+

CN119286942B - Nanometer carrier for aquatic animal gene editing and gene editing method - Google Patents

Nanometer carrier for aquatic animal gene editing and gene editing method Download PDF

Info

Publication number
CN119286942B
CN119286942B CN202411826906.9A CN202411826906A CN119286942B CN 119286942 B CN119286942 B CN 119286942B CN 202411826906 A CN202411826906 A CN 202411826906A CN 119286942 B CN119286942 B CN 119286942B
Authority
CN
China
Prior art keywords
tnp
nanoparticles
gene editing
mal
mrna
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
CN202411826906.9A
Other languages
Chinese (zh)
Other versions
CN119286942A (en
Inventor
李运东
曹紫洋
黄圣
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.)
South China Sea Fisheries Research Institute Chinese Academy Fishery Sciences
Guangzhou First Peoples Hospital
Original Assignee
South China Sea Fisheries Research Institute Chinese Academy Fishery Sciences
Guangzhou First Peoples Hospital
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 South China Sea Fisheries Research Institute Chinese Academy Fishery Sciences, Guangzhou First Peoples Hospital filed Critical South China Sea Fisheries Research Institute Chinese Academy Fishery Sciences
Priority to CN202411826906.9A priority Critical patent/CN119286942B/en
Publication of CN119286942A publication Critical patent/CN119286942A/en
Application granted granted Critical
Publication of CN119286942B publication Critical patent/CN119286942B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • 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
    • C12N15/873Techniques for producing new embryos, e.g. nuclear transfer, manipulation of totipotent cells or production of chimeric embryos
    • 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
    • C12N15/10Processes for the isolation, preparation or purification of DNA or RNA
    • C12N15/102Mutagenizing nucleic acids
    • 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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • 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
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases [RNase]; Deoxyribonucleases [DNase]

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Biophysics (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Developmental Biology & Embryology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The invention discloses a nano-carrier for gene editing of aquatic animals, which comprises a) TNP nano-particles, b) Cas9 mRNA and sgRNA. Also discloses the application of the TNP nano-particles as mRNA delivery vectors and a gene editing method based on the nano-vectors, the method realizes a high-efficiency and low-damage gene editing and delivering mode by encapsulating Cas9 mRNA and sgRNA by using TNP nano particles with good biocompatibility. The nanocarriers optimize transfection efficiency and specificity of gene editing by non-invasive delivery into aquatic animal embryos. In addition, the method can improve the efficiency and the precision of gene editing, enhance the survival rate of embryos, has simple and safe operation and wide applicability, can support customized gene editing, and is beneficial to promoting the application of gene editing in aquatic germplasm genetic improvement.

Description

一种用于水产动物基因编辑的纳米载体及基因编辑方法A nanocarrier for gene editing of aquatic animals and a gene editing method

技术领域Technical Field

本发明属于基因工程技术领域,具体涉及一种用于水产动物基因编辑的纳米载体及基因编辑方法。The present invention belongs to the technical field of genetic engineering, and in particular relates to a nanocarrier for gene editing of aquatic animals and a gene editing method.

背景技术Background Art

随着全球水产养殖业的快速发展,针对疾病抗性、生长速度和肉质改善等关键性状的遗传改良需求不断增长。当前,传统的育种方法如选择性繁殖已不能完全满足产业对快速、精确改良的需求。尤其面临新疾病威胁和环境适应性挑战时,市场对高质量水产品的需求推动了对更先进的基因编辑技术的探索。然而,传统的基因编辑技术,尤其是微注射等物理递送方法,尽管在一定程度上实现了基因编辑的目的,但由于其技术复杂性高、操作要求严格、对胚胎的损伤大以及低存活率等问题,这些方法的应用受到了显著限制。微注射技术的高依赖性和低适应性,尤其在需要处理大量样本的商业养殖场景中,成为其普及的主要障碍。With the rapid development of the global aquaculture industry, the demand for genetic improvement of key traits such as disease resistance, growth rate and meat quality improvement is growing. At present, traditional breeding methods such as selective breeding can no longer fully meet the industry's needs for rapid and precise improvement. Especially in the face of new disease threats and environmental adaptability challenges, the market demand for high-quality aquatic products has driven the exploration of more advanced gene editing technologies. However, although traditional gene editing technologies, especially physical delivery methods such as microinjection, have achieved the purpose of gene editing to a certain extent, their application has been significantly limited due to their high technical complexity, strict operating requirements, great damage to embryos, and low survival rates. The high dependence and low adaptability of microinjection technology, especially in commercial aquaculture scenarios that require the processing of large numbers of samples, have become the main obstacles to its popularization.

面对这些挑战,开发一种既高效又低损伤的新型基因递送技术成为当务之急。脂质纳米粒子(Lipid Nanoparticles, LNP)技术的引入,为解决这一难题提供了可行的解决方案。LNP技术通过将基因编辑工具如Cas9蛋白和单向导RNA(sgRNA)封装在纳米级脂质颗粒中,保护RNA不被体内酶降解,并有效地将其递送到目标细胞内部。相较于微注射技术,LNP递送系统具有非侵入性,大大减少了物理穿刺对胚胎的伤害,从而提高了操作的安全性和胚胎的存活率。Faced with these challenges, it is imperative to develop a new gene delivery technology that is both efficient and less damaging. The introduction of lipid nanoparticles (LNP) technology provides a feasible solution to this problem. LNP technology encapsulates gene editing tools such as Cas9 protein and single guide RNA (sgRNA) in nanoscale lipid particles, protects RNA from degradation by enzymes in the body, and effectively delivers it to the target cells. Compared with microinjection technology, the LNP delivery system is non-invasive, greatly reducing the damage to embryos caused by physical puncture, thereby improving the safety of the operation and the survival rate of the embryo.

此外,LNP递送系统的高递送效率和优异的细胞渗透性能使其在基因编辑中展现出高度的特异性和效率。通过精确控制LNP颗粒的大小、表面电荷和脂质组成,可以进一步优化其递送性能,增强其在特定细胞类型中的靶向性。LNP技术的另一个显著优势在于其可扩展性,它适合于大规模生产和应用,能够满足商业养殖中对快速、大量基因编辑的需求。这一特点使得LNP递送系统特别适合在水产养殖业中推广使用,有助于实现在不牺牲遗传多样性的前提下,快速提升水产动物的生产性状,如增强病害抵抗力、改善生长速度和肉质等。In addition, the high delivery efficiency and excellent cell penetration of the LNP delivery system enable it to exhibit high specificity and efficiency in gene editing. By precisely controlling the size, surface charge and lipid composition of LNP particles, its delivery performance can be further optimized and its targeting in specific cell types can be enhanced. Another significant advantage of LNP technology is its scalability. It is suitable for large-scale production and application, and can meet the needs of rapid and large-scale gene editing in commercial farming. This feature makes the LNP delivery system particularly suitable for promotion and use in the aquaculture industry, which helps to quickly improve the production traits of aquatic animals without sacrificing genetic diversity, such as enhancing disease resistance, improving growth rate and meat quality, etc.

当前的基因编辑技术,包括LNP和Cas9工具,虽然在细胞和一些动物模型中展现了较高的基因编辑效率,但在水产动物中应用时面临一定局限性。首先,传统的物理递送方法(如微注射)操作复杂且对技术人员要求高,尤其是在处理大量水产动物胚胎时效率较低。此外,这类物理递送方法容易对胚胎造成机械性损伤,导致存活率下降,增加了实验失败的风险。其次,虽然LNP技术能够改善递送效率,但它在水产动物中尚未完全优化,特别是在不同水产物种的应用上,可能还需要对颗粒的大小、表面修饰等进行调整,以确保递送的精准性和高效性。Although current gene editing technologies, including LNP and Cas9 tools, have demonstrated high gene editing efficiency in cells and some animal models, they face certain limitations when used in aquatic animals. First, traditional physical delivery methods (such as microinjection) are complex to operate and require high technical staff, especially when dealing with a large number of aquatic animal embryos, which are less efficient. In addition, this type of physical delivery method can easily cause mechanical damage to the embryos, resulting in a decrease in survival rate and increasing the risk of experimental failure. Secondly, although LNP technology can improve delivery efficiency, it has not yet been fully optimized in aquatic animals, especially in the application of different aquatic species. It may also be necessary to adjust the particle size, surface modification, etc. to ensure the accuracy and efficiency of delivery.

因此,直接采用LNP和Cas9虽然在一定程度上提升了基因编辑的效率,但在水产养殖环境下,现有的递送系统并不能完全解决效率和胚胎存活率低的问题。Therefore, although the direct use of LNP and Cas9 has improved the efficiency of gene editing to a certain extent, the existing delivery system cannot completely solve the problems of low efficiency and embryo survival rate in aquaculture environments.

本发明的改进旨在针对水产动物的特殊生物环境和应用需求,优化纳米载体的设计与递送路径,确保基因编辑工具能够更加高效地到达目标细胞,并减少对胚胎的损伤。此外,本发明还着重解决大规模应用中的实际操作问题,提供一种更加易于操作、适应性强且在商业环境中可行的基因编辑解决方案。这样不仅可以提高编辑效率,还能显著提升胚胎的存活率,满足水产养殖中快速改良的需求。The improvement of the present invention aims to optimize the design and delivery path of nanocarriers for the special biological environment and application requirements of aquatic animals, ensure that gene editing tools can reach target cells more efficiently, and reduce damage to embryos. In addition, the present invention also focuses on solving practical operational problems in large-scale applications, providing a gene editing solution that is easier to operate, highly adaptable, and feasible in a commercial environment. This can not only improve editing efficiency, but also significantly improve the survival rate of embryos, meeting the needs of rapid improvement in aquaculture.

发明内容Summary of the invention

本发明目的在于提供一种用于水产动物基因编辑的纳米载体。The present invention aims to provide a nanocarrier for gene editing of aquatic animals.

本发明的目的还在于提供一种基于上述纳米载体的水产动物Cas9基因编辑方法。The present invention also aims to provide a method for editing the Cas9 gene in aquatic animals based on the above-mentioned nanocarrier.

本发明的上述第一个目的可以通过以下技术方案来实现:一种用于水产动物基因编辑的纳米载体,包括 a) TNP纳米颗粒以及 b) Cas9 mRNA和sgRNA。The above-mentioned first object of the present invention can be achieved by the following technical scheme: a nanocarrier for gene editing of aquatic animals, comprising a) TNP nanoparticles and b) Cas9 mRNA and sgRNA.

优选地,所述a)中的TNP纳米颗粒通过以下方法制备获得:Preferably, the TNP nanoparticles in a) are prepared by the following method:

(1)取马来酰亚胺化聚乙二醇Mal-PEG-OH和丙交酯D, L-LA以及乙交酯GA加入反应容器中,加入磁子并且在油浴中加热至130~150℃,持续搅拌直至反应容器中固体全部溶解,随后,在搅拌条件下滴加异辛酸亚锡Sn(Oct)2后继续搅拌反应2~5 h,反应完毕浓缩产物沉淀在甲醇-乙醚混合液中,抽滤干燥得淡黄色Mal-PEG-b-PLGA;(1) Maleimidized polyethylene glycol Mal-PEG-OH, lactide D, L-LA and glycolide GA are added to a reaction vessel, a magnetic particle is added and the mixture is heated to 130-150°C in an oil bath, and stirring is continued until all the solids in the reaction vessel are dissolved. Subsequently, stannous isooctanoate Sn(Oct) 2 is added dropwise under stirring and the reaction is continued for 2-5 h. After the reaction is completed, the concentrated product is precipitated in a methanol-ether mixture, and filtered and dried to obtain light yellow Mal-PEG- b -PLGA;

(2)取Mal-PEG-b-PLGA加入圆底容器中,再加入搅拌磁子和二甲基亚砜DMSO溶解Mal-PEG-b-PLGA,然后在搅拌条件下加入超纯水并继续搅拌,制备得Mal-NP纳米颗粒;(2) Mal-PEG- b -PLGA was added to a round-bottom container, and a stirring magnetic bar and dimethyl sulfoxide (DMSO) were added to dissolve Mal-PEG- b -PLGA. Ultrapure water was then added under stirring conditions and stirring was continued to prepare Mal-NP nanoparticles.

(3)将Mal-NP纳米颗粒在超纯水中透析过夜除去二甲基亚砜DMSO,收集透析袋内的纳米颗粒溶液转移至圆底容器中,加入穿膜肽TAT在氮气保护下搅拌反应,反应结束后,收集颗粒溶液离心,收集下层颗粒沉淀,冻干后得到TAT修饰的TAT-Mal-NP材料,简称TNP纳米颗粒。(3) The Mal-NP nanoparticles were dialyzed in ultrapure water overnight to remove dimethyl sulfoxide (DMSO). The nanoparticle solution in the dialysis bag was collected and transferred to a round-bottom container. The cell-penetrating peptide TAT was added and stirred for reaction under nitrogen protection. After the reaction, the particle solution was collected and centrifuged. The particle precipitate in the lower layer was collected and freeze-dried to obtain TAT-modified TAT-Mal-NP material, referred to as TNP nanoparticles.

