CN116694721B - Ganoderma lucidum glycopeptide and preparation method and application thereof, and ganoderma lucidum glycopeptide microemulsion and face cream - Google Patents

Ganoderma lucidum glycopeptide and preparation method and application thereof, and ganoderma lucidum glycopeptide microemulsion and face cream Download PDF

Info

Publication number
CN116694721B
CN116694721B CN202310993467.XA CN202310993467A CN116694721B CN 116694721 B CN116694721 B CN 116694721B CN 202310993467 A CN202310993467 A CN 202310993467A CN 116694721 B CN116694721 B CN 116694721B
Authority
CN
China
Prior art keywords
ganoderma lucidum
glycopeptide
ganoderma
microemulsion
skin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202310993467.XA
Other languages
Chinese (zh)
Other versions
CN116694721A (en
Inventor
金叶
刘达
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Hanning Zhuhai Biotechnology Co ltd
Changchun University of Chinese Medicine
Original Assignee
Hanning Zhuhai Biotechnology Co ltd
Changchun University of Chinese Medicine
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 Hanning Zhuhai Biotechnology Co ltd, Changchun University of Chinese Medicine filed Critical Hanning Zhuhai Biotechnology Co ltd
Priority to CN202310993467.XA priority Critical patent/CN116694721B/en
Publication of CN116694721A publication Critical patent/CN116694721A/en
Application granted granted Critical
Publication of CN116694721B publication Critical patent/CN116694721B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/06Preparation of peptides or proteins produced by the hydrolysis of a peptide bond, e.g. hydrolysate products
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/02Cosmetics or similar toiletry preparations characterised by special physical form
    • A61K8/04Dispersions; Emulsions
    • A61K8/06Emulsions
    • A61K8/068Microemulsions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/64Proteins; Peptides; Derivatives or degradation products thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • A61Q19/08Anti-ageing preparations
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/145Extraction; Separation; Purification by extraction or solubilisation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/34Extraction; Separation; Purification by filtration, ultrafiltration or reverse osmosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/36Extraction; Separation; Purification by a combination of two or more processes of different types
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2800/00Properties of cosmetic compositions or active ingredients thereof or formulation aids used therein and process related aspects
    • A61K2800/80Process related aspects concerning the preparation of the cosmetic composition or the storage or application thereof
    • A61K2800/805Corresponding aspects not provided for by any of codes A61K2800/81 - A61K2800/95
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2800/00Properties of cosmetic compositions or active ingredients thereof or formulation aids used therein and process related aspects
    • A61K2800/80Process related aspects concerning the preparation of the cosmetic composition or the storage or application thereof
    • A61K2800/82Preparation or application process involves sonication or ultrasonication
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2800/00Properties of cosmetic compositions or active ingredients thereof or formulation aids used therein and process related aspects
    • A61K2800/80Process related aspects concerning the preparation of the cosmetic composition or the storage or application thereof
    • A61K2800/84Products or compounds obtained by lyophilisation, freeze-drying
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Analytical Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Birds (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Epidemiology (AREA)
  • General Engineering & Computer Science (AREA)
  • Water Supply & Treatment (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biotechnology (AREA)
  • Dispersion Chemistry (AREA)
  • Gerontology & Geriatric Medicine (AREA)
  • Dermatology (AREA)
  • Cosmetics (AREA)

Abstract

Ganoderma glycopeptide, its preparation method and application, and Ganoderma glycopeptide microemulsion and facial cream, and relates to the technical field of protein extraction, which improves Ganoderma application value and solves skin photoaging problem. Pulverizing dried Ganoderma fruiting body into coarse powder, mixing with pure water, heating for extraction, performing ultrasonic extraction, and mixing the supernatants to obtain coarse extract of Ganoderma glycopeptide; removing free protein, and dialyzing with dialysis bag; enzymolysis, eluting with wheat germ lectin resin until the molecular weight cut-off is less than 10000, and freeze drying to obtain Ganoderma glycopeptide lyophilized powder. The ganoderma lucidum glycopeptide and water are prepared into a water phase, a zwitterionic surfactant and a cosurfactant are mixed according to the proportion, then the mixture is mixed with an oil phase, and the water phase is added dropwise under magnetic stirring to obtain the ganoderma lucidum glycopeptide microemulsion. The invention can be applied to the preparation of products for resisting oxidation or skin photoaging.

