CN117624098A - Guaiane type sesquiterpene lactone derivative and preparation method and application thereof - Google Patents

Guaiane type sesquiterpene lactone derivative and preparation method and application thereof Download PDF

Info

Publication number
CN117624098A
CN117624098A CN202311579631.9A CN202311579631A CN117624098A CN 117624098 A CN117624098 A CN 117624098A CN 202311579631 A CN202311579631 A CN 202311579631A CN 117624098 A CN117624098 A CN 117624098A
Authority
CN
China
Prior art keywords
compound
preparation
alkyl
mice
inflammatory
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202311579631.9A
Other languages
Chinese (zh)
Inventor
胡立宏
刘健
吕祁
林伟江
王平
王娟
谢颖
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.)
Nanjing University of Chinese Medicine
Original Assignee
Nanjing University of 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 Nanjing University of Chinese Medicine filed Critical Nanjing University of Chinese Medicine
Priority to CN202311579631.9A priority Critical patent/CN117624098A/en
Publication of CN117624098A publication Critical patent/CN117624098A/en
Pending legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses a guaiane type sesquiterpene derivative, a preparation method and application thereof, wherein the guaiane type sesquiterpene derivative is shown in a general formula I, and is prepared from parthenolide as a raw material, has strong anti-inflammatory activity, and is applied to preparing medicines for treating NLRP3 inflammatory minibody Guan Yanzheng diseases.

Description

Guaiane type sesquiterpene lactone derivative and preparation method and application thereof
Technical Field
The invention relates to a guaiane type sesquiterpene lactone derivative, and also relates to a preparation method and application of the derivative.
Background
Inflammatory diseases are a group of chronic diseases affecting different organs and systems. Is characterized by common inflammatory mechanism and immune disorder, and chronic inflammation is the basic manifestation of the disease. Inflammatory diseases include more than 100 different diseases, such as sepsis, inflammatory bowel disease, nonalcoholic steatohepatitis, chronic kidney disease, and rheumatoid arthritis.
The above inflammatory diseases have different pathological parts and essentially different pathogenesis, and ulcerative colitis is an autoimmune disease, and the pathogenesis is complex, and is a disease caused by multiple factors including genetic factors, environmental factors, unbalanced intestinal flora, damaged epithelial barrier, abnormal intestinal immune response and the like. While some inflammatory diseases are mainly tissue injury caused by release of a large amount of inflammatory factors due to macrophage activation, and less involved in the T lymphocyte-mediated immune response process. Gout is mainly a disease caused by the migration of uric acid concentration in blood to joints due to abnormal uric acid excretion or purine metabolism, and the main disease part is at the joints. Gouty arthritis, which is more important, is mainly caused by endogenous metabolic abnormalities, while acute tubular injury, acute lung injury, acute liver injury, and sepsis are mostly caused by exogenous chemicals or microorganisms. The clinical treatment of the lesions of different parts and different degrees caused by different inflammatory diseases usually requires complex targeted treatment, at present, the clinical treatment medicaments for the inflammatory diseases mainly comprise glucocorticoid, immunosuppressant, biological preparation and the like, and the medicaments can cause various complications and immune dysfunction in gastrointestinal tracts after long-term administration, increase the susceptibility of organisms to infection and tumor, have high treatment cost and cause serious economic burden to patients.
The guaiane type sesquiterpene lactone is taken as an important natural product and has various pharmacological activities, and modern pharmacological researches find that the guaiane type sesquiterpene lactone michelia figo lactone (MCL) has various anti-inflammatory activities, however, the existing researches show that the anti-inflammatory activity in the MCL is weaker, and the main mechanism and the acting target protein of the anti-inflammatory activity are unknown. Another guaiane-type sesquiterpene lactone drug, arglabin (Arg), also has been shown to have versatile anti-inflammatory activity, however its solubility in water is only 7.9 μg/mL; the chemical stability in gastric juice environment is poor, the degradation rate in 8 hours reaches 50%, the oral bioavailability is only 5%, the clinical application of the compound is greatly limited by the defects in the aspects of drug formation, the proper anti-inflammatory indication is not yet confirmed, and a new guaiane type sesquiterpene lactone derivative with strong anti-inflammatory activity needs to be developed.
Disclosure of Invention
The invention aims to: the invention aims to provide a guaiane type sesquiterpene lactone derivative with strong anti-inflammatory activity, and also provides a preparation method and application of the derivative.
The technical scheme is as follows: the guaiane type sesquiterpene lactone derivative is shown as a general formula I:
wherein R is 1 、R 2 Together form a double bond; or R is 1 Is hydrogen or deuterium, R 2 Is thatWherein R is 3 And R is 4 Respectively C 1 ~C 3 Alkyl, or R 3 、R 4 And the N atom forms pyrrole;
R 5 is-OCONR 6 R 7 Wherein R is 6 Is hydrogen, C 1 ~C 6 An alkyl group; r is R 7 Is C 1 ~C 6 Alkyl, fluoro substituted C 1 ~C 3 Alkyl, cyclopropane, hydroxy-substituted C 1 ~C 6 Alkyl, methoxy substituted C 1 ~C 6 Alkyl, dimethylaminoethyl and morpholine substituted C 1 ~C 6 Alkyl, piperazine substituted C 1 ~C 6 Alkyl, phenyl ring substituted methyl; or R is 6 、R 7 And N atoms form a substituted 5-6 membered cyclic structure, wherein the cyclic structure is pyrrole, piperidine, piperazine or morpholine.
Preferably, the derivative is selected from the following compounds:
pharmaceutically acceptable salts of the above derivatives include hydrochloride, sulfate, phosphate, maleate, fumaric or citrate salts.
Preferably, the pharmaceutically acceptable salt is the hydrochloride or fumarate salt of the derivative:
the invention also discloses a pharmaceutical composition which comprises the guaiane type Bei-hemiterpene derivative shown in the general formula I or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, adjuvant or auxiliary material.
The preparation method of the derivative specifically comprises the following steps:
wherein R is 1 、R 2 、R 5 Is as defined in claim 1;
the active ester is obtained by condensation reaction of michelia lactone MCL, wherein the condensing agent is selected from N, N-carbonyl di (1, 2, 4-triazole), the catalyst is selected from pyridine and 4-dimethylaminopyridine, and the solvent is selected from dichloromethane. The activated ester is not purified and is combined with NHR 6 R 7 The reaction produces an intermediate III-A, wherein the base of the reaction is selected from N, N-diisopropylethylamine, and the solvent is selected from dichloromethane. Intermediate III-A, and NHR 3 R 4 The target compound III is obtained, the base of the reaction is selected from potassium carbonate, and the solvent is selected from dichloromethane.
The application of the derivatives in preparing medicaments for treating NLRP3 inflammatory small phase Guan Yanzheng diseases.
Preferably, the inflammatory disease is acute lung injury, acute tubular injury, renal interstitial fibrosis, acute liver injury or sepsis caused by bacterial and viral infections.
The principle of the invention: inflammatory minibodies are protein complexes that recognize PAMPs (pathogen-associated pattern molecules) or and DAMPs (injury-associated pattern molecules) in innate immune cells such as macrophages, monocytes and dendritic cells. Different types of inflammatory bodies such as NLRP1, NLRP3, NLRC4, pyrin, NLRP6, AIM2 and the like can mediate inflammatory responses, promote the release of cell inflammatory factors, transmit signals to an immune system, are the initiator of inflammation, and are the bridge of natural immunity and acquired immunity. Unlike other types of inflammatory corpuscles that recognize specific DAMPs or PAMPs, NLRP3 inflammatory corpuscles are able to recognize widely different sources of DAMPs and PAMPs, and thus, NLRP3 inflammatory corpuscles are the most deeply studied inflammatory corpuscles at present and are demonstrated to be involved in the occurrence and development of a variety of chronic inflammatory diseases. The guaiane type sesquiterpene lactone derivative shown in the general formula I has better inhibitory activity on NLRP3 inflammation small bodies, can be used for treating NLRP3 inflammation small body Guan Yanzheng diseases, and comprises the following components: inflammatory diseases such as acute lung injury, acute tubular injury, renal interstitial fibrosis, acute liver injury or sepsis caused by bacterial and viral infection.
The beneficial effects are that: compared with the prior art, the invention has the following remarkable advantages: (1) The guaiane type sesquiterpene lactone derivative has strong anti-inflammatory activity and is used for treating NLRP3 inflammatory small phase Guan Yanzheng diseases. After the guaiane type sesquiterpene lactone derivatives 36 (1 mg/kg) and 39 (1 mg/kg) are administrated by gastric lavage, the kidney injury of the sepsis mice can be obviously improved, and the liver injury and the lung injury of the sepsis mice can be obviously improved at the same dosage; (2) After the guaiane type sesquiterpene lactone derivatives 36 (1 mg/kg) and 39 (1 mg/kg) are administrated by stomach irrigation, the acute lung injury of mice can be obviously improved; (3) After the guaiane type sesquiterpene lactone derivatives 36 (1 mg/kg) and 39 (1 mg/kg) are administrated by stomach irrigation, the acute lung injury of mice can be obviously improved; (4) After the guaiane type sesquiterpene lactone derivative 36 (1 mg/kg) and 39 (1 mg/kg) are administrated by stomach, CCl can be inhibited to a certain extent 4 An induced liver fibrosis response; (5) After the guaiane type sesquiterpene lactone derivatives 36 (1 mg/kg) and 39 (1 mg/kg) are administrated by stomach irrigation, the kidney inflammatory reaction induced by folic acid or unilateral ureter ligation can be reduced to a certain extent, and the kidney fibrosis induced by folic acid is reduced; (6) The guaiane type sesquiterpene lactone derivative shown in the general formula I has the advantages of simple preparation method, mild condition and easy large-scale synthesis.
Detailed Description
The present invention will be described in detail with reference to specific examples.
