CN114831961B - Liver-targeted bionic cell membrane drug-loaded nano-particle and preparation method and application thereof - Google Patents

Liver-targeted bionic cell membrane drug-loaded nano-particle and preparation method and application thereof Download PDF

Info

Publication number
CN114831961B
CN114831961B CN202210347880.4A CN202210347880A CN114831961B CN 114831961 B CN114831961 B CN 114831961B CN 202210347880 A CN202210347880 A CN 202210347880A CN 114831961 B CN114831961 B CN 114831961B
Authority
CN
China
Prior art keywords
cell membrane
drug
liver
targeted
loaded
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
CN202210347880.4A
Other languages
Chinese (zh)
Other versions
CN114831961A (en
Inventor
周天华
刘祥瑞
刘子墨
李琪
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.)
Zhejiang University ZJU
Original Assignee
Zhejiang University ZJU
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 Zhejiang University ZJU filed Critical Zhejiang University ZJU
Priority to CN202210347880.4A priority Critical patent/CN114831961B/en
Publication of CN114831961A publication Critical patent/CN114831961A/en
Application granted granted Critical
Publication of CN114831961B publication Critical patent/CN114831961B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/5176Compounds of unknown constitution, e.g. material from plants or animals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/20Carboxylic acids, e.g. valproic acid having a carboxyl group bound to a chain of seven or more carbon atoms, e.g. stearic, palmitic, arachidic acids
    • A61K31/203Retinoic acids ; Salts thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1793Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5146Organic macromolecular compounds; Dendrimers obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyamines, polyanhydrides
    • A61K9/5153Polyesters, e.g. poly(lactide-co-glycolide)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5192Processes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70575NGF/TNF-superfamily, e.g. CD70, CD95L, CD153, CD154
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N13/00Treatment of microorganisms or enzymes with electrical or wave energy, e.g. magnetism, sonic waves
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/067Hepatocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2509/00Methods for the dissociation of cells, e.g. specific use of enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2509/00Methods for the dissociation of cells, e.g. specific use of enzymes
    • C12N2509/10Mechanical dissociation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15041Use of virus, viral particle or viral elements as a vector
    • C12N2740/15043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector
    • 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

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Zoology (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Engineering & Computer Science (AREA)
  • Nanotechnology (AREA)
  • Microbiology (AREA)
  • Optics & Photonics (AREA)
  • Cell Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Biophysics (AREA)
  • Molecular Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Botany (AREA)
  • Toxicology (AREA)
  • Virology (AREA)
  • Plant Pathology (AREA)
  • Medicinal Preparation (AREA)

Abstract

The invention provides a liver-targeted bionic cell membrane drug-loaded nanoparticle, a preparation method and application thereof, wherein the outside of the drug-loaded nanoparticle is coated by an engineering cell membrane, and an anti-liver fibrosis drug is encapsulated in the drug-loaded nanoparticle for treating liver fibrosis. The engineering cell membrane selects LX2 cells (human hepatic stellate cells) as source cells, and specifically expresses tumor necrosis factor related apoptosis-inducing ligand (TRAIL) protein through Lv-TRAIL-zsgreen over-expression lentivirus transfection. The drug-loaded nano-particles consist of lactic acid-glycolic acid copolymer (PLGA) and all-trans retinoic acid (ATRA), and the ATRA and TRAIL can exert synergistic effect to induce hepatic fibroblast apoptosis. The bionic cell membrane nano particles are mainly concentrated in liver tissues after entering the body, and have remarkable treatment effect in anti-hepatic fibrosis treatment, realize targeted delivery and efficient synergistic effect, and have wide application prospects.

