CN116675602B - Pinane diterpene and preparation method and application thereof - Google Patents

Pinane diterpene and preparation method and application thereof Download PDF

Info

Publication number
CN116675602B
CN116675602B CN202310961491.5A CN202310961491A CN116675602B CN 116675602 B CN116675602 B CN 116675602B CN 202310961491 A CN202310961491 A CN 202310961491A CN 116675602 B CN116675602 B CN 116675602B
Authority
CN
China
Prior art keywords
diterpene
component
spots
separating
silica gel
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.)
Active
Application number
CN202310961491.5A
Other languages
Chinese (zh)
Other versions
CN116675602A (en
Inventor
林永强
郭东晓
崔伟亮
张雪
薛菲
许丽丽
刘青芝
赵淑秀
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.)
Shandong Institute for Food and Drug Control
Original Assignee
Shandong Institute for Food and Drug Control
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 Shandong Institute for Food and Drug Control filed Critical Shandong Institute for Food and Drug Control
Priority to CN202310961491.5A priority Critical patent/CN116675602B/en
Publication of CN116675602A publication Critical patent/CN116675602A/en
Application granted granted Critical
Publication of CN116675602B publication Critical patent/CN116675602B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C62/00Compounds having carboxyl groups bound to carbon atoms of rings other than six—membered aromatic rings and containing any of the groups OH, O—metal, —CHO, keto, ether, groups, groups, or groups
    • C07C62/30Unsaturated compounds
    • C07C62/32Unsaturated compounds containing hydroxy or O-metal groups
    • 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]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2603/00Systems containing at least three condensed rings
    • C07C2603/02Ortho- or ortho- and peri-condensed systems
    • C07C2603/04Ortho- or ortho- and peri-condensed systems containing three rings
    • C07C2603/22Ortho- or ortho- and peri-condensed systems containing three rings containing only six-membered rings
    • C07C2603/26Phenanthrenes; Hydrogenated phenanthrenes

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention belongs to the technical field of natural product preparation, and in particular relates to a pinane diterpene and a preparation method and application thereof, wherein the new pinane diterpene is obtained by separating cortex acanthopanacis serving as a traditional Chinese medicine material through a combined column chromatography technology: 14-hydroxy-16-de-ethenyl-enantiomer-pimpinene-8, 11, 13-triene-17-carboxylic acid, the structure of which is determined by nuclear magnetic resonance, mass spectrum and other techniques. The Griess method is adopted to test a lipopolysaccharide-induced mouse mononuclear macrophage RAW264.7 cell release inflammatory factor model, and the anti-inflammatory activity of the compound is evaluated, so that the compound has certain anti-inflammatory activity. The invention obtains a new component, and defines the extraction and separation and structure identification method of the new component, the method is simple and convenient, and the component can be obtained repeatedly and continuously. Meanwhile, a material basis is provided for the research and development of new anti-inflammatory drugs.

