CN113024619B - A Pimenta officinalis leaf residue extract after distillation, and its extraction method and anti-tumor application - Google Patents

A Pimenta officinalis leaf residue extract after distillation, and its extraction method and anti-tumor application Download PDF

Info

Publication number
CN113024619B
CN113024619B CN202110269198.3A CN202110269198A CN113024619B CN 113024619 B CN113024619 B CN 113024619B CN 202110269198 A CN202110269198 A CN 202110269198A CN 113024619 B CN113024619 B CN 113024619B
Authority
CN
China
Prior art keywords
compound
column
cells
extract
cinnamomum
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
CN202110269198.3A
Other languages
Chinese (zh)
Other versions
CN113024619A (en
Inventor
李明乾
李飞
李群
陈观平
陈嘉斌
柴可群
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.)
Sichuan Normal University
Tongde Hospital of Zhejiang Province
Original Assignee
Sichuan Normal University
Tongde Hospital of Zhejiang Province
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 Sichuan Normal University, Tongde Hospital of Zhejiang Province filed Critical Sichuan Normal University
Priority to CN202110269198.3A priority Critical patent/CN113024619B/en
Publication of CN113024619A publication Critical patent/CN113024619A/en
Application granted granted Critical
Publication of CN113024619B publication Critical patent/CN113024619B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H17/00Compounds containing heterocyclic radicals directly attached to hetero atoms of saccharide radicals
    • C07H17/04Heterocyclic radicals containing only oxygen as ring hetero atoms
    • C07H17/06Benzopyran radicals
    • C07H17/065Benzo[b]pyrans
    • C07H17/07Benzo[b]pyran-4-ones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H1/00Processes for the preparation of sugar derivatives
    • C07H1/06Separation; Purification
    • C07H1/08Separation; Purification from natural products
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biotechnology (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Plant Substances (AREA)

Abstract

A targeted antitumor compound extracted from leaves residue of distilled Cinnamomum zeylanicum leaves, and its extraction method and application in preparing antitumor drugs are provided.