Description

Liver-targeted bionic cell membrane drug-loaded nano-particle and preparation method and application thereof
Technical Field
The invention belongs to the field of pharmaceutical preparations, and in particular relates to liver-targeted bionic cell membrane drug-loaded nano-particles, and a preparation method and application thereof.
Background
Activation of hepatic stellate cells (Hepatic Stellate Cells, HSC) is an effector of liver fibrosis and thus its continued progression leading to the development of cirrhosis and hepatocellular carcinoma. As the most important mesenchymal cells in the liver, HSCs are normally in a resting state in the liver, storing vitamin a in the form of retinol esters. Following liver injury, HSCs activate extracellular matrix such as alpha-smooth muscle actin (alpha-smooth muscle actin, alpha-SMA) and I, III type collagen fibers. The increased number of fibroblasts and excessive deposition of extracellular matrix proteins disrupt the normal structure of the liver further leads to liver fibrosis. Whereas retinoic acid loss may play a potential role in promoting cell activation, related derivatives of vitamin a help prevent liver fibrosis and carcinogenesis. ATRA as one of the activated products of vitamin a can inhibit HSC activation by inhibiting thioredoxin interacting protein expression, thereby reducing its oxidative stress level to play a therapeutic role.
In recent years, a bionic cell membrane nano-drug delivery system gradually realizes innovative application in a plurality of fields such as vaccine preparation, disease targeted therapy and the like. The genetically engineered cell membrane has higher biocompatibility and biological safety, can reduce the serial immune reactions such as opsonin and complement activation caused by exogenous substances entering the body, and effectively avoid capturing drugs by a mononuclear macrophage system. In addition, the engineering cells select LX2 cells, and can express specific proteins on cell membranes, so that the bionic nano-carrier has heterologous targeting characteristics, and simultaneously activates downstream Caspase cascade reaction and NF- κB and JNK/AP signal channels to induce corresponding fibroblast apoptosis through the combination of TRAIL proteins and death receptor DR 5.
The bionic cell membrane drug-loaded nano-particles are different from widely applied red cell membrane coated bionic nano-materials, and the functional therapeutic proteins are expressed on the cell membrane through genetic engineering design except for selection of cell types, so that the extension application of the bionic nano-system is further realized. The liver-targeted bionic cell membrane drug-loaded nano-particles can be used as an effective strategy for treating liver fibrosis, and a new idea is provided for treating tumors.
Disclosure of Invention
The invention aims at overcoming the defects of the prior art and provides a liver-targeted bionic cell membrane drug-loaded nanoparticle, and a preparation method and application thereof.
The aim of the invention is achieved by the following technical scheme:
in a first aspect, the invention provides a preparation method of liver-targeted bionic cell membrane drug-loaded nanoparticles, which comprises the following steps:
(1) Preparing drug-loaded nano-particles: dissolving a lactic acid-glycolic acid copolymer and all-trans retinoic acid in anhydrous dichloromethane, wherein the mass ratio of the lactic acid-glycolic acid copolymer to the all-trans retinoic acid is 8-10:1, using polyvinyl alcohol as an aqueous phase, and preparing the aqueous phase by dissolving the polyvinyl alcohol in deionized water, wherein 4g of the polyvinyl alcohol is added into every 100ml of deionized water; then emulsifying on ice by using probe ultrasound under the condition that the output power is 260-325W and the duration is 5-15min; slowly dripping the white emulsion formed by ultrasonic emulsification into 12-14mL of single distilled water, stirring for 4-5h at room temperature to volatilize dichloromethane and solidify the drug-loaded nano particles; finally, the drug-loaded nano particles are collected after ultrafiltration and centrifugation for 30min at 3000 rpm;
(2) Preparation of LX2cell line stably expressing TRAIL protein: culturing LX2 cells in a logarithmic growth phase until the cells adhere to the wall, then transfecting the LX2 cells by using a culture medium containing Lv-TRAIL-zsgreen over-expression lentivirus, culturing for 24 hours at 37 ℃, replacing the culture medium containing the Lv-TRAIL-zsgreen over-expression lentivirus by using a DMEM culture medium for further culturing for 48 hours, and then performing resistance screening by using a G418 culture medium containing 200 mu mol/L to obtain a LX2cell line which can stably express TRAIL protein by stable transfection;
