CN111202741B - Application of lignan compound in preparation of medicine for treating and/or preventing nephropathy - Google Patents

Application of lignan compound in preparation of medicine for treating and/or preventing nephropathy Download PDF

Info

Publication number
CN111202741B
CN111202741B CN202010068887.3A CN202010068887A CN111202741B CN 111202741 B CN111202741 B CN 111202741B CN 202010068887 A CN202010068887 A CN 202010068887A CN 111202741 B CN111202741 B CN 111202741B
Authority
CN
China
Prior art keywords
compound
nephropathy
treating
cells
solution
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
CN202010068887.3A
Other languages
Chinese (zh)
Other versions
CN111202741A (en
Inventor
欧阳冬生
黄琪
曾祥昌
郭飞
陈露露
李超鹏
陈思雨
刘琼
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.)
Xiangya Hospital of Central South University
Original Assignee
Xiangya Hospital of Central South University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Xiangya Hospital of Central South University filed Critical Xiangya Hospital of Central South University
Priority to CN202010068887.3A priority Critical patent/CN111202741B/en
Publication of CN111202741A publication Critical patent/CN111202741A/en
Application granted granted Critical
Publication of CN111202741B publication Critical patent/CN111202741B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Diabetes (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Endocrinology (AREA)
  • Emergency Medicine (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Urology & Nephrology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses an application of a lignan compound in preparing a medicament for treating and/or preventing nephropathy, wherein the compound is Isoeucommin A and has the following structural formula:
Figure DDA0002376775830000011
the nephropathy comprises at least one of diabetic nephropathy, nephritis, toxic nephropathy, and nephrotic syndrome. The action mechanism of the compound for treating nephropathy is to play an antioxidation role by enhancing intracellular GSH expression and SOD enzyme activity, inhibiting MDA generation and inducing activation of Nrf2/HO-1 signal path, and the discovery of the action mechanism of the compound has important research value and application prospect for treating nephropathy.