Description

Ganoderma lucidum glycopeptide and preparation method and application thereof, and ganoderma lucidum glycopeptide microemulsion and face cream
Technical Field
The invention relates to the technical field of protein extraction, in particular to a ganoderma lucidum glycopeptide and a preparation method and application thereof, and ganoderma lucidum glycopeptide microemulsion and face cream.
Background
Skin is the first line of defense of human body to the external environment, and in daily life, normal skin tissue structure and function are extremely important for body health. Ultraviolet rays, blue light, ozone, etc., which are exposed to the external environment, cause a series of premature aging phenomena of the skin, and ultraviolet rays are the factors which are most easily contacted by people. Ultraviolet rays in sunlight are mainly classified into three types, namely ultraviolet rays A (UVA), ultraviolet rays B (UVB) and ultraviolet rays C (UVC), wherein UVA (320-400 nm) and UVB (275-320 nm) have strong penetrating power, can directly reach the epidermis layer and dermis layer of skin, and are main causes of photoaging of skin.
Glycopeptides are binding proteins formed by connecting an oligosaccharide chain and a polypeptide chain through a covalent bond, and can be divided into two main types, namely N-glycosidic bonds and O-glycosidic bonds according to different glycosidic bonds formed by dehydration of hydroxyl groups on the carbon atoms of the hetero heads of the glycosyl groups and amide groups or hydroxyl groups on amino acid residues of the peptide chains. Glycopeptides are widely present in animals, plants and microorganisms, and are indispensable biological macromolecules for vital phenomena such as cell communication, immune regulation and the like in organisms. Because of the properties of both protein and sugar, the protein has various biological activities, such as antioxidation, anti-tumor, blood sugar and blood lipid reduction, organism immunity improvement and the like.
Ganoderma, also called Ganoderma, herba Mesonae chinensis, rumex, herba Hedyotidis Diffusae, herba Linderae Caesalpiniae, herba Cannabifoliae, and herba Cannabifoliae, is a fungus of Polyporaceae, and is widely distributed in nature, and its cultivation area is mainly distributed in southeast coast, middle and downstream of Yangtze river, and great Khingan area. The fresh ganoderma lucidum is rich in six nutritional elements of protein, carbohydrate, fat, vitamins, inorganic matters and water, also contains components such as ganoderma lucidum adenosine, ganoderma lucidum total alkali and the like, and is a fungus medicament which contains rich nutritional components and various development potentials. The ganoderma lucidum has wide application, can resist radiation, aging and oxidization, and has obvious effects of enhancing human immunity, regulating blood sugar, controlling blood pressure, protecting liver, promoting sleep and the like.
Unlike ganoderan (GL-PS), less research is carried out on ganoderan peptide at present, and patent CN1629186A (ganoderan protein, preparation method and application thereof) discloses ganoderan protein Gl-PP obtained by extracting from ganoderma, which is obtained by taking ganoderma as a raw material matrix to extract liquid, concentrating the extract liquid, and then, carrying out ultrafiltration or dialysis to intercept high molecular weight polysaccharide protein; also relates to a preparation method of the ganoderan protein capable of being industrially produced and application of the ganoderan protein Gl-PP in preparing anti-tumor drugs. The ultrafiltration method provided by the patent can be used for extracting ganoderan protein, so that the angiogenesis of tumor and the invasion of cancer cells can be inhibited, and the growth of tumor can be inhibited. Patent CN1715298 discloses that the crude product of ganoderma tsugae proteoglycan is a mixture containing three single proteoglycans P1, P2 and P3, the polysaccharide parts of the crude product are uniform glucosan, and the glucosidic bonds are beta- (1-3) configuration, which has the physiological effects of improving the immune function of the organism, resisting cell mutation, reducing blood fat and blood sugar, protecting liver, soothing the nerves, calming the nerves, resisting tumors and the like; through molecular biology and cell pharmacology research, it can regulate cell division and differentiation, and regulate cell growth and death. Is used for preparing antitumor drugs. In summary, the currently disclosed ganoderma lucidum glycopeptides are mostly used for anti-tumor aspects, and do not relate to the application in anti-aging and skin photoaging directions.
Disclosure of Invention
In order to further improve the use value of ganoderma lucidum and solve the problems of skin photoaging and the like, the invention provides ganoderma lucidum glycopeptide, a preparation method and application thereof, and ganoderma lucidum glycopeptide microemulsion and face cream.
The technical scheme of the invention is as follows:
a preparation method of ganoderma lucidum glycopeptides comprises the following steps:
s1, taking dried ganoderma lucidum fruiting bodies and crushing the dried ganoderma lucidum fruiting bodies into ganoderma lucidum coarse powder;
s2, mixing the ganoderma lucidum coarse powder with pure water according to 1g: uniformly mixing (50-70) mL of feed liquid ratio by shaking through a shaking table, heating and extracting for 10-12 h at 60-80 ℃, then performing ultrasonic extraction for at least 3 times for 15-30 min each time, and combining the supernatant to obtain ganoderma lucidum glycopeptide crude extract;
s3, removing free protein from the ganoderma lucidum glycopeptide crude extract, and then carrying out dialysis treatment by using a dialysis bag;
s4, carrying out enzymolysis on the dialyzed ganoderma lucidum glycopeptide, eluting with wheat germ lectin resin until the molecular weight cutoff is less than 10000, and freeze-drying to obtain ganoderma lucidum glycopeptide freeze-dried powder.
Preferably, the shaking table oscillates at a rate of 120 r/min-180 r/min.
Preferably, the removal of the free protein in step S3 is specifically performed by the Sevage method; the dialysis treatment specifically comprises the following steps: the solution was dialyzed in distilled water using a 0.1kDa dialysis bag for 48 hours.
Preferably, the enzymolysis is specifically: treating dialyzed ganoderma lucidum glycopeptides in a water bath at 50-60 ℃ for 30-35 min, adding trypsin with an enzyme-to-bottom ratio of 4%, magnetically stirring for 5-6 h, then placing in a constant-temperature water bath at 80-85 ℃ for 15-20 min, and finally cooling to room temperature.
Preferably, the temperature of the freeze-drying in step S4 is-86 ℃ and the time of the freeze-drying is at least 48 hours.
The invention also provides the ganoderma lucidum glycopeptide which is prepared by the preparation method, and the combination mode of the glycopeptide bond of the ganoderma lucidum glycopeptide is O-glycosidic bond.
The invention also provides application of the ganoderma lucidum glycopeptide, which is used for preparing an antioxidant or skin photo-aging resistant skin care product.
The invention also provides a ganoderma lucidum glycopeptide microemulsion which is prepared by using the ganoderma lucidum glycopeptide as the raw material.
The preparation method of the ganoderma lucidum glycopeptide microemulsion comprises the following steps:
the ganoderma lucidum glycopeptide and water are prepared into a water phase with the concentration of the medicine of 10 mg/mL-20 mg/mL, and a zwitterionic surfactant and a cosurfactant are mixed according to the proportion of 1: mixing (0.5-2) by mass ratio, mixing with oil phase, magnetically stirring at 23-27 ℃ and 500-1000 rpm, and dropwise adding water phase to obtain the ganoderma lucidum glycopeptide microemulsion.
Preferably, the zwitterionic surfactant is soybean phospholipid, the cosurfactant is ethanol, and the oil phase is isopropyl myristate.
The invention also provides a face cream which comprises the ganoderma lucidum glycopeptide microemulsion.
Compared with the prior art, the invention has the following specific beneficial effects:
1. the extraction method of the ganoderma lucidum glycopeptides is simple to operate, and the obtained ganoderma lucidum glycopeptides are high in extraction rate and stable in structure, so that the use value of ganoderma lucidum medicinal materials is further improved, and the development channel of the novel cosmetic field is expanded;
2. the ganoderma lucidum glycopeptide (GL-Gp) provided by the invention can slow down skin aging caused by ultraviolet rays, improve the activity and HYP content of SOD, GSH-Px in skin tissues, and reduce the content of MDA, MMP-1, MMP-3, IL-1 beta, IL-6 and TNF-alpha in skin tissues; pharmacological experiments show that the composition has remarkable antioxidant and skin photoaging repairing effects, and can be used for research and application of photoaging-resistant skin care products.
Drawings
FIG. 1 is a graph of ultraviolet spectra of ganoderma lucidum glycopeptides before and after alkali treatment;
FIG. 2 is an infrared spectrum of ganoderma lucidum glycopeptides;
FIG. 3 is an SDS-PAGE electrophoresis of ganoderma lucidum glycopeptides;
FIG. 4 is a diagram showing the amino acid composition of ganoderan;
