 된다. DNA와 결합한 DAPI는 DNA와 결합하지 않은 것에 비하여 20배 정도의 밝기를 가지며 형광 현미경에서 자외선에 의해 발광하는 특징을 가진다. DNA와 결합되었을 때, 358nm에서 흡광 최대치를 나타내고 461nm에서 발광최대치를 나타낸다. 본 실험에서는 네퍼린 처리에 따른 핵 응축 및 apoptosome 형성의 검출을 위해, 간암세포를 DAPI로 염색하였으며, 이를 형광 현미경 하에 분석하였다(×1000).DAPI is an abbreviation of 4, 6-diamidino-2-phenylindole dihydrochloride hydrate and is a probe that can be permeated to cells and used to stain living cell nuclei in a fixed state. It forms a bond with DNA and a minor groove, and prefers to bind with AT rich DNA, and becomes stable through hydrogen bonding. DAPI bound to DNA is about 20 times brighter than non-DNA bound and emits light by ultraviolet light under a fluorescence microscope. When combined with DNA, it exhibits an absorption maximum at 358 nm and an emission maximum at 461 nm. In this experiment, hepatic cancer cells were stained with DAPI for detection of nuclear condensation and apoptosome formation following neferrin treatment and analyzed under fluorescence microscope (× 1000).
본 실험을 위해서 먼저 Hep3B 세포들을 cover glass bottom culture dish에서 24시간 동안 배양한 후, 6시간 간격으로 네퍼린(20, 25μM )을 처리한 뒤 PBS 버퍼로 한번 세척한 후 DAPI 용액(1μg/ml)을 첨가하여 염색하였다. 37℃에서 20분간 암실에서 배양하고, 세포를 다시 메탄올로 한번 세척하였다. 핵의 변화는 ECLIPSE 50i fluorescence microscope (Nikon, Tokyo, Japan)를 이용하여 관찰하였다.For this experiment, Hep3B cells were first incubated in a cover glass bottom culture dish for 24 hours, treated with neferin (20, 25 μM) at 6 hour intervals, and washed once with PBS buffer, followed by DAPI solution (1 μg / ml). Staining was added. Incubated in the dark at 37 ° C. for 20 minutes, and the cells were washed once again with methanol. Nuclear changes were observed using an ECLIPSE 50i fluorescence microscope (Nikon, Tokyo, Japan).
그 결과 도 2에서 나타낸 바와 같이, 네퍼린를 처리하지 않은 무처리군과 비교하여 네퍼린을 처리한 후 6시간이 경과한 실험군에서 세포들의 응축(shrinkage) 및 파열(blebbing)과 같은 핵 막 변화가 나타나는 것을 확인할 수 있었다. 특히 네퍼린 처리 후 24시간이 경과한 실험군에서 사멸세포(apoptotic cells)의 전형적인 특징, 핵 응축(apoptosome formation)이 관찰되었다.As a result, as shown in FIG. 2, nuclear membrane changes such as shrinkage and blebbing of cells were observed in the experimental group 6 hours after the neferrin treatment compared to the non-nephrrine-treated group. It was confirmed that it appeared. In particular, a typical characteristic of apoptotic cells, nuclear condensation (apoptotic formation) was observed in the experimental group 24 hours after neferrin treatment.
<< 실험예Experimental Example 3> 3> 네퍼린의Neferrin 간암세포에 대한 세포주기중지( Cell cycle arrest for liver cancer cells cellcell cyclecycle arrestarrest ) 유도) Judo
본 실험에서는 네퍼린의 간암세포주인 Hep3B에 대한 세포주기중지(cell cycle arrest) 유도능을 살펴보기 위하여, 세포주기 분석; 웨스턴 블랏을 통한 세포주기와 관련된 단백질 발현 분석; 및 면역형광분석을 실시하였다.In this experiment, to investigate the cell cycle arrest induction of hep3B, hepatic cancer cell line, cell cycle analysis; Protein expression analysis associated with cell cycle via Western blot; And immunofluorescence analysis.
<3-1> 세포주기 분석<3-1> Cell Cycle Analysis
Hep3B 세포에 네퍼린을 농도별(15, 20, 25μM)로 처리하여 24시간 반응시킨 후, 상기 세포에 트립신을 처리한 다음, 4℃에서 70% 에탄올을 이용하여 밤새 고정하였다. 그 후, 상기 세포들은 0.2mg/ml RNase A를 포함하는 PBS 버퍼로 재현탁한 다음, 37℃에서 1시간 동안 추가 배양하였다. 상기 과정을 거친 세포들은 암실에서 30분 동안 40μg/ml의 프로피디움 요오드화물(Propidium iodide, PI)로 염색하였다. subgenomic DNA 함량의 분산은 유세포분석기(BD biosciences)를 이용하여 분석하였다.Hep3B cells were treated with nephrine at different concentrations (15, 20, 25 μM) for 24 hours, and then treated with trypsin, and then fixed overnight using 70% ethanol at 4 ° C. The cells were then resuspended in PBS buffer containing 0.2 mg / ml RNase A and then further incubated at 37 ° C. for 1 hour. The cells were stained with 40 μg / ml propidium iodide (PI) for 30 minutes in the dark. Dispersion of subgenomic DNA content was analyzed using flow cytometer (BD biosciences).
그 결과 도 3에서 나타낸 바와 같이, 네퍼린 처리 농도(15, 20, 25μM)에 비례하여 sub-G1 cell population이 증대되는 것을 확인할 수 있었다. 특히, 네퍼린을 처리하지 않은 무처리군에서는 sub-G1 cell population이 1.35%를 나타낸 반면, 네퍼린을 15μM 농도로 처리한 실험군은 4.78%, 네퍼린을 20μM 농도로 처리한 실험군에서는 16.01%까지 증대되었다. 또한, 네퍼린 무처리군과 비교하여 네퍼린을 20μM 농도로 처리한 실험군에서 G1/S phase가 대략 9%가 증가된 것을 확인할 수 있었다.As a result, as shown in Figure 3, it was confirmed that the sub-G1 cell population increases in proportion to the neferrin treatment concentration (15, 20, 25μM). In particular, the sub-G1 cell population showed 1.35% in the untreated group without neferrin, while 4.78% in the experimental group treated with 15 μM concentration of neferrin, and 16.01% in the experimental group treated with 20 μM concentration of neferin. Increased. In addition, it was confirmed that the G1 / S phase was increased approximately 9% in the experimental group treated with 20 μM concentration of neferrin compared to the untreated group.
이러한 결과를 통해, Hep3B 세포에 네퍼린을 처리하는 경우 sub-G1기 및 G1기의 중지를 통해 간세포암의 세포 성장을 억제할 수 있음을 확인하였다.Through these results, it was confirmed that when hep3B cells were treated with neferrin, cell growth of hepatocellular carcinoma was inhibited through the suspension of sub-G1 and G1 phases.
