CN113797355B - Preparation method and application of nano material with synovial inflammation inhibition function - Google Patents

Preparation method and application of nano material with synovial inflammation inhibition function Download PDF

Info

Publication number
CN113797355B
CN113797355B CN202111197427.1A CN202111197427A CN113797355B CN 113797355 B CN113797355 B CN 113797355B CN 202111197427 A CN202111197427 A CN 202111197427A CN 113797355 B CN113797355 B CN 113797355B
Authority
CN
China
Prior art keywords
shrna
leptinr
rpk
preparing
plasmid
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
CN202111197427.1A
Other languages
Chinese (zh)
Other versions
CN113797355A (en
Inventor
杨成莉
周凯
胡丹蓉
钱志勇
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.)
West China Hospital of Sichuan University
Original Assignee
West China Hospital of Sichuan 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 West China Hospital of Sichuan University filed Critical West China Hospital of Sichuan University
Priority to CN202111197427.1A priority Critical patent/CN113797355B/en
Publication of CN113797355A publication Critical patent/CN113797355A/en
Application granted granted Critical
Publication of CN113797355B publication Critical patent/CN113797355B/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
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/0008Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition
    • A61K48/0025Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic acid
    • A61K48/0033Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'non-active' part of the composition delivered, e.g. wherein such 'non-active' part is not delivered simultaneously with the 'active' part of the composition wherein the non-active part clearly interacts with the delivered nucleic acid the non-active part being non-polymeric
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y40/00Manufacture or treatment of nanostructures
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • 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

Abstract

The invention discloses a preparation method and application of a nano material with a synovitis inhibiting function, belonging to the technical field of biology, and the technical scheme is as follows: the method comprises the following steps: s1, construction of shRNA-leptinR: inserting the synthesized shRNA-LeptinR fragment into a plasmid vector to obtain a recombinant plasmid vector, then converting the recombinant plasmid vector into escherichia coli, and extracting to obtain shRNA-LeptinR plasmid; s2, preparing a macrophage membrane; s3, preparing the gene-carrying nanoparticles M2H@RPK, and preparing the nanoparticles M2H@RPK by adopting an electrostatic adsorption method; and (3) wrapping the nanoparticle H@RPK by using the M2 macrophage membrane extracted in the step S2, and repeatedly passing through the polycarbonate membrane by adopting a micro extruder to obtain the nanoparticle M2H@RPK. The nanometer system is mainly used for reducing inflammatory cascade reaction in the OA process, targets cartilage matrix, and can effectively reduce joint injury and inhibit the severity of overall arthritis.