Description

Pinane diterpene and preparation method and application thereof
Technical Field
The invention belongs to the technical field of natural product preparation, and particularly relates to a new chemical structure of a sea squirrel diterpene extracted from cortex acanthopanacis and application thereof.
Background
The disclosure of this background section is only intended to increase the understanding of the general background of the invention and is not necessarily to be construed as an admission or any form of suggestion that this information forms the prior art already known to those of ordinary skill in the art.
Cortex Acanthopancis of Acanthopanax gracilistylus of Acanthopanax of AraliaceaeAcanthopanax gracilistylusW.W.Smith) is mainly distributed in Hubei and He of ChinaThe Chinese medicinal materials are traditional Chinese medicinal materials commonly used in clinic in the provinces of south, shaanxi, anhui, zhejiang, liaoning, hebei and the like. The cortex acanthopanacis is originally carried in the book meridian, has the main effects of dispelling wind and removing dampness, tonifying liver and kidney, strengthening tendons and bones, inducing diuresis and relieving swelling, and is clinically mainly used for treating diseases such as rheumatic arthralgia, flaccidity of bones and muscles, infantile slow running, body deficiency and general power, edema, beriberi and the like, and the chemical components of the cortex acanthopanacis mainly comprise terpenes, sterols, organic acids, phenylpropanoids, flavonoids and the like. Modern pharmacology shows that cortex acanthopanacis has good anti-inflammatory activity.
In the prior art, the research on chemical components and medicinal effects of cortex acanthopanacis is less, and the plant cannot be reasonably and effectively utilized.
Disclosure of Invention
Based on the research background, the invention extracts the new-structure pimarane diterpenoid compound from the cortex acanthopanacis, performs in-vitro anti-inflammatory activity, and provides a material basis for the research and development of anti-inflammatory new drugs.
The invention discloses a kind of sea-pinane diterpene, the structural formula is shown in formula I,
the identification of the chemical components of formula I is referred to in the corresponding parts of the specific embodiments.
The invention discloses a preparation method of a pinane diterpene with a structural formula shown in formula I, which comprises the following steps:
(1) Preparing a crude extract;
(2) Carrying out fractional extraction;
(3) Separating and purifying;
(4) Identification of chemical components.
The method comprises the following steps:
(1) Pulverizing cortex Acanthopanacis, extracting with 70% ethanol under reflux, collecting middle and small polar parts, and steaming at 60deg.C to obtain crude extract;
(2) Dispersing the crude extract with warm water, extracting with diethyl ether, standing to completely layer, separating the extraction solvent, repeatedly extracting, mixing the extractive solutions, and concentrating under reduced pressure to obtain diethyl ether layer extract;
(3) Separating the ether layer extract by using an MCI resin column, performing gradient elution by using a methanol aqueous solution, wherein the fraction A passes through a forward silica gel chromatographic column, performing gradient elution by using a petroleum ether-acetone system, detecting by using a thin-layer silica gel plate, combining the same components, targeting to find spots with dark spots and fluorescence under an ultraviolet lamp in the silica gel thin-layer chromatography, and separating the component A1 with the target spots by using a liquid chromatography to obtain a target compound;
(4) The chemical structure of the target compound is determined by adopting high-resolution mass spectrum, 1D nuclear magnetic resonance spectrum and other technologies.
Further, in the step (1), the extraction mode of heating and refluxing is adopted for 3 times, 1 time h each time, so that the material extraction is ensured to be complete.
Further, in the step (1), the reduced pressure rotary steaming temperature is 57 ℃, so that the target component is prevented from being damaged by long-time high-temperature heating.
Further, in the step (2), the extraction is repeated for 3 times, and the volume ratio of diethyl ether to water layer used in the extraction is 1:1.
In the step (3), the MCI resin column is subjected to gradient elution by 40% -100% of methanol aqueous solution, so that the target component is separated from other interference components.
Further, in the step (3), the fraction A is subjected to forward silica gel chromatographic column and is subjected to gradient elution by petroleum ether-acetone system (100:1-1:1), so that the target component is further separated from other interference components.
