CN114209680A - Application of astaxanthin in preparation of medicine for treating vascular dementia - Google Patents

Application of astaxanthin in preparation of medicine for treating vascular dementia Download PDF

Info

Publication number
CN114209680A
CN114209680A CN202111405583.2A CN202111405583A CN114209680A CN 114209680 A CN114209680 A CN 114209680A CN 202111405583 A CN202111405583 A CN 202111405583A CN 114209680 A CN114209680 A CN 114209680A
Authority
CN
China
Prior art keywords
astaxanthin
vascular dementia
group
composition
mice
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.)
Withdrawn
Application number
CN202111405583.2A
Other languages
Chinese (zh)
Inventor
朱宁伟
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.)
Zhejiang University ZJU
Original Assignee
Zhejiang Medical College
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 Zhejiang Medical College filed Critical Zhejiang Medical College
Priority to CN202111405583.2A priority Critical patent/CN114209680A/en
Publication of CN114209680A publication Critical patent/CN114209680A/en
Withdrawn legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • A61K31/122Ketones having the oxygen directly attached to a ring, e.g. quinones, vitamin K1, anthralin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Biomedical Technology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Epidemiology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

Compared with the prior art, the experimental research of the invention shows that the astaxanthin can reduce the neurotoxic protein A beta in the brain42The expression and the inhibition of inflammatory reaction in the brain, thereby playing a role in protecting neurons and improving the learning and memory ability. Therefore, astaxanthin is used for treating vascular dementia and has a unique mechanism.