Description

Application of lignan compound in preparation of medicine for treating and/or preventing nephropathy
Technical Field
The invention relates to the field of pharmaceutical chemistry, in particular to application of a lignan compound in preparing a medicament for treating and/or preventing nephropathy.
Background
Diabetic Nephropathy (DN) is the most severe and common microvascular complication of diabetes, accounting for 20-40% of the total diabetic patients. The clinical manifestations of DN are mainly proteinuria, hypertension and progressive injury to renal function, edema, etc., and eventually end-stage renal failure can develop. However, the existing effective DN treatment medicines and means are relatively lack, so that the development of a novel effective DN prevention and treatment medicine becomes an important subject facing the global medical and pharmaceutical fields at present. The pathogenesis of DN is complex and the etiology is not clear. The role played by several biochemical mechanisms associated with hyperglycemia in DN has been demonstrated, with the most focused being the Polyol Pathway (PP) and its rate-limiting enzyme, Aldose Reductase (AR), in the pathway. Imbalance in polyol pathways can lead to a series of chain reactions, including oxidative stress mechanisms (oxidative stress), protein non-enzymatic glycosylation (protein non-enzymatic), Protein Kinase C (PKC), and the like. Among them, AR plays an important pivotal role, and has become a popular target for developing drugs for preventing and treating DN. Currently, Aldose Reductase Inhibitors (ARIs) are mainly chemical drugs, mainly including hydantoins, carboxylic acids and heterocyclic compounds. However, many of the drugs have the problems of low selectivity, obvious toxic and side effects, low bioavailability, large clinical dosage and the like. Therefore, the search for novel ARIs with economy, high efficiency, low toxicity and strong specificity has great academic value and clinical significance.
Disclosure of Invention
The present invention is directed to solving at least one of the problems of the prior art. Therefore, the invention provides the application of the lignan compound in preparing the medicine for treating and/or preventing the nephropathy, and the lignan compound has a good treatment effect on the nephropathy.
The invention also provides a medicament for treating and/or preventing nephropathy.
The invention also provides a pharmaceutical composition for treating and/or preventing nephropathy.
According to a use of an embodiment of the first aspect of the invention, the compound has the following structural formula:
Figure BDA0002376775810000021
the application of the embodiment of the invention has at least the following beneficial effects: the compound provided by the invention can effectively inhibit the proliferation of human glomerular mesangial cells induced by high sugar and inhibit H2O2The compound has the action mechanism that the antioxidant effect is exerted by enhancing the intracellular GSH expression and the enzyme activity of SOD, inhibiting the MDA generation and inducing the activation of the Nrf2/HO-1 signal path, provides a good lead compound for developing a new medicine for preventing and/or treating the nephropathy, and has important research value and application prospect for treating the nephropathy.
According to some embodiments of the invention, the kidney disease comprises at least one of diabetic nephropathy, nephritis, toxic nephropathy, and nephrotic syndrome.
According to some embodiments of the invention, preferably, the renal disease comprises diabetic nephropathy.
A medicament according to an embodiment of the second aspect of the invention, comprising a compound as described above and/or a pharmaceutically acceptable salt thereof.
According to some embodiments of the invention, the pharmaceutical dosage form is a tablet, capsule or injection.
A pharmaceutical composition according to an embodiment of the third aspect of the invention comprises a compound as described above and/or a pharmaceutically acceptable salt thereof.
Additional aspects and advantages of the invention will be set forth in part in the description which follows and, in part, will be obvious from the description, or may be learned by practice of the invention.
Drawings
FIG. 1 is a mass spectrum (HRESIMS) of the compound of example 1 of the present invention;
FIG. 2 shows a hydrogen spectrum of the compound of example 1 of the present invention (1H NMR);
FIG. 3 shows a carbon spectrum of a compound of example 1 of the present invention (13C NMR);
FIG. 4 is a circular dichroism plot (CD) of the compound of example 1 of the present invention;
FIG. 5 shows the results of the compound of example 2 of the present invention inhibiting high glucose-induced HRMC cell proliferation;
FIG. 6 shows the induction of expression of normal cells Nrf2 and HO-1 by the compound of example 2;
FIG. 7 shows the induction of expression of high sugar-inducible cells Nrf2 and HO-1 by the compound of example 2 of the present invention;
FIG. 8 shows the effect of the compound of example 2 on the expression of SOD in HRMC cells induced by high sugar;
FIG. 9 shows the effect of the compound of example 2 on MDA expression in HRMC cells induced by high sugar;
FIG. 10 shows the compound pair H in example 3 of the present invention2O2The effect of proliferation of damaged primary tubular cells;
