CN115960158A - Rare ginsenoside product of ginseng, anti-aging skin care emulsion, preparation method and application - Google Patents

Rare ginsenoside product of ginseng, anti-aging skin care emulsion, preparation method and application Download PDF

Info

Publication number
CN115960158A
CN115960158A CN202211666625.2A CN202211666625A CN115960158A CN 115960158 A CN115960158 A CN 115960158A CN 202211666625 A CN202211666625 A CN 202211666625A CN 115960158 A CN115960158 A CN 115960158A
Authority
CN
China
Prior art keywords
group
rare
preparation
extract
formula
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.)
Pending
Application number
CN202211666625.2A
Other languages
Chinese (zh)
Inventor
请求不公布姓名
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.)
Ningbo Quanshui Biotechnology Co ltd
Original Assignee
Ningbo Quanshui Biotechnology Co ltd
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 Ningbo Quanshui Biotechnology Co ltd filed Critical Ningbo Quanshui Biotechnology Co ltd
Priority to CN202211666625.2A priority Critical patent/CN115960158A/en
Publication of CN115960158A publication Critical patent/CN115960158A/en
Pending legal-status Critical Current

Links

Images

Landscapes

  • Saccharide Compounds (AREA)

Abstract

The application discloses a rare ginsenoside product, an anti-aging skin care emulsion, a preparation method and application. The preparation method comprises the following steps: obtaining water extract of ginseng flower; carrying out organic extraction on the water extract to obtain an extract; dissolving the extract in a reaction solution, and simultaneously adding a conversion agent into the reaction solution for reaction to obtain a solution containing rare saponin; purifying the solution containing rare saponin to obtain rare saponin; wherein the rare saponin comprises a compound shown as a formula (I) and/or a formula (II). The ginsenoside and related derivatives prepared by the preparation method disclosed by the application have oxidation resistance, and the rare ginsenoside and related derivatives thereof are found to have application prospects in preparing anti-aging skin care milk, such as face cream, skin care milk and the like.

