1
|
Yu L, Shi H, Gao T, Xu W, Qian H, Jiang J, Yang X, Zhang X. Exomeres and supermeres: Current advances and perspectives. Bioact Mater 2025; 50:322-343. [PMID: 40276541 PMCID: PMC12020890 DOI: 10.1016/j.bioactmat.2025.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/26/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
Recent studies have revealed a great diversity and complexity in extracellular vesicles and particles (EVPs). The developments in techniques and the growing awareness of the particle heterogeneity have spurred active research on new particle subsets. Latest discoveries highlighted unique features and roles of non-vesicular extracellular nanoparticles (NVEPs) as promising biomarkers and targets for diseases. These nanoparticles are distinct from extracellular vesicles (EVs) in terms of their smaller particle sizes and lack of a bilayer membrane structure and they are enriched with diverse bioactive molecules particularly proteins and RNAs, which are widely reported to be delivered and packaged in exosomes. This review is focused on the two recently identified membraneless NVEPs, exomeres and supermeres, to provide an overview of their biogenesis and contents, particularly those bioactive substances linked to their bio-properties. This review also explains the concepts and characteristics of these nanoparticles, to compare them with other EVPs, especially EVs, as well as to discuss their isolation and identification methods, research interests, potential clinical applications and open questions.
Collapse
Affiliation(s)
- Li Yu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Hui Shi
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Tingxin Gao
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Wenrong Xu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Clinical Laboratory, School of Medicine, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Jiajia Jiang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, 279 Jingang Road, Zhangjiagang, Suzhou, 215600, Jiangsu, China
| | - Xiao Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
2
|
Kim H, Lee J, Qian A, Ji YR, Zhang R, Hu Q, Williams CK, Chuang HY, Smalley MD, Xu Y, Gao L, Mayo MC, Zhang T, Posadas EM, Tan ZS, Vinters HV, Vossel K, Magaki S, Zhu Y, Tseng HR. Noninvasive Assessment of β-Secretase Activity Through Click Chemistry-Mediated Enrichment of Neuronal Extracellular Vesicles to Detect Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2415289. [PMID: 40245252 DOI: 10.1002/advs.202415289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/27/2025] [Indexed: 04/19/2025]
Abstract
Alzheimer's disease (AD), the most prevalent type of dementia, is characterized by a biological process that begins with the development of AD neuropathologic change (ADNPC) while individuals remain asymptomatic. A key molecular hallmark of ADNPC is the accumulation of amyloid-β plaques. β-secretase plays a critical role in the upstream pathological cleavage of amyloid precursor protein (APP), producing amyloid-β peptides that are prone to misfolding, ultimately contributing to plaque formation. Neuronal extracellular vesicles (NEVs) in the blood transport β-secretase and preserve its activity, allowing for noninvasive profiling of β-secretase activity for detecting early onset of ADNPC. In this study, a novel approach is approached for noninvasive assessment of β-secretase activity in AD patients using an NEV β-secretase activity assay. This assay identifies NEVs exhibiting colocalization of NEV markers with AD-associated β-secretase, generating a β-secretase activity profile for each patient. The NEV β-secretase activity assay represents a significant advancement in leveraging the diagnostic potential of NEVs, offering a noninvasive, quantitative method for reliably assessing β-secretase activity to detect the early onset of ADNPC.
Collapse
Affiliation(s)
- Hyoyong Kim
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Junseok Lee
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Audrey Qian
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - You-Ren Ji
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Ryan Zhang
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Qixin Hu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Christopher Kazu Williams
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Han-Yu Chuang
- Eximius Diagnostics Corp, Magnify Incubator, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Matthew D Smalley
- Eximius Diagnostics Corp, Magnify Incubator, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Yaya Xu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Liang Gao
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - Mary C Mayo
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Ting Zhang
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Edwin M Posadas
- Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Zaldy S Tan
- Departments of Neurology and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Harry V Vinters
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Keith Vossel
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Shino Magaki
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| |
Collapse
|
3
|
Li Q, Liu J, Qi Z, Zhou W, Li W, Wu D, Li G. Bioorthogonal Synthesis of Biomimetic Nanoparticles for Screening Chemical Hazards in Food Samples. Anal Chem 2025; 97:247-254. [PMID: 39810346 DOI: 10.1021/acs.analchem.4c03988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The ability to identify unknown risks is the key to improving the level of food safety. However, the conventional nontargeted screening methods for new contaminant identification and risk assessment remain difficult work. Herein, a toxic-oriented screening platform based on high-expression epidermal growth factor receptor HEK293 cell membrane-coated magnetic nanoparticles (EGFR/MNPs) was first used for the discovery of unknown contaminants from food samples. The EGFR served as a bait to bind the risk compounds, considering that the abnormal activation of EGFR was related to the incidence of numerous chronic inflammatory diseases. The cell membranes were specifically covalently immobilized on magnetic carriers through metabolic glycoengineering and strain-promoted alkyne-azide cycloaddition (SPAAC) to ensure the stability and bioactivity of the EGFR/MNPs. The synthesized EGFR/MNPs possessed an excellent receptor loading amount and were used to screen potential risk substances from thermally processed food. Finally, two compounds, harman and norharman, were rapidly identified. The toxicological experiments confirmed that the screened compounds could promote EGFR phosphorylation and further activate downstream signaling pathways, thereby inducing cellular oxidative damage. The proposed method provided a toxic-oriented method for identifying risk compounds from food samples.
Collapse
Affiliation(s)
- Qianyu Li
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Jianghua Liu
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Zihe Qi
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Wen Zhou
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Wenrui Li
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| | - Di Wu
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast BT9 5DL, U.K
| | - Guoliang Li
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China
| |
Collapse
|
4
|
Liu ZX, Chen G, Yu ZL. Advances in subpopulation separation and detection of extracellular vesicles: for liquid biopsy and downstream research. Theranostics 2025; 15:1135-1155. [PMID: 39776815 PMCID: PMC11700854 DOI: 10.7150/thno.106459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Extracellular vesicles (EVs) are carriers of a diverse array of bioactive molecules, making them valuable clinical tools for liquid biopsy in disease diagnosis and prognosis evaluation. These molecules play critical roles in various physiological and pathological conditions, and effective separation of EVs is essential to achieve these objectives. Due to the high heterogeneity of EVs, particularly with regard to their cargo molecules, merely isolating the general EV population is inadequate for liquid biopsy and biological function studies. Therefore, separating EV subpopulations becomes crucial. Traditional separation methods, such as differential ultracentrifugation and size exclusion chromatography, along with burgeoning techniques like classical microfluidic chips and covalent chemistry, often prove time-consuming, yield low purity, and have limited ability to address cargo heterogeneity. Thus, precise separation of EV subpopulations is of utmost importance. Additionally, detecting subpopulation-specific cargo is vital for validating the effectiveness of separation methods and supporting clinical biopsy applications. However, reviews that focus specifically on detection methods for EV subpopulations are limited. This paper provides a comprehensive overview of the methods for separating and detecting EV subpopulations with surface marker heterogeneity, comparing the advantages and limitations of each technique. Furthermore, it discusses challenges and future prospects for these methods in the context of liquid biopsy and downstream research. Collectively, this review aims to offer innovative insights into the separation and detection of EV subpopulations, guiding researchers to avoid common pitfalls and refine their investigative approaches.
Collapse
Affiliation(s)
- Zi-Xiu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Gang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Zi-Li Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
5
|
Liu Z, Pang B, Wang Y, Zheng J, Li Y, Jiang J. Advances of New Extracellular Vesicle Isolation and Detection Technologies in Cancer Diagnosis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2405872. [PMID: 39676429 DOI: 10.1002/smll.202405872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 11/25/2024] [Indexed: 12/17/2024]
Abstract
Cancer is a global health issue threatening people's lives. Currently, cancer detection methods still have a lot of room for improvement in both efficiency and accuracy. The development and application of new technologies are urgently required for early cancer diagnosis and prognosis. Extracellular vesicles (EVs) are a type of phospholipid bilayer vesicle secreted by cells and play an important role in cancer development and metastasis. These small vesicles participate in cancer information transmission, antigen presentation, angiogenesis, immune response, tumor invasion, and mediate signaling pathways in the tumor microenvironment. Liquid biopsy of EV cargo contents is a fast-developing research area, holding promise for early cancer diagnosis and monitoring cancer progression in real-time. However, current EV detection technologies for clinical translation are still facing many challenges. Recent advancements in developing techniques for EV isolation and detection have made significant progress and are paving the way toward clinical application. Here, the advantages and limitations of traditional EV detection and isolation technologies in cancer diagnosis and prognosis are reviewed. The review also focuses on emerging EV detection and isolation technologies in cancer, discusses the challenges faced by current methods, and explores the perspective of new EV detection techniques for future cancer diagnosis.