FIG. 11 shows the compound pair H in example 3 of the present invention2O2(ii) the effect of expression of SOD in damaged primary renal tubular cells;
FIG. 12 shows the compound pair H in example 3 of the present invention2O2The effect of MDA expression in damaged primary tubular cells;
FIG. 13 shows the compound pair H in example 3 of the present invention2O2Induction of Nrf2 and HO-1 expression in injured primary tubular cells;
FIG. 14 shows the compound pair H in example 3 of the present invention2O2The expression of GSH in injured primary tubular cells.
Detailed Description
In order to explain technical contents, achieved objects, and effects of the present invention in detail, the following description is made with reference to the accompanying drawings in combination with the embodiments.
MTT detection solution: 50mg of MTT powder was weighed, diluted with 10mL of PBS to 5mg/mL of assay solution, filtered through a 0.22 μm filter, dispensed, and stored at-20 ℃.
Glucose mother liquor: 0.26g D- (+) -glucose was dissolved in 8mL of PBS to prepare a 250mM glucose stock solution, which was filtered through a 0.22 μm filter and used as it was.
Culture medium: DMEM medium with different glucose concentrations of 10% FBS was prepared:
low-sugar DMEM medium (L-DMEM): 47.5mL, 500. mu.L of diabody, 500. mu.L of pyruvic acid, 500. mu.L of MEMNAEA, 500. mu.L of HEPES, 500. mu.L of FBS, and 5.6mM of glucose final concentration;
sugar-free DMEM medium (N-DMEM): 47.5mL, 500. mu.L of diabody, 500. mu.L of pyruvic acid, 500. mu.L of MEMNEAA, 500. mu.L of HEPES and 500. mu.L of FBS.
HRMC cell culture: HRMC cells were incubated at 37 deg.C with 5% CO2And culturing for 2-3 days in a cell culture box with 100% relative humidity. And (3) when the cells grow to 80% confluence degree by adherence, digesting with 0.25% pancreatin for 1-2 minutes, sucking out the pancreatin, and adding a culture medium containing 10% FBS to terminate digestion. The cell suspension was aspirated into a centrifuge tube, centrifuged at 1000rpm for 5min, and the medium was removed. Cells were resuspended in fresh medium at a 1: 4, putting the mixture into a culture flask for continuous subculture.
And (4) performing data analysis by adopting Graphpad 7.0 software, and calculating the survival rate of each group of cells.
Example 1: confirmation of the Structure of the target Compound
1. Mass spectrum: HRESIMS M + Na 573.1972(calculated 573.1948); the mass spectrum is shown in FIG. 1.
2. Nuclear magnetism: the data tables for the hydrogen spectra (FIG. 2) and carbon spectra (FIG. 3) are shown in Table 1 below, with MeOH-d as the solvent4Hydrogen spectrum 600MHz, carbon spectrum 150 MHz:
TABLE 1 assignment of hydrogen and carbon spectra signals (in ppm. delta.) of the target compounds
Figure BDA0002376775810000041
3. Circular dichroism spectrum: the spectrum of the ECD is shown in FIG. 4.
The known compound Isoeutommin A is obtained by analyzing the nuclear magnetism and mass spectrum data, and the structural formula is as follows:
Figure BDA0002376775810000051
example 2: isoeutomomin A (in the figure, labeled P16 to avoid over-space labeling) anti-hyperglycemia-induced human mesangial cell (HRMC) proliferation study
Construction of high-sugar induced HRMC cell proliferation model
1. Taking HRMC cells which are well proliferated and are in logarithmic phase, discarding culture solution, dispersing by digestive juice of 0.25% pancreatin and EDTA half, and counting to prepare cell suspension;
2. adjusting the cell density to 1X 10 with L-DMEM medium4Inoculating to 96-well plate at 80 μ L/well, standing at 37 deg.C and 5% CO2Pre-culturing for 24h in an incubator with 100% relative humidity;
3. adding 20 mu L/well L-DMEM into Blank group and Control group, adding culture medium (the final concentration of Glucose is 30mM) prepared by mixing 20 mu L/well L-DMEM with 250mM Glucose mother liquor into Glucose group, and continuously culturing for 24 h;
4. isoeutomomin A with different concentrations is added to make the final concentration of 250 μm, 125 μm, 62.5 μm, 31.25 μm,15.63 μm, 7.8 μm, 3.9 μm and 1.9 μm, and the cell viability is detected by using an MTT method after the continuous culture for 24 h.
The result of the cell survival rate is shown in figure 5, and the result proves that the compound Isoeutomomin A can effectively inhibit the proliferation of human glomerular mesangial cells induced by high sugar, has obvious dose effect, and has obvious effect when the dose is not less than 31.25 mu m.
II, regulation and control of Nrf2 and HO-1 protein level downstream of Nrf2 pathway
1. Regulation of normal HRMC cells Nrf2 and HO-1 by target compounds
1) Sample preparation
a) Taking HRMC cells which are well proliferated and are in logarithmic phase, discarding culture solution, washing with PBS once, digesting with digestive juice of 0.25% pancreatin and EDTA half, stopping with fresh culture medium, blowing, dispersing and counting to prepare cell suspension;
b) adjusting the cell density to 3X 10 with L-DMEM medium5Cells/well, inoculated into 6-well plate, 2 mL/well, placed at 37 ℃ with 5% CO2Culturing for 24 hours in an incubator with 100% relative humidity;
c) the Blank group, the Control group and the administration group were cultured in L-DMEM (2 ml/well) for 24 hours.
