CN113413467A - Carrier-free mRNA delivery method - Google Patents

Carrier-free mRNA delivery method Download PDF

Info

Publication number
CN113413467A
CN113413467A CN202110746485.9A CN202110746485A CN113413467A CN 113413467 A CN113413467 A CN 113413467A CN 202110746485 A CN202110746485 A CN 202110746485A CN 113413467 A CN113413467 A CN 113413467A
Authority
CN
China
Prior art keywords
mrna
metal
carrier
delivery
free
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202110746485.9A
Other languages
Chinese (zh)
Inventor
喻国灿
戚少龙
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.)
Tsinghua University
Original Assignee
Tsinghua 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 Tsinghua University filed Critical Tsinghua University
Priority to CN202110746485.9A priority Critical patent/CN113413467A/en
Publication of CN113413467A publication Critical patent/CN113413467A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/52Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an inorganic compound, e.g. an inorganic ion that is complexed with the active ingredient
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • 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
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/141Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers
    • A61K9/143Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers with inorganic compounds
    • 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
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation

Abstract

The invention discloses a carrier-free mRNA delivery method. The method comprises the following steps: synthesizing carrier-free Metal-mRNA nano particles by coordination-driven self-assembly effect of Metal cations and mRNA molecules, and further realizing delivery of the mRNA molecules. The invention relates to a universal carrier-free mRNA delivery technology, which does not need a loading material to wrap mRNA, but completes self-assembly by virtue of a supermolecule coordination effect between metal ions and the mRNA, and has sensitive micro-environment response, good dispersion stability, high-efficiency mRNA delivery efficiency, low biotoxicity and good biological safety. In addition, the technology is simple and feasible, has universality, is suitable for wide metal ion and mRNA coordination assembly and delivery of different nucleic acid molecular weight lengths and different nucleic acid sequences, is easy for clinical popularization, and has wide application prospect in the field of mRNA treatment.