Example 1
Preparation of parthenolide: pulverizing 5kg of dried root bark of Magnolia amurensis into coarse powder, soaking in 10 times of 95% ethanol for 12 hr, and reflux-extracting under heating twiceFiltering for 2 hr each time, mixing filtrates, concentrating under reduced pressure, and drying to obtain crude extract of Magnolia officinalis. Refining by silica gel column chromatography, gradient eluting with petroleum ether-ethyl acetate, collecting fraction rich in parthenolide and costunolide, mixing, concentrating, and recrystallizing to obtain parthenolide with a yield of 4.0% and a purity of 96.3%. 1 H NMR(500MHz,CDCl 3 ):δ6.31(d,J=2.9Hz,1H),5.62(d,J=3.4Hz,1H),5.20(d,J=11.8Hz,2H),3.85(t,J=8.6Hz,1H),2.78(d,J=8.9Hz,1H),2.45-2.32(m,2H),2.22-2.10(m,4H),1.70(s,3H),1.69-1.66(m,1H),1.29(s,3H),1.27-1.18(m,1H)。ESI-MS(m/z):[M+H] + =249.1(calcd:249.1)。
Example 2
Preparation of Compounds 1 to 39
The compounds 1 to 39 can be efficiently prepared by taking parthenolide as a raw material.
To a 250mL round bottom flask was added dichloromethane (100 mL), p-toluenesulfonic acid (250 mg,1.45 mmol) and parthenolide (10 g,40.32 mmol) in this order and stirred at room temperature until TLC detection was complete. The reaction solution was washed with water (20 mL. Times.3) and saturated brine (20 mL. Times.3) in this order, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and separated by silica gel column chromatography to give the intermediate michelia lactone MCL, yield: 90%. 1 H NMR(500MHz,CDCl 3 ):δ6.21(d,J=3.5Hz,1H),5.51(d,J=3.0Hz,1H),3.81(t,J=10.5Hz,1H),2.73(d,J=10.5Hz,1H),2.68-2.64(m,2H),2.42-2.37(m,1H),2.26-2.16(m,3H),2.11-2.08(m,1H),1.83-1.75(m,2H),1.69(s,3H),1.31(s,3H),1.27-1.25(m,1H)。ESI-MS(m/z):[M+Na] + =271.1(calcd:271.1)。
Preparation of Compound 1:
MCL (100 mg,0.40 mmol), methylene chloride (1.0 mL), N-carbonylbis (1,2, 4-triazole) (131 mg,0.80 mmol), pyridine (64 mg,0.8 mmol), 4-dimethylaminopyridine (4 mg,0.04 mmol), and stirring at room temperature until TLC detection was complete to give a crude intermediate solution. N, N-diisopropylethylamine (52 mg,0.4 mmol) and aqueous methylamine (47. Mu.L, 0.60mmol,40% aqueous solution) were added in this order, and stirred at room temperature until TLC detection was completed. The reaction solution was diluted with ethyl acetate (10 mL), washed successively with water (10 ml×3), a saturated citric acid solution (10 ml×3), a saturated copper sulfate solution if the subsequent product contains a basic group, a saturated sodium bicarbonate solution (10 ml×3), and a saturated brine (10 ml×3), dried over anhydrous sodium sulfate, concentrated under reduced pressure, and separated by silica gel column chromatography to give compound 1 in a yield of 72%. 1 H NMR(500MHz,CDCl 3 ):δ6.19(d,J=3.5Hz,1H),5.47(d,J=3.0Hz,1H),4.73(s,1H),3.80(t,J=10.5Hz,1H),3.11(d,J=9.5Hz,1H),2.74(d,J=4.5Hz,3H),2.70-2.65(m,1H),2.48-2.43(m,2H),2.28-2.20(m,3H),2.11-2.02(m,2H),1.72(s,3H),1.53(s,3H),1.40-1.32(m,1H)。ESI-MS(m/z):[M+H] + =306.16(calcd:306.16)。
Preparation of compound 2:
the preparation method of the compound 2 refers to the compound 1, and the yield is 72%. 1 H NMR(500MHz,CDCl 3 ):δ6.20(d,J=3.5Hz,1H),5.48(d,J=3.0Hz,1H),4.76(s,1H),3.80(t,J=10.5Hz,1H),3.21-3.11(m,3H),2.76-2.66(m,1H),2.48-2.43(m,2H),2.28-2.20(m,3H),2.12-2.06(m,2H),1.72(s,3H),1.53(s,3H),1.40-1.32(m,1H),1.14(t,J=7.0Hz,3H)。ESI-MS(m/z):[M+H] + =320.18(calcd:320.18)。
Preparation of compound 3:
the preparation method of the compound 3 refers to the compound 1, and the yield is 76%. 1 H NMR(500MHz,CDCl 3 ):δ6.20(d,J=3.5Hz,1H),5.47(d,J=3.0Hz,1H),4.80(s,1H),3.80(t,J=10.5Hz,1H),3.14-3.05(m,3H),2.70-2.66(m,1H),2.48-2.41(m,2H),2.27-2.21(m,3H),2.11-2.05(m,2H),1.72(s,3H),1.53(s,3H),1.55-1.48(m,2H),1.40-1.32(m,1H),0.92(t,J=7.5Hz,3H)。ESI-MS(m/z):[M+H] + =334.19(calcd:334.19)。
Preparation of Compound 4:
the preparation method of the compound 4 refers to the compound 1, and the yield is 74%. 1 H NMR(500MHz,CDCl 3 ):δ6.17(d,J=3.5Hz,1H),5.46(d,J=3.0Hz,1H),4.78(s,1H),3.79(t,J=10.5Hz,1H),3.17-3.10(m,3H),2.69-2.64(m,1H),2.46-2.41(m,2H),2.26-2.18(m,3H),2.10-2.04(m,2H),1.71(s,3H),1.51(s,3H),1.48-1.44(m,2H),1.37-1.31(m,3H),0.91(t,J=7.5Hz,3H)。ESI-MS(m/z):[M+H] + =348.21(calcd:348.21)。
Preparation of compound 5:
the preparation method of the compound 5 refers to the compound 1, and the yield is 41%. 1 H NMR(500MHz,CDCl 3 ):δ6.20(d,J=3.5Hz,1H),5.48(d,J=3.0Hz,1H),4.66(s,1H),3.83-3.74(m,2H),3.13(d,J=10.0Hz,1H),2.71-2.66(m,1H),2.48-2.44(m,2H),2.29-2.20(m,3H),2.13-2.05(m,2H),1.73(s,3H),1.53(s,3H),1.41-1.33(m,1H),1.16(t,J=7.0Hz,6H)。ESI-MS(m/z):[M+H] + =334.19(calcd:334.19)。
Preparation of Compound 6:
the preparation method of the compound 6 refers to the compound 1, and the yield is 33%. 1 H NMR(500MHz,CDCl 3 ):δ6.16(d,J=3.5Hz,1H),5.44(d,J=3.0Hz,1H),4.51(d,J=9.0Hz,1H),3.77(t,J=10.5Hz,1H),3.39-3.38(m,1H),3.19-3.17(m,1H),2.68-2.63(m,1H),2.45-2.34(m,2H),2.24-2.21(m,3H),2.17-2.06(m,2H),1.70(s,3H),1.54-1.47(m,2H),1.49(s,3H),1.41-1.30(m,3H),0.92-0.86(m,6H)。ESI-MS(m/z):[M+H] + =362.23(calcd:362.23)。
Preparation of compound 7:
the preparation method of the compound 7 refers to the compound 1, and the yield is 69%. 1 H NMR(500MHz,CDCl 3 )δ6.19(d,J=3.3Hz,1H),5.47(d,J=3.0Hz,1H),5.01(s,1H),3.80(t,J=10.1Hz,1H),3.09(s,1H),2.71-2.62(m,1H),2.56(s,1H),2.50-2.41(m,2H),2.32-2.16(m,3H),2.13-1.99(m,2H),1.72(s,3H),1.53(s,3H),1.39-1.32(m,1H),0.69(d,J=6.8Hz,2H),0.53(s,2H)。ESI-MS(m/z):[M+H] + =332.18(calcd:332.18)。
Preparation of Compound 8:
the preparation method of the compound 8 refers to the compound 1, and the yield is 71%. 1 H NMR(500MHz,CDCl 3 ):δ6.13(d,J=3.5Hz,1H),5.42(d,J=3.0Hz,1H),5.30(s,1H),4.66(s,1H),3.75(t,J=10.5Hz,1H),3.63-3.61(m,1H),3.24-3.20(m,1H),3.13-3.11(m,1H),2.63-2.59(m,1H),2.54-2.47(m,2H),2.40-2.28(m,2H),2.20-2.15(m,3H),2.06-1.98(m,2H),1.64(s,3H),1.42(s,3H),1.33-1.26(m,1H)。ESI-MS(m/z):[M+H] + =336.17(calcd:336.17)。
Preparation of Compound 9:
the preparation method of the compound 9 refers to the compound 1, and the yield is 62%. 1 H NMR(500MHz,CDCl 3 ):δ6.21(d,J=3.5Hz,1H),5.49(d,J=3.0Hz,1H),3.83(t,J=10.5Hz,1H),3.68(t,J=5.5Hz,2H),3.37-3.26(m,2H),3.19(d,J=10.5Hz,1H),2.71-2.67(m,1H),2.48-2.44(m,1H),2.40-2.37(m,1H),2.29-2.20(m,3H),2.16-2.10(m,2H),1.72(s,3H),1.71-1.68(m,2H),1.51(s,3H),1.40-1.35(m,1H)。ESI-MS(m/z):[M+H] + =350.19(calcd:350.19)。
Preparation of compound 10:
the preparation method of the compound 10 refers to the compound 1, and the yield is 59%. 1 H NMR(500MHz,CDCl 3 ):δ6.18(d,J=3.5Hz,1H),5.47(d,J=3.0Hz,1H),5.06(s,1H),4.49(s,1H),3.79(t,J=10.5Hz,1H),3.62(t,J=6.5Hz,2H),3.15-3.10(m,3H),2.68-2.64(m,1H),2.45-2.38(m,2H),2.25-2.22(m,3H),2.10-2.04(m,2H),1.70(s,3H),1.60-1.55(m,2H),1.53-1.50(m,2H),1.49(s,3H),1.43-1.33(m,3H)。ESI-MS(m/z):[M+H] + =377.24(calcd:377.24)。
Preparation of Compound 11:
the preparation method of the compound 11 refers to the compound 1, and the yield is 75%. 1 H NMR(500MHz,CDCl 3 ):δ6.20(d,J=3.5Hz,1H),5.47(d,J=3.0Hz,1H),5.51(s,1H),3.80(t,J=10.5Hz,1H),3.48-3.46(m,2H),3.37(s,3H),3.36-3.29(m,2H),3.14(d,J=10.0Hz,1H),2.71-2.66(m,1H),2.48-2.42(m,2H),2.27-2.20(m,3H),2.12-2.03(m,2H),1.73(s,3H),1.55(s,3H),1.40-1.32(m,1H)。ESI-MS(m/z):[M+H] + =350.19(calcd:350.19)。
Preparation of Compound 12:
the preparation method of the compound 12 refers to the compound 1, and the yield is 74%. 1 H NMR(500MHz,CDCl 3 )δ6.20(d,J=3.3Hz,1H),5.48(d,J=3.0Hz,1H),5.06(s,1H),3.80(t,J=10.1Hz,1H),3.46(t,J=5.8Hz,2H),3.35(s,3H),3.29-3.21(m,2H),3.14(d,J=10.1Hz,1H),2.71-2.66(m,1H),2.48-2.41(m,2H),2.27-2.20(m,3H),2.12-2.04(m,2H),1.80-1.76(m,2H),1.73(s,3H),1.53(s,3H),1.40-1.33(m,1H)。ESI-MS(m/z):[M+H] + =364.20(calcd:364.20)。
Preparation of Compound 13:
the preparation method of the compound 13 refers to the compound 1, and the yield is 62%. 1 HNMR(500MHz,CDCl 3 ):δ6.20(d,J=3.5Hz,1H),5.56(s,1H),5.49(d,J=3.0Hz,1H),3.82(t,J=10.5Hz,1H),3.62-3.58(m,4H),3.36-3.34(m,2H),3.12(d,J=10.5Hz,1H),2.98-2.87(m,1H),2.71-2.66(m,1H),2.48-2.43(m,2H),2.27-2.20(m,3H),2.13-1.97(m,3H),1.72(s,3H),1.52(s,3H),1.40-1.32(m,1H)。ESI-MS(m/z):[M+H] + =380.20(calcd:380.20)。
Preparation of compound 14:
the preparation method of the compound 14 refers to the compound 1, and the yield is 61%. 1 H NMR(500MHz,CDCl 3 ):δ6.20(d,J=3.5Hz,1H),5.48(d,J=3.0Hz,1H),5.22(s,1H),3.80(t,J=10.5Hz,1H),3.31-3.22(m,2H),3.16(d,J=10.0Hz,1H),2.70-2.67(m,1H),2.48-2.40(m,4H),2.34-2.20(m,3H),2.26(s,6H),2.11-2.04(m,2H),1.72(s,3H),1.55(s,3H),1.39-1.32(m,1H)。ESI-MS(m/z):[M+H] + =363.22(calcd:363.22)。
Preparation of compound 15:
the preparation method of the compound 15 refers to the compound 1, and the yield is 71%. 1 H NMR(500MHz,CDCl 3 ):δ6.18(d,J=3.5Hz,1H),5.46(d,J=3.0Hz,1H),5.18(s,1H),3.79(t,J=10.5Hz,1H),3.72-3.70(m,4H),3.25-3.23(m,2H),3.17(d,J=9.5Hz,1H),2.70-2.66(m,1H),2.48-2.37(m,8H),2.26-2.19(m,3H),2.12-2.07(m,2H),1.71(s,3H),1.53(s,3H),1.39-1.31(m,1H)。ESI-MS(m/z):[M+H] + =405.23(calcd:405.23)。
Preparation of Compound 16:
the preparation method of the compound 16 refers to the compound 1, and the yield is 69%. 1 H NMR(500MHz,CDCl 3 ):δ7.35-7.30(m,4H),7.28-7.25(m,1H),6.20(d,J=3.5Hz,1H),5.48(d,J=3.0Hz,1H),5.13(s,1H),4.43-4.39(m,1H),4.30-4.26(m,1H),3.81(t,J=10.5Hz,1H),3.18(d,J=9.5Hz,1H),2.68-2.65(m,1H),2.49-2.42(m,2H),2.28-2.21(m,3H),2.17-2.06(m,2H),1.73(s,3H),1.56(s,3H),1.40-1.32(m,1H)。ESI-MS(m/z):[M+H] + =382.19(calcd:382.19)。
Preparation of compound 17:
the preparation method of the compound 17 refers to the compound 1, and the yield is 70%. 1 H NMR(500MHz,CDCl 3 ):δ6.20(d,J=3.5Hz,1H),5.48(d,J=3.0Hz,1H),3.83(t,J=10.5Hz,1H),3.07(d,J=10.0Hz,1H),2.96(s,3H),2.89(s,3H),2.72-2.66(m,1H),2.54-2.44(m,2H),2.30-2.21(m,3H),2.14-2.09(m,1H),2.03-1.97(m,1H),1.73(s,3H),1.54(s,3H),1.42-1.34(m,1H)。ESI-MS(m/z):[M+H] + =320.18(calcd:320.18)。
Preparation of compound 18:
the preparation method of the compound 18 refers to the compound 1, and the yield is 69%. 1 H NMR(500MHz,CDCl 3 ):δ6.20(d,J=3.5Hz,1H),5.48(d,J=3.0Hz,1H),3.83(t,J=10.5Hz,1H),3.41-3.35(m,1H),3.30-3.22(m,3H),3.06(d,J=10.0Hz,1H),2.72-2.67(m,1H),2.57-2.53(m,1H),2.49-2.44(m,1H),2.30-2.22(m,3H),2.14-2.09(m,1H),2.04-1.97(m,1H),1.73(s,3H),1.55(s,3H),1.42-1.34(m,1H),1.14(s,6H)。ESI-MS(m/z):[M+H] + =348.21(calcd:348.21)。
Preparation of compound 19:
the preparation method of the compound 19 refers to the compound 1, and the yield is 71%. 1 H NMR(500MHz,CDCl 3 ):δ6.20(d,J=3.5Hz,1H),5.48(d,J=3.0Hz,1H),3.82(t,J=10.5Hz,1H),3.53-3.49(m,1H),3.37-3.30(m,3H),3.10(d,J=10.0Hz,1H),2.72-2.66(m,1H),2.53-2.44(m,2H),2.29-2.21(m,3H),2.13-2.08(m,1H),2.07-2.00(m,1H),1.88-1.83(m,4H),1.73(s,3H),1.56(s,3H),1.42-1.34(m,1H)。ESI-MS(m/z):[M+H] + =346.19(calcd:346.19)。
Preparation of compound 20:
the preparation method of the compound 20 refers to the compound 1, and the yield is 68%. 1 H NMR(500MHz,CDCl 3 ):δ6.20(d,J=3.5Hz,1H),5.47(d,J=3.0Hz,1H),3.82(t,J=10.5Hz,1H),3.51-3.42(m,4H),3.10(d,J=10.0Hz,1H),2.72-2.68(m,1H),2.52-2.44(m,2H),2.30-2.22(m,3H),2.14-2.01(m,2H),1.73(s,3H),1.60-1.56(m,6H),1.53(s,3H),1.41-1.35(m,1H)。ESI-MS(m/z):[M+H] + =360.21(calcd:360.21)。
Preparation of compound 21:
the preparation method of the compound 21 refers to the compound 1, and the yield is 67%. 1 H NMR(500MHz,CDCl 3 ):δ6.19(d,J=3.5Hz,1H),5.47(d,J=3.0Hz,1H),4.24-4.12(m,2H),3.81(t,J=10.5Hz,1H),3.08(s,1H),2.83-2.67(m,3H),2.51-2.43(m,2H),2.29-2.20(m,3H),2.12-2.09(m,1H),2.05-1.99(m,1H),1.72(s,3H),1.65-1.60(m,2H),1.55-1.48(m,1H),1.52(s,3H),1.41-1.33(m,1H),1.19-1.04(m,2H),0.94(d,J=6.5Hz,3H)。ESI-MS(m/z):[M+H] + =374.23(calcd:374.23)。
Preparation of compound 22:
/>
the preparation method of the compound 22 refers to the compound 1, and the yield is 52%. 1 H NMR(500MHz,CDCl 3 ):δ6.20(d,J=3.5Hz,1H),5.48(d,J=3.0Hz,1H),3.83(t,J=10.5Hz,1H),3.57-3.40(m,4H),3.08(d,J=10.0Hz,1H),2.71-2.67(m,1H),2.53-2.46(m,2H),2.43-2.38(m,3H),2.32(s,3H),2.30-2.22(m,4H),2.13-2.10(m,1H),2.04-2.02(m,1H),1.73(s,3H),1.53(s,3H),1.42-1.37(m,1H)。ESI-MS(m/z):[M+H] + =375.22(calcd:375.22)。
Preparation of compound 23:
the preparation method of the compound 23 refers to the compound 1, and the yield is 55%. 1 H NMR(500MHz,CDCl 3 ):δ6.20(d,J=3.5Hz,1H),5.48(d,J=3.0Hz,1H),3.82(t,J=10.5Hz,1H),3.71-3.41(m,4H),3.08(d,J=10.0Hz,1H),2.71-2.66(m,1H),2.52-2.43(m,8H),2.29-2.22(m,3H),2.13-2.10(m,1H),2.07-2.00(m,1H),1.72(s,3H),1.52(s,3H),1.42-1.34(m,1H),1.11(t,J=7.0Hz,3H)。ESI-MS(m/z):[M+H] + =389.24(calcd:389.24)。
Preparation of compound 24:
the preparation method of the compound 24 refers to the compound 1, and the yield is 71%. 1 H NMR(500MHz,CDCl 3 ):δ6.18(d,J=3.5Hz,1H),5.47(d,J=3.0Hz,1H),3.81(t,J=10.5Hz,1H),3.68-3.62(m,5H),3.49-3.41(m,3H),3.06(d,J=10.0Hz,1H),2.70-2.64(m,1H),2.51-2.44(m,2H),2.28-2.21(m,3H),2.13-2.00(m,2H),1.72(s,3H),1.52(s,3H),1.40-1.33(m,1H)。ESI-MS(m/z):[M+H] + =362.19(calcd:362.19)。
Preparation of compound 25:
the preparation method of the compound 25 refers to the compound 1, and the yield is 75%. 1 HNMR(500MHz,CDCl 3 )δ6.17(d,J=3.3Hz,1H),5.46-5.45(m,1H),4.82(s,1H),3.78(t,J=10.1Hz,1H),3.13(d,J=9.9Hz,1H),3.02-2.97(m,1H),2.90-2.85(m,1H),2.71-2.62(m,1H),2.48-2.35(m,2H),2.26-2.17(m,3H),2.12-2.02(m,2H),1.77-1.71(m,1H),1.70(s,3H),1.51(s,3H),1.38-1.30(m,1H),0.89(d,J=6.7Hz,6H)。ESI-MS(m/z):[M+H] + =348.21(calcd:348.21)。
Preparation of compound 26:
the preparation of compound 26 was carried out in 78% yield with reference to compound 1. 1 H NMR(500MHz,CDCl 3 )δ6.20(d,J=3.3Hz,1H),5.83(t,J=56.1Hz,1H),5.48(d,J=2.9Hz,1H),5.16(s,1H),3.80(t,J=10.1Hz,1H),3.62-3.38(m,2H),3.11(d,J=10.5Hz,1H),2.73-2.62(m,1H),2.48-2.41(m,2H),2.27-2.20(m,3H),2.12-1.99(m,2H),1.72(s,3H),1.53(s,3H),1.38-1.31(m,1H)。ESI-MS(m/z):[M+H] + =356.16(calcd:356.16)。
Preparation of compound 27:
the preparation method of the compound 27 refers to the compound 1, and the yield is 67%. 1 H NMR(500MHz,CDCl 3 )δ6.17(d,J=3.2Hz,1H),5.46(d,J=3.0Hz,1H),3.80(t,J=10.0Hz,1H),3.37-3.12(m,2H),3.10-3.00(m,1H),2.88(d,J=28.6Hz,3H),2.72-2.62(m,1H),2.54-2.41(m,2H),2.29-2.18(m,3H),2.11-2.07(m,1H),2.02-1.93(m,1H),1.71(s,3H),1.63-1.53(m,2H),1.51(s,3H),1.40-1.32(m,1H),0.88(t,J=7.4Hz,3H)。ESI-MS(m/z):[M+H] + =348.21(calcd:348.21)。
