留言板

尊敬的读者、作者、审稿人, 关于本刊的投稿、审稿、编辑和出版的任何问题, 您可以本页添加留言。我们将尽快给您答复。谢谢您的支持!

姓名
邮箱
手机号码
标题
留言内容
验证码

细菌外囊泡在疾病发生发展中的作用机制及应用前景

罗晓霞 覃思华 王晖迪 周宏伟 何彦

罗晓霞, 覃思华, 王晖迪, 周宏伟, 何彦. 细菌外囊泡在疾病发生发展中的作用机制及应用前景[J]. 协和医学杂志, 2023, 14(5): 915-924. doi: 10.12290/xhyxzz.2023-0260
引用本文: 罗晓霞, 覃思华, 王晖迪, 周宏伟, 何彦. 细菌外囊泡在疾病发生发展中的作用机制及应用前景[J]. 协和医学杂志, 2023, 14(5): 915-924. doi: 10.12290/xhyxzz.2023-0260
LUO Xiaoxia, QIN Sihua, WANG Huidi, ZHOU Hongwei, HE Yan. Mechanism of Bacterial Extracellular Vesicles in Disease Development and Prospects for Application[J]. Medical Journal of Peking Union Medical College Hospital, 2023, 14(5): 915-924. doi: 10.12290/xhyxzz.2023-0260
Citation: LUO Xiaoxia, QIN Sihua, WANG Huidi, ZHOU Hongwei, HE Yan. Mechanism of Bacterial Extracellular Vesicles in Disease Development and Prospects for Application[J]. Medical Journal of Peking Union Medical College Hospital, 2023, 14(5): 915-924. doi: 10.12290/xhyxzz.2023-0260

细菌外囊泡在疾病发生发展中的作用机制及应用前景

doi: 10.12290/xhyxzz.2023-0260
基金项目: 

国家自然科学基金 82272391

国家自然科学基金 82022044

详细信息
    通讯作者:

    何彦, E-mail: bioyanhe@gmail.com

    罗晓霞、覃思华对本文同等贡献

    罗晓霞、覃思华对本文同等贡献

  • 中图分类号: R446.5;R456;R378

Mechanism of Bacterial Extracellular Vesicles in Disease Development and Prospects for Application

Funds: 

National Natural Science Foundation of China 82272391

National Natural Science Foundation of China 82022044

More Information
  • 摘要: 细菌外囊泡(bacterial extracellular vesicles,BEVs)是细菌衍生的细胞外囊泡。作为细菌间或细菌-宿主间相互作用的关键新媒介,BEVs是一把“双刃剑”:一方面在宿主多种病理过程,包括肠道炎性疾病、中枢神经系统相关疾病、肝脏疾病、代谢性疾病、自身免疫性疾病及肿瘤中发挥“致病”作用;另一方面作为潜在生物标志物、疫苗、抗肿瘤制剂等发挥“治病”作用,为疾病的诊治打开了新思路。但关于BEVs的研究目前尚处于起步阶段,在囊泡分离技术、疾病诊断特异性、囊泡储运优化等方面仍面临挑战,未来需设计严谨的临床研究以证实其诊断和治疗价值。
    作者贡献:罗晓霞、覃思华负责文献检索及论文撰写;王晖迪负责论文构思及修订;周宏伟、何彦负责选题设计及论文审校。
    利益冲突:所有作者均声明不存在利益冲突
  • 图  1  细菌外囊泡形成过程示意图