Description

Carrier-free mRNA delivery method
Technical Field
The invention relates to the technical fields of biological medicine technology, nano medicine, supramolecular chemistry and mRNA delivery, in particular to a carrier-free mRNA delivery method, and specifically relates to a preparation method and application of a carrier-free Metal-mRNA nano material synthesized by coordination-driven self-assembly of Metal ions and mRNA molecules.
Background
Similar to DNA gene therapy, RNA-based therapy is the latest initiative for global medical innovation and has great potential. RNA therapy was marketed from the first antisense therapy (antisense therapy) concept proposed in 1978 to the first antisense oligonucleotide (ASO) drug fomivirsen in 1998, to the RNA interference (RNAi) mechanism research in 2006 with a nobel physiology or medical prize, and in 2020, the introduction of the ultra-individualized drug Technology represented by the ASO drug Milasen into MIT Technology Review ten major breakthroughs, the development of RNA therapy is tortuous and lengthy. RNA therapy refers to therapy for regulating the expression of pathogenic genes from the root using nucleic acids having a function of treating diseases. RNA therapy can be further subdivided into the following three major directions: 1. messenger RNA therapy (Messenger RNA, mRNA) encoding a therapeutic protein or antigen; 2. small nucleic acid therapies that target nucleic acids and inhibit pathogenic RNA activity or activate gene activity, including antisense oligonucleotides (ASOs), small interfering RNAs (sirnas), micro RNAs (mi RNAs), and the like; 3. RNA aptamer (aptamer) therapy which takes protein as a target and regulates the activity of the protein.
The principle of mRNA therapy is to introduce exogenous mRNA into target cells, which allows the target cells to self-synthesize target proteins. The mRNA therapy is simple and convenient to design and prepare and high in safety, compared with the traditional gene therapy, the expression of mRNA is safe due to no risk of inserting a genome, and compared with the traditional targeted drug taking protein as a target point, the mRNA therapy is short in development period and rich in candidate target points. More importantly, mRNA drugs are not confined to dividing cells, do not risk integration into the host genome, and are subject to auto-degradation in vivo. At present, the mRNA therapy is widely applied to the treatment of various diseases such as rare diseases, tumors, infectious diseases and the like by virtue of the characteristics of high efficiency, safety and rich treatment.
The research and development of the mRNA therapy generally include the processes of sequencing, target determination, sequence design and optimization, delivery technology research, nucleic acid synthesis, purification, preparation, production and the like, the current nucleic acid design and preparation processes are relatively mature, and the efficient delivery of mRNA to various tissue cells of a human body is a key to the performance of the therapeutic function of mRNA and is a main technical difficulty of the mRNA therapy. Currently, delivery technology difficulties include: nucleic acid is unstable, is easily degraded by ribonuclease (RNase) in blood plasma and tissues, and is quickly removed by liver and kidney; the immunogenicity of the nucleic acid itself and the carrier material, thereby causing a series of side effects; ③ the nucleic acid has negative charge, so that the nucleic acid is not easy to cross over the cell membrane with negative charge and enter the cytoplasm; fourthly, the card can not be released in vivo. In addition, how to achieve targeted delivery is also the focus of current research. The solutions of the above points are focused on three links of mRNA drug preparation, namely in vitro transcription, chemical modification and delivery vectors, and the most difficult, or technical barrier to construct, is the drug delivery system. Therefore, the reasonable design of the nano delivery technology has important significance for improving the delivery of the therapeutic genes and the treatment efficiency.
At present, technologies for delivering mRNA vectors mainly include liposome nanoparticle carrier technology and polymer carrier technology, wherein liposomes are the mainstream carrier materials with the most extensive applications, and mainly include cationic liposome complexes, liposome polymers, Liposome Nanoparticles (LNPs), cationic nanoemulsions, and the like. Cationic polymers have been widely used for nucleic acid delivery, such as poly (L-lysine), PEI, DEAE-dextran, PBAE and chitosan. In the simplest form, the cationic polymer is mixed with the nucleic acid in excess to form an electrostatically bound cationic polymer. Although many polymers have been developed, they are less advanced in nucleic acid delivery than lipid nanoparticles. LNP delivery technology has achieved good clinical results, but LNP vector technology still has great promotion space, for example, LNP has problems of anaphylaxis, easy oxidative degradation, poor preparation reproducibility, etc. In addition, the mRNA preparation prepared by taking LNP as a carrier can be gathered in the liver and spleen, is difficult to target other parts, and has potential toxic and side effects in clinical use. In recent years, researches show that although PEG in liposome nano materials can effectively prolong half-life and improve water solubility, more and more evidences prove that the great application of PEG can possibly cause PEG 'distress', accelerate the in vivo clearance rate of PEG modified nano-drugs and seriously influence the curative effect of the PEG nano-drugs. Therefore, developing a simple, universal and efficient method for synthesizing a nano-scale mRNA nano-delivery system with specific morphology and function has important significance for various applications.
Disclosure of Invention
The invention aims to provide a carrier-free mRNA delivery method.
In a first aspect, the invention claims a method for vector-free mRNA delivery.
The presently claimed vector-free mRNA delivery method may comprise the steps of:
(A1) synthesizing carrier-free Metal-RNA nano particles (hereinafter referred to as Metal-mRNA) by coordination-driven self-assembly effect of Metal cations and mRNA molecules;
(A2) the delivery of the RNA molecule is achieved in the form of the carrier-free Metal-mRNA nanoparticles.
The non-carrier metal-mRNA nanoparticles obtained in step (a1) can be prepared by the method of the second aspect.
In a second aspect, the invention claims a method of making carrier-free Metal-mRNA nanoparticles.
The method for preparing carrier-free Metal-mRNA nano particles, which is claimed by the invention, can comprise the following steps:
(B1) mixing the target mRNA and the metal cation according to the charge ratio of 500/1-1/500;
(B2) standing the treated system (B1) at 37-60 deg.C (such as 55 deg.C), and naturally cooling;
(B3) and (B2) centrifuging the system after treatment, and collecting precipitates to obtain the carrier-free Metal-mRNA nano particles.
Further, in the step (B1), the charge ratio of the mRNA of interest and the metal cation may be 100/1 to 1/10.
Further, the charge ratio of the mRNA of interest and the metal cation can be 1: 2.
further, in the step (B1), the mixing of the mRNA of interest and the metal cation may be achieved by: the aqueous solution containing the metal cation is added dropwise to the aqueous solution of the target mRNA, and stirred for 1 to 10min (e.g., 1 min).
Further, in the step (B2), the standing time may be 1.5 to 4 hours (e.g., 3 hours).
Further, in the step (B2), the temperature after the natural cooling is room temperature.
Further, in the step (B3), a step of washing the treated system of (B2) with deionized water may be further included before the centrifugation.
Further, in the step (B3), the centrifugation is 10000-. The centrifugation may be performed at room temperature.
Further, in the step (B3), the centrifugation may further include a step of dispersing the precipitate in deionized water for storage.
In the first and second aspects above, the valency of the metal cation may be from +2 to +8 (e.g. +2, + 4); the metal cation is any metal cation that can participate in the phosphate coordination reaction of RNA and can be bioavailable.
Further, the metal cation may be selected from the following: ca2+、Zr4+、Zn2+、Fe2+、Fe3+、Mn2+、 Cu2+、Al3+Or/and Hf4+And the like. The metal cation can be a single metal cation, or different metal cations can be mixed for use.
Further, the mRNA may be a naked mRNA or a modified mRNA. Wherein the modification includes, but is not limited to, phosphorylation and/or glycosylation.
In a specific embodiment of the present invention, the target mRNA is specifically the following mRNA: mouse IL-10, Human EPO, SV40 NLS-Cas9, NLS-Cre, Mouse ECD, EGFP, Luciferase.
