[go: up one dir, main page]

CN116763945A - Exosome diagnosis and treatment agent based on M2 microglial cells, preparation method and application thereof in preparation of medical imaging probes - Google Patents

Exosome diagnosis and treatment agent based on M2 microglial cells, preparation method and application thereof in preparation of medical imaging probes Download PDF

Info

Publication number
CN116763945A
CN116763945A CN202310776270.0A CN202310776270A CN116763945A CN 116763945 A CN116763945 A CN 116763945A CN 202310776270 A CN202310776270 A CN 202310776270A CN 116763945 A CN116763945 A CN 116763945A
Authority
CN
China
Prior art keywords
evs
exosomes
exosome
microglia
imaging
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
CN202310776270.0A
Other languages
Chinese (zh)
Inventor
汤耀辉
张璐
吴胜菊
施晓婧
王继先
张春富
杨国源
李浩宇
刘艳
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.)
Ankerui Shanxi Biological Cell Co ltd
Original Assignee
Ankerui Shanxi Biological Cell Co ltd
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 Ankerui Shanxi Biological Cell Co ltd filed Critical Ankerui Shanxi Biological Cell Co ltd
Priority to CN202310776270.0A priority Critical patent/CN116763945A/en
Publication of CN116763945A publication Critical patent/CN116763945A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/0002General or multifunctional contrast agents, e.g. chelated agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/0019Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules
    • A61K49/0021Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules the fluorescent group being a small organic molecule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/005Fluorescence in vivo characterised by the carrier molecule carrying the fluorescent agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/005Fluorescence in vivo characterised by the carrier molecule carrying the fluorescent agent
    • A61K49/0052Small organic molecules
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/08Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by the carrier
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/06Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations
    • A61K49/08Nuclear magnetic resonance [NMR] contrast preparations; Magnetic resonance imaging [MRI] contrast preparations characterised by the carrier
    • A61K49/10Organic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Engineering & Computer Science (AREA)
  • Optics & Photonics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Physics & Mathematics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Radiology & Medical Imaging (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

本发明提供一种基于M2型小胶质细胞的外泌体诊疗剂、制备方法及其在制备医学成像探针中的应用,制备M2小胶质细胞来源的外泌体,标记并修饰能够靶向脑部神经元的小分子配体能够特异性的在脑卒中区域高剂量主动靶向迁移,提升外泌体诊疗剂在脑卒中区域的滞留能力,提高其对脑卒中区域的修复疗效。首次实现了荧光染料、SPECT信号放射性核素125I和磁共振T2加权对比剂超顺磁性氧化铁的无螯合剂直接共标记,满足了荧光成像、核医学及磁共振成像设备对多模态成像探针的需求,实现了无创、精准、实时、动态体内监控不同给药途径下外泌体诊疗剂在靶区域迁移、归巢及分布情况,同时评估治疗效果,以便调整给药时间,实现对脑卒中的有效修复,进一步提升治疗效果。The present invention provides an exosome diagnostic and therapeutic agent based on M2 microglia, a preparation method and its application in preparing medical imaging probes. Exosomes derived from M2 microglia are prepared and labeled and modified to be able to target. Small molecule ligands to brain neurons can actively target and migrate specifically to the stroke area at high doses, improving the retention ability of exosome therapeutic agents in the stroke area and improving their repair efficacy in the stroke area. For the first time, chelator-free direct co-labeling of fluorescent dyes, SPECT signal radionuclide 125 I and magnetic resonance T2- weighted contrast agent superparamagnetic iron oxide has been achieved, which satisfies the requirements of multi-modality equipment for fluorescence imaging, nuclear medicine and magnetic resonance imaging. The demand for imaging probes enables non-invasive, accurate, real-time and dynamic in vivo monitoring of the migration, homing and distribution of exosome therapeutic agents in the target area under different administration routes, and at the same time evaluates the treatment effect in order to adjust the administration time and achieve Effective repair of stroke further improves the therapeutic effect.

Description

基于M2型小胶质细胞的外泌体诊疗剂、制备方法及其在制备 医学成像探针中的应用Exosome therapeutic agents based on M2 microglia, preparation methods and their preparation Applications in Medical Imaging Probes

技术领域Technical field

本发明属于生物医药领域,尤其涉及到一种基于M2型小胶质细胞的外泌体诊疗剂、制备方法及其在制备医学成像探针中的应用。The invention belongs to the field of biomedicine, and in particular relates to an exosome therapeutic agent based on M2 microglia, a preparation method and its application in preparing medical imaging probes.

背景技术Background technique

卒中在世界范围内是致残和致死的主要原因,由于治疗时间窗和治疗手段的限制,其诊断及治疗的形势严峻。近年来,包括外泌体在内的细胞外囊泡(EVs)在治疗卒中方面显示出巨大的潜力。我们之前的研究表明,M2小胶质细胞衍生的外泌体可以减轻神经炎症,保护小鼠大脑免受缺血再灌注损伤,并促进卒中小鼠的白质修复和重塑。然而由于缺乏基于EVs治疗的体内可视化有效策略,明确EVs治疗过程中的靶部位迁移效率,严重阻碍了其临床转化。此外,面对EVs具有前景性的治疗,目前针对卒中最佳治疗效果而言,哪种是最优的给药方式也存在争议。因此,对EVs治疗过程中的定向迁移和分布的全面可视化和理解对于其进一步的临床转化至关重要。Stroke is the main cause of disability and death worldwide. Due to the limitations of treatment time window and treatment methods, the diagnosis and treatment situation is grim. In recent years, extracellular vesicles (EVs), including exosomes, have shown great potential in treating stroke. Our previous studies have shown that M2 microglia-derived exosomes can reduce neuroinflammation, protect mouse brains from ischemia-reperfusion injury, and promote white matter repair and remodeling in stroke mice. However, the lack of effective strategies for in vivo visualization based on EVs treatment to clarify the target site migration efficiency during EVs treatment has seriously hindered its clinical translation. In addition, in the face of the promising treatment of EVs, there is currently controversy over which method of administration is optimal for the best therapeutic effect in stroke. Therefore, a comprehensive visualization and understanding of the directional migration and distribution of EVs during therapy is crucial for their further clinical translation.

为了解决如上尚未解决的需求,迫切需要先进的成像技术,能够对外源性引入的EVs进行无创、可重复和定量的监测。在影像学诊断方面,常规的计算机断层扫描(CT)成像和磁共振成像(MRI)是用于脑卒中患者诊断的主要成像手段。以PET,SPECT为代表的核医学成像,以及将核医学与常规成像技术结合的SPECT/CT,PET/CT,PET/MRI以及荧光成像等成像技术已被广泛用于无创EVs示踪,在脑卒中基础研究方面发挥着越来越重要的作用。尽管每种成像方式都获得了一定的进展,但它们在从全身到单细胞水平精确监测EVs在体内靶部位迁移和疗效发挥方面都有局限性。例如,荧光成像能够研究细胞和亚细胞过程,但总是受到非特异性“背景”信号的影响,多数条件下无法实现无创显像。MRI提供了超高的空间分辨率和软组织对比度,而其灵敏度不足以实现EVs的全身成像和定量跟踪。由于核医学成像灵敏度高,允许对外源性EVs的体内生物分布进行全身可视化和动态监测,但不能够提供足够的空间解剖学信息,特别是在缺血性脑病方面。因此,开发一种针对EVs的适当标记策略,允许可以对其进行多模态成像,全面监控不同给药途径后的EVs在体内靶部位的定向迁移和分布情况,对于剖析基于EVs的治疗机制至关重要,且有利于其进一步临床转化。In order to address the above unresolved needs, there is an urgent need for advanced imaging technologies that can perform non-invasive, reproducible and quantitative monitoring of exogenously introduced EVs. In terms of imaging diagnosis, conventional computed tomography (CT) imaging and magnetic resonance imaging (MRI) are the main imaging methods used for the diagnosis of stroke patients. Nuclear medicine imaging represented by PET and SPECT, as well as imaging technologies such as SPECT/CT, PET/CT, PET/MRI and fluorescence imaging that combine nuclear medicine with conventional imaging technologies, have been widely used for non-invasive EVs tracing in the brain. Basic research on stroke plays an increasingly important role. Although each imaging modality has made certain progress, they all have limitations in accurately monitoring the migration and efficacy of EVs at target sites in vivo from the whole body to the single cell level. For example, fluorescence imaging can study cellular and subcellular processes, but is always affected by non-specific "background" signals and cannot achieve non-invasive imaging under most conditions. MRI provides ultra-high spatial resolution and soft tissue contrast, while its sensitivity is insufficient to achieve whole-body imaging and quantitative tracking of EVs. Due to the high sensitivity of nuclear medicine imaging, it allows whole-body visualization and dynamic monitoring of the in vivo biodistribution of exogenous EVs, but it is unable to provide sufficient spatial anatomical information, especially in ischemic encephalopathy. Therefore, developing an appropriate labeling strategy for EVs allows for multi-modal imaging and comprehensive monitoring of the directional migration and distribution of EVs at target sites in the body after different administration routes, which is important for dissecting the mechanism of EVs-based treatment. is important and conducive to its further clinical transformation.

发明内容Contents of the invention

本发明的一个目的是提供一种基于M2型小胶质细胞的外泌体诊疗剂、制备方法及其在制备医学成像探针中的应用,并提供至少后面将说明的优点。An object of the present invention is to provide an exosome therapeutic agent based on M2 microglia, a preparation method and its application in preparing medical imaging probes, and to provide at least the advantages that will be explained below.

本发明的另一个目的是提供一种基于M2型小胶质细胞的外泌体诊疗剂、制备方法及其在制备医学成像探针中的应用,制备了M2小胶质细胞来源的外泌体,标记并修饰了能够靶向脑部神经元的的小分子配体能够特异性的在脑卒中区域高剂量主动靶向迁移,提升外泌体诊疗剂在脑卒中区域的滞留能力,提高其对脑卒中区域的修复疗效。首次实现了荧光染料、SPECT信号放射性核素碘-125和磁共振T2加权对比剂超顺磁性氧化铁(SPIO)的无螯合剂直接共标记,满足了荧光成像、核医学及磁共振成像设备对多模态成像探针的需求,实现了无创、精准、实时、动态的体内监控不同给药途径下(尾静脉i.a.、颈动脉i.v和/或鼻腔i.n.给药)外泌体诊疗剂在靶区域的迁移、归巢及分布情况,同时,评估了治疗效果,以便调整给药时间,实现对脑卒中的有效修复进一步提升其的治疗效果。Another object of the present invention is to provide an exosome therapeutic agent based on M2 microglia, a preparation method and its application in preparing medical imaging probes. Exosomes derived from M2 microglia are prepared. , labeled and modified small molecule ligands that can target brain neurons can specifically and actively target migration at high doses in the stroke area, improve the retention ability of exosome therapeutic agents in the stroke area, and improve their response to the stroke area. Restorative efficacy in stroke areas. For the first time, direct co-labeling of fluorescent dyes, SPECT signal radionuclide iodine-125, and magnetic resonance T2-weighted contrast agent superparamagnetic iron oxide (SPIO) without chelating was achieved, which meets the requirements of fluorescence imaging, nuclear medicine and magnetic resonance imaging equipment. The demand for multi-modal imaging probes enables non-invasive, accurate, real-time and dynamic in vivo monitoring of exosome therapeutic agents in the target area under different administration routes (tail vein i.a., carotid artery i.v and/or nasal cavity i.n. administration) migration, homing and distribution, and at the same time, the therapeutic effect was evaluated in order to adjust the administration time to achieve effective repair of stroke and further improve its therapeutic effect.

本发明的技术方案如下:The technical solution of the present invention is as follows:

基于M2型小胶质细胞的外泌体诊疗剂,其包括基于M2型小胶质细胞的外泌体、外泌体表面修饰的荧光染料、多巴胺或多巴胺衍生物、放射性核素、磁共振成像离子、靶向神经元的配体。M2-type microglia-based exosome therapeutic agents, which include M2-type microglia-based exosomes, exosome surface-modified fluorescent dyes, dopamine or dopamine derivatives, radionuclides, and magnetic resonance imaging ions, neuron-targeting ligands.