    表  1  BEVs作为潜在生物标志物的最新研究

    第一作者
    (发表时间)
    BEVs
    来源
    疾病 研究对象
    (实验组/对照组)
    研究方法 生物标志物 诊断模型 标志物
    类型
    Dey[43] (2019年) 血清 葡萄球菌菌血症 21例幸存者/23例因菌血症死亡者 对感染金黄色葡萄球菌菌血症幸存者和死亡者血清中的BEVs进行分离 BEVs含量 301相对荧光单位:特异度90%/灵敏度78% 预警标志物
    Wei[44] (2019年) 粪便 AD 9例AD患者/9名健康者 对来自AD患者和健康者的BEVs进行差异代谢物分析 4-半醛/PGE2/20-羟基-二十碳四烯酸/菌素/L -岩藻吡喃糖/吲哚-3-乙酸/4-甲基伞形酮/胆碱/咪唑-4-醋酸/ L-天冬氨酸/富马酸/十三酸 4-半醛/PGE2/20-羟基-二十碳四烯酸/菌素/L-岩藻吡喃糖/吲哚-3-乙酸/4-甲基伞形酮/胆碱:各自AUC>0.900咪唑-4-醋酸/ L-天冬氨酸/富马酸/十三酸:各自AUC>0.800 预测和诊断标志物
    Yang[45](2020年) 血清 脑肿瘤 152例脑肿瘤患者/198名健康者 对BEVs进行16S rRNA基因测序 微生物组成 - 诊断标志物
    Kim[46](2020年) 血清 卵巢癌 166例卵巢癌患者/76例良性肿瘤患者 对BEVs进行宏基因测序,构建卵巢癌与良性卵巢肿瘤的诊断模型 BEVs宏基因组图谱 年龄+ CA-125 +不动杆菌相对丰度:AUC=0.846 鉴别标志物
    Kim[47](2020年) 粪便 结直肠癌 32例结直肠癌患者/40名健康者 联合BEVs的宏基因组学数据和代谢组学数据构建诊断结直肠癌的模型 两种细菌(Collinsella、Solanum melongena)/两种代谢物(leucine、oxalic acid) 宏基因组学+代谢组学:AUC=1 诊断标志物
    Yang[48](2020年) 血清 特发性皮炎 24例特发性皮炎患者/49名健康者 16S rDNA宏基因组分析 BEVs细菌类群 血清BEVs生物标志物(LDA评分>3):AUC=1 诊断标志物
    Yang[49](2020年) 血清 哮喘、COPD、肺癌 239例哮喘患者、205例COPD患者、324例肺癌患者/88名健康者 16S rDNA宏基因组分析,基于ELISA的抗BEVs抗体滴度分析作为肺部疾病的诊断工具 BEVs细菌类群 哮喘:AUC=0.78; COPD:AUC=0.79; 肺癌:AUC=0.80 BEVs抗体滴度的肺部疾病诊断工具
    Han[50](2021年) 唾液 牙周炎 7例牙龈炎患者、8例牙周炎患者/7名健康者 对健康者、牙龈炎和牙周炎患者唾液小细胞BEVs的DNA表观遗传模式、LPS阳性OMVs群体和特异性牙周病原体来源的OMVs进行分析 具核梭杆菌OMVs 具核梭杆菌OMVs:AUC=0.94 鉴别牙龈炎和牙周炎的标志物
    Yoon[51](2023年) 尿液 结直肠癌 91例结直肠癌患者/116名健康者 对BEVs进行16S rRNA基因测序,分析两组间的差异微生物群 微生物组成 - 诊断标志物
    BEVs: 细菌外囊泡;AUC:受试者工作特征曲线下面积;AD: 阿尔茨海默病;CA-125:糖类抗原-125;LDA: 线性判别分析;COPD: 慢性阻塞性肺疾病;LPS: 脂多糖;OMVs: 外膜囊泡;-:未描述
    下载: 导出CSV