In a particular embodiment of the invention, when the metal cation is Ca2+When the metal cation-containing aqueous solution is CaCl2An aqueous solution. More specifically, the CaCl2CaCl in aqueous solution2The concentration of (3) was 798 mM. Accordingly, the mRNA of interest is an EGFP mRNA. More specifically, the EGFP mRNAThe concentration of the aqueous solution was 1 mM. In the step (B1), the EGFP mRNA aqueous solution and the CaCl2The dosage ratio of the aqueous solution is 1000 mu L: 1000 μ L, charge ratio 1: 2.
In a particular embodiment of the invention, when the metal cation is Al3+When the "aqueous solution containing the metal cation" is specifically AlCl3An aqueous solution. More specifically, the AlCl3AlCl in aqueous solution3The concentration of (3) was 798 mM. Accordingly, the mRNA of interest is an EGFP mRNA. More specifically, the EGFP mRNA aqueous solution concentration is 1.5 mM. In the step (B1), the EGFP mRNA aqueous solution and the AlCl3The dosage ratio of the aqueous solution is 1000 mu L: 1000 μ L, charge ratio 1: 2.
In a particular embodiment of the invention, when the metal cation is Zr4+When the "aqueous solution containing the metal cation" is ZrF4An aqueous solution. More specifically, the ZrF4ZrF in aqueous solution4The concentration of (3) was 798 mM. Accordingly, the mRNA of interest is an EGFP mRNA. More specifically, the concentration of the EGFP mRNA aqueous solution is 2 mM. In the step (B1), the EGFP mRNA aqueous solution and the ZrF4The dosage ratio of the aqueous solution is 1000 mu L: 1000 μ L, charge ratio 1: 2.
In a specific embodiment of the invention, the optimal charge ratio of the target mRNA to the metal cation is 1:2 and the volume ratio is 1: 1. The mRNA of interest is EGFP mRNA. The charging ratio of the other valence metal ions is similar to the three types: namely, 2-valent metal: mRNA (molar ratio) 798: 1; a 3-valent metal: mRNA 798: 1.5; 4-valent metal: mRNA 798: 2; other cases may be analogized depending on the specific RNA sequence and metal ion species.
In a third aspect, the invention claims an unsupported Metal-mRNA nanoparticle prepared by the method of the second aspect.
The particle size of the carrier-free Metal-mRNA nano particle obtained by the invention is 20-300nm, and the zeta potential is-100 to 40.
In a fourth aspect, the invention claims the use of the method of the first or second aspect above or the carrier-free Metal-mRNA nanoparticles of the third aspect above for carrier-free delivery of an mRNA of interest to a cell or tissue or organism.
The application may be a non-disease diagnostic therapeutic application.
The present invention is applicable to not only mRNA but also siRNA and/or microRNA.
The technology of the invention synthesizes the carrier-free Metal-mRNA nano particles by the coordination-driven self-assembly effect of Metal ions and mRNA molecules, and has the following advantages: 1. can easily penetrate cell membrane and quickly enter cell. 2. The Metal-mRNA nano particles have good biocompatibility and are more stable. 3. mRNA is easier to dissociate in cytoplasm, so that the endosome escape performance is enhanced, the cell delivery efficiency is improved, and the target protein expression is increased. 4. Metal-mRNA nanoparticles can realize carrier-free delivery and immune disturbance of carrier-free materials. 5. The one-pot supermolecule self-assembly strategy is extremely simple and easy to implement, saves the cost and is suitable for large-scale transformation and application. 6. The technology has universality and is suitable for mRNA delivery of different nucleic acid molecular weight lengths and different nucleic acid sequences.
In conclusion, the invention relates to a universal carrier-free mRNA delivery technology, which does not need a loading material to wrap mRNA, completes self-assembly by virtue of a supermolecule coordination effect between metal ions and mRNA, and has sensitive micro-environment response, good dispersion stability, high-efficiency mRNA delivery efficiency, low biological toxicity and good biological safety. In addition, the technology is simple and feasible, has universality, is suitable for wide metal ion and mRNA coordination assembly and delivery of different nucleic acid molecular weight lengths and different nucleic acid sequences, is easy for clinical popularization, and has wide application prospect in the field of mRNA treatment.
Drawings
FIG. 1 is a schematic diagram of the synthesis of Metal-mRNA nanoparticles by coordination-driven self-assembly of Metal cations and mRNA molecules according to the present invention.
FIG. 2 is an electron micrograph of Ca-mRNA (different sequences) nanoparticles
FIG. 3 shows the survival rate of cells in each group measured by MTT method.
FIG. 4 shows the liver function changes in mice of different groups.
FIG. 5 shows the expression of inflammatory factors in mice of different groups.
FIG. 6 shows EGFP mRNA delivery and quantitation of GFP protein expression for different Metal-EGFP mRNAs.
FIG. 7 is a graph showing the effect of Ca-EGFP mRNA and Lipo2000-mRNA protein expression under a fluorescence microscope.
Detailed Description
The present invention is described in further detail below with reference to specific embodiments, which are given for the purpose of illustration only and are not intended to limit the scope of the invention. The examples provided below serve as a guide for further modifications by a person skilled in the art and do not constitute a limitation of the invention in any way.
The experimental procedures in the following examples, unless otherwise indicated, are conventional and are carried out according to the techniques or conditions described in the literature in the field or according to the instructions of the products. Materials, reagents and the like used in the following examples are commercially available unless otherwise specified.
The invention provides a universal technology for synthesizing Metal-mRNA nano particles by coordination-driven self-assembly of Metal cations and mRNA molecules, which is a carrier-free mRNA delivery technology and a synthetic schematic diagram of the technology is shown in figure 1.
Wherein n is an integer between 2 and 8, and represents the valence potential number of the selected metal ion (i.e., the valence of the metal cation may be from +2 to + 8);
m represents a metal ion species including Mn2+,Cu2+,Zn2+,Fe2+,Ca2+,Zr2+,Fe3+,Zr3+, Zr4+,Hf4+And all the biologically available metal ions which can participate in the phosphate coordination reaction of RNA in the nature, and different metal ions can also be mixed for use, thereby achieving similar assembly effect.
mRNA Chain is used for delivering mRNA, and comprises mRNA chains with different nucleic acid molecular weight lengths and different nucleic acid sequences (including mRNA post-modification, such as phosphorylation and glycosylation and the like). Meanwhile, the discovery is also suitable for other RNA systems, such as siRNA, microRNA and the like.
In the invention, the Metal-mRNA nano-particle consists of Metal ions and mRNA fragments, has uniform particle size and stable property, and can be simply and efficiently used for mRNA delivery in organisms.
Example 1 Synthesis of Metal-mRNA nanoparticles (Ca)2+Example of the design reside in
In this example, the selected metal ion is Ca2+And the mRNA is EGFP mRNA (SEQ ID No. 6). Specifically, 1000. mu.L of CaCl was added2An aqueous solution (798mM) was added dropwise to 1000. mu.L of an aqueous solution of EGFP mRNA (1mM) so that EGFP mRNA was brought into contact with Ca2+Has a charge ratio of 1: 2. Stirring thoroughly for 1min to mix well, standing the mixture at 55 deg.C for 3 hr, and naturally cooling to room temperature. Washing the obtained product with deionized water, centrifuging at 12000 r/min for 5min at room temperature to obtain precipitate as Ca-EGFP mRNA nano particle, and dispersing in deionized water for storage.
The charge ratio of the metal cations and the mRNA sequences adopted in the reaction is determined according to the charge ratio relationship of the mRNA and the metal cations: mRNA-/M+=500/1~1/500(M+Represents the positive charge of the metal cation, as described in table 1: zn (ZnCl)2),Fe(FeCl2),Mn(MnCl2),Cu(CuSO4),Ca(CaCl2),Fe(FeCl3),Al(AlCl3), Zr(ZrF4),Hf(Hf Cl4) Etc., mRNA-Representing the negative charge of different base sequences. The mRNA is specifically Mouse IL-10 (the tail end carries 6 XHis), Human EPO, Mouse ECD (the tail end carries 6 XHis), NLS-Cre, SV40 NLS-Cas9, EGFP and Luciferase (the specific sequences of the mRNA are sequentially shown as SEQ ID No.1 to SEQ ID No. 7). The implementation method of coordination self-assembly of other metal ions and mRNA is similar to the above, according to the optimal charge ratio of the target mRNA and metal cations being 1:2, the mRNA is selected to be EGFP mRNA, the volume ratio of the two substances added is 1:1 as an example, the feeding ratio of the metal ions to the mRNA is as follows: 2-valent metal/mRNA (molar ratio, the same applies hereinafter) 798/1; metal 3/mRNA 798/1.5; 4 valence metal/mRNA 798/2; in specific implementation, the base number of mRNA with different sequences and lengths can be adjusted according to the principle according to the valence potential of the metal ion and the selected mRNA.