优选地,步骤(1)中所述马来酰亚胺化聚乙二醇Mal-PEG-OH、丙交酯D, L-LA与乙交酯GA的摩尔比为0.2:10.8~11.5:5.0~5.2。Preferably, in step (1), the molar ratio of maleimidized polyethylene glycol Mal-PEG-OH, lactide D, L-LA and glycolide GA is 0.2:10.8~11.5:5.0~5.2.

更佳地,步骤(1)中所述马来酰亚胺化聚乙二醇Mal-PEG-OH、丙交酯D, L-LA与乙交酯GA的摩尔比为0.2:11.111:5.173。More preferably, in step (1), the molar ratio of maleimidized polyethylene glycol Mal-PEG-OH, lactide D, L-LA and glycolide GA is 0.2:11.111:5.173.

更佳地,步骤(1)中加入磁子并且在油浴中加热至130℃。More preferably, in step (1), a magnetron is added and the mixture is heated to 130° C. in an oil bath.

更佳地,步骤(1)中在搅拌条件下滴加异辛酸亚锡Sn(Oct)2后继续搅拌反应3 h。More preferably, in step (1), Sn(Oct) 2 is added dropwise under stirring and the stirring reaction is continued for 3 h.

优选地,步骤(1)中所述甲醇-乙醚混合液中所述甲醇与乙醚的体积比为1:9~11。Preferably, the volume ratio of methanol to ether in the methanol-ether mixture in step (1) is 1:9-11.

更佳地,步骤(1)中所述甲醇-乙醚混合液中所述甲醇与乙醚的体积比为1:10。More preferably, the volume ratio of methanol to ether in the methanol-ether mixture in step (1) is 1:10.

优选地,步骤(2)中所述Mal-PEG-b-PLGA与所述二甲基亚砜DMSO的用量关系为145~155mg:15~25mL。Preferably, in step (2), the dosage of Mal-PEG- b -PLGA and dimethyl sulfoxide (DMSO) is 145-155 mg: 15-25 mL.

更佳地,步骤(2)中所述Mal-PEG-b-PLGA与所述二甲基亚砜DMSO的用量关系为150mg:20mL。More preferably, in step (2), the dosage of Mal-PEG- b -PLGA and dimethyl sulfoxide (DMSO) is 150 mg:20 mL.

优选地,步骤(2)中在搅拌条件下缓慢加入80~120 mL超纯水连续搅拌1.5~2.5 h,制备得Mal-NP纳米颗粒。Preferably, in step (2), 80-120 mL of ultrapure water is slowly added under stirring and stirred continuously for 1.5-2.5 h to prepare Mal-NP nanoparticles.

更佳地,步骤(2)中在搅拌条件下缓慢加入100 mL超纯水连续搅拌2 h,制备得Mal-NP纳米颗粒。More preferably, in step (2), 100 mL of ultrapure water is slowly added under stirring conditions and stirred continuously for 2 h to prepare Mal-NP nanoparticles.

优选地,步骤(3)中所述Mal-NP纳米颗粒与所述穿膜肽TAT的质量份配比为14~16:1.5~2.5。Preferably, in step (3), the mass ratio of the Mal-NP nanoparticles to the cell-penetrating peptide TAT is 14-16:1.5-2.5.

更佳地,步骤(3)中所述Mal-NP纳米颗粒与所述穿膜肽TAT的质量份配比为15:2。More preferably, in step (3), the mass ratio of the Mal-NP nanoparticles to the cell-penetrating peptide TAT is 15:2.

所述穿膜肽TAT为一种靶向配体,可以利用靶向配体增强纳米载体与水产动物胚胎细胞的亲和力,以及通过特定的浓度控制,提高基因编辑的精确性和操作的灵活性,其具有与所述水产动物胚胎细胞膜表面的高亲和力,可以与水产动物胚胎细胞膜进行融合或嵌入细胞膜,用于增强纳米载体与水产动物胚胎细胞的相互作用力。The transmembrane peptide TAT is a targeting ligand that can be used to enhance the affinity between nanocarriers and aquatic animal embryonic cells, and to improve the accuracy of gene editing and the flexibility of operation through specific concentration control. It has a high affinity with the surface of the aquatic animal embryonic cell membrane and can fuse with or embed into the cell membrane of the aquatic animal embryonic cell membrane to enhance the interaction between the nanocarrier and the aquatic animal embryonic cells.

优选地,步骤(3)中所述TNP纳米颗粒的平均粒径范围为50~200纳米。Preferably, the average particle size of the TNP nanoparticles in step (3) is in the range of 50 to 200 nanometers.

本发明的核心是使用经特殊设计的TNP纳米粒子,其具有良好的生物相容性和较低的免疫原性,可以有效减少对胚胎的物理和化学损伤。通过精确控制纳米粒子的大小、表面电荷和化学性质,优化其与胚胎膜的亲和力,从而提高转染效率和特异性。The core of the present invention is to use specially designed TNP nanoparticles, which have good biocompatibility and low immunogenicity, and can effectively reduce physical and chemical damage to the embryo. By precisely controlling the size, surface charge and chemical properties of the nanoparticles, the affinity with the embryo membrane is optimized, thereby improving the transfection efficiency and specificity.

本发明还提供了上述TNP纳米颗粒作为mRNA递送载体的应用。The present invention also provides the use of the TNP nanoparticles as mRNA delivery carriers.

应用时,可以将TNP材料先溶于氯仿中,再加入(2,3-二油酰基-丙基)-三甲胺DOTAP的氯仿溶液以及mRNA的超纯水溶液,得混合材料溶液,将所述混合材料溶液在冰浴中进行第一次超声,超声结束后加入DEPC水,混匀;随后将离心管放在冰浴中进行第二次超声,超声结束后,所得颗粒溶液即为包载mRNA物质的TNP纳米颗粒,其中TNP纳米颗粒为递送mRNA等基因物质的高分子纳米载体。When used, the TNP material can be first dissolved in chloroform, and then a chloroform solution of (2,3-dioleoyl-propyl)-trimethylamine DOTAP and an ultrapure aqueous solution of mRNA are added to obtain a mixed material solution. The mixed material solution is subjected to a first ultrasound in an ice bath, and after the ultrasound, DEPC water is added and mixed. Subsequently, the centrifuge tube is placed in an ice bath and subjected to a second ultrasound. After the ultrasound, the obtained particle solution is TNP nanoparticles encapsulating mRNA substances, wherein the TNP nanoparticles are polymer nanocarriers for delivering genetic substances such as mRNA.

本发明提供的用于水产动物基因编辑的纳米载体,可以解决当前水产动物基因编辑技术中存在的效率低下和胚胎存活率低的问题,其利用生物相容性良好的TNP纳米载体,可以有效封装Cas9 mRNA和sgRNA,通过非侵入式的方式递送到水产动物(如斑马鱼、斑节对虾等)的胚胎中。The nanocarrier for gene editing of aquatic animals provided by the present invention can solve the problems of low efficiency and low embryo survival rate in the current gene editing technology of aquatic animals. It utilizes TNP nanocarriers with good biocompatibility to effectively encapsulate Cas9 mRNA and sgRNA, and deliver them to the embryos of aquatic animals (such as zebrafish, giant tiger shrimp, etc.) in a non-invasive manner.

优选地,本发明所述用于水产动物基因编辑的纳米载体的制备方法,包括以下步骤:将Cas9mRNA和sgRNA的超纯水溶液与TNP和DOTAP混合,经过两次超声过程制备得到用于水产动物基因编辑的纳米载体。Preferably, the method for preparing a nanocarrier for gene editing of aquatic animals according to the present invention comprises the following steps: mixing an ultrapure aqueous solution of Cas9mRNA and sgRNA with TNP and DOTAP, and preparing a nanocarrier for gene editing of aquatic animals through two ultrasonic processes.

本发明的上述第二个目的可以通过以下技术方案来实现:一种基于上述纳米载体的水产动物Cas9基因编辑方法,包括以下步骤:The above second object of the present invention can be achieved by the following technical scheme: A method for editing the Cas9 gene in aquatic animals based on the above nanocarrier comprises the following steps:

1) 制备含有Cas9 mRNA和sgRNA的TNP纳米粒子;1) Preparation of TNP nanoparticles containing Cas9 mRNA and sgRNA;

2) 将所述纳米粒子递送至目标水产动物的胚胎中;2) delivering the nanoparticles into embryos of target aquatic animals;

3) 在所述胚胎中进行目标基因的编辑。3) Editing the target gene in the embryo.

优选地,步骤1)中所述的Cas9 mRNA和sgRNA是通过物理吸附、化学结合或生物相容性包埋技术完成封装在所述TNP纳米粒子中。Preferably, the Cas9 mRNA and sgRNA described in step 1) are encapsulated in the TNP nanoparticles by physical adsorption, chemical binding or biocompatible encapsulation technology.

作为本发明的一种优选的实施方式,步骤1)中制备含有Cas9 mRNA和sgRNA的TNP纳米粒子包括以下步骤:将TNP纳米颗粒先溶于氯仿,再加入(2,3-二油酰基-丙基)-三甲胺DOTAP的氯仿溶液以及Cas9 mRNA和sgRNA的超纯水溶液,得混合材料溶液,将得混合材料溶液在冰浴中进行第一次超声,超声结束后加入超纯水混匀,然后在冰浴中进行第二次超声,再经浓缩处理,得含有Cas9 mRNA和sgRNA的TNP纳米粒子。As a preferred embodiment of the present invention, the preparation of TNP nanoparticles containing Cas9 mRNA and sgRNA in step 1) comprises the following steps: first dissolving the TNP nanoparticles in chloroform, then adding a chloroform solution of (2,3-dioleoyl-propyl)-trimethylamine DOTAP and an ultrapure aqueous solution of Cas9 mRNA and sgRNA to obtain a mixed material solution, subjecting the obtained mixed material solution to a first ultrasound in an ice bath, adding ultrapure water after the ultrasound to mix, then subjecting the solution to a second ultrasound in an ice bath, and then concentrating the solution to obtain TNP nanoparticles containing Cas9 mRNA and sgRNA.

优选地,第一次超声时,设定为超声5s,停2s,超声时间1min,功率为80 W。Preferably, during the first ultrasound treatment, the ultrasound is set to be on for 5 seconds, off for 2 seconds, the ultrasound time is 1 minute, and the power is 80 W.

优选地,第二次超声时,设定为超声10s,停2s,超声时间1min,功率为80 W。Preferably, during the second ultrasound treatment, the ultrasound is set to be on for 10 seconds, pause for 2 seconds, the ultrasound time is 1 minute, and the power is 80 W.

本发明的递送系统可以通过调整纳米载体的浓度,进一步提升基因编辑的精确性和操作的灵活性。The delivery system of the present invention can further improve the accuracy of gene editing and the flexibility of operation by adjusting the concentration of nanocarriers.

优选地,步骤2)中将所述纳米粒子递送至目标水产动物的胚胎中的方法为浸泡、注射或电穿孔。Preferably, in step 2), the method for delivering the nanoparticles into the embryo of the target aquatic animal is immersion, injection or electroporation.

优选地,步骤2)中所述的目标水产动物为斑马鱼或斑节对虾。Preferably, the target aquatic animal in step 2) is zebrafish or giant tiger prawn.

优选地,步骤3)中目标基因的编辑旨在提高水产动物的疾病抗性、生长速率或肉质。Preferably, the editing of the target gene in step 3) is intended to improve the disease resistance, growth rate or meat quality of aquatic animals.

优选地,还进一步包括步骤4)验证所述基因编辑的效果,验证所述基因编辑的效果包括使用PCR或测序。Preferably, the method further comprises step 4) verifying the effect of the gene editing, wherein verifying the effect of the gene editing comprises using PCR or sequencing.

因此,本发明为不同的水产动物种类选择特定的Cas9 mRNA和sgRNA,以及为每种特定的编辑目标(如抗病性、生长速度或色素沉着)定制化设计纳米载体。还提供了评估编辑效果的快速筛选方法,允许在初期阶段快速识别和优化成功的基因编辑事件。Therefore, the present invention selects specific Cas9 mRNA and sgRNA for different aquatic animal species, as well as customizes the design of nanocarriers for each specific editing target (such as disease resistance, growth rate or pigmentation). A rapid screening method for evaluating the editing effect is also provided, allowing for rapid identification and optimization of successful gene editing events at an early stage.

本发明具有以下优点:The present invention has the following advantages:

(1)提高基因编辑效率和精度:通过使用专门设计的纳米载体,本发明方法显著提高了CRISPR/Cas9系统的递送效率和基因编辑的靶向性。这种方法能够减少离散的基因编辑事件,从而提高基因编辑的整体成功率和精确度;(1) Improving gene editing efficiency and accuracy: By using specially designed nanocarriers, the method of the present invention significantly improves the delivery efficiency of the CRISPR/Cas9 system and the targeting of gene editing. This method can reduce discrete gene editing events, thereby improving the overall success rate and accuracy of gene editing;

(2)增强胚胎存活率:与传统的微注射技术相比,TNP纳米载体提供了一种更为温和的基因递送方式,显著降低了胚胎受损的风险,从而提高了胚胎的整体存活率;(2) Enhanced embryo survival rate: Compared with traditional microinjection technology, TNP nanocarriers provide a milder gene delivery method, significantly reducing the risk of embryo damage, thereby improving the overall survival rate of the embryo;

(3)操作的简便性和安全性:本发明简化了基因编辑的操作过程,减少了对高技能操作人员的依赖。同时,由于纳米载体的生物相容性和低免疫原性,减少了潜在的生物安全风险;(3) Simplicity and safety of operation: The present invention simplifies the operation process of gene editing and reduces the reliance on highly skilled operators. At the same time, due to the biocompatibility and low immunogenicity of the nanocarrier, potential biosafety risks are reduced;

(4)适用性广泛:该方法可以广泛应用于多种水产动物,包括那些传统微注射方法难以操作的小型或敏感种类;(4) Wide applicability: This method can be widely applied to a variety of aquatic animals, including small or sensitive species that are difficult to operate with traditional microinjection methods;

(5)支持定制化基因编辑:通过对纳米载体的特性进行调整,可以为特定的基因编辑目标和不同的水产动物种类量身定制,提供了一种高度灵活的基因编辑解决方案。(5) Support customized gene editing: By adjusting the properties of the nanocarrier, it can be tailored for specific gene editing targets and different aquatic animal species, providing a highly flexible gene editing solution.