    表  2  益生菌细胞外囊泡在疾病治疗中可能发挥的作用

    第一作者(发表时间) 微生物名称 疾病 研究内容 作用
    Ñahui Palomino[56](2019年) 卷曲乳杆菌BC3(Lactobacillus crispatus) HIV感染 介导病毒-细胞相互作用的包膜蛋白暴露减少,减少HIV-1对宿主细胞的附着 抑制病原菌黏附
    加氏乳杆菌BC12(Lactobacillus gasseri)
    Kim[57](2018年) 植物乳杆菌APsulloc 331261(Lactobacillus plantarum) 特发性皮炎 诱导抗炎细胞因子IL-10的分泌及免疫调节细胞因子IL-1β和GM-CSF的分泌; 诱导单核细胞向巨噬细胞转变及巨噬细胞向M2b状态极化 抗炎作用/免疫调节
    Tong[58](2021年) 鼠李糖乳杆菌GG(Lacticaseibacillus rhamnosus) 结肠炎 抑制结肠组织TLR4-NF-κB-NLRP3轴的激活; 降低TNF-α、IL-1β、IL-2和IL-6水平 抗炎作用/免疫调节
    Hu[59](2021年) 罗伊氏乳杆菌BBC3(Lactobacillus reuteri) 结肠炎 降低TNF-α、IL-1β和IL-6的基因表达,增强IL-10和TGF-β的基因表达; 激活巨噬细胞,抑制Th1和Th17介导的炎症反应 抗炎作用/免疫调节
    Lee[60](2022年) 乳链球菌(Lactococcus lactis) 过敏性哮喘 气道高反应性、嗜酸性粒细胞数量、细胞因子分泌和黏液产生显著降低; 刺激DC产生IL-12,诱导Th2转变Th1,降低Ig4 免疫调节
    Croatti[61](2022年) 乳杆菌(lactobacilli) 阴道感染 支持有益物种的定植及防止大肠杆菌、金黄色葡萄球菌、无乳链球菌和粪肠球菌等条件致病菌的黏附 抑制病原菌
    Choi[62](2022年) 嗜黏蛋白阿克曼氏菌ATCC BA 835(Akkermansia muciniphil) 抑郁 恢复海马中MeCP2、Sirt1及神经营养因子的表达 抗抑郁作用
    枯草芽孢杆菌(Bacillus subtilis)
    Ye[63](2023年) 植物乳杆菌(Lactobacillus plantarum) 肝损伤 降低肝细胞氧化损伤、改善脂质代谢、调节肠道菌群 -
    HIV:人类免疫缺陷病毒;IL: 白细胞介素;GM-CSF:粒细胞巨噬细胞刺激因子;TLR4:Toll样受体4;TNF-α:肿瘤坏死因子α;TGF-β: 转化生长因子β;Th:辅助性T细胞;DC:树突状细胞;MeCP2:甲基CpG结合蛋白2;Sirt1:NAD依赖的脱乙酰化酶;-:不作分类
    下载: 导出CSV
  • [1] Toyofuku M, Nomura N, Eberl L. Types and origins of bacterial membrane vesicles[J]. Nat Rev Microbiol, 2019, 17: 13-24. doi:  10.1038/s41579-018-0112-2
    [2] Díaz-Garrido N, Badia J, Baldomà L. Microbiota-derived extracellular vesicles in interkingdom communication in the gut[J]. J Extracell Vesicles, 2021, 10: e12161. doi:  10.1002/jev2.12161
    [3] Toyofuku M, Schild S, Kaparakis-Liaskos M, et al. Composition and functions of bacterial membrane vesicles[J]. Nat Rev Microbiol, 2023, 21: 415-430. doi:  10.1038/s41579-023-00875-5
    [4] Hendrix A, De Wever O. Systemically circulating bacterial extracellular vesicles: origin, fate, and function[J]. Trends Microbiol, 2022, 30: 213-216. doi:  10.1016/j.tim.2021.12.012
    [5] Toyofuku M, Cárcamo-Oyarce G, Yamamoto T, et al. Prophage-triggered membrane vesicle formation through peptidoglycan damage in Bacillus subtilis[J]. Nat Commun, 2017, 8: 481. doi:  10.1038/s41467-017-00492-w
    [6] Xie J, Li Q, Haesebrouck F, et al. The tremendous biomedical potential of bacterial extracellular vesicles[J]. Trends Biotechnol, 2022, 40: 1173-1194. doi:  10.1016/j.tibtech.2022.03.005
    [7] Devos S, Van Putte W, Vitse J, et al. Membrane vesicle secretion and prophage induction in multidrug-resistant Stenotrophomonas maltophilia in response to ciprofloxacin stress[J]. Environ Microbiol, 2017, 19: 3930-3937. doi:  10.1111/1462-2920.13793
    [8] Brown L, Wolf JM, Prados-Rosales R, et al. Through the wall: extracellular vesicles in Gram-positive bacteria, mycobacteria and fungi[J]. Nat Rev Microbiol, 2015, 13: 620-630. doi:  10.1038/nrmicro3480
    [9] Lee EY, Choi DS, Kim KP, et al. Proteomics in gram-negative bacterial outer membrane vesicles[J]. Mass Spectrom Rev, 2008, 27: 535-555. doi:  10.1002/mas.20175
    [10] Zakharzhevskaya NB, Vanyushkina AA, Altukhov IA, et al. Outer membrane vesicles secreted by pathogenic and nonpathogenic Bacteroides fragilis represent different metabolic activities[J]. Sci Rep, 2017, 7: 5008. doi:  10.1038/s41598-017-05264-6
    [11] Kaplan GG. The global burden of IBD: from 2015 to 2025[J]. Nat Rev Gastroenterol Hepatol, 2015, 12: 720-727. doi:  10.1038/nrgastro.2015.150
    [12] Jones E, Stentz R, Telatin A, et al. The Origin of Plasma-Derived Bacterial Extracellular Vesicles in Healthy Individuals and Patients with Inflammatory Bowel Disease: A Pilot Study[J]. Genes (Basel), 2021, 12: 1636. doi:  10.3390/genes12101636
    [13] Liu L, Liang L, Yang C, et al. Extracellular vesicles of Fusobacterium nucleatum compromise intestinal barrier through targeting RIPK1-mediated cell death pathway[J]. Gut Microbes, 2021, 13: 1-20.
    [14] Engevik MA, Danhof HA, Ruan W, et al. Fusobacterium nucleatum Secretes Outer Membrane Vesicles and Promotes Intestinal Inflammation[J]. mBio, 2021, 12: e02706-20.
    [15] Kaparakis M, Turnbull L, Carneiro L, et al. Bacterial membrane vesicles deliver peptidoglycan to NOD1 in epithelial cells[J]. Cell Microbiol, 2010, 12: 372-385. doi:  10.1111/j.1462-5822.2009.01404.x
    [16] Patten DA, Hussein E, Davies SP, et al. Commensal-derived OMVs elicit a mild proinflammatory response in intestinal epithelial cells[J]. Microbiology (Reading), 2017, 163: 702-711. doi:  10.1099/mic.0.000468
    [17] Pritchard AB, Fabian Z, Lawrence CL, et al. An Investigation into the Effects of Outer Membrane Vesicles and Lipopolysaccharide of Porphyromonas gingivalis on Blood-Brain Barrier Integrity, Permeability, and Disruption of Scaffolding Proteins in a Human in vitro Model[J]. J Alzheimers Dis, 2022, 86: 343-364. doi:  10.3233/JAD-215054
    [18] Muraca M, Putignani L, Fierabracci A, et al. Gut microbiota-derived outer membrane vesicles: under-recognized major players in health and disease?[J]. Discov Med, 2015, 19: 343-348.
    [19] Choi HI, Choi JP, Seo J, et al. Helicobacter pylori-derived extracellular vesicles increased in the gastric juices of gastric adenocarcinoma patients and induced inflammation mainly via specific targeting of gastric epithelial cells[J]. Exp Mol Med, 2017, 49: e330. doi:  10.1038/emm.2017.47
    [20] Lee KE, Kim JK, Han SK, et al. The extracellular vesicle of gut microbial Paenalcaligenes hominis is a risk factor for vagus nerve-mediated cognitive impairment[J]. Microbiome, 2020, 8: 107. doi:  10.1186/s40168-020-00881-2
    [21] Ma X, Shin YJ, Yoo JW, et al. Extracellular vesicles derived from Porphyromonas gingivalis induce vagus nerve-mediated cognitive impairment[J]. J Adv Res, 2023. doi:  10.1016/j.jare.2023.02.006.
    [22] Park AM, Tsunoda I. Helicobacter pylori infection in the stomach induces neuroinflammation: the potential roles of bacterial outer membrane vesicles in an animal model of Alzheimer's disease[J]. Inflamm Regen, 2022, 42: 39. doi:  10.1186/s41232-022-00224-8
    [23] Yoshida K, Yoshida K, Seyama M, et al. Porphyromonas gingivalis outer membrane vesicles in cerebral ventricles activate microglia in mice[J]. Oral Dis, 2022. doi:  10.1111/odi.14413.
    [24] Wei S, Peng W, Mai Y, et al. Outer membrane vesicles enhance tau phosphorylation and contribute to cognitive impairment[J]. J Cell Physiol, 2019, 235: 4843-4855.
    [25] Gong T, Chen Q, Mao H, et al. Outer membrane vesicles of Porphyromonas gingivalis trigger NLRP3 inflammasome and induce neuroinflammation, tau phosphorylation, and memory dysfunction in mice[J]. Front Cell Infect Microbiol, 2022, 12: 925435. doi:  10.3389/fcimb.2022.925435
    [26] Younossi ZM, Golabi P, de Avila L, et al. The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: A systematic review and meta-analysis[J]. J Hepatol, 2019, 71: 793-801. doi:  10.1016/j.jhep.2019.06.021
    [27] Villard A, Boursier J, Andriantsitohaina R. Bacterial and eukaryotic extracellular vesicles and nonalcoholic fatty liver disease: new players in the gut-liver axis? [J]. Am J Physiol Gastrointest Liver Physiol, 2021, 320: G485-G495. doi:  10.1152/ajpgi.00362.2020
    [28] Fizanne L, Villard A, Benabbou N, et al. Faeces-derived extracellular vesicles participate in the onset of barrier dysfunction leading to liver diseases[J]. J Extracell Vesicles, 2023, 12: e12303. doi:  10.1002/jev2.12303
    [29] Natsui K, Tsuchiya A, Imamiya R, et al. Escherichia coli-derived outer-membrane vesicles induce immune activation and progression of cirrhosis in mice and humans[J]. Liver Int, 2023, 43: 1126-1140. doi:  10.1111/liv.15539
    [30] Choi Y, Kwon Y, Kim DK, et al. Gut microbe-derived extracellular vesicles induce insulin resistance, thereby impairing glucose metabolism in skeletal muscle[J]. Sci Rep, 2015, 5: 15878. doi:  10.1038/srep15878
    [31] Chelakkot C, Choi Y, Kim D, et al. Akkermansia muciniphila-derived extracellular vesicles influence gut permea-bility through the regulation of tight junctions[J]. Exp Mol Mede, 2018, 50: e450. doi:  10.1038/emm.2017.282
    [32] Luo Z, Ji Y, Gao H, et al. CRIg+ Macrophages Prevent Gut Microbial DNA-Containing Extracellular Vesicle-Induced Tissue Inflammation and Insulin Resistance[J]. Gastroenterology, 2021, 160: 863-874. doi:  10.1053/j.gastro.2020.10.042
    [33] Mcinnes IB, Schett G. The pathogenesis of rheumatoid arthritis[J]. N Engl J Med, 2011, 365: 2205-2219. doi:  10.1056/NEJMra1004965
    [34] Hong M, Li Z, Liu H, et al. Fusobacterium nucleatum aggravates rheumatoid arthritis through FadA-containing outer membrane vesicles[J]. Cell Host Microbe, 2023, 31: 798-810. e7. doi:  10.1016/j.chom.2023.03.018
    [35] Quirke AM, Lugli EB, Wegner N, et al. Heightened immune response to autocitrullinated Porphyromonas gin-givalis peptidylarginine deiminase: a potential mechanism for breaching immunologic tolerance in rheumatoid arthritis[J]. Ann Rheum Dis, 2014, 73: 263-269.
    [36] Gabarrini G, Palma Medina LM, Stobernack T, et al. There's no place like OM: Vesicular sorting and secretion of the peptidylarginine deiminase of Porphyromonas gingivalis[J]. Virulence, 2018, 9: 456-464.
    [37] Sepich-Poore GD, Zitvogel L, Straussman R, et al. The microbiome and human cancer[J]. Science, 2021, 371: eabc4552. doi:  10.1126/science.abc4552
    [38] Chronopoulos A, Kalluri R. Emerging role of bacterial extracellular vesicles in cancer[J]. Oncogene, 2020, 39: 6951-6960. doi:  10.1038/s41388-020-01509-3
    [39] Chen G, Gao C, Jiang S, et al. Fusobacterium nucleatum outer membrane vesicles activate autophagy to promote oral cancer metastasis[J]. J Adv Res, 2023. doi:  10.1016/j.jare.2023.04.002.
    [40] Schaack B, Hindré T, Quansah N, et al. Microbiota-Derived Extracellular Vesicles Detected in Human Blood from Healthy Donors[J]. Int J Mol Sci, 2022, 23: 13787. doi:  10.3390/ijms232213787
    [41] Yoo JY, Rho M, You YA, et al. 16S rRNA gene-based metagenomic analysis reveals differences in bacteria-derived extracellular vesicles in the urine of pregnant and non-pregnant women[J]. Exp Mol Med, 2016, 48: e208. doi:  10.1038/emm.2015.110
    [42] Vandendriessche C, Kapogiannis D, Vandenbroucke RE. Biomarker and therapeutic potential of peripheral extracel-lular vesicles in Alzheimer's disease[J]. Adv Drug Deliv Rev, 2022, 190: 114486. doi:  10.1016/j.addr.2022.114486
    [43] Dey S, Gudipati S, Giuliano C, et al. Reduced Production of Bacterial Membrane Vesicles Predicts Mortality in ST45/USA600 Methicillin-Resistant Staphylococcus aureus Bacteremia[J]. Antibiotics (Basel), 2019, 9: 2. doi:  10.3390/antibiotics9010002
    [44] Wei SC, Wei W, Peng WJ, et al. Metabolic Alterations in the Outer Membrane Vesicles of Patients with Alzheimer's Disease: An LC-MS/MS-based Metabolomics Analysis[J]. Curr Alzheimer Res, 2019, 16: 1183-1195.
    [45] Yang J, Moon HE, Park HW, et al. Brain tumor diagnostic model and dietary effect based on extracellular vesicle microbiome data in serum[J]. Exp Mol Med, 2020, 52: 1602-1613. doi:  10.1038/s12276-020-00501-x
    [46] Kim SI, Kang N, Leem S, et al. Metagenomic Analysis of Serum Microbe-Derived Extracellular Vesicles and Diagnostic Models to Differentiate Ovarian Cancer and Benign Ovarian Tumor[J]. Cancers (Basel), 2020, 12: 1309. doi:  10.3390/cancers12051309
    [47] Kim DJ, Yang J, Seo H, et al. Colorectal cancer diagnostic model utilizing metagenomic and metabolomic data of stool microbial extracellular vesicles[J]. Sci Rep, 2020, 10: 2860. doi:  10.1038/s41598-020-59529-8
    [48] Yang J, McdowelL A, Seo H, et al. Diagnostic Models for Atopic Dermatitis Based on Serum Microbial Extracellular Vesicle Metagenomic Analysis: A Pilot Study[J]. Allergy Asthma Immunol Res, 2020, 12: 792-805. doi:  10.4168/aair.2020.12.5.792
    [49] Yang J, Hong G, Kim YS, et al. Lung Disease Diagnostic Model Through IgG Sensitization to Microbial Extracellular Vesicles[J]. Allergy Asthma Immunol Res, 2020, 12: 669-683. doi:  10.4168/aair.2020.12.4.669
