CN115645430A - Application of delphinidin-3-O-galactoside in preparation of medicine or health food - Google Patents

Application of delphinidin-3-O-galactoside in preparation of medicine or health food Download PDF

Info

Publication number
CN115645430A
CN115645430A CN202211588207.6A CN202211588207A CN115645430A CN 115645430 A CN115645430 A CN 115645430A CN 202211588207 A CN202211588207 A CN 202211588207A CN 115645430 A CN115645430 A CN 115645430A
Authority
CN
China
Prior art keywords
ages
delphinidin
diseases
galactoside
health
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
CN202211588207.6A
Other languages
Chinese (zh)
Inventor
张旭光
贺瑞坤
岳中宝
吴新星
汪玉芳
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.)
BY Health Co Ltd
Original Assignee
BY Health 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 BY Health Co Ltd filed Critical BY Health Co Ltd
Priority to CN202211588207.6A priority Critical patent/CN115645430A/en
Publication of CN115645430A publication Critical patent/CN115645430A/en
Pending legal-status Critical Current

Links

Images

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The application provides an application of delphinidin-3-O-galactoside in preparation of medicines or health-care foods, wherein the medicines or the health-care foods are medicines or health-care foods for inhibiting AGEs, medicines or health-care foods for treating AGEs-related injuries, diseases or symptoms, or medicines or health-care foods for relieving AGEs-related injuries, diseases or symptoms. Experiments prove that the delphinidin-3-O-galactoside can inhibit AGEs, can treat AGEs-related injuries, diseases or symptoms, or relieve AGEs-related injuries, diseases or symptoms, and has higher activity compared with aminoguanidine which is a common AGEs inhibitor, so that a new idea is provided for inhibiting AGEs, treating AGEs-related injuries, diseases or symptoms, or relieving AGEs-related injuries, diseases or symptoms.