Description

Rare ginsenoside product, anti-aging skin care emulsion, preparation method and application
Technical Field
The invention relates to the technical field of anti-aging skin care cream, in particular to a preparation method of rare ginsenoside, anti-aging skin care cream and application thereof.
Background
In the age of 'anti-aging and anti-oxidation', more and more anti-aging components are added into the cosmetics, and the products with anti-aging and anti-oxidation effects occupy a large market in all countries. The skin has the advantages of excellent biocompatibility, easy absorption, safety, good water solubility, low molecular weight and the like, and has the effects of protecting and repairing oxidative damage cells, promoting cell growth and removing excessive free radicals in the cells, so the skin has great development potential as an anti-aging substance.
Disclosure of Invention
In view of the above, the present application provides an anti-aging product based on rare saponins of panax ginseng and a preparation method thereof.
In a first aspect, the embodiment of the application discloses a method for preparing rare ginsenoside, which comprises the following steps:
obtaining water extract of ginseng flower;
carrying out organic extraction on the water extract to obtain an extract;
dissolving the extract in a reaction solution, and simultaneously adding a conversion agent into the reaction solution for reaction to obtain a solution containing rare saponin; and
purifying the solution containing the rare saponin to obtain the rare saponin;
wherein said rare saponins comprise, for example
Figure BDA0004015337580000021
And/or
Figure BDA0004015337580000022
The compounds shown.
In the embodiment of the present application, the R1 is selected from at least one of monoglucosyl group, biglucosyl group, polyglucosyl group, monomennosyl group, bimannosyl group, polymannosyl group, monofructosyl group, bimannosyl group, polyfructosyl group, monomecosyl group, bissaccharasyl group, and polysaccharyl group; and R2 is selected from at least one of monoglucosyl, biglucosyl, polyglucosyl, monomannosyl, bimannosyl, polymannosyl, monofructosyl, bimavosyl, polyfructose, monosaccharidyl, bimavosyl and polysaccharidyl.
In an embodiment of the application, the transforming agent comprises at least one of glucose, mannose, fructose and sucrose.
In an embodiment of the application, the transforming agent further comprises a glycosyltransferase selected from at least one of UGT91D2, UGT91D2e, UGT76G1, EUGT11, UGT73C6, UGT85C2, UGT74G1, UGT76G1 and UGTSL2 and a sucrose synthase.
In the examples of the present application, the reaction solution was Tris-HCl of 0.1mol/L pH 7.5 to 10.0.
In the embodiment of the present application, the step of obtaining the aqueous extract of ginseng flower specifically comprises:
soaking dried flos Ginseng material in distilled water at room temperature overnight, decocting to 100 deg.C, decocting for not less than 6 hr, filtering, repeating the residue for several times, mixing extractive solutions, and centrifuging to obtain water extractive solution.
The method according to claim 1, wherein the step of obtaining the extract comprises concentrating the aqueous extract, and extracting with an organic solvent selected from at least one of methanol, ethanol, isopropanol, n-butanol, ethyl acetate, and diethyl ether.
In a second aspect, the present application discloses a ginsenoside product comprising rare saponins prepared by the preparation method disclosed in the first aspect.
In a third aspect, the embodiment of the application discloses anti-aging skin care cream, which comprises rare saponin prepared by the preparation method disclosed in the first aspect.
In a fourth aspect, the rare saponin obtained by the preparation method disclosed in the first aspect is applied to preparing an antioxidant product.
Compared with the prior art, the application has at least the following beneficial effects:
the ginsenoside and related derivatives prepared by the preparation method disclosed by the application have oxidation resistance, and the rare ginsenoside and related derivatives have application prospects in preparation of anti-aging skin care milk, such as face cream, skin care milk and the like.
Drawings
FIG. 1 shows a scheme for preparing a compound represented by the formula (I) (R1 is-Fru and R2 is-Suc) according to example 1 of the present invention 1 HNMR map.
FIG. 2 shows a compound represented by the formula (I) (R1 is-Fru, R2 isof-Suc) 13 A CNMR map.
FIG. 3 shows a diagram of a compound represented by the formula (II) (wherein R1 is-Fru and R2 is-Suc) provided in example 1 of the present invention 1 HNMR map.
FIG. 4 shows a diagram of a compound represented by the formula (II) (wherein R1 is-Fru and R2 is-Suc) provided in example 1 of the present invention 13 A CNMR map.
FIG. 5 shows a compound represented by the formula (I) (R1 is- (Fru)) 2 R2 is- (Suc) 3 ) Is 1 HNMR picture.
FIG. 6 shows a compound of formula (I) (R1 is- (Fru) according to example 1 of the present invention 2 R2 is- (Suc) 3 ) Is/are as follows 13 A CNMR map.
FIG. 7 shows a compound represented by the formula (II) (R1 is- (Fru)) 2 R2 is- (Suc) 3 ) Is/are as follows 1 HNMR map.
FIG. 8 shows a compound represented by the formula (II) (R1 is- (Fru)) 2 R2 is- (Suc) 3 ) Is/are as follows 13 A CNMR map.
Detailed Description
In order to make the objects, technical solutions and advantages of the present invention more apparent, the present invention is further described in detail with reference to the following embodiments. It should be understood that the specific embodiments described herein are merely illustrative of the invention and do not limit the invention.
The inventor finds that two kinds of rare ginsenosides and related derivatives thereof have oxidation resistance in the long-term practice process, and finds that the rare ginsenosides and the related derivatives thereof have application prospects in preparing anti-aging skin care milk, such as face cream, skin care milk and the like. Wherein the two rare saponins of Ginseng radix have structures of
Figure BDA0004015337580000041
As shown. Wherein R1 is selected from the group consisting of monoglucosyl group, biglucosyl group, polyglucosyl group, monomannosyl group, dimanmannosyl group, polymanosyl group, monofructosyl group, dimanfructosyl group, polyfructosyl group, monosaccharidyl group, and dimancosyl groupAt least one of sugar groups and polysaccharose groups; r2 is at least one selected from the group consisting of monoglucosyl group, biglucosyl group, polyglucosyl group, monomannosyl group, dimanmannosyl group, polymannan base group, monofructosyl group, dimannan base group, polyfructose base group, monosaccharidyl group, dimancosyl group and polysaccharose base group. Wherein the term "mono" refers to only 1 saccharide unit, e.g. 1 glucose group; the term "bis" refers to 2 saccharide units only, e.g., 2 glucosyl groups, with 2 glucoses being linked by glycosidic linkages; the term "poly" refers to 3 and more saccharide units, e.g., 3 glucosyl groups, with the glucoses being linked by glycosidic linkages.