Preparation of Compound 28:
the preparation method of the compound 28 refers to the compound 1, and the yield is 68%. 1 H NMR(500MHz,CDCl 3 )δ6.19(d,J=3.3Hz,1H),5.46(d,J=3.0Hz,1H),3.81(t,J=10.1Hz,1H),3.44-3.00(m,5H),2.69-2.66(m,1H),2.59-2.42(m,2H),2.29-2.20(m,3H),2.12-2.08(m,1H),2.03-1.97(m,1H),1.72(s,3H),1.61-1.54(m,2H),1.53(s,3H),1.42-1.33(m,1H),1.19-1.07(m,3H),0.89-0.87(m,3H)。ESI-MS(m/z):[M+H] + =362.23(calcd:362.23)。
Preparation of compound 29:
the preparation method of the compound 29 refers to the compound 1, and the yield is 55%. 1 H NMR(500MHz,CDCl 3 )δ6.20(d,J=3.3Hz,1H),5.72(s,1H),5.48(d,J=3.1Hz,1H),3.81(t,J=10.1Hz,1H),3.73(t,J=4.7Hz,4H),3.30-3.12(m,3H),2.72-2.65(m,1H),2.47-2.39(m,8H),2.29-2.20(m,3H),2.14-2.05(m,2H),1.72(s,3H),1.69-1.66(m,2H),1.53(s,3H),1.40-1.34(m,1H)。ESI-MS(m/z):[M+H] + =419.25(calcd:419.25)。
Preparation of compound 30:
the preparation method of the compound 30 refers to the compound 1, and the yield is 47%. 1 H NMR(500MHz,CDCl 3 )δ6.17(d,J=3.3Hz,1H),5.83(s,1H),5.46(d,J=3.0Hz,1H),3.78(t,J=10.1Hz,1H),3.26-3.11(m,3H),2.65(t,J=9.3Hz,1H),2.56-2.36(m,11H),2.26(s,7H),2.12-2.04(m,2H),1.71(s,3H),1.69-1.63(m,2H),1.51(s,3H),1.39-1.32(m,1H)。ESI-MS(m/z):[M+H] + =432.28(calcd:432.28)。
Preparation of Compound 31:
the preparation method of the compound 31 refers to the compound 1, and the yield is 62%. 1 H NMR(500MHz,CDCl 3 )δ6.18(d,J=3.2Hz,1H),5.46(d,J=3.0Hz,1H),4.28-4.14(m,2H),3.80(t,J=10.1Hz,1H),3.75-3.68(m,4H),3.06(s,1H),2.92-2.63(m,4H),2.49-2.42(m,2H),2.37-2.18(m,5H),2.14-2.07(m,2H),2.01(d,J=10.2Hz,1H),1.84-1.82(m,2H),1.71(s,3H),1.49(s,3H),1.46-1.32(m,3H)。ESI-MS(m/z):[M+H] + =445.26(calcd:445.26)。
Preparation of compound 32:
the preparation method of the compound 32 refers to the compound 1, and the yield is 44%. 1 H NMR(500MHz,CDCl 3 )δ6.17(d,J=3.3Hz,1H),5.45(d,J=3.0Hz,1H),5.22(s,1H),3.78(t,J=10.1Hz,1H),3.31-3.19(m,2H),3.15(d,J=9.5Hz,1H),2.83(s,1H),2.67(t,J=9.4Hz,1H),2.48-2.37(m,11H),2.29(s,3H),2.25-2.18(m,3H),2.11-2.04(m,2H),1.70(s,3H),1.52(s,3H),1.38-1.31(m,1H)。ESI-MS(m/z):[M+H] + =418.26(calcd:418.26)。
Preparation of compound 33:
in a 5mL reaction flask, compound 20 (100 mg,0.28 mmol) was dissolved in 1mL of methylene chloride, tetrahydropyrrole (197mg, 2.80 mmol) was added, and after stirring for 3 hours, ethyl acetate (10 mL) was diluted, the mixture was washed with water (10 mL. Times.3), saturated sodium bicarbonate solution (10 mL. Times.3) and saturated brine (10 mL. Times.3) in this order, dried over anhydrous sodium sulfate, concentrated under reduced pressure, and separated by silica gel column chromatography to give compound 33 in 66% yield. 1 H NMR(500MHz,CDCl 3 )δ3.80(t,J=10.2Hz,1H),3.57-3.30(m,4H),3.03-2.97(m,1H),2.91-2.81(m,2H),2.59-2.53(m,2H),2.52-2.37(m,5H),2.28-2.14(m,4H),2.14-1.97(m,2H),1.79-1.69(m,8H),1.61-1.53(m,5H),1.52(s,4H)。ESI-MS(m/z):[M+H] + =431.28(calcd:431.28)。
Preparation of compound 34:
starting from compound 20 and dimethylamine hydrochloride, the preparation method is the same as that of compound 33, and the yield of compound 34 is: 85%. 1 H NMR(500MHz,CDCl 3 )δ3.82(t,J=10.1Hz,1H),3.58-3.30(m,4H),3.05-2.96(m,1H),2.74(dd,J=12.9,4.9Hz,1H),2.60(dd,J=12.9,6.6Hz,1H),2.53-2.35(m,3H),2.26(s,6H),2.28-2.14(m,4H),2.09-1.97(m,2H),1.74-1.65(m,6H),1.59-1.54(m,6H),1.39-1.27(m,1H)。ESI-MS(m/z):[M+H] + =405.27(calcd:405.27)。
Preparation of hydrochloride 35 of Compound 15
Compound 15 is taken as a starting material, dissolved in dichloromethane (2 mL), stirred for 2 hours at room temperature, then hydrochloric acid solution is added dropwise until the pH value is 4-5, filtration is carried out, the obtained solid is washed by dichloromethane, and the obtained white solid is the hydrochloride of compound 15 (compound 35) with the yield of 86%. 1 H NMR(500MHz,Methanol-d 4 )δ6.15(d,J=6.9Hz,1H),5.62(d,J=14.3Hz,1H),4.14-4.08(m,2H),4.05-3.89(m,1H),3.88-3.76(m,2H),3.65-3.42(m,4H),3.28-3.16(m,3H),2.89-2.71(m,1H),2.57-2.46(m,1H),2.42-2.28(m,4H),2.25-2.15(m,1H),2.09-2.00(m,3H),1.76(s,3H),1.57(s,3H),1.44-1.37(m,1H)。ESI-MS(m/z):[M+H] + =405.23(calcd:405.23)。
Preparation of fumarate salt 36 of Compound 15
By fumaric acidInstead of hydrochloric acid, fumarate compound 36 of 15 was prepared with reference to the preparation method of compound 15 hydrochloride. The yield thereof was found to be 71%. 1 H NMR(500MHz,Methanol-d 4 )δ6.74(s,2H),6.14(d,J=10.2Hz,1H),5.61(d,J=10.5Hz,1H),4.01-3.78(m,5H),3.39-3.36(m,2H),3.19(d,J=11.5Hz,1H),3.11-3.05(m,2H),2.99-2.85(m,4H),2.82-2.72(m,1H),2.52-2.47(m,1H),2.43-2.24(m,4H),2.22-2.13(m,1H),2.06-2.00(m,1H),1.75(s,3H),1.56(s,3H),1.43-1.35(m,1H)。ESI-MS(m/z):[M+H] + =405.23(calcd:405.23)。
Preparation of hydrochloride 37 of Compound 33
Compound 37 can be prepared in 91% yield by the preparation method of hydrochloride of reference compound 15 using compound 33 as starting material. 1 H NMR(500MHz,Methanol-d 4 )δ4.13(t,J=10.0Hz,1H),3.81-3.72(m,2H),3.68(s,1H),3.64-3.56(m,1H),3.51-3.39(m,3H),3.26-3.19(m,2H),3.09(d,J=8.7Hz,2H),2.54-2.40(m,2H),2.34-2.06(m,8H),2.04-1.94(m,2H),1.75(s,3H),1.66-1.60(m,2H),1.55-1.51(m,7H),1.49-1.43(m,1H),1.42-1.26(m,1H)。ESI-MS(m/z):[M+H] + =431.28(calcd:431.28)。
Preparation of fumarate salt 38 of Compound 33
Fumaric acid was used instead of hydrochloric acid, and the fumarate compound 38 of 33 was prepared according to the preparation method of compound 15 hydrochloride, with a yield of 75%. 1 H NMR(500MHz,Methanol-d 4 )δ6.72(s,2H),4.17-4.07(m,2H),3.60-3.40(m,9H),3.11-3.01(m,2H),2.52-2.41(m,2H),2.35-2.21(m,3H),2.18-2.12(m,5H),2.02-1.93(m,2H),1.74(s,3H),1.66-1.61(m,2H),1.55-1.52(m,7H),1.49-1.39(m,1H)。ESI-MS(m/z):[M+H] + =431.28(calcd:431.28)。
Preparation of fumarate salt 39 of Compound 34
Fumaric acid was used instead of hydrochloric acid, and the fumarate compound 39 of 34 was prepared according to the preparation method of compound 15 hydrochloride, in 71% yield. 1 H NMR(500MHz,Methanol-d 4 )δ6.71(s,2H),4.14-4.09(m,1H),3.46-3.40(m,3H),3.31-3.27(m,1H),3.12-3.01(m,2H),2.90(s,6H),2.52-2.42(m,2H),2.34-2.22(m,3H),2.14(q,J=10.9,10.1Hz,1H),2.01-1.93(m,2H),1.75(s,3H),1.66-1.61(m,2H),1.55-1.51(m,7H),1.48-1.40(m,1H),1.40-1.29(m,2H)。ESI-MS(m/z):[M+H] + =405.27(calcd:405.27)。
Example 3
Test of Activity of Compounds 1 to 39 for inhibition of NLRP3 inflammatory body activation:
NLRP3 is an important pattern recognition receptor, and can form NLRP3 inflammatory corpuscles through linker protein ASC and pro-caspase-1, and can mediate caspase-1 activation after NLRP3 inflammatory corpuscle activation, thereby promoting maturation and secretion of IL-1 beta. To determine whether the prepared guaiane-type sesquiterpene lactone derivatives 1-39 can inhibit the activation of NLRP3 inflammatory bodies, we induced the activation of NLRP3 inflammatory bodies with LPS and ATP, and observed the effect of compounds 1-39 on IL-1β levels caused by the activation of NLRP3 inflammatory bodies.