Description

Application of delphinidin-3-O-galactoside in preparation of medicine or health food
Technical Field
The application relates to the field of pharmaceutical preparations, in particular to application of delphinidin-3-O-galactoside in preparation of medicines or health-care foods.
Background
Advanced glycation end products (advanced glycation end products AGEs) refer to stable covalent adducts formed by macromolecules such as proteins, lipids or nucleic acids which spontaneously react with glucose or other reducing monosaccharides without the participation of enzymes. It is the end product of the non-enzymatic glycosylation reaction (Maillard reaction), and is the product of the binding of excess sugar to the protein. There are dozens of identified AGEs, such as carboxymethyl lysine (CML), carboxyethyl lysine (CEL), pyrrol, etc.
AGEs are derived from two sources in the body, one is that excess sugar and protein synthesize AGEs in the body, and the other is that AGEs present in food are taken into the body by eating. AGEs are able to combine with and destroy the tissue cells of the body. Normally, AGEs in the body can be cleared by the kidneys. However, AGEs are accumulated in the body with the increase of age or under certain pathological conditions, which can cause obvious adverse effects such as damage to the body, intervene in normal physiological and biochemical processes in the body, affect the normal metabolism of the body and cause the occurrence and development of diseases. In pathological conditions, AGEs can be present in the corresponding pathological tissues.
There are two main pathogenic mechanisms of AGEs. First, AGEs accumulate both intracellularly and extracellularly in all tissues and fluids, altering the structure and function of cellular proteins through glycation and cross-linking. Glycosylation of proteins affects their normal function by disrupting molecular conformation, interfering with receptor function, or altering enzymatic activity. AGEs also cross-link with other intracellular and extracellular molecules (e.g., lipids and nucleic acids), resulting in structural and functional changes that affect their normal function. Secondly, AGEs can interact with specific cell surface receptors, thereby altering intracellular signaling, gene expression, promoting reactive oxygen species production and activation of inflammatory pathways.
The current research proves that AGEs can accelerate the aging of human bodies and cause a plurality of chronic degenerative diseases, such as cardiovascular and cerebrovascular diseases, neurodegenerative diseases, osteoarticular diseases, kidney diseases and the like. Studies have shown that elevated levels of AGEs are a risk factor for the development of diabetes and complications. AGEs are independent risk factors for insulin resistance, and multiple regression analysis results show that AGEs levels are independently correlated with insulin resistance in healthy subjects. In another study, over 300 non-diabetic patients were examined and demonstrated that serum AGEs levels were independently correlated with HOMA-IR. The results of a 1 year clinical trial with 138 obese subjects with metabolic syndrome showed that a low AGEs diet improved insulin resistance and potentially reduced risk of type 2 diabetes in the test population compared to the high AGEs diet group without a substantial weight loss. In addition, the hyperglycemia of the diabetic patients can further promote the glycosylation reaction process, promote the increase of AGEs level and further cause the occurrence of related complications. The marked increase of AGEs content in liver cells and renal tubules of diabetic patients is observed by staining. Overexpression of β -growth factor and increased vascular endothelial growth factor concentration were found in the glomeruli and tubules. AGEs binding to specific receptors results in the overexpression of cytokines that may play an important role in diabetic vascular complications, such as Diabetic Retinopathy (DR), diabetic Nephropathy (DN), atherosclerosis.
Increased AGEs levels are strongly associated with decreased cognitive abilities in the elderly. Studies have shown that the content of AGEs is significantly increased in the brain and central nervous system of patients with neurodegenerative diseases such as alzheimer's disease, and that the increased content of AGEs is mainly concentrated in tissue regions with distinct pathological features (e.g. hippocampus of alzheimer's disease). Furthermore, it was found that dietary AGEs or their precursors may lead to a loss of selective permeability of the blood brain barrier. Results of alzheimer mouse experiments show that exogenously ingested AGE can reduce SIRT1 expression, thereby increasing β -amyloid and plaque production through the depolymerin and metalloprotease pathways. A cross-sectional study with the elderly showed that high levels of AGEs diet were associated with a more rapid memory decline.
The increase of AGEs level is closely related to the occurrence of osteoarticular diseases. The glycosylation pathway is considered to be one of the important factors leading to osteoporosis. Elevated serum AGEs levels, such as pentoside and CML, are found in patients with osteoporosis. Studies have shown that non-enzymatic glycosylation is a new factor affecting bone remodeling. AGEs accumulated in the bone matrix affect osteoblast differentiation and proliferation by binding to its receptor. In these cells, binding of AGEs to RAGE activates NF-. Kappa.B, resulting in increased expression of cytokines, growth factors, and cell adhesion molecules. This triggers an inflammatory process and induces oxidative stress, leading to osteoblast dysfunction and bone remodeling disorders. One of the major age-related changes in articular cartilage is an increase in AGEs levels. From the age of 20 years ago, AGEs accumulate in the collagen and proteoglycans of articular cartilage. The accumulation of AGEs in articular cartilage also leads to an increase in the prevalence of osteoarthritis as it AGEs. Recent studies have found that elevated levels of AGEs can negatively affect articular cartilage by increasing its stiffness, increasing chondrocyte-mediated proteoglycan degradation, decreasing proteoglycan synthesis, and inducing degradation of the chondrocyte extracellular matrix (ECM).
In addition, numerous empirical studies have shown an association between elevated levels of AGEs and various diseases such as skin aging, polycystic ovary syndrome, wound healing, periodontitis, erectile dysfunction, anemia in elderly women, slow walking speed in elderly, peripheral neuropathy, peripheral arterial disease, obstructive sleep apnea, cancer, schizophrenia, alzheimer's disease, higher all-cause mortality, severity of coronary atherosclerosis, cardiovascular disease mortality, and metabolic syndrome in adults and children, among others.
AGEs play an important role in the development or progression of a variety of diseases, and lowering their levels is positive for health benefits. Methods for reducing circulating AGEs include inhibiting the formation of AGEs, accelerating the catabolism of existing AGEs or inhibiting the crosslinking of AGEs, and blocking the biological response of AGEs. Inhibition of AGEs formation may occur by several mechanisms, including aldose reductase, antioxidant activity, reactive dicarbonyl trapping, sugar autoxidation inhibition, and amino binding. The research result proves that the inhibition of the generation of AGEs is beneficial to slowing down the aging.
Anthocyanins are compounds formed by combining anthocyanidins and sugars, are widely present in cell sap of flowers, fruits, stems, leaves and root organs of plants, are different in color from red, purple red to blue and are water-soluble pigments with bright colors. The anthocyanin belongs to flavonoid compounds, and the skeleton structure of the anthocyanin is 2-phenyl benzopyran cation. At present, more than 20 anthocyanins are known, and the most common anthocyanins are 6 anthocyanins, namely cyanidin (Cy), pelargonidin (Pg), delphinidin (Dp), petuniain (Pt), peoniflorin (Pn) and malvidin (Mv), and the anthocyanins are mainly different from one another in a group R 1 And R 2 A change in (c).
Figure 449379DEST_PATH_IMAGE002
According to preliminary statistics, tens of thousands of plants of 27 families, 73 genera, have been found to contain anthocyanins. More than 500 anthocyanins are currently isolated from plants. Various anthocyanins can exhibit different shades of yellow, red, violet, black, etc., depending on their ability to form resonance structures, substituents on the C6-C3-C6 nucleus, and environmental factors.