d) The blank group was added with 2. mu.L of DMSO, the administration group was added with 2. mu.L of the objective compound at different concentrations so that the final concentrations were 31.25. mu.M, 62.5. mu.M and 125. mu.M, respectively, and the mixture was subjected to CO treatment2Culturing for 24h in the incubator to collect cell samples;
e) removing culture medium supernatant, washing with PBS once, digesting with 0.25% pancreatin, stopping digestion with 1mL culture medium, centrifuging at low speed, collecting cells, and washing with PBS once again;
f) each 1 × 106Adding 100 mu L of RIPA lysate into each cell, fully suspending, and placing on ice for cracking for 15-30 min; centrifuging at 12000g for 10min at 4 ℃;
g) and sucking a little supernatant for BCA quantification, and adding the A solution: preparing a detection working solution according to the proportion of 50:1 of the solution B, adding 5 mu L of supernatant or standard substance, incubating at 37 ℃ for 30min, and detecting the absorbance at the wavelength of 562 nm;
h) the total protein concentration in the supernatant was calculated and configured to the appropriate concentration using Buffer, plotted as a linear plot against the standard concentration. Adding 4 xSDS loading buffer solution according to the ratio of 3:1, boiling in a metal bath for 10min, and storing at-80 ℃.
2) Electrophoresis
a) Loading on the prepared gel;
b) electrophoresis: adjusting the voltage to 60V, adjusting the voltage to 120V after the protein is concentrated until the bromophenol blue front edge reaches the gel bottom, and ending electrophoresis;
c) film transfer: electrifying, keeping constant voltage, rotating the die for 2h at 18V;
d) and (3) sealing: placing the lower membrane in 5% skimmed milk powder sealing solution, and sealing for 1 hr at room temperature under shaking on a shaking table;
e) primary antibody incubation: dripping primary antibody according to the required dilution concentration, and incubating at 4 ℃ overnight or incubating at 37 ℃ for 3 h;
f) and (3) secondary antibody incubation: the PVDF membrane was removed and washed three times with TBST for 10min each. And adding the secondary antibody dropwise according to the required dilution concentration, and incubating for 1h at room temperature. Taking out the membrane, and washing with TBST for 10min for three times;
g) and opening the developing instrument, preparing an ECL developing solution, uniformly coating the ECL developing solution on the film, and developing.
The experimental result is shown in fig. 6, the compound Isoeucommin a can significantly induce the expression of Nrf2 and HO-1 of normal HRMC, wherein Tublin is tubulin.
2. Regulation of high sugar-induced HRMC cells Nrf2 and HO-1 by target compounds:
a) taking HRMC cells which are well proliferated and are in logarithmic phase, discarding culture solution, dispersing by digestive juice of 0.25% pancreatin and EDTA half, and counting to prepare cell suspension;
b) adding 2 mL/hole L-DMEM into Blank group and Control group, adding 2 mL/hole L-DMEM into Glucose group, adding a culture medium (the final concentration of Glucose is 30mM) prepared by mixing 250mM Glucose mother liquor into Glucose group, and continuously culturing for 24 h;
c)2 μ L DMSO was added to the blank group and the Glucose group, 2 μ L target compound was added to the administration group at different concentrations to give final concentrations of 62.5 μ M and 125 μ M, respectively, and CO was added2Culturing for 24h in the incubator to collect cell samples;
other processing steps and detection methods have the same regulating effect of the target compound on normal HRMC cells Nrf2 and HO-1.
The experimental result is shown in fig. 7, and it can be seen from the figure that the expression of Nrf2 in HRMC cells is significantly reduced under the induction of high sugar, and it is found that the phosphorylation modification level of GSK-3 β is significantly enhanced after isoeucomin a action, and the protein stability of GSK3 β is reduced, so that the inhibition effect of GSK 353 β on Nrf2 is weakened, thereby significantly enhancing the protein expression levels of Nrf2 and HO-1, and exerting an antioxidant effect, further inhibiting the proliferation of mesangial cells in a high sugar environment, and reducing the damage effect of high sugar on kidneys.
Third, detection of superoxide dismutase (SOD) Activity
1. Taking HRMC cells which are well proliferated and are in logarithmic phase, discarding culture solution, dispersing by digestive juice of 0.25% pancreatin and EDTA half, and counting to prepare cell suspension;
2. adding 2 ml/hole L-DMEM into Blank group and Control group, adding 2 ml/hole L-DMEM into Glucose group, adding a culture medium (the final concentration of Glucose is 30mM) prepared by mixing 250mM Glucose mother liquor into Glucose group, and continuously culturing for 24 h;
3. the blank group and the Glucose group were added with 2. mu.L DMSO, and the administration group was added with Isoeutomomin A2. mu.L at different concentrations to give final concentrations of 31.25. mu.M, 62.5. mu.M and 125. mu.M, respectively, and placed in CO2Culturing for 24h in the incubator to collect cell samples;
4. the cells were collected by trypsinization, washed 1-2 times with pre-cooled PBS, added with 40. mu.L PBS and disrupted with liquid nitrogen. Centrifuging at 7500rpm at 4 deg.C for 10min, and collecting supernatant as sample to be tested;