Experimental reagent: lipopolysaccharide (LPS) and adenosine triphosphate (Adenosine triphosphate, ATP), sigma; recombinant mouse macrophage colony-stimulating factor (recombinant mouse macrophage colony stimulating factor, rmM-CSF), pentTech; RPMI 1640 medium, DMEM medium and fetal bovine serum (Fetal bovine serum, FBS).
Experimental animals: c57BL/6 mice, female, 6-8 weeks old, body weight 18-20g, supplied by the Jiang Ningou Qinglong mountain animal farm in Nanjing, production license number: SCXK 2017-0001.
The experimental method comprises the following steps:
(1) Isolation and culture of mouse Bone Marrow Derived Macrophages (BMDMs): taking a C57BL/6 mouse, killing the mouse by adopting a cervical dislocation method, soaking the mouse in 75% alcohol for 5-10min, taking down two rear legs of the mouse by scissors, removing meat and remaining bones, washing the bones of the mouse three times by PBS, sucking serum-free RPMI 1640 culture medium into a 1mL syringe, clamping bones by forceps, cutting off the bones from two ends by scissors, blowing the bone marrow into a 15mL centrifuge tube by the syringe, and repeating for a plurality of times until the bone marrow is completely flushed out, and turning the bones into white by red. Subsequently, the mixture was centrifuged at 1500rpm for 5min, the supernatant was discarded, resuspended in 1mL of erythrocyte lysate, and repeatedly blown, followed by standing for 7min to lyse erythrocytes. 1500rpm, centrifuged for 5min and the supernatant was discarded, resuspended in RPMI 1640 medium containing 100ng/mLrmM-CSF and transferred to 6 well plates for cultivation. After 6-7 days, the cell state can be observed, and the cell is long-fusiform, which indicates that the cell state is good and can be used for subsequent experiments.
(2) And (3) establishing an NLRP3 inflammation small body activation model: preparing a DMEM medium containing 1% FBS and 100ng/mL LPS, taking BMDMs in the 6-hole culture plate, and adding the prepared medium for pre-stimulation for 3 hours. Subsequently, after incubation for 1h with Argatroban (1, 3,10,30,60,120 nM), ATP (5 mM) was added to stimulate for 1h. Supernatants were collected into 1.5mL EP tubes for subsequent detection of IL-1β levels.
TABLE 1 inhibition of NLRP3 inflammatory body activation by derivatives 1-39 and prodrugs thereof
Derivatives and their use as inhibitors of viral infection IC 50 (nM) Derivatives and their use as inhibitors of viral infection IC 50 (nM)
1 ++ 2 +
3 + 4 +
5 + 6 ++
7 ++ 8 +
9 + 10 +
11 + 12 ++
13 ++ 14 +
15 +++ 16 +
17 + 18 +
19 ++ 20 +++
21 ++ 22 +
23 ++ 24 +
25 ++ 26 +
27 + 28 +
29 ++ 30 ++
31 + 32 ++
33 + 34 +
35 + 36 ++
37 ++ 38 ++
39 ++
Argatroban (Argatroban) +++ Michelia lactone +++
Note that: + represents 80nM<IC 50 <100nM, ++represents 40nM<IC 50 <80nM of the total of two different molecules, ++ + representative IC 50 <40nM
The effect of compounds 1-39 on IL-1β expression levels was examined in BMDMs using LPS and ATP-induced NLRP3 inflammatory body activation models. As shown in Table 1, both compounds 15 and 20 have excellent inhibitory activity against IL-1β, and hydrochloride 36 of compound 15 also has excellent inhibitory activity against IL-1β, and prodrugs 37, 38 and 39 of compound 20 all have excellent inhibitory activity against IL-1β.
Example 4
Test of Compound 36 and 39 for Activity against mouse sepsis
Experimental reagent: lipopolysaccharide (LPS), sigma company; sodium carboxymethylcellulose (CMC-Na), a chemical plant of the ridge, guangdong, first city; RNA isolater total RNA extraction reagent, hiscript Q RT Supermix for qPCR, nanjing Norvigator Biotech Co., ltd; aceQ qPCR SYBR Green Master Mix the next holy biotechnology share, inc. on the Shanghai.
Experimental animals: c57BL/6 mice, male, 6-8 weeks old, body weight 18-20g, supplied by Shanghai Laek animal cultivation center, production license number: SCXK (Su) 2019-002. The animals eat and drink water freely, and feed standard pellet feed at room temperature of 22+ -2deg.C and humidity of 45+ -10%. The experiment was performed 7 days after the adaptation to the environment.
The experimental method comprises the following steps:
(1) Establishment of a mouse sepsis model and group dosing: mice were randomized into the normal, model, 36 (0.1, 1 mg/kg), 39 (0.1, 1 mg/kg) and dexamethasone (Dex, 5 mg/kg) groups, 6 each. Compound 36 (0.1, 1 mg/kg), 39 (0.1, 1 mg/kg) and Dex (5 mg/kg) were administered by gavage 5 days prior to molding. Normal and model mice were given equal amounts of vehicle 0.5% cmc-Na by gavage. Mice sepsis model was constructed 1h intraperitoneally injected with LPS (10 mg/kg) after the last day of dosing.
(2) Sample collection: 6h after injecting LPS into the abdominal cavity, killing the mouse by removing cervical vertebra, opening the abdominal cavity, taking out the lung, liver and kidney, washing twice by precooling PBS, shearing the lung tissue, left lobe tissue and kidney tissue of the mouse, fixing 4% paraformaldehyde for 24-48h, and freezing the rest tissue at-70 ℃ for later use.
Q-PCR analysis:
(1) Extraction of Total RNA
Accurately weighing 20mg of kidney, lung and liver tissues, rinsing with precooled PBS solution, shearing into small blocks by ophthalmic surgery, placing into a glass homogenizer, adding 1mL of Trizol reagent for grinding, and preparing tissue homogenate. Subsequently, the homogenate was transferred to a 1.5mL EP tube without RNase and lysed on ice for 10min. 200. Mu.L of chloroform was added thereto, and the mixture was vigorously shaken, allowed to stand on ice for 3 to 5 minutes, and centrifuged at 12000rpm at 4℃for 15 minutes. Carefully aspirate the supernatant into a fresh RNase-free 1.5mL EP tube, add an equal volume of pre-chilled isopropyl alcohol, leave it to stand in an ice bath for 30min, centrifuge at 12000rpm at 4℃for 10min, discard the supernatant. Adding 75% ethanol-DEPC solution to wash the precipitate, and reversely airing on the flat paper to obtain the total RNA. Subsequently, 10. Mu.L of DEPC treated water was added for dissolution. 1. Mu.L of the total RNA solution was diluted 100 times and OD values at 260nm and 280nm were measured using a full wavelength microplate reader. When the ratio of OD260/OD280 is between 1.8 and 2.0, the extracted RNA has higher purity, and can be used for subsequent experiments.
(2) Synthesis of cDNA: the following experiments were performed using 20 μl of reverse transcription reaction system according to the method described in the kit instructions:
the reagents were gently mixed by pipetting under the following reaction conditions:
the cDNA obtained after synthesis is stored at-80℃or immediately used for the subsequent Q-PCR reaction.