附图说明BRIEF DESCRIPTION OF THE DRAWINGS

下面参照附图结合实施例对本发明作进一步的描述。The present invention will be further described below in conjunction with embodiments with reference to the accompanying drawings.

图1中A为实施例1的Mal-PEG-b-PLGA聚合物材料的合成路线图;B为实施例1的Mal-PEG-b-PLGA聚合物材料和Mal-PEG-OH原料的凝胶渗透色谱图;C为实施例1的Mal-PEG-b-PLGA聚合物材料的核磁共振氢谱图;In Figure 1, A is a synthetic route diagram of the Mal-PEG-b-PLGA polymer material of Example 1; B is a gel permeation chromatogram of the Mal-PEG-b-PLGA polymer material and the Mal-PEG-OH raw material of Example 1; C is a nuclear magnetic resonance hydrogen spectrum of the Mal-PEG-b-PLGA polymer material of Example 1;

图2中A为实施例1的双乳化法制备包载mRNA的TNP纳米颗粒过程示意图; B为实施例1的动态光散射检测TNP纳米颗粒的粒径大小图;C为实施例1的RiboGreen试剂盒检测TNP对mRNA的包封效率图;D为实施例1的TNP纳米颗粒在海水中24 h的粒径图;E为实施例1的分散度监测图;F为实施例1的琼脂糖凝胶检测不同质量比的TNP纳米载体对质粒系统的包载效率图;In Figure 2, A is a schematic diagram of the process of preparing TNP nanoparticles encapsulating mRNA by double emulsion method in Example 1; B is a particle size diagram of TNP nanoparticles detected by dynamic light scattering in Example 1; C is a diagram of the encapsulation efficiency of TNP for mRNA detected by RiboGreen kit in Example 1; D is a particle size diagram of TNP nanoparticles in seawater for 24 hours in Example 1; E is a dispersion monitoring diagram of Example 1; F is a diagram of the encapsulation efficiency of TNP nanocarriers with different mass ratios for plasmid systems detected by agarose gel in Example 1;

图3是实施例2中斑马鱼鱼卵与TNP纳米载体封装的罗丹红颗粒共同孵育,孵育8h、24h、48h后观察斑马鱼卵红色荧光,测试TNP纳米颗粒与斑马鱼鱼卵卵膜融合效果;3 is a diagram of zebrafish eggs in Example 2 incubated with rhodamine red particles encapsulated by TNP nanocarriers, and red fluorescence of zebrafish eggs was observed after incubation for 8h, 24h, and 48h to test the fusion effect of TNP nanoparticles with zebrafish egg membranes;

图4是实施例2中斑马鱼鱼卵与TNP纳米载体封装的EGFP-mRNA共同孵育,孵育8h、48h后观察斑马鱼卵绿色荧光,测试TNP纳米颗粒递送EGFP-mRNA在斑马鱼鱼卵中表达效果;Figure 4 shows the co-incubation of zebrafish eggs and EGFP-mRNA encapsulated by TNP nanocarriers in Example 2, and the green fluorescence of zebrafish eggs was observed after 8h and 48h of incubation to test the expression effect of TNP nanoparticles delivering EGFP-mRNA in zebrafish eggs;

图5是实施例3中斑马鱼鱼卵与TNP纳米载体封装的Cas9mRNA、Tyr-sgRNA共同孵育,孵育72小时后提取基因组DNA进行NGS二代测序,测序突变结果展示。Figure 5 shows the zebrafish eggs in Example 3 incubated with Cas9mRNA and Tyr-sgRNA encapsulated by TNP nanocarriers, and the genomic DNA was extracted after 72 hours of incubation for NGS second-generation sequencing, and the sequencing mutation results are displayed.

具体实施方式DETAILED DESCRIPTION

下面结合具体实施例进一步说明本发明。下述实施例和附图仅用于示例性说明,不能理解为对本发明的限制。除非特别说明,下述实施例中使用的试剂原料为常规市购或商业途径获得的生试剂原料,除非特别说明,下述实施例中使用的方法和设备为本领域常规使用的方法和设备。The present invention is further described below in conjunction with specific examples. The following examples and drawings are only used for illustrative purposes and are not to be construed as limiting the present invention. Unless otherwise specified, the reagent raw materials used in the following examples are conventional commercially available or commercially available raw reagent raw materials, and unless otherwise specified, the methods and equipment used in the following examples are conventional methods and equipment used in the art.

以下实施例是对本发明的进一步说明,而不是对本发明的限制。The following examples are provided to further illustrate the present invention, rather than to limit the present invention.

实施例1Example 1

以下结合具体实施例,对本发明做进一步说明。应理解,以下实施例仅用于说明本发明而非用于限制本发明的范围。The present invention is further described below in conjunction with specific examples. It should be understood that the following examples are only used to illustrate the present invention and are not used to limit the scope of the present invention.

实施例1 制备纳米载体及包埋Example 1 Preparation of Nanocarriers and Embedding

1. 聚合物材料的合成1. Synthesis of polymer materials

首先,合成马来酰亚胺化聚乙二醇-聚(丙交酯-co-乙交酯)(Mal-PEG5k-b-PLGA10k)。First, maleimidized polyethylene glycol-poly(lactide-co-glycolide) (Mal-PEG 5k - b -PLGA 10k ) was synthesized.

具体合成步骤如下:将经甲苯共沸除水的马来酰亚胺化聚乙二醇(Mal-PEG5k(5k指PEG的分子量)-OH)和冻抽过夜除水的丙交酯(D, L-LA)和乙交酯(GA)转移至手套箱(H2O<0.1 ppm, O2<0.1ppm)中。称取Mal-PEG5k-OH(1 g, 0.2 mmol)和D, L-LA(1.6 g, 11.111mmol)以及GA(0.6 g, 5.173 mmol)加入预先烘烤除水汽的50 mL圆底烧瓶中,加入磁子并且用油浴锅加热至130 ℃,持续搅拌直至烧瓶中固体全部溶解。随后,在搅拌条件下滴加2滴(20 mg, 0.049 mmol)异辛酸亚锡(Sn(Oct)2)后继续搅拌反应3 h。反应完毕浓缩产物沉淀在甲醇/乙醚混合液(1/10, v/v)中,抽滤干燥得淡黄色Mal-PEG5k-b-PLGA10k(10k指Mal-PEG5k-b-PLGA的分子量),收集产物进行称重并通过1 H NMR测定化学结构。The specific synthesis steps are as follows: Maleimidized polyethylene glycol (Mal-PEG 5k (5k refers to the molecular weight of PEG)-OH) dehydrated by toluene azeotropy and lactide (D, L-LA) and glycolide (GA) dehydrated by freezing and pumping overnight were transferred to a glove box (H 2 O <0.1 ppm, O 2 <0.1 ppm). Mal-PEG 5k -OH (1 g, 0.2 mmol) and D, L-LA (1.6 g, 11.111 mmol) and GA (0.6 g, 5.173 mmol) were weighed and added to a 50 mL round-bottom flask that had been pre-baked to remove moisture. A magnetic bar was added and heated to 130 °C in an oil bath. Stirring was continued until all the solids in the flask were dissolved. Subsequently, 2 drops (20 mg, 0.049 mmol) of stannous isooctanoate (Sn(Oct) 2 ) were added under stirring and the reaction was continued for 3 h. After the reaction, the concentrated product was precipitated in a methanol/ether mixture (1/10, v/v), filtered and dried to obtain light yellow Mal-PEG 5k - b -PLGA 10k (10k refers to the molecular weight of Mal-PEG 5k - b -PLGA). The product was collected and weighed, and its chemical structure was determined by 1 H NMR.

进一步,称取150 mg Mal-PEG5k-b-PLGA10k加入250 mL圆底烧瓶中,加入搅拌磁子,加入10 mL二甲基亚砜(Dimethyl sulfoxide, DMSO)溶解上述聚合物材料,并在搅拌条件下缓慢加入100 mL超纯水连续搅拌2 h,分别制备得Mal-NP纳米颗粒。Furthermore, 150 mg Mal-PEG 5k - b -PLGA 10k was weighed and added into a 250 mL round-bottom flask, a stirring magnet was added, 10 mL dimethyl sulfoxide (DMSO) was added to dissolve the above polymer materials, and 100 mL ultrapure water was slowly added under stirring conditions and stirred continuously for 2 h to prepare Mal-NP nanoparticles.

将Mal-NP在超纯水中透析过夜除去DMSO,收集透析袋内的纳米颗粒溶液转移至圆底烧瓶中,称取TAT(穿膜肽,20 mg, 0.016 mmol)加入圆底烧瓶中在氮气保护下搅拌反应12 h,通过TAT末端的半胱氨酸残基中的巯基与马来酰亚胺之间的点击化学反应在Mal-NP表面修饰上TAT。反应结束后,收集颗粒溶液在30000 g离心力下离心颗粒2 h,收集下层颗粒沉淀,冻干后得到TAT修饰的TAT-PEG-b-PLGA材料(简称TNP)。Mal-NP was dialyzed in ultrapure water overnight to remove DMSO, the nanoparticle solution in the dialysis bag was collected and transferred to a round-bottom flask, TAT (membrane-penetrating peptide, 20 mg, 0.016 mmol) was weighed and added to the round-bottom flask, stirred and reacted for 12 h under nitrogen protection, and TAT was modified on the surface of Mal-NP by a click chemistry reaction between the thiol group in the cysteine residue at the end of TAT and maleimide. After the reaction, the particle solution was collected and centrifuged at 30,000 g for 2 h, the lower layer of particle precipitation was collected, and the TAT-PEG- b -PLGA material (abbreviated as TNP) modified with TAT was obtained after freeze-drying.

收集高速离心后的上层清液,利用Ellman’s试剂盒(pH 8.0, 70 μg/mL, 100 mMPB buffer含1 mM乙二胺四乙酸(EDTA,Ethylene diamine tetraacetic acid,)中的DTNB与上层清液中未反应的游离TAT末端巯基反应15 min,再检测反应溶液在412 nm处的吸收值计算TAT与Mal基团的键合效率。The supernatant was collected after high-speed centrifugation, and the DTNB in the Ellman’s kit (pH 8.0, 70 μg/mL, 100 mMPB buffer containing 1 mM ethylenediaminetetraacetic acid (EDTA)) was used to react with the unreacted free TAT terminal thiol in the supernatant for 15 min. The absorption value of the reaction solution at 412 nm was then detected to calculate the bonding efficiency of TAT and Mal groups.

本实施例中Mal-PEG-b-PLGA(TNP)聚合物材料的合成路线如图1中的A所示, Mal-PEG-b-PLGA聚合物材料和Mal-PEG-OH原料的凝胶渗透色谱图如图1中的B所示,Mal-PEG-b-PLGA的出峰时间早于原料Mal-PEG-OH,表明成功合成了Mal-PEG-b-PLGA嵌段聚合物材料;进一步,根据Mal-PEG-b-PLGA聚合物材料的核磁共振氢谱图如图1中的C所示,合成的Mal-PEG-b-PLGA聚合物中LA的聚合度为111,而GA的聚合度为40,得到最终PLGA段分子量为10000 道尔顿。The synthesis route of the Mal-PEG-b-PLGA (TNP) polymer material in this embodiment is shown in A of Figure 1, and the gel permeation chromatogram of the Mal-PEG-b-PLGA polymer material and the Mal-PEG-OH raw material is shown in B of Figure 1. The peak time of Mal-PEG- b -PLGA is earlier than that of the raw material Mal-PEG-OH, indicating that the Mal-PEG- b -PLGA block polymer material has been successfully synthesized; further, according to the nuclear magnetic resonance hydrogen spectrum of the Mal-PEG- b -PLGA polymer material as shown in C of Figure 1, the degree of polymerization of LA in the synthesized Mal-PEG- b -PLGA polymer is 111, while the degree of polymerization of GA is 40, and the molecular weight of the final PLGA segment is 10,000 Daltons.