Description

Application of astaxanthin in preparation of medicine for treating vascular dementia
Technical Field
The invention belongs to the field of new application of astaxanthin, and particularly relates to application of astaxanthin in preparation of a medicine for treating vascular dementia.
Background
Vascular dementia (VaD) is a deterioration in brain cell function primarily caused by cerebrovascular disease, and is the second common type of dementia following senile dementia. Oxidative stress injury and neuroinflammatory response caused by low cerebral blood flow perfusion in patients with VaD are important factors causing cognitive dysfunction of patients. In recent years, the incidence of cerebrovascular diseases is increasing, and researchers at home and abroad pay more and more attention to the research of VaD. At present, the pathogenesis of VaD is not completely clear, and the research of effective VaD drug treatment strategies has become the focus of international attention today.
Astaxanthin (AST) is a natural carotenoid existing in marine organisms such as fish, shrimp, crab and algae, and has strong biological activities such as oxidation resistance and inflammation resistance. The unique chemical structure of AST makes it easy to penetrate blood brain barrier to treat central nervous system diseases.
Disclosure of Invention
The purpose of the invention is as follows: aiming at the technical problems, the invention provides the application of astaxanthin in preparing the medicine for treating vascular dementia.
The technical scheme is as follows: in order to achieve the purpose of the invention, the technical scheme adopted by the invention is as follows:
application of astaxanthin in preparing medicine for treating vascular dementia is provided.
The chemical name of the astaxanthin is 3,3 ' -dihydroxy-4, 4 ' -diketone-beta, beta ' -carotene, and the chemical formula is C40H52O4CAS registry number 472-61-7.
Further, the present invention also provides:
the astaxanthin is applied to the preparation of the medicine for protecting cerebral neuron damage caused by ischemia-reperfusion in vascular dementia.
Application of astaxanthin in preparation of A beta for reducing vascular dementia42Application in protein expression drugs.
The astaxanthin is applied to the preparation of medicines for inhibiting intracerebral inflammatory reaction in vascular dementia.
A composition for treating vascular dementia, which comprises astaxanthin or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable auxiliary material.
Preferably, the dosage form of the composition is selected from one of tablets, powders, granules, capsules, oral liquid, sustained release preparations, injections, infusion solutions and sterile powder for injection.
Use of a composition comprising astaxanthin in the manufacture of a medicament for the treatment of vascular dementia.
Further preferred is:
the application of the composition containing astaxanthin in preparing medicines for protecting cerebral neuron damage caused by ischemia-reperfusion in vascular dementia is provided.
Application of composition containing astaxanthin in preparation of medicine for reducing A beta in vascular dementia42Application in protein expression drugs.
The application of the composition containing astaxanthin in preparing medicines for inhibiting intracerebral inflammatory reaction in vascular dementia.
Has the advantages that: compared with the prior art, the experimental research shows that the astaxanthin can reduce the brain neurotoxicity protein Abeta42The expression and the inhibition of inflammatory reaction in the brain, thereby playing a role in protecting neurons and improving the learning and memory ability. Therefore, astaxanthin is used for treating vascular dementia and has a unique mechanism.
Drawings
FIG. 1 shows the results of Neisseria staining of hippocampal tissues of mice in each group: 1a, sham group; 1B, model group; astaxanthin low dose group; astaxanthin high dose group; large scale 200 μm; the small scale indicates 50 μm, the neural cells containing nissl are blue, and n is 15.
FIG. 2 shows the hippocampal tissue Abeta of each group of mice42Immunohistochemical staining results: 1a, sham group; 1B, model group; astaxanthin low dose group; astaxanthin high dose group; the large icon indicates 200 μm; the small scale in the black box indicates 50 μm, the brown region is the positive expression region of a β 42 protein, and n is 15.
FIG. 3 shows Western blotting analysis of hippocampal tissue Abeta of each group of mice42Protein expression: western blotting result; B. quantitative analysis (F: 53.312, P: 0.000), n: 15, aP compared to sham group<0.05; bP compared to model group<0.05; cP compared to astaxanthin high dose group<0.05。
FIG. 4 shows IL-4, IL-6 and Abeta of hippocampal tissues of each group of mice42And (3) ELISA detection results: quantitative comparison of il-4 (F-65.613, P-0.000); quantitative comparison of il-6 (F-72.124, P-0.000); C.A.beta.42Quantitative comparison results (F ═ 81.425, P ═ 0.000); aP compared to sham group<0.05; bP compared to model group<0.05; cP compared to astaxanthin high dose group<0.05;n=10。
Detailed Description
The technical solutions of the present invention are further described in detail by the following specific examples, but it should be noted that the following examples are only used for describing the content of the present invention and should not be construed as limiting the scope of the present invention.
Example Experimental study of astaxanthin to improve learning and memory abilities of mice with vascular dementia
Materials and methods
1. Experimental Material
60 healthy male ICR cleaning-grade mice with the weight of 30-35 g are purchased from the center of Zhejiang experimental animals with the license number SCXK (Zhe) 2014-0001. AST (Shanghai Allantin reagent Co., Ltd., batch No. A114383, content not less than 98%), interleukin (interleukin, IL) -4, IL-6 cytokine determination kit (batch Nos. M150929-002a, M150826-004a, respectively, all purchased from Shenzhen Xin Bosheng Biotech Co., Ltd.), rabbit anti-mouse beta-amyloid peptide 42(amyloid beta-peptides42, Abeta. beta.) (Abeta.)42) Antibody (batch number: ab10148, abcam), streptavidin-biotin-peroxidase complex technology (SABC), immunohistochemical staining kit, DA color development kit (wuhan bosch & d bioengineering ltd, lot numbers SV1022, 12J25C22, respectively).
2. Method of producing a composite material
2.1 establishment and experimental grouping of mouse VaD model: the mouse VaD model is established by clamping bilateral common carotid arteries, and the specific process is briefly described as follows: 10% chloral hydrate (300mg/kg) is injected into an abdominal cavity to anaesthetize a mouse, the mouse is fixed in a supine position, an incision is made in the middle of the neck, muscles and fascia are separated along the intramuscular lateral margin of the sternocleidomastoid to find two lateral carotid sheaths, the common carotid artery is clamped by a non-invasive artery clamp after the common carotid artery and the vagus nerve are stripped, so that the double-sided common carotid artery is blocked to cause global cerebral ischemia, the artery clamp is removed after 20min, and the vagus nerve close to the common carotid artery is prevented from being damaged in the operation. After a model is established for 24 hours, randomly dividing mice in an operation group into a sham operation group (sham), a model group (VaD), an AST low-dose group (15mg/kg, AST low-dose group) and an AST high-dose group (30mg/kg, AST high-dose group) and 15 mice in each group, and respectively carrying out intraperitoneal injection on astaxanthin vegetable oil solution with corresponding dose for 3 times per week for 6 weeks; the sham group only exposes bilateral carotid arteries separately, but does not clamp, and the rest of the procedure is the same as the surgical group. Sham and model mice were given equal amounts of vegetable oil.