Description

Cinnamomum cassia distilled leaf residue extract, and extraction method and anti-tumor application thereof
[ technical field ] A method for producing a semiconductor device
The invention belongs to the technical field of medicines. The invention relates to a targeted antitumor compound extracted from leaf residues of distilled cinnamomum japonicum leaves, and also relates to an extraction method of the compound, an antitumor application of the compound and an action target signal path.
[ background of the invention ]
Cinnamomum cassia (Cinnamomum pauciflorum), also known as Cinnamomum cassia, Sanlianjin, Pelargonium graveolens and Cinnamomum minus, is a plant of Lauraceae, and can extract aromatic oil from leaves, branches, trunk and bark, and the leaf has the highest oil content and is a plant with the highest safrole. After cinnamomum longepaniculatum leaves are subjected to steam distillation to obtain sassafras oil, about 10 million tons of distilled leaf residues exist each year, the leaf residues are wasted in resources and pollute the environment, the cost of waste treatment of enterprises is increased, and the comprehensive utilization and development of the cinnamomum longepaniculatum industry are limited.
Tumors are important diseases threatening the global human health. In 2020, 1929 ten thousand new cases and 996 ten thousand dead cases all over the world exist, while 457 thousand new cases (accounting for 23.7 percent of the world) and 300 thousand dead cases (accounting for 30 percent of the world) exist in China. The natural product is an important source for finding the antitumor drug and the lead compound. The use of the cinnamomum japonicum as a medicine has already been recorded for more than one thousand years, and the efficacy and the main treatment of the cinnamomum japonicum are recorded in Chinese herbal medicine: regulating qi to alleviate pain, promoting blood circulation and dredging collaterals. It is used to treat various pain syndromes, traumatic injury … … ". The volatile oil of cinnamomum japonicum leaves and the safrole which is the main component thereof have stronger inhibition effect on the growth of liver cancer cells, also have the effects of treating burn, resisting bacteria and the like, and simultaneously the deoiled leaf residue alcohol extract also has the antibacterial effect.
[ summary of the invention ]
The invention aims to improve the industrial added value of the cinnamomum japonicum and the utilization rate of the leaf residues.
The invention solves another technical problem of providing a targeted antitumor compound.
The invention establishes a rapid pollution-free extraction method of the leaf residue of the distilled cinnamomum japonicum leaves, and obtains a compound (compound YC) with a targeted anti-tumor effect, which is shown in a formula 1.
Figure BDA0002973491030000011
The compound of the formula 1 acts on a cAMP/RAPGEF3/PDE3B/Rap-1 complex to inhibit a cAMP-mediated Rap-1 signal channel, thereby inhibiting the proliferation and the metastasis of tumor cells.
The present invention also provides a method for preparing a compound of formula 1, comprising the steps of:
A. crushing the oil-removed leaves of the cinnamomum japonicum into powder of 50-120 meshes by using a high-speed multifunctional crusher, soaking and extracting the powder on an absolute ethyl alcohol shaking table overnight, centrifuging the extracted cinnamomum japonicum leaves at 3000rpm for 15min, and collecting the supernatant to obtain the cinnamomum japonicum ethyl alcohol extract.
B. Concentration: the brown powder obtained after the crude extract of the cinnamomum japonicum is processed by a centrifugal concentrator is prepared into an extract with the concentration of 100mg/mL by absolute ethyl alcohol.
C. Separating effective parts: separating effective components by using macroporous adsorption resin or C18 column;
D. and (3) monomer purification: and C18 filler, mobile phase acetonitrile: and (3) water is 55:45, the wavelength is 275nm, the product preparation section is used for preparing refined products again to obtain a qualified section, and the refined products are used for preparing mobile phase acetonitrile: water 55: 45; the collected fractions were concentrated under reduced pressure at 40 ℃ to acetonitrile free, lyophilized to give a solid, and vacuum dried at room temperature to give the compound of formula 1.
In the step A, the ratio of the cinnamomum japonicum deoiled leaf residue powder in grams to the absolute ethyl alcohol in milliliters is 1:8-15, or 1: 9-12, or 1: 9.6-10.4.