The experimental results prove that the delivery technology has universality and is suitable for mRNA with different metal ions and different sequences and lengths (see table 1 and figure 2 for details).
TABLE 1 partial nanoparticle sizes of different metal ions coordinated with mRNA of different lengths and sequences
Figure RE-GDA0003193745160000061
FIG. 2 shows Ca2+Electron micrograph of mRNA (different sequence) nanoparticles. a is Ca-EGFP mRNA; b is Ca-Luciferase mRNA.
As can be seen from Table 1 and FIG. 2, the Metal ions with different valence states and RNAs with different lengths and sequences can be well assembled through coordination, so as to form Metal-mRNA nanoparticles with uniform particle size, controllable morphology, uniform dispersion and stability.
Example 2 Metal-mRNA nanoparticle Dispersion stability evaluation
According to the method described in example 1, the metal ions are Ca2+For example, the mRNA is EGFP mRNA. An aqueous solution of metal ions was chosen at a concentration of 798mM and an aqueous solution of EGFP mRNA at a concentration of 1mM, based on the ratio of EGFP mRNA to metal cation charge: 100/1-1/10, calculating the volume of the two materials added respectively. Specifically, when the charge ratio is 100/1, the volume ratio of mRNA/metal is 200/1; when the charge ratio is selected to be 50/1, the volume ratio of mRNA/metal is 100/1; when the charge ratio is selected to be 30/1, the volume ratio of mRNA/metal is 60/1; when the charge ratio is selected to be 20/1, the volume ratio of mRNA/metal is 40/1; when the charge ratio is selected to be 10/1, the volume ratio of mRNA/metal is 20/1; when the charge ratio is selected to be 5/1, the volume ratio of mRNA/metal is 10/1; when the charge ratio is selected to be 2/1, the volume ratio of mRNA/metal is 4/1; when the charge ratio is selected to be 1/1, the volume ratio of mRNA/metal is 2/1; when the charge ratio is selected to be 1/2, the volume ratio of mRNA/metal is 1/1; when the charge ratio is selected 1/5, mRThe volume ratio of NA/metal is 1/2.5; when the charge ratio was chosen to be 1/10, the mRNA/metal volume ratio was 1/5. The other valency metals may be converted according to the relationship given in example 1. Specifically, the desired Metal-mRNA nanoparticles were synthesized as described in example 1. The dispersion stability of Metal-mRNA nanoparticles under different charge ratios is evaluated by a zeta potential method, a light transmittance method (a spectrophotometer) and other methods. (see tables 2 and 3 for details).
TABLE 2 zeta potential changes of Metal-EGFP mRNA nanoparticles at different Metal particles and ratios
Figure RE-GDA0003193745160000071
Table 3, Metal-EGFP mRNA nanoparticle dispersion stability variation conditions under different Metal particles and ratios
Figure RE-GDA0003193745160000072
As can be seen from table 2: with the increase of the charge ratio of mRNA/Metal ions, the Metal-EGFP mRNA nano system gradually becomes neutral and weak electropositive. Correspondingly, the nano system can penetrate cell membranes more easily, and can enter the interior of cells more efficiently to finish mRNA delivery.
As can be seen from Table 3, the Metal-mRNA nanosystem is more stable in this range with increasing charge ratio of mRNA/Metal ion, and the system can be kept intact before reaching the target environment.
Example 3 in vitro cytotoxicity assay
HEK293 cells (CRL-1573-293) were seeded in 96-well plates in DMEM medium (10% fetal bovine serum and 1% penicillin). Cells were incubated at 37 ℃ in the presence of 5% CO2Is incubated in an atmosphere of (2). Cells were incubated for 24 hours and then replaced with fresh medium. Different Metal ion and mRNA assemblies (prepared in example 1, Metal-EGFP mRNA, 15. mu.g), Lipofectamin 2000-EGFP mRNA (15. mu.g) and PEI-EGFP mRNA (15. mu.g) were added as commercial mRNA delivery model controls (see below for specific preparation methods). Incubating the cellsAfter 48 hours, the original medium was replaced with 100. mu.L of medium containing 0.5mg/mL of 3- (4, 5-dimethylthiazol-2-yl) -2, 5-diphenyltetrazolium (MTT). After 4 hours of incubation, the medium containing MTT was removed and carefully washed 3 times with PBS. Then, DMSO (100. mu.L) was added, absorbance at a wavelength of 570nm was measured in a BioTek Synergy H4 reader, and cell viability was calculated to verify cytotoxicity of the assembly obtained in the present invention.
Attached: synthesis method of PEI-EGFR mRNA and Lipofectamin 2000-EGFP mRNA
An appropriate amount of PEI/Lipofectamin 2000 (all commercially available) was dissolved in 15ml PBS with EGFP mRNA in a mass ratio of PEI or Lipofectamin 2000/EGFP mRNA of 3/1. Stirring at room temperature overnight, and centrifuging the obtained precipitate to obtain PEI-EGFR mRNA and Lipofectamin 2000-EGFP mRNA.
FIG. 3 shows the survival rate of cells of each group measured by MTT method, a: Zr-EGFP mRNA; Zn-EGFP mRNA; fe (II) -EGFP mRNA; d Fe (III) -EGFP mRNA; e is Mn-EGFP mRNA; f is Cu-EGFP mRNA; Ca-EGFP mRNA; h is Al-EGFP mRNA; Hf-EGFP mRNA; j lipofectamine2000-EGFP mRNA; k is PEI-EGFP mRNA.
As can be seen from FIG. 3, the Metal-EGFP mRNA nanoparticles have significantly reduced cytotoxicity and exhibit good biological safety compared with Lipofectamine2000-EGFP mRNA and PEI-EGFP mRNA.
Example 4 Metal-mRNA nanoparticle biosafety analysis
Balb/c mice were divided into 5 groups of 5 mice each, and the Balb/c mice (4-6 weeks old, female, approximately 18-20g in weight) were fed under normal conditions with a 12 hour light-dark cycle per day. PBS, PEI-EGFP mRNA (50. mu.g), PEI-EGFP mRNA (10. mu.g), Ca-EGFP mRNA (50. mu.g) and Ca-EGFP mRNA (10. mu.g) were injected tail vein separately. The Ca-EGFP mRNA was prepared as described in example 1; see example 3 for PEI-EGFP mRNA preparation. After injection, venous blood of the mice is taken 5 to 7 days, and the supernatant is obtained by centrifugation, and various indexes of liver function are measured.
Mice were divided into groups under the same conditions, and injected into tail vein: LPS (15 μ g); II: r848(15 μ g); III: PBS (15. mu.g); IV: PEI-EGFP mRNA (15. mu.g); v: Ca-EGFP mRNA (15. mu.g). The Ca-EGFP mRNA was prepared as described in example 1; PEI-EGFP mRNA was prepared according to example 3. After injection, venous blood of the mice is taken 5 to 7 days, supernatant fluid is obtained by centrifugation, and the level of inflammatory factors in tissues is determined by enzyme-linked immunosorbent assay (ELISA).
FIG. 4 shows the liver function changes in mice of different groups. Wherein a is ALT (glutamic pyruvic transaminase); b is AST (aspartate aminotransferase); c is LDH (lactate dehydrogenase); d is Total bilirubin (Total bilirubin). I: PBS; II: PEI-EGFP mRNA (50. mu.g); III: PEI-EGFP mRNA (10. mu.g); IV: Ca-EGFP mRNA (50. mu.g); v: Ca-EGFP mRNA (10. mu.g). As can be seen from FIG. 4, the Ca-EGFP mRNA nanoparticles had substantially no effect on liver function, were significantly smaller than PEI-EGFP mRNA, and were substantially equivalent to the PBS group. The results show that Metal-mRNA has little liver metabolic toxicity and good biological safety.
FIG. 5 shows the expression of different inflammatory factors in different groups of mice. Wherein a is IL-1 β; b is IL-10; c is IL-6; d is IL-8. I: LPS (positive control); II: r848 (positive control); III: PBS; IV: PEI-EGFP mRNA; v: Ca-EGFP mRNA. As can be seen in FIG. 5, the expression of inflammatory factors in vivo after the injection of Ca-EGFP mRNA nanoparticles was maintained at a low level, which was substantially equal to that of PBS. The results show that Metal-mRNA has small immunogenicity and good biological safety.
Example 5 intracellular protein expression of Metal-mRNA nanoparticles
In this example, the Luciferase mRNA was selected as the target mRNA and the intracellular protein expression experiment was performed. Specifically, the ratio of Luciferase mRNA to metal cation charge was adjusted as described in example 1: 100/1-1/10. In particular, with Ca2+For example, molar concentration ratio: 1/1818, charge ratio: 1/2. The Luciferase mRNA is mixed with CaCl2The volume ratio of the aqueous solution was 1/1 (in the case of other charge ratios, the volume ratio was calculated by the method in example 2). In particular, with Al3+For example, the concentration ratio: 1.5/1818, charge ratio: 1/2. The Luciferase mRNA and AlCl3The volume ratio of the aqueous solution was 1/1 (in the case of other charge ratios, the volume ratio was calculated by the method in example 2). In particular, with Hf4+For example, asConcentration ratio of 1/909, charge ratio: 1/2. The Luciferase mRNA and HfCl4The volume ratio of the aqueous solution was 1/1 (in the case of other charge ratios, the volume ratio was calculated by the method in example 2). The obtained nanoparticles are used for verifying intracellular delivery efficiency and protein expression efficiency of Metal-mRNA. The feasibility and universality of this method were demonstrated by measuring the bioluminescent signal of intracellular luciferase (see table 4 for details).