Description

Preparation method and application of nano material with synovial inflammation inhibition function
Technical Field
The invention relates to the technical field of biology, in particular to a preparation method and application of a nano material with a synovitis inhibiting function.
Background
There is growing evidence that Osteoarthritis (OA) is an inflammatory-related immunological disorder in which the synovium of the joints plays an important role. The joint synovium can be divided into two layers, namely a synovial inner layer and a synovial lower layer which are thinner and close to the joint cavity, wherein the surface layer is provided with 2-3 layers of cells, and two types of cells can be confirmed under an electron microscope, but no obvious boundary exists between the two types of cells, namely macrophage-like synovial cells and fibroblast-like synovial cells.
During synovitis, the number of macrophages increases dramatically. This massive growth results in part from the original macrophages in the synovium escaping the apoptotic pathway, but more from the infiltrating macrophages during blood circulation. Macrophages are a highly variable cell, and mainly have two polarization forms, namely M1 type and M2 type, in response to the difference of microenvironments, M1 type macrophages are a main macrophage subgroup formed by osteoarthritic factors, cartilage degradation and osteophytes, and M2 type macrophages have a protective effect on osteoarthritis by secreting anti-inflammatory factors.
Synovium can secrete Leptin (Leptin) in large quantity, the expression level of Leptin is obviously related to the progress and severity of OA, leptin plays a regulating role through a receptor Leptin R, and participates in autoimmune inflammatory reaction through inducing proliferation and activation of mononuclear or macrophage, so that targeting and regulating Leptin signals provide a potential immune pathway for OA treatment.
OA is currently the most common joint disease in the elderly worldwide, is an important cause of affecting the quality of life of the elderly, and the major clinical manifestation of this disease is pain and gradual loss of function, and current treatments have certain limitations.
In order to solve the problems, the invention provides a preparation method and application of a nano material with a synovitis inhibiting function based on the prior art.
Disclosure of Invention
The invention aims to provide a preparation method of a nano material with a synovitis inhibiting function, and the nano material can effectively reduce joint injury and inhibit the severity of overall arthritis by constructing a shRNA-leptinR (Ob-Rb) nano system which is wrapped by a macrophage membrane and targets a cartilage matrix.
The technical aim of the invention is realized by the following technical scheme:
the preparation method of the nano material with the synovitis inhibiting function comprises the following steps:
s1, construction of shRNA-leptinR plasmid: and inserting the synthesized shRNA-LeptinR fragment into a plasmid vector to obtain a recombinant plasmid vector, then converting the recombinant plasmid vector into escherichia coli, and extracting to obtain the shRNA-LeptinR plasmid.
S2, preparing a macrophage membrane:
A. taking M0 type mouse mononuclear macrophage leukemia cells in logarithmic growth phase, adding IL-4 and IL-13 when the density reaches 70%, and collecting M2 type macrophages after 24h of action.
B. The cells were resuspended with pre-chilled Tris-magnesium salt buffer, then repeatedly extruded 20 times through a mini-extruder to destroy the cells, and then the cell membranes were extracted by sucrose gradient centrifugation.
S3, preparing the gene-carrying nanoparticles M2H@RPK, wherein the nanoparticles M2H@RPK are prepared by adopting an electrostatic adsorption method, and the specific steps are as follows:
a. mixing the shRNA-LeptinR plasmid in the step S1 with iRGD, and standing for 5min to form a blend of the iRGD and the shRNA-LeptinR plasmid; adding PEI and anti-inflammatory peptide KAFAKLAARLYRKALARQLGVAA (KAFAK) to obtain compound RPK (KAFAK-iRGD-PEI-shRNA-leptinR), standing and mixing for 5min.
b. Adding HA into the compound RPK, uniformly mixing, and obtaining nano-particles H@RPK (HA@KAFAK-iRGD-PEI-shRNA-leptinR) through self-assembly.
c. And (3) wrapping the nanoparticle H@RPK by using the M2 macrophage membrane extracted in the step S2, repeatedly passing through the polycarbonate membrane by adopting a micro extruder to obtain the nanoparticle M2H@RPK, and then preserving at 4 ℃ and using as soon as possible.