2. 制备罗丹明B(RhoB)标记的TNP纳米颗粒:2. Preparation of RhoB-labeled TNP nanoparticles:

称取25 mg TAT-PEG-b-PLGA(TNP)和1 mg罗丹明B标记的PLGA(PLGA指聚(丙交酯-co-乙交酯)均聚物,下同)(RhoB-PLGA)共溶于400 μL氯仿中,向其中加入(2,3-二油酰基-丙基)-三甲胺(DOTAP, 2 mg, 100 μL, 氯仿);取一50 mL离心管加入DEPC水(400 μL,超纯水);随后将上述混合材料溶液加至离心管底部。将离心管放在冰浴中进行第一次超声,超声探头没入液面。设定为超声5s,停2s,超声时间1min,功率为80 W;超声结束后加入5mLDEPC水,混匀;随后将离心管放在冰浴中进行第二次超声,设定为超声10s,停2s,超声时间1min,功率为80 W;超声结束后,将液体转移到圆底烧瓶中,接到旋蒸仪上,关闭进气口,打开抽气泵开低速旋转。在气泡抽完溶液变清之后将圆底烧瓶沉到水浴中,将颗粒溶液浓缩至1-2 mL。Weigh 25 mg of TAT-PEG- b -PLGA (TNP) and 1 mg of rhodamine B-labeled PLGA (PLGA refers to poly (lactide-co-glycolide) homopolymer, the same below) (RhoB-PLGA) and dissolve them in 400 μL of chloroform, add (2,3-dioleoyl-propyl)-trimethylamine (DOTAP, 2 mg, 100 μL, chloroform); take a 50 mL centrifuge tube and add DEPC water (400 μL, ultrapure water); then add the above mixed material solution to the bottom of the centrifuge tube. Place the centrifuge tube in an ice bath for the first ultrasound, and the ultrasound probe is submerged in the liquid surface. The setting was 5s ultrasonic, 2s off, 1min ultrasonic time, and 80W power. After the ultrasonic was finished, 5mL DEPC water was added and mixed. Then the centrifuge tube was placed in an ice bath for a second ultrasonic treatment, which was set to 10s ultrasonic, 2s off, 1min ultrasonic time, and 80W power. After the ultrasonic was finished, the liquid was transferred to a round-bottom flask, connected to a rotary evaporator, the air inlet was closed, and the vacuum pump was turned on at a low speed. After the bubbles were exhausted and the solution became clear, the round-bottom flask was sunk into a water bath to concentrate the particle solution to 1-2 mL.

3. 包载基因物质的TNP纳米载体制备:3. Preparation of TNP nanocarriers carrying genetic material:

称取25 mg TAT-PEG-b-PLGA(TNP)和罗丹明B标记的PLGA溶于400 μL氯仿中,向其中加入(2,3-二油酰基-丙基)-三甲胺(DOTAP, 2 mg, 100 μL, 氯仿);取一50 mL离心管加入DEPC水(400 µL),随后加入EGFP-mRNA (50 µg)(该基因来源于源自于水母Aequoreavictoria,它是一种经过优化的绿色荧光蛋白。EGFP常用于研究基因的表达效率和细胞功能,因为它在生物体内能够发出明显的绿色荧光,且无需添加外源性物质即可在活细胞中直接观察。这使得EGFP成为细胞生物学和基因工程中最常用的报告基因之一。);随后将上述混合材料溶液加至离心管底部。将离心管放在冰浴中进行第一次超声,超声探头没入液面。设定为超声5s,停2s,超声时间1min,功率为80 W;超声结束后加入5ml DEPC水,混匀;随后将离心管放在冰浴中进行第二次超声,设定为超声10s,停2s,超声时间1min,功率为80W;超声结束后,将液体转移到圆底烧瓶中,接到旋蒸仪上,关闭进气口,打开抽气泵开低速旋转。在气泡抽完溶液变清之后将圆底烧瓶沉到水浴中,将颗粒溶液浓缩至1-2 mL;最后,用RiboGreen试剂盒检测mRNA的包载效率,或者用琼脂糖凝胶检测质粒或siRNA的包载效率。进一步,将包载基因物质的TNP纳米颗粒溶液转移到EP管中置于4℃进行保存待用。Weigh 25 mg of TAT-PEG- b -PLGA (TNP) and rhodamine B-labeled PLGA and dissolve them in 400 μL of chloroform, add (2,3-dioleoyl-propyl)-trimethylamine (DOTAP, 2 mg, 100 μL, chloroform); take a 50 mL centrifuge tube and add DEPC water (400 μL), then add EGFP-mRNA (50 μg) (this gene comes from the jellyfish Aequoreavictoria, which is an optimized green fluorescent protein. EGFP is often used to study gene expression efficiency and cell function because it can emit obvious green fluorescence in vivo and can be directly observed in living cells without adding exogenous substances. This makes EGFP one of the most commonly used reporter genes in cell biology and genetic engineering.); then add the above mixed material solution to the bottom of the centrifuge tube. Place the centrifuge tube in an ice bath for the first ultrasound, and the ultrasound probe is submerged in the liquid surface. The setting is 5s ultrasonic, 2s stop, 1min ultrasonic time, and 80W power; after the ultrasonic, add 5ml DEPC water and mix; then put the centrifuge tube in an ice bath for a second ultrasonic, set to 10s ultrasonic, 2s stop, 1min ultrasonic time, and 80W power; after the ultrasonic, transfer the liquid to a round-bottom flask, connect it to a rotary evaporator, close the air inlet, turn on the vacuum pump and rotate at a low speed. After the bubbles are exhausted and the solution becomes clear, sink the round-bottom flask into a water bath and concentrate the particle solution to 1-2 mL; finally, use the RiboGreen kit to detect the mRNA encapsulation efficiency, or use agarose gel to detect the plasmid or siRNA encapsulation efficiency. Further, transfer the TNP nanoparticle solution containing genetic material to an EP tube and store it at 4°C for later use.

本实施例中双乳化法制备TNP纳米颗粒包载基因物质示意图如图2中的A所示,TNP纳米颗粒的粒径大小如图2中的B所示,RiboGreen试剂盒检测TNP对mRNA的包封效率如图2中的C所示,TNP纳米颗粒在海水中24 h粒径和分散度监测如图2中的D和E所示;琼脂糖凝胶检测不同质量比的TNP纳米载体对质粒系统的包载效率如图2中的F所示。The schematic diagram of preparing TNP nanoparticles encapsulating genetic material by the double emulsion method in this embodiment is shown in A of Figure 2, the particle size of the TNP nanoparticles is shown in B of Figure 2, the encapsulation efficiency of TNP for mRNA detected by the RiboGreen kit is shown in C of Figure 2, and the particle size and dispersion monitoring of TNP nanoparticles in seawater for 24 h are shown in D and E of Figure 2; the encapsulation efficiency of TNP nanocarriers with different mass ratios for the plasmid system detected by agarose gel is shown in F of Figure 2.

图2中的结果表明,TNP纳米载体能够有效包载mRNA或者质粒等基因物质,并且在海水中具有良好的颗粒稳定性,具有用于水产物种胚胎细胞的基因编辑应用潜力。The results in Figure 2 indicate that TNP nanocarriers can effectively encapsulate genetic materials such as mRNA or plasmids, and have good particle stability in seawater, and have the potential for application in gene editing of embryonic cells of aquatic species.

实施例2递送表达验证与基因编辑详细说明,Example 2 Delivery expression verification and gene editing detailed description,

1.斑马鱼卵膜融合的验证过程1. Verification process of zebrafish egg membrane fusion

为验证实施例1中TNP纳米材料与斑马鱼胚胎卵膜的融合效果,可采取以下步骤:To verify the fusion effect of the TNP nanomaterial and the zebrafish embryo egg membrane in Example 1, the following steps can be taken:

纳米颗粒的递送:将实施例1中用RhoB-PLGA标记的TNP纳米颗粒悬浮在适宜的介质中,然后将斑马鱼的胚胎放入含纳米颗粒的溶液中进行浸泡。Delivery of nanoparticles: The TNP nanoparticles labeled with RhoB-PLGA in Example 1 were suspended in a suitable medium, and then zebrafish embryos were immersed in the solution containing the nanoparticles.

荧光显微观察:在浸泡后8h、24h、48h后,使用荧光显微镜观察胚胎。通过罗丹明颗粒的荧光信号,检查纳米颗粒是否已与卵膜融合并附着在胚胎表面。Fluorescence microscopic observation: After 8h, 24h, and 48h of immersion, the embryos were observed using a fluorescence microscope. The fluorescence signal of the rhodamine particles was used to check whether the nanoparticles had fused with the egg membrane and attached to the embryo surface.

数据分析:通过比较处理组和对照组胚胎的荧光强度,评估卵膜融合的效率。高荧光强度表明良好的膜融合效果。Data analysis: The efficiency of egg membrane fusion was evaluated by comparing the fluorescence intensity of embryos in the treated and control groups. High fluorescence intensity indicates good membrane fusion effect.

斑马鱼鱼卵与TNP纳米载体封装的罗丹红颗粒共同孵育结果如图3所示,孵育8h、24h、48h后观察斑马鱼卵红色荧光,测试TNP纳米颗粒与斑马鱼鱼卵卵膜融合效果。The results of co-incubation of zebrafish eggs with Rhodamine red particles encapsulated by TNP nanocarriers are shown in FIG3 . The red fluorescence of zebrafish eggs was observed after incubation for 8 h, 24 h, and 48 h to test the fusion effect of TNP nanoparticles with the zebrafish egg membrane.

图3中的实验结果显示,封装罗丹明颗粒的纳米颗粒能够与斑马鱼鱼卵膜实现有效融合,且这种融合与纳米颗粒的浓度呈正相关。随着纳米颗粒浓度的逐步增加,斑马鱼卵膜的红色荧光强度显著增强,表明TNP纳米颗粒浓度越高,其与斑马鱼鱼卵卵膜的融合效果越好。The experimental results in Figure 3 show that the nanoparticles encapsulating rhodamine particles can effectively fuse with the zebrafish egg membrane, and this fusion is positively correlated with the concentration of the nanoparticles. As the concentration of the nanoparticles gradually increases, the red fluorescence intensity of the zebrafish egg membrane is significantly enhanced, indicating that the higher the concentration of TNP nanoparticles, the better the fusion effect with the zebrafish egg membrane.

2. mRNA递送的验证过程2. Validation process of mRNA delivery

为验证TNP纳米材料递送EGFP-mRNA到斑马鱼和斑节对虾胚胎的效果,可采取以下步骤:To verify the effectiveness of TNP nanomaterials in delivering EGFP-mRNA to zebrafish and giant shrimp embryos, the following steps can be taken:

递送与孵化:将封装有EGFP-mRNA的纳米颗粒悬浮在无酶水中,悬浮稀释浓度为包埋mRNA 0.5μg/mL和1μg/mL,然后将胚胎放入溶液中进行浸泡孵化。Delivery and incubation: Nanoparticles encapsulated with EGFP-mRNA were suspended in enzyme-free water at a suspension dilution concentration of 0.5 μg/mL and 1 μg/mL of embedded mRNA, and then the embryos were placed in the solution for immersion incubation.

荧光显微观察:在孵化后8小时,使用荧光显微镜观察斑马鱼胚胎中的绿色荧光蛋白表达。荧光的存在和强度可以反映mRNA递送的效率和表达水平。Fluorescence microscopy: 8 hours after hatching, green fluorescent protein expression in zebrafish embryos was observed using a fluorescence microscope. The presence and intensity of fluorescence can reflect the efficiency of mRNA delivery and expression level.

表达分析:通过图像分析软件量化荧光强度,评估EGFP-mRNA的递送效果和表达效率。同时,可进行RT-PCR或Western blot等生化实验验证mRNA表达的蛋白。Expression analysis: The fluorescence intensity is quantified by image analysis software to evaluate the delivery effect and expression efficiency of EGFP-mRNA. At the same time, biochemical experiments such as RT-PCR or Western blot can be performed to verify the protein expressed by mRNA.

斑马鱼鱼卵与TNP纳米载体封装的EGFP-mRNA共同孵育荧光显微观察如图4所示,孵育8h、48h后观察斑马鱼卵绿色荧光,测试TNP纳米颗粒递送EGFP-mRNA在斑马鱼鱼卵中表达效果。Fluorescence microscopic observation of zebrafish eggs co-incubated with EGFP-mRNA encapsulated by TNP nanocarriers is shown in Figure 4. Green fluorescence of zebrafish eggs was observed after incubation for 8 hours and 48 hours to test the expression effect of TNP nanoparticles delivering EGFP-mRNA in zebrafish eggs.

如图4所示,实验结果表明,转染了TNP纳米载体封装的EGFP-mRNA的斑马鱼鱼卵在孵育8小时和48小时后均出现了绿色荧光信号,而对照组则未观察到荧光表达。这表明EGFP-mRNA成功在斑马鱼鱼卵中得以表达。同时,随着封装EGFP-mRNA的纳米载体浓度的增加,斑马鱼鱼卵的绿色荧光强度也逐步增强,实验结果证明,TNP纳米载体能够有效转染EGFP-mRNA并在斑马鱼鱼卵中成功表达,且其表达效果与纳米载体的浓度密切相关。As shown in Figure 4, the experimental results show that zebrafish eggs transfected with EGFP-mRNA encapsulated by TNP nanocarriers showed green fluorescence signals after incubation for 8 hours and 48 hours, while no fluorescence expression was observed in the control group. This shows that EGFP-mRNA was successfully expressed in zebrafish eggs. At the same time, as the concentration of nanocarriers encapsulating EGFP-mRNA increased, the green fluorescence intensity of zebrafish eggs also gradually increased. The experimental results prove that TNP nanocarriers can effectively transfect EGFP-mRNA and successfully express it in zebrafish eggs, and its expression effect is closely related to the concentration of nanocarriers.