Further, in the step (3), the observation wavelength of the dark spots is 254.+ -.2 nm, the observation wavelength of the fluorescence is 365.+ -.2 nm, and the spots with special colors after the development are black brown spots after the development with 10% sulfuric acid-ethanol solution.
Further, component A1 in step (3) is purified by liquid chromatography TM RP18 chromatographic column (4.6 mm ×250 mm,5 μm) at 35 deg.C and methanol-water 70:30 to obtain the target component compound monomer.
The invention discloses application of a pinane diterpene with a structural formula shown in a formula I in preparation of anti-inflammatory drugs.
Further, anti-inflammatory activity is achieved by reducing NO release in the cell.
Compared with the prior art, the invention has the beneficial effects that:
the invention provides a new component obtained by using the compound with a new structure, which defines the extraction and separation and structure identification methods of the new component, and adopts separation methods such as reflux extraction, solvent extraction, MCI, silica gel column, HPLC chromatography and the like, thereby having simple method and being capable of repeatedly and continuously obtaining the component. Meanwhile, the component shows a certain in-vitro anti-inflammatory activity, and provides a material basis for the research and development of new anti-inflammatory drugs.
Drawings
The accompanying drawings, which are included to provide a further understanding of the invention and are incorporated in and constitute a part of this specification, illustrate embodiments of the invention and together with the description serve to explain the invention.
Fig. 1: critical HMBC (% and) of compounds 1 H- 1 H COSY (━) panels.
Fig. 2: the effect of the compound on RAW264.7 cell viability and the inhibition of NO release (n=3, MTT assay corresponds to the right ordinate, in% and NO assay corresponds to the left ordinate, in. Mu. Mol/L; diDOX: positive control, C: blank control, LPS: LPS addition).
Fig. 3: HR-ESI-MS of a Compoundm/z
Fig. 4: of compounds 1 H NMR spectrum.
Fig. 5: of compounds 13 C NMR spectrum.
Fig. 6: HMBC spectra of compounds.
Fig. 7: HSQC spectrum of the compound.
Fig. 8: of compounds 1 H- 1 H COSY profile.
Fig. 9: NOESY spectra of the compounds.
Fig. 10: key NOESY («) profile of compounds.
Detailed Description
It should be noted that the following detailed description is illustrative and is intended to provide further explanation of the invention. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
It is noted that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting of exemplary embodiments according to the present invention. The singular is also intended to include the plural unless the context clearly indicates otherwise, and furthermore, it is to be understood that when the terms "comprises" and/or "comprising" are used in this specification, they specify the presence of stated features, steps, operations, devices, components, and/or combinations thereof.
In order to enable those skilled in the art to more clearly understand the technical scheme of the present invention, the technical scheme of the present invention will be described in detail with reference to specific embodiments.
1 instrument, reagent and materials
1.1 Instrument and reagent
Bruker Avance III 600 Nuclear magnetic resonance spectrometer (Bruker, switzerland); METTLER TOLEDO XSE205 and 205; a switzerland Buchi R210 rotary evaporator; DP series oilless vacuum pump: shi take advantage of the world science and technology (Tianjin) limited company; agilent 1200 HPLC chromatograph; HPLC-IT-TOF-MS liquid chromatography-mass spectrometry (Shimadzu corporation, japan); bio Tek EPOCH2NS microplate reader.
Reagents such as diethyl ether, n-butanol, ethyl acetate, methanol, ethanol, petroleum ether, acetone, methylene dichloride and the like are all analytically pure; MCI GEL CHP20P: 37-75 μm, mitsubishi chemical corporation, japan; column layer chromatography silica gel: 200-300 mesh, qingdao Bay Fine chemical Co., ltd; sephadex LH-20 (Sephadex LH-20) Beijing Soy Bao technology Co., ltd; normal phase silica gel thin layer plate: the institute of chemical industry in the tobacco stand market; MTT solution: beijing Ding Guo prosperous biotechnology limited company; (Eagle) DMEM high sugar medium: gibco; fetal bovine serum: shanghai Datthe Biotech Co., ltd; dimethyl sulfoxide (DMSO): LIFE SCIENCE company; LPS lipopolysaccharide: sigma Aldrich(Shanghai) trade Limited; para-aminobenzenesulfonamide: beijing Ding Guo prosperous biotechnology limited company; pancreatic enzyme: beijing Ding Guo prosperous biotechnology limited company; DIDOX: MCE. Cortex Acanthopancis is obtained from Hubei, and is a plant of AraliaceaeAcanthopanax gracilistylusW.w. Smith dried root.