By adopting the technical scheme, the synovium can secrete a large amount of Leptin (Leptin), the expression level of Leptin is obviously related to the progress and severity of OA, the Leptin plays a role in regulating through a receptor (Leptin R) of Leptin, and the shRNA is an RNA sequence with a tight clasp, can be used for RNA interference to silence the expression of a target gene, the shRNA is introduced into a plasmid vector, shRNA-Leptin R plasmid carrying the target Leptin R gene is obtained, and a promoter in the plasmid vector ensures that the shRNA is always expressed, and simultaneously silences the Leptin R gene, so that the effect of reducing the level of Leptin R protein is achieved, inflammatory cascade reaction in the OA process can be reduced, joint injury is reduced, and the severity of overall arthritis is inhibited.
The extracted M2 macrophage membrane is used for wrapping the nanoparticle M2H@RPK, so that the biocompatibility of the nanoparticle M2H@RPK is improved while the complex biological function of a cell membrane is maintained, the monitoring and the removal of an immune system of an organism can be effectively avoided, the targeting specificity is improved, the toxic and side effects are reduced, the targeting cartilage matrix of a nano system can be effectively realized, and the aims of reducing joint injury and inhibiting the severity of overall arthritis are fulfilled.
The adoption of the scheme can reduce the inflammatory cascade reaction in the OA process, and the nano system targets the cartilage matrix, so that the joint injury can be effectively reduced, and the severity of the whole arthritis can be inhibited.
Further, the forward sequence of the shRNA construction primer in step S1 is 5'-CACAAAGAAGTTACATCTT-3'; the reverse sequence of the constructed primer is 3'-AAGATGTAACTTCTTTGTG-5'.
Further, in step S3, shRNA-LeptinR plasmid: iRGD: PEI: KAFAK: HA was added in an amount of 1:5:5:20:30.
further, in the step S3, the mass ratio of the M2 cell membrane to the shRNA-leptinR plasmid addition amount is 48:10.
further, the pore diameter of the polycarbonate film in the S3 step was 400nm.
The invention also provides application of the preparation method of the nano material with the synovitis inhibiting function, and the preparation method of the nano material with the synovitis inhibiting function is applied to inhibiting synovitis.
In summary, the invention has the following beneficial effects:
1. according to the invention, an shRNA-leptinR (Ob-Rb) nano system wrapped by a macrophage membrane is constructed, so that inflammatory cascade reaction in the OA process can be reduced.
2. And the nano system targets the cartilage matrix, so that the joint injury can be effectively reduced, and the severity of the whole arthritis can be effectively inhibited.
Drawings
FIG. 1 is a graph of Western Blot results of in vitro down-regulation of the LeptinR protein by shRNA-LeptinR constructed in example 1;
FIG. 2 is a fluorescent staining verification chart of the M2 cell membrane coated nanosystems in example 1;
FIG. 3 is a graph of cell biosafety statistics of the nanosystems prepared in example 1;
FIG. 4 is a graph showing the detection of IL-10 and IL-1βTNF- α secretion from synovial cell supernatants by nanosystems in example 1;
FIG. 5 shows PCR detection of mRNA expression levels of synovial cell inflammatory factors IL1 beta, IL10 and TNF-alpha by the nanosystems of example 1.
Detailed Description
The invention is described in further detail below with reference to the attached drawings and embodiments:
example 1: the preparation method of the nano material with the synovitis inhibiting function comprises the following steps:
s1, construction of shRNA-leptinR plasmid: inserting the synthesized shRNA-LeptinR fragment into a Plko.1-EGFP-PURO plasmid vector to obtain a recombinant plasmid vector, then converting the recombinant plasmid vector into escherichia coli, extracting to obtain shRNA-LeptinR plasmid which contains green fluorescent protein labeled fragments and has ampicillin resistance, and transferring the shRNA-LeptinR plasmid into the escherichia coli, wherein the forward sequence of a adopted construction primer is 5'-CACAAAGAAGTTACATCTT-3'; the reverse sequence of the constructed primer is 3'-AAGATGTAACTTCTTTGTG-5'.
S2, preparing a macrophage membrane:
A. taking M0 type mouse mononuclear macrophage leukemia cells in logarithmic phase, adding 20ng.mL when density reaches 70% -1 IL-4 and IL-13, after 24h of action, M2 type macrophages were collected.
B. The cells were resuspended with pre-chilled Tris-magnesium salt buffer (TM buffer, pH 7.4,0.01M Tris and 0.001M MgCl) to a cell concentration of 2X 107 cells/mL, and then repeatedly extruded 20 times through a mini-extruder to destroy the cells, followed by sucrose gradient centrifugation to extract the cell membranes. The sucrose gradient centrifugation method comprises the following specific steps:
diluting the TM buffer solution to the concentration of 0.25M sucrose by adopting 1M sucrose solution, and centrifuging for 10min at 4 ℃ with 2000 g; then collecting supernatant, centrifuging at 4000g and 4 ℃ for 30min, re-suspending and washing the lower precipitate with TM buffer of 0.25M sucrose, centrifuging at 4000g and 4 ℃ for 30min again, collecting cell membranes, and then adopting 100W power ultrasound for 2min to store the obtained cell membranes for later use. Protein quantification of the extracted M2 macrophage membrane using BCA protein quantification showed a protein content of about 0.24mg per 108 cells.