<3-2> 세포주기와 관련된 단백질 발현 분석<3-2> Analysis of Protein Expression Related to Cell Cycle
세포주기와 관련된 단백질(c-Myc, 사이클린 D1, 사이클린 D3, CDK4, E2F-1, P21Waf/Cip1, p-cdc2) 발현 분석을 위하여 웨스턴 블랏을 실시하였다.Western blots were performed for expression analysis of cell cycle-related proteins (c-Myc, cyclin D1, cyclin D3, CDK4, E2F-1, P21 Waf / Cip1 , p-cdc2).
본 실험을 위해서 먼저 Hep3B 세포에 네퍼린을 농도별(15, 20, 25μM)로 24시간 동안 처리한 다음, 차가운 PBS로 2번 세척한 후 차가운 용해버퍼로 용해시켰다. 그런 다음, 얼음에서 30분 동안 반응시킨 후, 14,000rpm에서 20분 동안 원심분리하여 불용성 물질 제거하였다. 세포 용해물의 단백질 함량은 Protein Quantification Kit (CBB solution)를 이용하여 측정하였다. 웨스턴 블랏은 12% 폴리아크릴아마이트 젤에서 전기영동을 통해 분리된 단백질 30μg을 이용하여 수행되었으며, 니트로셀룰로오즈 멤브레인으로 이동시키고 지시 항체로 면역 반응을 수행하였다. chemiluminescent (ECL) 시약을 사용하여 화학발광을 검출하였다.For this experiment, first, Hep3B cells were treated with neferin by concentration (15, 20, 25 μM) for 24 hours, washed twice with cold PBS, and then lysed with cold lysis buffer. Then, the reaction was carried out on ice for 30 minutes, followed by centrifugation at 14,000 rpm for 20 minutes to remove insoluble matters. Protein content of the cell lysate was measured using a Protein Quantification Kit (CBB solution). Western blots were performed using 30 μg of protein isolated via electrophoresis on a 12% polyacrylamide gel, transferred to nitrocellulose membrane and subjected to immune reaction with indicator antibody. Chemiluminescence was detected using chemiluminescent (ECL) reagent.
그 결과 도 4에서 나타낸 바와 같이, 농도별 네퍼린을 처리한 실험군에서 cdc2의 탈인산화; c-Myc, cyclin D1, D3, CDK4, E2F-1의 하향조절; 및 p21 Waf1/Cip1 단백질 발현이 증대되는 것을 확인할 수 있었다. 이러한 결과는 세포주기 중지를 나타내는 증거로써 상기 실험예<3-1>과 일치하는 결과이다. As a result, as shown in Figure 4, the dephosphorylation of cdc2 in the experimental group treated with neferrin for each concentration; down regulation of c-Myc, cyclin D1, D3, CDK4, E2F-1; And p21 Waf1 / Cip1 protein expression was increased. These results are in agreement with Experimental Example <3-1> as evidence of cell cycle arrest.
<3-3> <3-3> 면역형광분석Immunofluorescence
배양된 Hep3B 세포들을 cover-glass bottom dish 상에서 15μM 농도의 네퍼린을 처리하여 12시간동안 반응시켰다. 세포들은 37℃에서 20분 동안 1μg/ml DAPI로 전처리하였으며, 그 후 25℃에서 15분 동안 4% 포름알데하이드 (JUNSEI Chemical Co., Japan)로 고정한 다음, 5% 마우스 및 0.3% Triton X-100 (Sigma-Aldrich, St. Louis, MO, USA)을 가지는 정상 토끼 혈청 (Santa Cruz Biotechnology Inc.)을 함유하는 blocking 용액으로 1시간 동안 차단시켰다. 고정되고 차단된 세포들은 1차 항체(p21Waf1/Cip1)로 반응시킨 다음, PBS 버퍼로 3번 세척하였다. 세척 후, 상기 세포들을 항-마우스 IgG (H+L) 0.1 μg/ml, F(ab’)2 프래그먼트 (Alexa Fluor 555 Conjugate) 및 항-래빗 IgG (H+L), F(ab’)2 프래그먼트 (Alexa Fluor 488 Conjugate)로 1시간 동안 처리하였다. 염색된 세포들은 Prolong Gold Antifade Reagent (Invitrogen, Grand Island, NY, USA)로 슬라이드에 옮긴 후 charged-coupled device (CCD) camera가 장착된 ECLIPSE 50i fluorescence microscope을 이용하여 측정하였다. 이미지들은 켑쳐된 후 High-Content Analysis Software (Cambridge Healthtech Ins., Needham, MA, USA)로 처리하였다.Cultured Hep3B cells were reacted for 12 hours by treatment with 15 μM concentration of neferrin on a cover-glass bottom dish. Cells were pretreated with 1 μg / ml DAPI for 20 minutes at 37 ° C., then fixed with 4% formaldehyde (JUNSEI Chemical Co., Japan) at 25 ° C. for 15 minutes, then 5% mice and 0.3% Triton X-100 It was blocked for 1 hour with a blocking solution containing normal rabbit serum (Santa Cruz Biotechnology Inc.) with (Sigma-Aldrich, St. Louis, MO, USA). Fixed and blocked cells were reacted with the primary antibody (p21Waf1 / Cip1) and then washed three times with PBS buffer. After washing, the cells were treated with 0.1 μg / ml anti-mouse IgG (H + L), F (ab ') 2 fragment (Alexa Fluor 555 Conjugate) and anti-rabbit IgG (H + L), F (ab') 2. Fragment (Alexa Fluor 488 Conjugate) was treated for 1 hour. Stained cells were transferred to slides with Prolong Gold Antifade Reagent (Invitrogen, Grand Island, NY, USA) and measured using an ECLIPSE 50i fluorescence microscope equipped with a charged-coupled device (CCD) camera. Images were captured and processed with High-Content Analysis Software (Cambridge Healthtech Ins., Needham, MA, USA).
그 결과 도 5에서 나타낸 바와 같이, 네퍼린을 처리하지 않은 무처리군과 비교하여 네퍼린을 처리한 Hep3B 세포들은 세포질에서 핵 주변의 p21 Waf1 / Cip1 단백질 발현 수준이 두드러지게 증대되는 것을 확인할 수 있었다.As a result, as shown in Figure 5, compared to the non-neferrin-treated group, hep3B cells treated with neferin was found to significantly increase the expression level of p21 Waf1 / Cip1 protein around the nucleus in the cytoplasm. .
<< 실험예Experimental Example 4>  4> 네퍼린의Neferrin 간암세포에 대한 세포사멸( Apoptosis on Liver Cancer Cells apoptosisapoptosis ) 유도) Judo
본 실험에서는 네퍼린의 간암세포주인 Hep3B에 대한 세포사멸(apoptosis)을 유도하는지를 살펴보기 위하여, 세포사멸 조절과 관련된 Bcl-2 패밀리 단백질 발현 및 카스파아제 활성화 정도를 측정하기 위하여 웨스턴 블랏을 실시하였으며, 면역형광분석법을 추가로 실시하였다.In this experiment, we performed Western blot to measure the degree of Bcl-2 family protein expression and caspase activation related to apoptosis in order to investigate whether apoptosis of Hep3B, a hepatic cancer cell line of nephrin, was induced. Immunofluorescence was further performed.