Free anthocyanidins are very unstable and are generally present in nature in the form of glycoside conjugates. Its glycoside form is more stable than aglycone and therefore exists mainly as glycoside (glycoside), i.e., anthocyanin, in plants. Typically these glycosides include mono-glucosides, di-glucosides and acyl derivatives. In the known anthocyanins or anthocyanidins, the majority are present in glycosidated form, the glycoside-forming sugars being mainly glucose, galactose, rhamnose, arabinose, xylose and the disaccharides and trisaccharides composed of these monosaccharides, the common diglycosides being sophorose, rutinose and sambucobiose, etc. The sugar and anthocyanin are linked by O-bonds, mainly at the 3-, 5-and 7-carbon positions, with a small proportion being linked to the 3' -carbon position, and almost all anthocyanins will be glycosidated at the 3-position.
Further, anthocyanins in plants are also present as acylated anthocyanins, which are formed by ester bond bonding of organic acids, and the most common acids involved in glycosyl acylation are various hydroxycinnamic acid derivatives such as ferulic acid, caffeic acid, sinapic acid, etc., fatty acids such as malic acid, acetic acid, succinic acid, malonic acid, oxalic acid, etc., and p-hydroxybenzoic acid, etc. The number of hydroxyl groups in anthocyanin molecules, the methylation degree of hydroxyl groups, the type, quantity and position of sugars attached to anthocyanin molecules, the type and number of fatty acids or aromatic acids attached to sugar molecules, the different actions of anthocyanin molecules with other substances and the like cause the existence of various anthocyanins in nature.
Related research is also carried out on inhibition of AGEs by anthocyanin monomers. For example, researches prove the inhibition effect of cyanidin-3-O-galactoside, delphinidin-3-O-glucoside and paeoniflorin-3-O-glucoside on AGEs.
The master paper 'research on structure-activity relationship and mechanism of medium-pressure rapid separation preparation of anthocyanin monomers and inhibition of fluorescent glycosylation end products' experiments on the activity of seven anthocyanins in inhibition of fluorescent glycosylation end products, and the research shows that anthocyanins with large molecular weight and a large number of glycosyl groups have more hydroxyl groups, can form more hydrogen bonds and other intermolecular forces with protein, and anthocyanins with large molecular weight have larger steric hindrance, can effectively hinder glycosylation modification of beta-Lg by carbonyl compounds, and further has good effect of inhibiting glycosylation. In the research of the oxidation resistance and the structure-activity relationship of the procyanidin, the oxidation resistance of the procyanidin of which the B ring has a pyrogallol structure is higher than that of the procyanidin of which the B ring has catechol, and the influence of the pyrogallol structure on the oxidation resistance of the procyanidin is the largest. Since glycosylation is accompanied by oxidation change, it is presumed that anthocyanin having a pyrogallol structure in the mother nucleus has stronger oxidation resistance than anthocyanin having a catechol hydroxyl structure in the mother nucleus, and therefore, exhibits excellent effect of inhibiting fluorescent AGEs. Therefore, in order to enhance the effect of inhibiting AGEs, those skilled in the art generally screen anthocyanins having a large molecular weight and a large number of hydroxyl groups. For example, in the extraction and purification processes of anthocyanin, anthocyanin with large molecular weight and large number of hydroxyl groups is enriched by means of molecular sieve, column chromatography and membrane filtration, or specific anthocyanin is subjected to glycosylation modification and B-ring hydroxyl substitution to obtain anthocyanin with large molecular weight and large number of hydroxyl groups, so that the inhibition rate of anthocyanin AGEs is improved.
Disclosure of Invention
The application provides application of delphinidin-3-O-galactoside in preparation of medicines or health-care foods so as to improve the in-vivo inhibition effect on AGEs.
The first aspect of the application provides an application of delphinidin-3-O-galactoside in preparation of medicines or health foods, wherein the medicines or health foods are medicines or health foods for inhibiting AGEs, medicines or health foods for treating AGEs-related injuries, diseases or symptoms, or medicines or health foods for relieving AGEs-related injuries, diseases or symptoms.
Furthermore, the medicine also comprises pharmaceutically acceptable auxiliary materials, and the health-care food also comprises dietotherapy acceptable auxiliary materials.
Furthermore, the dosage form of the medicine or the health food is an oral preparation or an injection preparation.
Further, the oral preparation is tablet, powder, capsule, granule, pill, powder, paste, solid beverage or oral liquid.
Furthermore, the mass content of the delphinium-3-O-galactoside in the oral preparation is more than or equal to 0.013%.
Furthermore, the injection preparation is injection or powder injection for injection.
Furthermore, the mass content of delphinium-3-O-galactoside in the injection preparation is more than or equal to 0.013%.
Further, the inhibition of AGEs is inhibition of the production of AGEs or promotion of the decomposition of AGEs.
Further, the AGEs-related damage is AGEs-mediated apoptosis increase, active oxygen content increase, inflammatory factor TNF-alpha overexpression, inflammatory factor ICAM-1 overexpression, inflammatory factor VCAM-1 overexpression and/or mitochondrial ATP content reduction.
Furthermore, the AGEs-related diseases can be any one or more of cardiovascular and cerebrovascular diseases, neurodegenerative diseases and osteoarticular diseases.
Further, the cardiovascular and cerebrovascular diseases are selected from diabetes and its complications, and atherosclerosis.
Further, the neurodegenerative disease is selected from alzheimer's disease, parkinson's disease, and amyotrophic lateral sclerosis.
Further, the bone joint disease is osteoporosis and osteoarthritis.
Further, the aforementioned AGEs-related symptoms are aging.
Experiments prove that the delphinidin-3-O-galactoside can inhibit AGEs, can treat AGEs-related injuries, diseases or symptoms, or relieve AGEs-related injuries, diseases or symptoms, and has higher activity compared with aminoguanidine which is a common AGEs inhibitor, so that a new idea is provided for inhibiting AGEs, treating AGEs-related injuries, diseases or symptoms, or relieving AGEs-related injuries, diseases or symptoms.
Drawings
In order to more clearly illustrate the technical solutions of the embodiments of the present application, the drawings needed to be used in the embodiments of the present application will be briefly described below, and it is obvious that the drawings described below are only some embodiments of the present application, and it is obvious for a person skilled in the art to obtain other drawings based on the drawings without any creative effort.
FIG. 1 shows the results of detecting the content of AGEs in HUVEC cells of each test group by ELISA in test example 2, wherein # indicates that P is <0.05 compared with the control group; * Represents a ratio P <0.05 to the model group; and ^ represents the ratio P <0.05 to other anthocyanin groups.
FIG. 2 is a graph showing the results of the measurement of the cell survival rate of each test group by the CCK8 method in test example 2, in which # indicates that the ratio P to the control group is <0.05; * Represents a ratio P <0.05 to the model group; and a represents the ratio P <0.05 of other anthocyanin groups.
FIG. 3 shows the results of ROS production by the microplate reader in test example 2 for each test group, wherein # indicates that P is <0.05 as compared with the control group; * Represents a ratio P <0.05 to the model group; and a represents the ratio P <0.05 of other anthocyanin groups.
FIG. 4 shows the Propidium Iodide (PI) staining method in test example 3 for detecting the apoptosis rate of each test group, wherein # indicates that P <0.05 compared with the control group; * Represents a ratio P <0.05 to the model group; and ^ represents the ratio P <0.05 to other anthocyanin groups.
FIG. 5 shows the results of qPCR assay of inflammatory factor TNF-. Alpha.expression in each test group in test example 3, wherein # indicates that P <0.05 compared with the control group; * Represents a ratio P <0.05 to the model group; and ^ represents the ratio P <0.05 to other anthocyanin groups.
FIG. 6 shows the results of qPCR detection of the expression of inflammatory factor ICAM-1 in each test group in test example 3, wherein # indicates that the ratio P to the control group is <0.05; * Represents a ratio P <0.05 to the model group; and ^ represents the ratio P <0.05 to other anthocyanin groups.
FIG. 7 shows the results of qPCR assay for the expression of the inflammatory factor VCAM-1 in each test group in test example 3, wherein # indicates a P <0.05 to the control group; * Represents a ratio P <0.05 to the model group; and ^ represents the ratio P <0.05 to other anthocyanin groups.
