CN116440179A - Application of kaurene extract in preparation of anti-abdominal aortic aneurysm products - Google Patents

Application of kaurene extract in preparation of anti-abdominal aortic aneurysm products Download PDF

Info

Publication number
CN116440179A
CN116440179A CN202310584879.8A CN202310584879A CN116440179A CN 116440179 A CN116440179 A CN 116440179A CN 202310584879 A CN202310584879 A CN 202310584879A CN 116440179 A CN116440179 A CN 116440179A
Authority
CN
China
Prior art keywords
abdominal aortic
inflammatory
extract
application
inhibiting
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
CN202310584879.8A
Other languages
Chinese (zh)
Inventor
祁荣
张庆义
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.)
Peking University
Original Assignee
Peking University
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 Peking University filed Critical Peking University
Priority to CN202310584879.8A priority Critical patent/CN116440179A/en
Publication of CN116440179A publication Critical patent/CN116440179A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/13Coniferophyta (gymnosperms)
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2236/00Isolation or extraction methods of medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicine
    • A61K2236/30Extraction of the material
    • A61K2236/33Extraction of the material involving extraction with hydrophilic solvents, e.g. lower alcohols, esters or ketones
    • A61K2236/333Extraction of the material involving extraction with hydrophilic solvents, e.g. lower alcohols, esters or ketones using mixed solvents, e.g. 70% EtOH
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

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

Abstract

The invention discloses application of kaurena extract in preparing an anti-abdominal aortic aneurysm product, and belongs to the field of medicines. The invention discloses a new application of a kaurene extract (Agathis dammara extract, AD) and a monomer of Araucarone (AO) in medicaments. The new application is application of kaurene extract and monomer sequoyitol in resisting abdominal aortic aneurysm. The invention adopts an abdominal aortic aneurysm model related to clinic: porcine pancreatic elastase-induced mouse abdominal aortic aneurysm; experiments prove that the kaurene extract and the monomer of the kaurene can obviously inhibit the occurrence and development of the mouse abdominal aortic aneurysm induced by elastase.

Description

Application of kaurene extract in preparation of anti-abdominal aortic aneurysm products
Technical Field
The invention relates to the field of medicines, in particular to application of kaurene extract in preparation of an anti-abdominal aortic aneurysm product.
Background
Kauri (Latin name Agath dammara (lamb.) Rich), arbor, taxonomically belonging to the phylum gymnosperm, class Pinus, order Pinus, family Cunninghamiae, genus kauri, mainly distributed in tropical Asia, and introduced and cultivated in Xiamen, fuzhou, etc. of China. The volatile oil in the leaves mainly contains terpenoid compounds, wherein the beta-cubene is the main component, and the volatile oil also contains compounds such as alkane, alcohol, ketone, a small amount of aldehyde, ether, ester and the like, so that the volatile oil has important application in medicine, spice, daily chemicals and the like, and has important significance in phytochemical taxonomy. The pharmacological actions of kaurene can be broadly divided into the following aspects: antitumor, antibacterial, antiinflammatory, tranquilizing, and antiasthmatic.
Abdominal aortic aneurysm (abdominal aortic aneurysm, AAA) refers to the largest diameter of the abdominal aorta exceeding 30mm under imaging diagnosis such as ultrasound or CT, and is usually found in the infrarenal section of the abdominal aorta. Risk factors for abdominal aortic aneurysms include age, male, smoking, family history of AAA, hypertension, dyslipidemia, and other cardiovascular diseases such as ischemic heart disease and peripheral arterial disease, among others. Global abdominal aortic aneurysm the overall prevalence of (2) is 4.8%, and with the global trend of obesity, aging of the population and its associated metabolic syndrome, the incidence of AAA is on the rise worldwide. AAA is also characterized by underlying disease, and is usually asymptomatic in the early stages of the disease, sometimes accompanied by only minor abdominal, back, or leg pain. Thus, aneurysms are often burst without precursor symptoms, progress rapidly, resulting in very high mortality (-80%). Therefore, AAA is also one of the main causes of sudden death.
The pathogenesis of AAA mainly involves inflammation and oxidative stress of the membranous layer in the vessel wall, thereby causing local inflammatory cell infiltration, leading to the generation of local inflammatory microenvironments. Local inflammation of the blood vessels can lead to inflammatory states and apoptosis of vascular smooth muscle cells, and dysfunction of synthesis and secretion of extracellular matrix, thereby leading to changes in the wall structure of the blood vessels and initiation of AAA formation. For large or symptomatic AAA, the current mainstay of treatment is vascular replacement or intravascular stents. Although AAA surgery is mature, there are problems such as high surgical risk, high treatment cost, fewer adaptation groups, and the like. Moreover, for small and medium-sized AAA or AAA patients unsuitable for surgical treatment, there is no active treatment other than regular monitoring. Therefore, the search for drugs that inhibit the development of AAA has become an important research in the field of prevention and control of cardiovascular diseases.
There is no report of the effects of kaurene and its monomer components on vascular smooth muscle cell inflammatory response and AAA related effects.
Disclosure of Invention
The invention aims to provide a new application of a kauri pine extract, and experiments show that the kauri pine extract and a monomer, namely, the kauri pine ketone (AO) can treat abdominal aortic aneurysm.
The invention firstly provides application of kaurena extract in preparing a product for preventing and/or treating abdominal aortic aneurysm.
The abdominal aortic aneurysm may be a infrarenal abdominal aortic aneurysm.
More specifically, the abdominal aortic aneurysm is a porcine pancreatic elastase (porcine pancreatic elastase, PPE) -induced abdominal aortic aneurysm.
The invention also provides application of the kaurene extract in at least one of the following:
1) The application of the elastic plate in preparing products for inhibiting the rupture of the elastic plate of the abdominal aorta;
2) The application of the composition in preparing a product for inhibiting inflammatory cell infiltration of abdominal aortic vessel wall;
in particular, the inflammatory cells may be macrophages and/or leukocytes;
3) The application of the composition in preparing a product for inhibiting the expression of inflammatory factors in the wall of abdominal aortic blood vessel;
in particular, the inflammatory factor may be interleukin 6 (interleukin-6, IL-6), monocyte chemoattractant factor-1 (monocyte chemotactic protein, MCP-1), tumor necrosis factor alpha (tumor necrosis factor. Alpha., TNF-alpha.), IL-1. Beta. And IL-18;
4) The application of the preparation of a product for inhibiting the expression of matrix metalloproteinase in the abdominal aortic vessel wall;
in particular, the matrix metalloproteinase may be matrix metalloproteinase 9 (matrix metalloproteinase, mmp 9);
5) The application in preparing the product for inhibiting the activation of the inflammatory channel of the abdominal aortic vessel wall;
in particular, the inflammatory pathway may be the nuclear factor κb (NF- κb) and NOD-like receptor thermal protein domain related protein 3 (NOD-like receptor thermal protein domain associated protein 3, nlrp 3) inflammatory small body pathway;
6) The application of the matrix metalloproteinase inhibitor in preparing products for inhibiting the expression of vascular smooth muscle cell matrix metalloproteinase;
in particular, the matrix metalloproteinase may be MMP9;
7) The application of the preparation of the product for inhibiting the expression of vascular smooth muscle cell inflammatory factors;
specifically, the inflammatory factors may be IL-6, MCP-1, IL-1β, and IL-18;
8) The application of the composition in preparing products for inhibiting the activation of vascular smooth muscle cell inflammatory pathways;
in particular, the inflammatory pathways may be NF-. Kappa.B and NLRP3 inflammatory small body pathways.
In the above application, the active ingredient of the kaurene extract includes Araucarone (AO), and its structural formula is as follows:
in the above application, the kauri extract is ethanol water solution extract of kauri.
In particular, the method comprises the steps of, the volume fraction of the ethanol in the ethanol water solution is 60% -100%; specifically, the content may be 95%.
The kaurena extract of the present invention can be prepared according to the methods disclosed in the prior art.
The preparation can be carried out as follows: drying and pulverizing rhizome of kaurena, soaking in ethanol water solution, heating to slightly boiling state, reflux extracting, collecting extractive solution, concentrating, and drying to obtain kaurena extract.
The soaking time can be 1h; the reflux extraction is carried out at least 1 time, preferably 3 times; the time of each reflux extraction is 1-2 h.
In the above application, the product is a medicament or a pharmaceutical preparation.
The medicament may be introduced into the body by injection, oral, spray, osmotic, absorption, physical or chemical mediated means such as muscle, intradermal, subcutaneous, intravenous, mucosal tissue; or mixed or wrapped with other substances and introduced into the body.
If necessary, one or more pharmaceutically acceptable carriers can be added into the above medicines to prepare into pharmaceutical preparations. The carrier includes diluents, excipients, fillers, binders, wetting agents, disintegrants, absorption promoters, surfactants, adsorption carriers, lubricants, etc. which are conventional in the pharmaceutical field.
The medicine can be prepared into various forms such as injection, suspending agent, powder, tablet, granule and the like. The medicaments of the various formulations can be prepared according to the conventional method in the pharmaceutical field.
The invention adopts a clinical related AAA model: porcine pancreatic elastase-induced mouse AAA. Experiments prove that the injection of kaurena extract into stomach or abdominal cavity can obviously inhibit the generation and development of mouse AAA induced by elastase.
The anti-AAA medicine provided by the invention is safe, low in toxicity and strong in pharmacological action; the raw materials are rich in sources and can be extracted from kaurene plants; the invention provides a new medicine source for preventing, diagnosing, detecting, protecting, treating and researching abdominal aortic aneurysm, is easy to popularize and apply, and can generate huge social benefit and economic benefit in a shorter time.
Drawings
FIG. 1 shows the inhibition of TNF- α -induced expression of inflammatory factors and activation of MMP9 in vascular smooth muscle cells by kaurene extract (AD).
FIG. 2 shows the inhibition of gastric lavage by AD 100mg/kg/day of Porcine Pancreatic Elastase (PPE) -induced abdominal aortic aneurysm in mice.
FIG. 3 inhibition of inflammatory factor expression and MMP9 activation in TNF- α -induced vascular smooth muscle cells by Naquoyitol (AO).
FIG. 4 shows the inhibition of PPE-induced abdominal aortic aneurysm in mice by AO 100mg/kg/day lavage.
FIG. 5 shows the inhibition of PPE-induced abdominal aortic aneurysm in mice by AD/AO 50mg/kg/day intraperitoneal injection.
FIG. 6 shows the up-regulation of inflammatory factors and MMP9 in early inflammatory phase of abdominal aortic blood vessels in mice induced by AD/AO 100mg/kg/day gastric lavage inhibition PPE.
FIG. 7 shows the AD/AO inhibition of TNF- α -induced smooth muscle cells and PPE-induced activation of NF- κB/NLRP3 inflammatory pathways in abdominal aortic vascular tissue.
Detailed Description
The following detailed description of the invention is provided in connection with the accompanying drawings that are presented to illustrate the invention and not to limit the scope thereof.
The experimental methods in the following examples are conventional methods unless otherwise specified.
Materials, reagents and the like used in the examples described below are commercially available unless otherwise specified.
The percentages indicated in the examples below are by mass, unless otherwise indicated.
Porcine pancreatic elastase used in the following examples was purchased from Sigma-Aldrich under the designation E1250.
In the following examples, the solvent for AD or AO was 0.5% (m/v) sodium carboxymethyl cellulose, and the solvent for sodium carboxymethyl cellulose was 0.9% physiological saline.
The quantitative tests in the following examples were all set up for 3-6 replicates and the results averaged.
EXAMPLE 1 preparation of kaurene extract (AD) and monomer thereof
1. Preparation of kaurene extract
Dried and crushed kauren root powder (367 g) is put into a 3.0L round bottom flask, is soaked for 1h by adding 2.5L of 95% (v/v) ethanol, is heated to a micro-boiling state, is heated and refluxed for extraction for 2h, and is filtered when the extract is hot. The residue is extracted with 2.0L of 95% (v/v) ethanol under reflux for 2 times each for 1.5h. The filtrates were combined, concentrated by rotary evaporator, dried in vacuo, and weighed to give 105.3g (extraction yield: 28.7%) of kaurena extract.
2. Preparation of monomers
Taking the prepared kaurena shell extract (100.0 g), adding 1.0L of water for suspension, extracting with dichloromethane, extracting 3 times with 1.0L of extractant each time, and mixing the extracts to obtain 34.0g of dichloromethane layer extract.
Dichloromethane layer extract (34 g) was prepared according to 1:2, mixing the sample with silica gel (Qingdao ocean chemical, 300-400 mesh column chromatography silica gel; product number: 2.003447F6), purifying by column chromatography with 3.3L column volume of silica gel, and sequentially performing gradient elution with dichloromethane and dichloromethane/methanol, wherein the elution gradient is pure dichloromethane and dichloromethane/methanol 50: 1. 20: 1. 10: 1. 5: 1. 2: 1. 1:1, pure methanol (v/v), 6L per gradient. The same components were then combined according to the thin layer chromatography results to give 22 fractions in total. Fraction 1 (4.4 g) was stirred into 7.0g of silica gel, loaded onto a silica gel column (column volume 261 mL), and eluted with petroleum ether, petroleum ether/ethyl acetate, respectively, in a gradient of petroleum ether (600 mL), petroleum ether/ethyl acetate (v/v) 30: 1. 20: 1. 10: 1. 5: 1. 4: 1. 3: 1. 2: 1. 1:1 (400 mL each for each gradient). The same fractions were combined by thin layer chromatography to give 9 sub-fractions (A-I). And recrystallizing the subfraction H at room temperature to obtain the monomer compound.
The purity of the monomer was checked by HPLC, the purity is more than 95 percent. The detection conditions were as follows:
mobile phase: acetonitrile 100%; flow rate: 1mL/min; chromatographic column: welch Ultimate XB-C18 (250 mm. Times.4.6 mm. I.d.,5 μm); column temperature: 40 ℃; detection wavelength: 254nm,210nm.
And identifying the structure of the compound by utilizing mass spectrum and nuclear magnetic resonance spectrum.
Compound 1, ESI-MS m/z 301[ M+H ] + ], 1H-NMR (CDCl 3,600 MHz) δH:5.50
(1H,m),4.40(2H,s),3.26(1H,br.s),2.70(1H,td,J=14.6,5.3Hz),2.29(1H,br.d,J=11.5Hz),2.27(1H,dt,J=14.7,3.8Hz),2.14(1H,m),2.12(1H,m),2.10(1H,m),1.94(1H,m),1.75(1H,br.dd,J=6.2,3.1Hz),1.73(1H,m),1.68(1H,m),1.63(1H,td,J=12.8,3.7Hz),1.56(1H,dd,J=12.2,4.1Hz),1.50(1H,td,J=13.9,4.0Hz),1.45(1H,qd,J=12.5,4.2Hz),1.14(3H,s),1.10(6H,s),1.07(3H,s);
13C-NMR(CDCl3,151MHz)δC:216.5,214.9,133.3,123.3,64.1,51.5,50.7,47.4,45.9,41.7,38.0,35.3,34.6,32.5,25.6,23.9,22.6,19.5,18.9,14.8。
This compound was designated as southern cedar ketone (AO) and has the following structural formula:
example 2 pharmacodynamic test of Shell fir extract and monomer Nandina Kerata
1. Experimental materials
1.1 laboratory animals
The mice used in the experiments were male C57BL/6J mice with an average body weight of 20-25 g, purchased from Experimental animal center of the university of Beijing, followed the laboratory animal care principle (NIH publication No. 85-23, revision 1996), and the experimental protocol was approved by the animal ethics Committee of the university of Beijing (approval No. LA 2021045): all mice were kept under a 12 hour light/12 hour dark cycle, free to acquire food and water.
1.2 animal feed
Animal breeding feed is a commercially available normal feed for rodents.
2. In vivo experimental method
2.1 pig pancreatic elastase-induced mouse renal lower section abdominal aortic aneurysm model
Male C57BL/6J mice at 8 weeks of age were anesthetized with 4% chloral hydrate (m/v, 8. Mu.L/g by weight injection). The renal arteries of the mice were isolated to the abdominal aortic segment between the iliac arteries. The blood vessel was wrapped with 40. Mu.L of moistened gauze added dropwise to a stock of Porcine Pancreatic Elastase (PPE), after 1h incubation, the gauze was removed and excess porcine pancreatic elastase was rinsed off with PBS. After wrapping the blood vessel with 0.9% saline-wet gauze for 1h, the gauze was removed, rinsed with PBS, and used as a NaCl incubation control.
2.2 pharmacodynamic action of kaurene extract against Abdominal aortic aneurysm (intragastric administration)
Male C57BL/6J mice were randomly divided into control group (6), control drug treatment group (100 mg/kg/day AD, 6), elastase molding group (6), elastase molding while kaurena extract (100 mg/kg/day AD, 6) stomach-lavage administration group. The control group and the elastase molding group were administered with the same volume of 0.5% sodium carboxymethyl cellulose per day and the control drug-treated group and the elastase molding group were administered with 100mg/kg AD per day (AD was administered as a suspension at a concentration of 12.5mg/mL, and the solvent was 0.5% sodium carboxymethyl cellulose). Surgery was performed after the administration on day 3. The medicine is administrated by lavage once a day after the operation, and the animal is sacrificed after 14 days of molding.
2.3 pharmacodynamic effects of Nantarione (AO) against Abdominal aortic aneurysm (intragastric administration)
Male C57BL/6J mice were randomly divided into control groups (6), elastase-induced modules (6), elastase-induced simultaneous mode of administration of Nanyuan (100 mg/kg/day AO, 6) by intragastric administration. The control group and the elastase model group were infused with the same volume of 0.5% sodium carboxymethyl cellulose daily for 2 days, the elastase model was infused with 100mg/kg AO (AO was formulated as a suspension with a concentration of 12.5 mg/mL) daily in the group administered with the same volume of the stomach infused with the southern datone, and the solvent was 0.5% sodium carboxymethyl cellulose. Surgery was performed after the administration on day 3. The medicine is administrated by lavage once a day after the operation, and the animal is sacrificed after 14 days of molding.
2.4 pharmaceutical effects of kaurene (AD) and Nanchuone (AO) on abdominal aortic aneurysm (intraperitoneal injection)
Male C57BL/6J mice were randomly split into: control group (6), elastase molding while southern China fir ketone (50 mg/kg/day AO, 6) intraperitoneal injection administration group and elastase molding while kaurena extract (50 mg/kg/day AD, 6) intraperitoneal injection administration group. The control group and the elastase molding module were injected with the same volume of 0.5% sodium carboxymethyl cellulose daily for 2 days of pre-administration, the elastase molding module was simultaneously injected with the southern datone into the abdominal cavity, and the elastase molding module was simultaneously injected with the kaurene extract into the abdominal cavity, 50mg/kg of AO or kaurene extract AD (AO or AD was formulated into a suspension with a concentration of 6.25mg/mL, and the solvent was 0.5% sodium carboxymethyl cellulose) was injected into the abdominal cavity each day. Surgery was performed after the administration on day 3. Once daily administration is performed after the operation, and after 14 days of molding, the animals are sacrificed for material taking.
2.5 Shell fir extract (AD) and Nandina Kerata (AO) for inhibiting vascular inflammation
Male C57BL/6J mice were randomly divided into a control group, an elastase-induced module, an elastase-induced simultaneous douche administration group of kaurenone (100 mg/kg/day AO) and an elastase-induced simultaneous douche administration group of kaurene extract (100 mg/kg/day AD). The control group and the elastase molding module were subjected to gastric lavage daily for 2 days with 0.5% sodium carboxymethylcellulose, the elastase molding and southern datone gastric lavage administration group was subjected to gastric lavage daily for 100mg/kg AO, the elastase molding and kauri extract gastric lavage administration group was subjected to gastric lavage daily for 100mg/kg AD (AO or AD was prepared into suspension with concentration of 12.5mg/mL, and the solvent was 0.5% sodium carboxymethylcellulose). Surgery was performed after the administration on day 3. The medicine is administrated by lavage once a day after the operation, and the animal is sacrificed for sampling after 3 days of molding.
2.6 animal handling
2.6.1 collection of mouse serum
After the mice are anesthetized, blood is taken for about 600 mu L, incubated for 30min at 37 ℃, centrifuged for 30min at 4000rpm at room temperature by a centrifuge, and the upper serum is taken and frozen in a refrigerator at-30 ℃ for later use.
2.6.2 mouse draws
The body weight of the mice was weighed and the body weight value was recorded. After the sacrifice of excess anesthesia, the pin is fixed in an anatomic disc (plastic foam box). The skin and subcutaneous film of the mice are cut to expose the viscera and heart to take blood. After perfusion with cardiac phosphate buffered saline (phosphate buffered solution, PBS), the mice were isolated from the heart to the complete aorta at the iliac artery with scissors and forceps, the morphologically observed vascular tissue was left to fix overnight with 4% paraformaldehyde (m/v, PBS as solvent), and then transferred to 20% sucrose. After finely separating peripheral connective tissues from blood vessels, preparing frozen slices after dehydration, fixation and embedding, and using the frozen slices for HE and EVG staining to judge vascular injury and fracture degree of an elastic plate. The aorta with RNA or protein is not fixed, PBS is used for perfusion, the abdominal aortic subrenal segment is directly separated under an dissecting microscope, and the abdominal aortic subrenal segment is frozen at-80 ℃ for later use after being taken down.
3. In vitro experimental method
3.1 culture of primary rat vascular smooth muscle cells and establishment of in vitro cell inflammation models.
3.1.1 extraction of vascular smooth muscle cells from Primary rats
About 3 male Sprague-Dawley rats were prepared, and after the rats were anesthetized and sacrificed, the thoracic cavity was rapidly opened, and the aortic arch of the rats was isolated to the thoracic aortic vessel at the diaphragmatic site. Residual blood was washed off with pre-chilled PBS and extravascular fat and connective tissue was gently scraped off with forceps. The vessel was cut, the intima was gently scraped, and the intima layer was torn off. After shearing the vessel membrane, the vessel membrane was placed in 3mL of Dalberg modified eagle's medium (dulbecco's modified eagle medium, DMEM) containing 1mg/mL of type II collagenase (available from Gibco under the trade name 17101015) at 37℃in 5% CO 2 Digestion in incubator of (v/v) for 3h.1500g was centrifuged for 15 min, DMEM (v/v) containing 10% fetal bovine serum, also known as complete medium, and after resuspension, the cells were inoculated into petri dishes, and after 3 days of stationary culture, the solution was changed. Cells of passages 4 to 6 were used for the experiment.
3.1.2 establishment of in vitro vascular smooth muscle cell inflammation model
(1) Primary rat vascular smooth muscle cells 40 ten thousand cells per well (vascular smooth muscle cell, VSMC) were seeded in 6-well plates and cultured until cell attachment was achieved by addition of 1mL of DMEM complete medium.
(2) Then, after the DMEM complete medium is replaced by 1mL of serum-free DMEM medium to starve for 24 hours, the DMEM complete medium is replaced by 1mL of DMEM complete medium containing 100ng/mL of TNF-alpha (the TNF-alpha is diluted by sterile water, and the concentration of mother liquor is 10 mu g/mL) to stimulate for 24 hours (used for extracting RNA) or 48 hours (used for extracting protein); simultaneously administering dimethyl sulfoxide (Dimethyl Sulphoxide, DMSO) with the same volume in the culture medium of the control group and the model group, simultaneously administering AD or AO in the experimental group, and adding 1 mu L of mother solution into 1mL of DMEM complete culture medium for fully and uniformly mixing when the culture medium is used, wherein the concentration of the mother solution prepared by the AD or AO by DMSO is 25mg/mL and 25mmol/L respectively; the culture conditions are 37 ℃ and relative humidity 90%, CO 2 Incubating in incubator at 5% (v/v).
(3) The cells were washed 3 times with PBS and 1mL of RNA extraction reagent (Trizol) was added to each well for RNA extraction or 50. Mu.L of protein lysate was added for protein extraction and Western blot.
3.2MTT assay
(1) Vascular smooth muscle cells in the logarithmic growth phase were collected.
(2) Adjusting the concentration of the cell suspension: in an ultra clean bench, the cell counting plate and the cover glass are scrubbed by alcohol, 20 mu L of cell suspension is taken, and a gun head is used for propping against the middle part of one side of the cover glass and is driven into the counting plate. Cells within 4 squares of four square corners were counted and averaged under a low power field of view. The cell concentration in the suspension is: average value x 10 4 (individual/mL). The number of cells seeded per well of a 96-well plate was 1×10 4 Each (100 μl per well) was used to adjust the concentration of the cell suspension with DMEM complete medium.
(3) Grouping according to experimental requirements: zeroing (no cells), control, dosing (6 parallel wells).
(4) Incubating the 96-well plate in a incubator (relative humidity 90%, CO 2 Concentration 5%, v/v) until cells were fully adherent, after starvation with blank DMEM medium (without FBS) for 24h, changing to full DMEM with different concentration of drug (with 10% FBS, v/v) and incubation was continued for 48h. AD or AO were treated with DMSO to prepare solutions of different concentrations.
(5) The culture was terminated, the culture broth was aspirated, 5.0g/L of a solution of tetramethylazo-salt (3-2, 5-diphenyl-2-H-tetrazolium bromide, MTT) was added to each well, and the mixture was further cultured in a cell incubator at 37℃for 4 hours.
(6) After 4h the supernatant was aspirated (note that the purple crystals at the bottom of the well were not aspirated). 150 mu L of DMSO is added into each hole, the mixture is put into an enzyme labeling instrument and vibrated for 10min to completely dissolve blue crystalline formazan, then the absorbance is measured at the wavelength of 540nm, and the cell survival rate is calculated according to the following formula:
cell viability = (dosing group-zeroing group)/(control group-zeroing group)
3.3 pharmacodynamic effects of kaurene extract in inhibiting VSMC cell inflammation
VSMC was seeded in six well plates at a density of 2X 10 5 Each of the cells was cultured with 1mL of DMEM containing 10% (v/v) FBS per wellTo the wall, cells were starved for 24h with 1mL serum-free DMEM per well. VSMC were incubated with 1mL of complete DMEM (10% FBS, v/v) containing 100ng/mL TNF- α per well (TNF- α diluted with sterile water at a mother liquor concentration of 10. Mu.g/mL), and the drug administration group was incubated in an incubator (37 ℃ C., relative humidity 90%, CO) with 25. Mu.g/mL kauri extract (AD) or 25. Mu. Mol/L kauri ketone (AO) administered simultaneously with TNF- α stimulation 2 Concentration 5% (v/v)). Cells were harvested after 24h (for RNA extraction) or 48h (for protein extraction) for detection of inflammatory factor expression, MMP9 expression and NF- κ B, NLRP3 pathway activation. AD or AO was prepared as a mother solution with a concentration of 25mg/mL and 25mmol/L, respectively, with DMSO and then added to the cell culture solution.
4. Experimental results:
1. kaurene extract (AD) inhibits TNF-alpha-induced vascular smooth muscle cell inflammatory factor expression and MMP9 activation
A in fig. 1 is a cell viability test of vascular smooth muscle cells treated with AD at different concentrations for 48 h; as a result, it was found that AD was excellent in cell safety in the concentration range of 100. Mu.g/mL.
B-E in FIG. 1 was 100ng/mL TNF- α for 24h, respectively, with 25 μg/mL AD treatment, and relative gene expression levels of IL-6, MCP-1, IL-1. Beta. And IL-18 in the cells. From this, it is known that AD can significantly inhibit TNF- α -induced inflammatory response of vascular smooth muscle cells, thereby reducing inflammatory injury of smooth muscle cells.
F in FIG. 1 is the relative gene expression level of MMP9 in cells treated with 100ng/mL TNF- α for 24h, while treated with 25 μg/mL AD. It follows that AD can significantly inhibit TNF-alpha-induced upregulation of MMP9 mRNA expression levels in vascular smooth muscle cells, thereby inhibiting extracellular matrix component degradation.
G and H in FIG. 1 are representative bands and quantification of protein expression of MMP9 in cells treated with 100ng/mL TNF- α for 48H, respectively, while treated with 25 μg/mL AD. From this, it is shown that AD can significantly inhibit TNF-alpha-induced upregulation of protein expression of MMP9 in vascular smooth muscle cells, thereby inhibiting degradation of extracellular matrix components.
I and J in FIG. 1 are the relative gene expression levels of TIMP1 and TIMP2 in cells treated with 100ng/mL TNF-. Alpha.for 24h, respectively, and 25. Mu.g/mL AD. It was shown that AD significantly up-regulates the expression of TIMP1/2 in TNF-alpha induced vascular smooth muscle cells, thereby inhibiting extracellular matrix degradation.
K and L in FIG. 1 are representative bands and quantification of MMP9 activity in cell culture supernatant, respectively, for 48h at 100ng/mL TNF- α, while treated with 25 μg/mL AD. From this, it is clear that AD can significantly inhibit TNF-alpha-induced upregulation of MMP9 enzymatic activity in vascular smooth muscle cells, thereby inhibiting extracellular matrix component degradation.
2. Gastric lavage administration of kaurene extract (AD) inhibits PPE-induced abdominal aortic aneurysm in mice
A in fig. 2 is a flow chart of animal experiments. After 2 days of 100mg/kg/day AD pre-gavage, AAA molding was performed after day 3 gavage. The gastric lavage administration is continued every day after the operation, and the materials are obtained after 14 days.
B and C in fig. 2 are a rough photograph of the mouse abdominal aorta and statistics of maximum diameter of the renal lower section of the abdominal aorta, respectively, and the results show that AD gastric lavage administration can significantly inhibit PPE-induced dilation of the mouse abdominal aorta.
D in fig. 2 is a HE staining pattern of a mouse abdominal aortic section, and the results show that AD can significantly inhibit PPE-induced vascular remodeling, including pathological changes such as vessel wall thickening, adventitia massive cell infiltration, and media adventitia boundary blurring.
E in FIG. 2 is an EVG staining pattern, which shows that AD can significantly inhibit PPE-induced thinning, rupture and degradation of the abdominal aortic vascular spandex.
F in fig. 2 is a CD68 immunohistochemical staining pattern, and the results indicate that AD can significantly inhibit PPE-induced macrophage infiltration in the abdominal aortic blood vessels.
3. Nadaturone (AO) inhibits TNF-alpha-induced vascular smooth muscle cell inflammatory factor expression and MMP9 activation
A in FIG. 3 is a cell viability test of vascular smooth muscle cells treated with AO at various concentrations for 48h. From this, it was found that AO was excellent in cell safety in the concentration range of 100. Mu.M.
B-E in FIG. 3 is the relative gene expression levels of MCP-1, IL-1. Beta., IL-18 and MMP9 in cells treated with 100ng/mL TNF- α for 24h, respectively, and 25. Mu.M AO. From this, it is suggested that AO can significantly inhibit TNF- α -induced upregulation of mRNA expression levels of MMP9 in vascular smooth muscle cells, thereby inhibiting degradation of extracellular matrix components. Meanwhile, AO can obviously inhibit up-regulation of mRNA expression level of inflammatory factors in vascular smooth muscle cells induced by TNF-alpha, thereby inhibiting inflammatory injury of the smooth muscle cells.
F and G in FIG. 3 are representative bands and quantification of MMP9 activity in cell culture supernatant, respectively, for 48h at 100ng/mL TNF- α treatment, while at the same time treating with 25. Mu.M AO. From this, it is suggested that AO can significantly inhibit TNF- α -induced upregulation of MMP9 enzyme activity in vascular smooth muscle cells, thereby inhibiting extracellular matrix component degradation.
4. Gastric lavage administration of Nanymanone (AO) inhibits PPE-induced abdominal aortic aneurysm in mice
A in fig. 4 is a flow chart of animal experiments. 100mg/kg/day AO pre-lavage was performed for 2 days, and AAA molding was performed after the 3 rd day lavage. The gastric lavage administration is continued every day after the operation, and the materials are obtained after 14 days.
B and C in fig. 4 are a rough photograph of the mouse abdominal aorta and statistics of maximum diameter of the renal lower section of the abdominal aorta, respectively, and the results show that administration of AO by gastric lavage can significantly inhibit PPE-induced expansion of the mouse abdominal aorta.
D in FIG. 4 is a HE staining pattern of a mouse abdominal aortic section, showing that AO can significantly inhibit PPE-induced vascular remodeling, including pathological changes such as vessel wall thickening, adventitia massive cell infiltration, and media adventitia boundary blurring.
E in FIG. 4 is an EVG staining pattern, which shows that AO can significantly inhibit PPE-induced thinning, rupture and degradation of the abdominal aortic vascular spandex.
F in FIG. 4 is a CD68 immunohistochemical staining pattern, and the results show that AO can significantly inhibit PPE-induced macrophage infiltration in the abdominal aortic blood vessels.
ad/AO intraperitoneal injection can significantly inhibit PPE-induced abdominal aortic aneurysm in mice
A in fig. 5 is an experimental flowchart. AD or AO 50mg/kg/day pre-administration was performed 2 days later, AAA molding surgery was performed after administration on day 3, and mice were sacrificed after 14 days after still daily administration by intraperitoneal injection.
B in fig. 5 is a general photograph of AD and AO inhibiting PPE-induced abdominal aortic aneurysm in mice, and C in fig. 5 is a maximum diameter statistic of the lower abdominal aortic renal segment, which indicates that both AD and AO can significantly reduce PPE-induced abdominal aortic dilation.
D in FIG. 5 is a HE staining pattern of a mouse abdominal aortic section, and the results show that both AD and AO can significantly inhibit PPE-induced vascular remodeling, including pathological changes such as vessel wall thickening, adventitia massive cell infiltration, and media adventitia boundary blurring.
E in FIG. 5 is an EVG staining pattern, which shows that AD and AO can significantly inhibit PPE-induced thinning, rupture and degradation of the abdominal aortic vascular spandex.
F in fig. 5 is a CD68 immunohistochemical staining pattern, and the results indicate that AD and AO significantly inhibited PPE-induced macrophage infiltration in the abdominal aortic blood vessels.
AD/AO gastric lavage administration can significantly inhibit PPE-induced inflammatory factor and MMP9 expression in early inflammatory stage of abdominal aorta of mice
A in fig. 6 is a flow chart of a mouse experiment. AAA molding was performed 3 days after AD or AO gavage at 100mg/kg/day pre-gavage. The gastric lavage administration is continued every day after the operation, and the materials are obtained after 3 days. AD or AO significantly inhibited gene expression of MCP-1, IL-6, TNF- α, IL-1β, IL-18 and MMP9 in the abdominal aortic blood vessels of mice (see B-G in FIG. 6). AD or AO significantly inhibited MMP9 protein expression in the abdominal aortic blood vessels of mice (H in fig. 6) WB representative bands and their (I in fig. 6) quantification results. It can be seen that AD and AO can significantly inhibit PPE-induced inflammatory injury and extracellular matrix degradation in early inflammatory phases of the mouse abdominal aortic blood vessels.
AD/AO significantly inhibited TNF- α -induced vascular smooth muscle cell and PPE-induced activation of the NF-. Kappa.B/NLRP 3 pathway in the aortic blood vessels of mice.
A and B in FIG. 7 are 100ng/mL TNF- α for 24h, while vascular smooth muscle cells are treated with 25 μg/mL AD or 25 μM AO, which inhibits TNF- α induced gene expression of NLRP3 in vascular smooth muscle cells. 100ng/mL TNF- α was treated for 48h, while treated with 25 μg/mL AD or 25 μM AO, which inhibited the WB-representative band of TNF- α -induced protein expression of NLRP3 (C in FIG. 7) in vascular smooth muscle cells and its (D in FIG. 7) quantification results. It can be seen that both AD and AO significantly inhibit TNF- α -induced activation of NLRP3 in vascular smooth muscle cells.
E and F in FIG. 7 are representative bands of WB and quantitative results of phosphorylated protein of NF-. Kappa. B p65 in vascular smooth muscle cells of 25. Mu.g/mL AD or 25. Mu.M AO pretreated vascular smooth muscle cells for 12h, followed by 100ng/mL TNF-. Alpha.treated cells for 30min, and AD or AO inhibited the phosphorylated protein of NF-. Kappa. B p in vascular smooth muscle cells induced by TNF-. Alpha.and were quantified. It can be seen that both AD and AO significantly inhibit TNF- α -induced activation of NF- κB in vascular smooth muscle cells.
AAA molding was performed 3 days after AD/AO 100mg/kg/day pre-lavage. The gastric lavage administration is continued every day after the operation, and the materials are obtained after 3 days. AD or AO can significantly inhibit gene expression of NLRP3 in the abdominal aortic blood vessels (G in fig. 7) of mice. AD or AO significantly inhibited protein expression of NLRP3 and NF-. Kappa. B p65 phosphorylation expression in the abdominal aortic vessels of mice (H-J in FIG. 7). It can be seen that both AD and AO significantly inhibited PPE-induced activation of NF-. Kappa.B and NLRP3 in the lower abdominal aortic renal vessels of mice in mice.

Claims (8)

1. Application of kaurena extract in preparing product for preventing and/or treating abdominal aortic aneurysm is provided.
2. The use according to claim 1, characterized in that: the abdominal aortic aneurysm is a renal subrenal abdominal aortic aneurysm.
3. Use of kauri extract in at least one of:
1) The application of the elastic plate in preparing products for inhibiting the rupture of the elastic plate of the abdominal aorta;
2) The application of the composition in preparing a product for inhibiting inflammatory cell infiltration of abdominal aortic vessel wall;
3) The application of the composition in preparing a product for inhibiting the expression of inflammatory factors in the wall of abdominal aortic blood vessel;
4) The application of the preparation of a product for inhibiting the expression of matrix metalloproteinase in the abdominal aortic vessel wall;
5) The application in preparing the product for inhibiting the activation of the inflammatory channel of the abdominal aortic vessel wall;
6) The application of the matrix metalloproteinase inhibitor in preparing products for inhibiting the expression of vascular smooth muscle cell matrix metalloproteinase;
7) The application of the preparation of the product for inhibiting the expression of vascular smooth muscle cell inflammatory factors;
8) Use in the preparation of a product for inhibiting activation of vascular smooth muscle cell inflammatory pathways.
4. A use according to claim 3, characterized in that: 2) Wherein the inflammatory cells are macrophages and/or leukocytes;
3) Wherein the inflammatory factor is interleukin 6 (IL-6), monocyte chemoattractant factor-1 (MCP-1), tumor necrosis factor alpha (TNF-alpha), IL-1 beta and IL-18;
4) Wherein the matrix metalloproteinase is matrix metalloproteinase 9 (MMP 9);
5) Wherein the inflammatory pathway is a nuclear factor κb (NF- κb) and NOD-like receptor thermal protein domain related protein 3 (NLRP 3) inflammatory small pathway;
6) Wherein the matrix metalloproteinase is MMP9;
7) Wherein the inflammatory factors are IL-6, MCP-1, IL-1β and IL-18;
8) The inflammatory pathways are NF- κB and NLRP3 inflammatory small body pathways.
5. The use according to any one of claims 1-4, characterized in that: the active ingredient of the kaurene extract comprises Araucarone (AO), and the structural formula is shown as follows:
6. the use according to any one of claims 1-4, characterized in that: the kauri extract is ethanol water solution extract of kauri.
7. The use according to claim 6, characterized in that: the volume fraction of the ethanol in the ethanol water solution is 60% -100%.
8. The use according to any one of claims 1-7, characterized in that: the product is a drug or a pharmaceutical preparation.
CN202310584879.8A 2023-05-23 2023-05-23 Application of kaurene extract in preparation of anti-abdominal aortic aneurysm products Pending CN116440179A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310584879.8A CN116440179A (en) 2023-05-23 2023-05-23 Application of kaurene extract in preparation of anti-abdominal aortic aneurysm products