(3) Preparation of engineered cell membranes: washing the LX2cell line which is prepared in the step (1) and stably expresses TRAIL protein in a phosphate buffer solution at 4 ℃ for three times, re-suspending the cell line by using hypotonic cell lysate, then lysing the cell line on a shaker at 4 ℃ for 4-6 hours, outputting 260-325W of power, and performing ultrasonic treatment on the cell line by a probe for 1-3 minutes; then, differential centrifugation is carried out to extract cell membrane precipitation; finally, re-suspending the proposed cell membrane sediment by using phosphate buffer salt solution, and extruding the cell membrane suspension back and forth from the polycarbonate membrane for 9-13 times by a micro extruder to obtain an engineering cell membrane;
(4) Preparing liver-targeted bionic cell membrane drug-loaded nano-particles: and (3) carrying out ultrasonic treatment on the engineering cell membrane prepared in the step (3), then blending the engineering cell membrane with the drug-carrying nano-particles prepared in the step (1) according to the mass ratio of 1:1, and extruding the blending liquid back and forth from a 200 nm-aperture polycarbonate membrane for 9-12 times through a micro extruder to obtain the liver-targeted bionic cell membrane drug-carrying nano-particles.
Further, the differential centrifugation in step (3) includes: firstly, centrifuging for 10-15min under the centrifugal force of 1,000-1500g and the centrifugal condition of 4 ℃, and taking supernatant; centrifuging supernatant at 10000-12000g centrifugal force and 4deg.C for 30-40min; finally, the mixture is centrifuged for 1 to 3 hours under the centrifugal force of 900000 to 100000g and the centrifugal condition of 4 ℃.
Further, the hypotonic cell lysate comprises a 0.25 x phosphate buffered saline solution, a protease inhibitor, and a phosphatase inhibitor.
Further, the polycarbonate membrane in step (3) has a pore size of 50nm, 100nm, 200nm or 400nm.
Further, the polycarbonate membrane pore size is preferably 200nm.
In a second aspect, the invention provides liver-targeted bionic cell membrane drug-loaded nano-particles prepared by the method.
In a third aspect, the invention provides an application of liver-targeted bionic cell membrane drug-loaded nano-particles in preparing drugs for treating liver fibrosis.
The invention has the beneficial effects that: 1. the invention provides bionic cell membrane drug-loaded nano-particles capable of playing a synergistic targeting role to treat hepatic fibrosis. Liver targeting can be achieved by using LX2 cells as source cells. The therapeutic protein TRAIL expressed on the cell membrane can promote the activation of hepatic fibroblast apoptosis, and the activation inhibition of the drug ATRA on the fibroblast can effectively reduce the promotion of hepatic fibrosis. Meanwhile, the bionic cell membrane drug-loaded nano-particles are conveyed by virtue of a stable membrane structure, so that the residence time of the bionic cell membrane drug-loaded nano-particles in blood circulation can be prolonged, the bionic cell membrane drug-loaded nano-particles smoothly reach target cells, and effective permeation and treatment of drugs are realized;
2. the polymer material used by the engineering cell membrane nano-particles is PLGA, and the PLGA is used as a medical polymer material approved by the United states Food and Drug Administration (FDA) to ensure the safety of a drug delivery system, and has higher clinical application value with the coated cell membrane for reinforcing the biocompatibility.
Drawings
FIG. 1 is a schematic diagram of the preparation and structure of a liver-targeted biomimetic cell membrane drug-loaded nanoparticle;
FIG. 2 is a transmission electron microscope image of a liver-targeted biomimetic cell membrane drug-loaded nanoparticle;
FIG. 3 is a Western blot assay;
FIG. 4 is a graph showing the effect of detecting TM-ATRA/NP and NP, M-NP, ATRA/NP, M-ATRA/NP, and TM-NP induced apoptosis by crystal violet;
FIG. 5 is a graph showing the detection of apoptosis induced by TM-ATRA/NP and TM-NP by classical collagen fiber staining;
FIG. 6 is a graph showing the detection of TM-ATRA/NP and TM-NP-induced apoptosis by sirius red staining;
FIG. 7 is a graph showing the detection of TM-ATRA/NP and TM-NP-induced apoptosis by hematoxylin-eosin staining;
fig. 8 is a schematic diagram of HE staining of tissue sections of different organs to assess treatment safety.
Detailed Description