Therefore, the embodiment of the application also discloses a preparation method of the rare ginsenoside, which comprises the following steps: obtaining water extract of ginseng flower; carrying out organic extraction on the water extract to obtain an extract; dissolving the extract in a reaction solution, and simultaneously adding a conversion agent into the reaction solution for reaction to obtain a solution containing rare saponin; and purifying the solution containing the rare saponin to obtain the rare saponin. Wherein the rare saponin comprises a compound shown as a formula (I) and/or a formula (II).
According to the embodiment of the application, water extraction and organic extraction are carried out on ginseng flowers to obtain a primary solution containing ginsenoside, the solution is subjected to conversion reaction by a conversion agent, a very small amount of rare saponin is subjected to conversion reaction by the conversion agent, other common ginsenoside is converted into the rare saponin, an enrichment effect on the rare saponin is achieved, and meanwhile, an analogue similar to the rare saponin in structure can be obtained, so that a foundation is provided for enrichment and purification of the rare saponin, and the yield, purity and efficiency of the rare saponin are improved finally.
In the above-described production method, the conversion agent includes at least one of glucose (abbreviated as Glu), mannose (abbreviated as Man), fructose (abbreviated as Fru), and sucrose (abbreviated as Suc). These sugars provide the basis for the conversion reactions of the common saponins.
In the above production method, the transforming agent further comprises a glycosyltransferase selected from at least one of UGT91D2, UGT91D2e, UGT76G1, EUGT11, UGT73C6, UGT85C2, UGT74G1 and UGTSL 2. These enzymes act as catalysts for the conversion reaction of saponins.
In the above preparation method, the reaction solution is selected from 50mmol/L Tris-HCl with pH value of 7.5-10.0.
For this reason, the following description will be made with reference to a more specific embodiment, and the reagents and apparatuses involved in the following embodiment are generally commercially available unless otherwise specified.
Preparation of rare ginsenoside
1. Reagent and apparatus
Ginseng flower: is obtained from dried flower of Panax ginseng (Panax ginseng C.A. Meyer) produced in 2-year old Jilin province.
Glucose (L115560L- (-) -glucose, high purity grade, 98%), mannose (M120942L- (-) -mannose, high purity grade, 99%), fructose (F304579-5 g, high purity grade, ≧ 99.0%), sucrose (D274314D (+) sucrose, high purity grade, ≧ 99.0%), sucrose (high purity grade, ≧ 99.0%) and disodium uridine diphosphate (UDP, 99%, biotechnological grade) were all purchased from Alantin reagent Inc.
UGT91D2, UGT91D2e, UGT76G1, EUGT11, UGT73C6, UGT85C2, UGT74G1 and UGTSL2 immobilized cell preparations were all purchased from Beijing Baiolai Pabock technology, inc. Sucrose Synthase (SS) immobilized cell preparation was purchased from amresco, USA. The glycosyltransferase and the sucrose synthase are both recombinant Escherichia coli expression products, so that the actual use process is all immobilized cell preparations.
2. Preparation method
The preparation method of the rare ginsenoside disclosed in the embodiment of the application can be divided into the steps of water extraction, transformation and purification.
2.1, aqueous extraction
In a specific example 1:
soaking 1000g dried flos Ginseng material in 3L distilled water overnight, decocting to 100 deg.C for not less than 6 hr, and sealing in a sealed cooking tank for soaking and decocting. Filtering the decoction with four layers of gauze, repeating the above process for 3 times, mixing the 3 times extractive solutions to obtain water extractive solution with volume of 8.5L.
As other examples and comparative examples, the water extraction process was substantially the same.
2.2, extraction
In example 1 above, the aqueous extract was concentrated to 552mL under reduced pressure, and an organic reagent was added to the concentrate, the organic reagent comprising methanol, ethanol and isopropanol at a volume ratio of 1 to 1, the volume of the organic reagent added being 4 times that of the concentrate and about 2208mL. After fully mixing, standing for 30min, taking the supernatant, and carrying out reduced pressure concentration to remove the organic reagent, thereby obtaining 363mL of concentrated solution.
As another extraction process of example 2, an organic reagent containing methanol, ethanol and n-butanol was used, the volume ratio of methanol, ethanol and n-butanol was 1.
As another extraction process of example 3, an organic reagent containing methanol, ethanol and ethyl acetate was used in a volume ratio of 2.
As another extraction process of example 4, an organic reagent containing methanol, ethanol and diethyl ether was used in a volume ratio of 2.
In the extraction process of comparative example 1, ethanol was used as the organic reagent, and the volume of ethanol added was 4 times the volume of the concentrate, and the other steps were the same as those of example 1.
2.3 conversion reaction
Since the transformation reaction requires the use of immobilized cell preparations, the transformation reaction needs to be adapted to the growth of immobilized recombinant E.coli and to the soluble expression of glycosylases and sucrose synthases.
The conversion process of example 1 is specifically:
1) Taking 5.0mg of UGT91D2 immobilized cell preparation and 5.0mg of SS immobilized cell preparation, adding 0.2mmol/L IPTG, inducing temperature of 16 ℃ and inducing time of 12h, and after the induction expression is finished, centrifuging and collecting the immobilized cell preparation;
2) The immobilized cell preparation was diluted to 10g/L by washing with 50mmol/L Tris-HCl buffer (pH 8.5), then 25mL of the above-extracted concentrate was added, sucrose was added to 1.5mg/mL, fructose was added to 1.2mg/mL, UDP was added to 0.25mg/mL at 37 ℃ and 220r/min for 14h.
3) After the reaction is finished, the reaction is stopped by using methanol with the same volume, the mixture is centrifuged at 12 000r/min for 10min, and the supernatant is taken to be analyzed by HPLC and LC-MS to check the transformation condition.
The immobilized cell preparations of glycosyltransferase and sucrose synthase used in the transformation reaction processes of examples 2 to 4 and comparative example 1 were the same as in example 1, and the transformation conditions were the same.
The conversion reaction process of example 5 is specifically:
1) Taking 5.0mg of UGT91D2e immobilized cell preparation and 5.0mg of SS immobilized cell preparation, adding 0.2mmol/L IPTG, inducing at 16 ℃ for 12h, and after the induction expression is finished, centrifuging and collecting the immobilized cell preparation;
2) The diluted immobilized cell preparation was washed with 50mmol/L Tris-HCl buffer (pH 8.5) to 10g/L, then 25mL of the above-extracted concentrate (post-extraction concentrate prepared in example 1) was added, sucrose was added to 1.5mg/mL, fructose was added to 1.2mg/mL, UDP was added to 0.25mg/mL at 37 ℃ and 220r/min for 14h.