S3, preparing the gene-carrying nanoparticles M2H@RPK, wherein the nanoparticles M2H@RPK are prepared by adopting an electrostatic adsorption method, and the specific steps are as follows:
a. mixing 10 mu g shRNA-LeptinR plasmid in the step S1 with 48 mu g giRGD, and standing for 5min to form a blend of iRGD and shRNA-LeptinR plasmid; then 50. Mu.g PEI and 200. Mu.g anti-inflammatory peptide KAFAKLAARLYRKALARQLGVAA (KAFAK) were added to obtain a complex RPK (KAFAK-iRGD-PEI-shRNA-leptinR), and the mixture was allowed to stand and mixed for 5 minutes.
b. Adding 300 mu g of HA into the composite RPK, uniformly mixing, and obtaining the nanoparticle H@RPK (HA@KAFAK-iRGD-PEI-shRNA-leptinR) through self-assembly.
c. And (3) wrapping the nanoparticle H@RPK by using the M2 macrophage membrane extracted in the step (S2), adding 0.048mg M2 cell membrane into every 10 mug shRNA-leptin R plasmid, repeatedly passing through 400 polycarbonate membranes by adopting a micro extruder to obtain the nanoparticle M2H@RPK, wherein the extrusion times are 20 times, and then preserving at 4 ℃ and using as soon as possible.
The invention adopts western blotting to evaluate the down-regulation condition of the leptin R protein, and comprises the following steps:
cells were collected at the end of transfection, washed with cold PBS and then resuspended in 100. Mu.L of RIPA lysis buffer containing cocktail.
After incubation on ice for 30min, lysed cells were collected and centrifuged at 12000g for 15min. The BCA kit was used to determine the total protein concentration of each group. Total proteins were separated on a 15% SDS-PAGE gel (120V voltage, 2 h), transferred to PVDF membrane (80V voltage, 2 h) and blocked in 5% skim milk for 2h.
Incubation with LeptinR rabbit monoclonal antibody overnight at 4 ℃. After washing the PVDF membrane twice with TBST buffer, the PVDF membrane (goat anti-rabbit) was incubated with horseradish peroxidase conjugated secondary antibody, and analyzed with a chemiluminescent detection system.
The invention characterizes the physicochemical properties of the nano particles by a plurality of characterization modes.
d. Particle size, potential
The particle size, polydispersity (PDI) and Zeta potential of H@RPK, M2H@RPK were determined using a Markov laser scattering particle sizer. The test was repeated 3 times at 25 ℃.
e. Appearance and morphology determination
The morphology of H@RPK, M2H@RPK was observed using TEM. Before observation, the prepared sample dispersion liquid is dripped on a copper mesh covered by nitrocellulose, and after being negatively dyed by 2% phosphotungstic acid, the sample dispersion liquid is dried at room temperature, and the shape of the sample dispersion liquid is observed.
f.DNA gel blocking experiment
In order to determine the protection effect of the nanoparticles on shRNA-leptinR, a gel blocking experiment was performed. The shRNA-LeptinR, RR, RP, RPK, H@RPK and M2H@RPK are diluted by deionized water, adjusted to the same amount of shRNA-leptinR (200 ng of shRNA-leptinR is added into each hole), placed in agarose gel with the concentration of 1.0%, electrophoresed for 40 minutes under the constant pressure of 110V by adopting TAE electrophoresis buffer solution, and imaged by adopting a nucleic acid gel imaging system.
Identification of M2 cell membrane coating fluorescent staining
To visually identify whether the M2 cell membrane is successfully coated on the surface of the H@RPK nanoparticle. The invention adopts a cell membrane green fluorescent dye PKH67 to mark an M2 cell membrane, connects a red fluorescent dye cy5 with PEI to mark H@RPK nanoparticles, and observes the coating condition of the M2 cell membrane by observing the co-localization of red and green fluorescence, and comprises the following specific steps: centrifuging to collect polarized M2 cell membrane, washing with serum-free medium, and sucking to dry the cell to ensure that the residue is not more than 25 μL; then, M2 cell membranes were green fluorescent labeled with 2X 107 cells stained with 4. Mu.L PKH67 ethanol stock solution according to PKH67 cell stain kit; meanwhile, the cy5-NHS and PEI (PEI-cy 5) are connected through an amide bond, and H@RPK nanoparticles marked by the red fluorescent dye cy5 are prepared; then repeatedly passing through a 400nm polycarbonate film (20 times) through a micro extruder to prepare a fluorescent marker M2H@RPK; meanwhile, nanoparticles of M2 cell membranes which are not marked by PKH67 are prepared as a research control; finally, taking the prepared nanoparticles by using synovial cells, and imaging by using a confocal microscope.
The inhibition effect of the M2H@RPK nano system on osteoarthritis synovitis is also evaluated through various experiments:
(1) Nanoparticle cytotoxicity investigation
To examine the safety of different components of the nanoparticle to synovial membrane and chondrocyte in joint cavity. Seeding synovial membrane and chondrocytes in 96-well plates at a density of 3500 per well; after 24h PEI-containing (final concentrations 2, 4, 7.8, 15.6, 31.2, 62.5, 125, 250, 500. Mu.g.mL) was added -1 ) KAFAK (final concentrations 2, 4, 7.8, 15.6, 31.2, 62.5, 125, 250, 500. Mu.g.mL) -1 ) M2 (final concentration 0.25, 0.5, 1, 2, 3.75, 7.5, 15, 30, 60. Mu.g.multidot.mL) -1 ) M2H@RPK (final concentration 7.5, 15, 30, 60, 120, 225, 450, 900, 1800. Mu.g.mL) -1 ) Continuously culturing for 24 hours; mu.L MTT (5 mg. Multidot.mL) was added to each well -1 pH 7.4), incubation at 37℃for 4h; the supernatant was pipetted off and 150. Mu.L of DMSO was added to each well to completely dissolve the bottom crystals and absorbance was read at 570 nm. Cell viability was calculated from the blank wells (cells incubated without drug medium) and zeroed wells (medium alone without cells) set up, with 6 duplicate wells per sample.
(2) Dynamic observation of synovial cell uptake
To evaluate the effect of cell uptake time on cell uptake rate, synovial cells were seeded at 5×104 cells/well into a 24-well cell culture plate at the bottom of glass, after 24h, the cells were lysosomally labeled with a green fluorescent lysosome probe dye (lyso-tracker), after 2h staining, the medium was discarded, PBS was slightly washed twice, serum-Free medium containing Free cy5, PEI-cy5, cy5 labeled h@rpk and m2h@rpk nanoparticles was added, and subjected to real-time photographing observation under a confocal microscope.
(3) Intracellular positioning observation
To further examine the effect of measuring synovial cells on the intracellular distribution of Free cy5, PEI-cy5, cy 5-labeled H@RPK and M2H@RPK nanoparticles, synovial cells were seeded at 5X 104 cells/well into 24-well plates; after 24 hours, the culture medium is discarded and replaced by a serum-free culture medium containing the components; 2h after dosing, PBS was washed 2 times, fixed with 4% paraformaldehyde for 15min, PBS was washed 2 times, membrane disruption with 0.2% Triton X-100 for 10min, PBS was washed 3 times, then blocked with 1% BSA for 30min, PBS was washed 2 times, after washing with phalloidin for 1h, PBS was washed 2 times, finally after washing with 1. Mu.g.mL-1 DAPI for 10min, PBS was washed 3 times, and cell images were observed and obtained under confocal microscope.
(4) Synovial cell inflammatory factor detection
1) Synovial cell supernatant inflammatory factor detection
Synovial cells were seeded at 5X 104/well into 24-well cell culture plates and, after 24h, serum-free culture with 10 ng.mL-1 IL-1β was added for 24h to stimulate synovial cell inflammation formation. Inflammatory factor IL-10IL-1β was detected by the Meso QuickPlex SQ 120 kit (Meso Scale Discovery, rockville, md., USA) following the PBS, shRNA-LeptinR, KAFAK, M2, H@RPK, M2H@RPK packet interventions, collecting supernatant after 0.5d, 1d, 3d, 5d days.
2) Synovial cell inflammatory factor qPCR detection
The mRNA level of Ptpn2 in mouse tumor tissues is detected by adopting real-time fluorescence quantitative PCR (qPCR), and the specific steps are as follows: firstly extracting total RNA, performing grouping intervention, and adding Trizol lysate to lyse synovial cells after 0.5d, 1d, 3d and 5 d; adding 1/5 volume chloroform solution of Trizol, shaking, mixing, centrifuging at 12000rpm for 15min, and collecting supernatant; adding equal volume of 75% ethanol, centrifuging at 12000rpm for 15min, and collecting supernatant; then, the mixture was centrifuged at 12000rpm for 15min, the supernatant was discarded, and the precipitate was washed once with 0.5mL of 75% ethanol; the supernatant was discarded after centrifugation at 12000rpm for 15min again, and the total RNA concentration was determined by ultra clean bench air drying. Next, RNA was reverse transcribed into DNA according to the instructions of the RNA reverse transcription kit, and the DNA concentration was measured. Finally, qPCR was performed according to the primer sequences of the factors of Table 1.
TABLE 1 qPCR experimental primer sequences
In the embodiment of the invention, the shRNA-leptinR (Ob-Rb) nano system wrapped by the macrophage membrane is constructed, and the nano system targets the cartilage matrix, so that the joint injury can be effectively reduced, and the severity of the whole arthritis can be effectively inhibited.
The present embodiment is only for explanation of the present invention and is not to be construed as limiting the present invention, and modifications to the present embodiment, which may not creatively contribute to the present invention as required by those skilled in the art after reading the present specification, are all protected by patent laws within the scope of claims of the present invention.