实施例3Example 3

1.基因编辑斑马鱼胚胎中递送验证1. Delivery verification of gene editing in zebrafish embryos

1.1斑马鱼鱼卵准备1.1 Zebrafish egg preparation

在实施本发明方法之前,首先准备斑马鱼成鱼,从斑马鱼国家资源中心获取,使用健康、成熟的样本,这些斑马鱼大约生长120天。为了增强交配活力,雄性和雌性斑马鱼在产卵前一天被分开饲养。Before implementing the method of the present invention, adult zebrafish were first prepared and obtained from the National Zebrafish Resource Center, using healthy, mature samples, which were about 120 days old. In order to enhance mating activity, male and female zebrafish were separated and raised one day before spawning.

孵化使用专门的水族箱进行,水温维持在28-30°C之间。照明周期设定为每天14小时光照和10小时黑暗,模拟自然环境中的日夜变化,以促进斑马鱼的正常生理节律和繁殖行为。Hatching was carried out in a dedicated aquarium with water temperature maintained between 28-30°C. The lighting cycle was set to 14 hours of light and 10 hours of darkness per day, simulating the day-night changes in the natural environment to promote the normal physiological rhythm and reproductive behavior of zebrafish.

通常在开灯后30分钟后,通过移除隔板让斑马鱼交配。交配后40分钟内收集受精卵,并立即用蒸馏水冲洗三次,以清除鱼卵表面污染物。Usually, zebrafish were mated by removing the baffle 30 minutes after lights were turned on. Fertilized eggs were collected within 40 minutes after mating and immediately rinsed three times with distilled water to remove surface contaminants.

1.2Tyr基因靶点设计1.2 Tyr gene target design

为实现对斑马鱼Tyr基因的有效编辑,需要精确设计CRISPR/Cas9系统的靶向单向导RNA(sgRNA)。以下是Tyr基因CRISPR/Cas9靶点设计的步骤:To achieve effective editing of the zebrafish Tyr gene, it is necessary to accurately design the targeted single guide RNA (sgRNA) of the CRISPR/Cas9 system. The following are the steps for designing the CRISPR/Cas9 target site of the Tyr gene:

1.2.1基因序列获取:1.2.1 Gene sequence acquisition:

首先,需要获得斑马鱼Tyr基因的完整序列。可以通过公共数据库NCBI获取。First, we need to obtain the complete sequence of the zebrafish Tyr gene, which can be obtained through the public database NCBI.

斑马鱼tyr基因(酪氨酸酶基因,tyrosinase gene),tyr基因编码的酪氨酸酶是一种关键酶,参与黑色素的合成过程,其序列及来源序列信息如下:NCBI ReferenceSequence: NM_131013.3。Zebrafish tyr gene (tyrosinase gene), the tyrosinase encoded by the tyr gene is a key enzyme involved in the synthesis of melanin. Its sequence and source sequence information are as follows: NCBI Reference Sequence: NM_131013.3.

(a)序列特征,(a) Sequence characteristics,

*长度:1955碱基对;* Length: 1955 base pairs;

*类型:核苷酸;*Type: Nucleotide;

*链型:双链;*Chain type: double chain;

*拓扑结构:线性;*Topology: Linear;

(b)分子类型:核酸;(b) Molecule type: nucleic acid;

序列描述:NM_131013.3。Sequence description: NM_131013.3.

2.2 靶点识别和选择:2.2 Target identification and selection:

2.2.1靶点位置:在Tyr基因序列中,选择具有典型的PAM序列(NGG)的区域作为潜在的CRISPR靶点。通常,选择位于功能域或关键编码区附近的靶点,以增加编辑后的表型变化可能性。2.2.1 Target location: In the Tyr gene sequence, select regions with typical PAM sequences (NGG) as potential CRISPR targets. Usually, targets located near functional domains or key coding regions are selected to increase the possibility of phenotypic changes after editing.

2.2.2特异性分析:使用在线工具Chopchop进行靶点的特异性分析。这些工具能评估所选靶点在基因组中的唯一性,减少非目标编辑的风险。2.2.2 Specificity analysis: Use the online tool Chopchop to perform target specificity analysis. These tools can evaluate the uniqueness of the selected target in the genome and reduce the risk of non-target editing.

2.2.3 sgRNA设计:2.2.3 sgRNA design:

序列设计:根据所选靶点序列设计sgRNA。sgRNA通常包括20个核苷酸的靶向序列,紧邻PAM序列。Sequence design: sgRNA is designed according to the selected target sequence. sgRNA usually includes a 20-nucleotide targeting sequence, immediately adjacent to the PAM sequence.

优化:对sgRNA序列进行优化,可能包括提高GC含量以增强稳定性和结合效率,或在序列中引入修饰以提高抗核酸酶降解的能力。Optimization: Optimization of the sgRNA sequence may include increasing the GC content to enhance stability and binding efficiency, or introducing modifications into the sequence to improve resistance to nuclease degradation.

设计序列为:ggactggaggacttctgggg(如SEQ NO:1所示)。The designed sequence is: ggactggaggacttctgggg (as shown in SEQ NO: 1).

2.2.4sgRNA合成:根据优化后的设计,合成sgRNA,在合成mRNA时需进行化学修饰的引入,N1-甲基伪尿苷的引入具体包括:2.2.4sgRNA synthesis: According to the optimized design, sgRNA is synthesized. Chemical modification needs to be introduced when synthesizing mRNA. The introduction of N1-methyl pseudouridine specifically includes:

在合成mRNA的过程中,选择特定的尿嘧啶位点进行化学修饰,将尿苷(U)替换为N1-甲基伪尿苷(m1Ψ)。这种修饰有助于减少mRNA被人体内源性免疫系统识别的机会, m1Ψ修改减少了mRNA激活模式识别受体如RIG-I的能力。During the synthesis of mRNA, specific uracil sites are selected for chemical modification, replacing uridine (U) with N1-methyl pseudouridine (m1Ψ). This modification helps reduce the chance of mRNA being recognized by the human endogenous immune system. The m1Ψ modification reduces the ability of mRNA to activate pattern recognition receptors such as RIG-I.

使用转录酶在体外转录过程中引入m1Ψ,确保修饰的均匀性和高效率。The m1Ψ was introduced during in vitro transcription using a transcriptase, ensuring uniformity and high efficiency of modification.

合成序列:Synthetic sequence:

mG*mG*mA*rCrUrGrGrArGrGrArCrUrUrCrUrGrGrGrGrGrUrUrUrUrArGrArGrCrUrArGrArArArUrArGrCrArArGrUrUrArArArArUrArArGrGrCrUrArGrUrCrCrGrUrUrArUrCrArArCrUrUrGrArArArArArGrUrGrGrCrArCrCrGrArGrUrCrGrGrUrGrCrU*mU*mU*mU(如SEQ NO:2所示)。mG*mG*mA*rCrUrGrGrArGrGrArCrUrUrCrUrGrGrGrGrGrUrUrUrUrArGrArGrCrUrArGrArArArUrArGrCrArArGrUrUrArArArArUrA rArGrGrCrUrArGrUrCrCrGrUrUrArUrCrArArCrUrUrGrArArArArGrUrGrGrCrArCrCrGrArGrUrCrGrGrUrGrCrU*mU*mU*mU (as SEQ NO:2 shown).

2.2.5合成NLS-Cas9-NLS(核定位)mRNA:2.2.5 Synthesis of NLS-Cas9-NLS (nuclear localization) mRNA:

NLS序列为公开已知序列。The NLS sequence is a publicly known sequence.

(a)序列特征,(a) Sequence characteristics,

*长度:4272碱基对;* Length: 4272 base pairs;

*类型:核苷酸;*Type: Nucleotide;

*链型:单链;*Chain type: single chain;