Fig. 8 shows the results of the measurement of the mitochondria-associated ATP index of each test group in experimental example 3, wherein # indicates a P <0.05 compared with the control group; * Represents a ratio P <0.05 to the model group; and ^ represents the ratio P <0.05 to other anthocyanin groups.
Detailed Description
Embodiments of the present application will be described in further detail below with reference to the drawings and examples. The following detailed description of the embodiments and the accompanying drawings are provided to illustrate the principles of the application and are not intended to limit the scope of the application, i.e., the application is not limited to the described embodiments.
As described in the background of the present application, in order to improve the effect of anthocyanins on AGEs inhibition, those skilled in the art generally screen anthocyanins having a large molecular weight and a large number of hydroxyl groups. For example, in the extraction and purification processes of anthocyanin, anthocyanin with large molecular weight and large number of hydroxyl groups is enriched by means of molecular sieve, column chromatography and membrane filtration, or specific anthocyanin is subjected to glycosylation modification and B-ring hydroxyl substitution to obtain anthocyanin with large molecular weight and large number of hydroxyl groups, so that the inhibition rate of anthocyanin AGEs is improved. But such an operation causes an increase in research costs, industrialization costs, and practical application costs. In experiments, the inventor of the application accidentally finds that the delphinidin anthocyanin and the delphinidin-3-O-galactoside have better inhibiting effect on AGEs, and the inhibiting effect on the AGEs is obviously higher than the inhibiting effect of the delphinidin-3-O-glucoside, the delphinidin-3-O-arabinoside, the delphinidin-3-O-rutinoside and the delphinidin-3,5-O-diglucoside on the AGEs. Based on the findings, the application provides the application of delphinidin-3-O-galactoside in preparing medicines or health-care foods, wherein the medicines or health-care foods are medicines or health-care foods for inhibiting AGEs, medicines or health-care foods for treating AGEs-related injuries, diseases or symptoms, or medicines or health-care foods for relieving AGEs-related injuries, diseases or symptoms.
Experiments prove that the delphinidin-3-O-galactoside can inhibit AGEs, can treat AGEs-related injuries, diseases or symptoms, or relieve AGEs-related injuries, diseases or symptoms, and has higher activity compared with aminoguanidine which is a common AGEs inhibitor, so that a new idea is provided for inhibiting AGEs, treating AGEs-related injuries, diseases or symptoms, or relieving AGEs-related injuries, diseases or symptoms.
In combination with the above analysis of 6 anthocyanidin structures, delphinidin-3-O-galactoside is a monosaccharide glycoside having a small molecular weight, but shows a completely different inhibitory effect against AGEs than that presumed in the prior art. Although the structure-activity relationship of delphinidin-3-O-galactoside cannot be explained at present, experiments prove that the delphinidin-3-O-galactoside has an excellent AGEs inhibition effect.
In some embodiments, the above medicament further comprises pharmaceutically acceptable auxiliary materials, and the health food further comprises dietetically acceptable auxiliary materials.
The auxiliary materials can be selected from common corresponding auxiliary materials whether pharmaceutically acceptable auxiliary materials or dietotherapy acceptable auxiliary materials, and can be selected according to specific dosage forms of medicines or health-care foods.
In some embodiments, the above-mentioned medicament or health food is in the form of an oral preparation or an injection preparation. Preferably, the oral preparation is a tablet, powder, capsule, granule, pill, powder, paste, solid beverage or oral liquid. Or preferably, the injection preparation is injection or powder injection for injection.
In some embodiments, the oral preparation contains delphinidin-3-O-galactoside in an amount of 0.013% or more by mass for easy administration to a patient or user.
In some embodiments, the injection preparation contains delphinidin-3-O-galactoside in 0.013% or more by mass.
The application research proves that delphinidin-3-O-galactoside has an outstanding effect of inhibiting AGEs, and the inhibition of AGEs can inhibit the generation of AGEs or promote the decomposition of AGEs.
Further experimental studies by the applicant have found that in some embodiments, the aforementioned AGEs-related damage is AGEs-mediated increase in apoptosis, increase in active oxygen content, overexpression of inflammatory factor TNF- α, overexpression of inflammatory factor ICAM-1, overexpression of inflammatory factor VCAM-1, and/or decrease in mitochondrial ATP content.
Based on the association of AGEs with various diseases, in some embodiments, the AGEs-related diseases are any one or more of cardiovascular and cerebrovascular diseases, neurodegenerative diseases and osteoarticular diseases; preferably, the cardiovascular and cerebrovascular diseases are selected from diabetes and its complications, atherosclerosis; preferably the neurodegenerative disease is selected from alzheimer's disease, parkinson's disease, amyotrophic lateral sclerosis; preferably, the osteoarticular disease is osteoporosis or osteoarthritis. Or in some embodiments, the age-related condition is aging.
The beneficial effects of the present application will be further explained below in connection with the experiments.
TABLE 1 sources of anthocyanins used in the tests
Figure 166799DEST_PATH_IMAGE003
Test example 1, method for detecting efficacy of inhibiting AGEs production:
the experimental principle is as follows:
bovine Serum Albumin (BSA) and Methylglyoxal (MGO) interact to generate AGEs, a compound to be detected is incubated with the BSA and the MGO together, and the amount of the AGEs generated is judged by detecting the change of a fluorescence value in a system, so that whether the compound has an inhibiting effect on the generation of the AGEs is evaluated. Aminoguanidine (AG) was used as a positive control. The blank control group did not use any anthocyanin or aminoguanidine.
The study was carried out using sterile black non-transparent 96-well plates, in which 90. Mu.L of 10 mg/mL BSA solution and 10. Mu.L of 1.25M MGO solution were added to each well, and then 10. Mu.L of test solution or 10. Mu.L of physiological saline (blank) was added to each well, and positive drug solution was added to the positive control group, as shown in the table below. After the reaction solution was sufficiently mixed, fluorescence values at the start of the experiment (excitation wavelength Ex =370 nm, emission wavelength Em =440 nm) were detected by an M5 microplate reader, respectively. After the detection is finished, the sterile sealing plate is sealed by a membrane, and the black non-transparent 96-well plate is placed in a 37 ℃ dark place to react with 24h, and then the fluorescence value is detected again under the same condition. And 6-8 multi-well detection is carried out on each test object group and each control group, the increase of the fluorescence value of each well of each group is calculated, and the AGEs generation inhibition rate of each group is converted.
Table 2: 96-pore plate reaction system for preliminary screening test
Figure 499691DEST_PATH_IMAGE004
Under the same reaction system and conditions, the test substances are subjected to different gradient concentration (50, 10, 1, 0.1, 0.03 and 0.01 mg/mL) detection, 5 multi-well detection is carried out on each concentration group of the test substances and a control group, and the detection result is subjected to fitting treatment by using Graphpad to obtain the IC50 value of the test substances.
The main experimental results are:
TABLE 3 inhibitory Effect of different kinds of anthocyanins on AGEs production
Figure 750282DEST_PATH_IMAGE005
Figure 510427DEST_PATH_IMAGE006
Remarking:P<0.05 * 1 compared with malvidin-3-O-galactoside, paeoniflorin-3-O-galactoside, delphinidin-3-O-glucoside, delphinidin-3-O-arabinoside, delphinidin-3-O-rutinoside, delphinidin-3,5-O-diglucoside, petunia-3-O-rutinoside (p-coumaroyl) -5-O-glucoside and cowberry mixed anthocyanin, the composite anthocyanin has obvious difference.
P<0.05 * 2 Compared with delphinidin-3-O-glucoside, paeonin-3-O-glucoside, malvidin-3-O-galactoside, malvidin-3-O-arabinoside, malvidin-3,5-O-diglucoside, petunidin-3-O-rutinoside (p-coumaroyl) -5-O-glucoside and cowberry mixed anthocyanin, the compound has obvious difference.
P<0.05 * 3 Compared with malvidin-3-O-arabinoside, delphinidin-3-O-arabinoside, paeonin-3-O-glucoside, paeonin-3-O-galactoside, paeonin-3-O-rutinoside, paeonin-3,5-O-diglucoside, petunidin-3-O-rutinoside (p-coumaroyl) -5-O-glucoside and cowberry mixed glucoside, the anthocyanin has obvious difference.
