CN113816866B - Organic acid compound and preparation method and application thereof - Google Patents

Organic acid compound and preparation method and application thereof Download PDF

Info

Publication number
CN113816866B
CN113816866B CN202010566402.3A CN202010566402A CN113816866B CN 113816866 B CN113816866 B CN 113816866B CN 202010566402 A CN202010566402 A CN 202010566402A CN 113816866 B CN113816866 B CN 113816866B
Authority
CN
China
Prior art keywords
organic acid
acid compound
preparation
ethanol
water
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
CN202010566402.3A
Other languages
Chinese (zh)
Other versions
CN113816866A (en
Inventor
张鹏
张晗
柏自松
李楠
雷曦
姜苗苗
常艳旭
王晓明
吴红华
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.)
Tianjin University of Traditional Chinese Medicine
Original Assignee
Tianjin University of Traditional Chinese Medicine
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 Tianjin University of Traditional Chinese Medicine filed Critical Tianjin University of Traditional Chinese Medicine
Priority to CN202010566402.3A priority Critical patent/CN113816866B/en
Publication of CN113816866A publication Critical patent/CN113816866A/en
Application granted granted Critical
Publication of CN113816866B publication Critical patent/CN113816866B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C229/00Compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C229/02Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton
    • C07C229/04Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated
    • C07C229/22Compounds containing amino and carboxyl groups bound to the same carbon skeleton having amino and carboxyl groups bound to acyclic carbon atoms of the same carbon skeleton the carbon skeleton being acyclic and saturated the carbon skeleton being further substituted by oxygen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C227/00Preparation of compounds containing amino and carboxyl groups bound to the same carbon skeleton
    • C07C227/38Separation; Purification; Stabilisation; Use of additives
    • C07C227/40Separation; Purification
    • 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
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Abstract

The invention provides an organic acid compound, a preparation method and application thereof, wherein the organic acid compound is (2R) -2-amino-2-hydroxymethyl-3- [ (4-hydroxy-3-methoxybenzoyl) -O- ] -propionic acid, and has an inhibition effect on macrophage foaming, so that the occurrence and development of atherosclerosis are inhibited.