In the step A, the soaking time is not more than 12 hours, and the soaking temperature is 25-35 ℃.
Separating with C18 column in step C, specifically comprising binding the extract obtained in step B on C18 column for 3min, washing three times with ddH 2 Washing the C18 separating column with O, 50% ethanol and 100% ethanol, and collecting the distilled 50% ethanol component.
And C, separating by using macroporous adsorption resin, specifically comprising the step of adsorbing the extract obtained in the step B by using a macroporous adsorption resin column at the flow rate of 1.5-2.5 BV/h.
The invention also provides application of the compound shown in the formula 1 in preparing an anti-tumor medicament.
In the application of preparing antitumor drugs, the compound shown in the formula 1 acts on cAMP/EPAC-1/PDE3B/Rap-1 complex.
In the application of preparing the antitumor drugs, the compound shown in the formula I inhibits EPAC-1, RAP-1 or simultaneously inhibits EPAC-1 and RAP-1, inhibits the phosphorylation of the downstream signal path PI3K/AKT/mTOR and RAF-1/ERK genes of an EPAC-1/RAP-1 complex and promotes the expression of apoptosis related genes.
In the application of preparing the antitumor drugs, the tumors are selected from intestinal cancer, lung cancer, liver cancer, melanoma and pancreatic cancer.
The beneficial effects obtained by the invention are as follows: a targeted antitumor compound is extracted from the oil-removed leaf residue of cinnamomum japonicum, and the medicine effect and target spot analysis are carried out, and the compound acts on a cAMP/EPAC-1/PDE3B/Rap-1 complex, so that the proliferation and the metastasis of tumor cells are inhibited. The compound has strong inhibiting effect on intestinal cancer, lung cancer, liver cancer, melanoma and pancreatic cancer cells.
The invention also establishes a rapid pollution-free method for extracting the antitumor compound, and improves the industrial added value of the cinnamomum japonicum and the utilization rate of the leaf residues.
[ description of the drawings ]
FIG. 1: the deoiled leaf residue alcohol extract has inhibitory effect on different cancer cells.
FIG. 2: the inhibition effect of different separation parts on pancreatic cancer cells BXCP-3.
FIG. 3: and (3) separating 50% of alcohol extraction component monomers.
FIG. 4: the compound YC has the inhibition effect on BXCP-3.
FIG. 5: effect of compound YC on clonogenic of BXCP-3 cells.
FIG. 6: EDU proliferation assay of compound YC on BXCP-3 cells.
FIG. 7: the compound YC has the inhibition effect on AA/NE/IGF-1/PAF/VEGF and other tumor growth promoting factors.
FIG. 8: effect of compound YC on BXCP-3 transferability.
FIG. 9: the effect of compound YC on the apoptosis and cycle of BXCP-3 cells.
FIG. 10: effect of compound YC on BXCP-3 cell protein expression.
FIG. 11: effect of compound YC on BXCP-3 cell AKT protein phosphorylation and cell localization.
FIG. 12: the potential target of compound YC is the EPAC-1/PDE3B complex.
FIG. 13: the compound YC has antitumor effect in vivo.
[ detailed description ] embodiments
The invention will be better understood from the following examples.
Example 1: discovery of antitumor activity of deoiled leaf residue
Crushing the oil-removed leaves of the cinnamomum japonicum into powder of 50-120 meshes by using a high-speed multifunctional crusher, soaking and extracting the powder on an absolute ethyl alcohol shaking table overnight, centrifuging the extracted cinnamomum japonicum leaves at 3000rpm for 15min, and collecting the supernatant to obtain the cinnamomum japonicum ethyl alcohol extract. Human hepatoma cells HepG2, HUH7, human lung cancer cells A549, melanoma cells B16 and human pancreatic cancer cells BXPC-3 are used as test cell strains to screen the antitumor activity of the test cell strains.
As shown in fig. 1: the cinnamomum japonicum crude extract has an inhibiting effect on lung cancer cells, liver cancer cells, melanoma cells and pancreatic cancer cells, wherein the inhibiting effect on proliferation of human pancreatic cancer cells BXPC-3 is the best.
Example 2: isolation of the antitumor active Compounds
In order to verify the effective components of the crude extract of the cinnamomum japonicum. The different fractions of the crude extract of cinnamomum japonicum were separated by the following method and the effect of all the separated fractions on the cell proliferation activity of human pancreatic cancer cells BXPC-3 was determined.