3) After the reaction is finished, the reaction is stopped by using methanol with the same volume, the mixture is centrifuged at 12 000r/min for 10min, and the supernatant is taken for HPLC analysis to investigate the transformation.
The conversion reaction process of example 6 is specifically:
1) Taking 5.0mg of UGT76G1 immobilized cell preparation and 5.0mg of SS immobilized cell preparation, adding 0.2mmol/L IPTG, inducing at 16 ℃ for 12h, and after the induction expression is finished, centrifuging and collecting the immobilized cell preparation;
2) The diluted immobilized cell preparation was washed with 50mmol/L Tris-HCl buffer (pH 8.5) to 10g/L, and then 25mL of the above-extracted concentrate (post-extraction concentrate prepared in example 1) was added, sucrose was added to 0.75mg/mL, fructose was added to 0.55mg/mL, glucose was added to 0.50mg/mL, UDP was added to 0.25mg/mL at 37 ℃ for 14h at 220 r/min.
3) The same as in example 5.
The conversion reaction process of example 7 is specifically:
1) Taking 5.0mg of EUGT11 immobilized cell preparation and 5.0mg of SS immobilized cell preparation, adding 0.2mmol/LIPTG, inducing at 16 deg.C for 12h, centrifuging to collect immobilized cell preparation after induction expression;
2) The diluted immobilized cell preparation was washed with 50mmol/L Tris-HCl buffer (pH 8.5) to 10g/L, and then 25mL of the above-extracted concentrate (post-extraction concentrate prepared in example 1) was added, sucrose was added to 0.55mg/mL, fructose was added to 0.35mg/mL, glucose was added to 0.75mg/mL, UDP was added to 0.25mg/mL at 37 ℃ for a reaction time of 220r/min for 14 hours.
3) The same as in example 5.
The conversion reaction process of example 8 is specifically:
1) Taking 5.0mg of UGT73C6 immobilized cell preparation and 5.0mg of SS immobilized cell preparation, adding 0.2mmol/L IPTG, inducing temperature of 16 ℃ and inducing time of 12h, and after the induction expression is finished, centrifuging and collecting the immobilized cell preparation;
2) The diluted immobilized cell preparation was washed with 50mmol/L Tris-HCl buffer (pH 8.5) to 10g/L, and then 25mL of the above-extracted concentrate (post-extraction concentrate prepared in example 1) was added, sucrose was added to 0.75mg/mL, fructose was added to 0.55mg/mL, glucose was added to 0.50mg/mL, UDP was added to 0.25mg/mL at 37 ℃ for 14h at 220 r/min.
3) The same as in example 5.
The conversion reaction process of example 9 is specifically:
1) Taking 5.0mg of UGT85C2 immobilized cell preparation and 5.0mg of SS immobilized cell preparation, adding 0.2mmol/L IPTG, inducing at 16 ℃ for 12h, and after the induction expression is finished, centrifuging and collecting the immobilized cell preparation;
2) The diluted immobilized cell preparation was washed with 50mmol/L Tris-HCl buffer (pH 8.5) to 10g/L, and then 25mL of the above-extracted concentrate (post-extraction concentrate prepared in example 1) was added, sucrose was added to 0.75mg/mL, fructose was added to 0.55mg/mL, glucose was added to 0.50mg/mL, UDP was added to 0.25mg/mL at 37 ℃ for 14h at 220 r/min.
3) The same as in example 5.
The conversion reaction process of example 10 is specifically:
UGT74G1 was used to immobilize the cell preparation at 5.0mg, SS was used to immobilize the cell preparation at 5.0mg, and the concentrated solution after extraction prepared in example 1 was used, and other reaction conditions and procedures were the same as those in example 5.
The conversion reaction process of example 11 is specifically: 5.0mg of UGTSL2 immobilized cell preparation and 5.0mg of SS immobilized cell preparation were used, and the concentrated solution after extraction prepared in example 1 was used, and the other reaction conditions and procedures were the same as those in example 5.
The conversion reaction process of example 12 is specifically: the UGTSL2, eugt11, and ss immobilized cell preparations were used, 4.0mg, and 1.0mg, respectively, and the post-extraction concentrate prepared in example 1 was used, and other reaction conditions and steps were the same as in example 5.
The conversion reaction process of comparative example 2 was specifically:
1) Taking 5.0mg of UGT91D2e immobilized cell preparation and 5.0mg of SS immobilized cell preparation, adding 0.35mmol/L IPTG, inducing at 16 ℃ for 15h, and after the induction expression is finished, centrifuging and collecting the immobilized cell preparation;
2) The diluted immobilized cell preparation was washed with 50mmol/L Tris-HCl buffer (pH 8.5) to 10g/L, then 25mL of the above-extracted concentrate (post-extraction concentrate prepared in example 1) was added, sucrose was added to 2.5mg/mL, fructose was added to 1.8mg/mL, UDP was added to 0.25mg/mL at 37 ℃ and 220r/min for 14h.
3) After the reaction is finished, the reaction is stopped by using methanol with the same volume, the mixture is centrifuged at 12 000r/min for 10min, and the supernatant is taken for HPLC analysis to investigate the transformation.
The concentrated solution after extraction and the solutions obtained in examples 1 to 12 and comparative example 1 were purified by preparative liquid chromatography, and the content and purity of the compound represented by the formula (I) or the formula (II) were analyzed by HPLC. Subjecting the prepared product to 1 HNMR and 13 and (3) performing CNMR measurement to determine the structure.
The chromatographic conditions for the preparation of the liquid phase were: the column was a Waters reverse phase column (35 mm. Times.150mm, 5 μm), and mobile phase A was deionized water (plus 0.1% formic acid); mobile phase B was acetonitrile (plus 0.1% formic acid). The elution gradient is 59-66% of the organic phase B, and the gradient time is 15min.
The HPLC analysis conditions were: c18 column (4.6 mm. Times.250 mm) m,5 μm); column temperature: 25 ℃; mobile phase: acetonitrile (a) -0.1% aqueous phosphoric acid solution (B); the amount of the sample was 20. Mu.L. Elution procedure: 0 to 15min,20 to 21 percent (A percent); 15-33min, 21% (A%); 33-38min, 21-30 percent (A%); 38-43min, 30 percent (A percent); 43 to 73min,30 to 40 percent (A%).
The compounds shown in the formula (I) and the formula (II) obtained in each example and comparative example are respectively shown in Table 1, and R1 groups and R2 groups in the compounds shown in the formula (I) and the formula (II) obtained in examples 1-12 are n glycosyl groups, wherein n is more than or equal to 2 and less than or equal to 5. In the compounds shown in the formula (I) and the formula (II) obtained in the comparative example 1, the R1 group and the R2 group are n glycosyl groups, and the number of the glycosyl groups is more than 5. The compounds of formula (I) and formula (II) prepared in example 1 were all distributed through 1 HNMR and 13 the CNMR structure identification results are shown in figures 1-8; other examples and comparative examples provide compounds of formula (I) and formula (II) wherein the R1 and R2 substituents are as shown in Table 1.
TABLE 1
Figure BDA0004015337580000091
Figure BDA0004015337580000101