TABLE 4 luciferase expression levels in Metal-mRNA (luciferase mRNA) cells at different Metal particle and ratio
Figure RE-GDA0003193745160000091
Figure RE-GDA0003193745160000101
As can be seen from Table 4, the luciferase protein expression tends to increase as the ratio of metal ions increases. This is due to two reasons: firstly, the metal ion ratio is improved, which is beneficial to the stability of mRNA assembly and the intracellular expression of mRNA; and secondly, the proportion of metal ions is improved, the Zeta potential value of the assembly is close to neutral or electropositive, so that endocytosis of cells is facilitated, and the transfer efficiency is improved. The two cooperate to improve the delivery of the target mRNA and the protein expression.
Example 6 intracellular expression assay of Metal-EGFP mRNA
In this example, EGFP mRNA was selected as the delivery mRNA and Ca was selected2+As metal ions, Ca-EGFP mRNA nanoparticles were synthesized according to the method of example 1, then the Ca-EGFP mRNA (0.2. mu.g/ml) nanoparticles were incubated with HEK293 cells for 24h to express green fluorescent protein, the culture medium in the cell culture dish was removed, and washed 3 times with PBS. The expression efficiency of the green fluorescent protein of the Ca-EGFP mRNA delivery system is qualitatively and quantitatively detected by a fluorescence microscope and flow cytometry. The control group comprises Lipo2000-EGFP mRNA nanoparticles and single EGFP mRNA. Other Metal-EGFP mRNA intracellular expression experiment methodsSimilarly.
FIG. 6 shows EGFP mRNA delivery and GFP protein expression for different Metal-EGFP mRNAs, where a: Zr-EGFP mRNA, b: Zn-EGFP mRNA, c: fe (ii) -EGFP mRNA, d: fe (iii) -EGFP mRNA, e: Mn-EGFP mRNA, f: Cu-EGFP mRNA, g: Ca-EGFP mRNA, h: Al-EGFP mRNA, i: Hf-EGFP mRNA, j: lipo2000-EGFP mRNA, k: EGFP mRNA, l: PBS. As can be seen in FIG. 6, Metal-EGFP mRNA has similar high-efficiency mRNA delivery ability and protein expression efficiency compared with Lipo2000-EGFP mRNA.
FIG. 7 is a graph showing the effect of Ca-EGFP mRNA protein expression and Lipo2000-EGFP mRNA protein expression under a fluorescence microscope. As can be seen from FIG. 7, both expressed bright GFP fluorescence in the cells, and both expressed well. In conclusion, the Metal-mRNA has high-efficiency mRNA delivery capability and protein expression efficiency and excellent transformation application prospect.
The present invention has been described in detail above. It will be apparent to those skilled in the art that the invention can be practiced in a wide range of equivalent parameters, concentrations, and conditions without departing from the spirit and scope of the invention and without undue experimentation. While the invention has been described with reference to specific embodiments, it will be appreciated that the invention can be further modified. In general, this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure as come within known or customary practice within the art to which the invention pertains. The use of some of the essential features is possible within the scope of the claims attached below.
<110> Qinghua university
<120> a vector-free mRNA delivery method
<130> CGGNQALN216079
<160> 7
<170> PatentIn version 3.5
<210> 1
<211> 564
<212> RNA
<213> Artificial sequence
<400> 1
augccuggcu cagcacugcu augcugccug cucuuacuga cuggcaugag gaucagcagg 60
ggccaguaca gccgggaaga caauaacugc acccacuucc cagucggcca gagccacaug 120
cuccuagagc ugcggacugc cuucagccag gugaagacuu ucuuucaaac aaaggaccag 180
cuggacaaca uacugcuaac cgacuccuua augcaggacu uuaaggguua cuuggguugc 240
caagccuuau cggaaaugau ccaguuuuac cugguagaag ugaugcccca ggcagagaag 300
cauggcccag aaaucaagga gcauuugaac ucccugggug agaagcugaa gacccucagg 360
augcggcuga ggcgcuguca ucgauuucuc cccugugaaa auaagagcaa ggcaguggag 420
caggugaaga gugauuuuaa uaagcuccaa gaccaaggug ucuacaaggc caugaaugaa 480
uuugacaucu ucaucaacug cauagaagca uacaugauga ucaaaaugaa aagcggcggc 540
ucucaccacc accaucacca cuaa 564
<210> 2
<211> 582
<212> RNA
<213> Artificial sequence
<400> 2
augggggugc acgaaugucc ugccuggcug uggcuucucc ugucccugcu gucgcucccu 60
cugggccucc caguccuggg cgccccacca cgccucaucu gugacagccg aguccuggag 120
agguaccucu uggaggccaa ggaggccgag aauaucacga cgggcugugc ugaacacugc 180
agcuugaaug agaauaucac ugucccagac accaaaguua auuucuaugc cuggaagagg 240
auggaggucg ggcagcaggc cguagaaguc uggcagggcc uggcccugcu gucggaagcu 300
guccugcggg gccaggcccu guuggucaac ucuucccagc cgugggagcc ccugcagcug 360
cauguggaua aagccgucag uggccuucgc agccucacca cucugcuucg ggcucuggga 420
gcccagaagg aagccaucuc cccuccagau gcggccucag cugcuccacu ccgaacaauc 480
acugcugaca cuuuccgcaa acucuuccga gucuacucca auuuccuccg gggaaagcug 540
aagcuguaca caggggaggc cugcaggaca ggggacagau ga 582
<210> 3
<211> 2975
<212> RNA
<213> Artificial sequence
<400> 3
augcuagccc gcgccccucc ucgccgcccg ccgcggcugg ugcugcuccg uuugcuguug 60
cugcaucugc ugcugcucgc ccugcgcgcc cgcugccuga gcgcugagcc gggucagggc 120
gcgcagaccu gggcucgcuu cgcgcgcgcu ccugccccag aggccgcugg ccuccuccac 180
gacaccuucc ccgacgguuu ccucugggcg guaggcagcg ccgccuauca gaccgagggc 240
ggcuggcgac agcacggcaa aggcgcgucc aucugggaca cuuucaccca ucacucuggg 300
gcggccccgu ccgacucccc gaucgucgug gcgccgucgg gugccccguc gccuccccug 360
uccuccacug gagauguggc cagcgauagu uacaacaacg ucuaccgcga cacagagggg 420
cugcgcgaac ugggggucac ccacuaccgc uucuccauau cgugggcgcg ggugcucccc 480
aauggcaccg cgggcacucc caaccgcgag gggcugcgcu acuaccggcg gcugcuggag 540
cggcugcggg agcugggcgu gcagccggug guuacccugu accauuggga ccugccacag 600
cgccugcagg acaccuaugg cggaugggcc aaucgcgccc uggccgacca uuucagggau 660
uaugccgagc ucugcuuccg ccacuucggu ggucagguca aguacuggau caccauugac 720
aaccccuacg ugguggccug gcacggguau gccaccgggc gccuggcccc gggcgugagg 780
ggcagcucca ggcucgggua ccugguugcc cacaaccuac uuuuggcuca ugccaaaguc 840
uggcaucucu acaacaccuc uuuccgcccc acacagggag gccggguguc uaucgccuua 900
agcucccauu ggaucaaucc ucgaagaaug acugacuaua auaucagaga augccagaag 960
ucucuugacu uugugcuagg cugguuugcc aaacccauau uuauugaugg cgacuaccca 1020
gagaguauga agaacaaccu cucgucucuu cugccugauu uuacugaauc ugagaagagg 1080
cucaucagag gaacugcuga cuuuuuugcu cucuccuucg gaccaaccuu gagcuuucag 1140
cuauuggacc cuaacaugaa guuccgccaa uuggagucuc ccaaccugag gcagcuucug 1200
ucuuggauag aucuggaaua uaaccacccu cccauauuua uuguggaaaa uggcugguuu 1260
gucucgggaa ccaccaaaag ggaugaugcc aaauauaugu auuaucucaa gaaguucaua 1320
auggaaaccu uaaaagcaau cagacuggau ggggucgacg ucauugggua caccgcgugg 1380
ucgcucaugg acgguuucga guggcauagg ggcuacagca uccggcgagg acucuucuac 1440
guugacuuuc ugagucagga caaggagcug uugccaaagu cuucggccuu guucuaccaa 1500
aagcugauag aggacaaugg cuuuccuccu uuaccugaaa accagccccu ugaagggaca 1560
uuucccugug acuuugcuug gggaguuguu gacaacuacg uucaagugga cacuacucuc 1620
ucucaguuua cugacccgaa ugucuaucug ugggaugugc aucacaguaa gaggcuuauu 1680
aaaguagacg ggguuguagc caagaagaga aaaccuuacu guguugauuu cucugccauc 1740
cggccucaga uaaccuuacu ucgagaaaug cgggucaccc acuuucgcuu cucccuggac 1800
ugggcccuga ucuugccucu ggguaaccag acccaaguga accacacggu ucugcacuuc 1860
uaccgcugca ugaucagcga gcuggugcac gccaacauca cuccaguggu ggcccugugg 1920
cagccagcag ccccgcacca aggccugcca caugcccuug caaaacaugg ggccugggag 1980
aacccgcaca cugcucuggc guuugcagac uacgcaaacc uguguuuuaa agaguugggu 2040
cacuggguca aucucuggau caccaugaac gagccaaaca cacggaacau gaccuaucgu 2100
gccgggcacc accuccugag agcccaugcc uuggcuuggc aucuguacga ugacaaguuu 2160
agggcggcuc agaaaggcaa aauauccauc gccuugcagg cugacuggau agaaccggcc 2220
ugcccuuucu cucaaaauga caaagaagug gccgagagag uuuuggaauu ugauauaggc 2280
uggcuggcag agccuauuuu ugguuccgga gauuauccac gugugaugag ggacuggcug 2340
aaccaaaaaa acaauuuucu uuugcccuau uucaccgaag augaaaaaaa gcuaguccgg 2400
guuccuuuga cuuccuggcg gugagucauu acaccaccau ucugguagac ugggaaaagg 2460
aggauccgau gaaauacaac gauuacuugg agguacagga gaugacugac aucacauggc 2520
ucaacucucc cagucaggug gcaguggugc cuugggggcu gcgcaaagug cucaacuggc 2580
uaagguucaa guacggagac cucccgaugu augugacagc caauggaauc gaugaugacc 2640
cccacgccga gcaagacuca cugaggaucu auuauauuaa gaauuaugug aaugaggcuc 2700
ugaaagccua cguguuggac gacaucaacc uuuguggcua cuuugcguau ucacuuagug 2760
aucgcucagc ucccaagucu ggcuuuuauc gauaugcugc gaaucaguuu gagcccaaac 2820
caucuaugaa acauuacagg agaauuauug acagcaaugg cuuccugggu ucuggaacac 2880
ugggaagguu uuguccagaa gaauacacug ugugcaccga auguggauuu uuucaaaccc 2940
ggaagggcgg cucucaccac caccaucacc acuaa 2975
<210> 4
<211> 1053
<212> RNA
<213> Artificial sequence
<400> 4
augcccaaga agaagaggaa gguggccaau uuacugaccg uacaccaaaa uuugccugca 60
uuaccggucg augcaacgag ugaugagguu cgcaagaacc ugauggacau guucagggau 120
cgccaggcgu uuucugagca uaccuggaaa augcuucugu ccguuugccg gucgugggcg 180
gcauggugca aguugaauaa ccggaaaugg uuucccgcag aaccugaaga uguucgcgau 240
uaucuucuau aucuucaggc gcgcggucug gcaguaaaaa cuauccagca acauuugggc 300