5. sample detection method
1) Preparing WST-8/enzyme working solution: an appropriate amount of WST-8 enzyme working solution was prepared in a volume of 160. mu.L per reaction. 151 mu LSOD detection buffer solution, 8 mu LWST-8 and 1 mu L enzyme solution are uniformly mixed to prepare 160 mu L WST-8/enzyme working solution, and the prepared working solution is prepared as soon as possible.
2) Preparing a reaction starting working solution: dissolving the reaction starting solution of 40X, uniformly mixing, adding 1 mu L of the reaction starting solution (40X) into 39 mu L of SOD detection buffer solution, diluting, and uniformly mixing to obtain the reaction starting working solution.
3) And (3) sample determination:
a. the sample wells and various blank control wells were set using a 96-well plate with reference to table 2 below, and the sample and the working solution were added in sequence. Adding the reaction starting liquid and fully mixing the mixture.
TABLE 2 reaction System
Sample (I) Blank control 1 Blank control 2 Blank control 3
Sample to be tested 20μL 20μL
SOD detection buffer solution 20μL 40μL 20μL
WST-8/enzyme working solution 160μL 160μL 160μL 160μL
Reaction starting working solution 20μL 20μL
b.37 ℃ for 30 minutes, and the absorbance at 450 nm.
4) Calculation of total SOD activity in samples:
percent inhibition ═ 100% for [ (a blank 1-a blank 2) - (a sample-a blank 3) ]/(a blank 1-a blank 2) ×;
the SOD enzyme activity unit in the sample to be detected is the SOD enzyme activity unit in the detection system is the inhibition percentage/(1-inhibition percentage) units.
The experimental results are shown in fig. 8, and the results show that the expression of SOD in HRMC cells is remarkably reduced under the induction of high sugar, and the expression level of SOD can be enhanced after Isoeutomin A acts, so that the anti-oxidation effect is exerted.
Detection of Malondialdehyde (MDA) expression level
1. Taking HRMC cells which are well proliferated and are in logarithmic phase, discarding culture solution, dispersing by digestive juice of 0.25% pancreatin and EDTA half, and counting to prepare cell suspension;
2. adding 2 ml/hole L-DMEM into Blank group and Control group, adding 2 ml/hole L-DMEM into Glucose group and adding 250mM Glucose mother liquor (the final concentration of Glucose is 30mM), and continuously culturing for 24 h;
3. 2 μ L DMSO was added to the blank group and the Glucose group, and Isoeutomomin A2 μ L was added to the administration group to give final concentrations of 31.25 μ M, 62.5 μ M and 125 μ M, respectively, and CO was added2Culturing for 24h in the incubator to collect cell samples;
4. and (3) sample determination:
1) preparing TBA storage solution: weighing a proper amount of thiobarbituric acid (TBA), and preparing TBA storage solution with the concentration of 0.37% by using TBA preparation solution.
2) And (3) preparing an MDA detection working solution, namely dissolving 4X TBA solution, uniformly mixing, adding 150 mu L TBA diluent into 50 mu L TBA storage solution to dilute, adding 3 mu L antioxidant, and uniformly mixing to obtain the MDA detection working solution.
3) Adding 0.1ml of appropriate solution such as homogenate, lysate or PBS as blank control into a centrifuge tube or other appropriate container, adding 0.1ml of the standard substance with different concentrations for preparing standard curve, and adding 0.1ml of sample for determination; subsequently 0.2ml of MDA assay working solution was added.
a. The detection reaction system can be set up with reference to table 3 below:
TABLE 3 reaction System
Blank control Standard article Sample (I)
Homogenate, lysate or PBS 0.1ml
Standard article 0.1ml
Sample to be tested 0.1ml
MDA detection working solution 0.2ml 0.2ml 0.2ml
b. After mixing, heating for 15 minutes at 100 ℃ or in a boiling water bath.
c. The water bath was cooled to room temperature and 1000g was centrifuged at room temperature for 10 min. 200. mu.l of the supernatant was added to a 96-well plate, followed by measurement of absorbance at 532nm with a microplate reader. If it is not convenient to measure the absorbance at 532nm, the absorbance between 530 nm and 540nm may also be measured. The dual wavelength measurement can be performed by setting 450nm as a reference wavelength.
d, calculating the MDA content: the molar concentration of MDA was calculated from the standard curve.
The experimental result is shown in fig. 9, and the result shows that the expression of MDA in the HRMC cell is significantly increased under the induction of high sugar, and the induced expression of MDA can be reduced after the Isoeucommin a acts, so that the anti-oxidation effect is exerted.
Example 3: research on oxidation signal path in Isoeutommin A anti-SD rat primary renal tubular cells
First, target Compound Pair H2O2Effect of injury of primary renal tubular cells in SD rats:
a) inoculating the well-propagated primary renal tubular cells to a 96-well plate at a density of 1500 cells/well, respectively, placing at 37 deg.C and 5% CO2And pre-culturing for 24h in a 100% relative humidity cell culture box.
b) 10% FBS RPMI L-DMEM was added to Blank, Control and Model groups, and Isoeutommin A was added to the administration group so that the final concentrations were 31.25. mu.m, 62.5. mu.m and 125. mu.m. After shaking and mixing, the culture was continued for 24 h.
c) After 24h, the Blank and Control groups were changed to L-DMEM, Model and administration groups to 400. mu. mol/L H2O2The L-DMEM is cultured for 2 hours at 37 ℃ in an incubator. Adding 100 μ L of luminous cell activity detection reagent into each well, placing in a constant temperature oscillator, oscillating at room temperature for 15min, and detecting at 500nm of a microplate reader. The cell growth rate of each group was defined as (observed OD value-blank OD value)/(control OD value-blank OD value) × 100%. The experiment was repeated 3 times and the average was taken.
The results of cell activity experiments are shown in FIG. 10, and the compound Isoeutomomin A can effectively inhibit H2O2Killing primary renal tubules in a dosage effect.
II, H2O2Effect of Isoeucommin A on intracellular SOD, MDA, GSH and Nrf2 signaling pathways after Effect in injury model
1) Taking primary kidney tubule cells with good proliferation at 3X 105The density of each well is inoculated to 6-well plate, and the plate is placed at 37 ℃ and 5% CO2And pre-culturing for 24h in a 100% relative humidity cell culture box.
2) The Blank, Control and Model groups were added with 10% FBS RPMI L-DMEM, the administration group was added with Isoeutommin A to give final concentrations of 31.25 μm, 62.5 μm and 125 μm, and the mixture was shaken and mixed, and then cultured for 24 h.
3) After 24h, the Blank and Control groups were changed to L-DMEM, Model and administration groups to 400. mu. mol/L H2O2The L-DMEM is evenly mixed by shaking and cultured in an incubator at 37 ℃ for 2 hours. Collecting cells and detecting SOD, MDA, GSH and Western-Blot by the same method.
The SOD enzyme activity detection result is shown in FIG. 11, and Isoeutomomin A can effectively enhance H after acting2O2SOD enzyme activity in the damaged primary renal tubular cells presents dose effect; the results of MDA expression level detection are shown in FIG. 12, and H is effectively inhibited after Isoeucommin A acts2O2MDA production in damaged primary tubular cells, in a dose-effect; the expression results of the proteins Nrf2 and HO-1 are shown in FIG. 13, FIG. 13(a) is the result of Western-Blot protein gel electrophoresis, it can be clearly seen that when the dosage is increased, the protein contents of Nrf2 and HO-1 are increased, FIGS. 13(b) and (c) are the protein expression difference of Nrf2 and HO-1 under different dosages, respectively, and when the dosage is not less than 31.25 μ M, the difference is significant, therefore, the Isoeutominin A can effectively enhance H after the action2O2Protein expression of Nrf2 and HO-1 in injured primary tubular cells, in a dose-response.
The GSH detection operation process is as follows:
a) sample preparation:
washing with precooled PBS for 1-2 times, resuspending each cell sample with 60 μ L5% sulfosalicylic acid (SSA), quickly freezing with liquid nitrogen, repeatedly and rapidly freezing and thawing in 37 deg.C water bath for 2 times to lyse cells, standing on ice for 10min, centrifuging at 10000rpm for 10min, and collecting supernatant as sample to be tested;
b) sample detection:
1) preparing a working solution: mu.L of glutathione reductase was diluted to 250. mu.L with 1X of the assay solution, i.e., enzyme solution. Adding 228 mu L of enzyme solution and 228 mu L of DTNB stock solution into 8mL of 1X detection solution, and uniformly mixing;
2) preparing an NADPH solution: sucking 10 μ L of NADPH original solution, adding into 2.5mL of 1 × detection solution, and mixing;
3) and (3) glutathione standard substance: diluting GSH stock solution 200 times with 5% sulfosalicylic acid (SSA) to obtain 50 μ M standard 1, diluting 2 times of standard 1 to obtain standard 2, and sequentially diluting 2 times to obtain standard 5;
4) the samples and desired solutions were added as specified in table 4 below:
TABLE 4
Figure BDA0002376775810000111
The average absorbance change per 5min was measured with a microplate reader at a wavelength of 412 nm.
From the results shown in FIG. 14, Isoeutommin A was effective in enhancing H after the action2O2GSH expression in injured primary tubular cells, in a dose-response.
In conclusion, the compound Isoeutommin A provided by the invention can effectively inhibit the proliferation of human glomerular mesangial cells induced by high sugar and inhibit H2O2Further inhibits the apoptosis of the kidney cells by the damage effect on the primary renal tubular cells. The action mechanism of the compound Isoeutomomin A is that the compound Isoeutomomin A plays an antioxidation role by enhancing the expression of GSH in cells, the enzyme activity of SOD, inhibiting the generation of MDA and inducing the activation of Nrf2/HO-1 signal channel, provides a good lead compound for developing a new medicament for preventing and/or treating nephropathy, and has important research value and application prospect for treating nephropathy.
The above description is only an embodiment of the present invention, and not intended to limit the scope of the present invention, and all equivalent changes made by using the contents of the present specification and the drawings, or applied directly or indirectly to the related technical fields, are included in the scope of the present invention.