<4-1> <4-1> 웨스턴Weston 블랏Blot 분석 analysis
세포사멸 조절과 관련된 Bcl-2 패밀리 단백질(Bim, BID, Bax, Bak, Puma) 발현 및 카스파아제 활성화 정도를 분석하기 위하여 웨스턴 블랏을 실시하였다.Western blot was performed to analyze the degree of Bcl-2 family protein (Bim, BID, Bax, Bak, Puma) expression and caspase activation associated with apoptosis regulation.
실험 과정은 상기 실험예<3-2>과 동일하게 진행하였다.The experimental procedure was performed in the same manner as in Experimental Example <3-2>.
도 6a는 미토콘드리아-매개된 세포사멸 경로를 조절하는 것으로 알려진 Bcl-2 패밀리 단백질(Bim, Bid, Bax, Bak 및 Puma)의 웨스턴 블랏 결과를 나타낸 것으로, 이를 통해 Bak 및 Puma 단백질 발현이 네퍼린 처리 농도의 의존적으로 두드러지게 증대되는 것을 나타났으며, Bim, Bid 및 Bax은 미미하게 발현이 증대되는 것을 확인할 수 있었다. 또한 도 6b에서는 소포체 스트레스에 따른 세포사멸에 수반되는 apoptotic 단백질(cleaved caspase-8, -3, -6, -7 및 PARP)의 웨스턴 블랏 결과를 나타낸 것으로, 이를 통해 caspase-3 단백질 발현은 네퍼린 처리 농도에 의존적으로 감소되는 것으로 나타난 반면 cleaved caspase-6 및 cleaved PARP 단백질 발현이 두드러지게 증대되는 것을 확인할 수 있었다.FIG. 6A shows Western blot results of Bcl-2 family proteins (Bim, Bid, Bax, Bak and Puma) known to modulate mitochondrial-mediated apoptosis pathways, whereby Bak and Puma protein expression was treated with nephrin. It was shown that the concentration was significantly increased, and Bim, Bid and Bax were found to be slightly increased in expression. In addition, Figure 6b shows the Western blot results of apoptotic proteins (cleaved caspase-8, -3, -6, -7 and PARP) associated with apoptosis due to endoplasmic reticulum stress, through which caspase-3 protein expression is neferin It was found that the expression of cleaved caspase-6 and cleaved PARP protein was markedly increased while it was shown to decrease depending on the treatment concentration.
<4-2> <4-2> 면역형광분석Immunofluorescence
상기 실험예<4-1>을 통해 보여준 결과를 좀 더 확인하기 위하여, 본 실험에서는 면역형광분석법을 통해 몇몇 세포사멸 관련 단백질의 발현 수준을 관찰하였다.In order to further confirm the results shown in Experimental Example <4-1>, the expression levels of several apoptosis-related proteins were observed through immunofluorescence in this experiment.
실험 과정은 1차 항체로 cleaved caspase-3 및 cleaved caspase-8을 사용한 것을 제외하고는 상기 실험예<3-3>과 동일하게 진행하였다.The experimental procedure was performed in the same manner as in Experimental Example <3-3> except that cleaved caspase-3 and cleaved caspase-8 were used as primary antibodies.
그 결과 도 7에서 나타낸 바와 같이, 네퍼린을 처리하지 않은 무처리군과 비교하여 네퍼린을 처리한 Hep3B 세포에서 cleaved caspase-8, 및 -3이 두드러지게 발현된 것으로 나타났다. 이러한 결과를 통해 네퍼린에 의해 유도된 apoptosomes이 세포질에서 caspase-3 및 caspase-8을 활성화시켜 세포사멸을 야기하는 것이라 유추할 수 있었다. As a result, as shown in Figure 7, cleaved caspase-8, and -3 was found to be markedly expressed in Hep3B cells treated with neferrin compared to the untreated group not treated with neferrin. These results suggest that nephrine-induced apoptosomes induce apoptosis by activating caspase-3 and caspase-8 in the cytoplasm.
<< 실험예Experimental Example 5>  5> 네퍼린의Neferrin 간암세포에 대한 소포체 스트레스 및 자가포식( Vesicular Stress and Autophagy on Liver Cancer Cells autophagyautophagy ) 유도) Judo
자가포식(autophagy)은 진핵세포에서 세포 내 오래된 단백질, 손상된 소기관(organelles) 그리고 세포질이 탐식 및 소화되고 재사용되는 기전을 가지며, 이는 기본적인 세포질 구성요소들의 turnover에 관여하거나 외부적인 환경의 변화, 즉 영양분 고갈, 병원균 침윤, 산소 결핍 등으로 유도되기도 한다. 최근 발표되는 연구들은 암세포에서 자가포식을 유발하여 항암 효과를 볼 수 있다고 보고하고 있다. 자가포식은 여러 종양 세포주에서 종양 치료제에 의해 활성화된다고 보고되고 있지만, 암의 형성이나 억제와의 연관성에 대해서는 확실히 규명된 바가 없다.Autophagy has mechanisms by which eukaryotic cells, old organelles, damaged organelles, and cytoplasm are phagocytized, digested, and reused, which are involved in the turnover of basic cellular components or changes in the external environment, ie nutrients. It may also be induced by depletion, pathogen infiltration and oxygen starvation. Recently published studies have reported that cancer cells may have anti-cancer effects by inducing autophagy. Autophagy has been reported to be activated by tumor therapies in many tumor cell lines, but the association between cancer formation and inhibition has not been elucidated.
본 실험에서는 네퍼린의 간암세포에 대한 항암 효과로서 자가포식을 유도할 수 있는지를 확인하기 위하여, 네퍼린의 간암세포주인 Hep3B에 대한 소포체 스트레스를 야기 및 자가포식을 유무를 살펴보았다.In this experiment, to determine whether nephrine can induce autophagy as an anticancer effect on hepatocellular carcinoma cells, we investigated whether nephrine causes vesicle stress on Hep3B, a hepatocellular carcinoma cell line, and whether autophagy is present.
<5-1> <5-1> 네퍼린에In neferrin 의한 간암세포에서 소포체 스트레스 유도 Induction of Vesicular Stress in Liver Cancer Cells
먼저, 네퍼린에 의한 소포체 스트레스 유도를 확인하기 위하여, Hep3B 세포에 네퍼린을 처리한 후 소포체 샤페론 단백질의 발현량 웨스턴 블랏을 분석하였다. 웨스턴 블랏 분석은 상기 실험예<3-2>과 동일하게 진행하였다.First, in order to confirm the induction of endoplasmic reticulum stress by neferin, Hep3B cells were treated with neferin and analyzed for Western blot expression of endoplasmic reticulum chaperone protein. Western blot analysis was performed in the same manner as in Experimental Example <3-2>.