(1) Concentration: the brown powder obtained after the crude extract of the cinnamomum japonicum is processed by a centrifugal concentrator is prepared into an extract with the concentration of 100mg/mL by absolute ethyl alcohol.
(2) Separation: combining the extract on C18 column for 3min using C18 column, washing three times, collecting distillate (First), and adding ddH 2 The C18 separation column was washed with O, 50% ethanol (50%), and 100% ethanol (100%), and the distillate fractions were collected. The collected distillate fractions were dried as described above and re-dissolved in anhydrous ethanol to their original volume. For the cinnamool extract, it was found that the effect of the distilled 50% fraction was close to that of the stock solution (Stoste). The effective components of the alcohol extract are presumed to be concentrated in 50 percent of the components. The inhibition of pancreatic cancer cells BXCP-3 by different isolated sites is shown in FIG. 2.
The separation step can also be carried out by adopting macroporous adsorption resin, and specifically comprises that the extract obtained by concentration is adsorbed by a macroporous adsorption resin column at the flow rate of 1.5-2.5 BV/h.
(3) And (3) monomer purification: about 1g of the starting material was dissolved with 50% acetonitrile to give about 300ml of a green-brown liquid, prepared with preparative high performance liquid phase, C18 filler, mobile phase acetonitrile: and (3) water is 55:45, the wavelength is 275nm, the product preparation section is used for preparing refined products again to obtain a qualified section, and the refined products are used for preparing mobile phase acetonitrile: water 55: 45. The collected fractions were concentrated under reduced pressure at 40 ℃ until acetonitrile-free, freeze-dried to give a solid, and vacuum-dried at room temperature. The monomer shown in FIG. 3 was obtained. Detecting the liquid phase condition on line: acetonitrile: water 45:55, flow rate 1ml/min, column temperature: the column, Agilent ZORBAX SB-C18(4.6 x 250mm,5 μm) at 275nm was used at room temperature.
Example 3: structural identification of compounds
The compound is a light yellow powder, and is identified as (-) -HR-ESI-MS M/z 723.1712[ M-H ] -, (+) -HR-ESI-MS M/z 747.1695[ M + Na ] +, which indicates that the molecular composition is C39H32O14(calcd for C39H31O14,723.1714; calcd for C39H32O14Na,747.1690) through mass spectrum. In the 1H NMR spectrum, 2 sets of coumaroyl signals of formula E configuration are shown [ δ H7.63 (1H, d, J ═ 16.2Hz), 7.50(1H, d, J ═ 15.6Hz), 6.30(1H, d, J ═ 16.2Hz), 6.20(1H, d, J ═ 15.6Hz) ]; a set of tetrasubstituted benzene signals [ δ H6.42 (1H, d, J ═ 1.8Hz), 6.23(1H, d, J ═ 1.8Hz) ]; a group of para-substituted benzene signals δ H [7.85(2H, d, J ═ 9.0Hz), 6.78(2H, d, J ═ 9.0Hz) ], suggesting that in addition to containing two p-coumaroyl groups in the molecular structure, the B ring of the flavone parent nucleus is also para-substituted. δ H5.80 (1H, d, J ═ 1.8Hz), which is the terminal hydrogen signal of the saccharide, gives a methyl hydrogen signal in the high field portion, δ H0.85 (3H, d, J ═ 6.6Hz), suggesting that the structure may contain an α -rhamnose.
The 13C NMR spectrum of the compound shows 39 carbon signals, the hydrocarbon signals are completely assigned in combination with HSQC, and the 1H NMR and 13C NMR data of the compound are basically consistent with the literature reports, so the structure of the compound is preliminarily presumed to be Kaempferol-3-O- (3 ', 4' -di-E-p-coumaroyl) -alpha-L-rhamnopyranoside.
The structure of the compound was further confirmed by HMBC. H-6 is related to C-5, C-8 and C-10, and H-2'/6' is related to C-2, so that the structure of the flavone part is confirmed. H-1 ' is related to C-3 ' and C-5 ', and the structure of the rhamnose part is confirmed. H-7 'is related to C-2', C-6 ', C-8', C-9 ', H-7' is related to C-2 ', C-6', C-8 ', C-9', H-8 'is related to C-1', H-8 'is related to C-1', and the structures of two p-coumaroyl parts are confirmed. HMBC shows key relevant signals: h-3 is related to C-9 ', H-4 is related to C-9', and two p-coumaroyl groups are respectively connected at the C-3 position and the C-4 position. In addition, no signal related to H-1' and C-3 appears, probably because the dihedral angle is 90 degrees, the flavone part of the compound is a kaempferol mother nucleus, when the C-3 position is not connected with sugar, the chemical shifts of the C-2 and C-3 positions of the kaempferol are delta C148.1 and 137.8, but the chemical shifts of the C-2 and C-3 positions of the compound are delta C159.3 and 134.8, which accords with the characteristic of forming glycoside at the C-3 position, and therefore, the rhamnose is connected at the C-3 position. Finally, the structure of the compound is Kaempferol-3-O- (3 ', 4' -di-E-p-coumaroyl) -alpha-L-rhamnopyranoside (formula 1).