For the purposes of making the objects, technical solutions and advantages of the present invention more apparent, the present invention will be further described in detail with reference to the accompanying drawings and examples, it being understood that the specific examples described herein are for the purpose of illustrating the present invention only, and not all the examples. All other embodiments, which can be made by those skilled in the art based on the embodiments of the present invention without making any inventive effort, are within the scope of the present invention.
FIG. 1 is a schematic diagram of preparation and structure of a liver-targeted biomimetic cell membrane drug-loaded nanoparticle (TM-ATRA/NP) of the present invention.
The present invention is further described below with reference to the accompanying drawings, in which the embodiments described in the present specification are merely examples of implementation forms of the inventive concept, and the scope of protection of the present invention should not be construed as being limited to the specific forms set forth in the embodiments, but also equivalent technical means as will occur to those skilled in the art based on the inventive concept.
The experimental methods described in the following examples are all conventional methods unless otherwise specified; the reagents and materials, as described herein without any knowledge, are commercially available.
Example preparation of liver-targeting bionic cell membrane drug-loaded nanoparticle (TM-ATRA/NP)
(1) Preparation of drug-loaded nanoparticles (ATRA/NP)
10mg of all-trans retinoic acid (ATRA) and 100mg of lactic-co-glycolic acid (PLGA) were blended in 1mL of anhydrous methylene chloride, and 4% polyvinyl alcohol (PVA) was used as the aqueous phase prepared by dissolving polyvinyl alcohol in deionized water, and 4g of polyvinyl alcohol was added to every 100mL of deionized water; then emulsifying on ice by using probe ultrasound under the condition that the output power is 325W and the power is 720W for 15min; slowly dripping the white emulsion formed by ultrasonic emulsification into 14mL of single distilled water, and stirring for 5 hours at room temperature to volatilize dichloromethane and solidify drug-loaded nano particles (ATRA/NP); finally, the drug-loaded nano particles (ATRA-loaded nanoparticles, ATRA/NP) are collected after ultrafiltration and centrifugation at 3000rpm for 30min;
(2) Preparation of LX2cell line (LX 2-TRAIL-zsgreen) stably expressing TRAIL protein
Culturing LX2 cells in a logarithmic growth phase until the cells adhere to the wall, then transfecting the LX2 cells by using a culture medium containing Lv-TRAIL-zsgreen over-expression lentivirus, culturing for 24 hours at 37 ℃, replacing the culture medium containing the Lv-TRAIL-zsgreen over-expression lentivirus by using a fresh culture medium, continuously culturing for 48 hours, wherein the cell fusion degree in each hole reaches about 90%, and then performing resistance screening by using a G418 culture medium containing 200 mu mol/L to obtain a LX2cell line (LX 2-TRAIL-zsgreen) which can stably express TRAIL protein by stable transfection;
the fresh culture medium is a DMEM culture medium;
the Lv-TRAIL-zsgreen over-expression slow virus is from Shanghai Ji Kai gene medical science and technology Co., ltd, comprises a gene fragment capable of expressing TRAIL protein and a gene fragment capable of expressing zsgreen fluorescent protein.
(3) Preparation of engineered cell membranes (LX 2-zsgreen-trail cell membrane)
Washing the LX2cell line (LX 2-TRAIL-zsgreen) which is prepared in the step (1) and stably expresses TRAIL protein in phosphate buffer saline solution at 4 ℃ for three times, re-suspending the cell line by hypotonic cell lysate, then cracking the cell line on a shaking table at 4 ℃ for 4 hours, and then using a probe with output power of 260W for 1min; then, differential centrifugation is carried out to extract cell membrane precipitation; the differential centrifugation includes: firstly, centrifuging for 10min under the centrifugal force of 1000g and the centrifugal condition of 4 ℃, and taking supernatant; centrifuging supernatant at 10000g centrifugal force and 4deg.C for 30min; finally, under the centrifugal force of 100000g and the centrifugal condition of 4 ℃, the transparent cell membrane sediment is obtained after centrifugation for 1 h; finally, the proposed cell membrane pellet was resuspended in phosphate buffered saline, and the cell membrane suspension was squeezed back and forth 11 times from 200nm pore size polycarbonate membrane by a micro-extruder to give an engineered cell membrane (LX 2-zsgreen-trail cell membrane).
The hypotonic cell lysate comprises 0.25 x phosphate buffer salt solution, protease inhibitor and phosphatase inhibitor.
(4) Preparation of liver-targeted bionic cell membrane drug-loaded nano-particles (TM-ATRA/NP)