(3) Designing a primer: the sequence was designed based on the nucleotide sequence of mice in GenBank, and the primers were synthesized as follows:
(4) And (3) PCR amplification: the following experiments were performed using 20 μl of the reaction system according to the method described in the kit instructions:
after the reagent is added into a micro-reaction tube, a sealing film is sealed, and the micro-reaction tube is placed into a Q-PCR instrument, parameters are set according to the following reaction conditions, and an amplification experiment is carried out. The reaction conditions were as follows:
recording thresh of each groupThe old cycle (Ct) value is 2 -ΔΔCt The method was carried out for calculation analysis (the expression level of the control gene Gapdh was set to 1 to correct the expression level of the target gene).
All data are expressed in Means ± s.e.m. using analysis of variance, analysis of significance of differences, and one-way ANOVA and Dunnett's test were further used to compare differences between groups for those with significant differences between other groups. P values less than 0.05 are considered significant differences.
Experimental results:
effects on the sepsis mouse pneumonic response: as shown in table 2, the expression level of inflammatory factors (Tnf, il6, il 1) mRNA in lung tissue of sepsis mice was significantly increased compared to control group, indicating that significant inflammatory reaction occurred in lung of sepsis mice. mRNA expression of Tnf, IL6, IL1 was significantly down-regulated in mouse lung tissue after compound 36 (1 mg/kg) and 39 (1 mg/kg) were dried. Compounds 36 and 39 are available to inhibit the expression of inflammatory factors and ameliorate lung injury in sepsis mice.
TABLE 2 expression of pneumonic factor mRNAf
Group of Tnf Il1 Il6
Normal 1.00±0.07 1.00±0.04 1.00±0.01
LPS 10.92±1.57 ## 19.01±2.40 ## 20.34±1.96 ##
36(0.1mg/kg) 9.89±0.69 15.59±1.58 19.34±1.55 *
36(1mg/kg) 9.06±0.98 8.04±1.01 11.90±0.34 *
39(0.1mg/kg) 10.06±1.01 16.13±1.22 19.87±2.27
39(1mg/kg) 9.68±1.23 8.21±0.97 ** 12.14±0.58 **
Dex(5mg/kg) 8.99±0.76 7.20±0.36 * 9.88±0.80 **
Note that: # P <0.01 compared to Normal group; p <0.05, P <0.01 compared to LPS group.
Effects on the nephritis response in sepsis mice: as shown in table 3, the expression level of inflammatory factors (Tnf, il6, il 1) mRNA in kidney tissue of sepsis mice was significantly increased compared to control group, indicating that the sepsis mice kidney developed a significant inflammatory reaction. mRNA expression of mouse kidney tissues Tnf, IL6, IL1 was significantly down-regulated following the drying of compounds 36 (1 mg/kg) and 39 (1 mg/kg). Compounds 36 and 39 are available to inhibit the expression of inflammatory factors and ameliorate kidney injury in sepsis mice.
TABLE 3 nephritis factor mRNA expression
Group of Tnf Il1 Il6
Normal 1.00±0.04 1.00±0.04 1.00±0.03
LPS 7.30±0.51 ## 7.99±0.85 ## 8.80±1.67 ##
36(0.1mg/kg) 7.20±0.85 7.16±0.52 8.05±1.10
36(1mg/kg) 6.91±0.95 3.20±0.25 ** 3.26±0.55 *
39(0.1mg/kg) 7.29±0.25 6.56±0.88 6.05±0.19
39(1mg/kg) 7.01±0.65 3.89±0.89 ** 3.55±0.15 *
Dex(5mg/kg) 6.62±0.68 6.05±1.03 5.76±0.72 *
Note that: comparison with Normal group ## P<0.01; comparison with LPS group * P<0.05, ** P<0.01.
Effect on hepatitis response in sepsis mice: as shown in table 4, the expression level of inflammatory factors (Tnf, il6, il 1) mRNA in liver tissue of sepsis mice was significantly increased compared to control group, indicating that significant inflammatory reaction occurred in liver of sepsis mice. mRNA expression of Tnf, IL6, IL1 was significantly down-regulated in mouse liver tissues after compound 36 (1 mg/kg) and 39 (1 mg/kg) were dried. Compounds 36 and 39 are available to inhibit the expression of inflammatory factors and ameliorate liver injury in sepsis mice.
TABLE 4 expression of pneumonic factor mRNA
Group of Tnf Il1 Il6
Normal 1.00±0.01 1.00±0.11 1.00±0.08
LPS 8.92±1.93 ## 11.52±1.19 ## 11.34±0.75 ##
36(0.1mg/kg) 8.75±2.25 11.24±2.98 8.86±3.11
36(1mg/kg) 7.09±0.99 4.82±0.58 ** 5.20±0.87 **
39(0.1mg/kg) 7.63±1.97 10.46±1.55 9.13±0.80
39(1mg/kg) 7.10±1.09 4.36±0.94 ** 4.97±0.77 **
Dex(5mg/kg) 7.04±0.77 7.54±0.70 * 4.27±1.33 *
Note that: # P <0.01 compared to Normal group; p <0.01 compared to LPS group.
Example 5
Test of Activity of Compounds 36, 39 against acute Lung injury
Experimental reagent: LPS was purchased from sigma company of the United states; dexamethasone (dexamethasone, dex) is available from sigma company in the united states; ELISA kits were purchased from Daidae Biolimited.
Experimental animals: male C57BL/6 mice, 6-8 weeks old, body weight 18-20g, purchased from Jiangsu Jiuyaokang technologies Co., ltd. [ production license number: SCXK (threo) 2018-0008 ]. Animals were fed and drunk freely and kept in an environment with a room temperature of 22.+ -. 2 ℃ and a humidity of 45.+ -. 10%. After 7 days of adaptive feeding, the strain was used for the subsequent experiments.
The experimental method comprises the following steps:
(1) Establishment of mice acute lung injury model and group administration: intragastric administration 36 and 39 treats acute lung injury: male C57BL/6J mice, 6-8 weeks old, 18-20g weight, were randomly divided into normal, model, 36, 39 (0.1, 1 mg/kg) and positive control Dex (5 mg/kg) groups of 8 mice each according to body weight size. Besides normal group lavage PBS injection, other mice were intraperitoneally injected with LPS (7.5 mg/kg) to construct a mouse acute lung injury model. Compound 36 (0.1, 1 mg/kg), 39 (0.1, 1 mg/kg) and the positive drug Dex (5 mg/kg) were administered by gavage three days prior to molding, and LPS (7.5 mg/kg) was injected intraperitoneally half an hour after the third day of administration. Lung tissue was harvested 12 hours after molding and the effect of 36 and 39 on inflammatory factor expression in lung tissue of acute lung injury mice was examined by ELISA (table 5).
(2) Sample collection: after the last administration for 12 hours, the mice are killed by cervical vertebra removal, the abdominal cavity is opened, the lungs are taken out, the PBS is precooled and washed twice, the lung tissues of the mice are sheared, 4% paraformaldehyde is fixed for 24-48 hours, and the rest tissues are frozen at the temperature of-70 ℃ for later use.
(3) ELISA analysis:
extraction of total protein: accurately weighing 20mg of lung tissue, rinsing with precooled PBS solution, cutting into small pieces by ophthalmic surgery, placing into a glass homogenizer, adding 180 μl PBS (containing 1mM PMSF), 60HZ, homogenizing 180s, centrifuging at 3000rpm/min at 4deg.C for 20min, collecting 1 μl supernatant, measuring protein concentration by Nanodrop, and leveling, and can be used for subsequent experiments.
Data statistics: all data are expressed in means±s.e.m. and statistical differences between groups were measured using one-way ANONA and t-test in SPSS software. P values less than 0.05 are considered significant differences.
Experimental results
Effects of intragastric administration 36 and 39 on acute lung injury in mice:
as shown in Table 5, the expression level of inflammatory factor (IL-1. Beta., IL-6) protein in the lung tissue of the mice was significantly increased compared to the control group, indicating that a significant inflammatory reaction occurred in the lungs of the mice. IL-1β, IL-6 protein expression was significantly down-regulated in mouse lung tissue after compound 36 (0.1, 1 mg/kg) and 39 (0.1, 1 mg/kg) were dried. Compounds 36 and 39 are available to inhibit the expression of inflammatory factors and ameliorate acute lung injury in mice.
TABLE 5 expression of inflammatory factors (IL-1 beta and IL-6) in mouse serum after intraperitoneal injection treatment
Note that: * p <0.05, < p <0.01, < p <0.001 (compared to LPS group)
Example 6
Test of Activity of Compounds 36 and 39 against liver injury
Experimental reagent: olive oil (Olive oil), beijing kospe green Olive oil development center; analytically pure carbon tetrachloride (CCl) 4 ) Shanzhong chemical plant; sodium carboxymethylcellulose (CMC-Na), a chemical plant of the ridge, guangdong, first city; RNAisolater total RNAextraction reagent, hiscript Q RT Supermix for qPCR, nanjing Norvigator Biotech Co., ltd; aceQ qPCR SYBR Green Master Mix the next holy biotechnology share, inc. on Shanghai; glutamic-pyruvic transaminase (AST/GOT) colorimetric test kit, glutamic-pyruvic transaminase (ALT/GPT) colorimetric test kit, WU-Han Yi Rui Biotech Co.
Experimental animals: c57BL/6 mice, male, 6-8 weeks old, body weight 18-20g, supplied by Shanghai Laek animal cultivation center, production license number: SCXK (Su) 2019-002. The animals eat and drink water freely, and feed standard pellet feed at room temperature of 22+ -2deg.C and humidity of 45+ -10%. The experiment was performed 7 days after the adaptation to the environment.
Experimental method
(1) Establishment of mice acute liver injury model and group administration: mice were randomly divided into model groups, compound 36 (1 mg/kg) and 39 (0.1, 1 mg/kg), 6 each. The remaining mice were injected with 0.5mL CCl except for the normal group with 10mL olive oil/kg 4 +9.5mL olive oil/kg body weight intraperitoneal injection, i.e. CCl 4 The ratio of the olive oil to the olive oil is 1:19. Compounds 36 (0.1, 1 mg/kg) and 39 (0.1, 1 mg/kg) were administered parenterally for 3 consecutive days from day 1 of the initial molding. Normal and model mice were given equal amounts of vehicle 0.5% cmc-Na by gavage.