fig. 5 is a pseudo ternary phase diagram of each oil phase with mixed surfactant when km=1:1;
FIG. 6 is a pseudo ternary phase diagram of isopropyl myristate and mixed surfactant formation at different Km;
FIG. 7 is a pseudo-ternary phase diagram at different drug concentrations;
FIG. 8 is a contour plot of factors A, B, C versus OD;
FIG. 9 is a three-dimensional effect plane graph of the factors A, B, C versus OD values;
FIG. 10 is a flow chart of a process for preparing the ganoderma lucidum glycopeptide microemulsion cream;
FIG. 11 is a graph showing the moisture retention of a ganoderma lucidum glycopeptide microemulsion face cream, wherein A is a glycerol group, B is a blank group, C is sample 1, D is sample 2, E is sample 3, F is sample 4, and G is sample 5;
FIG. 12 is an external view of a ganoderma lucidum glycopeptide microemulsion cream;
FIG. 13 is a comparison of the appearance of a stability test, wherein, A is 25 ℃ for 24 hours, B is 40 ℃ for 24 hours, C is-20 ℃ for 24 hours, and D is after cold and heat resistance acceleration test;
FIG. 14 is a graph showing the morphological effects of ganoderan on the photoaging appearance of rats;
FIG. 15 is a HE staining chart of a pathological section of the photoaged skin of a rat;
FIG. 16 is a graph of aldehyde fuchsin staining of pathological sections of photoaged skin of rats;
FIG. 17 is a graph showing the effect of ganoderan on MDA content in photoaged skin tissue of rats;
FIG. 18 is a graph showing the effect of ganoderan on SOD activity in rat skin tissue;
FIG. 19 is a graph showing the effect of ganoderan on GSH-Px content in rat skin tissue;
FIG. 20 is a graph showing the effect of ganoderan on HYP content in rat skin tissue;
FIG. 21 is a graph showing the effect of ganoderan on MMP-1 content in rat skin tissue;
FIG. 22 is a graph showing the effect of ganoderan on MMP-3 content in rat skin tissue;
FIG. 23 is a graph showing the effect of ganoderan on IL-6 content in rat skin tissue;
FIG. 24 is a graph showing the effect of ganoderan on IL-1β content in rat skin tissue;
FIG. 25 is a graph showing the effect of ganoderan on TNF- α levels in rat skin tissue.
Detailed Description
In order to make the technical solution of the present invention clearer, the technical solution of the embodiments of the present invention will be clearly and completely described below with reference to the accompanying drawings of the present invention, and it should be noted that the following embodiments are only used for better understanding of the technical solution of the present invention, and should not be construed as limiting the present invention.
Example 1.
In the embodiment, the ganoderma lucidum glycopeptide GL-Gp is extracted, and the specific operation is as follows:
pulverizing dried fruiting body of Ganoderma, mixing with 54mL pure water at shaking table vibration rate of 167r/min, heating at 71 deg.C for extracting for 12 hr, ultrasonic extracting for 30min for 3 times, and mixing the supernatants to obtain coarse extract of Ganoderma glycopeptide;
removing free protein from the obtained crude extract of ganoderma lucidum glycopeptide by adopting a Sevage method to obtain ganoderma lucidum glycopeptide solution, placing the obtained ganoderma lucidum glycopeptide solution in a dialysis bag of 0.1kDa, dialyzing for 48h, carrying out enzymolysis on the dialyzate, treating for 35min in a water bath at 60 ℃, adding trypsin with the enzyme bottom ratio of 4%, magnetically stirring for 6h, then placing in a constant-temperature water bath at 85 ℃ for 20min, cooling to room temperature, eluting with wheat germ lectin resin until the molecular weight cutoff is less than 10000, and freeze-drying to obtain ganoderma lucidum glycopeptide powder.
The ganoderma lucidum glycopeptides obtained in this example are characterized as follows:
(1) Beta-elimination experiment:
weighing 2mg of ganoderma lucidum glycopeptide, dissolving in 10ml of 0.4mol/L NaOH, carrying out water bath for 30min at 60 ℃, and scanning at 190-350 nm by using an ultraviolet spectrophotometer; the ganoderma lucidum glycopeptides which were not treated with 0.4mol/L NaOH were scanned under the same conditions. The results are shown in FIG. 1, wherein curve A represents the glossy ganoderma glycopeptides treated by NaOH, curve B represents the glossy ganoderma glycopeptides not treated by NaOH, and the light absorption value of the glossy ganoderma glycopeptides solution treated by NaOH is increased at the wavelength of 240nm, which indicates that polysaccharide and protein in the glossy ganoderma glycopeptides are connected in a covalent bond mode, and the connection mode is O-glycosidic bond.
(2) And (3) infrared spectrum analysis:
grinding small amount of Ganoderma glycopeptide and KBr, tabletting, and infrared scanning to obtain scanning result shown in figure 2, which can be seen in 3300 and 3300 cm -1 、2900 cm -1 、1640 cm -1 、1400 c cm -1 And 1000 cm -1 Absorption peaks are arranged on the left and right sides, which indicates that polysaccharide and protein molecules are contained; at 2930 cm -1 There is an absorption, characteristic absorption peaks indicative of methyl, methylene and other alkyl vibrations; amide at 1640 and 1640 cm -1 Has absorption peak, and the stretching vibration of sulfuric acid group is 1400 cm -1 At 1050 cm -1 The presence of an absorption peak indicates the presence of a pyranose configuration in the sugar chain.
The above results demonstrate that the present example successfully extracts ganoderma lucidum glycopeptides having O-glycosidic bonds as the binding mode of glycopeptides.
(3) Molecular weight determination:
the relative molecular weight of the obtained ganoderma lucidum glycopeptides was determined by gel electrophoresis in combination with coomassie brilliant blue dye pairs, and the result is shown in fig. 3. It is obvious that after SDS polyacrylamide gel electrophoresis separation, the ganoderma lucidum glycopeptides contain proteins with different molecular weights and are mainly distributed in 55 kD-70 kD, 35 kD-55 kD and 15 kD-25 kD.
(4) Analysis of amino acid composition results:
further determination of the amino acid composition of Gl-Gp using an amino acid analyzer, the results are shown in FIG. 4, and it can be seen that Gl-Gp consists of 18 amino acids: asp (5.86 mg.g) -1 ),Glu(4.08mg·g -1 ),Ser(2.65mg·g -1 ),His(0.23mg·g -1 ),Gly(1.95mg·g -1 ),Thr(1.36mg·g -1 ),Arg(0.54mg·g -1 ),Ala(2.35mg·g -1 ),Tyr(0.28mg·g -1 ),Cys(0.64mg·g -1 ),Val(0.85mg·g -1 ),Met(0.049mg·g -1 ),Phe(0.83mg·g -1 ),Ile(0.55mg·g -1 ),Lys(0.43mg·g -1 ),Leu(1.45mg·g -1 ),Pro(1.32mg·g -1 ),Trp(1.01mg·g -1 )。
(5) Protein content in ganoderma lucidum glycopeptides was determined using BCA method:
preparing a protein standard solution of 0.5mg/mL, respectively sucking 0 mu L, 1 mu L, 2 mu L, 4 mu L, 8 mu L, 12 mu L, 16 mu L and 20 mu L, adding the protein standard solution into standard wells of a 96-well plate, adding a diluent to make up 20 mu L, adding 200 mu LBCA working solution into each well, standing for 20-30 min at 37 ℃, and measuring absorbance at 562nm wavelength. Drawing a standard curve by taking absorbance as an ordinate and the standard mass concentration of the protein as an abscissa, and obtaining a regression curve equation of y=0.8305x+0.2149, R 2 =0.9993。
(6) Acid sugar content determination:
accurately sucking 0.5mg/mL glucuronic acid quasi-product solution 0mL, 0.1mL, 0.25mL, 0.4mL, 0.5mL, 0.75mL, 1.0mL into 10mL test tubes, adding distilled water to 1.0mL, cooling in ice water bath, adding four materials2.5mL of sodium borate-sulfuric acid solution, uniformly mixing, heating in a boiling water bath for 10min, cooling to room temperature, adding 50 mu L of m-hydroxybiphenyl solution, carrying out vortex oscillation for 5min, standing at room temperature for 1h, taking distilled water as a blank group, and measuring absorbance at a wavelength of 525 nm. Drawing a standard curve by taking the concentration of D-galacturonic acid as an abscissa and the absorbance value as an ordinate to obtain a regression curve equation of y=0.001x+0.0557, R 2 =0.9952。
(7) Neutral sugar content determination:
accurately weighing 1mg of anhydrous glucose as a standard substance, fixing the volume by a volumetric flask with 25mL to prepare 40 mug/mL of standard substance solution, respectively sucking 0mL, 0.2mL, 0.4mL, 0.6mL, 0.8mL, 1.0mL, 1.2mL, 1.4mL and 1.6mL of glucose standard substance solution into a test tube, adding distilled water to supplement 2mL, adding 1.0mL of 6% phenol solution, adding 5.0mL of concentrated sulfuric acid, standing for 10min, shaking uniformly, reacting at room temperature for 20min again, and measuring absorbance at 490nm wavelength. Drawing a standard curve by taking absorbance value A as an ordinate and glucose concentration as an abscissa to obtain a regression curve equation of y=0.0096 x-0.017, R 2 =0.9962。
According to calculation, the yield of the ganoderma lucidum glycopeptides (namely the amount of the obtained glycopeptides freeze-dried powder/3 g ganoderma lucidum coarse powder) in the embodiment is 9.05 percent, and the purity is 86.12 percent (sugar content+protein content).
Example 2.
In the embodiment, the area size of the formed pseudo ternary phase diagram is assumed to be used as an investigation index, and the following single-factor investigation is carried out on the oil phase, the zwitterionic surfactant, the cosurfactant, the Km value and the drug concentration before the microemulsion is prepared:
(1) Oil phase selection:
ethyl oleate, oleic acid, isopropyl myristate, isopropyl palmitate, trioleate, castor oil, jojoba oil and neodecanoate glyceride were initially selected as the oil phase. The mass ratio of the fixed soybean phospholipids to the absolute ethyl alcohol is 1:1 (Km=1:1) as a mixed surfactant, the fixed soybean phospholipids and the absolute ethyl alcohol are mixed with each oil phase according to the mass ratio of 1:1, the water added at the critical point is recorded, the mass percentage of the water phase is calculated, and the mass percentage of the water phase when each oil phase reaches the critical point is obtained as shown in table 1.
TABLE 1
From the results, the water phase mass percentages of the triolein, the castor oil, the jojoba oil, the caprylic/capric glyceride and the oleic acid are too small, so that the triolein, the castor oil, the jojoba oil, the caprylic/capric glyceride and the oleic acid are not considered to be used as oil phases; when ethyl oleate, isopropyl myristate and isopropyl palmitate are used as oil phases to form the microemulsion, the mass percentage of the water phase is equivalent, and screening is carried out continuously through a pseudo ternary phase diagram.
When km=1:1, the pseudo-ternary phase diagram formed by each oil phase and the mixed surfactant is shown in fig. 5, and the area of the microemulsion region is compared by AutoCAD2015 software, it was found that the area of the microemulsion region formed is the largest when the oil phase is isopropyl myristate.
(2) Zwitterionic surfactant and cosurfactant selection:
the mutual dissolution of different types of zwitterionic surfactants (soybean phospholipid, span 80 and span 60) and cosurfactants (ethanol, propylene glycol, glycerol and PEG 400) is examined, the composition of the mixed surfactants is primarily screened, km=1 and isopropyl palmitate are fixed and mixed according to a certain proportion, then the water carrying capacity residual microemulsion is examined to form difficulty, and the composition of the microemulsion surfactant is determined by taking the difficulty as an index.
Through testing, span 60 is insoluble in propylene glycol, glycerol and PEG400, and after span 60 is mixed with ethanol, the water content in isopropyl myristate is 0.49%, 0.33% and 0.39%; layering phenomenon can occur after span 80 is uniformly mixed with propylene glycol, glycerol and PEG400, water is added to the span to be turbid immediately after the span 80 is mixed with oil phase, and the water content in isopropyl myristate is 3.56% after the span 80 is mixed with ethanol; the soybean phospholipid is dissolved very slowly with glycerol and PEG400, is dissolved relatively slowly with propylene glycol, is viscous after being mixed with oil phase, is not easy to observe phase inversion phenomenon, is dissolved rapidly in ethanol, has a water content of 28.15% in isopropyl myristate after being mixed with ethanol, has a high water content, and has the characteristics of good biocompatibility, safety and no toxicity. Therefore, soybean phospholipids are selected as the surfactant and ethanol is selected as the cosurfactant.
(3) Selection of Km values:
the area of the microemulsion region formed by the mixed surfactant and the preferable oil phase when the Km value is 1:2, 1:1 and 2:1 is examined respectively, and the optimal Km value is determined.
When km=2:1, 1:1, 1:2, the pseudo-ternary phase diagram of isopropyl myristate and mixed surfactant is shown in fig. 6, the areas of the micro-emulsion areas are compared by AutoCAD2015 software, and the areas of the micro-emulsion areas formed when km=1:1 are larger than km=2:1 and km=1:2, and km=1 is selected as the best.
(4) Drug concentration selection:
the sizes of the microemulsion areas formed when the concentrations are 5mg/mL, 15mg/mL and 25mg/mL are respectively examined, and the optimal drug concentration is determined according to the state of the ganoderma lucidum glycopeptide aqueous solution after centrifugation. The test shows that the pseudo-ternary phase diagram formed under different drug concentrations is shown in figure 7, and the drug concentration has little influence on the formation area of the microemulsion pseudo-ternary phase diagram, so that 15mg/mL is selected as the optimal drug concentration according to the state of the drug solution after centrifugation shown in Table 2.
TABLE 2
Drug concentration Phenomenon after centrifugation
5mg/mL No insoluble matter
15 mg/mL No insoluble matter
25 mg/mL Presence of insolubilityArticle (B)
(5) Optimally mixing and designing:
according to the single factor experimental investigation result, the preparation process is determined as follows: isopropyl myristate is used as an oil phase, the mass ratio of soybean phospholipid to absolute ethyl alcohol is 1:1 (Km=1), the drug concentration is 15mg/mL, and the sum of the three phases of water phase, oil phase and mixed surfactant is set to be 1 according to the microemulsion forming area, so that the optimal limit range of the mass fractions of each phase is obtained. The D-optimal mixing test in Design Expert 13 software is adopted, the water phase (A), the oil phase (B) and the mixed surfactant (C) are used as optimal components, and the experiment is designed to determine the optimal prescription of the ganoderma lucidum glycopeptide microemulsion, and experimental factors and levels are shown in Table 3.
TABLE 3 Table 3
Component (A) Water phase (A) Oil phase (B) Mixed surfactant (C)
Minimum value of 0.13 0.23 0.26
Maximum value of 0.27 0.61 0.51
Taking OD as a response value of process optimization, and carrying out mathematical conversion on the encapsulation efficiency EE and the drug loading DL to obtain a normalized value (d), wherein the formula is as follows:
,/>
wherein d 1 And d 2 Respectively represent glossy ganoderma glycopeptides EE and DL, Y 1 As measured value, Y max For the maximum value of EE and DL, Y mix And d values of all indexes are calculated according to a formula to obtain a total normalized value OD for the minimum value of the measured EE and DL. The experimental design and results are shown in Table 4.
TABLE 4 Table 4
Fitting and optimizing a model:
taking OD as a response value and A, B, C as a variable, the obtained fitting equation is OD=2.26A+0.0621B+1.15C-4.89 AB-1.63AC-2.24BC+21.25ABC-4.77AB (A-B) +3.19AC (A-C) +3.79BC (B-C). The model has significanceP<0.0001 No significance of the mismatching termP=0.7670>0.05),R 2 =0.9985,R adj 2 As can be seen from 0.9963, the model has a good fitting degree with the actual microemulsion prescription, the contour plot and the three-dimensional effect plot of the factors a, B, and C versus the OD values are shown in fig. 8 and 9, respectively, and the model fitting results are shown in table 5.
TABLE 5
(6) Optimal prescription and verification experiment:
according to the result, the optimal prescription of the ganoderma lucidum glycopeptide microemulsion is: the mass percent of the water phase is 0.260, the mass percent of the oil phase is 0.365, the mass percent of the mixed surfactant is 0.375, and the predicted OD value is 0.991.
The results of three verifiable experiments on the optimal prescription are shown in Table 6, and the stability of the prescription can be proved to be good.
TABLE 6
Thus, in this example, ganoderma glycopeptide was formulated with water to give an aqueous phase with a pharmaceutical concentration of 15mg/mL, and soybean phospholipid and absolute ethanol were mixed at a ratio of 1:1, mixing with isopropyl myristate, magnetically stirring at 25 ℃ at 800rpm, dropwise adding water phase to make the mass percent of the mixed water phase be 0.260, the mass percent of the oil phase be 0.365 and the mass percent of the mixed surfactant be 0.375, and stirring until clear emulsion is formed, thus obtaining the ganoderma lucidum glycopeptide microemulsion.
Effect example 1.
In vitro transdermal experiments were performed on the ganoderma lucidum glycopeptide microemulsion prepared in example 2:
removing epidermal hair and subcutaneous connective tissue from the isolated abdominal mouse skin, washing with physiological saline, and sucking to dryness for later use; in vitro transdermal experiments were performed using a Franz diffusion cell, contacting the stratum corneum of the isolated murine skin with a drug supply cell, the dermis layer facing downward with a receiving cell, and ensuring no air bubbles between the dermis layer and the receiving solution (normal saline); adding 1mL of Ganoderma glycopeptide water solution and Ganoderma glycopeptide microemulsion into the drug supply tank, respectively, and water-bathing at 37deg.C, 3000rmin -1 At the rotational speeds, 0.5mL was sampled at 2h, 4h, 8h, 12h, 18h, 24h, 36h in the EP tube, and the cumulative transdermal rates at the respective time points were measured and calculated according to the protein content test conditions in example 1.
The calculation formula is as follows:
,/>
in which Q n Cumulative transmission per unit area (μg cm) -2 ) Cn is the concentration of ganoderan (mg.mL) at each sampling time point -1 ),V 0 For receiving pool volume (mL), ci is the concentration of ganoderan at a time point prior to each sampling time point, V 1 A is the effective permeation area (cm) of the diffusion cell for the sample volume (mL) 2 ) W is the dosage (mg.cm) -2 )。