Analyzing the compound shown as the formula (I) or the formula (II) in the extract liquid before conversion and the solution after conversion by adopting the HPLC method, drawing a standard curve by taking a chromatographically pure sample with the content of more than 99.0 percent prepared by preparative chromatography as a standard product, measuring the content of the compound shown as the formula (I) or the formula (II) in the solutions according to the standard curve, and calculating the conversion rate = the content of the compound shown as the formula (I) or the formula (II) in the solution after conversion/the content of the compound shown as the formula (I) or the formula (II) in the extract liquid before conversion. The results are shown in tables 2 and 3. In tables 2 and 3, the corresponding contents and conversions of the compounds of formula (I) or (II) (different R1 and R2 groups) are listed, respectively. For example, the compound of formula (I) before transformation in example 1 has R1 and R2 groups selected from-Fru \ Suc and R1 and R2 groups selected from- (Fru) 2 \-(Suc) 3 The corresponding contents of both can be analyzed by the HPLC method described above.
TABLE 2 formula (I)
Figure BDA0004015337580000102
Figure BDA0004015337580000111
TABLE 3 formula (II)
Figure BDA0004015337580000112
Figure BDA0004015337580000121
As can be seen from tables 2 and 3, the examples 1-12 and the comparative examples 1-2 both allow the contents of the formula (I) and the formula (II) to be significantly increased during the conversion process of the preparation, and have extremely high conversion rates.
2.4, purification
In specific example 1:
weighing 25g of macroporous adsorption resin (
Figure BDA0004015337580000122
ADS-750, KEHAISI (Beijing)), soaking in 80% ethanol for 12 hr, wet loading into 6X60cm glass chromatographic column, and washing with distilled water until no alcohol smell is detected. And weighing the converted solution, concentrating the solution to the effect of 15mL, sucking the solution by using a dropper, and standing the solution overnight by surrounding the sample on the inner wall of a glass chromatographic column. Finally, washing the inner wall of the column with a small amount of distilled water until no sample adheres, and sealing the opening of the column with a preservative film to ensure that the saponin can be completely adsorbed by the resin. And then the saponins adsorbed on the resin are subjected to gradient elution by 20%, 40%, 60% and 80% ethanol solutions respectively. Mixing eluates of 40%, 60% and 80%, concentrating by rotary evaporation to a certain volume, evaporating in water bath to remove residual ethanol, and freeze drying the concentrated solution at-50 deg.C for 48 hr to obtain dried product which is rare saponin sample of Ginseng radix.
The purification steps of the other examples and comparative examples were substantially the same as example 1.
Anti-aging function
The embodiment studies the rare ginsenoside samples prepared in the above examples and the anti-aging function of the skin care milk prepared from the same. Human Skin Fibroblasts (HSF) and human immortalized keratinocytes (HaCat) were used as model cells to study their anti-aging efficacy.
1. Experimental materials and methods
1.1 cell recovery and passage
A cryopreserved tube (from Punosai) containing Human Skin Fibroblasts (HSF) and human immortalized keratinocytes (HaCat) was immediately placed in a 37 ℃ constant temperature water bath to melt, and then cultured in 7-8mL of MEM complete medium for 24 hours, followed by 1 passage for backup.
The MTT method was used to examine the effects of the rare saponin samples of ginseng prepared in examples 1-12 and comparative examples 1-2 on HSF cells and on HaCat cell viability. Will 10 5 HSF of one/mL and 10 5 Cells per mLHaCat were seeded in 96-well plates separately and after 24h incubation, the solution was removed from the wells. Adding 0 prepared from DMEMA ginsenoside solution (examples 1-12 and comparative examples 1-2) of 02mg/mL, 100. Mu.L per well, was cultured in an incubator for 24 hours, with DMEM instead of the sample as a control. The wells were removed from the solution, washed 2 times with PBS, and their absorbance OD at 490nm was measured using a microplate reader, and cell viability was calculated as follows.
Cell viability/% = (As-Ab)/(Ac-Ab) × 100%;
in the formula: as is the light absorption value of the sample group; ab is blank light absorption value; ac is absorbance of control.
1.2 construction of oxidative damage cell model
Respectively preparing different molar concentrations H by taking DMEM as solution 2 O 2 Solution, 100. Mu.L of sample solution was added to each well and incubated for 1h, and 100. Mu.L of DMEM was used instead of the sample as a control. The wells were removed and washed 2 times with PBS, 100. Mu.L of DMEM solution was added to each well and incubated in an incubator for 24h. The OD value was measured in the above manner, and the cell viability was calculated, and the optimal condition was selected when the cell viability was about 60%. The results showed 100. Mu.L of 1.4mM H 2 O 2 The solution can satisfy the optimal damage condition.
1.3 repair action on oxidative damage cells
After the inoculated cells were incubated in an incubator for 24H, the culture medium was removed from the wells, and 100. Mu.L of 1.4mM H was added to the wells containing HSF and HaCat cells, respectively 2 O 2 Solution, incubation for 1H, 100 μ L DMEM instead of H 2 O 2 As a control group. The wells were removed and washed 2 times with PBS, 100. Mu.L of each well of 0.02mg/mL ginsenoside solution prepared in DMEM as a solvent at different mass concentrations was added, incubated in an incubator for 24h, OD was measured as above, and cell viability was calculated. Cell viability was calculated from cell viability, cell viability = H 2 O 2 Cell viability after treatment or cell viability after repair/initial cell viability x 100%.
1.4 skin care cream
The preparation process of the skin care emulsion comprises the following steps:
weighing phase A (EDTA-2Na 0.1g,1, 3-butanediol 4g, xanthan gum 0.3g, hyaluronic acid 2g deionized water solution) according to the formula, sterilizing in boiling water for 15min, and supplementing evaporated water; weighing the phase B components (octadecanol 1g, avocado oil 4g, shea butter 2g, vitamin E acetate 0.25 g) in sequence according to the formula, and fully dissolving in boiling water for 15min;
cooling A \ B phases to 80 deg.C respectively, slowly pouring B phase into A phase, homogenizing at 4000r/min for 5min, cooling to 45C, adding essence 0.5g and ginsenoside 0.6g (prepared in examples 1-12 and comparative examples 1-2 respectively); vacuumizing to remove bubbles, stirring and cooling, and discharging and filling.
1.5 animal experiments
Experimental animals: healthy SPF-class Kunming (KM) mice, all 36, were young for about two weeks, weighing (20 + -10) g, purchased from Beijing Wittisley laboratory animals technologies, inc. After the animals are purchased, the room temperature is maintained at 20 +/-2 ℃, the relative humidity is 35%, good ventilation is kept, regular conventional pellet feed feeding is realized, water is freely drunk, and the day and night rhythm is realized.
Preparation of mouse skin photoaging model:
grouping experimental animals: the group was divided into a normal control group (NC), a UV model group (UV), a skin care milk group (HH), and a vitamin E positive control group (VE). The mice of each group are fed with water by conventional drinking to adapt to the environment. Wherein, the skin care milk group is prepared using the above method, and comprises ginsenosides prepared in examples 1-12 and comparative examples 1-2, respectively, and thus the skin care milk group specifically comprises 14 groups in total of examples 1-12 and comparative examples 1-2.
Modeling an experimental animal:
model group: the backs of the mice in each group were shaved to form an exposed skin in the range of 3cm x 3cm, keeping the backs smooth. Except NC group, the mice in other groups are placed in an ultraviolet lamp (under the condition that the lamp tube position is adjusted to ensure that the distance between a light source and the mice is about 30cm, the lamp tube is preheated for 5min before each time of irradiation, and the mouse cage position is rotated, so as to balance the ultraviolet ray quantity irradiated by each group of mice, the irradiation frequency is 6 times per week, the irradiation frequency is 12 weeks, the irradiation time is 0.25 hour per day in the first 1-3 weeks, the irradiation time is 1.5 hours per day in the 6-9 weeks, the irradiation time is 3 hours per day in the 10-12 weeks, the irradiation time is 3 hours per day until the 13 th week is finished, the irradiation time is 12 weeks, the UV irradiation intensity is about 261.79J/cm 2 After irradiation, the exposed skin of the mouse is dry and dark, the mouse has reduced activity, listlessness and fatigue, body rolling and unfolding, increased red round spots on the back, thickened texture and the like.
Skin care milk group: before modeling, the skin care lotion prepared by the skin care lotion group is smeared on the exposed skin of the back of the mouse every day for 3 times, and each smearing time is 2min.
Positive control group: before molding, the exposed skin of the back of the mouse is smeared with vitamin E3 times a day for 2min each time.
1.6 immunohistochemical detection of Fas and FasL expression
Preparing a paraffin sample of the exposed skin of a mouse by using a DAB staining kit (Fuzhou Mixin Biotechnology Co., ltd.), and detecting the expression amounts of Fas and FasL in the paraffin sample by using an immunohistochemical secondary antibody kit (Fuzhou Mixin Biotechnology Co., ltd.); fas, fast antibody Wuhan Sanying Biotechnology Ltd.
2. As a result, the
2.1 results of cell experiments
TABLE 4 Effect on HSF and HaCat
Detailed description of the preferred embodiments Cell viability Cell survival rate after injury Post repair cell survival
Example 1 103.22%,105.23% 37.11%,35.21% 72.33%,69.28%
Example 2 102.45%,102.19% 36.23%,34.22% 68.12%,64.15%
Example 3 102.18%,103.42% 35.26%,32.17% 71.15%,65.32%
Example 4 104.32%,102.23% 34.13%,34.29% 69.45%,68.72%
Example 5 145.23%,123.12% 35.23%,34.29% 86.42%,86.18%
Example 6 102.12%,104.15% 36.07%,36.42% 71.12%,64.03%
Example 7 105.96%,107.12% 34.63%,37.12% 79.21%,62.42%
Example 8 104.12%,106.32% 36.52%,34.75% 63.15%,67.46%
Example 9 163.14%,154.12% 36.02%,34.18% 91.02%,89.42%
Example 10 105.42%,103.62% 35.75%,37.14% 65.21%,67.34%
Example 11 104.14%,106.07% 36.74%,37.32% 70.15%,67.32%
Example 12 156.32%,155.71% 36.21%,35.27% 96.15%,95.17%
Comparative example 1 101.12%,93.14% 36.14%,36.18% 42.18%,35.21%
Comparative example 2 85.15%,76.42% 36.28%,37.13% 35.72%,36.47%
As can be seen from Table 4, examples 1-12 showed growth promotion in MTT experiments for HSF and HaCat cells, whereas comparative examples 1 and 2 did not show significant growth promotion for both. Examples 1-12 have a repairing effect on HSF and HaCat cells after oxidative damage by hydrogen peroxide, while comparative examples 1-2 have an unobvious repairing effect. Also, of examples 1-12, examples 5, 9 and 12 are particularly prominent in promoting HSF and HaCat cell growth and oxidative damage repair.
It can be seen that the ginsenosides prepared in examples 1-12 have specific effects on promoting the growth of HSF and HaCat cells and repairing oxidative damage, and the effects are determined by the R1 group and R2 group in the compounds represented by formula (I) and formula (II).
2.2 animal experiments
TABLE 4
Figure BDA0004015337580000161
Table 5 lists the Fas and FasL expression values of the exposed skin tissues of mice from the control, model, VE and HH groups, respectively, after the above animal experiments, which were measured 6 times, and the values were expressed as mean and standard deviation, while significant differences between the different groups were compared and labeled for each line of data, respectively. As is clear from Table 5, examples 1 to 12 have an accelerating effect on Fas and FasL expression in mouse skin tissue, whereas comparative examples 1 and 2 have no significant accelerating effect on both.
Fas is a type I membrane protein and FasL is a type II membrane protein, both of which belong to the Tumor Necrosis Factor (TNF)/nerve growth factor receptor family. The death receptors of the TNF receptor family include tumor necrosis factor receptors, TNF-related apoptosis-inducing ligand receptors, and Fas-mediated related apoptosis pathways. In the death receptor-mediated exogenous pathway, membrane ligand (FasL) cell death receptor (Fas) binds, transmits an apoptosis signal through a death-inducing signal complex, induces activation of Caspase, and finally leads to apoptosis.
The damage of UV to human skin is a process of damage and continuous accumulation of UV in skin, during UV irradiation of skin tissue, apoptosis genes playing relevant roles comprise apoptosis promoting genes, apoptosis inhibiting genes and apoptosis effect genes, when cell genes are expressed differently from normal, cell dysfunction can be caused to induce photoaging, the abnormality comprises increase of expression of apoptosis promoting genes, down-regulation of expression of apoptosis inhibiting genes and the like, for example, expression of Fas and FasL is down-regulated, and the ginsenoside provided by the embodiment of the application can promote the increase of expression of the Fas and FasL, so that the ginsenoside has repair effect on the UV mouse skin damage, and the effect is determined by R1 groups and R2 groups in the compounds shown in the formula (I) and the formula (II) contained in the ginsenoside, and the result is consistent with cell experiments.
Therefore, the compound shown in the formula (I) and the compound shown in the formula (II) have an anti-aging function through cell experiments and animal experiments, and the ginsenoside is used for preparing the skin care milk, so that the ginsenoside has positive significance for the application prospect of cosmetics in the field of skin care.
The above description is only for the preferred embodiment of the present invention, but the scope of the present invention is not limited thereto, and any changes or substitutions that can be easily conceived by those skilled in the art within the technical scope of the present invention are included in the scope of the present invention.