优选的是,所述的基于M2型小胶质细胞的外泌体诊疗剂中,Preferably, among the exosome therapeutic agents based on M2 microglia,

所述外泌体来源为M2型小胶质细胞;The source of exosomes is M2 type microglia;

所述荧光染料选自DiR、Cy5.5、Cy7中的一种;The fluorescent dye is selected from one of DiR, Cy5.5, and Cy7;

所述多巴胺衍生物选自盐酸多巴胺;The dopamine derivative is selected from dopamine hydrochloride;

所述放射性核素选自碘-125、碘-131、镓-68、铜-64中的一种;The radioactive nuclide is selected from one of iodine-125, iodine-131, gallium-68, and copper-64;

所述磁共振成像离子选自Gd3+、Mn2+、Fe3+、Cu2+、Ni3+中的一种或多种;The magnetic resonance imaging ions are selected from one or more of Gd 3+ , Mn 2+ , Fe 3+ , Cu 2+ , and Ni 3+ ;

所述靶向神经元的配体选自RVG。The neuron-targeting ligand is selected from RVG.

基于M2型小胶质细胞的外泌体诊疗剂的制备方法,其包括以下步骤:A method for preparing exosome therapeutic agents based on M2 microglia, which includes the following steps:

1)体外培养未分化的M0型细胞,通过加入极化因子促进其向M2型细胞分化,并分泌外泌体;1) Cultivate undifferentiated M0 cells in vitro, promote their differentiation into M2 cells by adding polarizing factors, and secrete exosomes;

2)通过离心分离步骤1)的产物,收集并纯化得到M2型细胞来源的外泌体;2) Separate the product of step 1) by centrifugation, collect and purify exosomes derived from M2 cells;

3)将步骤2)得到的外泌体全部或部分进行经荧光染料染色标记;3) All or part of the exosomes obtained in step 2) are stained and labeled with fluorescent dyes;

4)将步骤3)得到的经过荧光染料染色标记的外泌体全部或部分表面上包覆多巴胺或多巴胺衍生物层;4) Coat all or part of the surface of the fluorescent dye-labeled exosomes obtained in step 3) with a dopamine or dopamine derivative layer;

5)将放射性核素、磁共振成像离子与步骤4)的产物混合;5) Mix radionuclides, magnetic resonance imaging ions and the product of step 4);

6) 将步骤5)的产物表面接枝靶向神经元的配体,获得所述外泌体。6) Graft the surface of the product of step 5) with a neuron-targeting ligand to obtain the exosomes.

优选的是,所述的基于M2型小胶质细胞的外泌体诊疗剂的制备方法中,所述步骤1)中,M0型细胞可选为小胶质细胞,极化因子可选为IL4,以总培养体系的总体积为准,控制培养体积为15 mL ,极化因子加入的浓度为20 ng/mL,极化培养时间为36-48小时,培养结束后,收集刺激后M2型小胶质细胞的上清,保存-20 ℃待提取外泌体。Preferably, in the preparation method of the exosome therapeutic agent based on M2 type microglia, in step 1), the M0 type cells can be microglia, and the polarizing factor can be IL4. , based on the total volume of the total culture system, the culture volume is controlled to 15 mL, the concentration of polarization factors added is 20 ng/mL, and the polarization culture time is 36-48 hours. After the culture is completed, collect the stimulated M2 small cells The supernatant of glial cells was stored at -20°C until exosomes were extracted.

优选的是,所述的基于M2型小胶质细胞的外泌体诊疗剂的制备方法中,所述步骤2)具体为:Preferably, in the method for preparing the exosome therapeutic agent based on M2 microglia, the step 2) is specifically:

收集刺激后的M2小胶质细胞的条件培养基超速离心,条件为4°C下以300 g连续离心10分钟、2000 g连续离心15分钟去除死细胞、10000 g连续离心30分钟去除细胞碎片,以及以100000 g连续超速离心70分钟沉淀外泌体,将EV用100000g的PBS洗涤一次70分钟,并悬浮以进行进一步表征;Collect the conditioned medium of stimulated M2 microglia and perform ultracentrifugation at 4°C at 300g for 10 minutes, 2000g for 15 minutes to remove dead cells, and 10000g for 30 minutes to remove cell debris. And the exosomes were precipitated by continuous ultracentrifugation at 100,000 g for 70 minutes, and the EVs were washed once with 100,000 g of PBS for 70 minutes and suspended for further characterization;

收集所得外泌体采用120 kV的透射电子显微镜鉴定结构,纳米粒子跟踪分析测量外泌体的直径和颗粒,BCA法测定外泌体的含量以及蛋白质免疫印迹分析外泌体标记物CD63、TSG101。The structure of the collected exosomes was identified using 120 kV transmission electron microscopy, nanoparticle tracking analysis was used to measure the diameter and particles of exosomes, BCA method was used to determine the content of exosomes, and Western blotting was used to analyze exosome markers CD63 and TSG101.

优选的是,所述的基于M2型小胶质细胞的外泌体诊疗剂的制备方法中,所述步骤3)具体为使用近红外荧光染料DiR和红色荧光染料PKH26修饰外泌体;Preferably, in the method for preparing the exosome therapeutic agent based on M2 microglia, step 3) specifically uses the near-infrared fluorescent dye DiR and the red fluorescent dye PKH26 to modify the exosomes;

对于DiR的标记过程,外泌体和1 μL稀释好的DiR溶液在37℃中孵育5 mins;For the DiR labeling process, exosomes and 1 μL of diluted DiR solution were incubated at 37°C for 5 mins;

对于体内外泌体摄取实验,1:500稀释后的PKH26与外泌体在室温下孵育5 mins;For in vivo exosome uptake experiments, PKH26 diluted 1:500 was incubated with exosomes at room temperature for 5 min;

全程采用不含EVs的血清;EVs-free serum is used throughout the process;

将标记好的外泌体在100000 g的PBS中洗涤1小时以洗掉非特异标记,并将外泌体可以在PBS中重悬待用。The labeled exosomes were washed in 100,000 g of PBS for 1 hour to wash away non-specific labeling, and the exosomes were resuspended in PBS for later use.

优选的是,所述的基于M2型小胶质细胞的外泌体诊疗剂的制备方法中,所述步骤4)具体包括:Preferably, in the method for preparing the exosome therapeutic agent based on M2 microglia, step 4) specifically includes:

将200 μL EVs与PBS按照1:1混合,将溶于8 mL Tris缓冲液(pH=8.5)中的1 mgPDA加入溶液中反应10 mins,使多巴胺的浓度为0.1 mg/mL;Mix 200 μL EVs and PBS at a ratio of 1:1, add 1 mg PDA dissolved in 8 mL Tris buffer (pH=8.5) into the solution and react for 10 mins to make the concentration of dopamine 0.1 mg/mL;

之后,将包裹有PDA的EVs悬浮在透析管中,通过10000 g超滤分离8分钟,然后用PBS洗涤三次,最终产物中包裹多巴胺的外泌体浓度约为1.2×1011个粒子/mL。Afterwards, the PDA-coated EVs were suspended in a dialysis tube, separated by 10000 g ultrafiltration for 8 minutes, and then washed three times with PBS. The concentration of dopamine-coated exosomes in the final product was approximately 1.2 × 10 11 particles/mL.

优选的是,所述的基于M2型小胶质细胞的外泌体诊疗剂的制备方法中,Preferably, in the preparation method of the exosome therapeutic agent based on M2 microglia,

所述步骤5)中放射性碘-125标记是通过经典的Iodogen方法实现的,将PDA@EVs悬浮液(0.5 mL,≈1.2×1011个粒子/mL)加入底部涂有20 μg Iodogen的玻璃管中。将新制备的Na125I溶液(500 μCi,18.5 MBq)加入管中,间歇振荡以避免PDA@EVs沉积,在室温下孵育30 mins后,通过超离心纯化最终产物,并用PBS洗涤三次;Radioactive iodine-125 labeling in step 5) is achieved by the classic Iodogen method. PDA@EVs suspension (0.5 mL, ≈1.2×10 11 particles/mL) is added to a glass tube coated with 20 μg Iodogen at the bottom. middle. Add the newly prepared Na 125 I solution (500 μCi, 18.5 MBq) into the tube, oscillate intermittently to avoid PDA@EVs deposition, and after incubating at room temperature for 30 mins, purify the final product by ultracentrifugation and wash three times with PBS;

PDA@EVs用氨基官能化的超顺磁性氧化铁(SPIO)以20 μg/mL的浓度标记1小时,然后洗涤和超滤以丢弃过量的SPIO颗粒。PDA@EVs were labeled with amino-functionalized superparamagnetic iron oxide (SPIO) at a concentration of 20 μg/mL for 1 h, followed by washing and ultrafiltration to discard excess SPIO particles.

优选的是,所述的基于M2型小胶质细胞的外泌体诊疗剂的制备方法中,所述步骤6)具体包括:将50 μg RVG加入EVs悬浮液300 μL中,并在室温下反应2-4小时。然后通过超滤纯化RVG修饰的EVs,并用PBS洗涤。Preferably, in the preparation method of the exosome therapeutic agent based on M2 microglia, the step 6) specifically includes: adding 50 μg RVG to 300 μL of EVs suspension, and reacting at room temperature. 2-4 hours. RVG-modified EVs were then purified by ultrafiltration and washed with PBS.

基于M2型小胶质细胞的外泌体诊疗剂在制备医学成像探针中的应用,所述医学成像选自核医学成像、磁共振成像、CT成像中的任一种或多种。Application of exosome therapeutic agents based on M2 microglia in the preparation of medical imaging probes, where the medical imaging is selected from any one or more of nuclear medicine imaging, magnetic resonance imaging, and CT imaging.

本发明不论是在修复脑卒中方面,还是在神经退行性疾病,包括老年痴呆,帕金森疾病以及脑外伤疾病方面均有用。The present invention is useful both in repairing stroke and in neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease and brain trauma diseases.

本发明的其它优点、目标和特征将部分通过下面的说明体现,部分还将通过对本发明的研究和实践而为本领域的技术人员所理解。Other advantages, objects, and features of the present invention will be apparent in part from the description below, and in part will be understood by those skilled in the art through study and practice of the present invention.

附图说明Description of drawings

图1为本发明的外泌体诊疗剂RVG-125I/SPIO-PDA@DiR-EVs示意图。Figure 1 is a schematic diagram of the exosome therapeutic agent RVG- 125I /SPIO-PDA@DiR-EVs of the present invention.

图2为本发明的外泌体诊疗剂RVG-125I/SPIO-PDA@DiR-EVs标记示意图。Figure 2 is a schematic diagram of labeling of RVG- 125I /SPIO-PDA@DiR-EVs, the exosome therapeutic agent of the present invention.

图3 为本发明的125I/SPIO-PDA@EVs的表征;A:EVs和PDA@EVs的TEM图像。B:EVs(左)和PDA@EVs(右)的纳米颗粒鉴定;C:EVs标志物CD63、TSG101的鉴定;D:考马斯亮蓝鉴定细胞裂解物、EVs和PDA@EVs;E:TEM显示PDA和SPIO标记后EVs形态的变化;F&D:Zeta电位和粒径显示PDA和SPIO标记的稳定性;H&I:放射性元素碘-125标记的稳定性;J:EDS mapping后显示元素的分布情况显示标记成功。Figure 3 shows the characterization of 125I/SPIO-PDA@EVs of the present invention; A: TEM images of EVs and PDA@EVs. B: Nanoparticle identification of EVs (left) and PDA@EVs (right); C: Identification of EVs markers CD63 and TSG101; D: Coomassie brilliant blue identification of cell lysates, EVs and PDA@EVs; E: TEM showing PDA and changes in EVs morphology after SPIO labeling; F&D: Zeta potential and particle size show the stability of PDA and SPIO labeling; H&I: Stability of radioactive element iodine-125 labeling; J: The distribution of elements after EDS mapping shows successful labeling .

图4 为在MRI成像显示中采用本发明的SPIO标记的EVs通过不同给药途径在缺血性卒中后的不同分布模式;比较i.a.、i.v.和i.n.三种方式注射后缺血性卒中脑血流的变化。A:体内实验示意图。B&C: tMACO后激光散斑显示脑血流的变化和定量;D、E&F: SPIO-PDA@EVs在tMCAO后24小时通过i.a.(D)、i.v.(E)和i.n.注射脑卒中小鼠T2加权磁共振成像。Figure 4 shows the different distribution patterns of SPIO-labeled EVs of the present invention after ischemic stroke through different administration routes in MRI imaging; comparing the cerebral blood flow of ischemic stroke after injection in three ways: i.a., i.v. and i.n. The change. A: Schematic diagram of in vivo experiments. B&C: Laser speckle shows changes and quantification of cerebral blood flow after tMACO; D, E&F: T2-weighted magnetic resonance imaging of SPIO-PDA@EVs injected i.a. (D), i.v. (E) and i.n. 24 hours after tMCAO in stroke mice Resonance imaging.