그 결과 도 8에서 나타낸 바와 같이, 네퍼린 처리한 실험군에서 시간에 의존적으로 Bip, calnexin, PDI, calpain2 및 caspase-12 단백질 발현 수준이 두드러지게 상향 조절되는 것으로 나타났다. 이러한 결과는 본 발명의 네퍼린이 간암세포주에서 소포체 스트레스를 야기시킴으로 인해 소포체 스트레스 완화를 위한 세포내 기작으로 샤페론 단백질의 발현이 증대되는 것을 의미한다.As a result, as shown in Figure 8, the neferin-treated experimental group was found to significantly up-regulate Bip, calnexin, PDI, calpain2 and caspase-12 protein expression levels depending on time. These results mean that the expression of chaperon protein is increased by intracellular mechanisms for endoplasmic reticulum stress relief because nephrin of the present invention causes endoplasmic reticulum stress in liver cancer cell line.
또한 상기 네퍼린의 소포체 스트레스 유도를 다시 증명하기 위하여, Hep3B 세포에 15μM 농도의 네퍼린을 12시간 동안 처리한 후 소포체 막에 위치한 단백질 caspase-12의 발현 정도를 면역형광분석법을 통해 분석하였으며, 그 결과는 도 9에서 나타낸 바와 같이 소포체의 확장을 관찰할 수 있었다. 이러한 결과들은 네퍼린에 의해 유도된 세포질내 소체(cytoplasmic vacuoles)가 소포체의 시스테네(cisternae)로의 확장 가능성을 의미하는 것이다. 실험 과정은 1차 항체로 caspase-12 사용한 것을 제외하고는 상기 실험예<3-3>과 동일하게 진행하였다.In addition, in order to demonstrate the induction of the endoplasmic reticulum stress of the neferin, after treatment with Hep3B cells for 15 hours at 15 μM concentration of neferin, the expression level of the protein caspase-12 located on the endoplasmic reticulum membrane was analyzed by immunofluorescence. As a result, the expansion of the endoplasmic reticulum was observed as shown in FIG. These results indicate that the cytoplasmic vacuoles induced by nephrin may expand the endoplasmic reticulum to cysternae. The experimental procedure was performed in the same manner as in Experimental Example <3-3> except that caspase-12 was used as the primary antibody.
<5-2> <5-2> 네퍼린에In neferrin 의한 간암세포에서  Caused by liver cancer cells 자가포식Self-feeding (( autophagyautophagy ) 유도) Judo
LC3는 자가포식의 발생에 있어서 중요한 구성요소 중 하나로 자가포식소체(autophagosome) 이중막을 형성하는데 관여하여 자가포식의 특이적 마커로 사용되고 있다. LC3는 세포 내에서 2가지 형태로 존재한다. LC3-Ι은 세포질 내에서 선행으로 존재하다가 세포 내부나 외부에서 스트레스를 받으면 LC3-Ⅱ로 변형되며, 이는 자가포식소체 이중막 형성에 관여하게 된다. 따라서 LC3-Ⅱ가 많이 발현되면 자가용해소체 역시 많이 만들어지는 상호관계에 있다.LC3 is an important component in the development of autophagy and is used as a specific marker for autophagy by participating in the formation of autophagosome double membranes. LC3 exists in two forms in cells. LC3-Ι is pre-existing in the cytoplasm and is transformed into LC3-II when stressed inside or outside the cell, which is involved in the formation of autophagosome bilayers. Therefore, when much LC3-II is expressed, autolysates are also in a correlation.
본 실험에서는 LC3가 네퍼린에 의해서 유발되는 자가포식과 관련이 있는지를 알아보기 위하여 Hep3B 세포에 20μM 농도의 네퍼린을 처리한 후 시간 경과에 따라 플라스미드 GFP-LC3의 발현 분포를 확인하였으며, 이를 통해 세포질 내 자가포식소체 형성 정도를 확인할 수 있었다.In this experiment, we examined the expression distribution of plasmid GFP-LC3 over time after treatment with 20 μM of nephrine in Hep3B cells to determine whether LC3 is related to autophagy induced by nephrine. The degree of autophagosome formation in the cytoplasm was confirmed.
먼저, GFP-LC3B(green fluorescent protein-light chain 3B) 플라스미드를 제조한 후, 본 프라스미드를 Hep3B 세포 도입하여 형질전환시켰다. 자세하게는 PCR을 이용하여 인간 간 cDNA 도서관 (Clontech, Mountain View, CA, USA)로부터 LC3B 전장서열을 증폭하였다. PCR 증폭에 사용된 프라이머 서열은 하기 표 1에 나타내었다. 정제된 PCR 생산물은 pGEM-T Easy vector (Promega, Madison, WI, USA) 내로 도입한 후 HindⅢ 및 Bam HΙ로 다시 절단하여, pEGFP-N2 vector (Clontech) 내에 N-terminal GFP와 융합시켰다. 이렇게 제조된 구조물은 DNA 시퀀싱을 통해 확인하였으며, GFP-LC3B 플라스미드라 명명하였다. 한편, Hep3B 세포들은 살균된 6-웰 플레이트에서 24시간 배양한 후, FBS-프리 배지에 Fugene 6 reagent (Roche Diagnostics GmbH, Mannheim, Germany) 혼합물을 이용하여 GFP-LC3B 플라스미드로 형질전환시켰다. 형질전환된 세포들은 레귤러 완전 배지에서 하루 동안 배양한 다음, 일정 시간 간격으로 (6, 12, 18, 24) 20μM 네퍼린을 처리한 후 차가운 PBS 버퍼로 세척하고 실온에서 5분간 차가운 메탄올 내에서 고정시켰다. 상기 세포들을 다시 PBS 버퍼로 2번 세척한 후, Gold Antifade Reagent로 커버슬립에 놓았다. ECLIPSE 50i fluorescence microscope을 이용하여 세포들을 관찰하였다.First, GFP-LC3B (green fluorescent protein-light chain 3B) plasmids were prepared, and the present plasmid was transformed by introducing Hep3B cells. Specifically, PCR was used to amplify LC3B full-length sequences from human liver cDNA libraries (Clontech, Mountain View, CA, USA). Primer sequences used for PCR amplification are shown in Table 1 below. Purified PCR products were introduced into the pGEM-T Easy vector (Promega, Madison, Wis., USA) and then cleaved back with HindIII and Bam Hl and fused with N-terminal GFP in the pEGFP-N2 vector (Clontech). The structure thus prepared was confirmed through DNA sequencing and named GFP-LC3B plasmid. Meanwhile, Hep3B cells were cultured in sterile 6-well plates for 24 hours, and then transformed into GFP-LC3B plasmid using Fugene 6 reagent (Roche Diagnostics GmbH, Mannheim, Germany) mixture in FBS-free medium. Transformed cells were incubated in regular complete medium for one day, then treated with 20 μM neferrin at regular intervals (6, 12, 18, 24), washed with cold PBS buffer and fixed in cold methanol for 5 minutes at room temperature. I was. The cells were washed again with PBS buffer twice and placed on coverslips with Gold Antifade Reagent. The cells were observed using an ECLIPSE 50i fluorescence microscope.