Claims (5)

1. The preparation method of the nano material with the synovitis inhibiting function is characterized by comprising the following steps: the method comprises the following steps:
s1, construction of shRNA-leptinR plasmid: inserting the synthesized shRNA-LeptinR fragment into a plasmid vector to obtain a recombinant plasmid vector, then converting the recombinant plasmid vector into escherichia coli, and extracting to obtain shRNA-LeptinR plasmid;
s2, preparing a macrophage membrane:
A. taking M0 type mouse mononuclear macrophage leukemia cells in logarithmic growth phase, adding IL-4 and IL-13 when the density reaches 70%, and collecting M2 type macrophages after 24 hours of action;
B. re-suspending cells by adopting precooled Tris-magnesium salt buffer salt, repeatedly extruding for 20 times by a micro extruder to destroy the cells, and extracting cell membranes by adopting a sucrose gradient centrifugation method;
s3, preparing the gene-carrying nanoparticles M2H@RPK, wherein the nanoparticles M2H@RPK are prepared by adopting an electrostatic adsorption method, and the specific steps are as follows:
a. mixing the shRNA-LeptinR plasmid in the step S1 with iRGD, and standing for 5min to form a blend of the iRGD and the shRNA-LeptinR plasmid; adding PEI and anti-inflammatory peptide KAFAKLAARLYRKALARQLGVAA (KAFAK) to obtain composite RPK (KAFAK-iRGD-PEI-shRNA-leptinR), standing and mixing for 5min;
b. adding HA into the composite RPK, uniformly mixing, and obtaining nano particles H@RPK (HA@KAFAK-iRGD-PEI-shRNA-leptinR) through self-assembly;
c. and (3) wrapping the nanoparticle H@RPK by using the M2 macrophage membrane extracted in the step S2, repeatedly passing through the polycarbonate membrane by adopting a micro extruder to obtain the nanoparticle M2H@RPK, and then preserving at 4 ℃ and using as soon as possible.
2. The method for preparing a nanomaterial with synovitis inhibiting function according to claim 1, characterized in that: the forward sequence of the shRNA construction primer in the S1 step is 5'-CACAAAGAAGTTACATCTT-3'; the reverse sequence of the constructed primer is 3'-AAGATGTAACTTCTTTGTG-5'.
3. The method for preparing a nanomaterial with synovitis inhibiting function according to claim 1, characterized in that: in the S3 step, shRNA-LeptinR plasmid: iRGD: PEI: KAFAK: HA was added in an amount of 1:5:5:20:30.
4. the method for preparing a nanomaterial with synovitis inhibiting function according to claim 1, characterized in that: in the S3 step, the mass ratio of the M2 cell membrane to the shRNA-leptinR plasmid addition amount is 48:10.
5. the method for preparing a nanomaterial with synovitis inhibiting function according to claim 1, characterized in that: the pore diameter of the polycarbonate film in the S3 step was 400nm.
CN202111197427.1A 2021-10-14 2021-10-14 Preparation method and application of nano material with synovial inflammation inhibition function Active CN113797355B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202111197427.1A CN113797355B (en) 2021-10-14 2021-10-14 Preparation method and application of nano material with synovial inflammation inhibition function