Claims (1)

1. Use of a lignan compound in the manufacture of a medicament for the treatment and/or prevention of a kidney disease, wherein the compound has the formula:
Figure FDA0002973679520000011
the nephropathy is diabetic nephropathy.
CN202010068887.3A 2020-01-21 2020-01-21 Application of lignan compound in preparation of medicine for treating and/or preventing nephropathy Active CN111202741B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010068887.3A CN111202741B (en) 2020-01-21 2020-01-21 Application of lignan compound in preparation of medicine for treating and/or preventing nephropathy

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010068887.3A CN111202741B (en) 2020-01-21 2020-01-21 Application of lignan compound in preparation of medicine for treating and/or preventing nephropathy

Publications (2)

Publication Number Publication Date
CN111202741A CN111202741A (en) 2020-05-29
CN111202741B true CN111202741B (en) 2021-07-20

Family

ID=70782056

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010068887.3A Active CN111202741B (en) 2020-01-21 2020-01-21 Application of lignan compound in preparation of medicine for treating and/or preventing nephropathy

Country Status (1)

Country Link
CN (1) CN111202741B (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103479711A (en) * 2013-09-17 2014-01-01 欧阳冬生 Application of eucommia ulmoides lignans extracts in preparation of medicine treating renal interstitial fibrosis
KR20150039351A (en) * 2013-10-02 2015-04-10 충남대학교산학협력단 A composition comprising extract of Japanese apricot or compounds isolated therefrom for preventing or treating osteoporosis
CN105582000A (en) * 2016-01-12 2016-05-18 张忠立 Preparation method of terpenoid and lignan substances in eucommia ulmoides bark or eucommia ulmoides leaves and application of terpenoid and lignan substances in preparation of senile dementia treatment drug

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103479711A (en) * 2013-09-17 2014-01-01 欧阳冬生 Application of eucommia ulmoides lignans extracts in preparation of medicine treating renal interstitial fibrosis
KR20150039351A (en) * 2013-10-02 2015-04-10 충남대학교산학협력단 A composition comprising extract of Japanese apricot or compounds isolated therefrom for preventing or treating osteoporosis
CN105582000A (en) * 2016-01-12 2016-05-18 张忠立 Preparation method of terpenoid and lignan substances in eucommia ulmoides bark or eucommia ulmoides leaves and application of terpenoid and lignan substances in preparation of senile dementia treatment drug