2.2 training and positioning navigation and space exploration experiments: after 6 weeks, each group of experimental animals should be trained, positioned, navigated and subjected to space exploration experiments by using a water maze device. The Morris water maze mainly comprises a circular water pool and an automatic video recording and analyzing system, swimming tracks of mice can be recorded dynamically in real time, each group of mice are placed in the Morris water maze to search for a platform, the water maze is manually divided into 4 quadrants to record the time of the mice for searching for the platform within 2min, if the mice do not find the platform within 2min, the mice are guided to the platform by an experimenter and stay for 60s, the escape latency is recorded as 2min, and the mice are trained repeatedly for 5 d. Data are exported through experimental video analysis software, and the average escape latency is analyzed (the space acquisition capacity of the mouse is predicted, and the shorter the average escape latency is, the better the space acquisition capacity of the mouse is). And (3) removing the platform in the water maze after the 5-day water maze experiment is finished, allowing each group of mice to enter the water maze from the opposite side quadrant of the quadrant where the original platform is located, and measuring the times of crossing the original platform in 2min and the time of staying of the mice in each quadrant of the platform by using a Smart system so as to judge the memory storage and extraction reproduction capability of the mice.
2.3 Niey staining: after the behavioral testing of all experimental animals was completed, the rats were sacrificed by intraperitoneal injection of 10% excess chloral hydrate. Taking out whole brain, cutting coronal section by section, cutting off brain tissue before visual cross and after cerebellum, embedding and continuously slicing the left middle brain tissue, numbering the brain tissue slices of each group, selecting slices with the same serial number for Nie dyeing, dewaxing xylene, dehydrating with gradient ethanol, adding distilled water, dyeing neuron Nie body according to standard steps shown in Nie dyeing kit specification, dehydrating with anhydrous ethanol, removing xylene, sealing with neutral gum, and observing under an optical microscope.
2.4 Aβ42Immunohistochemical staining: when immunohistochemical staining was performed, 5 μm sections of the same sequence number were selected and the sections were guaranteed to have a classical hippocampal structure (contiguous CA1, CA3 and DG 3 regions). Taking the hippocampal tissue slices, and carrying out Abeta according to the specification of the SABC immunohistochemical staining kit42Immunohistochemical staining, the specific steps are briefly described as follows: tissue sections are processed by absolute ethyl alcohol until the gradient concentration ethanol is obtainedAdding distilled water for hydration, and incubating with 3% hydrogen peroxide at room temperature for 30min to inactivate endogenous peroxidase; washing with PBS for 20 min. Adding blocking solution (5% sheep serum + 1% BSA + 0.05% Tween-20) to block the slide for 30min at room temperature; removing excess blocking solution, adding rabbit anti-mouse Abeta42Polyclonal antibody (1:200), incubating at room temperature for 1h, washing with PBS for 10min, adding biotinylated goat anti-rabbit IgG (1:200), incubating at room temperature for 30min, and washing with PBS for 10 min. Adding HRP-labeled ovalbumin, and incubating for 30 min. And (3) after the PBS is washed, DAB is added for color development, ethanol is gradually dehydrated, and the neutral resin is sealed. Blank control was performed with PBS instead of primary antibody. Serial histological sections with similar cross-sectional levels were taken from each mouse for observation.
2.5 Western blotting analysis of Abeta42Expressing: 15 mice were taken from each group, brain hippocampal tissue was isolated, total protein was extracted from RIPA lysate, and the total protein was measured with BCA kit. Taking 70 mu g protein sample, 15 mu l system, adding rabbit anti-mouse Abeta after electrophoresis, membrane transfer and sealing42Incubating the antibody (1:500) at 4 ℃ overnight, adding goat anti-rabbit secondary antibody (1:3000) for hybridization reaction for 1h, incubating at room temperature for 2h, washing with membrane washing buffer (TBST) for 3 times, each time for 10min, and developing. Western blotting imaging was analyzed by quantitative One 4.6.2 software, and the relative expression of each histone and internal reference GAPDH was calculated.
2.6 Hippocampus brain tissues IL-4, IL-6 and Abeta42Content determination: respectively taking the brain hippocampus tissues of each group of mice, weighing, adding pure water according to a ratio of 1: 9(m/V), ultrasonically homogenizing the tissues at low temperature, centrifuging at 4 ℃ and 10000r/min for 10min, and taking the supernatant for later use. Determination of IL-4, IL-6 and Abeta in Hippocampus tissues by ELISA kit42Protein content, the specific operation steps are strictly carried out according to the kit instructions.
3. Statistical analysis
Processing was performed with the SPSS 17.0 statistical software package. Data of the experiment are mean. + -. standard deviation
Figure BDA0003372097930000041
The mean comparison between two groups adopts t test, the comparison between multiple sample means adopts One-Way ANOVA test, the water maze data adopts repeated design variance analysis, and P is less than 0.05, and the difference is statisticThe meaning of learning.
Second, result in
1 Water maze test results
As shown in table 1, on days 4 and 5, the average escape latency of mice in the VaD group was significantly higher than that of the sham group, and the difference was statistically significant (P was less than 0.05), while the platform crossing times and residence time of mice in the VaD group were significantly lower than those of the sham group (P was less than 0.05); compared with the VaD group, mice in the different concentration AST intervention group had significantly decreased escape latency on both day 4 and day 5 (P both <0.05), while platform crossing times and residence time were significantly increased (P < 0.05); in addition, the avoidance latency was significantly decreased on both day 4 and day 5 of the AST high dose group (P both <0.05), and the number of platform crossings and residence time were significantly increased (P both <0.05) compared to the AST low dose group.
TABLE 1 mean escape latency and space exploration experimental results for each group of mice
Figure BDA0003372097930000051
Figure BDA0003372097930000052
1. The residence time of the mouse in the quadrant where the platform is located in the space exploration experiment accounts for the percentage of the total experiment time; aP <0.05 compared to sham group; bP <0.05 compared to model group; cP <0.05 compared to astaxanthin high dose group
2. Results of brain histopathology