2 Experimental methods
2.1 Preparation of crude extract
Pulverizing the collected cortex Acanthopanacis medicinal material 4 Kg, placing into a 5L round bottom flask, extracting with 70% ethanol for 3 times by adopting a heating reflux extraction mode, each time 1 h, mixing the extractive solutions, and recovering solvent under reduced pressure and rotary steaming at 57 ℃ until the obtained solution is odorless, thus obtaining ethanol crude extract 600 g.
2.2 Graded extraction
Dispersing the crude extract with warm water, extracting with diethyl ether, standing to completely delaminate, separating the extraction solvent, repeating extraction for 3 times, mixing the extracts, and concentrating under reduced pressure to obtain diethyl ether layer extract 130.2 g.
2.3 Separation and purification
Dissolving the ether layer extract, then passing through an MCI resin column, sequentially carrying out gradient elution by 40% -100% methanol to obtain 7 fractions, wherein the fraction 4 is subjected to forward silica gel chromatographic column, gradient elution by petroleum ether-acetone system (100:1-1:1), and detection by a thin-layer silica gel plate, and combining the same components to obtain Fr. A-X total 24 components. The target compound is obtained by separating the component J through liquid chromatography (column temperature: 35 ℃ C., methanol-water 70:30).
2.4 identification of chemical Components
2.4.1 Nuclear magnetic resonance analysis
And taking a proper amount of the separated compounds, respectively dissolving the compounds by using deuterated chloroform, and then placing the compounds into a nuclear magnetic tube, and measuring the compounds by using a Bruker Avance III 600 nuclear magnetic resonance spectrometer.
2.4.2 Mass spectrometry analysis
A small amount of the test compound was taken and dissolved in methanol and analyzed by a mass spectrometer. Electrospray ion source (ESI), sample injection flow rate is set to be 0.3 mu L/h, and detection mode positive and negative ion switching is carried out simultaneously for detection: scanning rangem/z50-1500; ion accumulation time50 ms; the flow rate of the atomized gas (nitrogen) is 1.5L/min; the temperature of the curve desolventizing pipe and the temperature of the heating module are 200 ℃; CID energy 50%; detector voltage, 1.69 kV; the dry gas (nitrogen) pressure was 111 kPa.
2.5 anti-inflammatory Activity Studies
2.5.1 RAW264.7 cell culture
At 37℃with 5% CO 2 In the environment, fetal bovine serum was added to the medium cultured in high-sugar DMEM, and resuscitated RAW264.7 cells were added. Taking cells in logarithmic growth phase, counting and diluting the cell density to 8×10 5 mu.L. Add 96-well plates, 200 μl per well.
2.5.2 MTT method for detecting influence of drug on RAW264.7 cell viability
Regulating cell concentration of logarithmic growth phase cells to 8×10 5 mu.L, 200. Mu.L per well, were inoculated into 96-well plates, and the group treatment was performed as a blank control group, a positive control group (DiDO: 3, 4-dihydroxybenzoic hydroxamic acid), and an experimental group. After the cells added with the drugs are respectively cultured for 24 h, 20 mu L of MTT solution is added into each hole, the culture is continued for 2 h, the supernatant is removed, 200 mu L of DMSO is added and vibrated away from light for 10 min, the Optical Density (OD) value of each hole is detected at 570 nm wavelength by adopting a full-wavelength enzyme-labeled instrument, and the cell survival rate is calculated.
2.5.3 testing of the monomeric Compounds for the release of NO from RAW264.7 cells
Taking RAW264.7 cells in logarithmic growth phase, and adjusting cell concentration to 2×10 5 Per mL, 200. Mu.L per well in 96-well plates at 37℃with 5% CO 2 After 24. 24 h culture in the environment, the supernatant was removed and the cells were divided into a blank (blank medium), a lipopolysaccharide (5. Mu.g/mL lipopolysaccharide), and compounds of different mass concentrations (1.0. Mu.g/mL lipopolysaccharide+100, 50, 25, 12.5, 6.25, 3.125. Mu.g/mL). After each group of cells was added with drug or blank medium, the culture was continued for 24 h. Taking 100 mu L of supernatant, adding 100 mu L of Griess reagent, standing at room temperature for 10 min, setting the wavelength to 570 nm, measuring absorbance on a microplate reader, substituting the absorbance into a standard curve, and calculating the NO release amount.
3 results and analysis
3.1 isolation results
Separating and purifying the diethyl ether layer extract by MCI resin column, silica gel column chromatography, and thin layer plate preparation separation method and thin layer chromatography detection analysis to obtain the target compound 3.3 mg. The structural formula is shown as a formula I,