图5为体内SPECT/CT成像显示为本发明的125I标记的EVs通过不同给药途径在卒中小鼠中的动态分布;A-F: SPIO-PDA@EVs不同给药途径后体内的生物分布和各器官、组织的定量信号分析;G: 不同给药途径在不同时间点脑卒中小鼠脑内EVs的放射性强度。Figure 5 is in vivo SPECT/CT imaging showing the dynamic distribution of 125I-labeled EVs of the present invention in stroke mice through different administration routes; A-F: In vivo biodistribution and various organs of SPIO-PDA@EVs after different administration routes , Quantitative signal analysis of tissue; G: Radioactivity intensity of EVs in the brains of stroke mice by different administration routes at different time points.

图6 为通过不同的给药途径对DiR标记的EV进行荧光成像;A:DiR标记的EVs在卒中小鼠中的体内分布;B:脑和主要器官(肝、脾、肺、心和肾)中DiR标记的EVs的离体荧光信号;C:用DiR标记的EVs处理的卒中小鼠的脑和主要器官匀浆的定量;D:HE染色显示了卒中小鼠的脑、心、肺、肝、脾和肾的细胞结构(上图)。脑切片的共聚焦图像显示,在i.a.、i.v.或i.n.注射后48小时,脑卒中小鼠体内DiR标记的EV(红色)的离体分布;E:脑、心、肺、肝、脾和肾中DiR荧光强度的定量;F:脑切片的共聚焦成像显示,DiR标记的EVs在各类细胞内的分布;G:条形图显示了不同细胞类型的吞噬指数的定量。Figure 6 shows fluorescence imaging of DiR-labeled EVs through different administration routes; A: In vivo distribution of DiR-labeled EVs in stroke mice; B: Brain and major organs (liver, spleen, lung, heart and kidney) Ex vivo fluorescence signal of DiR-labeled EVs; C: Quantification of brain and major organ homogenates of stroke mice treated with DiR-labeled EVs; D: HE staining showing the brain, heart, lungs, and liver of stroke mice , spleen and kidney cellular structure (above). Confocal images of brain sections showing ex vivo distribution of DiR-labeled EVs (red) in stroke mice 48 hours after i.a., i.v., or i.n. injection; E: brain, heart, lungs, liver, spleen, and kidneys Quantification of DiR fluorescence intensity; F: Confocal imaging of brain slices shows the distribution of DiR-labeled EVs within various types of cells; G: Bar chart shows quantification of phagocytosis index in different cell types.

图7 为本发明的RVG修饰增加了体外EVs的神经元摄取;A:PDA@EVs和RVG-PDA@EVs的STEM图像;B:动态光散射(DLS)测量PDA@EVs和RVG-PDA@EVs;C:PDA@EVs和RVG-PDA@EVs的Zeta电位;D:共聚焦成像显示PKH26标记的EV(红色)与原代神经元和星形胶质细胞共标;E:RVG修饰增加了EVs对于神经元的靶向性;F:PKH26标记的EVs修饰前后与神经元、星形胶质细胞吞噬差异定量;G:定量显示不同组的神经元吞噬指数;H:用EVs、PDA@EVs, RVG-PDA@EVs和RVG+RVG-PDA@EVs处理的神经元的细胞活力。Figure 7 shows that the RVG modification of the present invention increases the neuronal uptake of EVs in vitro; A: STEM images of PDA@EVs and RVG-PDA@EVs; B: Dynamic light scattering (DLS) measurement of PDA@EVs and RVG-PDA@EVs ; C: Zeta potential of PDA@EVs and RVG-PDA@EVs; D: Confocal imaging shows that PKH26-labeled EVs (red) co-labeled with primary neurons and astrocytes; E: RVG modification increases EVs For neuron targeting; F: Quantification of the difference in phagocytosis between PKH26-labeled EVs and neurons and astrocytes before and after modification; G: Quantitative display of neuron phagocytosis index in different groups; H: Using EVs and PDA@EVs, Cell viability of neurons treated with RVG-PDA@EVs and RVG+RVG-PDA@EVs.

图8 为本发明的RVG修饰促进了缺血性脑中EVs的靶向递送。Figure 8 shows that the RVG modification of the present invention promotes the targeted delivery of EVs in ischemic brain.

A:SPIO-PDA@EVs和RVG-SPIO-PDA@EVs注射前后不同时间点脑卒中小鼠脑磁共振成像;B&C:注射后48小时脑切片的普鲁士蓝染色显示SPIO-PDA@EVs被细胞吞噬与量化;D&E:125I-PDA@EVs和RVG-125I-PDA@EVs动脉注射脑卒中小鼠后的成像和定量数据;F:125I-PDA@EVs和RVG-125I-PDA@EVs动脉注射后的代表性荧光图像;G:同侧脑匀浆中EVs荧光强度的定量;H&I:共聚焦成像显示EVs与神经元共定位和定量。A: Magnetic resonance imaging of the brain of stroke mice at different time points before and after injection of SPIO-PDA@EVs and RVG-SPIO-PDA@EVs; B&C: Prussian blue staining of brain sections 48 hours after injection shows that SPIO-PDA@EVs are phagocytosed by cells. and quantification; D&E: Imaging and quantitative data after arterial injection of 125 I-PDA@EVs and RVG- 125 I-PDA@EVs into stroke mice; F: 125 I-PDA@EVs and RVG- 125 I-PDA@EVs Representative fluorescence images after arterial injection; G: Quantification of fluorescence intensity of EVs in ipsilateral brain homogenates; H&I: Confocal imaging showing colocalization and quantification of EVs and neurons.

图9 为本发明的RVG修饰的M2 EVs进一步减少了tMCAO后小鼠的神经元凋亡并促进了神经功能恢复;A-D: TUNEL和Fluoro-Jeded B染色显示PBS、M2-EVs或RVG-M2-EVs处理的卒中小鼠中的神经元死亡与定量;E&F:M2-EVs和RVG-M2-EVs抑制了Cleaved caspase-3的表达及定量;G&H: M2-EVs和RVG-M2-EVs改善了卒中小鼠mNSS评分和转棒测试。Figure 9 shows that RVG-modified M2 EVs of the present invention further reduces neuronal apoptosis and promotes neurological function recovery in mice after tMCAO; A-D: TUNEL and Fluoro-Jeded B staining shows PBS, M2-EVs or RVG-M2- Neuronal death and quantification in stroke mice treated with EVs; E&F: M2-EVs and RVG-M2-EVs inhibited the expression and quantification of Cleaved caspase-3; G&H: M2-EVs and RVG-M2-EVs improved stroke mNSS score and rotarod test in mice.

图10 为本发明的M2-EVs 转录组学测序上调了凋亡相关的miRNA;A:M0-EVs、M2-EVs和RVG-M2-EVs的miRNA阵列数据的主成分(PC)分析;B:条形图显示了M2 EVs和M0 EVs中差异表达的miRNA;C&D:热图和火山图显示M2和M0组之间差异表达的miRNA最高;E:点图显示,与M0组相比,M2组上调的前20个富集通路。Figure 10 shows that M2-EVs transcriptomic sequencing of the present invention up-regulates apoptosis-related miRNAs; A: Principal component (PC) analysis of miRNA array data of M0-EVs, M2-EVs and RVG-M2-EVs; B: The bar graph shows the differentially expressed miRNAs in M2 EVs and M0 EVs; C&D: The heat map and volcano plot show that the differentially expressed miRNAs between the M2 and M0 groups are the highest; E: The dot plot shows that compared with the M0 group, the M2 group The top 20 enriched pathways that were upregulated.

具体实施方式Detailed ways

下面结合附图对本发明做进一步的详细说明,以令本领域技术人员参照说明书文字能够据以实施。The present invention will be further described in detail below with reference to the accompanying drawings, so that those skilled in the art can implement it with reference to the text of the description.

应当理解,本文所使用的诸如“具有”、“包含”以及“包括”术语并不配出一个或多个其它元件或其组合的存在或添加。It will be understood that terms such as "having," "comprising," and "including" as used herein do not connote the presence or addition of one or more other elements or combinations thereof.

目前已经开发了针对EVs直接和间接的标记策略以实现多模态成像示踪。然而间接标记方式需要预先对亲本细胞进行基因修饰,过程耗时且低效。因此相比之下直接标记方式标记分离的EVs更简便有效,但往往对外泌体造成不可逆的损伤,进一步影响其生物学活性。据报道,EVs可以通过物理吸附直接标记放射性碘-125,但是生理条件下物理吸附的放射性碘-125极其不稳定,非常容易脱标记,在体内实现放射性碘-125的非特异性摄取,进而影响成像效果。因此,在不损失EVs生物活性的情况下,建立一种简单但有效的多模式EVs可视化方法是必需的。近年来,聚多巴胺(PDA)因其自聚合性能和多功能化学反应性而引起了人们的广泛兴趣。与其他复杂的标记方法相比,多巴胺在弱碱性条件下可以通过邻苯二酚的自聚集,包裹在EVs表面, PDA涂层上的儿茶酚和蒽醌键可以很容易地与许多金属离子发生配位反应,如Fe3+、Gd3+、Pt4+。另外,PDA还可以通过迈克尔加成反应与卤素、巯基和氨基反应,从而有助于用放射性同位素、聚乙二醇(PEG)和靶向小分子配合物等修饰EVs,用于EVs的可视化成像,进一步提高EVs生理条件下的稳定性和治疗过程中的靶向能力。Direct and indirect labeling strategies for EVs have been developed to achieve multimodal imaging tracing. However, the indirect labeling method requires genetic modification of the parental cells in advance, which is time-consuming and inefficient. Therefore, direct labeling is more convenient and effective for labeling isolated EVs, but it often causes irreversible damage to exosomes and further affects their biological activity. According to reports, EVs can directly label radioactive iodine-125 through physical adsorption. However, physically adsorbed radioactive iodine-125 is extremely unstable under physiological conditions and is easily delabeled. Non-specific uptake of radioactive iodine-125 is achieved in the body, thereby affecting imaging. Effect. Therefore, it is necessary to establish a simple but effective multimodal EVs visualization method without losing the biological activity of EVs. In recent years, polydopamine (PDA) has attracted widespread interest due to its self-polymerizing properties and multifunctional chemical reactivity. Compared with other complex labeling methods, dopamine can be wrapped on the surface of EVs through the self-aggregation of catechol under weakly alkaline conditions, and the catechol and anthraquinone bonds on the PDA coating can easily bind to many metals. Coordination reactions occur with ions, such as Fe 3+ , Gd 3+ , and Pt 4+ . In addition, PDA can also react with halogens, sulfhydryl groups and amino groups through the Michael addition reaction, thereby helping to modify EVs with radioactive isotopes, polyethylene glycol (PEG) and targeted small molecule complexes for visual imaging of EVs. , to further improve the stability of EVs under physiological conditions and the targeting ability during treatment.