*拓扑结构:线性;AUGGACUAUAAGGACCACGACGGAGACUACAAGGAUCAUGAUAUUGAUUACAAAGACGAUGACGAUAAGAUGGCCCCAAAGAAGAAGCGGAAGGUCGGUAUCCACGGAGUCCCAGCAGCCGACAAGAAGUACAGCAUCGGCCUGGACAUCGGCACCAACUCUGUGGGCUGGGCCGUGAUCACCGACGAGUACAAGGUGCCCAGCAAGAAAUUCAAGGUGCUGGGCAACACCGACCGGCACAGCAUCAAGAAGAACCUGAUCGGAGCCCUGCUGUUCGACAGCGGCGAAACAGCCGAGGCCACCCGGCUGAAGAGAACCGCCAGAAGAAGAUACACCAGACGGAAGAACCGGAUCUGCUAUCUGCAAGAGAUCUUCAGCAACGAGAUGGCCAAGGUGGACGACAGCUUCUUCCACAGACUGGAAGAGUCCUUCCUGGUGGAAGAGGAUAAGAAGCACGAGCGGCACCCCAUCUUCGGCAACAUCGUGGACGAGGUGGCCUACCACGAGAAGUACCCCACCAUCUACCACCUGAGAAAGAAACUGGUGGACAGCACCGACAAGGCCGACCUGCGGCUGAUCUAUCUGGCCCUGGCCCACAUGAUCAAGUUCCGGGGCCACUUCCUGAUCGAGGGCGACCUGAACCCCGACAACAGCGACGUGGACAAGCUGUUCAUCCAGCUGGUGCAGACCUACAACCAGCUGUUCGAGGAAAACCCCAUCAACGCCAGCGGCGUGGACGCCAAGGCCAUCCUGUCUGCCAGACUGAGCAAGAGCAGACGGCUGGAAAAUCUGAUCGCCCAGCUGCCCGGCGAGAAGAAGAAUGGCCUGUUCGGAAACCUGAUUGCCCUGAGCCUGGGCCUGACCCCCAACUUCAAGAGCAACUUCGACCUGGCCGAGGAUGCCAAACUGCAGCUGAGCAAGGACACCUACGACGACGACCUGGACAACCUGCUGGCCCAGAUCGGCGACCAGUACGCCGACCUGUUUCUGGCCGCCAAGAACCUGUCCGACGCCAUCCUGCUGAGCGACAUCCUGAGAGUGAACACCGAGAUCACCAAGGCCCCCCUGAGCGCCUCUAUGAUCAAGAGAUACGACGAGCACCACCAGGACCUGACCCUGCUGAAAGCUCUCGUGCGGCAGCAGCUGCCUGAGAAGUACAAAGAGAUUUUCUUCGACCAGAGCAAGAACGGCUACGCCGGCUACAUUGACGGCGGAGCCAGCCAGGAAGAGUUCUACAAGUUCAUCAAGCCCAUCCUGGAAAAGAUGGACGGCACCGAGGAACUGCUCGUGAAGCUGAACAGAGAGGACCUGCUGCGGAAGCAGCGGACCUUCGACAACGGCAGCAUCCCCCACCAGAUCCACCUGGGAGAGCUGCACGCCAUUCUGCGGCGGCAGGAAGAUUUUUACCCAUUCCUGAAGGACAACCGGGAAAAGAUCGAGAAGAUCCUGACCUUCCGCAUCCCCUACUACGUGGGCCCUCUGGCCAGGGGAAACAGCAGAUUCGCCUGGAUGACCAGAAAGAGCGAGGAAACCAUCACCCCCUGGAACUUCGAGGAAGUGGUGGACAAGGGCGCUUCCGCCCAGAGCUUCAUCGAGCGGAUGACCAACUUCGAUAAGAACCUGCCCAACGAGAAGGUGCUGCCCAAGCACAGCCUGCUGUACGAGUACUUCACCGUGUAUAACGAGCUGACCAAAGUGAAAUACGUGACCGAGGGAAUGAGAAAGCCCGCCUUCCUGAGCGGCGAGCAGAAAAAGGCCAUCGUGGACCUGCUGUUCAAGACCAACCGGAAAGUGACCGUGAAGCAGCUGAAAGAGGACUACUUCAAGAAAAUCGAGUGCUUCGACUCCGUGGAAAUCUCCGGCGUGGAAGAUCGGUUCAACGCCUCCCUGGGCACAUACCACGAUCUGCUGAAAAUUAUCAAGGACAAGGACUUCCUGGACAAUGAGGAAAACGAGGACAUUCUGGAAGAUAUCGUGCUGACCCUGACACUGUUUGAGGACAGAGAGAUGAUCGAGGAACGGCUGAAAACCUAUGCCCACCUGUUCGACGACAAAGUGAUGAAGCAGCUGAAGCGGCGGAGAUACACCGGCUGGGGCAGGCUGAGCCGGAAGCUGAUCAACGGCAUCCGGGACAAGCAGUCCGGCAAGACAAUCCUGGAUUUCCUGAAGUCCGACGGCUUCGCCAACAGAAACUUCAUGCAGCUGAUCCACGACGACAGCCUGACCUUUAAAGAGGACAUCCAGAAAGCCCAGGUGUCCGGCCAGGGCGAUAGCCUGCACGAGCACAUUGCCAAUCUGGCCGGCAGCCCCGCCAUUAAGAAGGGCAUCCUGCAGACAGUGAAGGUGGUGGACGAGCUCGUGAAAGUGAUGGGCCGGCACAAGCCCGAGAACAUCGUGAUCGAAAUGGCCAGAGAGAACCAGACCACCCAGAAGGGACAGAAGAACAGCCGCGAGAGAAUGAAGCGGAUCGAAGAGGGCAUCAAAGAGCUGGGCAGCCAGAUCCUGAAAGAACACCCCGUGGAAAACACCCAGCUGCAGAACGAGAAGCUGUACCUGUACUACCUGCAGAAUGGGCGGGAUAUGUACGUGGACCAGGAACUGGACAUCAACCGGCUGUCCGACUACGAUGUGGACCAUAUCGUGCCUCAGAGCUUUCUGGCCGACGACUCCAUCGACAACAAGGUGCUGACCAGAAGCGACAAGAACCGGGGCAAGAGCGACAACGUGCCCUCCGAAGAGGUCGUGAAGAAGAUGAAGAACUACUGGCGGCAGCUGCUGAACGCCAAGCUGAUUACCCAGAGAAAGUUCGACAAUCUGACCAAGGCCGAGAGAGGCGGCCUGAGCGAACUGGAUAAGGCCGGCUUCAUCAAGAGACAGCUGGUGGAAACCCGGCAGAUCACAAAGCACGUGGCACAGAUCCUGGACUCCCGGAUGAACACUAAGUACGACGAGAAUGACAAGCUGAUCCGGGAAGUGAAAGUGAUCACCCUGAAGUCCAAGCUGGUGUCCGAUUUCCGGAAGGAUUUCCAGUUUUACAAAGUGCGCGAGAUCAACAACUACCACCACGCCCACGACGCCUACCUGAACGCCGUCGUGGGAACCGCCCUGAUCAAAAAGUACCCUGCGCUGGAAAGCGAGUUCGUGUACGGCGACUACAAGGUGUACGACGUGCGGAAGAUGAUCGCCAAGAGCGAGCAGGAAAUCGGCAAGGCUACCGCCAAGUACUUCUUCUACAGCAACAUCAUGAACUUUUUCAAGACCGAGAUUACCCUGGCCAACGGCGAGAUCCGGAAGGCGCCUCUGAUCGAGACAAACGGCGAAACCGGGGAGAUCGUGUGGGAUAAGGGCCGGGAUUUUGCCACCGUGCGGAAAGUGCUGAGCAUGCCCCAAGUGAAUAUCGUGAAAAAGACCGAGGUGCAGACAGGCGGCUUCAGCAAAGAGUCUAUCCUGCCCAAGAGGAACAGCGAUAAGCUGAUCGCCAGAAAGAAGGACUGGGACCCUAAGAAGUACGGCGGCUUCGACAGCCCCACCGUGGCCUAUUCUGUGCUGGUGGUGGCCAAAGUGGAAAAGGGCAAGUCCAAGAAACUGAAGAGUGUGAAAGAGCUGCUGGGGAUCACCAUCAUGGAAAGAAGCAGCUUCGAGAAGAAUCCCAUCGACUUUCUGGAAGCCAAGGGCUACAAAGAAGUGAAAAAGGACCUGAUCAUCAAGCUGCCUAAGUACUCCCUGUUCGAGCUGGAAAACGGCCGGAAGAGAAUGCUGGCCUCUGCCGGCGAACUGCAGAAGGGAAACGAACUGGCCCUGCCCUCCAAAUAUGUGAACUUCCUGUACCUGGCCAGCCACUAUGAGAAGCUGAAGGGCUCCCCCGAGGAUAAUGAGCAGAAACAGCUGUUUGUGGAACAGCACAAGCACUACCUGGACGAGAUCAUCGAGCAGAUCAGCGAGUUCUCCAAGAGAGUGAUCCUGGCCGACGCUAAUCUGGACAAAGUGCUGUCCGCCUACAACAAGCACCGGGAUAAGCCCAUCAGAGAGCAGGCCGAGAAUAUCAUCCACCUGUUUACCCUGACCAAUCUGGGAGCCCCUGCCGCCUUCAAGUACUUUGACACCACCAUCGACCGGAAGAGGUACACCAGCACCAAAGAGGUGCUGGACGCCACCCUGAUCCACCAGAGCAUCACCGGCCUGUACGAGACACGGAUCGACCUGUCUCAGCUGGGAGGCGACAAAAGGCCGGCGGCCACGAAAAAGGCCGGCCAGGCAAAAAAGAAAAAGUAG(如SEQ NO:3所示)。*Topology: linear; AUGGACUAUAAGGACCACGACGGAGACUACAAGGAUCAUGAUAUUGAUUACAAAGACGAUGACGAUAAGAUGGCCCCAAAGAAGAAGCGGAAGGUCGGUAUCCACGGAGUCCCAGCAGCCGACA AGAAGUACAGCAUCGGCCUGGACAUCGGCACCAACUCUGUGGGCUGGGCCGUGAUCACCGACGAGUACAAGGUGCCCAGCAAGAAAUUCAAGGUGCUGGGCAACACCGACCGGCACAGCAUCAAGAAGAACCUG AUCGGAGCCCUGCUGUUCGACAGCGGCGAAACAGCCGAGGCCACCCGGCUGAAGAGAACCGCCAGAAGAAGAUACACCAGACGGAAGAACCGGAUCUGCUAUCUGCAAGAGAUCUUCAGCAACGAGAUGGCCAA GGUGGACGACAGCUUCUUCCACAGACUGGAAGAGUCCUUCCUGGUGGAAGAGGAUAAGAAGCACGAGCGGCACCCAUCUUCGGCAACAUCGUGGACGAGGUGGCCUACCACGAGAAGUACCCCACCAUCUACC ACCUGAGAAAGAAACUGGUGGGACAGCACCGACAAGGCCGACCUGCGGCUGAUCUAUCUGGCCCUGGCCCACAUGAUCAAGUUCCGGGGCCACUUCCUGAUCGAGGGCGACCUGAACCCCGACAACAGCGACGUG GACAAGCUGUUCAUCCAGCUGGUGCAGACCUACAACCAGCUGUUCGAGGAAAACCCCAUCAACGCCAGCGGCGUGGACGCCAAGGCCAUCCUGUCUGCCAGACUGAGCAAGAGCAGACGGCUGGAAAAUCUGAU CGCCCAGCUGCCCGGCGAGAAGAAGAAUGGCCUGUUCGGAAACCUGAUUGCCCUGAGCCUGGGCCUGACCCCCAACUUCAAGAGCAACUUCGACCUGGCCGAGGAUGCCAAACUGCAGCUGAGCAAGGACACCU ACGACGACGACCUGGACAACCUGCUGGCCCAGAUCGGCGACCAGUACGCCGACCUGUUUCUGGCCGCCAAGAACCUGUCCGACGCCAUCCUGCUGAGCGACAUCCUGAGAGUGAACACCGAGAUCACCAAGGCC CCCCUGAGCGCCUCUAUGAUCAAGAGAUACGACGAGCACCACCAGGACCUGACCCUGCUGAAAGCUCUCGUGCGGCAGCAGCUGCCUGAGAAGUACAAAGAGAUUUUCUUCGACCAGAGCAAGAACGGCUACGC CGGCUACAUUGACGGCGGAGCCAGCCAGGAAGAGUUCUACAAGUUCAUCAAGCCCAUCCUGGAAAAGAUGGACGGCACCGAGGAACUGCUCGUGAAGCUGAACAGAGAGGACCUGCUGCGGAAGCAGCGGACCU UCGACAACGGCAGCAUCCCCCACCAGAUCCACCUGGGAGAGCUGCACGCCAUUCUGCGGCGGCAGGAAGAUUUUUACCCAUUCCUGAAGGACAACCGGGAAAAGAUCGAGAAGAUCCUGACCUUCCGCAUCCCC UACUACGUGGGCCCUCUGGCCAGGGGAAACAGCAGAUUCGCCUGGAUGACCAGAAAGAGCGAGGAAACCAUCACCCCCUGGAACUUCGAGGAAGUGGUGGACAAGGGCGCUUCCGCCCAGAGCUUCAUCGAGCG GAUGACCAACUUCGAUAAGAACCUGCCCAACGAGAAGGUGCUGCCCAAGCACAGCCUGCUGUACGAGUACUUCACCGUGUAUAACGAGCUGACCAAAGUGAAAUACGUGACCGAGGGAAUGAGAAAGCCCGCCU UCCUGAGCGGCGAGCAGAAAAAGGCCAUCGUGGACCUGCUGUUCAAGACCAACCGGAAAGUGACCGUGAAGCAGCUGAAAGAGGACUACUUCAAGAAAAUCGAGUGCUUCGACUCCGUGGAAAUCUCCGGCG GAAGAUCGGUUCAACGCCUCCCUGGGCACAUACCACGAUCUGCUGAAAAUUAUCAAGGACAAGGACUUCCUGGACAAUGAGGAAAACGAGGACAUUCUGGAAGAUAUCGUGCUGACCCUGACACUGUUUGAGGA CAGAGAGAUGAUCGAGGAACGGCUGAAAACCUAUGCCCACCUGUUCGACGACAAAGUGAUGAAGCAGCUGAAGCGGCGGAGAUACACCGGCUGGGGCAGGCUGAGCCGGAAGCUGAUCAACGGCAUCCGGGACA AGCAGUCCGGCAAGACAAUCCUGGAUUUCCUGAAGUCCGACGGCUUCGCCAACAGAAACUUCAUGCAGCUGAUCCACGACGACAGCCUGACCUUUAAAGAGGACAUCCAGAAAGCCCAGGUGUCCGGCCAGGG CGAUAGCCUGCACGAGCACAUUGCCAAUCUGGCCGGCAGCCCCGCCAUUAAGAAGGGCAUCCUGCAGACAGUGAAGGUGGUGGACGAGCUCGUGAAAGUGAUGGGCCGGCACAAGCCCGAGAACAUCGUGAUCG AAAUGGCCAGAGAGAACCAGACCACCCAGAAGGGACAGAAGAACAGCCGCGAGAGAAUGAAGCGGAUCGAAGAGGGCAUCAAAGAGCUGGGCAGCCAGAUCCUGAAAGAACACCCGGUGGAAAACACCCAGCUG CAGAACGAGAAGCUGUACCUGUACUACCUGCAGAAUGGGCGGGAUAUGUACGUGGACCAGGAACUGGACAUCAACCGGCUGUCCGACUACGAUGUGGACCAUAUCGUGCCUCAGAGCUUUCUGGCCGACGACUC CAUCGACAACAAGGUGCUGACCAGAAGCGACAAGAACCGGGGCAAGAGCGACAACGUGCCCUCCGAAGAGGUCGUGAAGAAGAUGAAGAACUACUGGCGGCAGCUGCUGAACGCCAAGCUGAUUACCCAGAGAA AGUUCGACAAUCUGACCAAGGCCGAGAGAGGCGGCCUGAGCGAACUGGAUAAGGCCGGCUUCAUCAAGAGACAGCUGGUGGAAACCCGGCAGAUCACAAAGCACGUGGCACAGAUCCUGGACUCCCGGAUGAAC ACUAAGUACGACGAGAAUGACAAGCUGAUCCGGGAAGUGAAAGUGAUCACCCUGAAGUCCAAGCUGGUGUCCGAUUUCCGGAAGGAUUUCCAGUUUUACAAAGUGCGCGAGAUCAACAACUACCACCACGCCCA CGACGCCUACCUGAACGCCGUCGUGGGAACCGCCCUGAUCAAAAAGUACCCUGCGCUGGAAAGCGAGUUCGUGUACGGCGACUACAAGGUGUACGACGUGCGGAAGAUGAUCGCCAAGAGCGAGCAGGAAAUCG GCAAGGCUACCGCCAAGUACUUCUUCUACAGCAACAUCAUGAACUUUUUCAAGACCGAGAUUACCCUGGCCAACGGCGAGAUCCGGAAGGCGCCUCUGAUCGAGACAAACGGCGAAACCGGGGAGAUCGUGUGG GAUAAGGGCCGGGAUUUUGCCACCGUGCGGAAAGUGCUGAGCAUGCCCCAAGUGAAUAUCGUGAAAAAGACCGAGGUGCAGACAGGCGGCUUCAGCAAAGAGUCUAUCCUGCCCAAGAGGAACAGCGAUAAGCU GAUCGCCAGAAAGAAGGACUGGGACCCUAAGAAGUACGGCGGCUUCGACAGCCCCACCGUGGCCUAUUCUGUGCUGGUGGUGGCCAAAGUGGAAAAGGGCAAGUCCAAGAAACUGAAGAGUGUGAAAGAGCUGC UGGGGAUCACCAUCAUGGAAAGAAGCAGCUUCGAGAAGAAUCCCAUCGACUUUCUGGAAGCCAAGGGCUACAAAGAAGUGAAAAAGGACCUGAUCAUCAAGCUGCCUAAGUACUCCCUGUUCGAGCUGGAAAAC GGCCGGAAGAGAAUGCUGGCCUCUGCCGGCGAACUGCAGAAGGGAAACGAACUGGCCCUGCCCUCCAAAUAUGUGAACUUCCUGUACCUGGCCAGCCACUAUGAGAAGCUGAAGGGCUCCCCCGAGGAUAAUGA GCAGAAACAGCUGUUUGUGGAACAGCACAAGCACUACCUGGACGAGAUCAUCGAGCAGAUCAGCGAGUUCUCCAAGAGAGUGAUCCUGGCCGACGCUAAUCUGGACAAAGUGCUGUCCGCCUACAACAAGCACC GGGAUAAGCCCAUCAGAGAGCAGGCCGAAUAUCAUCCACCUGUUUACCCUGACCAAUCUGGGAGCCCCUGCCGCCUUCAAGUACUUUGACACCACCAUCGACCGGAAGAGGUACACCAGCACCAAAGAGGUG CUGGACGCCACCCUGAUCCACCAGAGCAUCACCGGCCUGUACGAGACACGGAUCGACCUGUCUCAGCUGGGAGGCGACAAAAGGCCGGCGGCCACGAAAAAGGCCGGCCAGGCAAAAAAGAAAAAGUAG (as SEQ NO: 3 shown).