The experimental results are shown in Table 7, and the skin cumulative transmittance of the ganoderma lucidum glycopeptide microemulsion in 36h is (20.98 + -0.34)%. Experimental results show that the microemulsion can remarkably improve the percutaneous permeability of the ganoderma lucidum glycopeptides within 2-36 hours.
TABLE 7
Time Cumulative transmittance (%)
0 0
2 3.99±0.52*
4 5.39±0.28**
8 6.57±0.19***
12 9.31±0.48**
18 13.90±0.90**
24 17.34±0.38***
36 20.98±0.34***
Example 3.
The ganoderma lucidum glycopeptide microemulsion cream is prepared in the embodiment, the formula is shown in Table 8, the preparation process is shown in FIG. 10, A, B phases are respectively weighed in beakers, heated in water bath at 80-85 ℃ and stirred for dissolution, B phase is poured into A phase for homogenization, C phase is added for homogenization again after the temperature is reduced to 60 ℃, and D phase is added finally, and stirring is continued until the temperature is 38 ℃ to obtain the ganoderma lucidum glycopeptide microemulsion cream.
TABLE 8
In the embodiment, the ganoderma lucidum glycopeptide microemulsion is taken as a variable of the experiment, 1mL, 2mL, 3mL, 4mL and 5mL of ganoderma lucidum glycopeptide microemulsion are added into 100g of cosmetics, and the ganoderma lucidum glycopeptide microemulsion face cream is prepared respectively according to the preparation process.
Effect example 2.
The ganoderma lucidum glycopeptide microemulsion creams of different addition amounts of ganoderma lucidum glycopeptide microemulsion prepared in example 3 were evaluated by using sensory evaluation and skin moisturization as indexes.
(1) The sensory evaluation index is shown in Table 9.
TABLE 9
Taking the ganoderma lucidum glycopeptide microemulsion as a variable, adding a sample No. 1mL, adding a sample No. 2mL and the like, and summarizing sensory scores of 30 testers on ganoderma lucidum glycopeptide microemulsion face cream, wherein the sample No. 1 and the sample No. 2 are adopted, and the sensory scores are summarized in Table 10. The score of sample No. 3 was optimal in terms of appearance, smoothness, spreadability, wetness, thickness, and absorbency, and the score of sample No. 4 was optimal in terms of spreadability, but there was no significant difference (P > 0.05) from sample No. 3.
Table 10
(2) Skin moisturization evaluation:
test conditions: the temperature is (21+/-1) DEG C, and the humidity is 40% -60%.
Two hours before testing, the test area was kept dry and cleaned with dry facial tissues; the subject needs to sit still for 30min before applying the sample; the whole test period is monitored in real time, and the moisture index of the test area is kept at 15% -45% as effective.
The test area is set to be 3cm multiplied by 3cm, the interval between the areas is 1cm, the same experimenter smears different samples in equal quantity, and after smearing for 1h, 2h and 3h, the skin moisture indexes are measured respectively, and the moisture retention rate is calculated. 2% aqueous glycerol was used as a control group and distilled water was used as a blank control group.
The calculation formula is as follows:
。/>
the evaluation results are shown in fig. 11, wherein a is a glycerol group, B is a blank group, C is sample 1, d is sample 2, e is sample 3, f is sample 4, g is sample 5, and the moisture retention after 1 hour of application is 3 # 5 # 4 >2 # 1 > Gan Youzu > blank group; after smearing 2h and 3h, the moisture retention rate is 3 No. 4 >5 No. 2 >1 > Gan Youzu > blank. The 5 groups of creams added with the ganoderma lucidum glycopeptide microemulsion have obvious moisturizing capability (P is less than 0.001), and the moisturizing performance of the No. 3 sample added with the 3mL ganoderma lucidum glycopeptide microemulsion is the best.
Effect example 3.
Quality evaluation was performed on the ganoderma lucidum glycopeptide microemulsion cream prepared in example 3 to which 3mL of ganoderma lucidum glycopeptide microemulsion was added:
(1) Appearance characteristics:
the glossy ganoderma glycopeptide microemulsion cream is described in terms of appearance, texture, smell and use feeling. The texture of the cream is shown in figure 12, and the glossy ganoderma glycopeptide microemulsion cream is in a milky cream shape, and has uniform and glossy texture; the face cream has no special smell, and the face is moistened and moisturized.
(2) And (3) pH value measurement:
the pH value of the cream was measured using a pH meter, and the glossy ganoderma glycopeptide microemulsion cream was mixed with distilled water at a ratio of 1:9, and carrying out pH measurement on the ganoderma lucidum glycopeptide microemulsion face cream diluted solution, wherein the pH value of the ganoderma lucidum glycopeptide microemulsion face cream is (7.44+/-0.08) according with the specified range of the pH value of the cosmetic between 4.5 and 8.5.
(3) Heavy metal and microbiological examination:
heavy metals and microorganisms in ganoderma lucidum glycopeptide microemulsion face cream are detected according to cosmetic safety technical Specification (2015 edition). The results are shown in Table 11, and the heavy metal and microorganism detection of the ganoderma lucidum glycopeptide micro-emulsion cream meets the cosmetic standard.
TABLE 11
Test item Test results Standard value
Lead (mg kg-1) Not detected<2) ≤10
Mercury (mg kg-1) Not detected<0.01) ≤1
Arsenic (mg kg-1) Not detected<0.01) ≤2
Cadmium (mg kg-1) Not detected<0.5) ≤5
Colony count (CFU g-1) <10 ≤1000
Total number of mold and yeast (CFU g-1) <10 ≤100
Staphylococcus aureus (g) Not detected Cannot be detected
Heat resistant E.coli group (g) Not detected Cannot be detected
Pseudomonas aeruginosa (g) Not detected Cannot be detected
Effect example 4.
Stability experiments were performed on the ganoderma lucidum glycopeptide microemulsion cream prepared in example 3 to which 3mL ganoderma lucidum glycopeptide microemulsion was added:
(1) Heat resistance experiment:
placing the sealed Ganoderma glycopeptide microemulsion face cream in a 40 deg.C oven, taking out respectively at 1, 7, 15 days, comparing with Ganoderma glycopeptide microemulsion face cream placed at room temperature, and observing layering, dilution, and discoloration.
(2) Cold resistance experiment:
placing the sealed Ganoderma glycopeptide microemulsion face cream in a refrigerator at-20deg.C, taking out respectively at 1, 7, and 15 days, comparing with Ganoderma glycopeptide microemulsion face cream placed at room temperature, and observing layering, dilution, and discoloration.
(3) Cold and heat resistant acceleration experiment:
placing the sealed ganoderma lucidum glycopeptide microemulsion face cream in a baking oven at 40 ℃ for 24h, placing at room temperature for 12h, placing at a refrigerator at-20 ℃ for 24h and placing at room temperature for 12h, completing cold-resistant and heat-resistant acceleration experiments, comparing with the ganoderma lucidum glycopeptide microemulsion face cream placed at room temperature, and observing the phenomena of layering, dilution, color change and the like.
(4) Centrifugal experiment
At room temperature, the ganoderma lucidum glycopeptide microemulsion face cream is processed by the steps of 3000r min -1 Centrifuging for 30min, comparing with non-centrifuged Ganoderma glycopeptide microemulsion cream, and observing layering.
The stability test results are shown in table 12 and fig. 13, and it can be seen that the ganoderma lucidum glycopeptide microemulsion cream has no layering, dilution, color change and other phenomena, which indicates that the ganoderma lucidum glycopeptide microemulsion cream has a feasible preparation process and good stability.
Table 12
Effect example 5.
Skin photoaging resistance effect test is carried out on ganoderma lucidum glycopeptide microemulsion face cream:
1. building an animal photo-aging model:
removing back hair from the rat to expose back skin, wherein the area is about 4cm×4cm; radiating the shaved rats under an ultraviolet lamp (UVA+UVB), radiating for 0.5h on the 1 st day, radiating for 1h on the 2 nd to 4 th days, radiating for 2h on the 5 th to 10 th days, radiating for 3h on the 11 th to 21 th days, and finishing molding; after the molding is finished, the back of the positive control group is smeared with 0.5mL vitamin E10 mg/mL, and the back of the ganoderma lucidum glycopeptide microemulsion high/medium/low dose group (Gl-Gp H/M/L) is smeared with 25mg/mL, 10mg/mL and 1mg/mL respectively with 0.5mL for 7 days.
The skin morphology of the rats after ultraviolet injury is observed in fig. 14, the back skin state of the rats in the blank group is smooth, no wrinkles and no visible scars are formed; the rough and thickened epidermis of the model group can be seen as obvious red swelling or slight ulceration, and transverse rough wrinkles can be seen to exist continuously and cannot disappear along with the movement of rats; the skin of the rat treated by VE and ganoderan is improved to a certain extent, the skin wrinkles and skin lifting are reduced compared with those of the model group, and the skin appearance of the photo-aged rat is improved more obviously along with the increase of the concentration of the ganoderan.
2. Skin treatment:
after administration, the rat spine is dislocated, skin tissues are sheared, subcutaneous connective tissues are removed, and the rat spine is weighed; adding 9 times of physiological saline, grinding into homogenate, centrifuging at 4000r/min for 20min, collecting tissue supernatant, and preserving at-80deg.C.
3. Tissue section:
taking 1cm multiplied by 1cm of skin on the back of a rat, placing the rat in 4% paraformaldehyde fixing solution for fixing for more than 48 hours, then dehydrating skin tissues with a gradient of 50% -100% ethanol, degreasing in dimethylbenzene, embedding in paraffin, and cutting the embedded skin tissue wax blocks into slices with the thickness of 5 mu m for HE dyeing; HE staining was performed according to the kit instructions and morphological changes of skin cells were observed as shown in fig. 15. It can be seen that the blank group skin has compact and thin epidermis structure and obvious dermal papilla, so that the dermis and epidermis are tightly connected, and the dermis reticulation layer is tightly and uniformly arranged; the cuticle of the skin of the model group is obviously thickened, the structure is loose, the connection between the cuticle layer and the dermis layer is lost, the dermis nipple disappears, and the reticular layer is loosely arranged; the thickness of the epidermis of the VE group and the ganoderma lucidum glycopeptide low-dose group is slightly improved, but the structure is still loose, and the dermal papilla is not obvious yet; the epidermis is thinned in the ganoderma lucidum glycopeptide and is visible in the high-dose group, the papilla of the dermis is recovered, and the reticular layers are arranged tightly, wherein the dermis of the ganoderma lucidum glycopeptide high-dose group is connected with the epidermis more tightly, and the ganoderma lucidum glycopeptide high-dose group is close to the normal group.
The paraffin sections were then subjected to aldehyde fuchsin staining to observe the changes in the number and morphology of dermal spandex, as shown in fig. 16. The reticular structure of the skin elastic fiber of the blank group rat is relatively clear, the fiber shape is slender, and the fibers are orderly arranged; the arrangement rule of the elastic fibers of the skin of the rat in the model group disappears, which is the result of massive breakage and accumulation of the elastic fibers; the VE group and the ganoderma lucidum glycopeptide group can see wavy elastic fibers, the fibers are rarely broken, crushed, curled and twisted and aggregated into clusters, and the repairing effect of the ganoderma lucidum glycopeptide group is positively related to the dosage.
4. Determination of SOD Activity, MDA, HYP, GSH-Px content in rat skin tissue:
skin tissue supernatants of a blank group (CON), a model group (MOD), a positive control group (VE), a high-dose group (Gl-Gp H) of ganoderan, a medium-dose group (Gl-Gp M) of ganoderan, and a low-dose group (Gl-Gp L) of ganoderan were taken respectively, and the total protein content was measured by using a BCA kit, and the SOD activity and MDA, HYP, GSH-Px content were measured according to the kit instructions.
As shown in fig. 17, malondialdehyde (MDA) is the final product of lipid peroxidation, and to some extent, the extent of damage to cells by lipid peroxidation can be assessed. The MDA content of the model group is obviously higher than that of the blank group, and the model group has obvious difference (P is less than 0.05), which indicates that the MDA content of the skin tissue of the rat can be obviously increased by ultraviolet irradiation; the MDA content of the Gl-Gp L group is reduced compared with the model group, but no significant difference (P is more than 0.05), the MDA content of the skin tissue of the rat is obviously reduced compared with the model group in the Gl-Gp M group and the Gl-Gp H group, the significant difference exists, and the reduction of the Gl-Gp H group is most significant (P is less than 0.01), so that the dose dependency exists.
The antioxidant enzyme system is used as an important component of an organism for resisting oxidative stress, and is one of important indexes for evaluating the antioxidant capacity of a human body. As shown in fig. 18, the SOD activity of the model group is significantly lower than that of the normal control group, and the difference (P < 0.001) is significant, which indicates that the SOD activity of the skin tissue of the rat can be significantly inhibited by the ultraviolet irradiation; the groups Gl-Gp L, gl-Gp M and Gl-Gp H showed significantly improved SOD activity compared with the model group, and showed significant difference (P < 0.05) and dose dependence.
As shown in fig. 19, the GSH-Px content of the model group was significantly lower than that of the blank group, and there was a significant difference (P < 0.01), indicating that the GSH-Px content of the rat skin tissue was significantly reduced by the ultraviolet irradiation. The Gl-Gp L group and the model group have increased GSH-Px content and significant difference (P < 0.05), and the Gl-Gp M and Gl-Gp H groups and the model group have significantly increased GSH-Px content and significant difference (P < 0.01) and have dose dependence.
Hydroxyproline (HYP) is a precursor amino acid for synthesizing skin collagen, can promote skin collagen regeneration and repair damaged skin metabolism, and meanwhile, the content of HYP is an important measure for reflecting the skin aging degree. As shown in fig. 20, the skin HYP content of the rats in the model group was significantly reduced (P < 0.05) compared with the blank group, which indicates that the skin of the rats in the model group had reduced collagen synthesis ability, the network structure of collagen and elastin was broken, the collagen degradation was serious, and the typical characteristics of aging were shown. The content of HYP in the skin of the Gl-Gp group rat is increased, the concentration dependence trend is presented, the content of HPY in the skin of the Gl-Gp group rat is obviously improved (P is less than 0.05) compared with the model group, and the content of Gl-Gp M and Gl-Gp H groups is obviously different (P is less than 0.01) compared with the model group.
5. Determination of MMP-1, MMP-3, IL-1. Beta., IL-6, TNF-. Alpha.content in rat skin tissue:
skin tissue supernatants of a blank group (CON), a model group (MOD), a positive control group (VE), a high-dose group (Gl-Gp H) of ganoderan, a medium-dose group (Gl-Gp M) of ganoderan, and a low-dose group (Gl-Gp L) of ganoderan were taken, and the contents of MMP-1, MMP-3, IL-1. Beta., IL-6, and TNF-alpha in rat skin were measured by referring to ELISA kit instructions.
During skin aging, a very distinct feature is the degradation of collagen fibers and the breakdown of elastic fibers, which has been shown to be the result of matrix metalloproteinase action. As shown in fig. 21, the skin tissue of rats in the model group had significantly increased MMP-1 content (P < 0.01) compared to the blank group, indicating that uv irradiation was able to stimulate the expression of MMPs in cells. Compared with the model group, the content of MMP-1 in the skin tissues of rats in the Gl-Gp L, gl-Gp M and Gl-Gp H groups is reduced, but the content of MMP-1 in the Gl-Gp H group is reduced more obviously (P < 0.01).
As shown in fig. 22, MMP-3 levels were significantly elevated in skin tissues of rats in the model group compared to the blank group (P < 0.0001); compared with the model group, the content of MMP-3 in the skin tissues of rats in the Gl-Gp L, gl-Gp M and Gl-Gp H groups is reduced, and the content of MMP-3 in the ginseng total saponin liposome is reduced more (P < 0.0001). Taken together, the ganoderma lucidum glycopeptides have a certain inhibition effect on the increase of MMP-1 and MMP-3 content induced by ultraviolet rays.
When the skin is stimulated by ultraviolet rays, the skin epidermis and the dermal cells can be promoted to secrete a large amount of pro-inflammatory factors IL-6, IL-1 beta, tumor necrosis factor TNF-alpha and the like. As shown in fig. 23, the skin tissue of the model rats had significantly increased IL-6 content (P < 0.001) compared to the blank rats, which suggests that the uv irradiation could significantly increase IL-6 content in the skin tissue. The amount of IL-6 in the skin of rats in the Gl-Gp L, gl-Gp M and Gl-Gp H groups was reduced compared to the model group; however, MMP-1 content was reduced more significantly in the Gl-Gp M and Gl-Gp H groups (P < 0.001).
As shown in fig. 24, the skin tissue of the model group rats had significantly increased IL-1 β content (P < 0.0001) compared to the blank group, which also suggests that the ultraviolet irradiation could significantly increase the IL-1 β content in the skin tissue. The content of IL-1β in the skin of rats in the Gl-Gp L, gl-Gp M and Gl-Gp H groups was reduced compared to the model group; however, the decrease in IL-1β content was more pronounced in the Gl-Gp H group (P < 0.001).
As shown in fig. 25, the TNF- α content in the skin tissue of rats in the model group was significantly increased (P < 0.01) compared to the blank group, suggesting that the uv irradiation resulted in an increase in TNF- α content in the skin tissue. Compared with the model group, the TNF-alpha content in the skin of rats in the groups of Gl-Gp L, gl-Gp M and Gl-Gp H is reduced, and the rats have significance (P < 0.05).