基于上述前提,我们通过收集并纯化M2型小胶质细胞分泌的外泌体并对其进行荧光染料染色后,将上述产物用多巴胺自聚集包裹,通过将磁共振显像造影剂和放射性核素直接偶联到PDA包裹层上,成功的构建出一种可用于脑卒中修复和核医学/磁共振成像/荧光三功能成像诊疗剂,以无创和定量地跟踪EVs在不同给药途径下对脑卒中区域的定向迁移、修复和疗效评估。实现了诊疗剂在生物个体、器官/组织、细胞和分子水平上的体内实时可视化动态监测。通过对外泌体诊疗剂偶联靶向脑部神经元的特异性配体,显著提高了Evs对脑缺血部位的神经元的靶向迁移量,来提高对脑卒中的治疗效果。通过EVs的miRNA测序,鉴定出与神经元凋亡相关的miRNA,阐明了M2小胶质细胞来源的诊疗剂在缺血性脑中的潜在治疗机制。本发明为EVs未来在脑血管疾病甚至是神经退行性相关疾病中的临床应用推广提供了强有力的标记方法和功能化修饰途径。Based on the above premise, we collected and purified the exosomes secreted by M2 microglia and stained them with fluorescent dyes. Then, we wrapped the above products with dopamine self-aggregation, and used magnetic resonance imaging contrast agents and radionuclides to Directly coupled to the PDA wrapping layer, we successfully constructed a diagnostic and therapeutic agent that can be used for stroke repair and nuclear medicine/magnetic resonance imaging/fluorescence three-functional imaging to non-invasively and quantitatively track the impact of EVs on the brain under different administration routes. Targeted migration, repair, and efficacy assessment of stroke areas. Real-time visual dynamic monitoring of diagnostic and therapeutic agents in vivo at the individual, organ/tissue, cellular and molecular levels is achieved. By conjugating exosome therapeutic agents with specific ligands targeting brain neurons, the amount of targeted migration of Evs to neurons in cerebral ischemic areas is significantly increased, thereby improving the therapeutic effect on stroke. Through miRNA sequencing of EVs, miRNAs related to neuronal apoptosis were identified, elucidating the potential therapeutic mechanism of M2 microglia-derived therapeutic agents in ischemic brain. The present invention provides a powerful labeling method and functional modification approach for the future clinical application and promotion of EVs in cerebrovascular diseases and even neurodegenerative related diseases.

本发明针对外泌体在内的细胞外囊泡(EVs)在治疗卒中方面显示出巨大的潜力,为EVs在卒中治疗过程中的定向迁移和分布的全面可视化和理解提供解决方案。The present invention shows great potential for treating extracellular vesicles (EVs) including exosomes, and provides a solution for the comprehensive visualization and understanding of the directional migration and distribution of EVs during stroke treatment.

本发明提供一种纳米级尺寸的外泌体诊疗剂探针,所述外泌体诊疗剂至少包括外泌体,标记外泌体的荧光染料、外泌体表面的多巴胺或多巴胺衍生物、放射性核素、磁共振成像离子、靶向神经元的配体。The present invention provides a nanometer-sized exosome diagnostic and therapeutic agent probe. The exosome therapeutic agent at least includes exosomes, fluorescent dyes that mark exosomes, dopamine or dopamine derivatives on the surface of exosomes, radioactive Nuclides, magnetic resonance imaging ions, ligands targeting neurons.

所述外泌体的来源选自小胶质细胞The source of exosomes is selected from microglia.

所述外泌体的来源细胞做以下预处理: 白介素4对M0型小胶质细胞诱导至少48小时后定向分化为M2型小胶质细胞。The source cells of the exosomes are subjected to the following pretreatment: Interleukin 4 induces M0 type microglia for at least 48 hours and then differentiates into M2 type microglia.

在本发明的某些实施方式中,所获得的外泌体通过荧光染料染色,所述荧光染料选自DiR、Cy5.5、Cy7的一种或多种。In certain embodiments of the present invention, the obtained exosomes are stained with a fluorescent dye selected from one or more of DiR, Cy5.5, and Cy7.

所述多巴胺衍生物选自盐酸多巴胺。The dopamine derivative is selected from dopamine hydrochloride.

所述放射性核素选自碘-125、碘-131、镓-68、铜-64中的一种。The radioactive nuclide is selected from one of iodine-125, iodine-131, gallium-68, and copper-64.

所述磁共振成像离子为T1或T2造影剂,选自Gd3+、Mn2+、Fe3+、Cu2+、Ni3+中的一种或多种;所述T1造影剂或T2造影剂由顺磁性或超顺磁性造影剂颗粒提供,所述顺磁性金属盐包括氯化钆、硝酸钆、醋酸钆、氯化锰、硝酸锰、醋酸锰、四氧化三铁中的一种或多种。The magnetic resonance imaging ion is a T1 or T2 contrast agent, selected from one or more of Gd 3+ , Mn 2+ , Fe 3+ , Cu 2+ , and Ni 3+ ; the T1 contrast agent or T2 contrast agent The agent is provided by paramagnetic or superparamagnetic contrast agent particles, and the paramagnetic metal salt includes one or more of gadolinium chloride, gadolinium nitrate, gadolinium acetate, manganese chloride, manganese nitrate, manganese acetate, and ferric oxide. kind.

所述靶向神经元的配体为蛋白、单克隆抗体或小分子抑制剂的一种或多种。在本发明的某些实施方式中,所述靶向神经元的配体选自RVG的一种或多种。The neuron-targeting ligand is one or more of a protein, a monoclonal antibody, or a small molecule inhibitor. In certain embodiments of the invention, the neuron-targeting ligand is selected from one or more RVGs.

所述外泌体诊疗剂为可用于医学成像的探针。所述医学成像选自核医学成像(例如PET、SPECT)、磁共振成像(MRI)、CT成像中的任一种或多种。例如所述探针可以作为PET/MRI显像探针、PET/CT显像探针、SPECT/MRI显像探针、SPECT/CT显像探针。The exosome therapeutic agent is a probe that can be used for medical imaging. The medical imaging is selected from any one or more of nuclear medicine imaging (such as PET, SPECT), magnetic resonance imaging (MRI), and CT imaging. For example, the probe can be used as a PET/MRI imaging probe, a PET/CT imaging probe, a SPECT/MRI imaging probe, or a SPECT/CT imaging probe.

在本发明的某些实施方式中,所述探针的结构示意图如图1所示。In some embodiments of the present invention, a schematic structural diagram of the probe is shown in Figure 1.

本发明还提供所述外泌体的制备方法,包括如下步骤:The invention also provides a method for preparing the exosomes, which includes the following steps:

体外培养未分化的M0型细胞,通过加入极化因子促进其向M2型细胞分化,并分泌外泌体;通过离心分离步骤1)的小胶质细胞,收集并纯化得到M2型细胞来源的外泌体;将步骤2)得到的外泌体全部或部分进行经荧光染料染色标记;Undifferentiated M0 type cells are cultured in vitro, and polarizing factors are added to promote their differentiation into M2 type cells and secrete exosomes; the microglia cells in step 1) are separated by centrifugation, and exosomes derived from M2 type cells are collected and purified. Exosomes; all or part of the exosomes obtained in step 2) are stained and labeled with fluorescent dyes;

将步骤3)得到的经过荧光染料染色标记的外泌体全部或部分表面上包覆多巴胺或多巴胺衍生物层;将放射性核素、磁共振成像离子与步骤4)的产物混合;6) 将步骤5)的产物表面接枝靶向神经元的配体,获得所述外泌体诊疗剂。Coat all or part of the surface of the fluorescent dye-labeled exosomes obtained in step 3) with a dopamine or dopamine derivative layer; mix radionuclides and magnetic resonance imaging ions with the product of step 4); 6) 5) The surface of the product is grafted with ligands targeting neurons to obtain the exosome therapeutic agent.

在本发明的某些优选实施方式中,步骤1)体外定向分化M0型细胞的步骤包括:将极化因子加入到M0型细胞,促进其向M2型细胞分化,并分泌外泌体。在一种实施方式中,M0型细胞可选为小胶质细胞,极化因子可选为IL4,以总培养体系的总体积为准,控制培养体积为15 mL 极化因子加入的浓度为20 ng/mL,极化培养时间为36-48小时,培养结束后,收集刺激后M2型小胶质细胞的上清,保存-20℃待提取外泌体。In certain preferred embodiments of the present invention, step 1) directed differentiation of M0 type cells in vitro includes: adding polarizing factors to M0 type cells to promote their differentiation into M2 type cells and secrete exosomes. In one embodiment, M0 type cells can be selected as microglia, and the polarizing factor can be selected as IL4. Based on the total volume of the total culture system, the culture volume is controlled to be 15 mL, and the concentration of the polarizing factor added is 20 ng/mL, and the polarization culture time is 36-48 hours. After the culture, collect the supernatant of the stimulated M2 microglia and store it at -20°C until exosomes are extracted.

在本发明的某些优选实施方式中,步骤2)中收集刺激后的M2小胶质细胞的条件培养基超速离心,条件为4 °C下以300 g连续离心10分钟、2000 g连续离心15分钟去除死细胞、10000 g连续离心30分钟去除细胞碎片,以及以100000 g连续超速离心70分钟沉淀外泌体,将EV用100000g的PBS洗涤一次70分钟,并悬浮以进行进一步表征。收集所得外泌体采用120 kV的透射电子显微镜(TEM)鉴定结构,纳米粒子跟踪分析(NTA)测量外泌体的直径和颗粒,BCA法测定外泌体的含量以及蛋白质免疫印迹分析外泌体标记物CD63、TSG101。In some preferred embodiments of the present invention, the conditioned medium of the stimulated M2 microglia cells collected in step 2) is ultracentrifuged under the conditions of continuous centrifugation at 300 g for 10 minutes at 4°C and continuous centrifugation at 2000 g for 15 minutes. minutes to remove dead cells, continuous centrifugation at 10,000 g for 30 minutes to remove cell debris, and continuous ultracentrifugation at 100,000 g for 70 minutes to precipitate exosomes. EVs were washed once with 100,000 g of PBS for 70 minutes and suspended for further characterization. The collected exosomes were characterized by transmission electron microscopy (TEM) at 120 kV to identify the structure, nanoparticle tracking analysis (NTA) to measure the diameter and particles of exosomes, BCA method to determine the content of exosomes, and protein immunoblotting to analyze exosomes. Markers CD63, TSG101.

在本发明的某些优选实施方式中,步骤3)具体包括:根据说明书,使用近红外荧光染料DiR和红色荧光染料PKH26修饰外泌体。对于DiR的标记过程,外泌体和1 μL稀释好的DiR溶液在37 ℃中孵育5 mins。对于体内外泌体摄取实验,1:500稀释后的PKH26与外泌体在室温下孵育5 mins。上述过程中为防止过度标记,全程采用不含EVs的血清。将标记好的外泌体在100000 g的PBS中洗涤1小时以洗掉非特异标记,并将外泌体可以在PBS中重悬待用。In some preferred embodiments of the present invention, step 3) specifically includes: according to the instructions, using the near-infrared fluorescent dye DiR and the red fluorescent dye PKH26 to modify the exosomes. For the DiR labeling process, exosomes were incubated with 1 μL of diluted DiR solution at 37°C for 5 mins. For in vivo exosome uptake experiments, PKH26 diluted 1:500 was incubated with exosomes for 5 min at room temperature. In order to prevent over-labeling during the above process, EVs-free serum was used throughout the process. The labeled exosomes were washed in 100,000 g of PBS for 1 hour to wash away non-specific labeling, and the exosomes were resuspended in PBS for later use.

在本发明的某些优选实施方式中,步骤4)具体包括:将200 μL EVs与PBS(1:1)混合,将溶于8 mL Tris缓冲液(pH=8.5)中的1 mg PDA加入溶液中反应10 mins,使多巴胺的浓度为0.1 mg/mL。之后,将包裹有PDA的EVs悬浮在透析管(100 kDa-MWCO)中,通过10000 g超滤分离8分钟,然后用PBS洗涤三次,最终产物中包裹多巴胺的外泌体浓度约为1.2×1011个粒子/mL。将PDA与氨基官能化吲哚菁绿(ICG)连接以评估标记效率,荧光强度ICG-PDA@EVs在超滤之前和之后用微孔板读取器进行测量以评估PDA标记率。In some preferred embodiments of the present invention, step 4) specifically includes: mixing 200 μL EVs and PBS (1:1), and adding 1 mg PDA dissolved in 8 mL Tris buffer (pH=8.5) to the solution React for 10 mins to make the concentration of dopamine 0.1 mg/mL. After that, the PDA-coated EVs were suspended in a dialysis tube (100 kDa-MWCO), separated by 10000 g ultrafiltration for 8 minutes, and then washed three times with PBS. The concentration of dopamine-encapsulated exosomes in the final product was approximately 1.2 × 10 11 particles/mL. PDA was conjugated with amino-functionalized indocyanine green (ICG) to evaluate the labeling efficiency, and the fluorescence intensity of ICG-PDA@EVs was measured with a microplate reader before and after ultrafiltration to evaluate the PDA labeling efficiency.