Collapse
Affiliation(s)
- Zhihan Liu
- The First Affiliated Hospital of Ningbo University, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
- Translational Research Laboratory for Urology, Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
| | - Bairen Pang
- The First Affiliated Hospital of Ningbo University, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
- Translational Research Laboratory for Urology, Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
- Zhejiang Engineering Research Center of Innovative Technologies and Diagnostic and Therapeutic Equipment for Urinary System Diseases, Ningbo, Zhejiang, 315010, China
| | - Yuhui Wang
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering (NIMTE) of the Chinese, Chinese Academy of Sciences, Ningbo, 315000, China
| | - Jianping Zheng
- Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering (NIMTE) of the Chinese, Chinese Academy of Sciences, Ningbo, 315000, China
| | - Yong Li
- Cancer Care Centre, St. George Hospital, Kogarah, NSW, 2217, Australia
- St. George and Sutherland Clinical Campuses, School of Clinical Medicine UNSW Sydney, Kensington, NSW, 2052, Australia
| | - Junhui Jiang
- The First Affiliated Hospital of Ningbo University, Health Science Center, Ningbo University, Ningbo, Zhejiang, 315211, China
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
- Translational Research Laboratory for Urology, Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, 315010, China
- Zhejiang Engineering Research Center of Innovative Technologies and Diagnostic and Therapeutic Equipment for Urinary System Diseases, Ningbo, Zhejiang, 315010, China
| |
Collapse
|
6
|
Wang Z, Zhou X, Kong Q, He H, Sun J, Qiu W, Zhang L, Yang M. Extracellular Vesicle Preparation and Analysis: A State-of-the-Art Review. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401069. [PMID: 38874129 PMCID: PMC11321646 DOI: 10.1002/advs.202401069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/11/2024] [Indexed: 06/15/2024]
Abstract
In recent decades, research on Extracellular Vesicles (EVs) has gained prominence in the life sciences due to their critical roles in both health and disease states, offering promising applications in disease diagnosis, drug delivery, and therapy. However, their inherent heterogeneity and complex origins pose significant challenges to their preparation, analysis, and subsequent clinical application. This review is structured to provide an overview of the biogenesis, composition, and various sources of EVs, thereby laying the groundwork for a detailed discussion of contemporary techniques for their preparation and analysis. Particular focus is given to state-of-the-art technologies that employ both microfluidic and non-microfluidic platforms for EV processing. Furthermore, this discourse extends into innovative approaches that incorporate artificial intelligence and cutting-edge electrochemical sensors, with a particular emphasis on single EV analysis. This review proposes current challenges and outlines prospective avenues for future research. The objective is to motivate researchers to innovate and expand methods for the preparation and analysis of EVs, fully unlocking their biomedical potential.
Collapse
Affiliation(s)
- Zesheng Wang
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| | - Xiaoyu Zhou
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| | - Qinglong Kong
- The Second Department of Thoracic SurgeryDalian Municipal Central HospitalDalian116033P. R. China
| | - Huimin He
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| | - Jiayu Sun
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
| | - Wenting Qiu
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
| | - Liang Zhang
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| | - Mengsu Yang
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhenGuangdong518000P. R. China
- Department of Biomedical Sciencesand Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| |
Collapse
|
7
|
Zhao W, Li K, Li L, Wang R, Lei Y, Yang H, Sun L. Mesenchymal Stem Cell-Derived Exosomes as Drug Delivery Vehicles in Disease Therapy. Int J Mol Sci 2024; 25:7715. [PMID: 39062956 PMCID: PMC11277139 DOI: 10.3390/ijms25147715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Exosomes are small vesicles containing proteins, nucleic acids, and biological lipids, which are responsible for intercellular communication. Studies have shown that exosomes can be utilized as effective drug delivery vehicles to accurately deliver therapeutic substances to target tissues, enhancing therapeutic effects and reducing side effects. Mesenchymal stem cells (MSCs) are a class of stem cells widely used for tissue engineering, regenerative medicine, and immunotherapy. Exosomes derived from MSCs have special immunomodulatory functions, low immunogenicity, the ability to penetrate tumor tissues, and high yield, which are expected to be engineered into efficient drug delivery systems. Despite the promising promise of MSC-derived exosomes, exploring their optimal preparation methods, drug-loading modalities, and therapeutic potential remains challenging. Therefore, this article reviews the related characteristics, preparation methods, application, and potential risks of MSC-derived exosomes as drug delivery systems in order to find potential therapeutic breakthroughs.
Collapse
Affiliation(s)
- Wenzhe Zhao
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Kaixuan Li
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Liangbo Li
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Ruichen Wang
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Yang Lei
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| | - Hui Yang
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
| | - Leming Sun
- School of Life Sciences, Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi’an 710072, China; (W.Z.); (K.L.); (L.L.); (R.W.); (Y.L.)