TABLE 4 IC50 values for inhibition of AGEs production by different anthocyanins
Figure 715144DEST_PATH_IMAGE007
Figure 851727DEST_PATH_IMAGE008
As can be seen from the data in tables 3 and 4 above, delphinidin-3-O-galactoside showed a better inhibitory effect on AGE production than the other anthocyanins tested (excluding malvidin-3-O-glucoside and paeoniflorin-3-O-arabinoside), even though it showed a better inhibitory effect compared to petunidin-3-O-rutinoside (p-coumaroyl) -5-O-glucoside; meanwhile, when the delphinidin-linked sugar chain is galactose (hexose, relative molecular mass 160), it has a more significant advantage than when the linked sugar chain is glucose (hexose, relative molecular mass 160), arabinose (pentose, relative molecular mass 150), rutinose (dodecane, relative molecular mass 326), or diglucose (dihexaglucose, relative molecular mass 320). In addition, delphinidin-3-O-galactoside has obvious advantages compared with the positive control group, and the effects of inhibiting AGEs generation of other test groups are not the same as those of the positive control group except that cowberry mixed anthocyanin, malvidin-3-O-glucoside and paeoniflorin-3-O-arabinoside have advantages compared with the positive control group.
Moreover, as can be seen from the data in tables 3 and 4, the delphinidin-3-O-galactoside in delphinidin anthocyanin has the most outstanding effect of inhibiting the production of AGEs, the malvidin-3-O-glucoside in malvidin anthocyanin has the most outstanding effect of inhibiting the production of AGEs, and the paeonin-3-O-arabinoside in paeonin anthocyanin has the most outstanding effect of inhibiting the production of AGEs, i.e., the inhibitory effects of the small molecular anthocyanins such as monosaccharide anthocyanin or disaccharide anthocyanin tested in the application on AGEs are not consistent, and the delphin-3-O-galactoside, malvidin-3-O-glucoside and paeonin-3-O-arabinoside in the application realize unexpected technical effects on the production of AGEs.
Test example 2 inhibition of high sugar-induced AGEs production and protection of umbilical vein endothelial cells
The experimental principle is as follows: HUVEC cells cultured in vitro were randomly divided into a control group (without any induction and intervention), a model group (40 mmol/L glucose), an delphinidin-3-O-glucoside group, an delphinidin-3-O-galactoside group, an delphinidin-3-O-arabinoside group, an delphinidin-3-O-rutinoside group, a petunianin-3-O-rutinoside (p-coumaroyl) -5-O-glucoside group, a cowberry mixed anthocyanin group, a high sugar induction of 40mmol/L, and an intervention of endothelial cells with different anthocyanins of 100. Mu. Mol/L for 24h.
2.1 The HVUECs are subjected to primary culture and subculture by adopting a modified Jaffe method and the like, and 3-6 generations of well-grown HVUECs are taken for experiment.
2.2 Experimental grouping and conditional culture 3-6 generation HVUECs are taken to prepare cell suspension according to 4.0X 10 5 cell/well cell density was plated on 24-well plates, DMEM medium containing 10% fetal bovine serum was added, and the mixture was incubated at 37 ℃ and 5% CO 2 Culturing in an incubator, and changing a serum-free culture solution when the HUVECs grow to be in a sub-fusion state, and continuously culturing for 12-24 h. Then adding a sample with the concentration of 100 mu mol/L for pretreatment for 8 hours according to the experimental requirements, and then adding glucose with the concentration of 40mmol/L for acting for 24 hours. Grouping: (1) blank control group: adding DMEM culture solution with the same amount as the medicine; (2) model group glucose (40 mmol/L) was added; (3) and (3) a high-sugar + sample 1~6 group, setting more than 3 multiple holes, continuously culturing for 24 hours, and centrifugally collecting cells and culture solution.
And (3) testing:
1) And detecting the content change of AGEs by an enzyme-linked immunosorbent assay, wherein the result is shown in figure 1.
2) The cell survival rate was measured by the CCK8 method using a detection kit (CCK-8 cell proliferation and cytotoxicity detection kit, japan Co., ltd.) according to the instructions, and the results are shown in FIG. 2;
3) And the generation of Reactive Oxygen Species (ROS) is detected by using a reactive oxygen species detection kit (Biyunyan, product number: S0033S) and a microplate reader according to an operation instruction, and the result is shown in figure 3.
As can be seen from FIGS. 1 to 3, the content of AGEs is the least, the survival rate of cells is the highest, and the content of reactive oxygen species ROS is the least after the delphinidin-3-O-galactoside action, which is closest to the control group.
Test example 3 protective action of inhibiting AGEs on umbilical vein endothelial cell injury
The experimental principle is as follows: HUVEC cells cultured in vitro were randomly divided into a control group, a model group (200. Mu.g/mL AGEs), an delphinidin-3-O-glucoside group, an delphinidin-3-O-galactoside group, an delphinidin-3-O-arabinoside group, an delphinidin-3-O-rutinoside group, a petunidin-3-O-rutinoside (P-coumaroyl) -5-O-glucoside group, and a blueberry mixed anthocyanin group. The obtained mixture is pretreated by 100 mu mol/L of each group of anthocyanin for 8 hours, and then 200 mu g/mL of AGEs are added to act on endothelial cells for 24 hours.
And (3) testing:
1) Propidium Iodide (PI) is a nuclear staining reagent capable of staining DNA, and can release red fluorescence after being embedded into double-stranded DNA. PI cannot pass through a living cell membrane, but only penetrates through a broken cell membrane to stain nuclei. Therefore, PI is often used in conjunction with nuclear fluorescent probes such as DAPI to stain both live and dead cells. When PI and DAPI and other nuclear dyes are co-dyed, DAPI can be taken up by living cells and combined with DNA to be blue fluorescence under ultraviolet light; while PI stains dead cells to produce red fluorescence. The test results are shown in FIG. 4.
2) The expression levels of inflammatory factors TNF-. Alpha.ICAM-1 and VCAM-1 were detected using qPCR using a detection Kit (Biyun day, beyofast. RTM. SYBR Green One-Step qRT-PCR Kit) according to the instructions, and the test results are shown in FIGS. 5 to 7.
3) And performing mitochondrial related ATP index detection by using an ATP Assay Kit (ATP Assay Kit) (Biyunyan, product number S0026) according to an operation instruction, wherein a test result is shown in figure 8.
4) The enzyme activity data of endothelial cell function NO (Biyuntian, product number S0021) and eNOS (eNOS enzyme-linked immunosorbent assay kit, nanjing institute for bioengineering) are detected by using the detection kit according to the instruction, and the test results are shown in Table 5.
TABLE 5 endothelial cell function
Figure 959754DEST_PATH_IMAGE009
Remarking: comparison with control groupP<0.05(ii) a * In comparison with model groupP<0.05(ii) a A ratio of other anthocyaninsP<0.05。
As can be seen from FIG. 4, the least apoptosis rate after delphinidin-3-O-galactoside action was closest to the control group; as can be seen from FIGS. 5 to 7, the expression of each inflammatory factor was minimal after the action of delphinidin-3-O-galactoside, closest to the control group; as can be seen in FIG. 8, delphinidin-3-O-galactoside acted with the highest mitochondrial content, closest to the control group.
Test example 4 animal experiments (Delphinidin-3-O-galactoside acute toxicity experiment)
The experimental standard is as follows: the experiment was carried out according to the method specified in GB 15193.3-2014 "national Standard for food safety acute oral toxicity test".
Animal grouping: 20 SPF-grade SD rats are half male and female, and the individual values of the same sex individual weight are within the range of average +/-20%.
The experimental method comprises the following steps: weighing a proper amount of sample, adding a proper amount of deionized water, and fully and uniformly mixing to prepare a test object solution with the concentration of 0.25 g/mL. Before the test, animals were fasted overnight (about 16 h) and allowed free access to water. In the actual test, the test animals are gavaged with 20 mL/kg body weight to give the test solution, and the test solution is gavaged in 24h for 2 times, wherein the interval between two gavages is about 4h, a small amount of feed is given in the period, and after the test object is given for the first time, the test animals continue to fast for about 3 h. After administration of the test subjects, the time of appearance and disappearance of signs of intoxication and the time of death were observed and recorded for a period of 14 days. The dead animals should be roughly dissected and visually examined for further histopathological examination if the abnormal tissue or organs are found. Weighing was carried out on days 0, 1, 3, 7 and 14, respectively.