표 1 PCR에서 사용한 프라이머 서열
유전자 프라이머 서열
LC3B sense 5’-CCGGAATTCCATGCCCTCAGACCGGCCTTT-3’
antisense 5’-CGCGGATCCTCAGAAGCCGAAGGTTTCCTG-3’
Table 1 Primer sequence used in PCR
gene Primer sequence
LC3B sense 5'-CCGGAATTCCATGCCCTCAGACCGGCCTTT-3 '
antisense 5'-CGCGGATCCTCAGAAGCCGAAGGTTTCCTG-3 '
그 결과 도 10에서 나타낸 바와 같이, 네퍼린을 처리한 실험군에서 시간 경과에 따라 자가포식의 형태학적 특징 중 하나인 세포질에서의 소체(vacuole) 형성이 발생되었다. 이러한 소체들은 시간 의존적으로 더 크고 많이 증가하는 것을 확인할 수 있었다. 또한 자가포식은 자가포식소체(autophagosomes)라 불리는 이중막으로 둘러싸인 소포를 형성하는데, 네퍼린을 처리한 실험군에서 시간 경과에 따라 강력한 형광 스팟(GFP-LC3B "dots")이 나타남으로써 자가포식소체 수가 세포질 내에서 증가함을 확인할 수 있었다. 비록 FBS(-) 세포의 세포질 내 몇몇 녹색 형광들이 보이나, 이것을 자가포식소체 형태가 아니다.As a result, as shown in FIG. 10, in the experimental group treated with neferrin, formation of vacuole in the cytoplasm, which is one of morphological characteristics of autophagy, occurred over time. These bodies were found to grow larger and larger in time. Autophagy also forms vesicles enclosed by a double membrane called autophagosomes, which show strong fluorescence spots (GFP-LC3B "dots") over time in the nephrine treated group. It was confirmed that the increase in the cytoplasm. Although some green fluorescence in the cytoplasm of FBS (-) cells is seen, it is not in the form of autophagosomes.
또한 상기 네퍼린의 처리에 따른 LC3B 발현 증가를 웨스턴 블랏을 통해 분석한 결과, 도 11에서 나타낸 바와 같이 네퍼린을 처리하지 않은 무처리군과 비교하여 네퍼린 처리 실험군에서 막 주위 LC3B의 발현양이 네퍼린 처리된 농도 의존적으로 증가되는 것을 확인할 수 있었다.In addition, as a result of analyzing the increase in the expression of LC3B according to the treatment of the neferrin through Western blot, as shown in FIG. It was confirmed that the concentration increased depending on the neferrin treatment.
<< 실험예Experimental Example 6>  6> 네퍼린의Neferrin HUVECsHUVECs 세포에서 신생혈관형성 억제 효과( Inhibitory effect on neovascularization in cells inin vitroin vitro ))
본 실험에서는 네퍼린의 신생혈관형성 억제 효과를 평가하기 위하여 튜브 형성 에세이(tube formation assay)를 실시하였다.In this experiment, a tube formation assay was performed to evaluate the effect of nephrine inhibition of angiogenesis.
Lab-Tek 챔버 슬라이드 (Thermo Fisher Scientific, NY, USA)는 웰당 200μl Matrigel (BD Biosciences)로 코팅하였으며 고화시키기 위하여 37℃에서 30분 동안 반응시켰다. 인간제대정맥내피세포(Human Umbilical Vein Endothelial Cell, HUVECs)들은 Matrigel 상에 웰 당 5×104의 밀도로 분주하였으며, 5% CO2, 37℃에서 18시간 동안 다양한 네퍼린 농도를 포함하는 배양 배지에서 인큐베이션되었다. 튜브 형성은 phase contrast inverted microscopy (Olympus CKX41; Olympus Optical Co. Ltd, Tokyo, Japan)를 이용하여 ×40 배율에서 관찰하였다. 튜브 형성 이미지 분석은 the Wimasis Image Analysis software (Wimasis GmbH, Munich, Germany)를 이용하여 수행되었다.Lab-Tek chamber slides (Thermo Fisher Scientific, NY, USA) were coated with 200 μl Matrigel per well (BD Biosciences) and reacted for 30 minutes at 37 ° C. to solidify. Human Umbilical Vein Endothelial Cells (HUVECs) were dispensed at a density of 5 × 10 4 per well on Matrigel and cultured at 5% CO 2 , at 37 ° C. for 18 hours at varying nepherin concentrations. Incubated at Tube formation was observed at × 40 magnification using phase contrast inverted microscopy (Olympus CKX41; Olympus Optical Co. Ltd, Tokyo, Japan). Tube formation image analysis was performed using the Wimasis Image Analysis software (Wimasis GmbH, Munich, Germany).
그 결과 도 12에서 나타낸 바와 같이, 튜브 길이(px)가 네퍼린 농도에 의존적으로 감소되는 것을 확인할 수 있었다. 자세하게는, 네퍼린 처리한 실험군에서 농도에 의존하여 세포 혈관 구조의 형성의 억제가 일어나는 것을 관찰할 수 있었으며(도 12A 참조), 이러한 튜브 형성 정도를 총 관(튜브) 길이를 Wimasis Image Aalysis 소프트웨어를 이용하여 퍼센트로 변환한 결과, 네퍼린을 처리하지 않은 실험군에서는 222로 나타난 반면, 네퍼린을 10μM 농도로 처리한 실험군에서 216, 네퍼린을 20μM 농도로 처리한 실험군에서 202, 네퍼린을 30μM 농도로 처리한 실험군에서 146으로 감소되는 것을 확인할 수 있었다(도 12B 참조). As a result, as shown in Figure 12, it was confirmed that the tube length (px) is reduced depending on the neferrin concentration. In detail, it was observed that the suppression of the formation of cellular vascular structures occurred in the neferrin-treated experimental group (see FIG. 12A), and the degree of tube formation was determined using the Wimasis Image Aalysis software. As a result of the conversion to percent, 222 was shown in the experimental group not treated with nephrin, while 216 in the experimental group treated with 10 μM concentration of neferrin, 202 in the experimental group treated with 20 μM concentration of neferrin, and 30 μM concentration of neferin In the experimental group treated with it was confirmed that the reduction to 146 (see Fig. 12B).
<실험예 7>Experimental Example 7
네퍼린의Neferrin 간암세포에 대한 세포이동 억제 효과 Cell migration inhibitory effect on liver cancer cells