Claims (8)

1. The application of the ganoderma lucidum glycopeptide is characterized by being used for preparing an antioxidant or skin photoaging resisting skin care product;
the combination mode of the glycopeptide bond of the ganoderma lucidum glycopeptide is O-glycosidic bond;
the ganoderma lucidum glycopeptide is prepared by the following process:
s1, taking dried ganoderma lucidum fruiting bodies and crushing the dried ganoderma lucidum fruiting bodies into ganoderma lucidum coarse powder;
s2, mixing the ganoderma lucidum coarse powder with pure water according to 1g: uniformly mixing (50-70) mL of feed liquid ratio by shaking through a shaking table, heating and extracting for 10-12 h at 60-80 ℃, then performing ultrasonic extraction for at least 3 times for 15-30 min each time, and combining the supernatant to obtain ganoderma lucidum glycopeptide crude extract;
s3, removing free protein from the ganoderma lucidum glycopeptide crude extract, and then carrying out dialysis treatment by using a dialysis bag;
s4, carrying out enzymolysis on the dialyzed ganoderma lucidum glycopeptides, carrying out water bath treatment at 60 ℃ for 35min, adding trypsin with the enzyme-to-bottom ratio of 4%, magnetically stirring for 6h, then placing in a constant-temperature water bath at 85 ℃ for 20min, and cooling to room temperature; then eluting with wheat germ agglutinin resin until the molecular weight cut-off is less than 10000Da, and freeze-drying to obtain Ganoderma glycopeptide freeze-dried powder.
2. The use of a ganoderma lucidum glycopeptide according to claim 1, wherein the shaking speed of the shaking table is 120r/min to 180r/min.
3. The use of a ganoderma lucidum glycopeptide according to claim 1, wherein the removing of the free protein in step S3 is performed by a Sevage method; the dialysis treatment specifically comprises the following steps: the solution was dialyzed in distilled water using a 0.1kDa dialysis bag for 48 hours.
4. The use of a ganoderma lucidum glycopeptide according to claim 1 wherein the temperature of the freeze drying in step S4 is-86 ℃ and the time of the freeze drying is at least 48 hours.
5. A ganoderma lucidum glycopeptide microemulsion, characterized in that the ganoderma lucidum glycopeptide microemulsion is prepared by taking the ganoderma lucidum glycopeptide as the raw material in any one of claims 1-4.
6. A method for preparing the ganoderma lucidum glycopeptide microemulsion according to claim 5, comprising the following steps:
the ganoderma lucidum glycopeptide and water are prepared into a water phase with the concentration of the medicine of 10 mg/mL-20 mg/mL, and a zwitterionic surfactant and a cosurfactant are mixed according to the proportion of 1: mixing (0.5-2) by mass ratio, mixing with oil phase, magnetically stirring at 23-27 ℃ and 500-1000 rpm, and dropwise adding water phase to obtain the ganoderma lucidum glycopeptide microemulsion.
7. The method of claim 6, wherein the zwitterionic surfactant is soybean lecithin, the cosurfactant is ethanol, and the oil phase is isopropyl myristate.
8. A cream comprising the ganoderma lucidum glycopeptide microemulsion of claim 5.
CN202310993467.XA 2023-08-09 2023-08-09 Ganoderma lucidum glycopeptide and preparation method and application thereof, and ganoderma lucidum glycopeptide microemulsion and face cream Active CN116694721B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310993467.XA CN116694721B (en) 2023-08-09 2023-08-09 Ganoderma lucidum glycopeptide and preparation method and application thereof, and ganoderma lucidum glycopeptide microemulsion and face cream