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202111197427.1A CN113797355B (en) 2021-10-14 2021-10-14 Preparation method and application of nano material with synovial inflammation inhibition function

Publications (2)

Publication Number Publication Date
CN113797355A CN113797355A (en) 2021-12-17
CN113797355B true CN113797355B (en) 2023-07-21

Family

ID=78937516

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111197427.1A Active CN113797355B (en) 2021-10-14 2021-10-14 Preparation method and application of nano material with synovial inflammation inhibition function

Country Status (1)

Country Link
CN (1) CN113797355B (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114081962A (en) * 2021-11-02 2022-02-25 四川大学华西医院 Preparation method and application of nano material with synovial membrane immunoregulation function
CN114904057B (en) * 2022-04-20 2023-06-09 华中科技大学同济医学院附属协和医院 Preparation method and application of composite fiber ring gel for inhibiting nerve ingrowth
CN116286996A (en) * 2023-03-24 2023-06-23 暨南大学附属第一医院(广州华侨医院) Medicine carrier targeting CXCL16+ macrophages and preparation method and application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101044164A (en) * 2004-07-20 2007-09-26 健泰科生物技术公司 Inhibitors of angiopoietin-like 4 protein, combinations, and their use
CN113197880A (en) * 2021-05-07 2021-08-03 皖南医学院第一附属医院(皖南医学院弋矶山医院) Macrophage exosome membrane-coated bionic nanoparticle and preparation method and application thereof

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL157624A0 (en) * 2001-03-22 2004-03-28 Bristol Myers Squibb Co Polynucleotide encoding apex4 polypeptides and diagnostic and therapeutic compositions containing apex4 polypeptides
WO2010068692A1 (en) * 2008-12-10 2010-06-17 Purdue Research Foundation Cell-permeant peptide-based inhibitor of kinases
US20190117799A1 (en) * 2016-04-01 2019-04-25 The Brigham And Women's Hospital, Inc. Stimuli-responsive nanoparticles for biomedical applications
CN114668743A (en) * 2021-08-04 2022-06-28 贵州医科大学附属医院 Preparation method and application of nano drug delivery system for inhibiting growth of melanoma
CN113995887B (en) * 2021-10-14 2022-06-28 四川大学华西医院 Preparation method and application of cartilage repair nanogel composite system
CN114081962A (en) * 2021-11-02 2022-02-25 四川大学华西医院 Preparation method and application of nano material with synovial membrane immunoregulation function