- Dongguan Sanhang Innovation Institute, Dongguan 523808, China
| |
Collapse
|
8
|
Grishaev NA, Moiseeva EO, Chernyshev VS, Komlev AS, Novoselov AM, Yashchenok AM. Studying the small extracellular vesicle capture efficiency of magnetic beads coated with tannic acid. J Mater Chem B 2024; 12:6678-6689. [PMID: 38894640 DOI: 10.1039/d4tb00127c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
The isolation of small extracellular vesicles (sEVs), including those secreted by pathological cells, with high efficiency and purity is highly demanded for research studies and practical applications. Conventional sEV isolation methods suffer from low yield, presence of contaminants, long-term operation and high costs. Bead-assisted platforms are considered to be effective for trapping sEVs with high recovery yield and sufficient purity for further molecular profiling. In this study, magnetically responsive beads made of calcium carbonate (CaCO3) particles impregnated with iron oxide (Fe3O4) nanoparticles are fabricated using a freezing-induced loading (FIL) method. The developed magnetic beads demonstrate sufficient magnetization and can be collected by a permanent magnet, ensuring their rapid and gentle capture from an aqueous solution. The tannic acid on the surface of magnetic beads is formed by a layer-by-layer (LbL) method and is used to induce coupling of sEVs with the surface of magnetic beads. These tannic acid coated magnetic beads (TAMB) were applied to capture sEVs derived from MCF7 and HCT116 cell lines. Quantitative data derived from nanoparticle tracking analysis (NTA) and BCA methods revealed the capture efficiency and recovery yield of about 60%. High-resolution transmission electron microscopy (HRTEM) imaging of sEVs on the surface of TAMBs indicated their structural integrity. Compared with the size exclusion chromatography (SEC) method, the proposed approach demonstrated comparable efficiency in terms of recovery yield and purity, while offering a relatively short operation time. These results highlight the high potential of the TAMB approach for the enrichment of sEVs from biological fluids, such as cell culture media.
Collapse
Affiliation(s)
- Nikita A Grishaev
- Skoltech Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology Skolkovo Innovation Center, 121205 Moscow, Russia.
| | - Ekaterina O Moiseeva
- Skoltech Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology Skolkovo Innovation Center, 121205 Moscow, Russia.
| | - Vasiliy S Chernyshev
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov, 117997 Moscow, Russia
| | - Aleksei S Komlev
- Faculty of Physics, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Anton M Novoselov
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Alexey M Yashchenok
- Skoltech Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology Skolkovo Innovation Center, 121205 Moscow, Russia.
| |
Collapse
|
9
|
Lim W, Lee S, Koh M, Jo A, Park J. Recent advances in chemical biology tools for protein and RNA profiling of extracellular vesicles. RSC Chem Biol 2024; 5:483-499. [PMID: 38846074 PMCID: PMC11151817 DOI: 10.1039/d3cb00200d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 04/25/2024] [Indexed: 06/09/2024] Open
Abstract
Extracellular vesicles (EVs) are nano-sized vesicles secreted by cells that contain various cellular components such as proteins, nucleic acids, and lipids from the parent cell. EVs are abundant in body fluids and can serve as circulating biomarkers for a variety of diseases or as a regulator of various biological processes. Considering these characteristics of EVs, analysis of the EV cargo has been spotlighted for disease diagnosis or to understand biological processes in biomedical research. Over the past decade, technologies for rapid and sensitive analysis of EVs in biofluids have evolved, but detection and isolation of targeted EVs in complex body fluids is still challenging due to the unique physical and biological properties of EVs. Recent advances in chemical biology provide new opportunities for efficient profiling of the molecular contents of EVs. A myriad of chemical biology tools have been harnessed to enhance the analytical performance of conventional assays for better understanding of EV biology. In this review, we will discuss the improvements that have been achieved using chemical biology tools.
Collapse
Affiliation(s)
- Woojeong Lim
- Department of Chemistry, Kangwon National University Chuncheon 24341 Korea
| | - Soyeon Lee
- Department of Chemistry, Kangwon National University Chuncheon 24341 Korea
| | - Minseob Koh
- Department of Chemistry, Pusan National University Busan 46241 Republic of Korea
| | - Ala Jo
- Center for Nanomedicine, Institute for Basic Science Seoul 03722 Republic of Korea
| | - Jongmin Park
- Department of Chemistry, Kangwon National University Chuncheon 24341 Korea
- Institute for Molecular Science and Fusion Technology, Kangwon National University Chuncheon 24341 Republic of Korea
- Multidimensional Genomics Research Center, Kangwon National University Chuncheon 24341 Republic of Korea
| |
Collapse
|
10
|
Qin X, Xiang Y, Mao L, Yang Y, Wei B, Lu H, Li X, Zhang Y, Yang F. Buoyant Metal-Organic Framework Corona-Driven Fast Isolation and Ultrasensitive Profiling of Circulating Extracellular Vesicles. ACS NANO 2024; 18:14569-14582. [PMID: 38781132 DOI: 10.1021/acsnano.4c02339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Accurately assaying tumor-derived circulating extracellular vesicles (EVs) is fundamental in noninvasive cancer diagnosis and therapeutic monitoring but limited by challenges in efficient EV isolation and profiling. Here, we report a bioinspired buoyancy-driven metal-organic framework (MOF) corona that leverages on-bubble coordination and dual-encoded surface-enhanced Raman scattering (SERS) nanotags to streamline rapid isolation and ultrasensitive profiling of plasma EVs in a single assay for cancer diagnostics. This integrated bubble-MOF-SERS EV assay (IBMsv) allows barnacle-like high-density adhesion of MOFs on a self-floating bubble surface to enable fast isolation (2 min, near 90% capture efficiency) of tumor EVs via enhanced EV-MOF binding. Also, IBMsv harnesses four-plexed SERS nanotags to profile the captured EV surface protein markers at a single-particle level. Such a sensitive assay allows multiplexed profiling of EVs across five cancer types, revealing heterogeneous EV surface expression patterns. Furthermore, the IBMsv assay enables cancer diagnosis in a pilot clinical cohort (n = 55) with accuracies >95%, improves discrimination between cancer and noncancer patients via an algorithm, and monitors the surgical treatment response from hepatocellular carcinoma patients. This assay provides a fast, sensitive, streamlined, multiplexed, and portable blood test tool to enable cancer diagnosis and response monitoring in clinical settings.
Collapse
Affiliation(s)
- Xiaojie Qin
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Yuanhang Xiang
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Linfeng Mao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning 530021, China
| | - Yu Yang
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Binqi Wei
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Hao Lu
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Xinchun Li
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Yuanqing Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Fan Yang
- Guangxi Key Laboratory of Pharmaceutical Precision Detection and Screening, Key Laboratory of Micro-Nanoscale Bioanalysis and Drug Screening of Guangxi Education Department, State Key Laboratory of Targeting Oncology, Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
11
|
Dai L, Zhou S, Yang C, Li J, Wang Y, Qin M, Pan L, Zhang D, Qian Z, Wu H. A bioorthogonal cell sorting strategy for isolation of desired cell phenotypes. Chem Commun (Camb) 2024; 60:1916-1919. [PMID: 38259188 DOI: 10.1039/d3cc05604j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Here we describe a cost-effective and simplified cell sorting method using tetrazine bioorthogonal chemistry. We successfully isolated SKOV3 cells from complex mixtures, demonstrating efficacy in separating mouse lymphocytes expressing interferon and HeLa cells expressing virally transduced green fluorescent protein post-infection.
Collapse
Affiliation(s)
- Liqun Dai
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Siming Zhou
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Cheng Yang
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Jie Li
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yayue Wang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China
| | - Meng Qin
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lili Pan
- Department of Nuclear Medicine, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Dan Zhang
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Zhiyong Qian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Haoxing Wu
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province and Frontiers Science Center for Disease Related Molecular Network West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
12
|
Chen H, Pang B, Zhou C, Han M, Gong J, Li Y, Jiang J. Prostate cancer-derived small extracellular vesicle proteins: the hope in diagnosis, prognosis, and therapeutics. J Nanobiotechnology 2023; 21:480. [PMID: 38093355 PMCID: PMC10720096 DOI: 10.1186/s12951-023-02219-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/18/2023] [Indexed: 12/17/2023] Open
Abstract
Current diagnostic tools for prostate cancer (PCa) diagnosis and risk stratification are insufficient. The hidden onset and poor efficacy of traditional therapies against metastatic PCa make this disease a heavy burden in global men's health. Prostate cancer-derived extracellular vesicles (PCDEVs) have garnered attention in recent years due to their important role in communications in tumor microenvironment. Recent advancements have demonstrated PCDEVs proteins play an important role in PCa invasion, progression, metastasis, therapeutic resistance, and immune escape. In this review, we briefly discuss the applications of sEV proteins in PCa diagnosis and prognosis in liquid biopsy, focus on the roles of the PCa-derived small EVs (sEVs) proteins in tumor microenvironment associated with cancer progression, and explore the therapeutic potential of sEV proteins applied for future metastatic PCa therapy.
Collapse
Affiliation(s)
- Haotian Chen
- Health Science Center, Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
- Translational Research Laboratory for Urology, Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
| | - Bairen Pang
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
- Translational Research Laboratory for Urology, Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
| | - Cheng Zhou
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
- Translational Research Laboratory for Urology, Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
| | - Meng Han
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
- Translational Research Laboratory for Urology, Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
| | - Jie Gong
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
- Translational Research Laboratory for Urology, Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
| | - Yong Li
- Cancer Care Centre, St George Hospital, Kogarah, NSW, 2217, Australia.
- School of Clinical Medicine, St. George and Sutherland Clinical Campuses, UNSW Sydney, Kensington, NSW, 2052, Australia.
| | - Junhui Jiang
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China.
- Translational Research Laboratory for Urology, Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China.
- Department of Urology, Ningbo First Hospital, The First Affiliated Hospital of Ningbo University, Haishu District, Ningbo, 315600, Zhejiang, People's Republic of China.
| |
Collapse
|
13
|
Flora K, Ishihara M, Zhang Z, Bowen ES, Wu A, Ayoub T, Huang J, Cano-Ruiz C, Jackson M, Reghu K, Ayoub Y, Zhu Y, Tseng HR, Zhou ZH, Hu J, Wu L. Exosomes from Von Hippel-Lindau-Null Cancer Cells Promote Metastasis in Renal Cell Carcinoma. Int J Mol Sci 2023; 24:17307. [PMID: 38139136 PMCID: PMC10743428 DOI: 10.3390/ijms242417307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/18/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Exosomes are extracellular vesicles that modulate essential physiological and pathological signals. Communication between cancer cells that express the von Hippel-Lindau (VHL) tumor suppressor gene and those that do not is instrumental to distant metastasis in renal cell carcinoma (RCC). In a novel metastasis model, VHL(-) cancer cells are the metastatic driver, while VHL(+) cells receive metastatic signals from VHL(-) cells and undergo aggressive transformation. This study investigates whether exosomes could be mediating metastatic crosstalk. Exosomes isolated from paired VHL(+) and VHL(-) cancer cell lines were assessed for physical, biochemical, and biological characteristics. Compared to the VHL(+) cells, VHL(-) cells produce significantly more exosomes that augment epithelial-to-mesenchymal transition (EMT) and migration of VHL(+) cells. Using a Cre-loxP exosome reporter system, the fluorescent color conversion and migration were correlated with dose-dependent delivery of VHL(-) exosomes. VHL(-) exosomes even induced a complete cascade of distant metastasis when added to VHL(+) tumor xenografts in a duck chorioallantoic membrane (dCAM) model, while VHL(+) exosomes did not. Therefore, this study supports that exosomes from VHL(-) cells could mediate critical cell-to-cell crosstalk to promote metastasis in RCC.
Collapse
Affiliation(s)
- Kailey Flora
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA;
| | - Moe Ishihara
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; (M.I.); (Z.Z.); (C.C.-R.)
| | - Zhicheng Zhang
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; (M.I.); (Z.Z.); (C.C.-R.)
| | - Elizabeth S. Bowen
- Department of Computational and Systems Biology, University of California, Los Angeles, CA 90095, USA;
| | - Aimee Wu
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA; (A.W.); (J.H.); (M.J.); (K.R.)
| | - Tala Ayoub
- Department of Physiology, University of California, Los Angeles, CA 90095, USA;
| | - Julian Huang
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA; (A.W.); (J.H.); (M.J.); (K.R.)
| | - Celine Cano-Ruiz
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; (M.I.); (Z.Z.); (C.C.-R.)
| | - Maia Jackson
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA; (A.W.); (J.H.); (M.J.); (K.R.)
| | - Kaveeya Reghu
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA; (A.W.); (J.H.); (M.J.); (K.R.)
| | - Yasmeen Ayoub
- School of Medicine, Saint Louis University, St. Louis, MO 63104, USA;
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA; (Y.Z.); (H.-R.T.); (Z.H.Z.)
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA; (Y.Z.); (H.-R.T.); (Z.H.Z.)
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Z. Hong Zhou
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA; (Y.Z.); (H.-R.T.); (Z.H.Z.)
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA
| | - Junhui Hu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; (M.I.); (Z.Z.); (C.C.-R.)
| | - Lily Wu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; (M.I.); (Z.Z.); (C.C.-R.)