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310993467.XA CN116694721B (en) 2023-08-09 2023-08-09 Ganoderma lucidum glycopeptide and preparation method and application thereof, and ganoderma lucidum glycopeptide microemulsion and face cream

Publications (2)

Publication Number Publication Date
CN116694721A CN116694721A (en) 2023-09-05
CN116694721B true CN116694721B (en) 2023-10-20

Family

ID=87837947

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310993467.XA Active CN116694721B (en) 2023-08-09 2023-08-09 Ganoderma lucidum glycopeptide and preparation method and application thereof, and ganoderma lucidum glycopeptide microemulsion and face cream

Country Status (1)

Country Link
CN (1) CN116694721B (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1429837A (en) * 2001-12-29 2003-07-16 福州绿谷生物药业技术研究所 Grass planted ganoderma polysaccharide peptide and its extraction method
CN1786023A (en) * 2005-07-27 2006-06-14 西安绿谷制药有限公司 Lucid ganoderma poly saccharide peptide standard product, sts preparation method and application
CN105418781A (en) * 2015-11-23 2016-03-23 宁国千方中药发展有限公司 Ultrasonic extraction method of ganoderan
CN110016086A (en) * 2019-03-14 2019-07-16 合肥工业大学 A kind of process for refining of ganoderma lucidum polysaccharide
CN113151389A (en) * 2021-04-24 2021-07-23 长春中医药大学 Ginseng glycopeptide, preparation method and medical application thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2405923A1 (en) * 2009-03-10 2012-01-18 Palmed Teva Ltd Novel coprinus comatus and tremella mesenterica mushroom strains, products and extracts thereof and compositions comprising them

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1429837A (en) * 2001-12-29 2003-07-16 福州绿谷生物药业技术研究所 Grass planted ganoderma polysaccharide peptide and its extraction method
CN1786023A (en) * 2005-07-27 2006-06-14 西安绿谷制药有限公司 Lucid ganoderma poly saccharide peptide standard product, sts preparation method and application
CN105418781A (en) * 2015-11-23 2016-03-23 宁国千方中药发展有限公司 Ultrasonic extraction method of ganoderan
CN110016086A (en) * 2019-03-14 2019-07-16 合肥工业大学 A kind of process for refining of ganoderma lucidum polysaccharide
CN113151389A (en) * 2021-04-24 2021-07-23 长春中医药大学 Ginseng glycopeptide, preparation method and medical application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
灵芝多糖肽GL-PPS 的结构研究;王赛贞等;西南农业学报;第28卷(第2期);793-796 *

Also Published As

Publication number Publication date
CN116694721A (en) 2023-09-05

Similar Documents

Publication Publication Date Title
RU2492850C2 (en) Fibroblast growth agent
EP2175833B1 (en) Dermocosmetic compositions based on tamarind seed polysaccharide and vegetable extracts
CN111135117A (en) Small-molecule skin softening essence and preparation method thereof
CN115531272A (en) Antioxidant composition derived from alpine plants and preparation method and application thereof
KR101834795B1 (en) Cosmetic composition containing nanoliposome encapsulating extracts of Tremella fuciformis and Dioscorea japonica Thunb
CN1823724A (en) Salamander face beautifying skin protection product and its preparation method
CN116694721B (en) Ganoderma lucidum glycopeptide and preparation method and application thereof, and ganoderma lucidum glycopeptide microemulsion and face cream
KR102130149B1 (en) Cosmetic composition containing peach gum extract
KR101073284B1 (en) A composition for skin external application
CN116869898A (en) Protective cream for sensitive skin with enhanced barrier and preparation method thereof
KR101794419B1 (en) Osmotic enzyme fermentation natural liposome containing mushroom, process for the preparation thereof, and cosmetic, food or pharmaceutical composition comprising the same
KR101662272B1 (en) Natural cosmetic composition for skin moisturizing comprising Mixed extract of barley and germ and rice bran of black rice and preparation method thereof
CN107174536A (en) A kind of salamander face beautifying skin protection product and preparation method thereof
CN114317651B (en) Polygonatum sibiricum polypeptide, preparation method and application thereof
CN115040448A (en) Skin care product with enhanced anti-aging effect and preparation method thereof
KR102595837B1 (en) Cosmetic composition comprising spore oil extracted from antler-shaped Ganoderma lucidum and the method for preparing the same
KR20200001686A (en) Anti-aging and anti-wrinkle of skin improving cosmetic composition comprising extracts of Moschus, Angelica gigas Nakai, Cornus officinalis, Cervi Parvum Cornu and Phellinus linteus, Ganoderma Lucidum, Inonotus obliquus, Grifola Frondosa, Sparassis crispa
KR100532633B1 (en) Cosmetic composition having anti-inflammatory, skin-protecting, skin-elastic effects which comprise mixed plants extract
CN114652628A (en) Anti-aging cream for beautifying chest and body and enhancing skin immune barrier and preparation method thereof
KR102157210B1 (en) The functional cosmetic composition comprising extract of natural micro organism-fermented black ginseng
KR20060104807A (en) A method for producing extract of germinated brown rice and cosmetic composition comprising the extract
KR102073943B1 (en) A skin-care agent or cosmetics containig Lyophyllum ulmarium fruit body extract or Lyophyllum ulmarium mycelium extract
KR20210092587A (en) Liposome composition comprising skin physiologically active substances and method of preparing the same
CN111700818A (en) Wrinkle-removing and anti-aging ginseng polypeptide toning lotion and preparation method thereof
CN111419779A (en) Pure natural stretch mark protective cream and preparation method thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant