CN115504950A - Lignans compound and preparation method and application thereof - Google Patents

Lignans compound and preparation method and application thereof Download PDF

Info

Publication number
CN115504950A
CN115504950A CN202110690684.2A CN202110690684A CN115504950A CN 115504950 A CN115504950 A CN 115504950A CN 202110690684 A CN202110690684 A CN 202110690684A CN 115504950 A CN115504950 A CN 115504950A
Authority
CN
China
Prior art keywords
ethanol
water
compound
gradient elution
eluate
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.)
Granted
Application number
CN202110690684.2A
Other languages
Chinese (zh)
Other versions
CN115504950B (en
Inventor
王团结
李海波
李贵平
顾莎莎
李旭
张全昌
王振中
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.)
Jiangsu Kanion Pharmaceutical Co Ltd
Original Assignee
Jiangsu Kanion Pharmaceutical 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 Jiangsu Kanion Pharmaceutical Co Ltd filed Critical Jiangsu Kanion Pharmaceutical Co Ltd
Priority to CN202410066479.2A priority Critical patent/CN117986309A/en
Priority to CN202110690684.2A priority patent/CN115504950B/en
Publication of CN115504950A publication Critical patent/CN115504950A/en
Application granted granted Critical
Publication of CN115504950B publication Critical patent/CN115504950B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/77Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D307/78Benzo [b] furans; Hydrogenated benzo [b] furans
    • C07D307/86Benzo [b] furans; Hydrogenated benzo [b] furans with an oxygen atom directly attached in position 7
    • 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

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pain & Pain Management (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Rheumatology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines Containing Plant Substances (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)

Abstract

The invention discloses a lignan compound which is a new chemical component found in Illicium verum leaves of safflower. The invention also carries out structural identification on the compound separated by the method through physicochemical properties and modern spectral means. The invention also utilizes an LPS to induce an activity screening system such as a RAW 264.7 cell inflammation model and the like to carry out activity evaluation, and discovers that the compound has a certain protection effect on a mouse macrophage system RAW 264.7 and can obviously inhibit PGE 2 The release of (2) shows a strong anti-inflammatory effect.

Description

Lignans compound and preparation method and application thereof
Technical Field
The invention relates to the technical field of medicines, and particularly relates to a novel compound and a preparation method and application thereof.
Background
The Illicium tinctorium (Illicium dunnianum Tutcher) is plant of Illicium genus (Illicium) of Magnoliaceae family (Magnoliaceae). The illicium plants are evergreen trees or shrubs with aromatic odor, 34 varieties exist all over the world, 28 varieties exist in China, most varieties are distributed in east and south east of Asia, and few varieties are distributed in southeast and central south America of North America. The star anise flower is peculiar to China, is distributed in places such as Guangxi, fujian, guizhou, hunan and Guangdong, and is usually grown along the coasts of rivers, beside valley waters, in mountain forests, in wet hills or in rock seams, and has the elevation of 400-1000 meters. The fruit shape is similar to that of the red fennel, the fruit is thin and small, and generally consists of 7-8 fruits, and a small number of 13 fruits have obvious diamond-shaped pointed ends and are slightly bent; the fruit stalks are fine but the fruit stalks are shorter, the seeds are smaller, and the fruit stalks are easy to distinguish. The medicine is bitter and pungent in taste and warm in nature, and has the effects of removing blood stasis, relieving swelling, dispelling wind, removing dampness and relieving pain, so roots and barks of the medicine are commonly used as medicines in folk, and the medicine is externally used for treating rheumatism bone pain, traumatic injury, contusion and fracture; toxic, the shikimic toxin and the new shikimic toxin separated from the root are convulsion components. Pharmacological experimental research shows that the alcohol extract of the aniseed leaves of the safflower has the central and peripheral analgesic effects and has better analgesic and detumescence effects on various pains and acute soft tissue injuries.
The safflower anise is a peculiar plant in China, is commonly used for treating rheumatic osteodynia, traumatic injury, contusion and fracture as folk medicine, and is not recorded by Chinese pharmacopoeia, but the safflower anise leaves are one of the prescription medicinal tastes of Chinese patent medicine Jinhong tablets, the Jinhong tablets have the effects of soothing liver and relieving depression, regulating vital energy and invigorating blood circulation, and harmonizing stomach and relieving pain, are mainly used for treating chronic superficial gastritis liver-stomach disharmony clinically, have definite curative effect and obvious effect, but the research on the chemical components of the safflower anise She Hejin red tablets is very few at present, the chemical components of the safflower anise tablets cannot be comprehensively clarified, the deep research on the medicinal effect substances and the action mechanism of the Jinhong tablets is limited, the improvement on the quality control standard cannot be realized, and the deep research on the active ingredients in the safflower anise leaves is further carried out.
Disclosure of Invention
The invention aims to carry out more intensive research on the anti-inflammatory active ingredients in the Illicium verum leaves of safflower, and finds the active ingredients.
In view of the above, the present invention provides a lignan compound, or a pharmaceutically acceptable salt, solvate, tautomer, stereoisomer, prodrug molecule, or metabolite thereof, wherein the compound has the following structure, and the structure of the compound is shown in formula I:
Figure BDA0003126052320000021
another object of the present invention is to provide a method for producing the above compound, which comprises:
a) Taking the aniseed leaves of the safflower, carrying out reflux extraction by 40-60% of ethanol, and removing the solvent to obtain a total extract;
b) Dissolving the total extract in water, separating by macroporous adsorption resin column chromatography, eluting with water, 25-35% ethanol, 45-55% ethanol and 90-100% ethanol in sequence, collecting eluates respectively, and concentrating under reduced pressure to obtain water eluate, 25-35% ethanol eluate, 45-55% ethanol eluate and 90-100% ethanol eluate; 4 column volumes per gradient elution (same below);
c) Taking the 45-55% ethanol elution part, separating by silica gel column chromatography, performing gradient elution and collection by using dichloromethane-methanol to obtain 15 fractions of 3A-3O, performing gradient elution on fraction 3L by using ODS column chromatography methanol-water to obtain 5 fractions of 3L1-3L5, performing gradient elution on fraction 3L4 by using polyamide column chromatography ethanol-water to obtain 6 fractions of 3L4A-3L4F, performing semi-preparative liquid chromatography on 3L4A to obtain 8 fractions of 3L4A2-3L4A9, performing semi-preparative liquid chromatography on 3L4A4 to obtain a compound 1,3L4A6, and performing semi-preparative liquid chromatography on the compound 2.
Specifically, the Illicium palmatum leaves can be dried leaves of Illicium palmatum.
Further, the step a) includes: taking dry illicium griseum leaves, carrying out reflux extraction for 1-3 times by 3-5 times of 40-60% ethanol, each time for 1-3 hours, combining extracting solutions, and removing the solvent under reduced pressure to obtain the total extract.
Preferably, the step B) includes: sequentially eluting with water, 30% ethanol, 50% ethanol and 95% ethanol, respectively collecting eluates, and concentrating under reduced pressure until no ethanol smell exists to obtain water eluate, 30% ethanol eluate, 50% ethanol eluate and 95% ethanol eluate.
The dichloromethane-methanol gradient elution of the step C) is that the gradient elution is carried out in a volume ratio of 100-90 to 0-10 to 0; the methanol-water gradient elution is performed in a volume ratio of 15-30; the ethanol-water gradient elution is performed by gradient elution according to the volume ratio of 5-15.
Preferably, said dichloromethane-methanol gradient elution of step C) is a gradient elution performed at a volume ratio of 100; the methanol-water gradient elution is performed by gradient elution at a volume ratio of 30; the ethanol-water gradient elution is performed by gradient elution at a volume ratio of 10.
Specifically, said dichloromethane-methanol gradient of step C) is eluted at a rate of 100; 95; 90; 85; 80; 70, 30to 60; the methanol-water gradient eluted as a 30; 40, mixing the raw materials in parts by weight; 50; 70; the ethanol-water gradient eluted as a gradient of 10; 25, 75;40, mixing the raw materials in parts by weight; 50;70, preparing a mixture of; 95/5 volume ratio for gradient elution.
Specifically, the macroporous adsorption resin comprises one or more of D101 type macroporous adsorption resin, HP-20 type macroporous adsorption resin, HPD-100A type macroporous adsorption resin or HPD-300 type macroporous adsorption resin.
Further, the semi-preparative liquid chromatography conditions include:
compound 1: specification C 18 5 μm, 10X 250mm Phenomenex Gemini column; the mobile phase comprises the following volume proportions: 17-27, 0.05-0.5, the detection wavelength is 240-260nm, and the flow rate is 2-4mL/min.
Compound 2: specification C 18 5 μm, 10X 250mm Phenomenex Gemini column(ii) a The mobile phase comprises the following volume proportions: 11-21, 89-79, the detection wavelength is 240-260nm, and the flow rate is 2-4mL/min.
Preferably, said step a) is a 2-time reflux extraction with 50% ethanol for 2 hours each;
the semi-preparative liquid chromatography conditions include: the mobile phases of the compounds 1 to 2 are acetonitrile-water-formic acid with a volume ratio of 22.1 and acetonitrile-water-formic acid with a volume ratio of 16.1.
The invention also aims to provide application of the compound or pharmaceutically acceptable salts, solvates, tautomers, stereoisomers, prodrug molecules and metabolites thereof in preparing anti-inflammatory drugs.
The invention also provides a medicament for treating inflammation, which comprises the compound shown in the formula (I) or pharmaceutically acceptable salt, solvate, tautomer, stereoisomer, prodrug molecule and metabolite thereof.
Further, the medicament contains a therapeutically effective amount of a compound of formula (I) as described above, or a pharmaceutically acceptable salt, solvate, tautomer, stereoisomer, prodrug molecule, metabolite, and one or more pharmaceutically acceptable carriers thereof.
Specifically, the medicament can be any one of the dosage forms in pharmaceutics, including tablets, capsules, soft capsules, gels, oral preparations, suspensions, granules, patches, ointments, pills, powders, injections, infusion solutions, freeze-dried injections, intravenous emulsions, liposome injections, suppositories, sustained-release preparations or controlled-release preparations.
Further, the pharmaceutically acceptable carrier refers to a pharmaceutical carrier conventional in the pharmaceutical field, such as: diluents, excipients, and water, and the like, fillers such as starch, sucrose, lactose, microcrystalline cellulose, and the like; binders such as cellulose derivatives, alginates, gelatin, and polyvinylpyrrolidone; humectants such as glycerol; disintegrating agents such as sodium carboxymethyl starch, hydroxypropyl cellulose, crosslinked carboxymethyl cellulose, agar, calcium carbonate and sodium bicarbonate; absorption enhancers such as quaternary ammonium compounds; surfactants such as cetyl alcohol, sodium lauryl sulfate; adsorption carriers such as kaolin and bentonite; lubricants such as talc, calcium and magnesium stearate, micronized silica gel, polyethylene glycol, and the like. Other adjuvants such as flavoring agent, sweetener, etc. can also be added into the composition.
The lignan compound is a new chemical component found in Illicium palmatum leaves by researchers, and is found to stably exist in Illicium palmatum leaves of each batch. <xnotran> (MS, </xnotran> 1 H-NMR、 13 C-NMR, etc.), and carrying out structural identification on the compound obtained by the method, thereby confirming that the compound is four new compounds with the structure shown in the formula (I). The invention also utilizes an LPS to induce an activity screening system such as a RAW 264.7 cell inflammation model and the like to carry out activity evaluation, and discovers that the compound has a certain protection effect on a mouse macrophage system RAW 264.7 and can obviously inhibit PGE 2 The release of (2) shows a strong anti-inflammatory effect. Has good research and development prospect.
Drawings
FIG. 1 is a HR-ESI-Q-TOF-MS spectrum of Compound 1 of the present invention;
FIG. 2 is a UV spectrum of Compound 1 of the present invention;
FIG. 3 is an IR spectrum of Compound 1 of the present invention;
FIG. 4 shows Compound 1 of the present invention 1 H-NMR spectrum
FIG. 5 shows Compound 1 of the present invention 13 A C-NMR spectrum;
FIG. 6 shows Compound 1 of the present invention 13 C-NMR and DEPT-135 spectra;
FIG. 7 is H of Compound 1 of the present invention 1 -H 1 COSY spectrum;
FIG. 8 is an HSQC spectrum of Compound 1 of the present invention;
FIG. 9 is an HMBC spectrum of compound 1 of the present invention;
FIG. 10 is a NOESY spectrum of Compound 1 of the present invention;
FIG. 11 is a CD map of Compound 1 of the present invention;
FIG. 12 is a HR-ESI-Q-TOF-MS spectrum of Compound 2 of the present invention;
FIG. 13 is a UV spectrum of Compound 2 of the present invention;
FIG. 14 is an IR spectrum of Compound 2 of the present invention;
FIG. 15 is a drawing of Compound 2 of the present invention 1 H-NMR spectrum
FIG. 16 shows Compound 2 of the present invention 13 A C-NMR spectrum;
FIG. 17 is a drawing of Compound 2 of the present invention 13 C-NMR and DEPT-135 spectra;
FIG. 18 is H of Compound 2 of the present invention 1 -H 1 COSY spectrum;
FIG. 19 is an HSQC spectrum of Compound 2 of the present invention;
FIG. 20 is a HMBC spectrum of compound 2 of the invention;
FIG. 21 is a NOESY spectrum of Compound 2 of the present invention;
FIG. 22 is a CD map of Compound 2 of the present invention.
Detailed Description
The following will specifically describe the contents of the experimental examples.
It is specifically noted that similar alternatives and modifications will be apparent to those skilled in the art, which are also intended to be included within the present invention. It will be apparent to those skilled in the art that the techniques of the present invention may be implemented and applied by modifying or appropriately combining the methods and applications described herein without departing from the spirit, scope, and content of the present invention. It is to be understood that the described embodiments are merely exemplary of the invention, and not restrictive of the full scope of the invention.
If the specific conditions are not indicated, the method is carried out according to the conventional conditions or the conditions suggested by manufacturers, and the used raw material medicines or auxiliary materials and the used reagents or instruments are the conventional products which can be obtained commercially.
EXAMPLE 1 preparation of the Compounds of the invention
(1) Extracting dry leaves of Illicium verum L.with 40% ethanol under reflux for 2 times (2 hr each time), mixing extractive solutions, and removing solvent under reduced pressure to obtain total extract. Dissolving the total extract in water, separating by HP-20 macroporous adsorbent resin column chromatography, sequentially eluting with water, 25% ethanol, 45% ethanol, and 90% ethanol, each gradient eluting for 4 column volumes (the same below), collecting eluates, respectively, concentrating under reduced pressure until no alcohol smell exists to obtain water eluate, 25% ethanol eluate, 45% ethanol eluate and 90% ethanol eluate;
(2) Taking the 45% ethanol elution fraction of step (1), separating by silica gel column chromatography, eluting with a dichloromethane-methanol gradient (95.
Wherein, the semi-preparative liquid chromatography conditions in the step (2) are as follows: phenomenex Gemini (C) 18 5 μm,10 × 250 mm), semi-preparative high performance liquid chromatography [ shimadzu, japan pump: LC-6AD (SHIMADZU, LIQUID CHROMATOGRAPH); a detector: SPD-20A (timing UV/VIS DETECTOR); a workstation: LC solution)]. The mobile phases of the compounds 1 to 2 are respectively in volume ratio: 17, 83.05 and 11.89, the detection wavelength being 240nm, the flow rate being 3mL/min.
EXAMPLE 2 preparation of Compounds of the invention
(1) Extracting dry leaves of Illicium verum L.with 50% ethanol under reflux for 2 times (2 hr each time), mixing extractive solutions, and removing solvent under reduced pressure to obtain total extract. Dissolving the total extract in water, separating by HP-20 macroporous adsorbent resin column chromatography, sequentially eluting with water, 30% ethanol, 50% ethanol, and 95% ethanol, collecting eluates, respectively, and concentrating under reduced pressure until no ethanol smell exists to obtain water eluate, 30% ethanol eluate, 50% ethanol eluate and 95% ethanol eluate;
(2) Taking the 50% ethanol elution fraction of step (1), separating by silica gel column chromatography, eluting with a dichloromethane-methanol gradient (100: 95: 5: 70) from 20.
Wherein, the semi-preparative liquid chromatography conditions in the step (2) are as follows: phenomenex Gemini (C) 18 5 μm,10 × 250 mm), semi-preparative high performance liquid chromatography [ shimadzu, japan pump: LC-6AD (SHIMADZU, LIQUID CHROMATOGRAPH); a detector: SPD-20A (timing UV/VIS DETECTOR); a workstation: LC solution)]. The mobile phases of the compounds 1 to 2 are respectively in the volume ratio of: 22, acetonitrile-water-formic acid of 0.1, acetonitrile-water-formic acid of 16.1, detection wavelength 254nm, flow rate 3mL/min.
EXAMPLE 3 preparation of the Compounds of the invention
(1) Taking dry leaves of Illicium verum, extracting with 60% ethanol under reflux for 2 times, each for 2 hr, mixing extractive solutions, and removing solvent under reduced pressure to obtain total extract. Dissolving the total extract in water, separating by HP-20 macroporous adsorbent resin column chromatography, sequentially eluting with water, 35% ethanol, 55% ethanol, and 100% ethanol, collecting eluates, respectively, and concentrating under reduced pressure until no ethanol smell exists to obtain water eluate, 35% ethanol eluate, 55% ethanol eluate, and 100% ethanol eluate;
(2) Taking the 55% ethanol elution fraction of step (1), separating by silica gel column chromatography, eluting with a dichloromethane-methanol gradient (90 20.
Wherein, the semi-preparative liquid chromatography conditions in the step (2) are as follows: phenomenex Gemini (C) 18 5 μm,10 × 250 mm), semi-preparative high performance liquid chromatography [ shimadzu, japan pump: LC-6AD (SHIMADZU, LIQUID CHROMATOGRAPH); a detector: SPD-20A (timing UV/VIS DETECTOR); a workstation: LC solution)]. The mobile phases of the compounds 1 to 4 are respectively in volume ratio: 27.73, acetonitrile-water-formic acid of 21.5, detection wavelength of 260nm, flow rate of 3mL/min.
EXAMPLE 4 structural characterization of the Compounds of the invention
Compound 1 as a pale yellow oil, [ alpha ]]25D-52.89 (c 0.45, meOH), HR-ESI-MS gives the excimer peak m/z 771.2847[ m ] +Na ] in positive ion mode] + (calculated: 771.2840) and the molecular formula of the compound is determined to be C 37 H 48 O 16 The unsaturation degree was 14.
As shown in FIGS. 1 to 11, of Compound 1 1 H-NMR(400MHz,in CD 3 OD) profile, showing three sets of aromatic signals: a group of 1,2,4-trisubstituted benzene ring signals [ delta ] H 7.09(1H,d,J=8.2Hz,H-5″),7.05(1H,d,J=1.1Hz,H-2″),6.89(1H,dd,J=8.2,1.1Hz,H-6″)]A group of symmetrical tetra-substituted benzene rings [ delta ] H 6.70(2H,s,H-2′,6′)]And a group of asymmetric 1,2,3,5-tetrasubstituted benzene ring signals [ delta ] H 6.74(1H,s,H-6),6.72(1H,s,H-2)](ii) a 1 group of sugar terminal signals and 4 methoxy hydrogen signals [ delta ] are observed in the high field region H 3.87(3H,s,5-OCH 3 ),3.83(3H,s,3″-OCH 3 ),3.78(6H,s,3′,5′-OCH 3 )]. Bonding of 13 C-NMR(100MHz,in CD 3 OD) spectrum gives 37 carbon signals including 3 benzene ring carbon signals (delta) C 154.5 × 2,150.4,147.5,147.2,145.3,139.6,137.4,137.2,136.2,129.6,120.9,117.9,117.4,114.2,112.3,104.0 × 2); 4 methine carbon signals (. Delta.) C 88.7,87.0,73.8,55.7); 5 methylene carbon signals (. Delta.) C 65.0,62.2,61.6,35.8,32.9) and 6 glycosyl carbon signals(δ C 102.8,78.2,77.8,74.9,71.4,62.6)。
NOESY patterns show the correlation peaks H-8'/H-2', H-8'/H-6' and H-7'/H-9', in combination with the coupling constant J at the 7',8' positions 7′,8′ =6.1Hz, relative configuration at the 7',8' positions is determined trans, coupling constant J at the 7',8' positions 7″,8″ =5.4Hz, relative configuration of the 7 ", 8" positions is erythro.
In a CD spectrum, the compound 1 shows a negative Cotton effect at 225nm (-2.38), and the absolute configuration of the 7',8' position is 7'S,8' R determined by combining relative configurations; shows a negative Cotton effect at 242nm (-1.47), determines 8'R, and determines 7' S in combination with relative configuration [43,45] Named as illiumignan N, the structure is as follows:
Figure BDA0003126052320000101
compound 2 white amorphous powder, [ alpha ]]25D-15.43 (c 0.7, meOH), having the same molecular formula as Compound 1 (m/z 771.2833, [ M ] +Na ]] + Calculating the value: 771.2840 And planar structures, taking into account differences in their configuration. NOESY patterns show the correlation peaks H-8'/H-2', H-8'/H-6' and H-7'/H-9', in combination with the coupling constant J at the 7',8' positions 7′,8′ =5.9Hz, determination of the relative configuration of the 7',8' positions as trans, coupling constant J in the 7 ", 8" positions 7″,8″ =5.2Hz, the relative configuration at position 7',8' is erythroid. In a CD spectrum, the compound 2 shows a negative Cotton effect at 228nm (-2.91), and the absolute configuration of the 7',8' position is 7'S,8' R determined by combining relative configurations; shows a positive Cotton effect at 243nm (+ 2.50), and determines the absolute configuration of 7 'R, 8' S, which is named as illiumignan P, and has the following structure:
Figure BDA0003126052320000102
TABLE 1 preparation of Compounds 1-2 1 H and 13 C NMR data
Figure BDA0003126052320000111
b:Measured at 400MHz for 1 H and 100MHz for 13 C in CD 3 OD
Multiplets and or overlapped signals are reported without designating multiplicity
EXAMPLE 5 in vitro anti-PGE of Compounds of the invention 2 Experiment of
1. Material
1.1 pharmaceutical compounds 1-2;
1.2 cell model mouse macrophage line RAW 264.7, from Chinese medicine academy of sciences; the culture conditions are as follows: DMEM +10% Fetal Bovine Serum (FBS), 37 ℃,5% CO 2
2. Principles and methods
2.1 principle of the experiment
Lipopolysaccharide (LPS) of gram-negative outer membrane (Sigma, USA, 114M 4009) is one of the most main pathogenic molecules mediating infectious inflammatory lesions, and many diseases are closely related to LPS-induced persistent subclinical inflammation. LPS is widely used to induce inflammation in animals and in cellular experiments.
Macrophages play a crucial role in the inflammatory response, and after stimulation, macrophages produce large amounts of inflammatory factors and mediators, such as: TNF-alpha, IL-1 beta, IL-6,NO and PGE 2 And the like. Activation of these inflammatory factors and mediators is a key process of inflammation, and their inhibition is often used as an important index for evaluating the anti-inflammatory activity of drugs.
2.2 drug Pair secretion of PGE 2 Inhibition test of
The method comprises the following steps:
(1) Preparing a liquid medicine: the compounds of the present invention were dissolved in 10% FBS in DMEM medium to prepare 2mg/ml stock solutions.
(2) The experimental method comprises the following steps: the cells were digested with 0.25% pancreatic enzymes (containing 0.02% EDTA), the cell density was adjusted to 1X 10 in DMEM medium containing 10% FBS 5 Each/ml, inoculated evenly into a 24-well plate, 400. Mu.l per well, and placed into an incubator for 24 hours after plating.
Blank control group (N group): 495 μ l serum-free DMEM medium was added to each well;
vehicle group/solvent control group (RM group): 495 mul serum-free DMEM medium containing one thousandth of DMSO was added to each well;
model group (group M): add 495. Mu.l of LPS 100. Mu.g/ml per well;
administration sample group: 495 mul of culture medium containing different concentrations of medicaments is added into each well;
simultaneously arranging 6 multiple holes, and putting CO into the 24-hole plate after the medicine is added 2 The cell culture box was incubated for 1 hour. After 1 hour, 5. Mu.l of LPS (final concentration: 1. Mu.g/ml) of 100. Mu.g/ml was added to each well except for the blank control and the solvent control, 5. Mu.l of serum-free DMEM medium was added to each well of the solvent control and the blank control, and after the addition of the reagents, the 24-well plate was placed in CO 2 The cell incubator was continued for 18 hours.
After 18 hours, cell culture fluid is collected, and PGE in cell supernatant is detected by ELISA method according to the kit instructions 2 The content of (b).
PGE 2 Inhibition ratio (%) = (model group PGE) 2 Average content of-sample group PGE 2 Average content of (3)/(model group PGE) 2 Average content of-solvent group PGE 2 Average content) x 100%.
3. Results of the experiment
3.1 drug sample on mouse macrophage line RAW 264.7 cell supernatant PGE 2 Influence of (2)
The result shows that the drug sample can obviously inhibit LPS to induce mouse macrophage RAW 264.7PGE 2 Shows strong anti-inflammatory action. Data results are shown in table 2.
TABLE 2 Compound 1-2 concentrations PGE supernatant of mouse macrophage cell line RAW 264.7 2 Influence of (A), (B)
Figure BDA0003126052320000131
n=6)
Figure BDA0003126052320000132
The invention adopts Graphad prism 7.00 analysis software, and compounds 1-2 in the invention measured by a linear regression analysis method inhibit LPS in vitro to induce mouse macrophage RAW 264.7 to secrete inflammatory medium PGE 2 Average IC of 50 19.04. Mu.M and 16.53. Mu.M, respectively.
4. Conclusion
The compound of the invention induces mouse macrophage RAW 264.7 to secrete inflammatory medium PGE by LPS 2 Has remarkable inhibitory effect, shows strong anti-inflammatory effect, and can treat PGE with the increase of drug concentration 2 Increased inhibition of secretion, IC of Compounds 1-2 50 19.04. Mu.M and 16.53. Mu.M, respectively.
The foregoing is only a preferred embodiment of the present invention, and it should be noted that, for those skilled in the art, various modifications and decorations can be made without departing from the principle of the present invention, and these modifications and decorations should also be regarded as the protection scope of the present invention.