Description

Organic acid compound and preparation method and application thereof
Technical Field
The invention relates to the technical field of traditional Chinese medicines, in particular to an organic acid compound and a preparation method and application thereof.
Background
The pericarpium Trichosanthis is dried mature pericarp of fructus Trichosanthis (Trichosanthes kirilowii Maxim) or fructus Trichosanthis (Trichosanthes rosthornii Harms) of Cucurbitaceae (Cucurbitaceae). The earliest recorded in Shennong Ben Cao Jing (Shennong's herbal meridian), mainly distributed in Shandong and Henan, the Shandong Changqing and Feicheng are the areas of the road and the land. It is sweet, slightly bitter and cold in nature, and has the effects of clearing heat and resolving phlegm, relieving chest stuffiness and resolving masses, and relaxing bowel. The ancient books are often used for treating cough, chest distress, hypochondriac pain and other diseases caused by lung heat; modern clinic is often used for treating various cardiovascular diseases, such as coronary heart disease, hyperlipidemia, angina pectoris, etc. Modern pharmacological researches have proved that the pericarpium Trichosanthis has various pharmacological activities such as lipid metabolism regulation, atherosclerosis resistance, vascular endothelial protection, platelet aggregation resistance, anoxia resistance, inflammation resistance, phlegm elimination and bacteria inhibition. The chemical components of the snakegourd peel comprise flavone, saponin, alkaloid, amino acid, polysaccharide and the like. The snakegourd peel flavonoid has the function of protecting vascular endothelial injury, the snakegourd peel total saponins have a certain antibacterial function, the snakegourd peel total amino acids have a good phlegm eliminating function, and the research of the snakegourd peel oligosaccharides has the angiotensin converting enzyme inhibiting function. However, the researches on the chemical components and the biological activity of the organic acids in the snakegourd peel are less, and the researches show that the organic acid compound has the physiological activities of anti-inflammatory, antibacterial, anticancer, antioxidation and the like.
Disclosure of Invention
The invention aims to provide an organic acid compound.
Another technical problem to be solved by the present invention is to provide a method for preparing the above organic acid compound.
Another technical problem to be solved by the present invention is to provide an application of the above organic acid compound.
The technical scheme adopted by the invention is as follows:
an organic acid compound is (2R) -2-amino-2-hydroxymethyl-3- [ (4-hydroxy-3-methoxybenzoyl) -O- ] -propionic acid, and has a structure shown in a structural formula (I):
Figure BDA0002547985900000021
preferably, the organic acid compound has hydrogen spectrum data [ ] 1 H-NMR(DMSO-d 6 ,600MHz):δ H 7.60(1H,dd,J=7.8,1.8Hz,H-6'),7.48(1H,d,J=1.8Hz,H-2'),6.94(1H,d,J=7.8Hz,H-5'),δ H 4.42(1H,d,J=11.4Hz,H-3a),4.37(1H,d,J=11.4Hz,H-3b),3.74(1H,d,J=11.4Hz,H-4a),3.70(1H,d,J=11.4Hz,H-4b),3.80(1H,S,H-8’)。
Preferably, the organic acid compound and the carbon spectrum data are [ (] 13 C-NMR(DMSO-d 6 ,150MHz):δ C 119.8(C-1'),113.3(C-2'),147.4(C-3'),152.3(C-4'),115.2(C-5'),124.3(C-6'),165.5(C-7'),55.8(C-8'),63.5(C-1);δ C 65.4 (C-3), 62.3 (C-4) are two methylene carbon signals; delta C 169.0 (C-1) is a carboxyl carbon signal.
Preferably, the organic acid compound is white powder and is easily dissolved in methanol.
The preparation method of the organic acid compound comprises the following specific steps:
(1) Soaking dried pericarpium Trichosanthis in ethanol at room temperature, filtering, and concentrating under reduced pressure to obtain extract;
(2) The extract is dispersed by D101 macroporous resin to obtain a water layer, a 30% ethanol layer, a 70% ethanol layer and a 95% ethanol layer;
(3) The 30% ethanol layer is eluted by an ODS chromatographic column with methanol-water to obtain three fractions F1, F2 and F3, F1 is eluted by a liquid phase methanol-water 9:91 to obtain three fractions F1-1, F1-2 and F1-3, and F1-3 is purified by a liquid phase methanol-water 7:93 to obtain the novel compound.
The organic acid compound has an inhibitory effect on macrophage foaming.
The application of the organic acid compound in preparing atherosclerosis inhibiting medicine.
The beneficial effects of the invention are as follows:
the organic acid compound is (2R) -2-amino-2-hydroxymethyl-3- [ (4-hydroxy-3-methoxybenzoyl) -O- ] -propionic acid, has an inhibiting effect on macrophage foaming, and has a preventing and treating effect on cardiovascular and cerebrovascular diseases such as coronary heart disease with atherosclerosis as pathological characteristics.