cagcuaaaca ugcuucaucg ucgguccggg cugccacgac caagugacag caaugcuguu 360
ucacugguua ugcggcguau ccgaaaagaa aacguugaug ccggugaacg ugcaaaacag 420
gcucuagcgu ucgaacgcac ugauuucgac cagguucguu cacucaugga aaauagcgau 480
cgcugccagg auauacguaa ucuggcauuu cuggggauug cuuauaacac ccuguuacgu 540
auagccgaaa uugccaggau caggguuaaa gauaucucac guacugacgg ugggagaaug 600
uuaauccaua uuggcagaac gaaaacgcug guuagcaccg cagguguaga gaaggcacuu 660
agccuggggg uaacuaaacu ggucgagcga uggauuuccg ucucuggugu agcugaugau 720
ccgaauaacu accuguuuug ccgggucaga aaaaauggug uugccgcgcc aucugccacc 780
agccagcuau caacucgcgc ccuggaaggg auuuuugaag caacucaucg auugauuuac 840
ggcgcuaagg augacucugg ucagagauac cuggccuggu cuggacacag ugcccguguc 900
ggagccgcgc gagauauggc ccgcgcugga guuucaauac cggagaucau gcaagcuggu 960
ggcuggacca auguaaauau ugucaugaac uauauccgua accuggauag ugaaacaggg 1020
gcaauggugc gccugcugga agauggcgau uaa 1053
<210> 5
<211> 4203
<212> RNA
<213> Artificial sequence
<400> 5
auggccccaa agaagaagcg gaaggucggu auccacggag ucccagcagc cgacaagaag 60
uacagcaucg gccuggacau cggcaccaac ucugugggcu gggccgugau caccgacgag 120
uacaaggugc ccagcaagaa auucaaggug cugggcaaca ccgaccggca cagcaucaag 180
aagaaccuga ucggagcccu gcuguucgac agcggcgaaa cagccgaggc cacccggcug 240
aagagaaccg ccagaagaag auacaccaga cggaagaacc ggaucugcua ucugcaagag 300
aucuucagca acgagauggc caagguggac gacagcuucu uccacagacu ggaagagucc 360
uuccuggugg aagaggauaa gaagcacgag cggcacccca ucuucggcaa caucguggac 420
gagguggccu accacgagaa guaccccacc aucuaccacc ugagaaagaa acugguggac 480
agcaccgaca aggccgaccu gcggcugauc uaucuggccc uggcccacau gaucaaguuc 540
cggggccacu uccugaucga gggcgaccug aaccccgaca acagcgacgu ggacaagcug 600
uucauccagc uggugcagac cuacaaccag cuguucgagg aaaaccccau caacgccagc 660
ggcguggacg ccaaggccau ccugucugcc agacugagca agagcagacg gcuggaaaau 720
cugaucgccc agcugcccgg cgagaagaag aauggccugu ucggaaaccu gauugcccug 780
agccugggcc ugacccccaa cuucaagagc aacuucgacc uggccgagga ugccaaacug 840
cagcugagca aggacaccua cgacgacgac cuggacaacc ugcuggccca gaucggcgac 900
caguacgccg accuguuucu ggccgccaag aaccuguccg acgccauccu gcugagcgac 960
auccugagag ugaacaccga gaucaccaag gccccccuga gcgccucuau gaucaagaga 1020
uacgacgagc accaccagga ccugacccug cugaaagcuc ucgugcggca gcagcugccu 1080
gagaaguaca aagagauuuu cuucgaccag agcaagaacg gcuacgccgg cuacauugac 1140
ggcggagcca gccaggaaga guucuacaag uucaucaagc ccauccugga aaagauggac 1200
ggcaccgagg aacugcucgu gaagcugaac agagaggacc ugcugcggaa gcagcggacc 1260
uucgacaacg gcagcauccc ccaccagauc caccugggag agcugcacgc cauucugcgg 1320
cggcaggaag auuuuuaccc auuccugaag gacaaccggg aaaagaucga gaagauccug 1380
accuuccgca uccccuacua cgugggcccu cuggccaggg gaaacagcag auucgccugg 1440
augaccagaa agagcgagga aaccaucacc cccuggaacu ucgaggaagu gguggacaag 1500
ggcgcuuccg cccagagcuu caucgagcgg augaccaacu ucgauaagaa ccugcccaac 1560
gagaaggugc ugcccaagca cagccugcug uacgaguacu ucaccgugua uaacgagcug 1620
accaaaguga aauacgugac cgagggaaug agaaagcccg ccuuccugag cggcgagcag 1680
aaaaaggcca ucguggaccu gcuguucaag accaaccgga aagugaccgu gaagcagcug 1740
aaagaggacu acuucaagaa aaucgagugc uucgacuccg uggaaaucuc cggcguggaa 1800
gaucgguuca acgccucccu gggcacauac cacgaucugc ugaaaauuau caaggacaag 1860
gacuuccugg acaaugagga aaacgaggac auucuggaag auaucgugcu gacccugaca 1920
cuguuugagg acagagagau gaucgaggaa cggcugaaaa ccuaugccca ccuguucgac 1980
gacaaaguga ugaagcagcu gaagcggcgg agauacaccg gcuggggcag gcugagccgg 2040
aagcugauca acggcauccg ggacaagcag uccggcaaga caauccugga uuuccugaag 2100
uccgacggcu ucgccaacag aaacuucaug cagcugaucc acgacgacag ccugaccuuu 2160
aaagaggaca uccagaaagc ccaggugucc ggccagggcg auagccugca cgagcacauu 2220
gccaaucugg ccggcagccc cgccauuaag aagggcaucc ugcagacagu gaagguggug 2280
gacgagcucg ugaaagugau gggccggcac aagcccgaga acaucgugau cgaaauggcc 2340
agagagaacc agaccaccca gaagggacag aagaacagcc gcgagagaau gaagcggauc 2400
gaagagggca ucaaagagcu gggcagccag auccugaaag aacaccccgu ggaaaacacc 2460
cagcugcaga acgagaagcu guaccuguac uaccugcaga augggcggga uauguacgug 2520
gaccaggaac uggacaucaa ccggcugucc gacuacgaug uggaccauau cgugccucag 2580
agcuuucuga aggacgacuc caucgacaac aaggugcuga ccagaagcga caagaaccgg 2640
ggcaagagcg acaacgugcc cuccgaagag gucgugaaga agaugaagaa cuacuggcgg 2700
cagcugcuga acgccaagcu gauuacccag agaaaguucg acaaucugac caaggccgag 2760
agaggcggcc ugagcgaacu ggauaaggcc ggcuucauca agagacagcu gguggaaacc 2820
cggcagauca caaagcacgu ggcacagauc cuggacuccc ggaugaacac uaaguacgac 2880
gagaaugaca agcugauccg ggaagugaaa gugaucaccc ugaaguccaa gcuggugucc 2940
gauuuccgga aggauuucca guuuuacaaa gugcgcgaga ucaacaacua ccaccacgcc 3000
cacgacgccu accugaacgc cgucguggga accgcccuga ucaaaaagua cccuaagcug 3060
gaaagcgagu ucguguacgg cgacuacaag guguacgacg ugcggaagau gaucgccaag 3120
agcgagcagg aaaucggcaa ggcuaccgcc aaguacuucu ucuacagcaa caucaugaac 3180
uuuuucaaga ccgagauuac ccuggccaac ggcgagaucc ggaagcggcc ucugaucgag 3240
acaaacggcg aaaccgggga gaucgugugg gauaagggcc gggauuuugc caccgugcgg 3300
aaagugcuga gcaugcccca agugaauauc gugaaaaaga ccgaggugca gacaggcggc 3360
uucagcaaag agucuauccu gcccaagagg aacagcgaua agcugaucgc cagaaagaag 3420
gacugggacc cuaagaagua cggcggcuuc gacagcccca ccguggccua uucugugcug 3480
gugguggcca aaguggaaaa gggcaagucc aagaaacuga agagugugaa agagcugcug 3540
gggaucacca ucauggaaag aagcagcuuc gagaagaauc ccaucgacuu ucuggaagcc 3600
aagggcuaca aagaagugaa aaaggaccug aucaucaagc ugccuaagua cucccuguuc 3660
gagcuggaaa acggccggaa gagaaugcug gccucugccg gcgaacugca gaagggaaac 3720
gaacuggccc ugcccuccaa auaugugaac uuccuguacc uggccagcca cuaugagaag 3780
cugaagggcu cccccgagga uaaugagcag aaacagcugu uuguggaaca gcacaagcac 3840
uaccuggacg agaucaucga gcagaucagc gaguucucca agagagugau ccuggccgac 3900
gcuaaucugg acaaagugcu guccgccuac aacaagcacc gggauaagcc caucagagag 3960
caggccgaga auaucaucca ccuguuuacc cugaccaauc ugggagcccc ugccgccuuc 4020
aaguacuuug acaccaccau cgaccggaag agguacacca gcaccaaaga ggugcuggac 4080
gccacccuga uccaccagag caucaccggc cuguacgaga cacggaucga ccugucucag 4140
cugggaggcg acaaaaggcc ggcggccacg aaaaaggccg gccaggcaaa aaagaaaaag 4200
uaa 4203
<210> 6
<211> 798
<212> RNA
<213> Artificial sequence
<400> 6
auggugagca agggcgagga gcuguucacc gggguggugc ccauccuggu cgagcuggac 60
ggcgacguaa acggccacaa guucagcgug uccggcgagg gcgagggcga ugccaccuac 120
ggcaagcuga cccugaaguu caucugcacc accggcaagc ugcccgugcc cuggcccacc 180
cucgugacca cccugaccua cggcgugcag ugcuucagcc gcuaccccga ccacaugaag 240
cagcacgacu ucuucaaguc cgccaugccc gaaggcuacg uccaggagcg caccaucuuc 300
uucaaggacg acggcaacua caagacccgc gccgagguga aguucgaggg cgacacccug 360
gugaaccgca ucgagcugaa gggcaucgac uucaaggagg acggcaacau ccuggggcac 420
aagcuggagu acaacuacaa cagccacaac gucuauauca uggccgacaa gcagaagaac 480
ggcaucaagg ugaacuucaa gauccgccac aacaucgagg acggcagcgu gcagcucgcc 540
gaccacuacc agcagaacac ccccaucggc gacggccccg ugcugcugcc cgacaaccac 600
uaccugagca cccaguccgc ccugagcaaa gaccccaacg agaagcgcga ucacaugguc 660
cugcuggagu ucgugaccgc cgccgggauc acucucggca uggacgagcu guacaagucc 720
ggagcugcgg ccgcugccgc ugcggcagcg gccgaauucc ccgggcucga gaagcuugga 780
uccaccggau cuagauaa 798
<210> 7
<211> 1818
<212> RNA
<213> Artificial sequence
<400> 7
gaaaccgaca ccaccaugaa gauaauaauu cugucuguua uauuggccua cugugucacc 60
gacaacuguc aagaugcaug uccuguagaa gcggaaccgc caucaaguac accaacaguu 120
ccaacuucuu gugaagcuaa agaaggagaa uguauagaua ccagaugcgc aacauguaaa 180
cgagauauac uaucagaugg acugugugaa aauaaaccag ggaagacaug cuguagaaug 240
ugccaguaug ugauugaaug cagaguagaa gcagcugguu auuuuagaac guuuuacggc 300
aaaagauuua auuuucagga accugguaaa uaugugcugg cuaggggaac caaggguggc 360
gauuggucug uaacccucac cauggagaau cuagauggac agaagggagc ugugcugacu 420
aagacaacac uggagguugc aggagacgua auagacauua cucaagcuac ugcagauccu 480
aucacaguua acggaggagc ugacccaguu aucgcuaacc cguucacaau uggugaggug 540
accauugcug uuguugaaau accgggcuuc aauaucacag ucaucgaauu cuuuaaacua 600
aucgugauug auauucuggg aggaagaucu gugagaauug cuccagacac agcaaacaaa 660
ggacugauau cugguaucug ugguaaucug gagaugaaug acgcugauga cuuuacuaca 720
gaugcagauc agcuggcgau ccaacccaac auaaacaaag aguucgacgg cugcccauuc 780
uauggcaauc cuucugauau cgaauacugc aaaggucuga uggagccaua cagagcugua 840
ugucguaaca auaucaacuu cuacuauuac acucuauccu gugccuucgc uuacuguaug 900
ggaggagaag aaagagcuaa acacguccuu uucgacuaug uugagacaug cgcugcgccg 960
gaaacgagag gaacgugugu uuuaucagga cauacuuucu augacacauu cgacaaagca 1020
agauaucaau uccagggccc augcaaggag auucugaugg ccgcagacug uuacuggaac 1080
acaugggaug uaaagguuuc acauagagac gucgaaucau acacugaggu agagaaagua 1140
acaaucagga aacagucaac uguaguagau cucauugugg auggcaagca ggucaagguu 1200
ggaggagugg auguaucuau cccguacagc ucugagaaca cuuccauaua cuggcaggau 1260
ggagacaucc ugacgacggc cauccuaccu gaagcucuug ucguuaaguu caacuuuaag 1320
cagcuccuug uaguucauau cagagaucca uucgauggaa agacaugcgg cauauguggu 1380
aacuauaauc aagauucaac ugaugauuuc uuugacgcag aaggagcaug cgcucuaacc 1440
cccaaccccc caggauguac agaggaacag aaaccagaag cugagcgacu uugcaauaau 1500
cucuuugauu cuucuaucga cgagaaaugu aaugucugcu acaagccuga ccggauugcc 1560
cgauguaugu acgaguauug ccugagggga caacaaggau uuugugacca ugcuugggag 1620
uucaagaaag aaugcuacau aaaacaugga gacacucuag aaguaccacc ugaaugucaa 1680
uaaacguaca aagauacaga agcuaaggcu acuacagcag aagauaaaaa agaaacugua 1740
guucuucaaa aaccguguau uuuauguacu cauuguuuaa uuagaggaaa auaaauuguu 1800
auuaucauaa cuuaaagu 1818