    [50] Han P, Bartold PM, Salomon C, et al. Salivary Outer Membrane Vesicles and DNA Methylation of Small Extracellular Vesicles as Biomarkers for Periodontal Status: A Pilot Study[J]. Int J Mol Sci, 2021, 22: 2423. doi:  10.3390/ijms22052423
    [51] Yoon H, Kim NE, Park J, et al. Analysis of the gut microbiome using extracellular vesicles in the urine of patients with colorectal cancer[J]. Korean J Intern Med, 2023, 38: 27-38. doi:  10.3904/kjim.2022.112
    [52] VesikarI T, Esposito S, Prymula R, et al. Immunogenicity and safety of an investigational multicomponent, recombin-ant, meningococcal serogroup B vaccine (4CMenB) administered concomitantly with routine infant and child vaccinations: results of two randomised trials[J]. Lancet, 2013, 381: 825-835. doi:  10.1016/S0140-6736(12)61961-8
    [53] Kaparakis-Liaskos M, Ferrero RL. Immune modulation by bacterial outer membrane vesicles[J]. Nat Rev Immunol, 2015, 15: 375-387. doi:  10.1038/nri3837
    [54] Wo J, Lv ZY, Sun JN, et al. Engineering probiotic-derived outer membrane vesicles as functional vaccine carriers to enhance immunity against SARS-CoV-2[J]. iScience, 2023, 26: 105772. doi:  10.1016/j.isci.2022.105772
    [55] Chen L, Valentine JL, Huang CJ, et al. Outer membrane vesicles displaying engineered glycotopes elicit protective antibodies[J]. Proc Natl Acad Sci U S A, 2016, 113: E3609-E3618. doi:  10.1073/pnas.1523686113
    [56] Ñahui Palomino RA, Vanpouille C, Laghi L, et al. Extracellular vesicles from symbiotic vaginal lactobacilli inhibit HIV-1 infection of human tissues[J]. Nat Commun, 2019, 10: 5656. doi:  10.1038/s41467-019-13468-9
    [57] Kim MH, Choi S J, Choi HI, et al. Lactobacillus plantarum-derived Extracellular Vesicles Protect Atopic Dermatitis Induced by Staphylococcus aureus-derived Extracellular Vesicles[J]. Allergy Asthma Immunol Res, 2018, 10: 516-532. doi:  10.4168/aair.2018.10.5.516
    [58] Tong L, Zhang X, Hao H, et al. Lactobacillus rhamnosus GG Derived Extracellular Vesicles Modulate Gut Microbiota and Attenuate Inflammatory in DSS-Induced Colitis Mice[J]. Nutrients, 2021, 13: 3319. doi:  10.3390/nu13103319
    [59] Hu R, Lin H, Wang M, et al. Lactobacillus reuteri-derived extracellular vesicles maintain intestinal immune homeostasis against lipopolysaccharide-induced inflammatory responses in broilers[J]. J Anim Sci Biotechnol, 2021, 12: 25. doi:  10.1186/s40104-020-00532-4
    [60] Lee DH, Park HK, Lee HR, et al. Immunoregulatory effects of Lactococcus lactis-derived extracellular vesicles in allergic asthma[J]. Clin Transl Allergy, 2022, 12: e12138. doi:  10.1002/clt2.12138
    [61] Croatti V, Parolin C, Giordani B, et al. Lactobacilli extracellular vesicles: potential postbiotics to support the vaginal microbiota homeostasis[J]. Microb Cell Fact, 2022, 21: 237. doi:  10.1186/s12934-022-01963-6
    [62] Choi J, Kwon H, Kim YK, et al. Extracellular Vesicles from Gram-positive and Gram-negative Probiotics Remediate Stress-Induced Depressive Behavior in Mice[J]. Mol Neurobiol, 2022, 59: 2715-2728. doi:  10.1007/s12035-021-02655-9