And (3) carrying out ultrasonic treatment on 10mg of the engineering cell membrane (LX 2-zsgreen-trail cell membrane) prepared in the step (3), then blending with 10mg of the drug-loaded nano-particles (ATRA/NP) prepared in the step (1), and extruding the blending liquid back and forth from a 200 nm-aperture polycarbonate membrane for 11 times through a micro extruder to obtain the liver-targeted bionic cell membrane drug-loaded nano-particles (TM-ATRA/NP).
FIG. 2 is a transmission electron microscopy image of liver-targeted biomimetic cell membrane drug-loaded nanoparticles (TM-ATRA/NP); the liver-targeted bionic cell membrane drug-loaded nano-particles prepared by the invention in figure 2 are spherical particles and have a classical core-shell structure.
FIG. 3 is a Western blot analysis. The membrane protein concentrations of LX2cell lysate (LX 2cell lysate, CL), LX2cell line lysate stably expressing TRAIL protein (LX 2-TRAIL-zsgreen CL), LX2cell membrane (LX 2cell membrane) and engineered cell membrane (LX 2-zsgreen-TRAIL cell membrane) were measured using BCA kit, respectively, and it can be seen that LX2cell line lysate stably expressing TRAIL protein (LX 2-TRAIL-zsgreen CL) and engineered cell membrane (LX 2-zsgreen-TRAIL cell membrane) all expressed TRAIL protein as shown in fig. 3.
Application example 1
(1) Cytotoxicity test
The crystal violet dyeing detection process comprises the following steps: inoculating Huh7 cells into a 12-well plate according to the number of 5X 10-4 cells/well, culturing for 12 hours, and standing for cell attachment; medium treatments of 24h containing no-nanoparticle treated cells, empty Nanoparticle (NP) cell membrane-coated empty nanoparticle (nanoparticles coated with fibroblast membrane, M-NP), TRAIL protein-expressing cell membrane-coated empty nanoparticle (nanoparticles coated with TRAIL-expressing fibroblast membrane, TM-NP), drug-loaded nanoparticle (ATRA-encapsulated nanoparticles, ATRA/NP), cell membrane-coated ATRA-loaded nanoparticle (ATRA-encapsulated nanoparticles coated with fibroblast membrane, M-ATRA/NP), and liver-targeted biomimetic cell-loaded nanoparticle (ATRA-encapsulated nanoparticles coated with TRAIL-expressing fibroblast membrane, TM-ATRA/NP) were used as experimental groups for Huh7 cells, and fresh medium treatments were used as control groups (control) for Huh7 cells, respectively; then the culture medium containing NP, M-NP, TM-NP, ATRA/NP, M-ATRA/NP or TM-ATRA/NP is discarded, and after washing twice with phosphate buffer solution, the plates are placed on filter paper in an inverted manner, and the water is drained; to the wells of 6 experimental groups (NP, M-NP, TM-NP, ATRA/NP, M-ATRA/NP, or TM-ATRA/NP) and one control group (control), 50. Mu.L of crystal violet staining solution was added per well and incubated at room temperature on a shaker at a frequency of 20 oscillations per minute for 20min. Discarding crystal violet staining solution in the plate, washing with phosphate buffer solution for four times, pouring the plate on filter paper, draining water, and observing cells under a microscope; the detection results are shown in FIG. 4.
FIG. 4 is a graph showing the effects of detection of TM-ATRA/NP and apoptosis induced by NP, M-NP, TM-NP, ATRA/NP, and M-ATRA/NP by crystal violet staining solution. As can be seen from fig. 4, TRAIL cell membranes themselves have an apoptotic effect. The cytotoxicity of TM-ATRA/NP was maximal, indicating that ATRA could synergistically potentiate the cell killing effect of TRAIL.
(2) Liver fibrosis experiment
A mouse liver fibrosis model was constructed. CCl4 was dissolved in corn oil in a 1:4 ratio, and corresponding doses were given to mice according to the weight of the mice in a proportion of 0.5. Mu.L/g of the volume of CCl4, but for practical reasons (range of the syringe) mice were intraperitoneally injected with a constant volume of 50. Mu.L, with the remaining volume being made up with corn oil. Intraperitoneal injections were given every 7 days and weighed in time. Mice were treated with Phosphate Buffered Saline (PBS), TM-NP, ATRA/NP, and TM-ATRA/NP, respectively, and the mice from each group were weighed and averaged.
FIG. 5 is the effect of staining TM-ATRA/NP with control vector TM-NP, ATRA/NP on a mouse liver fibrosis model by classical collagen fiber staining method (Masson) to induce apoptosis. As can be seen from the experimental results, the control group, i.e. healthy mice not receiving treatment, had no obvious blue collagen fibers, indicating that the liver was healthy, whereas the PBS group had a large amount of blue collagen fiber deposited, extending outward from the periphery of the manifold area, the fibrids were thicker and stained darker, indicating that collagen fibers were more, false lobules had formed, indicating that liver fibrosis was severe, and no therapeutic effect. While the TM-NP group had some blue fibril deposition extending outward from around the manifold region, but had some therapeutic effect compared to the PBS group. The ATRA/NP group showed significant blue fibril deposition, but with time the blue fibril decreased, indicating a certain therapeutic effect. Collagen fibers were far less in the TM-ATRA/NP group than in the other groups, indicating that the liver-targeted biomimetic cell membrane drug-loaded nanoparticles (TM-ATRA/NP) were most pronounced to reduce collagen fiber deposition.