在本发明的放的某些优选实施方式中,步骤5)具体包括:放射性碘-125标记是通过经典的Iodogen方法实现的。将PDA@EVs悬浮液(0.5 mL,≈1.2×1011个粒子/mL)加入底部涂有20 μg Iodogen的玻璃管中。将新制备的Na125I溶液(500 μCi,18.5 MBq)加入管中,间歇振荡以避免PDA@EVs沉积。在室温下孵育30 mins后,通过超离心纯化最终产物,并用PBS洗涤三次。使用γ检测器进行放射性薄层色谱以评估标记效率。通过在37 °C的DMEM高糖溶液中,将5 μL样品在200 μL 10% FBS中共孵育不同时间(1、3、6、12和24小时)来评估放射性探针的稳定性。孵育后,通过离心收集探针,并对保留在颗粒上的放射性进行计数。硅胶色谱纸和0.9%氯化钠用作固定相和流动相。125I标记的SPIO-PDA@EVs保留在原点,而游离的125I随流动相保留在前沿, 125I标记的SPIO-PDA@EVs的放射化学纯度表示为保留在原点的SPIO-PDA@EVs探针的放射性剂量与总放射性剂量的百分比。In certain preferred embodiments of the present invention, step 5) specifically includes: radioactive iodine-125 labeling is achieved by the classic Iodogen method. PDA@EVs suspension (0.5 mL, ≈1.2 × 10 11 particles/mL) was added to a glass tube coated with 20 μg Iodogen at the bottom. Add freshly prepared Na 125 I solution (500 μCi, 18.5 MBq) into the tube and oscillate intermittently to avoid PDA@EVs deposition. After incubation at room temperature for 30 mins, the final product was purified by ultracentrifugation and washed three times with PBS. Radioactive thin-layer chromatography was performed using a gamma detector to assess labeling efficiency. The stability of the radioactive probe was evaluated by incubating 5 μL samples in 200 μL 10% FBS for different times (1, 3, 6, 12, and 24 h) in DMEM high glucose solution at 37 °C. After incubation, the probes were collected by centrifugation and the radioactivity retained on the particles was counted. Silica gel chromatography paper and 0.9% sodium chloride were used as stationary and mobile phases. The 125 I-labeled SPIO-PDA@EVs remains at the origin, while the free 125 I remains at the front with the mobile phase. The radiochemical purity of the 125 I-labeled SPIO-PDA@EVs is expressed as the SPIO-PDA@EVs probe retained at the origin. The radioactive dose to the needle as a percentage of the total radioactive dose.

在本发明的某些优选实施方式中,步骤5)具体包括:PDA@EVs用氨基官能化的超顺磁性氧化铁(SPIO)以20 μg/mL的浓度标记1小时,然后洗涤和超滤以丢弃过量的SPIO颗粒。为了检查SPIO的标记稳定性SPIO-PDA@EVs在37 °C下悬浮在含有10% FBS的DMEM高糖培液中,并在0、1、3、6、12、24和48小时用电感耦合等离子体发射光谱仪(ICP-OES)测量分离的SPIO颗粒。使用1.41 T minispec mq 60 NMR分析仪在37 °C下测定标记EVs(1×109个粒子/mL)的T2弛豫时间。通过将1×109个探针在含有10% FBS的5 mL DMEM高糖培液中在37 °C下共孵育不同时间(0、1、3、6、12、24和48小时),均分三份研究SPIO从探针中的可能释放。孵育后,通过超滤收集探针,并通过ICP-OES测量血清中的游离SPIO。SPIO的稳定性表示为保留在PDA@EVs相对于探针上SPIO的总量。使用DLS测量探针制备过程中每个步骤的标记EV的Zeta电位。为了评估SPIO标记的稳定性、EV、PDA@EVs和SPIO-PDA@EVs的流体动力学直径和表面电荷,在0、1、2、3、4天的10% FBS(v/v)溶液中通过DLS进行检查。In certain preferred embodiments of the present invention, step 5) specifically includes: PDA@EVs are labeled with amino-functionalized superparamagnetic iron oxide (SPIO) at a concentration of 20 μg/mL for 1 hour, followed by washing and ultrafiltration to Discard excess SPIO pellets. To check the labeling stability of SPIO, SPIO-PDA@EVs were suspended in DMEM high glucose culture medium containing 10% FBS at 37 °C and inducted with inductor at 0, 1, 3, 6, 12, 24 and 48 h. Coupled plasma optical emission spectrometry (ICP-OES) measures isolated SPIO particles. The T2 relaxation time of labeled EVs (1 × 10 particles/mL) was measured using a 1.41 T minispec mq 60 NMR analyzer at 37 °C. By co-incubating 1 × 10 probes in 5 mL DMEM high-glucose culture medium containing 10% FBS at 37 °C for different times (0, 1, 3, 6, 12, 24, and 48 hours), all The possible release of SPIO from the probe was studied in triplicate. After incubation, the probe was collected by ultrafiltration and free SPIO in serum was measured by ICP-OES. The stability of SPIO is expressed as the total amount of SPIO retained on PDA@EVs relative to the probe. Use DLS to measure the zeta potential of labeled EVs at each step during probe preparation. To evaluate the stability of SPIO labeling, hydrodynamic diameter and surface charge of EVs, PDA@EVs and SPIO-PDA@EVs in 10% FBS (v/v) solution at 0, 1, 2, 3 and 4 days Check via DLS.

在本发明的某些优选实施方式中,步骤6)具体包括:将50 μg RVG加入EVs悬浮液(300 μL)中,并在室温下反应2-4小时。然后通过超滤纯化RVG修饰的EVs,并用PBS洗涤。荧光强度FITC-RVG-PDA@EVs在纯化之前和之后使用微孔板系统进行定量以评估接枝速率。In certain preferred embodiments of the present invention, step 6) specifically includes: adding 50 μg RVG to the EVs suspension (300 μL), and reacting at room temperature for 2-4 hours. RVG-modified EVs were then purified by ultrafiltration and washed with PBS. The fluorescence intensity of FITC-RVG-PDA@EVs before and after purification was quantified using a microplate system to evaluate the grafting rate.

在本发明的表征外泌体的实施方式中,具体包括:In the embodiment of the present invention, the characterization of exosomes specifically includes:

ICP-OES测量。向已经处理45 mins PDA的外泌体中加入100 μL的SPIO,让其反应5分钟。由此产生的SPIO-PDA@EVs悬浮于含有10% FBS的DMEM高葡萄糖中,并在37°C下孵育1、3、6、12、24和48小时。通过测量SPIO-PDA@EVs在加入KI后、超速离心和去离子水稀释,使用电感耦合等离子体发射光谱仪检测标记稳定性。此外SPIO-PDA@EVs(≈3.8×1012个粒子/mL)和等量的纯SPIO在120 °C下加热,每毫升产物中加入50 μL硝酸消融1小时后进行比较。标记率计算为(SPIO-PDA@EVs中Fe离子浓度/SPIO的离子浓度)* 100%。ICP-OES measurement. Add 100 μL of SPIO to the exosomes that have been treated with PDA for 45 min and let it react for 5 minutes. The resulting SPIO-PDA@EVs were suspended in DMEM high glucose containing 10% FBS and incubated at 37°C for 1, 3, 6, 12, 24, and 48 h. The labeling stability was detected using an inductively coupled plasma optical emission spectrometer by measuring SPIO-PDA@EVs after adding KI, ultracentrifugation and dilution with deionized water. In addition, SPIO-PDA@EVs (≈3.8 × 10 12 particles/mL) and an equal amount of pure SPIO were heated at 120 °C, and 50 μL nitric acid was added to each ml of the product for ablation for 1 hour before comparison. The labeling rate is calculated as (Fe ion concentration in SPIO-PDA@EVs/ion concentration of SPIO) * 100%.

STEM成像。5 μL SPIO-PDA@EVs将其放置在亲水性碳膜上并使其温育2分钟。孵育后的铜网和样品用去离子水洗涤三次,每次在室温下干燥10 s以减少盐沉积。接下来,将4μL磷钨酸加入到样品中,干燥1分钟。为了防止SPIO-PDA@EVs在成像过程中进入极靴,在样品顶部盖上一层碳膜。使用重量百分比(wt%)或原子百分比(at%)扫描进行明场和高角度环形暗场成像(HAADF)成像5-10分钟。能量色散光谱仪-探测器(EDS)映射分析SPIO-PDA@EVs与STEM具有相同铁、碘、氮、碳和氧元素的元素分布STEM imaging. 5 μL SPIO-PDA@EVs were placed on the hydrophilic carbon membrane and allowed to incubate for 2 min. The incubated copper grid and sample were washed three times with deionized water and dried at room temperature for 10 s each time to reduce salt deposition. Next, 4 μL of phosphotungstic acid was added to the sample and dried for 1 min. In order to prevent SPIO-PDA@EVs from entering the pole shoe during imaging, a carbon film was covered on the top of the sample. Perform brightfield and high-angle annular darkfield (HAADF) imaging for 5-10 minutes using weight percent (wt%) or atomic percent (at%) scanning. Energy dispersive spectrometer-detector (EDS) mapping analysis SPIO-PDA@EVs and STEM have the same elemental distribution of iron, iodine, nitrogen, carbon and oxygen elements

在本发明的外泌体成像的实施方式中,具体包括:In the embodiment of exosome imaging of the present invention, it specifically includes:

磁共振成像。使用T2加权自旋回波序列,参数如下:重复时间(TR)=2500 ms,回波时间(TE)=33 ms,视场(FOV)=20×20 mm,矩阵=256×256,切片厚度=700 μm。使用ParaVision 6软件分析MR图像。为每个图像计算在肿瘤周围绘制的感兴趣区域(ROI)内的平均信号强度。相对信号强度增强(rSIE)被定义为注射后大脑中的平均强度与注射前的平均强度之比。在最终的体内成像后,立即处死小鼠,收集大脑并固定在4%多聚甲醛溶液中进行组织学分析。Magnetic resonance imaging. A T2-weighted spin echo sequence was used with the following parameters: repetition time (TR) = 2500 ms, echo time (TE) = 33 ms, field of view (FOV) = 20 × 20 mm, matrix = 256 × 256, slice thickness = 700 μm. MR images were analyzed using ParaVision 6 software. The average signal intensity within a region of interest (ROI) drawn around the tumor was calculated for each image. Relative signal intensity enhancement (rSIE) is defined as the ratio of the average intensity in the brain after injection to the average intensity before injection. After final in vivo imaging, mice were immediately sacrificed, and brains were collected and fixed in 4% paraformaldehyde solution for histological analysis.

SPECT/CT成像。通过不同给药方式(尾静脉、颈动脉和/或鼻腔给药)给小鼠注射125I标记的EVs,并在注射后0.5、6、24和48小时成像,每只小鼠的辐射剂量为300 μCi。125I-PDA@EVs的量通过来自感兴趣区域(ROI)的每体积器官的注射剂量密度来测量。CT图像为小鼠的位置提供了解剖学参考。SPECT图像是在360°C的32个映射获得的(旋转半径=7.6 cm,30秒/映射)。使用InVivoScope对来自SPECT和CT的重建数据进行可视化和共同配准。ROI是在表现出明显放射性的主要器官中绘制的。SPECT/CT imaging. Mice were injected with 125I -labeled EVs via different administration methods (tail vein, carotid artery and/or nasal administration) and imaged at 0.5, 6, 24 and 48 hours after injection. The radiation dose per mouse was 300 μCi. The amount of 125 I-PDA@EVs was measured by the injected dose density per volume of organ from the region of interest (ROI). The CT images provide an anatomical reference for the mouse's position. SPECT images were acquired for 32 mappings at 360°C (radius of rotation = 7.6 cm, 30 sec/mapping). Use InVivoScope to visualize and co-register reconstructed data from SPECT and CT. ROIs were drawn in major organs exhibiting significant radioactivity.

