CN116159067A - Application of cyclovirobuxine D in preparing medicament for treating cervical cancer - Google Patents

Application of cyclovirobuxine D in preparing medicament for treating cervical cancer Download PDF

Info

Publication number
CN116159067A
CN116159067A CN202211500660.7A CN202211500660A CN116159067A CN 116159067 A CN116159067 A CN 116159067A CN 202211500660 A CN202211500660 A CN 202211500660A CN 116159067 A CN116159067 A CN 116159067A
Authority
CN
China
Prior art keywords
cyclovirobuxine
cervical cancer
cell
cells
medicine
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
CN202211500660.7A
Other languages
Chinese (zh)
Inventor
沈卫阳
林春霞
洪思祺
张旭
杨勇
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 Edberg Pharmaceutical Technology Co ltd
China Pharmaceutical University
Original Assignee
Nanjing Edberg Pharmaceutical Technology Co ltd
China Pharmaceutical 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 Nanjing Edberg Pharmaceutical Technology Co ltd, China Pharmaceutical University filed Critical Nanjing Edberg Pharmaceutical Technology Co ltd
Priority to CN202211500660.7A priority Critical patent/CN116159067A/en
Publication of CN116159067A publication Critical patent/CN116159067A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/57Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of two carbon atoms, e.g. pregnane or progesterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Abstract

The invention discloses an application of cyclovirobuxine D in preparing a medicament for treating cervical cancer, and belongs to the technical field of medicines. The invention also discloses a medicine for treating cervical cancer, which comprises cyclovirobuxine D and derivatives or pharmaceutically acceptable salts thereof. According to the invention, MTT (methyl thiazolyl tetrazolium) experiments, cell cycle experiments, scratch experiments, hoechst33342 staining experiments and mitochondrial membrane potential detection experiments show that cyclovirobuxine D can remarkably inhibit proliferation and migration of cervical cancer cells and induce apoptosis. The application of the cyclovirobuxine D in the preparation of the medicine for treating cervical cancer not only digs out the new medicinal value of the cyclovirobuxine D, but also has certain medical prospect and economic value.