(2) Sample collection: after the last administration for 1h, the fundus vein Cong Caixie is stood for 2h at room temperature, centrifuged at 3000rpm for 20min, and serum is sucked and split charging is carried out, and the fundus vein is frozen at-70 ℃ for standby. After blood collection, the mice are killed by cervical vertebra removal, the abdominal cavity is opened, the liver is taken out, precooled PBS is used for washing twice, the left leaf tissue of the liver of the mice is sheared, 4% blood polyformaldehyde is fixed for 24 to 48 hours, and the rest tissue is frozen at the temperature of minus 70 ℃ for standby.
(3) Serum AST and ALT assays: 10. Mu.L of each sample was taken for the experiment. The levels of mouse serum AST and ALT were measured according to the operating manual, absorbance values were measured at 510nm wavelength by the microplate reader, and the sample concentration (U/L) was calculated according to the standard curve.
Q-PCR analysis: the method for extracting total RNA from tissues, cDNA synthesis and PCR amplification were the same as in example 4. Primer addition information is as follows:
data statistics: all data are expressed in Means ± s.e.m. using analysis of variance, analysis of significance of differences, and one-way ANOVA and Dunnett's test were further used to compare differences between groups for those with significant differences between other groups. P values less than 0.05 are considered significant differences.
Experimental results
Effects on serum ALT and AST levels
Serum ALT and AST level changes are important indicators for liver injury, in order to determine CCl 4 Whether acute liver injury in mice was indeed induced, the study first examined serum ALT and AST levels in mice from the model group. The results show that CCl compared to the normal olive oil group 4 Serum ALT and AST levels were significantly elevated in mice after treatment. The method can be used for successfully establishing the acute liver injury model of the mice. In addition, administration of compound 36 (1 mg/kg), and 39 (1 mg/kg) significantly reduced ALT and AST levels in mouse serum.
TABLE 6 ALT and AST levels
Group of ALT AST
Normal 9.47±3.21 41.42±4.62
LPS 171.46±3.42 ## 430.64±54.91 ##
36(0.1mg/kg) 174.83±22.05 425.20±178.43
36(1mg/kg) 105.14±5.39 ** 239.08±39.59 *
39(0.1mg/kg) 134.21±34.25 387.71±29.17
39(1mg/kg) 99.46±2.69** 216.14±65.51*
Silybin(200mg/kg) 121.58±15.45* 301.45±38.44*
Note that: comparison with Normal group ## P<0.01; with CCl 4 Group comparison * P<0.05, ** P<0.01.
Effects on hepatic tissue fibrosis-related factor mRNA expression: the effect of compounds 36 and 39 on mouse liver pro-inflammatory factors was evaluated by detecting changes in mRNA expression levels of the liver tissue fibrosis related factors (Acta 2, col1a1, tgfb 1) in each group of mice. As shown in Table 7, compared with Normal group, CCl was injected into the abdominal cavity 4 mRNA levels of Acta2, col1a1, tgfb1 were significantly increased in liver tissue after 72h, and expression was significantly reduced in 36 (1 mg/kg) and 39 (1 mg/kg) treated groups. Compounds 36 and 39 are available which inhibit cci to some extent 4 Induced liver fibrosis response.
TABLE 7 mRNA expression of hepatic fibrosis related factors
Group of Acta2 Col1a1 Tgfb1
Normal 1.00±0.00 1.00±0.00 1.00±0.00
LPS 4.35±0.71 ## 2.75±0.25 ## 1.66±0.12 ##
36(0.1mg/kg) 3.78±0.90 2.33±0.87 1.43±0.71
36(1mg/kg) 2.41±0.18 * 1.42±0.29 ** 1.12±0.19 **
39(0.1mg/kg) 2.97±0.26 1.83±0.53 1.08±0.29 **
39(1mg/kg) 2.15±0.44 * 1.33±0.31 ** 0.97±0.13 **
Silybin(200mg/kg) 2.91±0.48 * 1.84±0.17 * 1.11±0.08 *
Note that: comparison with Normal group ## P<0.01; with CCl 4 Group comparison ** P<0.01.
Example 9
Effects of Compounds 36, 39 on Folic acid and unilateral ureteral ligation (UUO) induced kidney injury in mice
Experimental reagent: folic Acid (FA), MP Biomedicals company; sodium carboxymethylcellulose (CMC-Na), a chemical plant of the ridge, guangdong, first city; RNAisolater total RNAextraction reagent, hiscript Q RT Supermix for qPCR, nanjing Norvigator Biotech Co., ltd; aceQ qPCR SYBR Green Master Mix the next holy biotechnology share, inc. on Shanghai; urea colorimetric test cartridges, creatinine colorimetric test cartridges, martial arts, inc.
Experimental animals: c57BL/6 mice, male, 6-8 weeks old, body weight 18-20g, supplied by Shanghai Laek animal cultivation center, production license number: SCXK (Su) 2019-002. The animals eat and drink water freely, and feed standard pellet feed at room temperature of 22+ -2deg.C and humidity of 45+ -10%. The experiment was performed 7 days after the adaptation to the environment.
Experimental method
(1) Establishment of mouse kidney injury model and group administration:
acute kidney injury: mice were randomly assigned to the normal, model, compound 36 (0.1, 1 mg/kg) and 39 (0.1, 1 mg/kg) groups of 6 animals each. Except for normal group mice, the other mice were intraperitoneally injected with a solution of 0.9% NaHCO 3 Folic acid (250 mg/kg) of the solution was used to model acute kidney injury in mice. The gavage was administered 36 (0.1, 1 mg/kg) and 39 (0.1, 1 mg/kg) for 3 consecutive days before 1h of modeling. Normal and model mice were given equal amounts of vehicle 0.5% cmc-Na by gavage.
Chronic kidney injury: mice were randomly divided into 7 groups, 1 of which was arbitrarily selected as a normal group, and the remaining 6 groups were model groups, compound 36 (0.1, 1 mg/kg) and 39 (0.1, 1 mg/kg), respectively, of 6 groups each. Except for the normal group of mice which are subjected to false operation (namely, only left ureter is exposed and other operations are not performed), the other mice are subjected to unilateral ureter ligation to establish a chronic kidney injury model of the mice, and the chronic kidney injury model is specifically as follows: sterilizing abdominal skin with iodophor, sterilizing with alcohol, and separating abdominal skin layer by layer. After the left ureter was fully exposed and isolated, double ligation was performed at the proximal bladder end of the ureter using digestible sutures, followed by suturing. Modeling 36 (0.1, 1 mg/kg) and 39 (0.1, 1 mg/kg) were given intragastrically for 14 consecutive days. Normal and model mice were given equal amounts of vehicle 0.5% cmc-Na by gavage.
Sample collection: after the last administration for 1h, the fundus vein Cong Caixie is stood for 2h at room temperature, centrifuged at 3000rpm for 20min, and serum is sucked and split charging is carried out, and the fundus vein is frozen at-70 ℃ for standby. After blood collection, the mice are killed by cervical vertebra removal, the abdominal cavity is opened, the kidneys are taken out, perirenal fat and envelopes are stripped, after the mice are washed twice by precooled PBS, the mice are fixed for 24-48 hours by 4% paraformaldehyde, after paraffin embedding, HE staining is carried out, and the residual tissues are frozen at the temperature of-70 ℃ for later use.
Measurement of blood creatinine (Scr) and urea (BUN) levels: for creatinine and urea level detection 10 μl of serum was taken per sample. And (3) sequentially adding samples according to the instruction of the kit, incubating at 37 ℃, measuring absorbance light values by using an enzyme-labeled instrument, and calculating the content level of each group of detection indexes according to a standard curve.
Q-PCR analysis: animal tissue total RNA extraction, cDNA synthesis, and PCR amplification as in the previous examples. Primer addition information is as follows:
experimental results
Effects on serum creatinine and urea levels: as shown in tables 8 and 9, serum levels of urea and creatinine were significantly elevated in mice 48h after folic acid administration compared to Normal mice, whereas urea and creatinine levels were significantly lower in the treatment groups of compounds 36 (1 mg/kg) and 39 (1 mg/kg) than in the model group, with differences statistically significant (P < 0.05). It is known that compounds 36 and 39 have good protective effects on folic acid or unilateral ureteral ligation induced acute and chronic kidney injury.
TABLE 8 serum creatinine and Urea levels-acute model
Group of Scr BUN
Normal 44.90±5.63 7.07±0.76
FA 124.49±12.85 ## 33.18±2.54 ##
36(0.1mg/kg) 118.25±4.99** 31.98±2.09*
36(1mg/kg) 73.98±6.03* 22.59±1.09**
39(0.1mg/kg) 120.56±3.89** 32.65±2.02*
39(1mg/kg) 79.69±3.98* 23.55±1.23**
Note that: comparison with Normal group ## P<0.01; comparison with the FA group * P<0.05, ** P<0.01.
TABLE 9 serum creatinine and Urea levels 8-chronic model
Group of Scr BUN
Normal 37.07±7.66 7.60±0.98
UUO 201.57±33.25 ## 20.36±1.44 ##
36(0.1mg/kg) 180.87±8.53 ** 18.91±1.59 *
36(1mg/kg) 72.59±7.89 * 13.52±0.88 **
39(0.1mg/kg) 189.26±7.89 ** 19.56±2.34 *
39(1mg/kg) 85.33±8.03 * 14.69±0.98 **
Note that: comparison with Normal group ## P<0.01; comparison with UUO group * P<0.05, ** P<0.01.
Effects on kidney injury marker mRNA expression: the effect of compounds 36 and 39 on mouse kidney function was assessed by detecting changes in the level of kidney injury marker (Ngal, havcr 1) mRNA expression in kidney tissue of each group of mice. As shown in tables 10 and 11, the levels of Ngal and Havcr1 mRNA were significantly increased in the kidney tissue of mice after 48h of folic acid administration or unilateral ureteral ligation, and significantly decreased in the treated groups of compounds 36 (1 mg/kg) and 39 (1 mg/kg) compared to the Normal group. Compounds 36 and 39 are available to inhibit the elevation of kidney injury markers caused by folic acid or unilateral ureteral ligation.