The results of Niger staining of brain tissue of each group of mice are shown in FIG. 1. Compared with the sham group, after 6 weeks, neurons in CA1, CA3 and DG regions of hippocampal tissues of the brain of mice in the VaD group are obviously solidified and contracted and deeply stained, the neurons are obviously lost, gaps around the cells are enlarged and become vacuole-like lesions, and the cells are not arranged in a hierarchical manner. After the dry prognosis of AST treatment with different concentrations, the number of mouse brain tissue Neisseria staining positive neurons is obviously increased, the neuron shrinkage degree is reduced, the intercellular spaces are not vacuolated, particularly, the neurons in the CA1, CA3 and DG areas of mouse brain tissue hippocampus in an AST high-dose intervention group are clearly visible, and the neurons and the interstitial structures thereof are complete and approximate to the level of a sham group.
Immunohistochemical results for a β 42 protein in various regions of hippocampal brain tissue of each group of mice are shown in fig. 2. Abeta (beta)42The protein expression region was dark brown. A beta of each region of hippocampus of VaD group mice42The positive expression of the protein is obviously higher than that of the sham group; after the prognosis of different AST concentrations, Abeta of each region of mouse hippocampal tissues42The protein expression is obviously reduced compared with the VaD group, and the effect is obviously dose-dependent.
Western blotting analysis of Hippocampus tissues Abeta of various groups of mice42Protein expression
Hippocampus tissue Abeta of each group of mice42The Western blotting detection results of protein expression are shown in FIG. 3, and the statistical results show that compared with the sham group, the hippocampal Abeta of mice in the VaD group42Protein expression is significantly increased (P)<0.05); after different forms of AST treatment intervention, Abeta of mouse hippocampal region42The protein is obviously reduced compared with the VaD group (P is equal to<0.05), and the mice hippocampal tissue Abeta with AST high dose group prognosis42Protein expression is obviously reduced compared with AST low dose group and model group (P is equal to<0.05)。
4. Hippocampus cerebri tissue IL-4, IL-6 and Abeta42Results of content measurement
Hippocampus tissues IL-4, IL-6 and Abeta of each group of mice42The results of the content measurement are shown in FIG. 4. Mouse hippocampal IL-4 expression was significantly reduced in the VaD group compared to the sham group (P)<0.05), IL-6 and Abeta42Significantly increased expression (P)<0.05); compared with the VaD group, the IL-4 content of the hippocampal tissue of the mice subjected to different-concentration AST treatment prognosis is remarkably increased (P)<0.05), and IL-6 and Abeta42The content is remarkably reduced (P)<0.05), and the results are dose-dependent, AST high dose groups mice hippocampal IL-4, IL-6 and Abeta42The content was close to the sham group level.
The Morris water maze experiment utilizes the astronomy that a mouse does not like swimming and tends to escape water to search for a platform hidden in the water, and the mouse uses different surrounding markers to distinguish the position of the platform after continuous training, so that stable spatial cognitive memory is formed and is widely used for evaluating the learning and memory functions of the mouse. The main reason for the obvious difference of the average escape latency of the platforms of each group on the 4 th day and the 5 th day of the water maze is that as the training days increase, the sham group with normal learning and memory ability gradually forms stable memory for the platform, so that the time for searching the platform (average escape latency) is obviously shorter than that of the mice in the VaD group with impaired learning and memory ability and without stable memory for the platform (P < 0.05). The results of the space exploration experiments also further confirm that mice in the VaD group have obvious decline in learning and memory abilities compared with those in the sham group. In addition, consistent with previous literature reports, pathological results show that neuronal cells in hippocampal CA1, CA3 and DG regions are shrunk and the number of neurons positive for Niger staining is obviously reduced after 6 weeks in mice in VaD group, which indicates that ischemia reperfusion can cause the mice to be damaged in learning and memory functions. After AST treatment with different concentrations is used for prognosis, water maze and space exploration experiments show that the learning memory ability and the space memory ability of the mouse are obviously improved compared with those of a VaD group, the effect is dose-dependent, meanwhile, Niger dyeing shows that the quantity of hippocampal tissue neurons and Niger corpuscles of different dose intervention groups of AST is obviously higher than that of the VaD group, and the neurons are arranged in order, which shows that the AST can protect cerebral neuron damage caused by ischemia reperfusion, thereby improving the learning and cognitive dysfunction of VaD animals.
The present invention also initially investigates the molecular mechanism of AST treatment of VaD. Long-term hypoxic hypoperfusion of VaD results in high expression of a β. A beta is obtained by the proteolytic cleavage of beta Amyloid Precursor Protein (APP), and under pathological conditions, abnormal hydrolysis of APP leads to the increasing of A beta, and after aggregation, A beta oligomer is formed or high beta folding is aggregated to form insoluble A beta fiber or plaque, so that neurotoxicity is generated. A beta is mainly A beta40And Abeta42Two subtypes. And Abeta40In contrast, Abeta42Has the characteristics of low solubility, higher neurotoxicity and the like, and plays a key role in plaque polymerization. It has been shown that A.beta.42Deposition can activate microglia to release proinflammatory cytokines such as IL-6 and the like, so that astrocytes are activated, proliferated and released to inflammatory proteins, and A beta is promoted42Conversion to neurotoxic insolublePrecipitating to further activate microglia to release inflammatory factors so as to form a chronic inflammatory response. IL-4 is a major anti-inflammatory factor secreted by glial cells and is capable of providing a danger signal to neural stem cells, the level of which indirectly reflects the level of inflammation in the brain. Immunohistochemical staining and Western blotting analysis of groups of mice from 6 weeks later, and IL-4, IL-6 and Abeta42ELISA detection results show that the expression of hippocampal Abeta protein and the content of inflammatory factor IL-6 of mice in the VaD group are obviously higher than those in the sham group, and the content of anti-inflammatory factor IL-4 is lower than that in the sham group, which indicates that animals in the VaD group have obvious inflammatory response, and the results are consistent with the above documents. After different dose of AST treatment, the mouse hippocampal region Abeta42The protein expression and IL-6 content are obviously reduced compared with that of the VaD group, the IL-4 content is obviously increased, and the effect is dose-dependent, which indicates that the AST can reduce the A beta in the brain of the VaD mouse42The expression of protein and inflammation related factor IL-6 can raise the content of anti-inflammatory factor IL-4, reduce inflammation reaction and play the role of protecting neuron.
In conclusion, the research of the invention proves that astaxanthin can reduce A beta42The expression of the protein and the inhibition of inflammatory reaction in brain play a role in protecting neurons and improve the learning and cognitive functions of a mouse model with vascular dementia.