Drawings
FIG. 1 is a HMBC correlation diagram of compound TP 1;
FIG. 2 is a comparison of TP1 experimental ECD spectra with TP1 enantiomer calculated ECD spectra;
FIG. 3 is the effect of compound TP1 on cell viability;
FIG. 4 is the effect of compound TP1 on macrophage foaming;
FIG. 5 shows lipid content of compound TP 1.
Fig. 6 is a graph showing the inhibitory effect of aortic atherosclerosis in rats, wherein the three graphs from left to right correspond to a blank control group, a model group and an administration group respectively.
Detailed Description
To further illustrate the invention, the following examples are provided in connection with:
example 1 preparation of Compounds
Experimental instrument and reagent
Fourier transform Nuclear magnetic resonance spectrometer (type AVIII; bruker, switzerland); mass spectrometer (Waters map G2 mass; waters, USA); automatic polarimeter (Autopol type III; rudolph Research, USA); analytical high performance liquid chromatography (Agilent 1260; agilent technologies Co., ltd., U.S.); preparation high performance liquid chromatograph (Agilent 1260; agilent technologies Co., ltd., U.S.); one ten thousandth balance (AL 204; METTER TOLEDOO Instrument Shanghai Co., ltd.); one ten million balance (XP 6; METTER TOLEDOPO Instrument Shanghai Co., ltd.); rotary evaporator (RE-52A; shanghai Asia Biochemical instruments Co.); temperature-regulating electrothermal sleeve (ZDHW; beijing Zhongxing Wei industry instruments Co., ltd.); dark box ultraviolet analyzer (ZF-20D; shanghai Gu Cun electro-optical instruments factory); thin layer heater (TH-II type; shanghai family hucho technology Co., ltd.); the display agent comprises: 2%
FeCl 3 -K 3 [Fe(CN) 6 ]An ethanol solution; bismuth potassium iodide.
Specific preparation method
The dried snakegourd peel (30 kg) is added with 130L of 70% ethanol to be soaked for three times at room temperature, each time for one week, filtered, decompressed and concentrated to obtain extract, and D101 macroporous resin is dispersed with proper amount of water to obtain a water layer, a 30% ethanol layer, a 70% ethanol layer and a 95% ethanol layer. Subjecting 30% ethanol layer to ODS chromatography column, eluting with methanol-water (0:100-100:0) to obtain three fractions (F1, F)2, f 3). F1 is eluted by preparing liquid phase methanol-water (9:91) to obtain three fractions (F1-1, F1-2, F1-3); f1-3 is purified by preparing liquid phase methanol-water (7:93) to obtain new compound TP1 (t) R =19min,16mg)。
Structural identification of TP1
TP1 white powder (methanol), readily soluble in methanol. Dark spots at UV 254nm by silica gel thin layer chromatography, 2% FeCl 3 -K 3 [Fe(CN) 6 ]Positive color development of ethanol shows that the compound is an organic acid compound; the bismuth potassium iodide shows positive color development, which indicates that the compound is a nitrogen-containing compound. HR-ESI-MS gives an excimer ion peak [ M+H ] at M/z 286.0932] + (molecular formula C 12 H 16 NO 7 Calculated as 286.0927), combined with 1 H NMR 13 C NMR to confirm that the compound is of formula C 12 H 15 NO 7 . Comparing the experimental ECD spectrum of compound TP1 with the calculated ECD spectrum of the TP1 enantiomer, as shown in FIG. 2, (2R) -TP1 calculated spectrum is characterized at 285 and 214nm, qualitatively consistent with the experimental spectrum, and incorporates the optical rotation value [ alpha ]]2 D0= -2.00 (c 0.40, meOH) and literature (Yajima T, aizawa Y, nishida, et al preparation of optically active-aminobutanoic acid via ptical resolution by replacing crystallization. Bioscience, biotechnology, and biochemistry.2007,71 (5): 1338-1341)
Figure BDA0002547985900000051
Figure BDA0002547985900000062
(c 1.00, methanol) symbols are identical, so the 2-position is defined as the R configuration.
1 H-NMR(DMSO-d 6 600 MHz) hydrogen spectral data delta H 7.60(1H,dd,J=7.8,1.8Hz,H-6'),7.48(1H,d,J=1.8Hz,H-2'),6.94(1H,d,J=7.8Hz,H-5'),δ H 4.42(1H,d,J=11.4Hz,H-3a),4.37(1H,d,J=11.4Hz,H-3b),3.74(1H,d,J=11.4Hz,H-4a),3.70(1H,d,J=11.4Hz,H-4b),3.80(1H,S,H-8’)。
13 C-NMR(DMSO-d 6 150 MHz) carbon spectrum data:δ C 119.8(C-1'),113.3(C-2'),147.4(C-3'),152.3(C-4'),115.2(C-5'),124.3(C-6'),165.5(C-7'),55.8(C-8'),63.5(C-1);δ C 65.4 (C-3), 62.3 (C-4) are two methylene carbon signals; delta C 169.0 (C-1) is a carboxyl carbon signal. Specific nuclear magnetic spectrum data are shown in table 1.