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
Chemical constituents from Alisma plantago-aquatica subsp. orientale (Sam.) Sam and their anti-inflammatory and antioxidant activities;Xin-Yu Zhao 等;《Natural Product Research》;20181231;第32卷(第23期);第2749-2755页 *
LIGNANS AND A STILBENE FROM FESTUCA ARGENTINA;ADRIANA C. CASABUON 等;《Phytochemistry》;19941231;第35卷(第2期);第479-483页 *
Xin-Yu Zhao 等.Chemical constituents from Alisma plantago-aquatica subsp. orientale (Sam.) Sam and their anti-inflammatory and antioxidant activities.《Natural Product Research》.2018,第32卷(第23期), *
弯管列当的化学成分研究;杨美珍 等;《中药学》;20140930;第45卷(第17期);第2447-2452页 *
活血化瘀类中药治疗糖尿病肾病机制的研究进展;白宇 等;《中国实验方剂学杂志》;20181231;第24卷(第23期);第200-206页 *

Also Published As

Publication number Publication date
CN111202741A (en) 2020-05-29

Similar Documents

Publication Publication Date Title
Hou et al. Mangiferin suppressed advanced glycation end products (AGEs) through NF-κB deactivation and displayed anti-inflammatory effects in streptozotocin and high fat diet-diabetic cardiomyopathy rats
Wu et al. Identification of novel dynamin‐related protein 1 (Drp1) GTPase inhibitors: therapeutic potential of Drpitor1 and Drpitor1a in cancer and cardiac ischemia‐reperfusion injury
Sun et al. Taxifolin prevents diabetic cardiomyopathy in vivo and in vitro by inhibition of oxidative stress and cell apoptosis
Leontieva et al. Yeast-like chronological senescence in mammalian cells: phenomenon, mechanism and pharmacological suppression
Capdevila et al. The Cyp2c44 epoxygenase regulates epithelial sodium channel activity and the blood pressure responses to increased dietary salt
Li et al. ROS and iron homeostasis dependent ferroptosis play a vital role in 5-Fluorouracil induced cardiotoxicity in vitro and in vivo
Tugcu et al. Protective effect of a potent antioxidant, pomegranate juice, in the kidney of rats with nephrolithiasis induced by ethylene glycol
US20160074373A1 (en) Methods and compositions for inhibiting human copper trafficking proteins atox1 and ccs
Tang et al. Salidroside attenuates CoCl2-simulated hypoxia injury in PC12 cells partly by mitochondrial protection
Wang et al. Inhibition of glycogen synthase kinase 3beta ameliorates triptolide-induced acute cardiac injury by desensitizing mitochondrial permeability transition
Liu et al. Conversion of 2-deoxyglucose-induced growth inhibition to cell death in normoxic tumor cells
CN111202741B (en) Application of lignan compound in preparation of medicine for treating and/or preventing nephropathy
Zhang et al. Tubeimoside I Ameliorates Doxorubicin‐Induced Cardiotoxicity by Upregulating SIRT3
Guan et al. Troxerutin alleviates kidney injury in rats via PI3K/AKT pathway by enhancing MAP4 expression
Yuan et al. Palmitate impairs the autophagic flux to induce p62-dependent apoptosis through the upregulation of CYLD in NRCMs
Jian et al. The effect of 17-methoxyl-7-hydroxy-benzene-furanchalcone isolated from Millettia pulchra on myocardial ischemia in vitro and in vivo
Liu et al. Effects of Rosa laevigata Michx. extract on reactive oxygen species production and mitochondrial membrane potential in lens epithelial cells cultured under high glucose
Zhang et al. Cardiotoxicity of patulin was found in H9c2 cells
CN108578702B (en) Mixture of rare ginsenoside and application thereof
Harlow et al. Insulin-like activity from the sparganum of Spirometra mansonoides
CN110551156A (en) specific group modified N1, N3 substituted 4, 5-diaryl imidazole ring carbene rhodium complex and preparation method and application thereof
US20220249450A1 (en) Inhibitors of Mitochondrial Fission
Santana Filho et al. Seleno-indoles trigger reactive oxygen species and mitochondrial dysfunction in Leishmania amazonensis
TWI738334B (en) Use of naphthoquinone derivative for inhibiting proliferation and metastasis of cancer cells
Taheri et al. Neuroprotective Effect of Thiazolidine-2, 4-dione Derivatives on Memory Deficits and Neuropathological Symptoms of Dementia on a Scopolamine-Induced Alzheimer’s Model in Adult Male Wistar Rats

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