3.2 structural identification of Compounds
White amorphous powder, easily dissolved in methanol, developed with a thin layer of petroleum ether-ethyl acetate (7:3), had dark spots under 254 nm, fluorescence under 365 nm, and developed black brown spots after heating by spraying 10% sulfuric acid-ethanol. HR-ESI-MSm/ z) 287.1641 [M-H] - (FIG. 3), theoretical value 287.1653, deviation 4.2 ppm, and its molecular formula C 18 H 24 O 3 The unsaturation was 7. 1 H NMR (FIG. 4, table 1) shows that there are 2 angular methyl groups [δ H 1.12(s), 1.27(s)]And 1 double bond-linked methyl groupδ H 2.14(s)]Is present. By HSQC (FIG. 7) and 13 c NMR (FIG. 5)δ C 122.7 (C-8), 147.2 (C-9), 116.6 (C-11), 127.4 (C-12), 120.3 (C-13), 151.7 (C-14) 6C signals can determine the presence of a benzene ring,δ H 6.73(J=8.4 Hz) andδ H 6.84(J=8.4 Hz) are two adjacent hydrogen protons on the benzene ring. By HMBC (FIG. 6)δ H 6.84(J=8.4 Hz) is correlated with the C-9, C-11 and C-14 signals,δ H 6.73(J=8.4 Hz) with C-8, C-9 and C-13 signals, it was determined that the phencyclized hydrogens are at the 11 and 12 positions; the hydroxyl group at the 14 position can be deduced through the related signals of C-14, H-7 and H-15 in HMBC; by H-16 in HMBCδ C 180.1 the correlation may speculate that the carboxilic group is at position 17. According to HMBCδ C 147.2 to H-7, H-12, H-18,δ C 122.7 to H-6, H-7, H-11, H-14,δ C the correlation of 38.1 with H-1, H-2, H-5, H-18 can further confirm its structure. According to 1 H- 1 Correlation of H-1 with H-2, H-2 with H-3, H-5 with H-6, H-6 with H-7, H-11 with H COSY (FIG. 8)H-12, in combination with information in HMBC, can determine its planar structure (see FIG. 1). By the NOESY (FIG. 9) spectra of H-5 and H-1β、H-7βH-16, and deducing its relative configuration (see FIG. 10). Thus, the structure of the compound was identified as 14-hydroxy-16-de-ethenyl-enantiomer-pimo-8, 11, 13-triene-17-carboxylic acid.
The compounds of Table 1 1 H and 13 c NMR data
Position of δ H δ C
1α 2.23(m) 39.7
1β 1.31(m)
2α 1.98(m) 19.9
2β 1.56(m)
3α 2.21(m) 37.5
3β 1.07(dt,J= 13.2, 4.2 Hz)
4 43.5
5 1.47(d,J= 12.6 Hz) 52.5
6α 2.19(m) 20.4
6β 1.96(m)
7α 2.90(dd,J= 16.8, 5.4 Hz) 25.8
7β 2.45(m)
8 122.7
9 147.2
10 38.1
11 6.73(d,J= 8.4 Hz) 116.6
12 6.84(d,J= 8.4 Hz) 127.4
13 120.3
14 151.7
15 2.14(s) 14.7
16 1.27(s) 27.9
17 180.1
18 1.12(s) 22.3
Note that: 1 h NMR 13 C NMR was 600 and 150 MHz respectively; the solvent is CD 3 OD。
Note that: 1 h NMR 13 C NMR was 600 and 150 MHz respectively; the solvent is CD 3 OD。
3.3 screening results for anti-inflammatory Activity
3.3.1 Effect of monomer Compounds on cell RAW264.7 cell viability
Cells were divided into a blank control group, a positive control group (DiDO: 3, 4-dihydroxybenzohydroxamic acid) and an experimental group. Compared with a blank control group, the compound has the drug concentration within the range of 3.125-100 mug/ml, is nontoxic to cells, and has the cell activity of more than 80%. The results are shown in FIG. 2.
3.3.2 determination of NO content in LPS stimulated RAW264.7 cells by monomeric Compounds
Cells were divided into a blank (blank medium, without lipopolysaccharide LPS), a lipopolysaccharide group (5. Mu.g/mL lipopolysaccharide LPS), a positive control (1.0. Mu.g/mL lipopolysaccharide+100, 50. Mu.g/mL 3, 4-dihydroxybenzoic hydroxamate DiDO), compounds of different mass concentrations (1.0. Mu.g/mL lipopolysaccharide+100, 50, 25, 12.5, 6.25, 3.125. Mu.g/mL compounds). As a result, as shown in FIG. 2, the compound had a certain anti-inflammatory activity at 50, 100. Mu.g/ml, and the amount of NO released decreased with increasing concentration, as compared with LPS group. At a concentration of 100. Mu.g/ml, the anti-inflammatory activity was comparable to that of the positive drug DiDO 50. Mu.g/ml.
The anti-inflammatory activity shows that the compound has weaker anti-inflammatory activity, and can obviously inhibit NO release in RAW264.7 cells induced by LPS when reaching a certain concentration (more than 50 mu g/ml), thereby inhibiting NO-mediated inflammatory reaction. Meanwhile, experiments show that the component has no toxicity to cells, so that the component can be developed into an anti-inflammatory drug.
The above description is only of the preferred embodiments of the present invention and is not intended to limit the present invention, but various modifications and variations can be made to the present invention by those skilled in the art. Any modification, equivalent replacement, improvement, etc. made within the spirit and principle of the present invention should be included in the protection scope of the present invention.