荧光成像。对于体内荧光成像,通过不同给药方式(尾静脉、颈动脉和/或鼻腔给药)给小鼠注射DiR标记的EVs,并在注射后0.5、6、24和48小时使用IVIS系统分别在748和780nm的激发和发射波长下成像。同侧和对侧大脑、心脏、肺、胃、肝、脾、肾和背景的ROI是从包含相同像素数的同等大小的区域中选择的。摄取的定量是通过绘制每个器官的ROI来确定的。去除的脏器放置在冰上并称重并保存至负80℃以验证匀浆荧光情况。简言之,将80-150 mg的每个器官添加到1 mL裂解物缓冲液中,其中1×RIPA缓冲液含有50 mM Tris-HCl、150 mM NaCl、1% Ige-pal(NP40)、0.5% 脱氧胆酸钠、0.1% SDS和1% 蛋白酶抑制剂混合物。然后将样品均化并离心以收集上清液,取100 µL匀浆加入黑色96孔不透明微孔板中,立即测量每个孔中的荧光强度,从而计算每克组织荧光强度。Fluorescence imaging. For in vivo fluorescence imaging, mice were injected with DiR-labeled EVs via different administration methods (tail vein, carotid artery, and/or nasal administration), and the IVIS system was used at 0.5, 6, 24, and 48 hours after injection at 748 and imaged at excitation and emission wavelengths of 780nm. ROIs for ipsilateral and contralateral brain, heart, lung, stomach, liver, spleen, kidney, and background were selected from equally sized regions containing the same number of pixels. Quantification of uptake was determined by plotting the ROI for each organ. The removed organs were placed on ice, weighed, and stored at minus 80°C to verify homogenate fluorescence. Briefly, 80-150 mg of each organ was added to 1 mL of lysate buffer containing 1× RIPA buffer containing 50 mM Tris-HCl, 150 mM NaCl, 1% Ige-Pal (NP40), 0.5 % sodium deoxycholate, 0.1% SDS, and 1% protease inhibitor cocktail. The sample was then homogenized and centrifuged to collect the supernatant, 100 µL of the homogenate was added to a black 96-well opaque microplate, and the fluorescence intensity in each well was immediately measured to calculate the fluorescence intensity per gram of tissue.

在本发明的外泌体治疗的实施方式中,具体包括:成年ICR小鼠进行短暂性大脑中动脉缺血模型,在造模后一天,分别通过静脉、动脉和鼻腔分别注射100 ug外泌体(1.0×1011个颗粒)。在术后0,1,和3天进行神经行为学检测和脑片收集进行免疫荧光染色,鉴定摄取外泌体的神经细胞类型,并进一步明确标记后的外泌体诊疗体对于缺血性卒中后神经元细胞的靶向性和治疗。In the embodiment of the exosome treatment of the present invention, it specifically includes: adult ICR mice are subjected to a transient middle cerebral artery ischemia model, and one day after the modeling, 100 ug of exosomes are injected through the vein, artery and nasal cavity respectively. (1.0×10 11 particles). Neurobehavioral testing and brain slices were collected for immunofluorescence staining at 0, 1, and 3 days after surgery to identify the types of nerve cells that take up exosomes, and to further clarify the role of labeled exosomes in ischemic stroke. Postneuronal cell targeting and therapy.

本发明经过透射电镜和NTA分别检测了正常和包被PDA后外泌体的形态结构和粒径大小,(如图3A-B所示),并发现PDA包被后不影响外泌体中外显子标记CD63、TSG101的表达变化(图3C-D)。STEM成像显示外泌体膜上存在10 nm左右的超顺磁性四氧化三铁的颗粒覆盖(图3E),以便进行磁共振成像显示外泌体的空间分布。通过检测标记后的外泌体随时程Zeta电位和粒径(图3F-G),可以得知我们的铁颗粒标记是成功且稳定的。为了使我们能够进行SPET/CT成像,我们用经典的碘-125标记外泌体,核素成像显示标记率高达90%(图3H)。放射性薄层色谱显示24 h后标记率仍然保持85%左右,表明放射性标记稳定(图3I)。另外,能谱仪mapping显示碘元素和Fe元素聚集在单个外泌体表面,表明标记成功(图3J)。The present invention detected the morphological structure and particle size of normal and PDA-coated exosomes through transmission electron microscopy and NTA respectively (as shown in Figure 3A-B), and found that PDA coating did not affect the expression of exosomes in exosomes. Expression changes of sub-markers CD63 and TSG101 (Figure 3C-D). STEM imaging showed that there was a particle coverage of about 10 nm superparamagnetic iron tetroxide on the exosome membrane (Figure 3E), so that magnetic resonance imaging could show the spatial distribution of exosomes. By detecting the zeta potential and particle size of labeled exosomes over time (Figure 3F-G), we can know that our iron particle labeling is successful and stable. To enable us to perform SPET/CT imaging, we labeled exosomes with the classic iodine-125, and radionuclide imaging showed that the labeling rate was as high as 90% (Figure 3H). Radioactive thin-layer chromatography showed that the labeling rate remained about 85% after 24 h, indicating that the radioactive labeling was stable (Figure 3I). In addition, energy spectrometer mapping showed that iodine and Fe elements were accumulated on the surface of individual exosomes, indicating successful labeling (Figure 3J).

利用多模态成像系统,本发明通过在短暂性缺血卒中小鼠中验证标记的外泌体示踪效果。首先激光散斑脑血流定量证实了tMCAO手术期间小鼠闭塞和再灌注的成功(图4B-C)。为了确定最佳给药途径,卒中小鼠分别通过动脉内(i.a.)、静脉内(i.v.)和鼻内(i.n.)注射100 μg(1×1011个颗粒)外泌体。利用7T磁共振,在造模后一天对小鼠注射前后进行T2加权成像(图4E-F),可以看到动脉内注射30 min后,在梗死区检测到强烈的对比度,但24 h后几乎消失,表明缺血脑中EVs的快速摄取和降解。相比之下,静脉注射和鼻内注射后,大脑摄取量远低于动脉内注射。Using a multimodal imaging system, the present invention verified the tracking effect of labeled exosomes in mice with transient ischemic stroke. First, laser speckle cerebral blood flow quantification confirmed the success of occlusion and reperfusion in mice during tMCAO surgery (Figure 4B-C). To determine the optimal route of administration, stroke mice were injected with 100 μg (1 × 10 11 particles) of exosomes via intraarterial (ia), intravenous (iv), and intranasal (in) respectively. Using 7T magnetic resonance, T2-weighted imaging was performed on mice before and after injection one day after modeling (Figure 4E-F). It can be seen that strong contrast was detected in the infarct area 30 minutes after intra-arterial injection, but almost no contrast was detected after 24 hours. disappeared, indicating rapid uptake and degradation of EVs in the ischemic brain. In contrast, brain uptake after intravenous and intranasal injections was much lower than intraarterial injections.

为了观察和追踪EVs在主要器官和组织中的迁移,本发明用放射性标记的外泌体治疗卒中小鼠,并用SPECT/CT扫描。如图5所示,可以看到检测到EVs在不同给药途径中具有显著的器官靶向性和生物分布特征。SPECT/CT成像结果标明:动脉给药30 min后,标记的外泌体主要出现在肝脏/脾脏和膀胱,少量积累在肺部和心脏,在同侧脑内也有大量累积。24h后,经肾脏系统的清除,仍有大量EVs积聚在肝脏/脾脏中,同时,大脑中的信号显著减少了近50%;48 h后,脑内几乎没有发现EVs,结果表明动脉注射后的EVs,摄取和细胞内化是高效的。相较于动脉注射,静脉注射30 mins后,EVs在肝脏、肾脏和甲状腺强烈积累,但是脑内的信号要低得多。24 h后信号消失,表明静脉注射后EVs在大脑中的初始摄取较低,药代动力学加速更快。与前两种注射方式相比,鼻腔给EVs除甲状腺外,没有迁移到其他器官或组织,6 h后EVs倾向于迁移至胃肠道,并在注射后48 h排出体外。SPET/CT结果显示,相比于静脉和鼻腔注射,动脉注射EVs后6 h,大脑内的核信号分别增加2.4倍和2.0倍。In order to observe and track the migration of EVs in major organs and tissues, the present invention treated stroke mice with radiolabeled exosomes and scanned them with SPECT/CT. As shown in Figure 5, it can be seen that EVs were detected to have significant organ targeting and biodistribution characteristics in different administration routes. SPECT/CT imaging results showed that 30 minutes after arterial administration, labeled exosomes mainly appeared in the liver/spleen and bladder, with a small amount accumulated in the lungs and heart, and a large amount accumulated in the ipsilateral brain. After 24 hours, after being cleared by the renal system, a large number of EVs still accumulated in the liver/spleen. At the same time, the signal in the brain was significantly reduced by nearly 50%; after 48 hours, almost no EVs were found in the brain. The results showed that after arterial injection EVs, uptake and cellular internalization are efficient. Compared with arterial injection, EVs accumulated strongly in the liver, kidney and thyroid 30 min after intravenous injection, but the signal in the brain was much lower. The signal disappeared after 24 h, indicating lower initial uptake of EVs in the brain and faster pharmacokinetic acceleration after intravenous injection. Compared with the first two injection methods, EVs administered intranasally did not migrate to other organs or tissues except the thyroid gland. EVs tended to migrate to the gastrointestinal tract after 6 hours and were excreted 48 hours after injection. SPET/CT results showed that compared with intravenous and nasal injection, 6 h after intraarterial injection of EVs, nuclear signals in the brain increased 2.4 times and 2.0 times respectively.

通过使用荧光成像验证了EVs在不同器官中分布的动力学。将近红外染料DiR插入EVs的膜内,利用小动物活体成像系统进行离体和在体检测。小动物活体成像结果表明(图6A-B),动脉和静脉注射外泌体的前6 h,DIR信号出现在腹部和盆骨的位置,24 h后信号显著降低,而鼻腔注射首先在注射部位信号较高,随后持续下降。离体成像显示了不同的分布特征,动脉和静脉注射主要集中在肝脏、肺部、肾脏和受损脑半球,而鼻腔注射在胃、肺和脾脏强烈出现信号,脑部信号极少。结果与MRI及SPECT/CT成像一致,表明三种注射方式相比,动脉内注射在同侧大脑中产生更强的信号,这意味着动脉内注射具有显著的脑靶向优势。同时,在共聚焦显微镜下对不同器官的冷冻切片进行成像(图6D),以验证EVs的分布。HE染色显示了主要器官的结构,共聚焦显微照片显示,动脉内注射和静脉注射小鼠的肝脏和脾脏中的荧光强度水平相当,但动脉内注射小鼠损伤纹状体中积聚的EVs明显增加,这进一步支持了前述成像结果。不同脏器和组织中DiR荧光信号统计显示(图6E),与静脉注射或鼻内注射相比,动脉内注射后小鼠大脑中EVs的荧光强度显著更高。本发明通过免疫染色,以确定动脉内注射脑中EVs的细胞摄取特性。3D重建图像表明,大多数EV被小胶质细胞(Iba-1+)和神经元(MAP2+)吸收,而不是内皮细胞(CD31+)或星形胶质细胞(GFAP+)吸收。The dynamics of EVs distribution in different organs was verified by using fluorescence imaging. The near-infrared dye DiR is inserted into the membrane of EVs, and small animal in vivo imaging systems are used for in vitro and in vivo detection. In vivo imaging results of small animals show (Figure 6A-B) that in the first 6 hours of arterial and intravenous injection of exosomes, DIR signals appear in the abdomen and pelvis, and the signals decrease significantly after 24 hours, while nasal injection first occurs at the injection site. The signal is high and then continues to decrease. Ex vivo imaging showed different distribution characteristics, with arterial and intravenous injections mainly concentrated in the liver, lungs, kidneys and damaged brain hemispheres, while nasal injections showed strong signals in the stomach, lungs and spleen, with minimal signals in the brain. The results were consistent with MRI and SPECT/CT imaging, indicating that compared with the three injection methods, intra-arterial injection produced stronger signals in the ipsilateral brain, which means that intra-arterial injection has significant brain targeting advantages. At the same time, frozen sections of different organs were imaged under a confocal microscope (Figure 6D) to verify the distribution of EVs. HE staining showed the structure of the major organs, and confocal micrographs showed that the fluorescence intensity levels in the liver and spleen of mice injected intra-arterially and intravenously were similar, but the accumulated EVs in the injured striatum of mice injected intra-arterially were obvious. increased, which further supports the aforementioned imaging results. Statistics of DiR fluorescence signals in different organs and tissues showed (Figure 6E) that compared with intravenous or intranasal injection, the fluorescence intensity of EVs in the mouse brain was significantly higher after intra-arterial injection. The present invention uses immunostaining to determine the cellular uptake characteristics of intra-arterially injected EVs in the brain. 3D reconstructed images showed that most EVs were taken up by microglia (Iba-1 + ) and neurons (MAP2 + ) rather than endothelial cells (CD31 + ) or astrocytes (GFAP + ).