Example 4: antitumor Activity of Compounds
The inhibition effect of separated Kaempferol-3-O- (3 ', 4' -di-E-p-coumaroyl) -alpha-L-rhamnopyranoside (compound YC) on five different pancreatic cancer cells such as BXPC-3, SW1990, PANC-1, CFPAC-1, ASPC-1 and the like is researched, and the inhibition effect of YC on cells such as intestinal cancer HCT116, SW480, non-small cell lung cancer H1975, HCC827, A549, liver cancer HEPG2, HEP1-6, HUH7 and the like is screened. The results show that: YC has the most obvious effect of inhibiting the human pancreatic cancer cell strain BXPC3, and a human pancreatic cancer cell strain BXPC-3 is selected as a tested cell strain in subsequent related experiments, and the influence of Clone and EDU on the proliferation of the BXPC-3 cell strain is researched.
(1) CCK8 cell proliferation assay: pancreatin digestion of cells, counting by a blood cell counting plate, seeding BXPC-3 cells in a 96-well plate in a system of 5000 cells/well/100 uL, and placing in an incubator for overnight culture for 24H; after 24H, changing the culture medium (containing 10% FBS), adding YC with different concentrations into the BXPC-3 cells respectively, and treating for 12, 24, 48 and 72H respectively; adding 10uLCCK8 into each well, detecting the absorbance within 4h by using a microplate reader, counting IC50 processed by YC at different time points, and displaying that the inhibition effect of YC on the pancreatic cancer BXPC-3 cell line has time and dose dependence as shown in figure 4; the IC50 was 14.44 ug/mL for 72 h.
IC50 values were calculated for each cell as shown in table 1:
Figure BDA0002973491030000051
Figure BDA0002973491030000061
(2) colony formation assay
Pancreatin digestion of cells, counting by a blood cell counting plate, seeding BXPC-3 normal cells in a system of 1000 cells/hole/2000 uL, and putting the cells into an incubator for 24 hours at night; after 24h, the culture medium (containing 10% FBS) is replaced, YC with different concentrations is respectively added into BXPC3 cells, after 15 days of treatment, the culture medium is removed, methanol is fixed for 20min, 0.1% crystal violet solution is used for staining for 20min, and PBS is used for washing out the crystal violet solution to take pictures. The effect of compound YC on the clonogenic behavior of BXCP-3 cells was dose-dependent, with 10ug/mL compound YC forming less cell clonogenic clumps, as shown in figure 5.
(3) EDU cell proliferation assay
Pancreatin digestion cells, counting by a blood cell counting plate, inoculating BXCP-3 cells in a system of 10000 cells/hole/100 uL, and putting the cells into an incubator for overnight culture for 24 hours; after 24H, the medium (containing 10% FBS) was changed, YC was added to BXCP-3 cells at a concentration gradient of 10, 5, 0ug/mL, 50uL of medium was aspirated after 24H treatment, 50uL of EDU-containing medium was added thereto, the incubator was incubated for 2H, and subsequent experiments were performed according to the reagent instructions. EDU proliferation assay of compound YC on BXCP-3 cells, newly proliferated BXCP-3 cells were significantly less than 10ug/mL treated and untreated groups after 10ug/mL treatment with compound YC, as shown in fig. 6.
(4) YC inhibiting tumor cell growth factor
Through experimental verification, Arachidonic Acid (AA)/Neurotensin (NE)/recombinant human insulin-like growth factor 1 (IGF-1)/platelet factor (PAF)/Vascular Endothelial Growth Factor (VEGF) are found to promote the proliferation of BXPC-3 cells, and through the combined administration of the polypeptide and YC, the YC is found to inhibit the proliferation caused by the growth factors. The compound YC has the inhibitory effect on tumor growth promoting factors such as AA/NE/IGF-1/PAF/VEGF, and the like, as shown in figure 7.
(5) Cell invasion and wound healing experiments
Cells were trypsinized, resuspended in serum-free medium, counted on a hemacytometer plate, seeded into the chamber in a5 × 104/well/200 uL system, 650uL of medium containing 10% FBS was added to the lower layer, and YC was added to the chamber at 0, 5ug/mL, 10ug/mL, 20 ug/mL. Putting into an incubator for 48 h. After 48h, the chamber was removed, fixed in methanol for 20min, washed in PBS, stained with 0.1% crystal violet for 20min, washed in PBS and then swabbed off the cells in the chamber with a cotton swab, and photographed under an inverted microscope, as shown in FIG. 8.