Claims (8)

1. A kind of sea pine alkane diterpene is characterized in that the structural formula is shown in the formula I,
2. the process for the preparation of a norpimaran diterpene of formula I of claim 1, characterized by the steps of:
(1) Preparing a crude extract: pulverizing cortex Acanthopanacis, extracting with 70% ethanol under reflux, collecting polar part, and steaming at 60deg.C under reduced pressure to obtain crude extract;
(2) And (3) carrying out fractional extraction: dispersing the crude extract with warm water, extracting with diethyl ether, standing to completely layer, separating the extraction solvent, repeatedly extracting, mixing the extractive solutions, and concentrating under reduced pressure to obtain diethyl ether layer extract;
(3) And (3) separating and purifying: separating the ether layer extract by using an MCI resin column, performing gradient elution by using a methanol aqueous solution, wherein the fraction A passes through a forward silica gel chromatographic column, performing gradient elution by using a petroleum ether-acetone system, detecting by using a thin-layer silica gel plate, combining the same components, targeting to find spots with dark spots and fluorescence under an ultraviolet lamp in the silica gel thin-layer chromatography, and separating the component A1 with the target spots by using a liquid chromatography to obtain a target compound;
(4) Identification of chemical components: the chemical structure of the target compound is determined using high resolution mass spectrometry, 1D and 2D nmr spectroscopy.
3. The process according to claim 2, wherein the reduced pressure rotary evaporation temperature in step (1) is 57 ℃.
4. The method according to claim 2, wherein the MCI resin column in step (3) is eluted with a 40% -100% aqueous methanol gradient to separate the target component from other interfering components.
5. The preparation method according to claim 2, wherein the fraction A in the step (3) is subjected to forward silica gel chromatographic column, and is subjected to gradient elution by a petroleum ether-acetone system, and the volume ratio of petroleum ether to acetone is 100:1-1:1, so that the target component is further separated from other interference components.
6. The method according to claim 2, wherein the dark spots observed in the step (3) have a wavelength of 254.+ -.2 nm and a fluorescence observed wavelength of 365.+ -.2 n, and the spots having a specific color after the development are spots which develop a black brown color in sunlight after the development with a 10% sulfuric acid-ethanol solution.
7. The process according to claim 2, wherein component A1 in step (3) is purified by liquid chromatography TM RP18 chromatographic column with temperature of 35 deg.C and methanol-water separation of 70:30 to obtain the target component compound monomer.
8. Use of a norpimaran-type diterpene of formula I as claimed in claim 1 for the manufacture of an anti-inflammatory agent whose anti-inflammatory activity is achieved by reducing NO release in cells.
CN202310961491.5A 2023-08-02 2023-08-02 Pinane diterpene and preparation method and application thereof Active CN116675602B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310961491.5A CN116675602B (en) 2023-08-02 2023-08-02 Pinane diterpene and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310961491.5A CN116675602B (en) 2023-08-02 2023-08-02 Pinane diterpene and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN116675602A CN116675602A (en) 2023-09-01
CN116675602B true CN116675602B (en) 2023-10-20