TABLE 10 expression of kidney injury marker mRNA-acute model
Note that: comparison with Normal group ## P<0.01; comparison with the FA group * P<0.05, ** P<0.01.
TABLE 11 mRNA expression of kidney injury markers-chronic model
Group of Havcr1 Ngal
Normal 1.00±0.04 1.00±0.05
UUO 14.49±2.23 ## 45.13±7.89 ##
36(0.1mg/kg) 12.99±0.77 * 38.67±1.23
36(1mg/kg) 8.77±1.23 21.89±0.48
39(0.1mg/kg) 13.06±0.78 ** 38.98±1.85 *
39(1mg/kg) 8.88±0.79 * 22.92±0.28 **
Note that: # P <0.01 compared to Normal group; p <0.05, P <0.01 compared to UUO group.
Effects on kidney tissue inflammatory factor mRNA expression: the effect of compounds 36 and 39 on mouse kidney pro-inflammatory factors was evaluated by detecting changes in the expression levels of inflammatory factors (Tnf, il6, il 1) mRNA in the kidney tissue of each group of mice. As shown in tables 12 and 13, the levels of Tnf, il6, il1 mRNA were significantly increased in kidney tissue after 48h of folic acid administration or unilateral ureteral ligation, and the expression was significantly reduced in the treatment groups of compounds 36 (1 mg/kg) and 39 (1 mg/kg) compared to the Normal group. Compounds 36 and 39 may be available to reduce to some extent the renal inflammatory response induced by folic acid or unilateral ureteral ligation.
Table 12, nephritis factor mRNA expression-acute model
Group of Tnf Il1 Il6
Normal 1.00±0.06 1.00±0.05 1.00±0.02
FA 3.10±0.56 ## 2.94±0.35 ## 8.87±0.91 ##
36(0.1mg/kg) 2.99±0.88 * 2.76±1.11 8.15±1.02 *
36(1mg/kg) 2.77±1.12 1.89±0.59 4.87±0.98 *
39(0.1mg/kg) 3.06±0.68 ** 2.79±1.09 * 8.19±1.58
39(1mg/kg) 2.79±0.89 * 1.92±0.38 ** 5.04±0.68 **
Note that: # P <0.01 compared to Normal group; p <0.05, P <0.01 compared to FA group.
TABLE 13 expression of nephritis factor mRNA-chronic model
Note that: # P <0.01 compared to Normal group; p <0.05, P <0.01 compared to UUO group.
TABLE 14 mRNA expression of kidney fibrosis related factors-acute model
Group of α-Sma Col1a1 Tgfb1
Normal 1.00±0.06 1.00±0.04 1.00±0.03
Folic acid 2.16±0.26 ## 1.95±0.11 ## 2.11±0.29 ##
36(0.1mg/kg) 1.89±0.69 1.69±1.58 2.01±1.55 *
36(1mg/kg) 1.60±0.98 1.42±1.01 1.56±0.34 *
39(0.1mg/kg) 1.96±1.01 1.73±1.22 2.17±2.27
39(1mg/kg) 1.68±1.23 1.51±0.97 ** 1.84±0.58 **
Note that: # P <0.01 compared to Normal group; p <0.05, P <0.01 compared to folic acid group.
TABLE 15 mRNA expression of kidney fibrosis related factors-chronic model
α-Sma Col1a1 Tgfb1
Normal 1.00±0.04 1.00±0.01 1.00±0.02
UUO 2.37±0.26 ## 16.51±1.71 # 2.12±0.16 ##
36(0.1mg/kg) 2.33±0.25 16.18±1.95 * 2.08±1.05 *
36(1mg/kg) 1.59±0.48 10.12±0.98 ** 1.58±0.23 *
39(0.1mg/kg) 2.35±0.59 16.25±1.02 * 2.10±1.95
39(1mg/kg) 1.63±1.13 10.51±0.97 ** 1.79±0.49 **
Note that: # P <0.01 compared to Normal group; p <0.05, P <0.0 compared to UUO group
Effects on kidney tissue fibrosis-related factor mRNA expression: the effect of compounds 33 and 39 on mouse kidney fibrosis was evaluated by detecting changes in the expression level of mRNA of fibrosis-related factors (. Alpha. -Sma, col1a1, tgfb 1) in kidney tissue of each group of mice. As shown in tables 14 and 15, the Acta2, col1a1, tgfb1 mRNA levels in the kidney tissue were significantly increased after 48h administration of folic acid or single side ureter ligation, and the expression was significantly reduced in the treatment group of compounds 33 (1 mg/kg) and 39 (1 mg/kg) compared to the Normal group, and it was seen that compounds 33 and 39 could reduce folate-induced renal fibrosis to some extent.

Claims (7)

1. A guaiane type sesquiterpene lactone derivative, which is characterized by being shown in a general formula I:
wherein R is 1 、R 2 Together form a double bond; or R is 1 Is hydrogen or deuterium, R 2 Is thatWherein R is 3 And R is 4 Respectively C 1 ~C 3 Alkyl, or R 3 、R 4 And the N atom forms pyrrole;
R 5 is-OCONR 6 R 7 Wherein R is 6 Is hydrogen, C 1 ~C 6 An alkyl group; r is R 7 Is C 1 ~C 6 Alkyl groupFluorine substituted C 1 ~C 3 Alkyl, cyclopropane, hydroxy-substituted C 1 ~C 6 Alkyl, methoxy substituted C 1 ~C 6 Alkyl, dimethylaminoethyl and morpholine substituted C 1 ~C 6 Alkyl, piperazine substituted C 1 ~C 6 Alkyl, phenyl ring substituted methyl; or R is 6 、R 7 And N atoms form a substituted 5-6 membered cyclic structure, wherein the cyclic structure is pyrrole, piperidine, piperazine or morpholine.
2. The derivative according to claim 1, characterized in that it is selected from the following compounds:
3. a pharmaceutically acceptable salt of the derivative of claim 1, wherein the pharmaceutically acceptable salt comprises a hydrochloride, sulfate, phosphate, maleate, fumarate, or citrate salt.
4. A pharmaceutically acceptable salt according to claim 3, wherein the pharmaceutically acceptable salt is the hydrochloride or fumarate salt of a derivative:
5. a pharmaceutical composition comprising a guaiane-type sesquiterpene derivative according to any one of claims 1-4 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, adjuvant or adjuvant.
6. Use of a derivative according to claim 1 for the manufacture of a medicament for the treatment of a disease of the NLRP3 inflammatory miniphase Guan Yanzheng.
7. The use according to claim 6, wherein the inflammatory disease is acute lung injury, acute tubular injury, renal interstitial fibrosis, acute liver injury or sepsis caused by bacterial and viral infections.
CN202311579631.9A 2023-11-24 2023-11-24 Guaiane type sesquiterpene lactone derivative and preparation method and application thereof Pending CN117624098A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202311579631.9A CN117624098A (en) 2023-11-24 2023-11-24 Guaiane type sesquiterpene lactone derivative and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202311579631.9A CN117624098A (en) 2023-11-24 2023-11-24 Guaiane type sesquiterpene lactone derivative and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN117624098A true CN117624098A (en) 2024-03-01

Family

ID=90033142

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202311579631.9A Pending CN117624098A (en) 2023-11-24 2023-11-24 Guaiane type sesquiterpene lactone derivative and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN117624098A (en)

Similar Documents

Publication Publication Date Title
US20220331348A1 (en) Method of preparing pentacyclic triterpenoid saponins and drug composition
EP3505526B1 (en) Myricetin derivative and preparing method thereof, and application of same for treating colitis, preventing and treating colitis tumorigenesis, and treating colorectal cancer
HU230727B1 (en) Intermediates for the synthesis of c-aryl glucoside sglt2 inhibitors
CN112156102B (en) Crystal form of NUC-1031 single isomer and preparation method thereof
CN115154476B (en) Cyclocarya paliurus extract and application thereof in resisting gout and reducing uric acid
WO2014154131A1 (en) Caffeoyl substituted pentacyclic triterpene derivative and use thereof
CN110256522B (en) Tripterine cinnamate derivatives, and preparation method and application thereof
CN111484504B (en) Optical isomer of ACC inhibitor and application thereof
CN114591214A (en) Isatin derivative and preparation method thereof
US10759809B2 (en) Deuterated compound and medical use thereof
US20240124409A1 (en) Guaianolide sesquiterpene lactone derivatives and pharmaceutical use thereof
CN111333696B (en) Compound for treating gout and preparation method and application thereof
CN117624098A (en) Guaiane type sesquiterpene lactone derivative and preparation method and application thereof
CN108503678A (en) A kind of iridoid and its preparation method and application
CN100398544C (en) Aqueous triptolide alcohol derivative with high immunesuppressive activity and its application
CN105503988B (en) Natural antiepileptic activity compound and its purposes in pharmaceutical preparation
CN111410679B (en) Preparation and application of Daphnane type macrocyclic diterpenoid compounds
RU2492173C2 (en) Novel compounds with spirochiral carbon base, methods of their obtaining and pharmaceutical compositions which contain such compounds
CN111170980B (en) Calycosin derivative and synthesis method and application thereof
WO2018058863A1 (en) Use of polyether compounds
CN110590779B (en) 3, 10 di-p-chlorophenyl 6, 12 diazatetracubane compound, and synthetic method, application and pharmaceutical composition thereof
CN116559458A (en) Active natural product target spot identification method based on PROTAC protein targeted degradation technology and PROTAC compound
CN113004253B (en) Di- (benzimidazole) -1,2, 3-triazole derivative, preparation method thereof and application thereof in inflammatory skin diseases
CN114409673A (en) Morphinan compound and preparation method and application thereof
CN108785317B (en) Use of cortex Periplocae Radicis C21 steroid compound in preparation of IDO inhibitor

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