The experimental results are as follows: the animals have no abnormal symptoms during the period of sample administration and the period of observation of 14 d, the weight is normally increased, no animal death is seen, and LD50 is more than 10 g/kg weight. At the end of the test, all animals were roughly dissected and visually observed without abnormality.
The experimental conclusion is that: the sample is detected according to GB 15193.3-2014 acute oral toxicity test of national standard for food safety, the acute oral toxicity LD50 of SD rats is more than 10 g/kg body weight, and the sample is practically nontoxic.
Test example 5 animal experiments (Delphinidin-3-O-galactoside and blood AGEs level)
The experimental method comprises the following steps:
animal grouping: 2-month-old mice were grouped by body weight and randomly divided into 1 model group, delphinidin-3-O-glucoside group, delphinidin-3-O-arabinoside group, delphinidin-3-O-galactoside group, delphinidin-3-O-rutinoside group, petunidin-3-O-rutinoside (p-coumaroyl) -5-O-glucoside, blueberry mixed anthocyanin and 1 blank control group.
Preparing a feed: common feed: SPF grade maintenance feed; high AGEs feed: will be provided with 60 The SPF-grade maintenance feed sterilized by Co irradiation is baked at 160 ℃ for 40 minutes to prepare a high advanced glycation end product feed (high AGEs feed), and the concentration of carboxymethyl lysine and carboxyethyl lysine in the high AGEs feed is determined to be more than 2 times of that of a control feed.
The experimental method comprises the following steps: the samples were dissolved in purified water to prepare 1mg/mL stock solutions and administered by gavage at a dose of 0.9-3 mg/kg. Wherein the model group and the sample group were given high AGEs feed and the blank control group was given normal feed for 1 month. Meanwhile, the sample group is perfused with the tested sample, and the model group and the blank control group are dosed with the same volume of solvent. Taking 0.4mL of tail tip blood before intervention as a baseline sample, killing animals at the end of the experiment, taking 0.4mL of blood sample, and detecting the content of conjugated Carboxymethyllysine (CML) and conjugated Carboxyethyllysine (CEL) in the blood pressure sample by using a UPLC-MS method.
The experimental results are as follows: CML and CEL in blood of model mice were significantly elevated compared to the blank control group (P < 0.05); compared with a model group, the delphinidin-3-O-galactoside and the cowberry mixed anthocyanin can obviously reduce the CML and CEL content (P is less than 0.05) in blood, and the effect of the delphinidin-3-O-galactoside is obviously better than that of the cowberry mixed anthocyanin (P is less than 0.05); petunidin-3-O-rutinoside (p-coumaroyl) -5-O-glucoside can significantly reduce the CML content in blood, but the change in CEL content is not significant. delphinidin-3-O-glucoside, delphinidin-3-O-arabinoside and delphinidin-3-O-rutinoside have no significant change (P > 0.05) to AGEs in mouse blood.
And (4) experimental conclusion: delphinidin-3-O-galactoside can significantly reduce AGEs in mouse blood, and is significantly superior to other sample groups.
Test example 6 animal experiments (delphinidin-3-O-galactoside and type II diabetes)
Based on the fact that the increase of the levels of AGEs is a risk factor of diabetes and complications, and AGEs is an independent risk factor causing insulin resistance, the application further researches the relationship between delphinidin-3-O-galactoside and type II diabetes.
Experimental animal models and groups: streptozotocin (STZ, product of Sigma, dissolved in citrate buffer at pH4.5 at a concentration of 0.1 mmol/L, ready for use) was injected into the lower left abdominal cavity of rats at a dose of 60 mg/kgBW. The normal control group was injected with an equal volume of citrate buffer. After 72 hours, the tail blood of the rat is taken for measuring the blood sugar, and the rat with the blood sugar concentration of more than 16.7mmol/L is taken as the diabetic rat. Diabetic rats were randomly divided into 1 model group, delphinidin-3-O-galactoside group, and 1 blank control group.
The administration method comprises the following steps: the samples were dissolved in purified water to prepare 1mg/ml stock solutions and administered by gavage at a dose of 0.9-3 mg/kg. The sample group was administered with the test sample, the model group and the blank control group with the same volume of solvent by gavage.
Detection indexes are as follows: the main measurement indexes include body weight, glucose tolerance, fasting Blood Glucose (FBG), serum Insulin (INS) content, etc.
The experimental results are as follows: delphinidin-3-O-galactoside can reduce FBG level and serum INS content in type II diabetic rat obviously and improve weight loss and insulin resistance of type II diabetic rat.
Test example 7 animal experiments (delphinidin-3-O-galactoside and Alzheimer's disease)
Based on the close correlation between the increase of AGEs level and the decline of cognitive ability of the old, the relation between delphinidin-3-O-galactoside and Alzheimer's disease is further researched.
Experimental animal models and groups: 3x Tg-AD mice (Psen 1tm1Mpm Tg (APPSwe, tauP 301L) triple-conversion AD mice on C57BL6 background), 4-month-old AD mice were randomly divided into 1 model group, delphinidin-3-O-galactoside group, and 1 blank control group.
The administration method comprises the following steps: the samples were mixed into the mouse feed at a dose of 0.9-3mg/kg and administered by free feeding for a period of 3 months. The sample group was given the feed to which the test sample was added, and the model group and the blank control group were given the maintenance feed to which the placebo was added.
Detection indexes are as follows: the learning and memory level of each group of mice is detected by adopting behavioural experiments such as Morris water maze, diving platform and dark avoidance; and detecting related pathological indexes of Abeta and Tau in the brain of the mouse.
The experimental results are as follows: delphinidin-3-O-galactoside can improve the learning and memory ability of AD mouse obviously, lower the expression level of brain A beta and Tau related pathological protein and improve Alzheimer's disease.
Test example 8 animal experiments (Delphinidin-3-O-galactoside and Atherosclerosis)
Based on the elevated AGEs levels and the close correlation between endothelial cell damage and atherosclerosis, the present application further investigated the relationship between delphinidin-3-O-galactoside and atherosclerosis.
Experimental animal models and groups: c57BL/6J mice of males at 4 weeks of age were fed with a high-fat diet for 12 weeks to establish an atherosclerosis model. The atherosclerosis model mice were randomly divided into 1 model group, delphinidin-3-O-galactoside group, and 1 blank control group.
The administration method comprises the following steps: the samples were dissolved in purified water to prepare 1mg/ml stock solutions and administered by gavage at a dose of 0.9-3 mg/kg. The sample group was administered with the test sample, the model group and the blank control group with the same volume of solvent by gavage. Prophylactic administration was started from 6 weeks after the start of high-fat diet feeding for 6 weeks.
Detection indexes are as follows: detecting mouse blood lipid level, inflammation and active oxygen index (ROS, TNF alpha, ICAM, VCAM) and the like.
The experimental results are as follows: delphinidin-3-O-galactoside can significantly improve blood lipid metabolism of atherosclerotic mice, including reduction of TC and LDL-C levels; by reducing the levels of ROS and inflammatory factors (TNF alpha, ICAM and VCAM), the endothelial function of the mice is improved, thereby slowing the occurrence and the development of atherosclerosis.
Therefore, as can be seen from the results of the above tests, delphinidin-3-O-galactoside has a superior effect in inhibiting AGES, treating AGES-associated injuries, diseases or symptoms, or alleviating AGES-associated injuries, diseases or symptoms, relative to other test groups; compared with cowberry mixed anthocyanin, the single-component substance has the effect of inhibiting AGES generation more stably and controllably.
The present application is not limited to the above embodiments. The above embodiments are merely examples, and embodiments having substantially the same configuration as the technical idea and exhibiting the same operation and effect within the technical scope of the present application are included in the technical scope of the present application. In addition, various modifications that can be conceived by those skilled in the art are applied to the embodiments and other embodiments are also included in the scope of the present application, in which some of the constituent elements in the embodiments are combined and constructed, without departing from the scope of the present application.