Description

Application of cyclovirobuxine D in preparing medicament for treating cervical cancer
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to application of cyclovirobuxine D in preparing a medicine for treating cervical cancer.
Background
Cervical cancer is a malignant tumor occurring in the cervix, which is a global fourth-size female cancer, with a female morbidity of 6.6% and mortality of 7.5%, which is a significant global health challenge. Cervical cancer is a complex and slow process, closely related to infection with human papillomavirus (Human papillomavirus, HPV). In addition to vaccines for preventing HPV infection, clinically common modes of treatment include excision, radiation therapy, chemo-radiation complications and gene therapy. Although the current operation treatment and auxiliary treatment modes have certain progress, the development and recurrence of diseases still plagues women suffering from cervical cancer, so that the search for new anti-cervical cancer drugs has great clinical demands and is also a hotspot of the current international research.
The development of drugs for inhibiting proliferation and migration of cervical cancer cells and inducing apoptosis has important practical significance for treating cervical cancer.
Cyclovirobuxine D (CVB-D) is an alkaloid refined from Buxus microphylla and other congeneric plants. Studies have shown that cyclovirobuxine D inhibits the tumorigenesis of colorectal cancer through CTHRC1-AKT/ERK-Snail signaling pathway and exerts anticancer effects through inhibiting EGFR-FAK-AKT/ERK1/2-Slug signaling pathway of human hepatocellular carcinoma. In addition, cyclovirobuxine D inhibits cell proliferation and induces mitochondrial-mediated apoptosis in human gastric cancer cells and induces autophagy-related cell death via Akt/mTOR pathway in MCF-7 human breast cancer cells.
At present, cyclovirobuxine D is not reported as an active ingredient for treating cervical cancer.
Disclosure of Invention
The invention aims to: the invention aims at overcoming the defects of the prior art and provides application of cyclovirobuxine D in preparing medicaments for treating cervical cancer. According to the invention, through related researches, the cyclovirobuxine D can obviously inhibit proliferation, migration and apoptosis of human cervical cancer cells Hela. The application of the cyclovirobuxine D in the preparation of the medicine for treating cervical cancer not only digs out the new medicinal value of the cyclovirobuxine D, but also has certain medical prospect and economic value.
The technical scheme is as follows: the aim of the invention is achieved by the following technical scheme:
the invention provides an application of cyclovirobuxine D in preparing a medicament for treating cervical cancer.
Cyclovirobuxine D is a natural small molecule compound extracted from plant of Buxaceae. The plant of Buxaceae is Buxaceae or its congeneric plant.
The invention also provides a medicine for treating cervical cancer, which comprises cyclovirobuxine D and derivatives or pharmaceutically acceptable salts thereof.
Preferably, the pharmaceutically acceptable salt comprises a hydrochloride, sulfate, phosphate, oxalate or citrate salt.
The invention relates to the pharmaceutically acceptable salts, which are converted into cyclovirobuxine D in vivo. For example, within the scope of the present invention, the cyclovirobuxine D of the invention is converted into a pharmaceutically acceptable salt form and used in salt form according to processes known in the art.
Preferably, the medicament further comprises pharmaceutically acceptable excipients.
Further, the auxiliary materials comprise one or more of preservative, diluent, disintegrating agent, antioxidant, wetting agent, emulsifying agent, adhesive, lubricant and solubilizer.
The preservative is at least one selected from benzoic acid and salts thereof, sorbic acid and salts thereof and parabens. The diluent is at least one selected from starch, saccharide, cellulose and inorganic salts. The disintegrating agent is at least one selected from starch, sodium carboxymethyl starch, crosslinked povidone, low-substituted hydroxypropyl cellulose and crosslinked polyvinylpyrrolidone. The antioxidant is at least one selected from ascorbic acid, sulfite, bisulfite, gallic acid and lipid thereof. The wetting agent is at least one selected from water and ethanol. The emulsifier is at least one selected from tween, span, glycerol fatty acid esters, pectin, agar, sulfate, sodium alginate and silicon dioxide. The binder is at least one selected from starch slurry, sodium carboxymethyl cellulose, povidone, hydroxypropyl cellulose, methyl cellulose and ethyl cellulose. The lubricant is at least one selected from magnesium stearate, talcum powder, hydrogenated vegetable oil, polyethylene glycol and micropowder silica gel. The solubilizer is one of tween, polyoxyethylene fatty alcohol ether, sulfate and sulfonate.
Preferably, the dosage form of the medicament is granules, tablets, capsules, pills, suspensions, solutions, injections or infusion solutions.
The medicaments of the present invention may be administered in a variety of known ways, for example orally, by injection, by inhalation spray. The medicine of the invention can be used alone or in combination with other medicines. The oral composition may be any orally acceptable dosage form including, but not limited to, granules, tablets, capsules, pills, suspensions, and solutions.
Sterile injectable compositions can be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. Pharmaceutically acceptable carriers and solvents that can be used include water, sodium chloride solution, and the like.
The medicine of the invention can be prepared into common preparations, and also can be prepared into sustained release preparations, controlled release preparations, targeted preparations and various microparticle administration systems.
The actual dosage level of the active ingredient in the medicament of the present invention may be varied to obtain an amount of active ingredient that is effective to achieve the desired therapeutic response for the particular patient, composition and mode of administration, which is non-toxic to the patient. The selected dosage level depends on a variety of factors including the route of administration, the time of administration, the rate of excretion, the duration of the treatment, other drugs, compounds and/or materials used in combination with the cyclovirobuxine D, the age, sex, weight, general health and past medical history of the patient being treated, and like factors well known in the medical arts.
The cyclovirobuxine D can inhibit proliferation and migration of human cervical cancer cells Hela cells and induce apoptosis of cervical cancer cells, and is used for treating cervical cancer.
The beneficial effects are that:
the invention discovers a novel anti-cervical cancer natural small molecular compound cyclovirobuxine D and provides application thereof in preparing medicaments for treating cervical cancer. The results of MTT experiment, cell cycle experiment, scratch experiment, hoechst33342 staining experiment and mitochondrial membrane potential detection experiment show that: the cyclovirobuxine D can obviously inhibit proliferation and migration of human cervical cancer cells Hela cells, induce apoptosis and have the effect of resisting cervical cancer. The application of the cyclovirobuxine D in the preparation of the medicine for treating cervical cancer not only digs out the new medicinal value of the cyclovirobuxine D, but also has certain medical prospect and economic value.
Drawings
FIG. 1 is a graph showing experimental results of inhibiting human cervical cancer cell Hela proliferation by cyclovirobuxine D.
FIG. 2 is a diagram showing the experimental cloning of cyclovirobuxine D acting on Hela cells, a human cervical cancer cell.
FIG. 3 is a graph showing the cell cycle distribution experiment of flow cytometry analysis of human cervical cancer cells Hela cells.
FIG. 4 is a graph showing a scratch experiment of cyclovirobuxine D acting on Hela cells, a human cervical cancer cell.
FIG. 5 is a chart showing the experiment of staining hoechst33342 of cyclovirobuxine D acting on Hela cells, a human cervical cancer cell.
Fig. 6 is a mitochondrial membrane potential detection experimental diagram of cyclovirobuxine D acting on human cervical cancer cell Hela cells.
Detailed Description
The technical scheme of the present invention is described in detail below through specific examples, but the scope of the present invention is not limited to the examples.
The specific techniques or conditions are not identified in the examples and are described in the literature in this field or are carried out in accordance with the product specifications. The reagents or equipment used were conventional products available for purchase through regular channels, with no manufacturer noted.
The experimental methods in the following examples are conventional methods unless otherwise specified. The test materials used in the examples described below, unless otherwise specified, are all commercially available products.
The specifications and sources of reagents used in the examples: cyclovirobuxine D, purchased from Chinese food and drug testing institute; 4% paraformaldehyde was purchased from Biosharp corporation; crystal violet, purchased from alaa Ding Gongsi.
DMEM high sugar medium and foetal calf serum were purchased from Gibco,
penicillin-streptomycin (diabody), PBS buffer and pancreatin cell digests, cell cryopreservation, hoechst33342 live cell staining solution (100×) were purchased from yunnan biological reagent company;
thiazole blue (MTT) was purchased from Sigma, usa;
cell cycle kits were purchased from Kaiyi organisms;
c2006 mitochondrial membrane potential detection kit (JC-1) was purchased from the bi yun tian biological reagent company and included: JC-1 (200X), JC-1 staining buffer (5X), CCCP (10 mM), ultrapure water.
Cell culture broth (10% fetal bovine serum, 1% diabody): adding 50mL of fetal bovine serum and 5mL of diabody into a 500mL sterile container, and fixing the volume to 500mL by using a DMEM high-sugar culture medium to obtain the feed;
low serum cell culture broth (2% fetal bovine serum, 1% diabody): adding 10mL of fetal bovine serum and 5mL of diabody into a 500mL sterile container, and fixing the volume to 500mL by using a DMEM high-sugar culture medium to obtain the feed;
MTT working solution (5 mg/mL): under the condition of avoiding light, 0.6g of MTT powder is completely dissolved in 12mL of 1 XPBS in a sterile super clean bench, filtered and sterilized by a 0.22 mu m filter, and then split-packed in a 1.5mL centrifuge tube (wrapped with tin foil) and preserved in a dark place at-20 ℃;
crystal violet staining solution: 0.05g of crystal violet powder is weighed and dissolved in 10mL of absolute methanol to prepare 0.5% crystal violet stock solution, and the stock solution is stored in a refrigerator at 4 ℃ in a dark place. The working concentration of crystal violet is 0.1%, and the crystal violet is diluted by PBS when in use;
hoechst33342 (1×): taking 0.1mL of cell culture solution, and adding 0.9ml Hoechst33342 living cell staining solution (100X) to obtain the cell culture solution;
JC-1 dyeing working solution: diluting JC-1 by adding 8mL of ultrapure water into 50 mu L of JC-1 (200X), vigorously swirling to mix the JC-1 evenly, then adding 2mL of JC-1 dyeing buffer solution (5X), and mixing evenly to obtain JC-1 dyeing working solution;
CCCP working fluid (10 μm): taking 2 mu L of 10mM CCCP, and adding 998 mu L of cell culture solution to obtain the compound;
JC-1 staining buffer (1×): 1mL of JC-1 staining buffer (5X) is taken, and 4mL of distilled water is added to be uniformly mixed to obtain the dye.
Hela cell line (human cervical cancer cell) was purchased from the China academy of sciences cell bank.
The test detection instruments used in the examples were: flow cytometer, FACSCalibur, BD company, usa; fluorescent inverted microscope, TS2, NOKON company; the enzyme-labeled instrument, tecan Sunrise, austria.
Example 1MTT assay for detecting anti-tumor Activity of cyclovirobuxine D
Selecting human cervical cancer Hela cells in logarithmic growth phase, discarding old culture solution, washing the cells twice with 1mL PBS buffer solution respectively, adding 1mL pancreatin cell digestive solution to eliminate cell mass into single cells, adding 3mL cell culture solution to stop digestion, centrifuging for 5min, discarding supernatant, adding 1mL cell culture solution again to blow the precipitate into single cell suspension, and diluting to proper cell concentration.
Using 96-well plate, adding 3000 logarithmic phase Hela cells (100 μL) into each of control group and experimental group, adding 100 μL fresh cell culture solution into blank group, setting three parallel groups into control group, experimental group and blank group, respectively adding 100 μL fresh cell culture solution into control group after overnight growth of cells, respectively adding 100 μL cyclovirobuxine D liquid with final concentration of 20 μmol/L, 40 μmol/L, 60 μmol/L and 80 μmol/L diluted with cell culture solution into experimental group, respectively adding 100 μL fresh culture solution into blank group, and adding 5% CO at 37deg.C 2 After incubation for 24, 48, 72h under the conditions, 20. Mu.L of MTT (5 mg/ml) working solution was added to each group, and after placing the 96-well plates in an incubator for 4h,the supernatant was aspirated, 200. Mu.L of DMSO was added to dissolve the precipitate, and the mixture was shaken on a shaker at 37℃for 10min at low speed, and the absorbance at 492nm was measured with an ELISA reader.
Cell viability = (experimental OD value-blank OD value/control OD value-blank OD value) ×100%.
FIG. 1 is a graph showing the experimental results of inhibiting Hela cell proliferation by cyclovirobuxine D. As can be seen from fig. 1: cyclovirobuxine D has 24, 48 and 72h of IC on Hela cells 50 The values were 58.82. Mu. Mol/L, 35.69. Mu. Mol/L and 18.93. Mu. Mol/L, respectively.
Experimental results show that the survival rate of the Hela cells is gradually reduced along with the increase of the concentration of the cyclovirobuxine D, and the Hela cells form a dose-response relationship.
Example 2 cloning experiments to test the antiproliferative Capacity of cyclovirobuxine D
2000 logarithmic phase HeLa cells were added to each well of a 6-well plate and placed at 37℃with 5% CO 2 Culturing in an incubator. After the cells are attached, the original culture medium is discarded, 2mL of fresh cell culture solution is added into a control group, 2mL of cyclovirobuxine D liquid medicine which is diluted to the final concentration of 20 mu mol/L, 40 mu mol/L, 60 mu mol/L and 80 mu mol/L by the cell culture solution is respectively added into an experiment group, and the experiment group is placed into an incubator at 37 ℃ and contains 5% CO 2 After 24h incubation, the liquid was discarded, washed twice with 1mL of PBS buffer, and 2mL of cell culture medium was added to each well for further culture. The fluid was changed every 3d and the proliferation of the cell clone was observed. After 14d, sufficient colonies have formed in each well of the 6-well plate (a plurality of cell masses greater than 10 cells have formed under microscopic observation). After the medium is discarded, 1mL of PBS buffer solution is used for carefully washing twice, 1mL of 4% paraformaldehyde is used for fixing cells for 10min in each hole, 1mL of 0.5% crystal violet staining solution is added in each hole, the staining solution is sucked and removed after the light-shielding staining for 10min, the residual staining solution is washed by tap water, and after the cells are naturally dried, a digital camera is used for photographing and recording.
FIG. 2 is a graph showing the cloning experiments of cyclovirobuxine D acting on Hela cells.
The experimental results show that: along with the increase of the concentration of the cyclovirobuxine D, the clone number of the Hela cell colony is reduced, and the cloning capacity is gradually weakened, which shows that the cyclovirobuxine D has an inhibition effect on the growth and proliferation of Hela cells and has the characteristic of concentration dependence.
Example 3 flow cytometry determination of the effects of cyclovirobuxine D on Hela cell cycle
Taking logarithmic growth phase Hela cells, centrifuging at 1000rpm for 3min, removing supernatant, adding 1mL cell culture solution, gently blowing to uniform cell suspension, counting with microscope, and collecting 1×10 cells 5 The density of each/mL was evenly added to a 6-well plate with 2mL per well and placed in an incubator overnight. Discarding the original culture medium, washing twice with 1mL PBS buffer solution, adding 2mL cell culture solution into control group, adding 2mL cell culture solution into medicine group, diluting with 40 μmol/L cyclovirobuxine D liquid medicine, standing at 37deg.C, and adding 5% CO 2 After incubation in an incubator for 24h under the conditions, the culture medium was discarded, washed twice with 1mL of PBS buffer, and after digestion of the cells with 500 μl of EDTA-free pancreatin per well, 1mL of cell culture medium was added to gently blow the cells, and the cell suspension was transferred to a 2mL centrifuge tube, placed in 4 ℃, and centrifuged at 2000rpm for 5min. Cells were washed 2 times with pre-chilled 1mL PBS, the supernatant was discarded, and 1mL of 70% absolute ethanol was used to resuspend cells overnight at-20 ℃.
Before testing, at 4 ℃,2000rpm, centrifuging for 5min to remove the fixing solution, adding 500 mu L of PI/RNase A staining working solution (before using, the PI/RNase A working solution is prepared into the staining working solution according to the volume of 9:1 and is derived from a cell cycle kit) into each sample, incubating for 15min at room temperature in a dark place, filtering the cell suspension by a 200-mesh filter screen, and detecting the sample by a flow cytometer.
FIG. 3 is a graph showing cell cycle distribution experiments of flow cytometry analysis of Hela cells.
The results of fig. 3 show that: compared with the control group, the cyclovirobuxine D group has statistical difference * P < 0.05). Experiments show that: under the action of cyclovirobuxine D, the mitotic cycle of Hela cells is changed and is a G2/M phase block. Thus, the cell G2/M phase retardation may be the mechanism of action of cyclovirobuxine D in inhibiting cancer cell proliferation.
Example 4 scratch assay to determine the Effect of cyclovirobuxine D on cervical cancer cell migration ability
Five lines are drawn on the back of each hole of the six-hole plate at uniform intervals along the ruler by using a marker. Taking HeLa cells in logarithmic growth phase according to 1X 10 5 The density of each/mL was evenly added to a 6-well plate with 2mL per well and placed in an incubator overnight. After the cell wall had grown full, a 200. Mu.L gun head was used to scratch vertically along the line drawn (note that the gun head remained vertical during this process). Slowly washing with 1mL PBS for 2 times, washing away the scratched cells, adding 2mL of low-serum culture solution into the medicine group to dilute the medicine group to obtain final concentration of 20 mu mol/L, 40 mu mol/L, 60 mu mol/L and 80 mu mol/L cyclovirobuxine D liquid, adding the same amount of low-serum culture solution into the control group, continuously placing the control group into an incubator for culturing, observing the scratch condition among the cells under a fluorescent inverted microscope at 0h, 24h and 48h respectively, and photographing and recording the experimental result.
FIG. 4 is a graph showing a scratch test of cyclovirobuxine D acting on Hela cells. Figure 4 shows that cyclovirobuxine D is able to inhibit Hela cell migration and has a dose-dependent effect on migration capacity.
The experimental result shows that the wound healing capacity of the Hela cells is reduced after the Hela cells are treated by the cyclovirobuxine D, which shows that the cyclovirobuxine D can inhibit the migration of the Hela cells, and the migration inhibiting effect is related to the acting concentration of the medicine.
Example 5hoechst 33342 staining experiment to determine the Effect of cyclovirobuxine D on the ability to induce apoptosis in cervical cancer cells
Taking HeLa cells in logarithmic growth phase, 5×10 4 Each cell/mL was inoculated into 6-well plates at 2mL per well, and placed at 37℃in 5% CO 2 Culturing in an incubator. After the cells are attached, the original cell culture solution is discarded, 2mL of cell culture solution is added into the medicine group to dilute the medicine group to obtain the final concentration of 20 mu mol/L, 40 mu mol/L, 60 mu mol/L and 80 mu mol/L cyclovirobuxine D medicine liquid, the control group is added with the equivalent cell culture solution, and the medicine group is placed at 37 ℃ and contains 5% CO 2 After incubation for 24h in an incubator under the condition, the liquid medicine is discarded, and after washing twice with 1mL of PBS, 2mL of 1 Xhoechst 33342 staining solution is added into each hole, and after the incubation for 10min in the incubator, the dye-containing culture solution is sucked out and separatedAfter washing twice with 1mL of PBS, each well was added with 1mL of cell culture solution, the change of the cell nucleus morphology was observed under a fluorescence microscope, and the result was recorded by photographing.
FIG. 5 is a chart showing an experiment of staining of hoechst33342 of cyclovirobuxine D acting on HeLa cells.
The results of fig. 5 show that: along with the increase of the concentration of the cyclovirobuxine D, chromatin in cells is condensed, the brightness is improved, and nucleus cleavage occurs in cell nuclei, which indicates that the cyclovirobuxine D can induce hela cell apoptosis, and the higher the drug concentration is, the more the number of apoptotic cells is increased.
Example 6 mitochondrial Membrane potential detection experiments to determine the effects of cyclovirobuxine D on the ability of inducing apoptosis in cervical cancer cells
Taking HeLa cells in logarithmic growth phase 1×10 5 Each cell/mL was inoculated into a 6-well plate at 2mL per well and placed in an incubator overnight. The original cell culture solution is discarded, 2mL of cell culture solution is added into the medicine group to dilute the medicine group, the final concentration is 20 mu mol/L, 40 mu mol/L, 60 mu mol/L and 80 mu mol/L of cyclovirobuxine D medicine liquid, the equivalent cell culture solution is added into the control group, and the equivalent cell culture solution is added into the positive control group. Placing at 37deg.C, containing 5% CO 2 After 24h in an incubator under the condition, the positive control group is firstly washed twice with 1mL of PBS respectively, 2mL of CCCP working solution is added, the incubation is carried out for 20min in the incubator under the condition of avoiding light, old cell culture solution in a six-hole plate is removed, 1mL of PBS is respectively washed twice, 1mL of cell culture solution and 1mL of JC-1 staining working solution are respectively added, the incubation is carried out for 20min in the incubator under the condition of avoiding light, after the incubation is finished, the supernatant is sucked, 1mL of 1 XJC-1 staining buffer solution is used for washing twice at 4 ℃, 2mL of cell culture solution is added into each hole, and the cells are observed under a fluorescence microscope.
FIG. 6 is a graph showing the mitochondrial membrane potential assay of cyclovirobuxine D acting on HeLa cells.
When the mitochondrial membrane potential is high, JC-1 gathers in the matrix of mitochondria to form a polymer, and red fluorescence can be generated; at low mitochondrial membrane potential, JC-1 cannot be accumulated in the matrix of mitochondria, and JC-1 is a monomer and can generate green fluorescence. This allows for a very convenient detection of changes in mitochondrial membrane potential by fluorescence color transitions. The decrease in cell membrane potential can be readily detected by the transition of JC-1 from red to green fluorescence, and the transition of JC-1 from red to green fluorescence can also be used as a detection indicator for early apoptosis.
As shown in the results of FIG. 6, after Hela cells were treated with CCCP as a positive control, the red fluorescence was weaker (FIG. 6-a), high-density green fluorescence appeared in the visual field (FIG. 6-b), and high-density red fluorescence was observed after JC-1 staining of cells in the control group (FIG. 6-c), and the green fluorescence intensity was weaker (FIG. 6-d); while as the concentration of cyclovirobuxine D increases, hela cells treated with cyclovirobuxine D observed a decrease in red fluorescence (fig. 6-e, 6-g, 6-i, 6-k) and an increase in green fluorescence intensity (fig. 6-f, 6-h, 6-j, 6-l), indicating that the membrane potential of Hela cells after treatment with cyclovirobuxine D decreased to a degree related to the drug concentration. It can be seen that cyclovirobuxine D has the ability to induce apoptosis of cervical cancer cells.
The results of MTT experiment, cell cycle experiment, scratch experiment, hoechst33342 staining experiment and mitochondrial membrane potential detection experiment show that: the cyclovirobuxine D can obviously inhibit proliferation and migration of human cervical cancer Hela cells, induce apoptosis, have the effect of resisting cervical cancer and can be used for preparing medicaments for treating cervical cancer.
As described above, although the present invention has been shown and described with reference to certain preferred embodiments, it is not to be construed as limiting the invention itself. Various changes in form and details may be made therein without departing from the spirit and scope of the invention as defined by the appended claims.

Claims (7)

1. Application of cyclovirobuxine D in preparing medicine for treating cervical cancer is provided.
2. A medicament for treating cervical cancer, which is characterized by comprising cyclovirobuxine D or a derivative or pharmaceutically acceptable salt thereof.
3. The medicament of claim 2, wherein the pharmaceutically acceptable salt comprises a hydrochloride, sulfate, phosphate, oxalate, or citrate salt.
4. The medicament of claim 2, further comprising a pharmaceutically acceptable adjuvant.
5. The medicine according to claim 4, wherein the auxiliary materials comprise one or more of preservative, diluent, disintegrating agent, antioxidant, wetting agent, emulsifying agent, adhesive, lubricant and solubilizing agent.
6. The use according to claim 1, wherein the medicament is in the form of granules, tablets, capsules, pills, suspensions, solutions, injections or infusions.
7. The use according to claim 1, wherein cyclovirobuxine D is used for the treatment of cervical cancer by inhibiting proliferation, migration and induction of apoptosis of human cervical cancer cells Hela cells.
CN202211500660.7A 2022-11-28 2022-11-28 Application of cyclovirobuxine D in preparing medicament for treating cervical cancer Pending CN116159067A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202211500660.7A CN116159067A (en) 2022-11-28 2022-11-28 Application of cyclovirobuxine D in preparing medicament for treating cervical cancer

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202211500660.7A CN116159067A (en) 2022-11-28 2022-11-28 Application of cyclovirobuxine D in preparing medicament for treating cervical cancer

Publications (1)

Publication Number Publication Date
CN116159067A true CN116159067A (en) 2023-05-26

Family

ID=86418963

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202211500660.7A Pending CN116159067A (en) 2022-11-28 2022-11-28 Application of cyclovirobuxine D in preparing medicament for treating cervical cancer

Country Status (1)

Country Link
CN (1) CN116159067A (en)

Similar Documents

Publication Publication Date Title
CN106580987B (en) The purposes of dihydro palmatine anti-ulcerative colitis
US11077126B2 (en) Use of carrimycin and pharmaceutically acceptable salts of carrimycin in manufacturing medicament for treating and/or preventing tumor
RU2554497C2 (en) Composition for injections containing hydroxychloroquine for local application in treating cancer
US20200163984A1 (en) Use of isovaleryl spiramycin i, ii and/or iii in manufacturing medicament for treating and/or preventing tumor, and medicament
CN113143913A (en) Application of eudesmane type sesquiterpene compound in preparation of anti-pancreatic cancer drugs
CN110563679B (en) Sesquiterpene lactone compound, preparation method thereof and application of sesquiterpene lactone compound in preparation of medicine for preventing and treating nasopharyngeal carcinoma
CN116159067A (en) Application of cyclovirobuxine D in preparing medicament for treating cervical cancer
CN116407531A (en) Application of anhydroicaritin in preparation of drugs for inhibiting macrophage iron death to treat atherosclerosis
CN117159560A (en) Application of cyclovirobuxine D in preparing medicament for treating breast cancer
CN112402413B (en) Application of lindley eupatorium herb sesquiterpene lactone B in preparation of anti-liver cancer medicine and anti-liver cancer medicine
CN111000851B (en) Application of tanshinone I in preparation of tumor cell autophagy inducer drug
CN110327317B (en) Application of alkannin in preparing medicine for resisting rotavirus infection
CN108451905B (en) Gambogic acid nanoemulsion preparation and preparation method and application thereof
CN109223801B (en) Novel gastric cancer tumor stem cell killing agent and application thereof
CN113318112A (en) Anti-tumor medicine composition and application thereof
CN113750083B (en) Application of metformin in preparation of medicine for treating hand-foot-and-mouth disease
CN113908148B (en) Application of nobiletin in preparation of anti-cholangiocarcinoma drugs
US10004713B2 (en) Uses of chlorogenic acid in the preparation of medicaments for treatment of oligodendroglioma
CN114796114B (en) Antitumor drug micelle and preparation method and application thereof
CN112972455B (en) Application of compound in preparation of antitumor drugs
CN110974826B (en) Application of ouabain or 12-epi-ouabain in preparation of medicine for treating leukemia
CN110283138B (en) Compound, preparation method of compound, application of compound and product applying compound
CN111729074B (en) Application of polypeptide RL25 in preparation of antitumor drugs
CN115737817A (en) Application of tyrosine kinase inhibitor in preparation of echinococcosis treatment drug
CN117442599A (en) Application of coltsfoot ketone in preparation of medicine for treating breast cancer

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