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
14
|
Chen C, Cai N, Niu Q, Tian Y, Hu Y, Yan X. Quantitative assessment of lipophilic membrane dye-based labelling of extracellular vesicles by nano-flow cytometry. J Extracell Vesicles 2023; 12:e12351. [PMID: 37525378 PMCID: PMC10390660 DOI: 10.1002/jev2.12351] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 07/16/2023] [Indexed: 08/02/2023] Open
Abstract
Although lipophilic membrane dyes (LMDs) or probes (LMPs) are widely used to label extracellular vesicles (EVs) for detection and purification, their labelling performance has not been systematically characterized. Through concurrent side scattering and fluorescence detection of single EVs as small as 40 nm in diameter by a laboratory-built nano-flow cytometer (nFCM), present study identified that (1) PKH67 and PKH26 could maximally label ∼60%-80% of EVs isolated from the conditioned cell culture medium (purity of ∼88%) and ∼40%-70% of PFP-EVs (purity of ∼73%); (2) excessive PKH26 could cause damage to the EV structure; (3) di-8-ANEPPS and high concentration of DiI could achieve efficient and uniform labelling of EVs with nearly 100% labelling efficiency for di-8-ANEPPS and 70%-100% for DiI; (4) all the four tested LMDs can aggregate and form micelles that exhibit comparable side scatter and fluorescence intensity with those of labelled EVs and thus hardly be differentiate from each other; (5) as the LMD concentration went up, the particle number of self-aggregates increased while the fluorescence intensity of aggregates remained constant; (6) PKH67 and PKH26 tend to form more aggregated micelles than di-8-ANEPPS and DiI, and the effect of LMD self-aggregation can be negligible at optimal staining conditions. (7) All the four tested LMDs can label almost all the very-low-density lipoprotein (VLDL) particles, indicating potential confounding factor in plasma-EV labelling. Besides, it was discovered that DSPE-PEG2000 -biotin can only label ∼50% of plasma-EVs. The number of LMP inserted into the membrane of single EVs was measured for the first time and it was confirmed that membrane labelling by lipophilic dyes did not interfere with the immunophenotyping of EVs. nFCM provides a unique perspective for a better understanding of EV labelling by LMD/LMP.
Collapse
Affiliation(s)
- Chen Chen
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical EngineeringXiamen UniversityXiamenPeople's Republic of China
| | - Niangui Cai
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical EngineeringXiamen UniversityXiamenPeople's Republic of China
| | - Qian Niu
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical EngineeringXiamen UniversityXiamenPeople's Republic of China
| | - Ye Tian
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical EngineeringXiamen UniversityXiamenPeople's Republic of China
| | - Yunyun Hu
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical EngineeringXiamen UniversityXiamenPeople's Republic of China
| | - Xiaomei Yan
- Department of Chemical Biology, MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, College of Chemistry and Chemical EngineeringXiamen UniversityXiamenPeople's Republic of China
| |
Collapse
|
15
|
Zhang Y, Wu Q, Huang Y, Wang W, Lu Y, Kang S, Yang C, Song Y. Reliable Detection of Extracellular PD-L1 by DNA Computation-Mediated Microfluidics. Anal Chem 2023. [PMID: 37276048 DOI: 10.1021/acs.analchem.3c01686] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Extracellular vesicle PD-L1 (programmed death-1 ligand 1) is of greater value in tumor diagnosis, prognosis, and efficacy monitoring of anti-PD-1/PD-L1 immunotherapy. However, soluble PD-L1 interferes with the accurate detection of extracellular vesicle (EV) PD-L1. Here, we developed a microfluidic differentiation method for the detection of extracellular PD-L1, without the interference of soluble, by DNA computation with lipid probes and PD-L1 aptamer as inputs (DECLA). For the developed DECLA method, a cholesterol-DNA probe was designed that efficiently embeds into the EV membrane, and an aptamer-based PD-L1 probe was used for PD-L1 recognition. Due to the stable secondary structure of the designed connector, only cobinding of cholesterol-DNA and PD-L1 affinity probe induced biotin-labeled connector activation, while soluble PD-L1 cannot hybridize. As a result, PD-L1 EVs can be efficiently captured by streptavidin-functioned herringbone chip and quantified by anti-CD63-induced fluorescence signal. The high specificity of dual-input DNA computation allied to the high sensitivity of microfluidic-based detection was suitable for distinguishing lung cancer patients from healthy donors, highlighting its potential translation to clinical diagnosis and therapy monitoring.