本发明通过多种成像方式监测多功能EVs探针的药代动力学,证明动脉内注射是在全身和器官/组织水平上实现EVs输送到卒中大脑的最大可视化的最佳方式。为了改善EVs被神经元摄取的靶向性,将RVG连接到PDA上便于和神经元表达的乙酰胆碱受体特异性结合。电镜和DLS的检测证明RVG的修饰并不影响外泌体的形态特征。共聚焦3D荧光图显示(图6F),RVG修饰后显著增加了MAP2+神经元摄取率,表明RVG肽成功地竞争了神经元结合,但未能逆转神经元的吞噬作用。为了检查RVG结合是否会影响神经元的生存能力,进行了CCK-8测定,结果发现10 μg/mL EVs孵育6 h不会影响神经元的细胞生存能力。The present invention monitors the pharmacokinetics of a multifunctional EVs probe through multiple imaging modalities, demonstrating that intra-arterial injection is the best way to achieve maximum visualization of EVs delivery to the stroke brain at the systemic and organ/tissue levels. In order to improve the targeting of EVs uptake by neurons, RVG was connected to PDA to facilitate specific binding to acetylcholine receptors expressed by neurons. Electron microscopy and DLS testing proved that RVG modification did not affect the morphological characteristics of exosomes. Confocal 3D fluorescence images showed (Figure 6F) that RVG modification significantly increased the MAP2 + neuronal uptake rate, indicating that the RVG peptide successfully competed for neuronal binding but failed to reverse neuronal phagocytosis. To examine whether RVG binding would affect the viability of neurons, a CCK-8 assay was performed, and it was found that incubation with 10 μg/mL EVs for 6 h did not affect the cell viability of neurons.

为了研究RVG修饰是否可以改善脑内EVs的摄取,本发明通过MR/SPECT/FL成像检查了用RVG修饰的EVs和未经处理的EVs治疗的卒中小鼠。MRI显示(图8A),注射后4.5 h,更多RVG修饰的EVs在缺血脑的梗死区积聚,即使在注射后48 h后,仍有许多分散的低强度点定位在病变脑中。普鲁士蓝染色也显示(图8B-C),SPIO标记的EVs明确定位于大脑梗死核心和梗死周围区域,RVG修饰显著增加了EVs在同侧脑半球的积聚。从SPECT/CT图像中(图8D-E),我们注意到RVG修饰显著改善了注射后0.5 h的脑靶向性。注射后6 h,最初聚集的EVs倾向于在病变脑区内迁移和扩散。离体成像显示RVG修饰确实改善了脑内EVs的靶向性。RVG修饰不仅改善了脑内EVs的摄取,进一步染色表明RVG显著增加了纹状体梗塞区神经元对EV的摄取。缺血性卒中后,缺血半影中的大量神经元在数小时内发生凋亡,并持续数天。考虑到卒中的时间限制治疗窗口,评估EVs给药的适当时间点至关重要。我们在卒中后2 h注射外泌体(图9A-D),与PBS组和M2-EVs对比,发现3 d后FJ-B+ 和 TUNEL+ 细胞数显著减少,RVG-M2-EVs治疗进一步减少了神经元变性和凋亡。WB结果显示(图9E),RVG修饰后的外泌体治疗降低了裂解的胱天蛋白酶-3(cleaved-caspase3)的表达,并且检测到较低的神经严重程度评分和更好的旋转棒试验表现。结果表明RVG修饰增强了EVs对缺血性卒中的神经保护作用。To investigate whether RVG modification can improve the uptake of EVs in the brain, we examined stroke mice treated with RVG-modified EVs and untreated EVs by MR/SPECT/FL imaging. MRI showed (Figure 8A) that more RVG-modified EVs accumulated in the infarct area of the ischemic brain at 4.5 h after injection, and even after 48 h after injection, there were still many scattered low-intensity spots localized in the diseased brain. Prussian blue staining also showed (Figure 8B-C) that SPIO-labeled EVs were clearly located in the brain infarct core and peri-infarct areas, and RVG modification significantly increased the accumulation of EVs in the ipsilateral cerebral hemisphere. From the SPECT/CT images (Figure 8D-E), we noticed that RVG modification significantly improved brain targeting 0.5 h after injection. At 6 h after injection, the initially accumulated EVs tended to migrate and spread within the diseased brain area. Ex vivo imaging showed that RVG modification indeed improved the targeting of EVs in the brain. RVG modification not only improved the uptake of EVs in the brain, but further staining showed that RVG significantly increased the uptake of EVs by neurons in the infarcted area of the striatum. After ischemic stroke, a large number of neurons in the ischemic penumbra undergo apoptosis within hours and persist for days. Given the time-limited therapeutic window for stroke, it is critical to evaluate the appropriate time point for EVs administration. We injected exosomes 2 h after stroke (Figure 9A-D). Compared with the PBS group and M2-EVs, we found that the number of FJ-B + and TUNEL + cells was significantly reduced after 3 d, and was further reduced after RVG-M2-EVs treatment. neuronal degeneration and apoptosis. WB results showed (Figure 9E) that RVG-modified exosome treatment reduced the expression of cleaved-caspase3 (cleaved-caspase3), and lower neurological severity scores and better rotarod test were detected Performance. The results indicate that RVG modification enhances the neuroprotective effect of EVs on ischemic stroke.

为了研究M2 EVs介导的神经保护作用的潜在机制,本发明进行了EVs-miRNA测序(图10)。主成分分析表明除RVG-M2-EVs一个样本被排除在外,其他样本都分散良好。M0-EVs和M2-EVs表现出不同的基因表达谱,而RVG-M2-EVs和M2 EV具有相似的基因组成,表明RVG-PDA 标记策略不会改变 EV miRNA 含量。与M0 EVs相比,在M2 EVs中鉴定出35个差异表达的miRNA,其中21个miRNA上调,14个miRNA下调。进一步的KEGG分析表明,一些代谢信号传导、突触、血管功能和细胞凋亡相关途径在M2 EVs中富集。细胞凋亡相关的miRNA,包括miR-423-3p、miR-7688-5p、miR-106b-3p、miR-532-5p、miR-151-3p和miR-146a-5p等尤为明显。这些结果表明,富含M2 EVs的抗凋亡相关miRNA对急性缺血性卒中小鼠具有神经保护作用。In order to study the potential mechanism of M2 EVs-mediated neuroprotection, the present invention performed EVs-miRNA sequencing (Figure 10). Principal component analysis showed that except for one sample of RVG-M2-EVs, which was excluded, the other samples were well dispersed. M0-EVs and M2-EVs exhibit different gene expression profiles, while RVG-M2-EVs and M2 EVs have similar genetic composition, indicating that the RVG-PDA labeling strategy does not change EV miRNA content. Compared with M0 EVs, 35 differentially expressed miRNAs were identified in M2 EVs, of which 21 were up-regulated and 14 were down-regulated. Further KEGG analysis showed that some metabolic signaling, synapse, vascular function and apoptosis-related pathways were enriched in M2 EVs. Apoptosis-related miRNAs, including miR-423-3p, miR-7688-5p, miR-106b-3p, miR-532-5p, miR-151-3p and miR-146a-5p, are particularly evident. These results indicate that anti-apoptosis-related miRNAs enriched in M2 EVs have neuroprotective effects in mice with acute ischemic stroke.

尽管本发明的实施方案已公开如上,但其并不仅仅限于说明书和实施方式中所列运用,它完全可以被适用于各种适合本发明的领域,对于熟悉本领域的人员而言,可容易地实现另外的修改,因此在不背离权利要求及等同范围所限定的一般概念下,本发明并不限于特定的细节和这里示出与描述的图例。Although the embodiments of the present invention have been disclosed above, they are not limited to the applications listed in the description and embodiments. They can be applied to various fields suitable for the present invention. For those familiar with the art, they can easily Additional modifications may be made, and the invention is therefore not limited to the specific details and illustrations shown and described herein without departing from the general concept defined by the claims and equivalent scope.

Claims (10)