在合成Cas9-mRNA时需进行化学修饰的引入,N1-甲基伪尿苷的引入具体包括:When synthesizing Cas9-mRNA, chemical modifications need to be introduced. The introduction of N1-methylpseudouridine specifically includes:

在合成mRNA的过程中,选择特定的尿嘧啶位点进行化学修饰,将尿苷(U)替换为N1-甲基伪尿苷(m1Ψ)。这种修饰有助于减少mRNA被人体内源性免疫系统识别的机会, m1Ψ修改减少了mRNA激活模式识别受体如RIG-I的能力。During the synthesis of mRNA, specific uracil sites are selected for chemical modification, replacing uridine (U) with N1-methyl pseudouridine (m1Ψ). This modification helps reduce the chance of mRNA being recognized by the human endogenous immune system. The m1Ψ modification reduces the ability of mRNA to activate pattern recognition receptors such as RIG-I.

使用转录酶在体外转录过程中引入m1Ψ,确保修饰的均匀性和高效率。The m1Ψ was introduced during in vitro transcription using a transcriptase, ensuring uniformity and high efficiency of modification.

聚腺苷酸尾部的添加:Addition of poly(A) tail:

在mRNA的3'端非编码区添加Poly(A)尾部,长度一般为100-250个腺苷酸单元。这一结构不仅可以保护mRNA免受外界酶的降解,还能增强其翻译效率。A poly(A) tail is added to the 3' non-coding region of mRNA, which is generally 100-250 adenylate units in length. This structure not only protects mRNA from degradation by external enzymes, but also enhances its translation efficiency.

在5'端,添加修饰的5'帽结构(如m^7G帽),这是通过共转录机制实现的,进一步提升mRNA的稳定性和效率。At the 5' end, a modified 5' cap structure (such as the m^7G cap) is added, which is achieved through a co-transcriptional mechanism, further improving the stability and efficiency of mRNA.

修饰mRNA的纯化与验证,Purification and verification of modified mRNA,

纯化过程:Purification process:

使用RNA纯化柱进行层析技术处理,从反应混合物中去除未反应的核苷酸、酶和其他杂质。RNA purification columns are used for chromatography to remove unreacted nucleotides, enzymes, and other impurities from the reaction mixture.

修饰后的mRNA通过离子交换柱或亲和层析柱进行进一步的净化,确保获取高纯度的产品。The modified mRNA is further purified by ion exchange columns or affinity chromatography columns to ensure high-purity products.

验证修饰的完整性与效率:Verify the integrity and efficiency of the modification:

利用凝胶电泳技术对修饰后的mRNA进行分析,通过比较修饰前后的迁移率差异来初步验证修饰的成功与否。The modified mRNA was analyzed using gel electrophoresis technology, and the success of the modification was preliminarily verified by comparing the difference in mobility before and after modification.

使用质谱分析进一步确认mRNA上各种修饰的存在和修饰率,确保每一步修饰的准确性和一致性。Mass spectrometry analysis is used to further confirm the presence and modification rate of various modifications on mRNA, ensuring the accuracy and consistency of each modification step.

实施末端测序分析,对修饰后的mRNA序列进行精确核对,确保无意外的序列变化或错配。Perform terminal sequencing analysis to accurately check the modified mRNA sequence to ensure there are no unexpected sequence changes or mismatches.

将上述合成修饰好的Cas9mRNA与Tyr-sgRNA共同包埋,通过上述类似的双乳化过程,将含50 µg Cas9mRNA+30 µg Tyr-sgRNA的DEPC水与Mal-PEG-b-PLGA/DOTAP混合材料,经过两次超声过程制备得到TNP/Cas9-mRNA&Tyr-sgRNA纳米颗粒。The above-mentioned synthesized and modified Cas9mRNA and Tyr-sgRNA were co-embedded, and through a double emulsification process similar to the above, DEPC water containing 50 µg Cas9mRNA + 30 µg Tyr-sgRNA and Mal-PEG-b-PLGA/DOTAP mixed material were prepared through two ultrasonic processes to obtain TNP/Cas9-mRNA&Tyr-sgRNA nanoparticles.

4. 递送方法及验证4. Delivery Method and Verification

使用浸泡法递送纳米载体。具体操作是将斑马鱼胚胎放入含有纳米载体(包埋Cas9mRNA与Tyr-sgRNA的TAT-PEG-b-PLGA材料)的溶液中,允许纳米粒子自然与胚胎接触并被胚胎细胞摄取。递送过程中,需要精确控制纳米载体的浓度,通常在1至100 μg/mL范围内,具体浓度依据胚胎的接受能力和细胞类型而定。此外,浸泡时间也是一个关键参数,一般持续时间从1-12h不等,根据不同的胚胎发育速率来调整。Nanocarriers are delivered using the immersion method. The specific operation is to place zebrafish embryos in a solution containing nanocarriers (TAT-PEG- b -PLGA material encapsulating Cas9mRNA and Tyr-sgRNA), allowing the nanoparticles to naturally contact the embryo and be taken up by embryonic cells. During the delivery process, the concentration of the nanocarrier needs to be precisely controlled, usually in the range of 1 to 100 μg/mL, and the specific concentration depends on the embryo's receptive capacity and cell type. In addition, the immersion time is also a key parameter, generally lasting from 1-12 hours, and is adjusted according to different embryonic development rates.

为了最大限度地提高转染效率并减少细胞毒性,可以在递送过程中使用微动或低速离心等物理方法帮助纳米粒子更均匀地分布和接触胚胎细胞。这种方法的优势在于其非侵入性和可重复性,适合于规模化操作。To maximize transfection efficiency and reduce cytotoxicity, physical methods such as microfluidization or low-speed centrifugation can be used during delivery to help nanoparticles distribute and contact embryonic cells more evenly. The advantages of this method are its non-invasiveness and reproducibility, which is suitable for large-scale operations.

基因编辑的执行,Implementation of gene editing,

在纳米载体递送到胚胎细胞后,Cas9蛋白和sgRNA将在细胞内释放。Cas9蛋白与sgRNA形成复合体,通过sgRNA指导的序列特异性结合到目标DNA上,Cas9酶随后在指定位置切割双链DNA,启动细胞的DNA修复机制。这一过程中,可以通过同源重组(HDR)或非同源末端连接(NHEJ)介导的修复路径引入突变或修正特定的基因错误。After the nanovector is delivered to the embryonic cells, the Cas9 protein and sgRNA will be released inside the cells. The Cas9 protein forms a complex with the sgRNA and binds to the target DNA through the sequence specificity guided by the sgRNA. The Cas9 enzyme then cuts the double-stranded DNA at the specified position and initiates the cell's DNA repair mechanism. In this process, mutations can be introduced or specific genetic errors can be corrected through homologous recombination (HDR) or non-homologous end joining (NHEJ)-mediated repair pathways.

5.效果验证详细说明5. Detailed description of effect verification

本发明方法的效果验证是一个关键步骤,以确保基因编辑的正确性和有效性。以下是对4中各步骤的详细扩展:The effect verification of the method of the present invention is a key step to ensure the correctness and effectiveness of gene editing. The following is a detailed expansion of each step in 4:

5.1. 样本采集5.1. Sample collection

在进行基因编辑的水产动物胚胎中,基因编辑后的特定时间点(通常是编辑后72小时以上,根据物种和胚胎的发育速度定)进行样本采集。使用精细的显微操作工具,在显微镜下轻柔地从每个编辑过的胚胎中提取少量组织样本,或在不影响胚胎发育的情况下提取整个胚胎。为了防止样本污染和DNA降解,所有操作应在无菌环境下进行,并且迅速将样本转移到适宜的保存液中或直接进行DNA提取。In gene-edited embryos of aquatic animals, samples are collected at specific time points after gene editing (usually more than 72 hours after editing, depending on the species and the development speed of the embryo). Using fine micromanipulation tools, a small amount of tissue samples are gently extracted from each edited embryo under a microscope, or the entire embryo is extracted without affecting embryonic development. To prevent sample contamination and DNA degradation, all operations should be performed in a sterile environment, and the samples should be quickly transferred to a suitable preservation solution or directly subjected to DNA extraction.

5.2. 基因编辑验证5.2. Gene Editing Validation

从采集的样本中提取DNA后,利用特定的引物通过聚合酶链反应(PCR)扩增目标基因区域。扩增的DNA片段应包括CRISPR/Cas9预期编辑的位点。使用特定设计的引物对Tyr基因的目标编辑区域进行PCR扩增。这一步骤是为了确保测序覆盖所有潜在的编辑位点,引物设计需要包括PAM序列附近及可能的编辑区域。After extracting DNA from the collected samples, the target gene region is amplified by polymerase chain reaction (PCR) using specific primers. The amplified DNA fragment should include the site expected to be edited by CRISPR/Cas9. PCR amplification of the target editing region of the Tyr gene is performed using specifically designed primers. This step is to ensure that sequencing covers all potential editing sites, and the primer design needs to include the vicinity of the PAM sequence and possible editing regions.

测序库准备:使用PCR产品进行测序库构建。在此过程中,将加入适当的接头和索引标签,以便在Illumina平台上进行多重测序。Sequencing library preparation: Sequencing library construction is performed using PCR products. During this process, appropriate adapters and index tags are added to allow for multiplex sequencing on the Illumina platform.

测序库准备,Sequencing library preparation,

准确的库构建是高通量测序成功的关键。Accurate library construction is critical to the success of high-throughput sequencing.

末端修饰和接头连接:PCR扩增产物首先进行末端修饰,通常包括加A尾处理和接头连接。在此过程中,将特定的接头序列连接到PCR产物的两端,这些接头包含用于Illumina测序的序列和索引标签,允许在同一测序反应中并行处理多个样品。End modification and adapter ligation: PCR amplification products are first subjected to end modification, usually including A-tailing and adapter ligation. In this process, specific adapter sequences are ligated to both ends of the PCR product. These adapters contain sequences and index tags for Illumina sequencing, allowing multiple samples to be processed in parallel in the same sequencing reaction.

文库纯化和验证:接头连接后,使用凝胶电泳或磁珠净化方法去除未连接的接头和过小的PCR产物。纯化后的文库通过qPCR和毛细管电泳分析其浓度和大小分布,确保文库质量符合高通量测序的要求。Library purification and validation: After the adapter is connected, gel electrophoresis or magnetic bead purification is used to remove unconnected adapters and PCR products that are too small. The purified library is analyzed for concentration and size distribution by qPCR and capillary electrophoresis to ensure that the library quality meets the requirements of high-throughput sequencing.

数据处理和分析,Data processing and analysis,

高通量测序数据的处理和分析是确定基因编辑效果的核心环节。The processing and analysis of high-throughput sequencing data is the core link in determining the effect of gene editing.

数据质控:使用CASAVA等软件处理原始测序数据,进行基础的质量控制,包括去除低质量读段和修剪接头序列,以保证数据分析的准确性。Data quality control: Use CASAVA and other software to process raw sequencing data and perform basic quality control, including removing low-quality reads and trimming adapter sequences to ensure the accuracy of data analysis.

序列比对:利用MAFFT或BWA软件将清洗后的读段与参考基因组进行比对,精确识别编辑区域的序列变化。比对结果帮助确定CRISPR/Cas9介导的特定编辑事件,如单核苷酸变异、插入或缺失。Sequence alignment: Use MAFFT or BWA software to align the cleaned reads with the reference genome to accurately identify sequence changes in the edited region. The alignment results help determine specific CRISPR/Cas9-mediated editing events, such as single nucleotide variations, insertions, or deletions.

编辑效率和特异性分析:通过比对结果计算编辑位点的变异频率,评估编辑效率。同时,检查潜在的脱靶位点,确保编辑的特异性和精确性。Editing efficiency and specificity analysis: Calculate the variation frequency of editing sites through comparison results to evaluate editing efficiency. At the same time, check potential off-target sites to ensure the specificity and accuracy of editing.

测序结果的分析包括比较编辑前后的基因序列,特别关注Cas9酶引导的sgRNA所靶向的基因区域。分析是否存在预期的插入、缺失或替换突变(indels),以及这些变化的类型和频率。通过计算编辑效率(即成功编辑的胚胎数量与总胚胎数量的比例)和验证特异性编辑(即检查是否有非目标编辑发生),可以评估基因编辑的精确性和效果。Analysis of sequencing results includes comparing gene sequences before and after editing, with a particular focus on the gene region targeted by the sgRNA guided by the Cas9 enzyme. Analyze whether there are expected insertion, deletion, or substitution mutations (indels), as well as the type and frequency of these changes. The accuracy and effectiveness of gene editing can be evaluated by calculating the editing efficiency (i.e., the ratio of the number of successfully edited embryos to the total number of embryos) and verifying specific editing (i.e., checking whether non-target editing occurs).