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310584879.8A CN116440179A (en) 2023-05-23 2023-05-23 Application of kaurene extract in preparation of anti-abdominal aortic aneurysm products

Publications (1)

Publication Number Publication Date
CN116440179A true CN116440179A (en) 2023-07-18

Family

ID=87127494

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310584879.8A Pending CN116440179A (en) 2023-05-23 2023-05-23 Application of kaurene extract in preparation of anti-abdominal aortic aneurysm products

Country Status (1)

Country Link
CN (1) CN116440179A (en)

Similar Documents

Publication Publication Date Title
Yang et al. Astragaloside IV attenuates inflammatory cytokines by inhibiting TLR4/NF-кB signaling pathway in isoproterenol-induced myocardial hypertrophy
JP5331962B2 (en) Herbal composition for inflammatory diseases
CN110172064B (en) Flavone derivatives, preparation method and medical application thereof
CN114716346B (en) 4-nitroaniline derivative and application thereof
KR20160123130A (en) Composition comprising Chrisanthemum indicum extract or fraction for treating, improving or preventing obesity or obesity-related disease
CN116440179A (en) Application of kaurene extract in preparation of anti-abdominal aortic aneurysm products
JP7326561B1 (en) Use of an extract of the active site of Gardenia japonicum in the preparation of a medicament for treating inflammatory diseases or tumors
CN103735550B (en) A kind of pharmaceutical composition and application thereof for the treatment of chronic kidney disease myocardial damage
CN110693880A (en) Urolithin preparation and application thereof
CN105147677B (en) One group of 2- replaces formamido -3- alkoxy pyridines analog derivative and its pharmaceutical applications
PT1931366E (en) Herbal composition for inflammatory disorders
US8692023B2 (en) Sugar metabolism improving composition, and pharmaceutical preparation containing said composition
CN112716986A (en) Application of inonotus obliquus in preparation of anti-ventricular remodeling medicine
CN110934864A (en) Application of 4-methyl-6, 7-dimethoxycoumarin in preparing medicine for preventing and treating diabetic nephropathy
Fan et al. Celastrol relieves myocardial infarction-induced cardiac fibrosis by inhibiting NLRP3 inflammasomes in rats
CN106580951B (en) A kind of new application of flavanone compound
CN114159449B (en) Application of 3 beta, 23-O-isopropylidene hydroxyl betulinic acid in preparation of anti-hepatic fibrosis drugs
CN110051671B (en) Application of purslane amide E in preparation of medicine for treating ischemic heart disease
CN103356637B (en) Chaetocin is preparing the application in prevention and therapy cardiotropic formulation
KR102283093B1 (en) Composition for prevention and treatment of metabolic diseases including ginger extract
CN112933120B (en) Application of kauri pine extract in preparing product for resisting non-alcoholic fatty liver disease
WO2023125719A1 (en) Veronica undulata extract as well as preparation method therefor and use thereof
KR20230155649A (en) Pharmaceutical composition for preventing or treating metabolic diseases comprising Licochalcone D or analogs thereof
CN104940220B (en) The pharmaceutical composition and preparation method thereof for preventing and treating diabetic complication
CN115844933A (en) Application of clinacanthus nutans total flavonoids in preparation of anti-heart failure drugs

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