1.基于M2型小胶质细胞的外泌体诊疗剂,其特征在于,包括基于M2型小胶质细胞的外泌体、外泌体表面修饰的荧光染料、多巴胺或多巴胺衍生物、放射性核素、磁共振成像离子、靶向神经元的配体。1. Exosome therapeutic agents based on M2 microglia, characterized by including exosomes based on M2 microglia, fluorescent dyes modified on the exosome surface, dopamine or dopamine derivatives, radioactive nuclei ligands, magnetic resonance imaging ions, and neuron-targeting ligands. 2.如权利要求1所述的基于M2型小胶质细胞的外泌体诊疗剂,其特征在于,2. The exosome therapeutic agent based on M2 type microglia according to claim 1, characterized in that, 所述外泌体来源为M2型小胶质细胞;The source of exosomes is M2 type microglia; 所述荧光染料选自DiR、Cy5.5、Cy7中的一种;The fluorescent dye is selected from one of DiR, Cy5.5, and Cy7; 所述多巴胺衍生物选自盐酸多巴胺;The dopamine derivative is selected from dopamine hydrochloride; 所述放射性核素选自碘-125、碘-131、镓-68、铜-64中的一种;The radioactive nuclide is selected from one of iodine-125, iodine-131, gallium-68, and copper-64; 所述磁共振成像离子选自Gd3+、Mn2+、Fe3+、Cu2+、Ni3+中的一种或多种;The magnetic resonance imaging ions are selected from one or more of Gd 3+ , Mn 2+ , Fe 3+ , Cu 2+ , and Ni 3+ ; 所述靶向神经元的配体选自RVG。The neuron-targeting ligand is selected from RVG. 3.如权利要求1或2所述的基于M2型小胶质细胞的外泌体诊疗剂的制备方法,其特征在于,包括以下步骤:3. The preparation method of exosome therapeutic agent based on M2 type microglia according to claim 1 or 2, characterized in that it includes the following steps: 1)体外培养未分化的M0型细胞,通过加入极化因子促进其向M2型细胞分化,并分泌外泌体;1) Cultivate undifferentiated M0 cells in vitro, promote their differentiation into M2 cells by adding polarizing factors, and secrete exosomes; 2)通过离心分离步骤1)的产物,收集并纯化得到M2型细胞来源的外泌体;2) Separate the product of step 1) by centrifugation, collect and purify exosomes derived from M2 cells; 3)将步骤2)得到的外泌体全部或部分进行经荧光染料染色标记;3) All or part of the exosomes obtained in step 2) are stained and labeled with fluorescent dyes; 4)将步骤3)得到的经过荧光染料染色标记的外泌体全部或部分表面上包覆多巴胺或多巴胺衍生物层;4) Coat all or part of the surface of the fluorescent dye-labeled exosomes obtained in step 3) with a dopamine or dopamine derivative layer; 5)将放射性核素、磁共振成像离子与步骤4)的产物混合;5) Mix radionuclides, magnetic resonance imaging ions and the product of step 4); 6) 将步骤5)的产物表面接枝靶向神经元的配体,获得所述外泌体。6) Graft the surface of the product of step 5) with a neuron-targeting ligand to obtain the exosomes. 4.如权利要求1所述的基于M2型小胶质细胞的外泌体诊疗剂的制备方法,其特征在于,所述步骤1)中,M0型细胞可选为小胶质细胞,极化因子可选为IL4,以总培养体系的总体积为准,控制培养体积为15 mL ,极化因子加入的浓度为20 ng/mL,极化培养时间为36-48小时,培养结束后,收集刺激后M2型小胶质细胞的上清,保存-20 ℃待提取外泌体。4. The preparation method of exosome therapeutic agent based on M2 type microglia as claimed in claim 1, characterized in that in step 1), M0 type cells can be selected as microglia, polarized The factor can be selected as IL4, based on the total volume of the total culture system. The control culture volume is 15 mL. The concentration of the polarization factor added is 20 ng/mL. The polarization culture time is 36-48 hours. After the culture is completed, collect The supernatant of M2 type microglia after stimulation was stored at -20°C until exosomes were extracted. 5.如权利要求4所述的基于M2型小胶质细胞的外泌体诊疗剂的制备方法,其特征在于,所述步骤2)具体为:5. The preparation method of exosome therapeutic agent based on M2 type microglia according to claim 4, characterized in that the step 2) is specifically: 收集刺激后的M2小胶质细胞的条件培养基超速离心,条件为4 °C下以300 g连续离心10分钟、2000 g连续离心15分钟去除死细胞、10000 g连续离心30分钟去除细胞碎片,以及以100000 g连续超速离心70分钟沉淀外泌体,将EV用100000g的PBS洗涤一次70分钟,并悬浮以进行进一步表征;Collect the conditioned medium of stimulated M2 microglia and perform ultracentrifugation at 4°C at 300g for 10 minutes, 2000g for 15 minutes to remove dead cells, and 10000g for 30 minutes to remove cell debris. And the exosomes were precipitated by continuous ultracentrifugation at 100,000 g for 70 minutes, and the EVs were washed once with 100,000 g of PBS for 70 minutes and suspended for further characterization; 收集所得外泌体采用120 kV的透射电子显微镜鉴定结构,纳米粒子跟踪分析测量外泌体的直径和颗粒,BCA法测定外泌体的含量以及蛋白质免疫印迹分析外泌体标记物CD63、TSG101。The structure of the collected exosomes was identified using 120 kV transmission electron microscopy, nanoparticle tracking analysis was used to measure the diameter and particles of exosomes, BCA method was used to determine the content of exosomes, and Western blotting was used to analyze exosome markers CD63 and TSG101. 6.如权利要求5所述的基于M2型小胶质细胞的外泌体诊疗剂的制备方法,其特征在于,所述步骤3)具体为使用近红外荧光染料DiR和红色荧光染料PKH26修饰外泌体;6. The preparation method of exosome therapeutic agent based on M2 type microglia as claimed in claim 5, characterized in that step 3) specifically involves using near-infrared fluorescent dye DiR and red fluorescent dye PKH26 to modify the exosomes. secretion body; 对于DiR的标记过程,外泌体和1 μL稀释好的DiR溶液在37 ℃中孵育5 mins;For the DiR labeling process, exosomes and 1 μL of diluted DiR solution were incubated at 37°C for 5 mins; 对于体内外泌体摄取实验,1:500稀释后的PKH26与外泌体在室温下孵育5 mins;For in vivo exosome uptake experiments, PKH26 diluted 1:500 was incubated with exosomes at room temperature for 5 min; 全程采用不含EVs的血清;EVs-free serum is used throughout the process; 将标记好的外泌体在100000g的PBS中洗涤1小时以洗掉非特异标记,并将外泌体可以在PBS中重悬待用。The labeled exosomes were washed in 100,000 g of PBS for 1 hour to wash away non-specific labeling, and the exosomes were resuspended in PBS for later use. 7.如权利要求6所述的基于M2型小胶质细胞的外泌体诊疗剂的制备方法,其特征在于,所述步骤4)具体包括:7. The method for preparing exosome therapeutic agents based on M2 microglia according to claim 6, characterized in that said step 4) specifically includes: 将200 μL EVs与PBS按照1:1混合,将溶于8 mL Tris缓冲液(pH=8.5)中的1 mg PDA加入溶液中反应10 mins,使多巴胺的浓度为0.1 mg/mL;Mix 200 μL EVs and PBS at a ratio of 1:1, add 1 mg PDA dissolved in 8 mL Tris buffer (pH=8.5) into the solution and react for 10 mins to make the concentration of dopamine 0.1 mg/mL; 之后,将包裹有PDA的EVs悬浮在透析管中,通过10000 g超滤分离8分钟,然后用PBS洗涤三次,最终产物中包裹多巴胺的外泌体浓度约为1.2×1011个粒子/mL。Afterwards, the PDA-coated EVs were suspended in a dialysis tube, separated by 10000 g ultrafiltration for 8 minutes, and then washed three times with PBS. The concentration of dopamine-coated exosomes in the final product was approximately 1.2 × 10 11 particles/mL. 8.如权利要求7所述的基于M2型小胶质细胞的外泌体诊疗剂的制备方法,其特征在于,8. The preparation method of exosome therapeutic agent based on M2 type microglia according to claim 7, characterized in that, 所述步骤5)中放射性碘-125标记是通过经典的Iodogen方法实现的,将PDA@EVs悬浮液(0.5 mL,≈1.2×1011个粒子/mL)加入底部涂有20 μg Iodogen的玻璃管中。将新制备的Na125I溶液(500 μCi,18.5 MBq)加入管中,间歇振荡以避免PDA@EVs沉积,在室温下孵育30mins后,通过超离心纯化最终产物,并用PBS洗涤三次;Radioactive iodine-125 labeling in step 5) is achieved by the classic Iodogen method. PDA@EVs suspension (0.5 mL, ≈1.2×10 11 particles/mL) is added to a glass tube coated with 20 μg Iodogen at the bottom. middle. Add the newly prepared Na 125 I solution (500 μCi, 18.5 MBq) into the tube, oscillate intermittently to avoid PDA@EVs deposition, and after incubating at room temperature for 30 mins, purify the final product by ultracentrifugation and wash three times with PBS; PDA@EVs用氨基官能化的超顺磁性氧化铁(SPIO)以20 μg/mL的浓度标记1小时,然后洗涤和超滤以丢弃过量的SPIO颗粒。PDA@EVs were labeled with amino-functionalized superparamagnetic iron oxide (SPIO) at a concentration of 20 μg/mL for 1 h, followed by washing and ultrafiltration to discard excess SPIO particles. 9.如权利要求8所述的基于M2型小胶质细胞的外泌体诊疗剂的制备方法,其特征在于,所述步骤6)具体包括:将50 μg RVG加入EVs悬浮液300 μL中,并在室温下反应2-4小时。然后通过超滤纯化RVG修饰的EVs,并用PBS洗涤。9. The preparation method of exosome therapeutic agent based on M2 type microglia as claimed in claim 8, characterized in that the step 6) specifically includes: adding 50 μg RVG to 300 μL of EVs suspension, And react at room temperature for 2-4 hours. RVG-modified EVs were then purified by ultrafiltration and washed with PBS. 10.基于M2型小胶质细胞的外泌体诊疗剂在制备医学成像探针中的应用,其特征在于,所述医学成像选自核医学成像、磁共振成像、CT成像中的任一种或多种。10. Application of exosome therapeutic agents based on M2 microglia in the preparation of medical imaging probes, characterized in that the medical imaging is selected from any one of nuclear medicine imaging, magnetic resonance imaging, and CT imaging. or more.
CN202310776270.0A 2023-06-28 2023-06-28 Exosome diagnosis and treatment agent based on M2 microglial cells, preparation method and application thereof in preparation of medical imaging probes Pending CN116763945A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202310776270.0A CN116763945A (en) 2023-06-28 2023-06-28 Exosome diagnosis and treatment agent based on M2 microglial cells, preparation method and application thereof in preparation of medical imaging probes

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310776270.0A CN116763945A (en) 2023-06-28 2023-06-28 Exosome diagnosis and treatment agent based on M2 microglial cells, preparation method and application thereof in preparation of medical imaging probes

Publications (1)

Publication Number Publication Date
CN116763945A true CN116763945A (en) 2023-09-19

Family

ID=87987683

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310776270.0A Pending CN116763945A (en) 2023-06-28 2023-06-28 Exosome diagnosis and treatment agent based on M2 microglial cells, preparation method and application thereof in preparation of medical imaging probes

Country Status (1)

Country Link
CN (1) CN116763945A (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101475461A (en) * 2009-02-09 2009-07-08 山东省医学科学院放射医学研究所 Iodogen method for preparation of hypericin radioactive iodine marker
US20190093105A1 (en) * 2015-10-01 2019-03-28 University Of Ottawa Exosome packaging of nucleic acids
CN113337468A (en) * 2021-05-27 2021-09-03 上海市伤骨科研究所 Dual-functional exosome, preparation method thereof and application thereof in cerebral apoplexy repair
CN113679850A (en) * 2020-05-19 2021-11-23 首都医科大学附属北京天坛医院 A kind of exosomes modified by targeting and loaded with drugs and preparation method and application thereof
US20210369858A1 (en) * 2017-08-16 2021-12-02 Board Of Regents, The University Of Texas System Use of exosomes for targeted delivery of therapeutic agents

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101475461A (en) * 2009-02-09 2009-07-08 山东省医学科学院放射医学研究所 Iodogen method for preparation of hypericin radioactive iodine marker
US20190093105A1 (en) * 2015-10-01 2019-03-28 University Of Ottawa Exosome packaging of nucleic acids
US20210369858A1 (en) * 2017-08-16 2021-12-02 Board Of Regents, The University Of Texas System Use of exosomes for targeted delivery of therapeutic agents
CN113679850A (en) * 2020-05-19 2021-11-23 首都医科大学附属北京天坛医院 A kind of exosomes modified by targeting and loaded with drugs and preparation method and application thereof
CN113337468A (en) * 2021-05-27 2021-09-03 上海市伤骨科研究所 Dual-functional exosome, preparation method thereof and application thereof in cerebral apoplexy repair

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
JIE WANG等: "Designer exosomes enabling tumor targeted efficient chemo/gene/photothermal therapy", BIOMATERIALS, vol. 276, 31 July 2021 (2021-07-31), pages 1 - 10 *
刘朝阳等: "外泌体标记方法的研究现状", 现代免疫学, vol. 40, no. 3, 31 December 2020 (2020-12-31), pages 240 - 244 *

Similar Documents

Publication Publication Date Title
Meng et al. Functional metal–organic framework-based nanocarriers for accurate magnetic resonance imaging and effective eradication of breast tumor and lung metastasis
Zhang et al. Noninvasive monitoring of orthotopic glioblastoma therapy response using RGD-conjugated iron oxide nanoparticles
Abakumov et al. VEGF-targeted magnetic nanoparticles for MRI visualization of brain tumor
McConnell et al. Ferumoxytol nanoparticle uptake in brain during acute neuroinflammation is cell-specific
JP6174603B2 (en) Contrast agent for T2 * -weighted magnetic resonance imaging (MRI)
Kim et al. Target-switchable Gd (III)-DOTA/protein cage nanoparticle conjugates with multiple targeting affibody molecules as target selective T1 contrast agents for high-field MRI
EP2671841A2 (en) Nanoparticle coated with ligand introduced with long hydrophobic chain and method for preparing same
C Manley et al. Tracking stem cells for cellular therapy in stroke
US20060204443A1 (en) Methods for tumor treatment using dendrimer conjugates
WO2024165076A1 (en) Multi-modal molecular imaging nanoprobe for early warning and dynamic monitoring of atherosclerotic plaques and use thereof
Yang et al. Quantitative visualization of myocardial ischemia-reperfusion-induced cardiac lesions via ferroptosis magnetic particle imaging
US10117956B2 (en) Radiolabeled active targeting pharmaceutical composition and the use thereof
Stojanov et al. Imaging of cells and nanoparticles: implications for drug delivery to the brain
CN104225630A (en) Multi-mode self-assembly nanoprobe suitable for MRI (magnetic resonance imaging)/PA (optical activation) and other imaging
US20140228552A1 (en) Mri contrast enhancing agent
Lahooti et al. Preliminary studies of 68Ga-NODA-USPION-BBN as a dual-modality contrast agent for use in positron emission tomography/magnetic resonance imaging
CN102350002A (en) Glioma-targeting molecule magnetic resonance contrast agent as well as preparation method and application thereof
Re et al. Nanoparticles for neuroimaging
CN106880846A (en) A kind of cancer target multifunctional nano delivery system and preparation method and purposes
Ding et al. Sensitive photoacoustic/magnetic resonance dual imaging probe for detection of malignant tumors
CN116763945A (en) Exosome diagnosis and treatment agent based on M2 microglial cells, preparation method and application thereof in preparation of medical imaging probes
Wu et al. The feasibility of targeted magnetic iron oxide nanoagent for noninvasive IgA nephropathy diagnosis
KR101386715B1 (en) Advanced biocompatible, non-polymeric surface modified, iron oxide nanoparticles with application of optimal amount of gluconic acid and a composition for diagnosing and treating cancer comprising the same
Su et al. In vivo magnetic resonance and fluorescence dual-modality imaging of tumor angiogenesis in rats using GEBP11 peptide targeted magnetic nanoparticles
Fadón-Padilla et al. Magnetic nanoradiotracers for targeted neutrophil detection in pulmonary arterial hypertension

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