斑马鱼鱼卵与TNP纳米载体封装的Cas9mRNA、Tyr-sgRNA共同孵育如图5所示,孵育72小时后提取基因组DNA进行NGS二代测序,测序突变结果展示。Zebrafish eggs were co-incubated with Cas9mRNA and Tyr-sgRNA encapsulated by TNP nanocarriers as shown in Figure 5. After 72 hours of incubation, genomic DNA was extracted for NGS second-generation sequencing, and the sequencing mutation results were displayed.

通过对斑马鱼幼体进行NGS(下一代测序)分析,结果进一步揭示了斑马鱼Tyr基因在靶点区域发生了明显的基因编辑变化。具体而言,在靶点区域检测到单个碱基的缺失以及大量的SNP(单核苷酸多态性)突变,这表明基因编辑工具成功引发了遗传变异。分析数据显示,Tyr基因的编辑效率达到36%,意味着在被分析的斑马鱼幼体中,约有36%的基因序列发生了预期的编辑或突变。图5中的这些结果表明,TNP纳米载体封装的EGFP-mRNA不仅成功进入了斑马鱼细胞,并在细胞内表达,而且能够有效地在靶向基因处引发突变和缺失。By performing NGS (next generation sequencing) analysis on zebrafish larvae, the results further revealed that the zebrafish Tyr gene had undergone significant gene editing changes in the target region. Specifically, single base deletions and a large number of SNP (single nucleotide polymorphism) mutations were detected in the target region, indicating that the gene editing tool successfully caused genetic variation. Analysis data showed that the editing efficiency of the Tyr gene reached 36%, which means that in the zebrafish larvae analyzed, about 36% of the gene sequences underwent the expected editing or mutation. These results in Figure 5 show that the EGFP-mRNA encapsulated by the TNP nanocarrier not only successfully entered the zebrafish cells and was expressed in the cells, but also was able to effectively induce mutations and deletions at the targeted gene.

以上实施例仅用于阐述本发明,而本发明的保护范围并非仅仅局限于以上实施例。所属技术领域的普通技术人员依据以上本发明公开的内容均可实现本发明的目的,任何基于本发明构思基础上做出的改进和变形,均落入本发明的保护范围之内,具体保护范围以权利要求书记载的为准。The above embodiments are only used to illustrate the present invention, and the protection scope of the present invention is not limited to the above embodiments. A person skilled in the art can achieve the purpose of the present invention based on the above disclosure of the present invention, and any improvement and deformation based on the concept of the present invention shall fall within the protection scope of the present invention, and the specific protection scope shall be subject to the claims.

Claims (3)

1.一种用于水产动物基因编辑的纳米载体,其特征在于,包括a)TNP纳米颗粒以及b)Cas9mRNA和sgRNA;1. A nanocarrier for gene editing in aquatic animals, comprising a) TNP nanoparticles and b) Cas9 mRNA and sgRNA; 所述a)中的TNP纳米颗粒通过以下方法制备获得:The TNP nanoparticles in a) are prepared by the following method: (1)取马来酰亚胺化聚乙二醇Mal-PEG-OH和丙交酯D,L-LA以及乙交酯GA加入反应容器中,加入磁子并且在油浴中加热至130~150℃,持续搅拌直至反应容器中固体全部溶解,随后,在搅拌条件下滴加异辛酸亚锡Sn(Oct)2后继续搅拌反应2~5h,反应完毕浓缩产物沉淀在甲醇-乙醚混合液中,抽滤干燥得淡黄色Mal-PEG-b-PLGA;(1) Maleimidized polyethylene glycol Mal-PEG-OH, lactide D, L-LA and glycolide GA are added to a reaction vessel, a magnetic particle is added and the mixture is heated to 130-150°C in an oil bath, and stirring is continued until all the solids in the reaction vessel are dissolved. Subsequently, stannous isooctanoate Sn(Oct) 2 is added dropwise under stirring and the stirring reaction is continued for 2-5 hours. After the reaction is completed, the concentrated product is precipitated in a methanol-ether mixture, and filtered and dried to obtain light yellow Mal-PEG-b-PLGA; (2)取Mal-PEG-b-PLGA加入圆底容器中,再加入搅拌磁子和二甲基亚砜DMSO溶解Mal-PEG-b-PLGA,然后在搅拌条件下加入超纯水并继续搅拌,制备得Mal-NP纳米颗粒;(2) Mal-PEG-b-PLGA is added to a round-bottom container, and a stirring magnetic bar and dimethyl sulfoxide (DMSO) are added to dissolve the Mal-PEG-b-PLGA, and then ultrapure water is added under stirring and continued to stir to prepare Mal-NP nanoparticles; (3)将Mal-NP纳米颗粒在超纯水中透析过夜除去二甲基亚砜DMSO,收集透析袋内的纳米颗粒溶液转移至圆底容器中,加入穿膜肽TAT在氮气保护下搅拌反应,反应结束后,收集颗粒溶液离心,收集下层颗粒沉淀,冻干后得到TAT修饰的TAT-Mal-NP材料,简称TNP纳米颗粒;(3) The Mal-NP nanoparticles were dialyzed in ultrapure water overnight to remove dimethyl sulfoxide (DMSO), and the nanoparticle solution in the dialysis bag was collected and transferred to a round-bottom container. The cell-penetrating peptide TAT was added and stirred for reaction under nitrogen protection. After the reaction was completed, the particle solution was collected and centrifuged, and the lower particle precipitate was collected and freeze-dried to obtain TAT-modified TAT-Mal-NP material, referred to as TNP nanoparticles; 步骤(1)中所述马来酰亚胺化聚乙二醇Mal-PEG-OH、丙交酯D,L-LA与乙交酯GA的摩尔比为0.2:10.8~11.5:5.0~5.2;The molar ratio of maleimidized polyethylene glycol Mal-PEG-OH, lactide D,L-LA and glycolide GA in step (1) is 0.2:10.8-11.5:5.0-5.2; 步骤(3)中所述Mal-NP纳米颗粒与所述穿膜肽TAT的质量份配比为14~16:1.5~2.5;In step (3), the mass ratio of the Mal-NP nanoparticles to the cell-penetrating peptide TAT is 14-16:1.5-2.5; 步骤(3)中所述TNP纳米颗粒的平均粒径范围为50~200纳米;The average particle size of the TNP nanoparticles in step (3) is in the range of 50 to 200 nanometers; 将Cas9mRNA和sgRNA的超纯水溶液与TNP纳米颗粒和DOTAP混合,经过两次超声过程制备得到用于水产动物基因编辑的纳米载体。An ultrapure aqueous solution of Cas9mRNA and sgRNA was mixed with TNP nanoparticles and DOTAP, and a nanocarrier for gene editing in aquatic animals was prepared after two ultrasonic processes. 2.权利要求1所述TNP纳米颗粒作为水产动物mRNA递送载体的应用;所述水产动物为斑马鱼或斑节对虾。2. Use of the TNP nanoparticles according to claim 1 as a carrier for delivering mRNA to aquatic animals; the aquatic animals are zebrafish or giant tiger prawns. 3.一种基于权利要求1所述纳米载体的基因编辑方法,其特征在于,包括以下步骤:3. A gene editing method based on the nanocarrier according to claim 1, characterized in that it comprises the following steps: 1)将Cas9mRNA和sgRNA的超纯水溶液与TNP纳米颗粒和DOTAP混合,经过两次超声过程制备得到用于水产动物基因编辑的纳米载体;1) An ultrapure aqueous solution of Cas9 mRNA and sgRNA was mixed with TNP nanoparticles and DOTAP, and a nanocarrier for gene editing in aquatic animals was prepared through two ultrasonic processes; 2)将所述纳米载体递送至目标水产动物的胚胎中;2) delivering the nanocarrier into embryos of target aquatic animals; 3)在所述胚胎中进行目标基因的编辑;3) editing a target gene in the embryo; 步骤2)中所述的目标水产动物为斑马鱼或斑节对虾;The target aquatic animal in step 2) is zebrafish or giant tiger prawn; 步骤2)中将所述纳米载体递送至目标水产动物的胚胎中的方法为浸泡。In step 2), the method for delivering the nanocarrier into the embryo of the target aquatic animal is immersion.
CN202411826906.9A 2024-12-12 2024-12-12 Nanometer carrier for aquatic animal gene editing and gene editing method Active CN119286942B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202411826906.9A CN119286942B (en) 2024-12-12 2024-12-12 Nanometer carrier for aquatic animal gene editing and gene editing method

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202411826906.9A CN119286942B (en) 2024-12-12 2024-12-12 Nanometer carrier for aquatic animal gene editing and gene editing method

Publications (2)

Publication Number Publication Date
CN119286942A CN119286942A (en) 2025-01-10
CN119286942B true CN119286942B (en) 2025-07-04

Family

ID=94167640

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202411826906.9A Active CN119286942B (en) 2024-12-12 2024-12-12 Nanometer carrier for aquatic animal gene editing and gene editing method

Country Status (1)

Country Link
CN (1) CN119286942B (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102988295A (en) * 2011-09-09 2013-03-27 复旦大学 Cell-penetrating peptide modified nanoparticle and its preparation method
WO2020092725A1 (en) * 2018-11-01 2020-05-07 Montana State University Gene modulation with crispr system type i

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3074047A2 (en) * 2013-11-26 2016-10-05 The Brigham and Women's Hospital, Inc. Receptor-targeted nanoparticles for enhanced transcytosis mediated drug delivery
EP3155116A4 (en) * 2014-06-10 2017-12-27 Massachusetts Institute Of Technology Method for gene editing
WO2017143042A2 (en) * 2016-02-16 2017-08-24 Yale University Compositions for enhancing targeted gene editing and methods of use thereof
WO2020033951A1 (en) * 2018-08-10 2020-02-13 Yale University Compositions and methods for embryonic gene editing in vitro
WO2023108076A1 (en) * 2021-12-08 2023-06-15 Yale University Surface conjugation to poly(amine-co-ester) nanoparticles for targeting to cells and tissues

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102988295A (en) * 2011-09-09 2013-03-27 复旦大学 Cell-penetrating peptide modified nanoparticle and its preparation method
WO2020092725A1 (en) * 2018-11-01 2020-05-07 Montana State University Gene modulation with crispr system type i

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
可生物降解TAT-PEG-PLA的制备及其性能研究;王婷;《中国优秀硕士学位论文全文数据库 工程科技Ⅰ辑》;20130415(第04期);摘要,第二章-第四章 *

Also Published As

Publication number Publication date
CN119286942A (en) 2025-01-10

Similar Documents

Publication Publication Date Title
CN107586835B (en) Single-chain-linker-based construction method and application of next-generation sequencing library
JP6531239B2 (en) Cell transfection method
CN113278619B (en) Double sgRNA, gene knockout vector, porcine fibroblast cell line with gene knockout STING gene and its construction method
CN112094868B (en) Method for preparing CD163 gene edited pig by using single base editor SpRY-BE4
CN113278618B (en) gRNA that specifically recognizes pig COL1A1 gene and its biological material, kit and application
CN105018523A (en) ZB (zebrafish) transposon system and gene transfer method mediated by same
CN112094866B (en) Method for preparing CD163 gene editing pig by using SpRY-Cas9 system
CN113736787A (en) gRNA of targeted mouse Atp7b gene and method for constructing Wilson disease mouse model
CN110250108B (en) RPRM gene knockout mouse model and its construction method and application
CN119286942B (en) Nanometer carrier for aquatic animal gene editing and gene editing method
CN110115248A (en) A kind of immunodeficient mouse, the Its Preparation Method And Use of somatostatin gene defect
CN112891365A (en) Preparation and application of 3D bionic cell implant capable of releasing microRNA nucleic acid drug
CN117859703A (en) Knock-in mouse model of new mutation pathogenic site C104G of Anxa11 gene, and construction method and application thereof
CN114410691B (en) Construction method and application of SLC35E1 gene knockout mouse animal model
CN116179543A (en) sgRNA and its application based on CRISPR specifically targeting porcine Cavin-1 gene
CN118562807B (en) A sgRNA for targeted editing of pig PNPLA3 gene and its application
CN109423500B (en) A kind of Mdr1a/1b double gene knockout method and application
CN113005147A (en) Construction method and application of mouse animal model with USP8 mutation
CN111471680A (en) Reconstructed eggs of zebrafish model and construction method and application thereof, construction method of zebrafish model
CN118599843B (en) SgRNA for targeted editing of pig HNF1A gene and application thereof
CN118910061B (en) sgRNA targeting porcine PLIN1 gene and its application
KR20200001022A (en) IRX1 Knock-out Transgenic Zebrafish Model and Method for Producing Thereof
CN112997966A (en) Mouse model knocking-in miRNA-125a based on CRISPR/Cas9 technology and construction method
CN118531065A (en) Targeting vector and method for pig UTY gene and application of targeting vector and method
CN119464262A (en) Development and application of high-efficiency and high-fidelity adenine base editors

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
点击 这是indexloc提供的php浏览器服务,不要输入任何密码和下载