본 실험에서는 네퍼린의 간암세포에 대한 항암 효과로서 세포이동 억제를 유도할 수 있는지를 확인하기 위하여, 네퍼린을 간암세포주인 Hep3B에 처리한 후 암세포의 이동 정도를 상처 치유 분석(wound-healing assay)을 실시하였다.In this experiment, to determine whether nephrine can induce cell migration as an anticancer effect on hepatocellular carcinoma cells, wound-healing assay was performed after treating neferin to Hep3B, a hepatic cancer cell line. ) Was performed.
Hep3B(각 웰 당 3.5×104 cells) 세포를 50μm 격막에 의해 분리된 2개의 reservoirs로 구성된 IBIDI culture insert (Ibidi GmbH, M, Germany) 챔버 내에 분주한 후 5% CO2, 37℃ 조건에서 24시간 동안 인큐베이션시켰다. 인큐베이션 후, 내용물을 제거한 후 세포들을 수거하여 배양배지에서 다시 배양하였다. 세포 이동은 도립현미경을 이용하여 네퍼린 처리 후 12시간 간격으로 측정되었다. 이미지들을 Wimasis Image Analysis software를 이용하여 cell-covered area의 퍼센트로 산출하였다.Hep3B (3.5 × 10 4 cells per well) cells were dispensed into an IBIDI culture insert (Ibidi GmbH, M, Germany) chamber consisting of two reservoirs separated by a 50 μm septum, followed by 5% CO 2 at 24 ° C. Incubate for hours. After incubation, the contents were removed and the cells harvested and re-cultured in culture medium. Cell migration was measured at 12 hour intervals after neferrin treatment using an inverted microscope. Images were calculated as percentage of cell-covered area using Wimasis Image Analysis software.
그 결과 도 13에서 나타낸 바와 같이, 네퍼린 처리 농도에 의존하여 세포이동이 억제되는 것을 확인할 수 있었다. 특히 네퍼린을 처리하지 않은 무처리군에서 세포 이동이 증대되는 것과 비교하여, 네퍼린을 15μM 농도로 처리한 후 72시간 반응시킨 실험군에서는 cell covered area가 약 20% 가까이 감소되는 것을 확인할 수 있었다(도 13B).As a result, as shown in FIG. 13, it was confirmed that cell migration was suppressed depending on the neferrin treatment concentration. In particular, the cell coverage area was reduced by about 20% in the experimental group treated with neferrin at a concentration of 15 μM after 72 hours of treatment, compared to the increase in cell migration in the untreated group without nephrin treatment. 13B).
이제까지 본 발명에 대하여 그 바람직한 실시예들을 중심으로 살펴보았다. 본 발명이 속하는 기술 분야에서 통상의 지식을 가진 자는 본 발명이 본 발명의 본질적인 특성에서 벗어나지 않는 범위에서 변형된 형태로 구현될 수 있음을 이해할 수 있을 것이다. 그러므로 개시된 실시예들은 한정적인 관점이 아니라 설명적인 관점에서 고려되어야 한다. 본 발명의 범위는 전술한 설명이 아니라 특허청구범위에 나타나 있으며, 그와 동등한 범위 내에 있는 모든 차이점은 본 발명에 포함된 것으로 해석되어야 할 것이다.So far I looked at the center of the preferred embodiment for the present invention. Those skilled in the art will appreciate that the present invention can be implemented in a modified form without departing from the essential features of the present invention. Therefore, the disclosed embodiments should be considered in descriptive sense only and not for purposes of limitation. The scope of the present invention is shown in the claims rather than the foregoing description, and all differences within the scope will be construed as being included in the present invention.
부호의 설명Explanation of the sign
HCC: hepatocellular carcinomaHCC: hepatocellular carcinoma
CDKs: Cyclin-dependent kinasesCDKs: Cyclin-dependent kinases
ER: endoplasmic reticulumER: endoplasmic reticulum
HBV: hepatitis B virusHBV: hepatitis B virus
DMSO: Dimethyl sulfoxideDMSO: Dimethyl sulfoxide
BSA: bovine serum albuminBSA: bovine serum albumin
HUVECs: human umbilical vein endothelial cellsHUVECs: human umbilical vein endothelial cells
WST-1:2-(4-iodophenyl)-3-(4-nitrophenyl)-5-(2,4-disulfophenyl)-2H-tetrazolium, monosodium saltWST-1: 2- (4-iodophenyl) -3- (4-nitrophenyl) -5- (2,4-disulfophenyl) -2H-tetrazolium, monosodium salt
DAPI: 4, 6-diamidino-2-phenylindole dihydrochloride hydrateDAPI: 4, 6-diamidino-2-phenylindole dihydrochloride hydrate
GFP: Green fluorescent proteinGFP: Green fluorescent protein
PBS: phosphate-buffered salinePBS: phosphate-buffered saline
DFF: DNA fragmentation factorDFF: DNA fragmentation factor
GAPDH: glyceraldehyde-3-phosphate dehydrogenaseGAPDH: glyceraldehyde-3-phosphate dehydrogenase
PARP: poly (ADP-ribose) polymerasePARP: poly (ADP-ribose) polymerase
PDI: protein disulfide isomerasePDI: protein disulfide isomerase
LC3B: light chain 3 BLC3B: light chain 3 B
SirT6: sirtuin-6SirT6: sirtuin-6
PCD: programmed cell deathPCD: programmed cell death
MOMP: mitochondrial outer membrane permeabilizationMOMP: mitochondrial outer membrane permeabilization
본 발명의 네퍼린은 간암을 예방 또는 치료할 수 있는 물질로서 약리학적 조성물과 같은 의약품은 물론이고, 건강기능식품 등으로 활용할 수 있어 식품산업 및 의약산업에 응용될 경우 그 가치가 매우 높을 것으로 기대된다. 또한 본 발명의 네퍼린(Neferine)은 특정 간암세포 외에는 세포독성을 보이지 않기 때문에, 이를 유효성분으로 포함하는 본 발명의 조성물은 장기적 사용에도 안전한 이점을 가진다.Neferin of the present invention is a substance capable of preventing or treating liver cancer, as well as pharmaceuticals such as pharmacological compositions, and can be used as a health functional food, etc. It is expected that its value will be very high when applied to food industry and pharmaceutical industry . In addition, since the neferine of the present invention does not show cytotoxicity except certain liver cancer cells, the composition of the present invention comprising the same as an active ingredient has a safe advantage even for long-term use.