FIG. 6 is the effect of detecting TM-ATRA/NP on a mouse liver fibrosis model by sirius red staining with control vector TM-NP and ATRA/NP induced apoptosis. Sirius red staining showed that various types of collagen fibers spread outward from the manifold region, and collagen deposition and spread were observed under polariscope to form prosthetic leaflets, with a large amount of red type I collagen fibers, a small amount of open mesh type II collagen fibers, and yellowish type IV collagen fibers. As can be seen from the experimental results, the control group, i.e. healthy mice not receiving treatment, have no yellow collagen fibers deposited, and the red collagen fibers are less and less stained, which indicates that the liver is healthy and has no liver fibrosis. The PBS group has a large amount of red and faint yellow collagen fibers deposited, and extends outwards from the periphery of a manifold area, and the fiber ropes are thicker and are dyed deeply, so that the PBS group has more collagen fibers, which indicates that liver fibrosis is serious and has no treatment effect. While the TM-NP group had some red collagen fiber deposition, there was still some fibrosis compared to the control group and some therapeutic effect compared to the PBS group. The ATRA/NP group was similar to the TM-NP group, and red collagen fiber deposition was reduced compared to the PBS group, indicating a therapeutic effect. Collagen fibers in the TM-ATRA/NP group are far less than those in other groups, which indicates that liver-targeted bionic cell membrane drug-loaded nano particles (TM-ATRA/NP) have remarkable treatment effect in the liver fibrosis process, and collagen fiber deposition is remarkably reduced.
FIG. 7 shows the effect of detecting TM-ATRA/NP on the control vector TM-NP, ATRA/NP on apoptosis induction by Hematoxylin (hemaloxin) -Eosin (Eosin) staining on the mouse liver fibrosis model. The control group, i.e., healthy mice not receiving treatment, was found to have no apparent tubular structure, indicating no liver fibrosis. The liver slice tissue structure of the PBS group is shown that liver lobule and manifold area structures are not obvious, a large amount of collagen fibers are generated, and the liver slice tissue structure is connected with and surrounds liver cell groups to form a ring-shaped and non-uniform artificial lobule structure; the liver cells in the pseudo-leaflet structure are scattered, the chordal arrangement is not obvious, the gap is large, the liver slice presents a large-area pseudo-leaflet tissue structure, and obvious circular vacuoles are visible in the liver cells. While the TM-NP group had some collagen fiber deposition, and had a certain therapeutic effect but no significant effect compared to the PBS group. The reduced deposition of red collagen fibers in the ATRA/NP group compared with the PBS group indicates a certain therapeutic effect. TM-ATRA/NP still shows normal liver tissue structure, which indicates that the liver-targeted bionic cell membrane drug-loaded nano-particles (TM-ATRA/NP) have remarkable effect of treating liver fibrosis.
Organ distribution experiment of nanocarriers: mice that constructed the liver fibrosis model were cervical sacrificed. Taking Heart (Heart), liver (Liver), spleen (Spleen), lung (Lung) and Kidney (Kidney), fixing in 10% neutral formalin for 4h, taking out, dehydrating with conventional ethanol step by step, and embedding with xylene transparent paraffin; after serial sections, conventional xylenes were dewaxed and washed with ethanol to water at various levels. HE staining was performed and the staining results are shown in fig. 8. Fig. 8 is a schematic diagram of HE staining of tissue sections of different organs to assess treatment safety. As can be seen from fig. 8, the groups were found to be indistinguishable and in physiologically normal condition in other organs except for the liver slice staining. Proves the biological safety and liver targeting of the liver targeting bionic cell membrane drug-loaded nano-particles (TM-ATRA/NP).
The liver-targeted bionic cell membrane drug-loaded nanoparticle prepared by the preparation method of the liver-targeted bionic cell membrane drug-loaded nanoparticle can be used for preparing drugs for treating liver fibrosis.