Claims (12)

1. The delphinidin-3-O-galactoside is applied to preparation of medicines or health-care foods, wherein the medicines or the health-care foods are medicines or health-care foods for inhibiting AGEs, medicines or health-care foods for treating AGEs-related injuries, diseases or symptoms, or medicines or health-care foods for relieving AGEs-related injuries, diseases or symptoms.
2. The use of claim 1, wherein the medicament further comprises pharmaceutically acceptable excipients and the health food further comprises dietetically acceptable excipients.
3. The use according to claim 1, wherein the medicament or health food is in the form of an oral preparation or an injectable preparation.
4. The use according to claim 3, wherein the oral formulation is a tablet, powder, capsule, granule, pill, powder, paste, solid beverage or oral liquid.
5. The use of claim 3, wherein the oral formulation comprises delphinidin-3-O-galactoside in an amount of 0.013% or more by weight.
6. The use of claim 3, wherein the injectable formulation is an injectable solution or powder for injection.
7. The use as claimed in claim 3, wherein the mass content of delphinidin-3-O-galactoside in the injectable preparation is not less than 0.013%.
8. The use according to any one of claims 1 to 7, wherein the inhibition of AGEs is the inhibition of AGEs production or the promotion of AGEs degradation.
9. The use according to any one of claims 1 to 7, wherein the AGEs-related damage is AGEs-mediated increase in apoptosis, increase in active oxygen content, overexpression of the inflammatory factor TNF- α, overexpression of the inflammatory factor ICAM-1, overexpression of the inflammatory factor VCAM-1 and/or decrease in mitochondrial ATP content.
10. The use according to any one of claims 1 to 7, wherein the AGEs-related diseases are any one or more of cardiovascular and cerebrovascular diseases, neurodegenerative diseases, osteoarticular diseases.
11. The use according to claim 10, wherein the cardiovascular and cerebrovascular diseases are selected from diabetes and its complications, atherosclerosis; and/or the neurodegenerative disease is selected from alzheimer's disease, parkinson's disease, amyotrophic lateral sclerosis; and/or the osteoarticular disease is osteoporosis, osteoarthritis.
12. The use according to any one of claims 1 to 7, wherein the AGEs-related symptoms are aging.
CN202211588207.6A 2022-12-12 2022-12-12 Application of delphinidin-3-O-galactoside in preparation of medicine or health food Pending CN115645430A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202211588207.6A CN115645430A (en) 2022-12-12 2022-12-12 Application of delphinidin-3-O-galactoside in preparation of medicine or health food