Wound healing experiments: cells were trypsinized, counted on a hemacytometer, plated in a six-well plate with a mold in a 4X 105/well/2000 uL system, and cultured in an incubator for 24 hours. Removing the mold after culturing for 24h, replacing culture medium (10% FBS), and adding YC with final concentration of 0, 2.5ug/mL, 5ug/mL, 10 ug/mL; the time of drug addition was taken as 0h, and after 24h of incubation, it was seen that the invasion ability and wound healing ability were weakened as the concentration of YC was increased, as shown in FIG. 8.
(6) Flow detection of apoptosis and cycle
Planting cells into a six-well plate in a system of 30 ten thousand per well/2 mL in the first day, culturing for 24H overnight, replacing a culture medium after 24H, and adding YC with final concentration of 40ug/mL, 20ug/mL, 10ug/mL, 5ug/mL, 2.5ug/mL and 0 ug/mL; and (3) treating the 24H pancreatin for digestion, centrifuging at 1000rpm/min for 5min, collecting dead and live cells by apoptosis, and periodically collecting the live cells. Staining according to apoptosis and cycle kit; the results are as follows: YC apparently caused apoptosis with little effect on cycle as shown in fig. 9.
(7) Effect of Signaling pathway protein expression
Planting 40 million cells per well into a six-well plate in the first day, and adding YC of 20, 15, 10 and 0ug/mL after 24 hours; and after 24 hours of treatment, protein is collected and quantified. Protein loading concentration: 30ug/10uL, separation gel concentration: 8%/12%, electrophoresis conditions: 90V, 130V, and the rotating die condition: constant pressure 100V, 90 min. The influence of the compound YC on the expression of BXCP-3 cell protein, the compound YC can obviously inhibit the phosphorylation of the downstream signal path PI3K-AKT-mTOR and ERK signal path of EPAC-1/PDE3B/RAP1 complex, and promote the expression of related genes of an apoptosis signal path caspase family and the like, as shown in figure 10.
(8) Indirect immunofluorescence
Trypsinizing the cells, counting by a hemocytometer, seeding BXPC3 cells in a system of 2X104 cells/200 uL in 48-well plates, placing in an incubator for 24H overnight; after 24H, changing the culture medium (containing 10% FBS), adding YC with different concentrations of 0, 10 and 15ug/mL into BXPC3 cells respectively, and treating the 24H; the medium was aspirated off, fixed in methanol for 15min, permeabilized for 15min, blocked with 3% BSA for 30min, incubated at room temperature for 1h for the first antibody, incubated at fluorescence for 1h for the second antibody, stained with DAPI for 20min, mounted, and photographed under confocal microscope. The effect of compound YC on BXCP-3 cell AKT protein phosphorylation and cell localization decreased the level of AKT phosphorylation and phosphorylated AKT nuclear entry, as shown in fig. 11.
Example 5: target of action of the compounds
In order to determine the potential target of YC, experiments such as WB, co-immunoprecipitation, flow cytometry and the like find that the cAMP/EPAC-1/PDE3B/Rap-1 complex is the potential target of YC acting on BXPC-3. When YC was acted on BXPC-3, both EPAC-1 and RAP-1 were decreased, as shown in FIG. 10. We found that by immunoprecipitating RAP-1 and EPAC-1, activated RAP-1, and PI3K γ 110 bound to EPAC-1 decreased, while PDE3B increased. After the EPAC-1 and the PDE3B are knocked down, apoptosis is detected through flow cytometry, and the apoptosis is increased after the EPAC-1 is knocked down and the apoptosis is reduced after the PDE3B is knocked down, which is consistent with the result of immunoprecipitation. And CCK8 results were consistent with apoptotic results, as shown in fig. 12.
Example 6: results of in vivo animal experiments
At 1.5x10 6 100 uL/the preparation is inoculated to nude mice subcutaneously when subcutaneous tumor is about 100mm 3 Drugs were given 5d at times, 5 per group, into 3 groups: a model group, a drug high concentration group, a drug low concentration group; weighing every day after administration, measuring tumor size, taking tumor tissue after administration for 5 days, and detecting P-AKT and Ki67 by immunohistochemistry; extracting tissue protein and quantitatively detecting related protein: as a result, the high concentration group significantly inhibited tumor growth, and immunohistochemistry showed that the administered group had decreased cell proliferation and decreased AKT phosphorylation compared to the model group, as shown in fig. 13.