Claims (10)

1. Application of astaxanthin in preparing medicine for treating vascular dementia is provided.
2. The use according to claim 1, wherein the astaxanthin is used in the preparation of a medicament for protecting cerebral neuronal damage caused by ischemia-reperfusion in vascular dementia.
3. Use according to claim 1, wherein astaxanthin is used in the preparation of a composition for reducing A β in vascular dementia42Application in protein expression drugs.
4. The use according to claim 1, wherein the use of astaxanthin in the manufacture of a medicament for inhibiting an inflammatory response in the brain in vascular dementia.
5. A composition for treating vascular dementia, which comprises astaxanthin or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable auxiliary material.
6. The composition according to claim 5, wherein the composition is in a dosage form selected from one of tablets, powders, granules, capsules, oral liquids, sustained release preparations, injections, infusions, and sterile powders for injections.
7. Use of a composition comprising astaxanthin in the manufacture of a medicament for the treatment of vascular dementia.
8. The use according to claim 7, wherein the composition comprises astaxanthin for the preparation of a medicament for the protection of cerebral neuronal damage caused by ischemia-reperfusion in vascular dementia.
9. Use according to claim 7, wherein the composition comprising astaxanthin is used in the preparation of a composition for reducing A β in vascular dementia42Application in protein expression drugs.
10. Use according to claim 7, wherein the composition comprises astaxanthin for the preparation of a medicament for the inhibition of inflammatory reactions in the brain in vascular dementia.
CN202111405583.2A 2021-11-24 2021-11-24 Application of astaxanthin in preparation of medicine for treating vascular dementia Withdrawn CN114209680A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111405583.2A CN114209680A (en) 2021-11-24 2021-11-24 Application of astaxanthin in preparation of medicine for treating vascular dementia