Claims (6)

1. The preparation method of the liver-targeted bionic cell membrane drug-loaded nanoparticle is characterized by comprising the following steps of:
(1) Preparing drug-loaded nano-particles: dissolving a lactic acid-glycolic acid copolymer and all-trans retinoic acid in anhydrous dichloromethane, wherein the mass ratio of the lactic acid-glycolic acid copolymer to the all-trans retinoic acid is 8-10:1, using polyvinyl alcohol as an aqueous phase, and preparing the aqueous phase by dissolving the polyvinyl alcohol in deionized water, wherein 4g of the polyvinyl alcohol is added into every 100ml of deionized water; then emulsifying on ice by using probe ultrasound under the condition that the output power is 260-325W and the duration is 5-15min; slowly dripping the white emulsion formed by ultrasonic emulsification into 12-14mL of single distilled water, stirring for 4-5h at room temperature to volatilize dichloromethane and solidify the drug-loaded nano particles; finally, the drug-loaded nano particles are collected after ultrafiltration and centrifugation for 30min at 3000 rpm;
(2) Preparation of LX2cell line stably expressing TRAIL protein: culturing LX2 cells in a logarithmic growth phase until the cells adhere to the wall, then transfecting the LX2 cells by using a culture medium containing Lv-TRAIL-zsgreen over-expression lentivirus, culturing for 24 hours at 37 ℃, replacing the culture medium containing the Lv-TRAIL-zsgreen over-expression lentivirus by using a DMEM culture medium for further culturing for 48 hours, and then performing resistance screening by using a G418 culture medium containing 200 mu mol/L to obtain a LX2cell line which can stably express TRAIL protein by stable transfection;
(3) Preparation of engineered cell membranes: washing the LX2cell line which is prepared in the step (2) and stably expresses TRAIL protein in a phosphate buffer solution at 4 ℃ for three times, re-suspending the cell line by using hypotonic cell lysate, then lysing the cell line on a shaker at 4 ℃ for 4-6 hours, outputting 260-325W of power, and performing ultrasonic treatment on the cell line by a probe for 1-3 minutes; then, differential centrifugation is carried out to extract cell membrane precipitation; finally, re-suspending the proposed cell membrane sediment by using phosphate buffer salt solution, and extruding the cell membrane suspension back and forth from the polycarbonate membrane for 9-13 times by a micro extruder to obtain an engineering cell membrane;
the differential centrifugation in step (3) includes: firstly, centrifuging for 10-15min under the centrifugal force of 1,000-1500g and the centrifugal condition of 4 ℃, and taking supernatant; centrifuging supernatant at 10000-12000g centrifugal force and 4deg.C for 30-40min; finally, centrifuging for 1-3h under the centrifugal force of 900000-100000g and the centrifugal condition of 4 ℃;
(4) Preparing liver-targeted bionic cell membrane drug-loaded nano-particles: and (3) carrying out ultrasonic treatment on the engineering cell membrane prepared in the step (3), then blending the engineering cell membrane with the drug-carrying nano-particles prepared in the step (1) according to the mass ratio of 1:1, and extruding the blending liquid back and forth from a 200 nm-aperture polycarbonate membrane for 9-12 times through a micro extruder to obtain the liver-targeted bionic cell membrane drug-carrying nano-particles.
2. The method for preparing liver-targeted biomimetic cell membrane drug-loaded nanoparticles according to claim 1, wherein the hypotonic cell lysate comprises 0.25 x phosphate buffer salt solution, protease inhibitor and phosphatase inhibitor.
3. The method for preparing liver-targeted drug-loaded nanoparticles with bionic cell membrane according to claim 1, wherein the pore diameter of the polycarbonate membrane in the step (3) is 50nm, 100nm, 200nm or 400nm.
4. The method for preparing liver-targeted bionic cell membrane drug-loaded nanoparticles according to claim 3, wherein the pore diameter of the polycarbonate membrane is preferably 200nm.
5. A liver-targeted biomimetic cell membrane drug-loaded nanoparticle prepared by the method of any one of claims 1-4.
6. Use of the liver-targeted biomimetic cell membrane drug-loaded nanoparticle of claim 5 in the preparation of a medicament for treating liver fibrosis.
CN202210347880.4A 2022-04-01 2022-04-01 Liver-targeted bionic cell membrane drug-loaded nano-particle and preparation method and application thereof Active CN114831961B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202210347880.4A CN114831961B (en) 2022-04-01 2022-04-01 Liver-targeted bionic cell membrane drug-loaded nano-particle and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202210347880.4A CN114831961B (en) 2022-04-01 2022-04-01 Liver-targeted bionic cell membrane drug-loaded nano-particle and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN114831961A CN114831961A (en) 2022-08-02
CN114831961B true CN114831961B (en) 2023-08-29