Claims (10)

1. A lignan compound or a pharmaceutically acceptable salt, solvate, tautomer, stereoisomer, prodrug molecule and metabolite thereof is disclosed, wherein the structure of the compound is shown in formula I:
Figure FDA0003126052310000011
2. a process for preparing a compound of claim 1, comprising:
a) Taking Illicium verum leaves of safflower, carrying out reflux extraction by 40-60% ethanol, and removing a solvent to obtain a total extract;
b) Dissolving the total extract in water, separating by macroporous adsorption resin column chromatography, eluting with water, 25-35% ethanol, 45-55% ethanol and 90-100% ethanol in sequence, collecting eluates respectively, and concentrating under reduced pressure to obtain water eluate, 25-35% ethanol eluate, 45-55% ethanol eluate and 90-100% ethanol eluate;
c) Taking the 45-55% ethanol elution part, separating by silica gel column chromatography, performing gradient elution and collection by using dichloromethane-methanol to obtain 15 fractions of 3A-3O, performing gradient elution on fraction 3L by using ODS column chromatography methanol-water to obtain 5 fractions of 3L1-3L5, performing gradient elution on fraction 3L4 by using polyamide column chromatography ethanol-water to obtain 6 fractions of 3L4A-3L4F, performing semi-preparative liquid chromatography on 3L4A to obtain 8 fractions of 3L4A2-3L4A9, performing semi-preparative liquid chromatography on 3L4A4 to obtain a compound 1,3L4A6, and performing semi-preparative liquid chromatography on the compound 2.
3. The method of claim 2, wherein the step a) comprises:
taking dry illicium griseum leaves, carrying out reflux extraction for 1-3 times by 3-5 times of 40-60% ethanol, each time for 1-3 hours, combining extracting solutions, and removing the solvent under reduced pressure to obtain the total extract.
4. The method of manufacturing according to claim 2, wherein the step B) includes: sequentially eluting with water, 30% ethanol, 50% ethanol and 95% ethanol, respectively collecting eluates, and concentrating under reduced pressure until no ethanol smell exists to obtain water eluate, 30% ethanol eluate, 50% ethanol eluate and 95% ethanol eluate.
5. The process according to claim 2, wherein the dichloromethane-methanol gradient elution of step C) is a gradient elution performed at a volume ratio of 100; the methanol-water gradient elution is performed by gradient elution at a volume ratio of 30; the ethanol-water gradient elution is performed in a gradient from 10 to 90 to 95.
6. The preparation method according to claim 2, wherein the macroporous adsorption resin comprises one or more of D101 type macroporous adsorption resin, HP-20 type macroporous adsorption resin, HPD-100A type macroporous adsorption resin or HPD-300 type macroporous adsorption resin.
7. The method of claim 2, wherein the semi-preparative liquid chromatography conditions include: specification C 18 5 μm, 10X 250mm Phenomenex Gemini column; the mobile phase volume ratios for preparing compounds 1 to 2 were respectively: 17-27, acetonitrile-water-formic acid of 0.05-0.5 and acetonitrile-water-formic acid of 11-21, 89-0.05-0.5, the detection wavelength is 240-260nm, and the flow rate is 2-4mL/min.
8. The method according to claim 7, wherein the step A) is a reflux extraction with 50% ethanol for 2 times, each for 2 hours;
preparation of compounds 1-2 acetonitrile-water-formic acid with mobile phase volume ratio of 22.1 and acetonitrile-water-formic acid of 16.1.
9. Use of the lignan-type compound of claim 1, or a pharmaceutically acceptable salt, solvate, tautomer, stereoisomer, prodrug molecule, or metabolite thereof for the manufacture of an anti-inflammatory agent.
10. A medicament comprising the lignan-type compound of claim 1, or a pharmaceutically acceptable salt, solvate, tautomer, stereoisomer, prodrug molecule, metabolite thereof.
CN202110690684.2A 2021-06-22 2021-06-22 Lignan compound and preparation method and application thereof Active CN115504950B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN202410066479.2A CN117986309A (en) 2021-06-22 2021-06-22 Lignan compound and preparation method and application thereof
CN202110690684.2A CN115504950B (en) 2021-06-22 2021-06-22 Lignan compound and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110690684.2A CN115504950B (en) 2021-06-22 2021-06-22 Lignan compound and preparation method and application thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
CN202410066479.2A Division CN117986309A (en) 2021-06-22 2021-06-22 Lignan compound and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN115504950A true CN115504950A (en) 2022-12-23
CN115504950B CN115504950B (en) 2024-01-26