Claims (9)

1. A method for preparing rare ginsenoside comprises the following steps:
obtaining water extract of ginseng flower;
carrying out organic extraction on the water extract to obtain an extract;
dissolving the extract in a reaction solution, and simultaneously adding a conversion agent into the reaction solution for reaction to obtain a solution containing rare saponin; and
purifying the solution containing the rare saponin to obtain the rare saponin;
wherein said rare saponins compriseSuch as
Figure FDA0004015337570000011
And/or
Figure FDA0004015337570000012
The compounds shown.
2. The production method according to claim 1, wherein R1 is selected from at least one of monoglucosyl group, biglucosyl group, polyglucosyl group, monomannosyl group, bimannosyl group, polymannosyl group, monofructosyl group, bimavosyl group, polyfructosyl group, monosaccharidyl group, bistalosyl group, and polysaccharidyl group; and R2 is selected from at least one of monoglucosyl, biglucosyl, polyglucosyl, monomannosyl, bimannosyl, polymannosyl, monofructosyl, bimavosyl, polyfructose, monosaccharidyl, bimavosyl and polysaccharidyl.
3. The production method according to claim 1, wherein the conversion agent includes at least one of glucose, mannose, fructose, and sucrose.
4. The production method according to claim 3, wherein the transforming agent further comprises a glycosyltransferase selected from at least one of UGT91D2, UGT91D2e, UGT76G1, EUGT11, UGT73C6, UGT85C2, UGT74G1, UGT76G1 and UGTSL2, and a sucrose synthase.
5. The method according to claim 4, wherein the reaction solution is Tris-HCl having a pH of 7.5 to 10.0 at 0.1 mol/L. The preparation method according to claim 1, wherein the step of obtaining the aqueous extract of ginseng flower specifically comprises:
soaking dried flos Ginseng material in distilled water at room temperature overnight, decocting to 100 deg.C, decocting for not less than 6 hr, filtering, repeating the residue for several times, mixing extractive solutions, and centrifuging to obtain water extractive solution.
6. The method according to claim 1, wherein the step of obtaining the extract comprises concentrating the aqueous extract, and extracting with an organic solvent selected from at least one of methanol, ethanol, isopropanol, n-butanol, ethyl acetate, and diethyl ether.
7. A ginsenoside product comprising rare saponins prepared by the method of any one of claims 1-7.
8. An anti-aging skin care cream comprising rare saponins prepared by the preparation method according to any one of claims 1 to 7.
9. Use of rare saponins obtained by the process according to any one of claims 1 to 7 for the preparation of an antioxidant product.
CN202211666625.2A 2022-12-23 2022-12-23 Rare ginsenoside product of ginseng, anti-aging skin care emulsion, preparation method and application Pending CN115960158A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202211666625.2A CN115960158A (en) 2022-12-23 2022-12-23 Rare ginsenoside product of ginseng, anti-aging skin care emulsion, preparation method and application