Family

ID=82563984

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210347880.4A Active CN114831961B (en) 2022-04-01 2022-04-01 Liver-targeted bionic cell membrane drug-loaded nano-particle and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN114831961B (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015164217A1 (en) * 2014-04-21 2015-10-29 Theraly Pharmaceuticals Inc. Trail receptor agonists for treatment of fibrotic diseases
CN113925856A (en) * 2021-10-13 2022-01-14 郑州大学 Preparation method and application of bionic nano-drug targeting hepatic stellate cells
CN114225027A (en) * 2021-11-16 2022-03-25 浙江大学 Genetically engineered liver cancer targeted cell membrane bionic nano-microsphere and preparation method thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060275838A1 (en) * 2003-07-12 2006-12-07 Ranjit Ray Compositions and methods for inhibiting liver stellate cell growth

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015164217A1 (en) * 2014-04-21 2015-10-29 Theraly Pharmaceuticals Inc. Trail receptor agonists for treatment of fibrotic diseases
CN106573049A (en) * 2014-04-21 2017-04-19 Theraly制药公司 TRAIL receptor agonists for treatment of fibrotic diseases
CN113925856A (en) * 2021-10-13 2022-01-14 郑州大学 Preparation method and application of bionic nano-drug targeting hepatic stellate cells
CN114225027A (en) * 2021-11-16 2022-03-25 浙江大学 Genetically engineered liver cancer targeted cell membrane bionic nano-microsphere and preparation method thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Yesi Shi等.TRAIL-expressing cell membrane nanovesicles as an anti-inflammatory platform for rheumatoid arthritis therapy.《Journal of Controlled Release》.2020,第320卷第304-313页. *

Also Published As

Publication number Publication date
CN114831961A (en) 2022-08-02

Similar Documents

Publication Publication Date Title
CN106924755B (en) Activated neutral particle cell membrane coated bionic nano particle and preparation method thereof
Jay et al. Engineering of multifunctional gels integrating highly efficient growth factor delivery with endothelial cell transplantation
CN112386709B (en) Targeting polypeptide modified drug-loaded lipoprotein nano drug delivery system and preparation and application thereof
Jin et al. Rapid Extracellular Matrix Remodeling via Gene‐Electrospun Fibers as a “Patch” for Tissue Regeneration
CN113952461B (en) Neutrophil-imitating nano drug delivery system and preparation method and application thereof
CN109666695A (en) A kind of excretion body carrier and its preparation method and application of targeted integration element α v β 3
NL2033447B1 (en) Brain-targeting erythrocyte membrane-enveloped salvianolic acid b nanoparticles as well as preparation method and application thereof
CN115487167B (en) Preparation of multifunctional bionic nano nerve antidote and application of multifunctional bionic nano nerve antidote in Alzheimer disease treatment medicine
CN111588704B (en) Targeted responsive release system and preparation method and application thereof
CN113041356A (en) Hemopsin targeting drug-loading system, preparation method and application thereof
CN113355290A (en) Anti-tumor engineered exosome, preparation method and application
Ren et al. Mechanism of miR-30b-5p-Loaded PEG-PLGA nanoparticles for targeted treatment of heart failure
CN114831961B (en) Liver-targeted bionic cell membrane drug-loaded nano-particle and preparation method and application thereof
Lee et al. Alveolar macrophage phagocytosis-evading inhaled microgels incorporating nintedanib-PLGA nanoparticles and pirfenidone-liposomes for improved treatment of pulmonary fibrosis
CN111529505B (en) Functional chimeric apoptotic body and preparation method and application thereof
CN111529504B (en) Functional chimeric apoptotic body and preparation method and application thereof
CN114668742B (en) Bacteria-imitating nano medicine delivery system and preparation method and application thereof
CN116286640A (en) Application of apoptotic bodies in inducing macrophage polarization and promoting wound healing
Hu et al. Immunomodulatory effect and safety of TNF-α RNAi mediated by oral yeast microcapsules in rheumatoid arthritis therapy
CN107837400A (en) A kind of novel nano genophore and preparation method thereof
CN114903870B (en) Engineered cell membrane nano-particles and preparation method and application thereof
Wang et al. Biomimetic nanoparticles of platelet membranes carrying bFGF and VEGFA genes promote deep burn wound healing
CN115737557B (en) miR-55 preparation and preparation method and application thereof
CN115779101B (en) Preparation and application of nanoparticle for resisting tumor metastasis
CN117180443B (en) Application of cell membrane of synovial myofibroblast in preparation of osteoarthritis medicine

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