Claims (10)

1. A method of carrier-free RNA delivery comprising the steps of:
(A1) synthesizing carrier-free Metal-mRNA nanoparticles by coordination-driven self-assembly effect of Metal cations and RNA molecules;
(A2) the delivery of the RNA molecule is achieved in the form of the carrier-free Metal-mRNA nanoparticles.
2. A method of making carrier-free Metal-mRNA nanoparticles, comprising the steps of:
(B1) mixing the target mRNA and the metal cation according to the charge ratio of 500/1-1/500;
(B2) standing the treated system (B1) at 37-60 ℃ and naturally cooling;
(B3) and (B2) centrifuging the system after treatment, and collecting precipitates to obtain the carrier-free Metal-mRNA nano particles.
3. The method of claim 2, wherein: in the step (B1), the charge ratio of the mRNA of interest and the metal cation is 100/1 to 1/10;
further, the charge ratio of the target mRNA and the metal cation is 1: 2.
4. A method according to claim 2 or 3, characterized in that: in the step (B1), mixing the mRNA of interest and the metal cation is achieved by: and dropwise adding the aqueous solution containing the metal cations into the target mRNA aqueous solution, and stirring for 1-10 min.
5. The method according to any one of claims 2-4, wherein: in the step (B2), the standing time is 1.5-4 h; and/or
In the step (B2), the temperature after the natural cooling is room temperature.
6. The method according to any one of claims 2-5, wherein: in the step (B3), before the centrifugation, a step of washing the treated system of (B2) with deionized water is further included; and/or
In the step (B3), the centrifugation is 10000-; and/or
In the step (B3), the centrifugation further comprises a step of dispersing the precipitate in deionized water for storage.
7. The method according to any one of claims 1-6, wherein: the valence of the metal cation is from +2 to + 8;
the metal cation is any metal cation that can participate in the phosphate coordination reaction of RNA and can be bioavailable.
8. The method according to any one of claims 1-7, wherein: the mRNA is naked mRNA or modified mRNA.
9. Carrier-free Metal-mRNA nanoparticles prepared by the method of any one of claims 2-8.
10. Use of the method of any one of claims 1-8 or the carrier-free Metal-mRNA nanoparticles of claim 8 for carrier-free delivery of an mRNA of interest to a cell or tissue or organism.
CN202110746485.9A 2021-07-01 2021-07-01 Carrier-free mRNA delivery method Pending CN113413467A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110746485.9A CN113413467A (en) 2021-07-01 2021-07-01 Carrier-free mRNA delivery method