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111405583.2A CN114209680A (en) 2021-11-24 2021-11-24 Application of astaxanthin in preparation of medicine for treating vascular dementia

Publications (1)

Publication Number Publication Date
CN114209680A true CN114209680A (en) 2022-03-22

Family

ID=80698171

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111405583.2A Withdrawn CN114209680A (en) 2021-11-24 2021-11-24 Application of astaxanthin in preparation of medicine for treating vascular dementia

Country Status (1)

Country Link
CN (1) CN114209680A (en)

Similar Documents

Publication Publication Date Title
US20130189382A1 (en) Use of fucoxanthin in the preparation of product for improving memory and having neuroprotective effect associated with neurodegenerative disorder
Hu et al. JD-30, an active fraction extracted from Danggui–Shaoyao–San, decreases β-amyloid content and deposition, improves LTP reduction and prevents spatial cognition impairment in SAMP8 mice
Wu et al. Hippocampal overexpression of TREM2 ameliorates high fat diet induced cognitive impairment and modulates phenotypic polarization of the microglia
CN110248646A (en) Slow releasing composition comprising pirfenidone is used to treat and reverse the medicinal usage of people&#39;s steatohepatitis (NAFLD/NASH)
CN104922164B (en) Pharmaceutical composite preparation for treating Alzheimer disease
Zhang et al. Therapeutic effect of a hydroxynaphthoquinone fraction on dextran sulfate sodium-induced ulcerative colitis
Li et al. Danggui buxue decoction ameliorates inflammatory bowel disease by improving inflammation and rebuilding intestinal mucosal barrier
Tan et al. Effect of chaihu shugan powder-contained serum on glutamate-induced autophagy of interstitial cells of cajal in the rat gastric antrum
CN111437323A (en) Application of vine tea extract in medicine for preventing and treating Alzheimer disease
CN116898970A (en) Application of ATP5D activator and B lymphocyte in preparation of medicines
CN114209680A (en) Application of astaxanthin in preparation of medicine for treating vascular dementia
Zhang et al. The role of CD38 in inflammation-induced depression-like behavior and the antidepressant effect of (R)-ketamine
CN1316976C (en) Use of kaempferol-3-O-glubulariacitrin in preparing medicine for senile dementia
CN103751197A (en) New use of stibene glucoside in preparing medicament for treating Alzheimer disease
CN103181954A (en) Medicine composition used for treating neurodegenerative disease as well as preparation method and application thereof
Zhang et al. Icariin upregulates phosphorylated cyclic adenosine monophosphate response element binding protein levels in the hippocampus of the senescence-accelerated mouse★
Ren et al. Neuroprotective effects of ZiBuPiYin recipe on db/db mice via PI3K-Akt signaling pathway by activating Grb2
CN107582574B (en) Application of nostoc commune alcohol extract in preparation of medicine for treating inflammatory bowel disease
CN105688031A (en) Application of compound traditional Chinese medicine preparation in treatment of Alzheimer&#39;s disease
CN109908151A (en) A kind of pentacyclic triterpene compound treats the application of the drug of inflammatory bowel disease in preparation
CN104083345A (en) Application of lycopene in preparation of medicine for prevention and treatment senile dementia
WO2023137672A1 (en) L-n-isobutyryl cysteine (l-nibc) -bound gold cluster au 18 (l-nibc) 14, compositions and applications thereof
Pellegrini et al. Targeting of NLRP3 inflammasome with a novel selective inhibitor as a suitable strategy for the pharmacological treatment of bowel inflammation
CN112830947B (en) Stilbene compounds isolated from Rheum lhasaense and their use in treating nervous system diseases
CN112791089B (en) Experimental method for influence of ellagic acid combined with autophagy inhibitor on aging of mouse model

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
WW01 Invention patent application withdrawn after publication

Application publication date: 20220322

WW01 Invention patent application withdrawn after publication