Claims (1)

1. A method for extracting a compound shown as a formula I, which is characterized in that the compound is quickly separated from waste cinnamomum japonicum deoiled leaf residues, and the extraction method comprises the following steps:
Figure 100001-1657789256930-1
formula I
A. Crushing the oil-removed leaves of the cinnamomum japonicum into powder of 50-120 meshes by using a high-speed multifunctional crusher, soaking and extracting the powder overnight on an absolute ethyl alcohol shaking table, centrifuging the extracted cinnamomum japonicum leaves at 3000rpm for 15min, and collecting supernatant to obtain an cinnamomum japonicum ethyl alcohol extract;
B. and (3) concentrating: preparing brown powder obtained by passing the crude extract of cinnamomum japonicum through a centrifugal concentrator into an extract with the concentration of 100mg/mL by using absolute ethyl alcohol;
C. separation of effective parts: separating effective components by using macroporous adsorption resin or C18 column;
D. and (3) monomer purification: and C18 filler, mobile phase acetonitrile: and (3) water is 55:45, the wavelength is 275nm, the product preparation section is used for preparing refined products again to obtain a qualified section, and the refined products are used for preparing mobile phase acetonitrile: water 55: 45; concentrating the collected part at 40 deg.C under reduced pressure until no acetonitrile is present, freeze-drying to obtain a solid, and vacuum-drying at room temperature to obtain the compound of formula 1; wherein the ratio of the cinnamomum micranthum deoiled leaf residue powder in grams to the absolute ethyl alcohol in milliliters in the step A is 1: 8-15; wherein the soaking time in the step A is not more than 12 hours, and the soaking temperature is 25-35 ℃; when the C18 column is used for separation in step C, the method comprises combining the extract obtained in step B on C18 column for 3min, washing three times, washing C18 column with ddH2O, 50% ethanol and 100% ethanol respectively, and collecting distillate 50% ethanol fraction; or when the step C is separated by using macroporous adsorption resin, the extraction liquid obtained in the step B is adsorbed by a macroporous adsorption resin column at the flow rate of 1.5-2.5 BV/h.
CN202110269198.3A 2021-03-12 2021-03-12 A Pimenta officinalis leaf residue extract after distillation, and its extraction method and anti-tumor application Active CN113024619B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110269198.3A CN113024619B (en) 2021-03-12 2021-03-12 A Pimenta officinalis leaf residue extract after distillation, and its extraction method and anti-tumor application