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101044164A (en) * 2004-07-20 2007-09-26 健泰科生物技术公司 Inhibitors of angiopoietin-like 4 protein, combinations, and their use
CN113197880A (en) * 2021-05-07 2021-08-03 皖南医学院第一附属医院(皖南医学院弋矶山医院) Macrophage exosome membrane-coated bionic nanoparticle and preparation method and application thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Leptin与免疫应答及老年免疫性疾病;徐小玉;林季;颜光涛;;中华老年多器官疾病杂志(05);第441-444页 *
携iRGD肽阳离子微泡的制备及其基本性质检测;曾欣欣;徐金锋;刘莹莹;周玉丽;罗慧;;中国超声医学杂志(05);第471-473页 *
瘦素在类风湿关节炎发病机制中的研究进展;杨林;乔永霞;李红艳;刘爱京;;医学综述(03);第421-427页 *

Also Published As

Publication number Publication date
CN113797355A (en) 2021-12-17

Similar Documents

Publication Publication Date Title
CN113797355B (en) Preparation method and application of nano material with synovial inflammation inhibition function
CN111249449B (en) Extracellular vesicle-interleukin-10 nano targeted drug and preparation method and application thereof
Liu et al. The use of antibody modified liposomes loaded with AMO-1 to deliver oligonucleotides to ischemic myocardium for arrhythmia therapy
AU2011271830B2 (en) Microvesicles derived from cell protoplast, and use thereof
CN112168973B (en) Aptamer delivery vehicle, preparation method and application thereof
Cai et al. Activated CD4+ T cells-derived exosomal miR-142-3p boosts post-ischemic ventricular remodeling by activating myofibroblast
CN114081962A (en) Preparation method and application of nano material with synovial membrane immunoregulation function
JP7420708B2 (en) Methods for producing therapeutic extracellular vesicles from nanoelectroporation and other non-endocytic cell transfections
Bear et al. Nuclear poly (A)-binding protein PABPN1 is associated with RNA polymerase II during transcription and accompanies the released transcript to the nuclear pore
CN110484615B (en) Application of lncRNA in regulation and control of macrophage polarization in viral myocarditis
CN113197880B (en) Macrophage exosome membrane-coated bionic nanoparticle and preparation method and application thereof
KR102450791B1 (en) Method for producing exosomes by electrical stimulation
Zhang et al. Airway epithelial cell-specific delivery of lipid nanoparticles loading siRNA for asthma treatment
Patil et al. Synthesis and efficient siRNA delivery of polyamine-conjugated cationic nucleoside lipids
US20210113641A1 (en) Extraction of plant source "medicinal soup" and manual preparation of "herbal medicine" and related products
Jiang et al. Characterization of the microRNA transcriptomes and proteomics of cochlear tissue-derived small extracellular vesicles from mice of different ages after birth
CN116942700A (en) Multi-target modified engineering M2 macrophage outer vesicle and preparation method and application thereof
Banerjee et al. Hydrogel nanocarrier encapsulated recombinant IκBα as a novel anticancer protein therapeutics
CN111686124B (en) Application of miR-486-3p in preparation of product for treating neuroinflammation caused by SAH (neuroinflammation)
CN110732028A (en) Preparation method and application of double-locked nanoparticles capable of restricting activation of CRISPR/Cas13a
CN110123795A (en) Load purposes of the human pluripotent stem cells excretion body of resveratrol on preparation treatment skin disease drug
CN113975287A (en) Application of rhizoma paridis saponin as lysosome targeting inhibitor
CN115919816B (en) Application of curcumin-tetrahedron framework nucleic acid drug carrier system in preparation of drugs for preventing or treating radioactive oral mucositis
CN114796258B (en) Application of hucMSC-sEV loaded with miR13896 in sepsis acute liver injury
CN114306367B (en) Composition containing C/EBP alpha-saRNA

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