Family

ID=84499169

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202410066479.2A Pending CN117986309A (en) 2021-06-22 2021-06-22 Lignan compound and preparation method and application thereof
CN202110690684.2A Active CN115504950B (en) 2021-06-22 2021-06-22 Lignan compound and preparation method and application thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CN202410066479.2A Pending CN117986309A (en) 2021-06-22 2021-06-22 Lignan compound and preparation method and application thereof

Country Status (1)

Country Link
CN (2) CN117986309A (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1634025A (en) * 2004-09-27 2005-07-06 深圳中药及天然药物研究中心 Use of erberry and its lignanoids compound in drug for osteoporosis
CN106083770A (en) * 2016-06-14 2016-11-09 江苏康缘药业股份有限公司 A kind of Lignanoids compounds and its preparation method and application

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1634025A (en) * 2004-09-27 2005-07-06 深圳中药及天然药物研究中心 Use of erberry and its lignanoids compound in drug for osteoporosis
CN106083770A (en) * 2016-06-14 2016-11-09 江苏康缘药业股份有限公司 A kind of Lignanoids compounds and its preparation method and application

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
HAI JIANG等: ""New phenylpropanoid derivatives from the fruits of Xanthium sibiricum and their anti-inflammatory activity"", FITOTERAPIA, vol. 117, pages 11 - 15 *

Also Published As

Publication number Publication date
CN115504950B (en) 2024-01-26
CN117986309A (en) 2024-05-07

Similar Documents

Publication Publication Date Title
CN113105388B (en) Euphorbia lathyris diterpene alkyl compound and extraction method and application thereof
CN115154476B (en) Cyclocarya paliurus extract and application thereof in resisting gout and reducing uric acid
CN109134486B (en) Coumarin lignan, preparation method and application thereof
CN111704641B (en) Iridoid glycoside compound and preparation method and application thereof
CN112047954B (en) Iridoid compound and preparation method and application thereof
CN115160251B (en) N-N-bis-oxazolidinone alkaloid compound, preparation method and application thereof in medicine field
CN111909228B (en) Alkaloid compound and preparation method and application thereof
CN115504950B (en) Lignan compound and preparation method and application thereof
CN113956229B (en) Lignan compound in lilac and preparation method and application thereof
CN115703753B (en) Benzofuran derivative and preparation method and application thereof
CN111732619B (en) Iridoid glycoside compound and preparation method and application thereof
CN116637102B (en) Application of flavane compound in preparation of antitumor drugs
CN108948040B (en) Gilmaxane type sesquiterpene compound extracted from herba Centellae and application thereof
CN109206392B (en) Coumarin compound and preparation method and application thereof
CN116730919A (en) Diterpene alkaloid compound and extraction method and application thereof
CN115703814A (en) Phenolic glycoside compound and preparation method and application thereof
CN111995645B (en) Phenylpropanoid compound and preparation method and application thereof
CN115505015A (en) Lignans compound and preparation method and application thereof
CN112409260B (en) Alkaloid compound and preparation method and application thereof
CN114853712B (en) Chromane or chromene type hetero-terpenoid, and extraction method and application thereof
CN114394931B (en) Monoterpene alkaloid with vasodilation activity and extraction method and application thereof
CN113816857B (en) Caffeoyl quinic acid ester compound and preparation method and application thereof
CN108530505A (en) A kind of flavonoid glycoside compound and its preparation method and application
CN116854653A (en) Diphenyl heptane compound and its preparation method and application
EP1411919B1 (en) (-)-olivil as antioxidant

Legal Events

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