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110746485.9A CN113413467A (en) 2021-07-01 2021-07-01 Carrier-free mRNA delivery method

Publications (1)

Publication Number Publication Date
CN113413467A true CN113413467A (en) 2021-09-21

Family

ID=77720053

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110746485.9A Pending CN113413467A (en) 2021-07-01 2021-07-01 Carrier-free mRNA delivery method

Country Status (1)

Country Link
CN (1) CN113413467A (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114748446A (en) * 2022-02-18 2022-07-15 广东粤港澳大湾区国家纳米科技创新研究院 Aluminum-based self-assembly delivery system of mRNA (messenger ribonucleic acid), and preparation method and application thereof
CN116059182A (en) * 2022-10-24 2023-05-05 荣灿生物医药技术(上海)有限公司 Nanoparticle, and preparation method and application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101180400A (en) * 2004-05-24 2008-05-14 Mida科技有限公司 Nanoparticles comprising rna ligands
CN111110846A (en) * 2018-10-30 2020-05-08 国家纳米科学中心 Metal-nucleic acid nano-particle and preparation method and application thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101180400A (en) * 2004-05-24 2008-05-14 Mida科技有限公司 Nanoparticles comprising rna ligands
CN111110846A (en) * 2018-10-30 2020-05-08 国家纳米科学中心 Metal-nucleic acid nano-particle and preparation method and application thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BEI LIU等: "《Metal–DNA Coordination-Driven Self-Assembly: A Conceptual Methodology to Expand the Repertoire of DNA Nanobiotechnology》", 《CHEM. EUR. J.》 *
MENGYUAN LI等: "《Engineering Multifunctional DNA Hybrid Nanospheres through Coordination-Driven Self-Assembly》", 《ANGEW. CHEM. INT. ED.》 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114748446A (en) * 2022-02-18 2022-07-15 广东粤港澳大湾区国家纳米科技创新研究院 Aluminum-based self-assembly delivery system of mRNA (messenger ribonucleic acid), and preparation method and application thereof
WO2023155862A1 (en) * 2022-02-18 2023-08-24 广东粤港澳大湾区国家纳米科技创新研究院 Aluminum-based self-assembled delivery system for mrna, method for preparing same, and use thereof
CN114748446B (en) * 2022-02-18 2023-11-28 广州瑞贝斯药业有限公司 Aluminum-based self-assembly delivery system of mRNA (messenger ribonucleic acid) as well as preparation method and application thereof
CN116059182A (en) * 2022-10-24 2023-05-05 荣灿生物医药技术(上海)有限公司 Nanoparticle, and preparation method and application thereof

Similar Documents

Publication Publication Date Title
Lee et al. Synthetically modified guide RNA and donor DNA are a versatile platform for CRISPR-Cas9 engineering
Rui et al. Reducible branched ester-amine quadpolymers (rBEAQs) codelivering plasmid DNA and RNA oligonucleotides enable CRISPR/Cas9 genome editing
Rojanarata et al. Chitosan-thiamine pyrophosphate as a novel carrier for siRNA delivery
CN113413467A (en) Carrier-free mRNA delivery method
JP5697123B2 (en) Method for introducing nucleic acid into cells using acidified polyethylenimine
Sun et al. Enhancing the therapeutic delivery of oligonucleotides by chemical modification and nanoparticle encapsulation
CN106572974B (en) Compositions and methods having lipid aggregates for efficient delivery of molecules to cells
CN1178973C (en) Method for preparing functionalised poly alkylenimides, composition containing same and uses thereof
CN110478322B (en) Nucleic acid drug compound and preparation method and application thereof
WO2014057432A9 (en) Multicomponent lipid nanoparticles and processes for the preparation thereof
CN110638759A (en) A preparation for in vitro transfection and in vivo mRNA delivery
CN109762821B (en) Interfering RNA for inhibiting expression of AFAP1-AS1 and application of interfering RNA in increasing sensitivity of breast cancer radiotherapy
Yoo et al. Large-scale preparation of extracellular vesicles enriched with specific microRNA
Xia et al. Tissue-specific delivery of oligonucleotides
CN112941072A (en) Nucleic acid self-assembly structure and preparation method and application thereof
US20230287408A1 (en) Nucleic acid constructs for delivering polynucleotides into exosomes
CN106188223B (en) Compound containing dipeptide lipid cations and preparation method and application thereof
JP6868306B2 (en) Cell introduction agent
Yao et al. Efficient Self‐Assembled DNA Nanoparticles through Rolling Circle Amplification for siRNA Delivery in vitro
US20230000990A1 (en) Non-viral vectors comprising polypropyleneimine
JP4827358B2 (en) Method for producing functional polyalkyleneimine, composition containing said compound and use thereof
CN112168980B (en) Gene-activated antibacterial bioactive bone repair material and preparation method and application thereof
CN106916316B (en) Preparation of nano material and preparation method of gene vector system thereof
CN106924746B (en) Composite gene vector and application thereof
CN114306278A (en) Preparation method of EV-loaded CPP-siRNA nanoparticles, product and application thereof

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