Family

ID=87784054

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310961491.5A Active CN116675602B (en) 2023-08-02 2023-08-02 Pinane diterpene and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN116675602B (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016114707A1 (en) * 2015-01-12 2016-07-21 Elinder Fredrik Dehydroabietic acid (dhaa) derivatives for use as ion channel openers
CN109908133A (en) * 2019-04-04 2019-06-21 山东大学 A kind of pimarane type diterpene is preparing the application in anti-oxidation stress medicine

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016114707A1 (en) * 2015-01-12 2016-07-21 Elinder Fredrik Dehydroabietic acid (dhaa) derivatives for use as ion channel openers
EP3245184A1 (en) * 2015-01-12 2017-11-22 Elinder, Fredrik Dehydroabietic acid (dhaa) derivatives for use as ion channel openers
CN109908133A (en) * 2019-04-04 2019-06-21 山东大学 A kind of pimarane type diterpene is preparing the application in anti-oxidation stress medicine

Also Published As

Publication number Publication date
CN116675602A (en) 2023-09-01

Similar Documents

Publication Publication Date Title
Huo et al. Neolignan glycosides from Symplocos caudata
CN101732383B (en) Total sesquiterpene lactone extract of centipeda minima, preparation method and application thereof
Liu et al. Three new monoterpene glycosides from oil peony seed cake
Zhao et al. Neuroprotective polyhydroxypregnane glycosides from Cynanchum otophyllum
CN110452113B (en) (4 → 2) rearranged clerodane diterpenoid compound and preparation method and application thereof
CN109824489A (en) A kind of compound with anti-inflammatory activity extracted from Radix Glycyrrhizae and its application
Fan et al. Diterpenoid alkaloids from the whole plant of Aconitum tanguticum (Maxim.) Stapf
CN106831384B (en) Cadinane sesquiterpene compound and preparation method and application thereof
Xiang et al. Chemical Constituents from the Roots of Polygala arillata and Their Anti‐Inflammatory Activities
CN116606273B (en) Preparation method of oleander flavone with whitening effect
CN116675602B (en) Pinane diterpene and preparation method and application thereof
Li et al. Enhanced recovery of four antitumor ganoderic acids from Ganoderma lucidum mycelia by a novel process of simultaneous extraction and hydrolysis
CN111943845A (en) Two sesquiterpenes with antitumor activity and preparation method and application thereof
Baek et al. Chemical constituents of Abies koreana leaves with inhibitory activity against nitric oxide production in BV2 microglia cells
CN114276364B (en) Sesquiterpenoids in Artemisia mongolica, and preparation method and application thereof
CN112274522B (en) New anti-inflammatory application of phenolic glycoside compounds in garden burnet root and extraction and separation method thereof
CN112079897A (en) Two steroid compounds and preparation method and application thereof
CN111675682A (en) Benzopyran compound and preparation method and application thereof
Tangmouo et al. Norbergenin derivatives from the stem bark of Diospyros sanza-minika (Ebenaceae) and their radical scavenging activity
CN114478683B (en) Schisandra chinensis lactone A and preparation and application thereof
Lee et al. Anti-inflammatory and cytotoxic compounds from solanum macaonense
Park et al. A new phenyl ethyl glycoside from the twigs of Acer tegmentosum
Wang et al. Bioactive ellagitannins and phenylpropanoid glycosides from the seed of Torreya grandis
CN108218933B (en) New phenolic glycoside compound in Guannan Tiancaojiao stem and preparation method and application thereof
CN111410645B (en) Lignans compound and preparation and application thereof

Legal Events

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