Claims (6)

  1. 하기 화학식 1로 표시되는 네퍼린(Neferine)을 유효성분으로 포함하는 간암 예방 또는 치료용 약제학적 조성물;A pharmaceutical composition for preventing or treating liver cancer comprising Neferine represented by Formula 1 as an active ingredient;
    <화학식 1><Formula 1>
    Figure PCTKR2013008639-appb-I000002
    Figure PCTKR2013008639-appb-I000002
  2. 제1항에 있어서,The method of claim 1,
    상기 네퍼린(Neferine)은 연꽃 배아로부터 유래된 것을 특징으로 하는 간암 예방 또는 치료용 약제학적 조성물.The neferine (Neferine) is a pharmaceutical composition for preventing or treating liver cancer, characterized in that derived from the lotus embryo.
  3. 제1항에 있어서,The method of claim 1,
    상기 간암은 바이러스감염에 의해 유발되는 간세포암(hepatocellular carcinoma)인 것을 특징으로 하는 간암 예방 또는 치료용 약제학적 조성물.The liver cancer is hepatocellular carcinoma (hepatocellular carcinoma) caused by viral infection, characterized in that for preventing or treating liver cancer.
  4. 제1항 내지 제3항 중 어느 한 항에 있어서,The method according to any one of claims 1 to 3,
    상기 약제학적 조성물은 암세포의 세포주기중지(cell cycle arrest), 세포사멸(apoptosis) 유도, 자가포식(autophagy) 유도, 신생혈관형성 억제 및 세포이동 억제 활성을 통해 항암 효과를 가지는 것을 특징으로 하는 간암 예방 또는 치료용 약제학적 조성물.The pharmaceutical composition is characterized in that it has anticancer effect through cell cycle arrest, induction of apoptosis, induction of autophagy, inhibition of angiogenesis and cell migration of cancer cells. Prophylactic or therapeutic pharmaceutical compositions.
  5. 하기 화학식 1로 표시되는 네퍼린(Neferine)을 유효성분으로 포함하는 간암의 예방 또는 개선용 건강기능식품;Health functional food for the prevention or improvement of liver cancer comprising Neferine (Neferine) represented by the formula (1) as an active ingredient;
    <화학식 1><Formula 1>
    Figure PCTKR2013008639-appb-I000003
    Figure PCTKR2013008639-appb-I000003
  6. 제5항에 있어서, The method of claim 5,
    상기 식품은 음료류, 육류, 초코렛, 식품류, 과자류, 피자, 라면, 기타 면류, 껌류, 사탕류, 아이스크림류, 알코올 음료류, 비타민 복합제 및 건강보조식품류로 이루어진 군으로부터 선택되는 것을 특징으로 하는 건강기능식품.The food is a health functional food, characterized in that selected from the group consisting of beverages, meat, chocolate, food, confectionery, pizza, ramen, other noodles, gum, candy, ice cream, alcoholic beverages, vitamin complexes and health supplements.
PCT/KR2013/008639 2012-09-26 2013-09-26 Pharmaceutical composition comprising neferine as active ingredient for preventing or treating hepatoma WO2014051359A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
KR10-2012-0106857 2012-09-26
KR1020120106857A KR101418161B1 (en) 2012-09-26 2012-09-26 Pharmaceutical compositions for prevention or treatment of liver cancer comprising neferine