    [63] Ye W, Chen Z, He Z, et al. Lactobacillus plantarum-Derived Postbiotics Ameliorate Acute Alcohol-Induced Liver Injury by Protecting Cells from Oxidative Damage, Improving Lipid Metabolism, and Regulating Intestinal Microbiota[J]. Nutrients, 2023, 15: 845. doi:  10.3390/nu15040845
    [64] Wang X, Lin S, Wang L, et al. Versatility of bacterial outer membrane vesicles in regulating intestinal homeostasis[J]. Sci Adv, 2023, 9: eade5079.
    [65] Starnes CO. Coley's toxins[J]. Nature, 1992, 360: 23.
    [66] Kim OY, ParK HT, Dinh NTH, et al. Bacterial outer membrane vesicles suppress tumor by interferon-γ-mediated antitumor response[J]. Nat Commun, 2017, 8: 626. doi:  10.1038/s41467-017-00729-8
    [67] Qing S, Lyu C, Zhu L, et al. Biomineralized Bacterial Outer Membrane Vesicles Potentiate Safe and Efficient Tumor Microenvironment Reprogramming for Anticancer Therapy[J]. Adv Mater, 2020, 32: e2002085. doi:  10.1002/adma.202002085
    [68] Li Y, Ma X, Yue Y, et al. Rapid Surface Display of mRNA Antigens by Bacteria-Derived Outer Membrane Vesicles for a Personalized Tumor Vaccine [J]. Adv Mater, 2022, 34: e2109984. doi:  10.1002/adma.202109984
    [69] Meng F, Li L, Zhang Z, et al. Biosynthetic neoantigen displayed on bacteria derived vesicles elicit systemic antitumour immunity[J]. J Extracell Vesicles, 2022, 11: e12289. doi:  10.1002/jev2.12289
    [70] Kuerban K, Gao X, Zhang H, et al. Doxorubicin-loaded bacterial outer-membrane vesicles exert enhanced anti-tumor efficacy in non-small-cell lung cancer[J]. Acta Pharm Sin B, 2020, 10: 1534-1548.
    [71] Liu XZ, Wen ZJ, Li YM, et al. Bioengineered Bacterial Membrane Vesicles with Multifunctional Nanoparticles as a Versatile Platform for Cancer Immunotherapy[J]. ACS Appl Mater Interfaces, 2023, 15: 3744-3759. doi:  10.1021/acsami.2c18244
    [72] Bauman SJ, Kuehn MJ. Purification of outer membrane vesicles from Pseudomonas aeruginosa and their activation of an IL-8 response[J]. Microbes Infect, 2006, 8: 2400-2408. doi:  10.1016/j.micinf.2006.05.001
    [73] Northrop-Albrecht EJ, Taylor WR, Huang BQ, et al. Assessment of extracellular vesicle isolation methods from human stool supernatant[J]. J Extracell Vesicles, 2022, 11: e12208. doi:  10.1002/jev2.12208
    [74] Hong J, Dauros-Singorenko P, Whitcombe A, et al. Analysis of the Escherichia coli extracellular vesicle proteome identifies markers of purity and culture conditions[J]. J Extracell Vesicles, 2019, 8: 1632099.
    [75] Gelibter S, Marostica G, Mandelli A, et al. The impact of storage on extracellular vesicles: A systematic study[J]. J Extracell Vesicles, 2022, 11: e12162. doi:  10.1002/jev2.12162
  • 加载中
图(1) / 表(2)
计量
  • 文章访问数:  1673
  • HTML全文浏览量:  146
  • PDF下载量:  169
  • 被引次数: 0
出版历程
  • 收稿日期:  2023-05-31
  • 录用日期:  2023-06-27
  • 网络出版日期:  2023-07-12
  • 刊出日期:  2023-09-30

目录

    /

    返回文章
    返回

    【温馨提醒】近日,《协和医学杂志》编辑部接到作者反映,有多名不法人员冒充期刊编辑发送见刊通知,鼓动作者添加微信,从而骗取版面费的行为。特提醒您,本刊与作者联系的方式均为邮件通知或电话,稿件进度通知邮箱为:mjpumch@126.com,编辑部电话为:010-69154261,请提高警惕,谨防上当受骗!如有任何疑问,请致电编辑部核实。谢谢!