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202211666625.2A CN115960158A (en) 2022-12-23 2022-12-23 Rare ginsenoside product of ginseng, anti-aging skin care emulsion, preparation method and application

Publications (1)

Publication Number Publication Date
CN115960158A true CN115960158A (en) 2023-04-14

Family

ID=87359594

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211666625.2A Pending CN115960158A (en) 2022-12-23 2022-12-23 Rare ginsenoside product of ginseng, anti-aging skin care emulsion, preparation method and application

Country Status (1)

Country Link
CN (1) CN115960158A (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105087739A (en) * 2014-05-12 2015-11-25 中国科学院上海生命科学研究院 Novel catalytic system for preparing rare ginsenosides and application thereof
CN111575333A (en) * 2020-04-03 2020-08-25 东北林业大学 Biological preparation method of ginsenoside

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105087739A (en) * 2014-05-12 2015-11-25 中国科学院上海生命科学研究院 Novel catalytic system for preparing rare ginsenosides and application thereof
CN111575333A (en) * 2020-04-03 2020-08-25 东北林业大学 Biological preparation method of ginsenoside

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
白钰等: "人参花化学成分研究进展", 人参研究, no. 5, 18 October 2021 (2021-10-18), pages 54 - 57 *

Similar Documents

Publication Publication Date Title
Wang et al. Comparison of antitumor activities of different polysaccharide fractions from the stems of Dendrobium nobile Lindl
Wang et al. Structural and enhanced memory activity studies of extracts from Panax ginseng root
CN103923149A (en) Gynostemma pentaphylla saponin XLV Ed and preparation method thereof
CN113201081B (en) Gynura procumbens polysaccharide with immunological activity and preparation method and application thereof
CN111704544B (en) Labdane diterpenoid compound and separation method and application thereof
CN115960158A (en) Rare ginsenoside product of ginseng, anti-aging skin care emulsion, preparation method and application
CN101333239B (en) Anti-glioma compounds of triterpenoid saponin extracted from ardipusilloside
CN111671780A (en) Preparation method and application of tuberculate speranskia herb lignan with nerve cell protection activity
CN106589158A (en) Helicteres angustifolia polysaccharide and preparation method and application thereof
CN103610762A (en) Extract of corydalis impatiens total alkaloids and extraction method thereof
CN101028322A (en) Use of Maoliefengdou extract for preparing anti-cancer medicine
CN106749492B (en) A kind of steroid saponin compound and its preparation method and application
CN112794925B (en) Amomum villosum polysaccharide and preparation method and application thereof
CN101708201A (en) Medicament for treating gastric ulcer
CN101863953B (en) Method for separating and preparing Liriope graminifolia saponin A from Liriope graminifolia
CN101333238B (en) Triterpene compounds separated from seed of cowherb of Chinese traditional medicine and uses thereof
CN104387403A (en) Diterpenoid compounds having anti-tumor activity and application of diterpenoid compounds
CN105481932A (en) Triterpenoid saponins compound, and preparation method and uses thereof
CN111187309A (en) Preparation process and application of four components in cistanche
CN104292351A (en) Phellinus ribis polysaccharide, as well as preparation method and applications thereof
CN102584805B (en) Preparation method of one new active ingredient in natural plant
CN103623066A (en) Corydalis impatiens total alkaloid extractive for preparing anti-cancer drugs and application thereof
CN104059038A (en) Sesquiterpene compounds and application thereof
CN117247472B (en) Fucoidin and preparation method and application thereof
CN108676013B (en) Flavonol ketone compound with autophagy activation activity, preparation method and pharmaceutical application 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