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110269198.3A CN113024619B (en) 2021-03-12 2021-03-12 A Pimenta officinalis leaf residue extract after distillation, and its extraction method and anti-tumor application

Publications (2)

Publication Number Publication Date
CN113024619A CN113024619A (en) 2021-06-25
CN113024619B true CN113024619B (en) 2022-09-09

Family

ID=76470034

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110269198.3A Active CN113024619B (en) 2021-03-12 2021-03-12 A Pimenta officinalis leaf residue extract after distillation, and its extraction method and anti-tumor application

Country Status (1)

Country Link
CN (1) CN113024619B (en)

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101218340B1 (en) * 2010-07-13 2013-01-03 이지훈 Composition comprising extracts or fractions of specific plants, use thereof and process for preparing the extracts
CN103223018B (en) * 2013-05-08 2015-09-30 浙江大学 Flos Osmanthi Fragrantis polyphenol extract and its production and use
CN103446294B (en) * 2013-09-18 2015-06-24 海南华拓天涯制药有限公司 Murrayae folium ET Cacumen extract preparation method, Murrayae folium ET Cacumen extract obtained thereby and application thereof
CN109232681A (en) * 2018-11-08 2019-01-18 佳木斯大学 The separation of verbascoside and identification method in a kind of sweet osmanthus

Also Published As

Publication number Publication date
CN113024619A (en) 2021-06-25

Similar Documents

Publication Publication Date Title
Xu et al. Immunomodulatory mechanism of a purified polysaccharide isolated from Isaria cicadae Miquel on RAW264. 7 cells via activating TLR4-MAPK-NF-κB signaling pathway
Liu et al. Tumor inhibition and improved immunity in mice treated with flavone from Cirsium japonicum DC
CN101647842B (en) Fructus Podophylli extract and application thereof
Cao et al. Ginkgo biloba exocarp extracts induces apoptosis in Lewis lung cancer cells involving MAPK signaling pathways
Zhang et al. Sesquiterpenes and polyphenols with glucose-uptake stimulatory and antioxidant activities from the medicinal mushroom Sanghuangporus sanghuang
CN108129295A (en) A kind of Diterpene derivative and its pharmaceutical composition and purposes
CN101648934B (en) Isopentenyl flavone and application thereof
Li et al. Transformation mechanism of rare ginsenosides in American ginseng by different processing methods and antitumour effects
CN113024619B (en) A Pimenta officinalis leaf residue extract after distillation, and its extraction method and anti-tumor application
CN1775267A (en) Kaikoujian extract, Its preparing method and use
CN101028322A (en) Use of Maoliefengdou extract for preparing anti-cancer medicine
CN111808153B (en) Monoterpene glycoside compound and application thereof in preparation of anti-inflammatory drugs
CN104173390A (en) Use of a flavanol glycoside for suppressing activation of stellate cells
CN100528870C (en) Internal ester monomer compound and its application in preparing anti cancer medicine
CN103610682A (en) Preparation method of 3(alpha)-hydroxyl-30-olive-12,20(29)-diene-28-acid and application in preparing anti-tumor drug
CN103301167B (en) Application of babysbreath isoorientin for preparing medicines for treating alcoholic liver injury
CN108948040B (en) Gilmaxane type sesquiterpene compound extracted from herba Centellae and application thereof
JP2015515993A (en) Pharmaceutical composition containing human cyclooxygenase and doxorubicin or doxorubicins, process for its preparation and use in the manufacture of multiple drugs
CN112939912A (en) Preparation method and application of lactucin extracted from cichorium intybus
CN111892634A (en) Glucoside compound from raw materials, extraction method and application thereof
CN108997473A (en) A kind of non-sea cucumber alkane type selenka and the preparation method and application thereof
US20090022827A1 (en) Agent and method for eliminating malignance of cancer cells without harmful effect to normal cells
Peng et al. Antitumor effect of the ethanol extract of Scutellaria baicalensis on the mice bearing U14 cervical cancer
CN111494397B (en) Application of ophiopogonin compounds in preparing medicines for preventing and treating tumors
CN108014119B (en) Application of phenylpropanoid glycoside Smiglaside A in preparation of medicine for treating sepsis

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