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202211588207.6A CN115645430A (en) 2022-12-12 2022-12-12 Application of delphinidin-3-O-galactoside in preparation of medicine or health food

Publications (1)

Publication Number Publication Date
CN115645430A true CN115645430A (en) 2023-01-31

Family

ID=85018090

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211588207.6A Pending CN115645430A (en) 2022-12-12 2022-12-12 Application of delphinidin-3-O-galactoside in preparation of medicine or health food

Country Status (1)

Country Link
CN (1) CN115645430A (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005077176A1 (en) * 2004-02-11 2005-08-25 The Trustees Of Columbia University In The City Of New York Anthocyanin compounds and methods of use thereof
CN108409805A (en) * 2018-05-31 2018-08-17 浙江大学 A kind of isolation and purification method of delphinidin -3-O- galactosides and its application
US20220322719A1 (en) * 2016-10-27 2022-10-13 Nse Products, Inc. Intestinal health promoting compositions

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005077176A1 (en) * 2004-02-11 2005-08-25 The Trustees Of Columbia University In The City Of New York Anthocyanin compounds and methods of use thereof
US20220322719A1 (en) * 2016-10-27 2022-10-13 Nse Products, Inc. Intestinal health promoting compositions
CN108409805A (en) * 2018-05-31 2018-08-17 浙江大学 A kind of isolation and purification method of delphinidin -3-O- galactosides and its application

Similar Documents

Publication Publication Date Title
KR100908626B1 (en) Method of utilizing physiological activity of rare saccharide and compositions containing rare saccharide
KR101089957B1 (en) Composition comprising arabinose from arabinoxylan for prevention and treatment of ischemic diseases and degenerative brain diseases
KR101627001B1 (en) Cognitive disorder-ameliorating agent
KR101946526B1 (en) Composition of preventing or improving UV-induced skin damage comprising hydrangenol
KR101965061B1 (en) Composition for prevention or treatment of metabolic disorder comprising peanut sprout extracts and fractions thereof
KR101332074B1 (en) Composition Comprising Esculetin for Inhibition of Bone Loss
CN115645431B (en) Composition and application thereof
JP2023009106A (en) Composition for protecting retinal nerve cells containing processed bilberry fruit and processed pine bark
CN115645430A (en) Application of delphinidin-3-O-galactoside in preparation of medicine or health food
CN115581702A (en) Application of paeoniflorin-3-O-arabinoside in preparation of medicines or health foods
KR101051085B1 (en) Parkinson&#39;s disease prevention and treatment composition containing cinnamon extract, fractions thereof or trans-cinnaaldehyde isolated from cinnamon as an active ingredient
CN115590874A (en) Application of malvidin-3-O-glucoside in preparation of medicines or health-care foods
CN115645429B (en) Anthocyanin composition and application thereof
KR20200066443A (en) A pharmaceutical composition for preventing or treating aging or an aging-related disease comprising an anthocyanin-polysaccharide complex as an active ingredient
KR20160081189A (en) Composition comprising an extract of Eisenia bicyclis for preventing and treating Alzheimers disease
KR101526435B1 (en) Compositions for skin-whitening comprising extract of Vitis amurensis ruprecht
KR102357449B1 (en) Composition for preventing or protecting liver damage comprising dieckol against methylglyoxal
KR20180085204A (en) Composition for prevention and treatment of liver disease or obesity comprising Rheum undulatum Linne extract and Glycyrriza uralensis Fischer extract as active ingredients
KR101722367B1 (en) Composition comprising an extract of Eisenia bicyclis for preventing and treating Alzheimers disease
KR20170068221A (en) A composition for antioxidant and inflammatory neurodegenerative diseases comprising fagopyrum tataricum extract
Cheung Impact of In Vitro Digestion on Pecan [Carya illinoinensis (Wangenh.) K. Koch] Phenolics and Evaluating Their Absorption Using Caco-2 Monolayers
KR20240131928A (en) Albumin Nanocomposite Comprising Phytochemical and Composition for Improving Muscle Disease Comprising Same
KR101348411B1 (en) A composition for treatment or prevention of diabetic complications or antiaging comprising brandioside or pheliposide
KR101429861B1 (en) Cosmetic composition for anti-wringkle containing lithospermate derivative
JP2024022503A (en) Nampt-activating agent

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