HMBC Spectrum methine Signal delta H 7.48(H-2'),δ H 7.60 (H-6') and delta C 165.5 (C-7') correlation; methyl hydrogen signal delta H 3.80 (H-8') and delta C 147.4 (C-3') correlation; methylene hydrogen signal delta H 4.42 (H-3 a) and 4.37 (H-3 b) and delta C 165.5 (C-7') and 169.0 (C-1); methylene hydrogen signal delta H 3.74 (H-4 a), 3.70 (H-4 b) and delta C 65.4 (C-8) and 169.0 (C-1). As shown in FIG. 1, the structure of the compound TP1 was determined as (2R) -2-amino-2-hydroxymethyl-3- [ (4-hydroxy-3-methoxybenzoyl) -O-]Propionic acid.
TABLE 1 Compound TP1 1 H NMR and 13 C NMR data
Figure BDA0002547985900000061
Figure BDA0002547985900000071
a measured in DMSO-d 6
Example 2 macrophage foaming assay
And (3) cells: RAW264.7 (China academy of sciences of Shanghai life sciences cell resource center)
Experimental apparatus and reagent
Carbon dioxide incubator (IL-161HI; STIK); desk-top refrigerated centrifuges (D-375200 Osterode; thermo); inverted fluorescence microscope (Nikon Ti-u; nikon); microscope (CKX 31SF; OLYMPUS); biosafety cabinet (HR 40-IIA 2; haier); a microplate reader (SPARK; tecan); cell scraping (Corning); 96 well plates (Corning); DMEM high sugar medium (Gibco); foetal Bovine Ser μm (Gibco); diabody (Hyclone); PBS (bi yun tian); DMSO (Solarbio); CCK8 ( Bao Bio); oil red O dye powder (Sigma); isopropyl alcohol (conrad); simvastatin (aletin); OX-LDL (Yiyuan).
Experimental method
1 drug toxicity screening
(1) Cell seed plates: scraping cells in logarithmic phase to obtain cell suspension, adding into 96-well plate, adding 100 μl of the suspension into each well to obtain density of 1×10 4 The edge wells were filled with sterile PBS, 5% CO 2 Culturing in an incubator at 37 ℃ for 24 hours.
(2) Administration: after 24 hours of incubation, quan Pei was aspirated, and the drug to be tested was added and incubated for 24 hours. Divided into blank groups, control group, and dosing group (TT 1 (0.1 μm,1 μm,10 μm)).
(3) After the cell culture and treatment, CCK8 reagent was added rapidly, and 10. Mu.L of CCK8 reagent was added to 100. Mu.L of medium per well.
(4) The plates were returned to the incubator and incubated at 37℃for 2h.
(5) OD was measured at 490 nm.
(6) Analysis of results: cell viability% = (dosing cell OD-blank OD)/(control cell OD-blank OD) ×100%
2 foam cell formation
(1) Cell seed plates: scraping cells in logarithmic phase to obtain cell suspension, adding into 96-well plate, adding 100 μl of the suspension into each well to obtain density of 1×10 4 The edge wells were filled with sterile PBS, 5% CO 2 Culturing in an incubator at 37 ℃ for 24 hours;
(2) Administration: after 24h incubation, quan Pei was aspirated, the drug to be tested was added and divided into blank (basal) model (OX-LDL 50 μm) and the drug-dosed (drug+OX-LDL) was incubated for 48h;
(3) Preparing an oil red O dye liquor: 500mg of oil red O is dissolved in 100mL of 70% ethanol to prepare 0.5%,
filtering the oil red O dye liquor with a 0.22 mu m filter membrane, sealing and keeping the oil red O dye liquor away from light at 4 ℃ for 24 hours to form saturated liquor, and then using the saturated liquor;
(4) Gently sucking the liquid medicine, and gently rinsing with PBS for 3 times;
(5) Adding 4% paraformaldehyde, and fixing at room temperature for 30min;
(6) Stock solution of diluent oil red O: diluting according to the volume ratio of the oil red O stock solution to the ultrapure water of 3:2, filtering with a 0.2 mu m filter membrane after dilution, and avoiding light in the preparation process;
(7) After the fixation, the fixation solution was discarded, rinsed gently 3 times with PBS, 100. Mu.L of 60% isopropyl alcohol solution was added to each well, and immersed for 15s;
(8) The isopropanol solution was aspirated and 80 μl of the diluted oil red O dye was added directly. Performing whole-process shading operation, and dyeing for 1h at 37 ℃;
(9) Decolorizing, absorbing oil red O dye, rinsing with PBS for 3 times, absorbing, adding 100 μl PBS solution, and directly observing with fluorescent inverted microscope;
(10) After photographing, PBS was removed from the wells, 100% isopropanol was added, the wells were immersed for 5min, the liquid was transferred to another 96-well plate, and OD was measured at 492 nm.
Experimental results
1 cytotoxicity
As shown in fig. 3, according to the cytotoxicity results of the compound TP1, the cell viability of TP1 at three concentrations was not significantly different from that of the control group at a medium concentration of 1 μm, and both low and high concentrations reduced the cell viability, and finally 1 μm was selected for subsequent activity test experiments.
Inhibition of macrophage foaming by Compounds 2