Publications (1)

Publication Number Publication Date
WO2014051359A1 true WO2014051359A1 (en) 2014-04-03

Family

ID=50388653

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2013/008639 WO2014051359A1 (en) 2012-09-26 2013-09-26 Pharmaceutical composition comprising neferine as active ingredient for preventing or treating hepatoma

Country Status (2)

Country Link
KR (1) KR101418161B1 (en)
WO (1) WO2014051359A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017142283A1 (en) * 2016-02-15 2017-08-24 서울대학교산학협력단 Composition for treating or preventing liver cancer.

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102348878B1 (en) * 2014-08-13 2022-01-06 주식회사 엘지생활건강 Cosmetic or pharmaceutical composition for skin whitening, elasticity, anti-wrinkle, or skin moisturizing comprising neferine or a pharmaceutically acceptable salt thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20030031729A (en) * 2001-10-16 2003-04-23 학교법인 원광학원 Extract of the seeds of nelumbo nucifera gaertn. having liver cell protective effect and liver injury preventive or therapeutic effect, and a composition containing same
US20090214683A1 (en) * 2008-02-27 2009-08-27 Chau-Jong Wang Extracts of Sacred Water Lotus for the Treatment of Cancer
US20120108629A1 (en) * 2009-04-14 2012-05-03 Institute of Chinese Materia Medica, China Academy New use of neferine

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20030031729A (en) * 2001-10-16 2003-04-23 학교법인 원광학원 Extract of the seeds of nelumbo nucifera gaertn. having liver cell protective effect and liver injury preventive or therapeutic effect, and a composition containing same
US20090214683A1 (en) * 2008-02-27 2009-08-27 Chau-Jong Wang Extracts of Sacred Water Lotus for the Treatment of Cancer
US20120108629A1 (en) * 2009-04-14 2012-05-03 Institute of Chinese Materia Medica, China Academy New use of neferine

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
YOON, J.-S. ET AL.: "Neferine isolated from Nelumbo nucifera enhances anti-cancer activities in Hep3B cells: molecular mechanisms of cell cycle arrest, ER stress induced apoptosis and anti-angiogenic response", PHYTOMEDICINE, vol. 20, 15 August 2013 (2013-08-15), pages 1013 - 1022 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017142283A1 (en) * 2016-02-15 2017-08-24 서울대학교산학협력단 Composition for treating or preventing liver cancer.
CN109069513A (en) * 2016-02-15 2018-12-21 首尔大学校产学协力团 For treating or preventing the composition of liver cancer
CN109069513B (en) * 2016-02-15 2021-12-28 硕氏医科有限公司 Composition for treating or preventing liver cancer

Also Published As

Publication number Publication date
KR20140040359A (en) 2014-04-03
KR101418161B1 (en) 2014-07-09

Similar Documents

Publication Publication Date Title
WO2017078405A9 (en) Pharmaceutical composition for treatment of lung cancer comprising glucocorticoid-based compound
WO2012008788A2 (en) Composition containing serine as an active ingredient for the prevention and treatment of fatty liver diseases, and use thereof
WO2024039164A1 (en) Composition for preventing, ameliorating, or treating cancer comprising steppogenin as active ingredient
WO2014051359A1 (en) Pharmaceutical composition comprising neferine as active ingredient for preventing or treating hepatoma
US20100297760A1 (en) Pharmaceutical composition and a method for treatment of prostate cancer
WO2018155921A1 (en) Pharmaceutical composition for preventing and treating pancreatic cancer, containing gossypol and phenformin as active ingredients
WO2020071795A1 (en) Anticancer pharmaceutical composition containing if1 as active ingredient
WO2020166779A1 (en) Composition for fat formation inhibition and body fat reduction, containing hydrangenol as active ingredient
WO2020122391A1 (en) Composition for preventing or treating cellular senescence-associated diseases, containing homoharringtonine as active ingredient
WO2021167175A1 (en) Pharmaceutical composition for preventing or treating cancer, comprising mtor-signaling inhibitor as active ingredient
WO2014168458A1 (en) Use of compounds isolated from morus bark
WO2021071334A1 (en) Pharmaceutical composition for preventing and treating type 2 diabetes, containing quercetin-3-o-xyloside
KR101720610B1 (en) The pharmaceutical composition Inula helenium hexane fractions thereof or compound isolated from the fraction comprising inhibitory activity of STAT for prevention or treatment of breast cancer
WO2016159627A1 (en) Peptide having anticancer activity, and pharmaceutical composition and dietary supplement composition for preventing and treating cancer, both of which contain same as active ingredient
WO2020032452A1 (en) Composition comprising gold compound as effective ingredient for inhibiting osteoclast differentiation
KR20210004133A (en) Compositions for preventing or treating fatty liver disease comprising Allomyrina dichotoma larva extract
WO2020122393A1 (en) Composition for preventing or treating cellular senescence-related diseases comprising salinomycin as effective component
WO2017222252A1 (en) Pharmaceutical composition for preventing or treating colon cancer, containing extract of telectadium dongnaiense as active ingredient
WO2023171998A1 (en) Saikosaponin c as composition for treating or preventing aging vascular diseases including arteriosclerosis
WO2022191656A1 (en) Novel indolizine derivative and composition for prevention or treatment of cancer comprising same
WO2022025709A1 (en) Preventive, relief or therapeutic use of 2,3,5-substituted thiophene compound against gastrointestinal stromal tumor
WO2022080729A1 (en) Composition for preventing or treating lung cancer, comprising compound isolated from cephalotaxaceae extract as active ingredient
WO2011059294A2 (en) Anticancer and immunity-enhancing composition containing morphinan alkaloids as active ingredients
WO2024058551A1 (en) Pharmaceutical composition for treating liver cancer comprising cannabidiol and anticancer agent as active ingredients and use thereof
Yagubova et al. Dimeric NGF mimetic attenuates hyperglycaemia and DNA damage in mice with streptozotocin-induced early-stage diabetes

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 13841897

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13841897

Country of ref document: EP

Kind code of ref document: A1