As shown in FIG. 4, intracellular lipids can be stained with oil red when the intracellular lipid droplets exceed 10 or more, and foam cell molding can be considered successful (ref: goswami R, michael M, sheta S, et al TRPV4calcium-permeable channel is a novel regulator of oxidized LDL-induced macrophage foam cell formation. Free radio Biol Med,2017,110:142-150;Tumurkhuu G,Jargalsaikhan D,Rebecca A,et al.Chlamydia pneumoniae Hijacks a Host Autoregulatory IL-1beta Loop to Drive Foam Cell Formation and Accelerate Atherosclerosis.Cell Metab,2018,28 (3): 432-448e 4). Compared with a normal group, the cell lipid drops in the model group are dense, and the number of the cell lipid drops is far more than 10, which indicates that the modeling is successful; both positive drug group (simvastatin), TP1 resulted in decreased intracellular lipid droplets relative to the model group. As shown in fig. 5, it is seen from the OD value results that both positive drug groups (simvastatin, P < 0.001) and TP1 (P < 0.05) significantly reduced intracellular lipid content relative to the model group. The results show that TP1 can inhibit foam cell formation and reduce lipid content.
Atherosclerosis is the primary cause of cardiovascular disease, and the formation of intimal macrophage foam is a hallmark of atherosclerosis. Can reduce the deposition of lipid in macrophage by inhibiting the foaming of macrophage, thereby inhibiting the occurrence and development of atherosclerosis, and achieving the effects of preventing and treating cardiovascular and cerebrovascular diseases. The mouse mononuclear macrophage RAW264.7 is easy to obtain and convenient to culture, is a common cell for researching macrophage foaming, adopts a RAW264.7 cell system to screen compounds for inhibiting macrophage foaming, and is a common technical means for developing medicaments for treating cardiovascular and cerebrovascular diseases such as atherosclerosis, coronary heart disease and the like.
Example 3 rat aortic atherosclerosis inhibition experiments
Rats with atherosclerosis caused by high fat feed and vitamin D3 were studied for the pharmacodynamic effects of the compounds described in example 1.
The experimental process comprises the following steps: the wistar rats were acclimatized for 3 days and were divided into three groups, (1) blank control group: normal diet was 8 weeks. (2) The model group is fed with high-fat feed for 8 weeks, and is injected intraperitoneally before molding to administer 60 ten thousand IU/kg of vitamin D3, and then 30 ten thousand IU/kg of vitamin D3 is supplemented every two weeks, and physiological saline is administered by lavage every day. (3) The administration group was fed with the high-fat diet for 8 weeks, and was given 60 ten thousand IU/kg of vitamin D3 by intraperitoneal injection before molding, followed by 30 ten thousand IU/kg of vitamin D3 every two weeks, and the compound described in example 1 (10 mg/kg/D) was administered by daily intragastric administration.
Animal material selection: after the rats fasted for 12 hours, the body weight, 10% chloral hydrate and 5mL/kg of abdominal cavity injection anesthesia were weighed, one section of the aorta was taken and fixed in 4% paraformaldehyde solution, and the rest was transferred to-80 ℃ for HE staining after being quenched by liquid nitrogen, and histopathology was detected.
Experimental results: as shown in fig. 6, the vascular endothelium of the blank group (C) is complete, the inner membrane, the middle membrane and the outer membrane are clearly demarcated, no lipid is deposited under the inner membrane, the middle membrane is a fusiform smooth muscle cell, the structure of the elastic fiber layer is clear and complete, the elastic fiber layer is circularly arranged, and the outer membrane is loose connective tissue. The model group (Mod) has the advantages that the aortic intima is thickened, obvious calcification is visible, the tube wall protrudes into the tube cavity, and the protruding part contains a large amount of foam cells. The tunica media is atrophic, smooth muscle cells proliferate, arrangement disorder, elastin is denatured, broken and disintegrated, and inflammatory cells infiltrate. The administration group improved the pathological changes to some extent compared to the model group, indicating that the compound of example 1 has an inhibitory effect on aortic lesions in atherosclerotic rats.
The above examples are only illustrative of the preferred embodiments of the present invention and are not intended to limit the scope of the present invention, and various modifications and improvements made by those skilled in the art to the technical solutions of the present invention should fall within the scope of protection defined by the claims of the present invention without departing from the spirit of the design of the present invention.

Claims (4)

1. An organic acid compound, characterized in that: is (2R) -2-amino-2-hydroxymethyl-3- [ (4-hydroxy-3-methoxybenzoyl) -O ]]Propionic acid having the structure of formula (I):
Figure QLYQS_1
2. the method for producing an organic acid compound according to claim 1, wherein: the method comprises the following specific steps:
(1) Soaking dried pericarpium Trichosanthis in ethanol at room temperature, filtering, and concentrating under reduced pressure to obtain extract;
(2) The extract is dispersed by D101 macroporous resin to obtain a water layer, a 30% ethanol layer, a 70% ethanol layer and a 95% ethanol layer;
(3) The 30% ethanol layer is eluted by an ODS chromatographic column with methanol-water to obtain three fractions F1, F2 and F3, F1 is eluted by a liquid phase methanol-water 9:91 to obtain three fractions F1-1, F1-2 and F1-3, and F1-3 is purified by a liquid phase methanol-water 7:93 to obtain the novel compound.
3. Use of the organic acid compound according to claim 1 for the preparation of a medicament having an inhibitory effect on macrophage foaming.
4. Use of the organic acid compound according to claim 1 for the preparation of an atherosclerosis-inhibiting drug.
CN202010566402.3A 2020-06-19 2020-06-19 Organic acid compound and preparation method and application thereof Active CN113816866B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010566402.3A CN113816866B (en) 2020-06-19 2020-06-19 Organic acid compound and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010566402.3A CN113816866B (en) 2020-06-19 2020-06-19 Organic acid compound and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN113816866A CN113816866A (en) 2021-12-21
CN113816866B true CN113816866B (en) 2023-07-07

Family

ID=78911973

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010566402.3A Active CN113816866B (en) 2020-06-19 2020-06-19 Organic acid compound and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN113816866B (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5431904A (en) * 1993-09-09 1995-07-11 The Gillette Company O-acyl serines as deodorants
CN105669480A (en) * 2016-03-02 2016-06-15 上海上药第一生化药业有限公司 Extracting method of gamma-aminobutyric acid

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5431904A (en) * 1993-09-09 1995-07-11 The Gillette Company O-acyl serines as deodorants
CN105669480A (en) * 2016-03-02 2016-06-15 上海上药第一生化药业有限公司 Extracting method of gamma-aminobutyric acid

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CHARLOTTES. H UNNECHE,等.Antioxidant Activity of a Combinatorial Library of Emulsifier -Antioxidant Bioconjugates.《J. Agric. Food Chem.》.2008,第56卷9258–9268. *
和焕香,等.瓜蒌化学成分和药理作用研究进展及质量标志物预测分析.《中草药》.2019,第50卷(第19期),4808-4820. *
巢志茂,等.五种瓜篓皮挥发性有机酸的分析.《中国中药杂志》.1992,第17卷(第11期),673-674. *

Also Published As

Publication number Publication date
CN113816866A (en) 2021-12-21

Similar Documents

Publication Publication Date Title
Ye et al. Immunomodulatory sesquiterpene glycosides from Dendrobium nobile
Lam et al. α-Glucosidase inhibitors from the seeds of Syagrus romanzoffiana
Li et al. Anti‐inflammatory effects of chemical components from Ginkgo biloba L. male flowers on lipopolysaccharide‐stimulated RAW264. 7 macrophages
Du et al. Pharmacologically active compounds in the Anoectochilus and Goodyera species
Song et al. Terpenoids and hexenes from the leaves of Crataegus pinnatifida
Vo et al. Protein tyrosine phosphatase 1B (PTP1B) inhibitory constituents from the aerial parts of Tradescantia spathacea Sw.
Li et al. Polyflavanostilbene A, a new flavanol-fused stilbene glycoside from Polygonum cuspidatum
Dudek et al. Hydroxycinnamoyl derivatives and secoiridoid glycoside derivatives from Syringa vulgaris flowers and their effects on the pro-inflammatory responses of human neutrophils
Zhang et al. Clerodane furanoditerpenoids from the stems of Tinospora sinensis
Kim et al. Screening ginseng saponins in progenitor cells identifies 20 (R)-ginsenoside Rh2 as an enhancer of skeletal and cardiac muscle regeneration
Cheng et al. Cardiac glycosides from the seeds of Thevetia peruviana and their pro-apoptotic activity toward cancer cells
CN113816866B (en) Organic acid compound and preparation method and application thereof
Xie et al. A new flavonol glycoside from the florets of Carthamus tinctorius L.
Ha et al. 12, 23-Dione dammarane triterpenes from Gynostemma longipes and their muscle cell proliferation activities via activation of the AMPK pathway
Chen et al. Antihyperlipidemic glycosides from the root bark of Lycium chinense
Pollo et al. A new polyacetylene glucoside from Vernonia scorpioides and its potential antihyperglycemic effect
Qiu et al. Sesquiterpenoids from the fruits of Alpinia oxyphylla Miq. and their neuroprotective effect
Alqahtani et al. Glycosylated phenols and an unprecedented diacid from the Saudi plant Cissus rotundifolia
Gaidi et al. Junceosides A− C, New Triterpene Saponins from Arenaria j uncea
Huang et al. Megastigmane glycosides from Streblus ilicifolius (S. Vidal) Corner and their anti-inflammatory activity
Kitajima et al. Monoterpenoids and their glycosides from the leaf of thyme
Ji et al. Chemical constituents from the stems of Securidaca inappendiculata Hassk
Li et al. Involucrasin C, Anti‐Inflammatory 2, 3‐Dihydro‐1H‐Indene Derivative from Chinese Dai Ethnic Medicine Shuteria involucrata
CN106565444A (en) Extraction method and application of phenanthrene compounds from overground part of Chinese yam
Chen et al. One new protocatechuic acid methyl ester and one enantiomeric pair of dihydroflavones isolated from Phymatopteris hastata

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