Collapse
Affiliation(s)
- Yuqian Zhang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Collaborative Innovation Center of Chemistry for Energy Materials, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Qiuyue Wu
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Collaborative Innovation Center of Chemistry for Energy Materials, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yihao Huang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Collaborative Innovation Center of Chemistry for Energy Materials, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Wencheng Wang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Collaborative Innovation Center of Chemistry for Energy Materials, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yinzhu Lu
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Collaborative Innovation Center of Chemistry for Energy Materials, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Siyin Kang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Collaborative Innovation Center of Chemistry for Energy Materials, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Chaoyong Yang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Collaborative Innovation Center of Chemistry for Energy Materials, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yanling Song
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Collaborative Innovation Center of Chemistry for Energy Materials, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- Innovation Laboratory for Sciences and Technologies of Energy Materials of Fujian Province (IKKEM), Xiamen 361005, China
| |
Collapse
|
16
|
Hu M, Brown V, Jackson JM, Wijerathne H, Pathak H, Koestler DC, Nissen E, Hupert ML, Muller R, Godwin AK, Witek MA, Soper SA. Assessing Breast Cancer Molecular Subtypes Using Extracellular Vesicles' mRNA. Anal Chem 2023; 95:7665-7675. [PMID: 37071799 PMCID: PMC10243595 DOI: 10.1021/acs.analchem.3c00624] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023]
Abstract
Extracellular vesicles (EVs) carry RNA cargo that is believed to be associated with the cell-of-origin and thus have the potential to serve as a minimally invasive liquid biopsy marker for supplying molecular information to guide treatment decisions (i.e., precision medicine). We report the affinity isolation of EV subpopulations with monoclonal antibodies attached to the surface of a microfluidic chip that is made from a plastic to allow for high-scale production. The EV microfluidic affinity purification (EV-MAP) chip was used for the isolation of EVs sourced from two-orthogonal cell types and was demonstrated for its utility in a proof-of-concept application to provide molecular subtyping information for breast cancer patients. The orthogonal selection process better recapitulated the epithelial tumor microenvironment by isolating two subpopulations of EVs: EVEpCAM (epithelial cell adhesion molecule, epithelial origin) and EVFAPα (fibroblast activation protein α, mesenchymal origin). The EV-MAP provided recovery >80% with a specificity of 99 ± 1% based on exosomal mRNA (exo-mRNA) and real time-droplet digital polymerase chain reaction results. When selected from the plasma of healthy donors and breast cancer patients, EVs did not differ in size or total RNA mass for both markers. On average, 0.5 mL of plasma from breast cancer patients yielded ∼2.25 ng of total RNA for both EVEpCAM and EVFAPα, while in the case of cancer-free individuals, it yielded 0.8 and 1.25 ng of total RNA from EVEpCAM and EVFAPα, respectively. To assess the potential of these two EV subpopulations to provide molecular information for prognostication, we performed the PAM50 test (Prosigna) on exo-mRNA harvested from each EV subpopulation. Results suggested that EVEpCAM and EVFAPα exo-mRNA profiling using subsets of the PAM50 genes and a novel algorithm (i.e., exo-PAM50) generated 100% concordance with the tumor tissue.
Collapse
Affiliation(s)
- Mengjia Hu
- Department of Cancer Biology, The University of Kansas Medical Center, Cancer Center, Kansas City, Kansas 66160, United States
- Center of BioModular Multi-Scale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
- Department of Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Virginia Brown
- Center of BioModular Multi-Scale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
- Bioengineering Program, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Joshua M Jackson
- Center of BioModular Multi-Scale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
- Department of Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Harshani Wijerathne
- Center of BioModular Multi-Scale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
- Department of Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
| | - Harsh Pathak
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Devin C Koestler
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
- Department of Biostatistics & Data Science, The University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Emily Nissen
- Department of Biostatistics & Data Science, The University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | | | - Rolf Muller
- BioFluidica, Inc., San Diego, California 92121, United States
| | - Andrew K Godwin
- Center of BioModular Multi-Scale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Malgorzata A Witek
- Center of BioModular Multi-Scale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
- Department of Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
| | - Steven A Soper
- Department of Cancer Biology, The University of Kansas Medical Center, Cancer Center, Kansas City, Kansas 66160, United States
- Center of BioModular Multi-Scale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66045, United States
- Department of Chemistry, The University of Kansas, Lawrence, Kansas 66045, United States
- Bioengineering Program, The University of Kansas, Lawrence, Kansas 66045, United States
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, United States
- BioFluidica, Inc., San Diego, California 92121, United States
- Department of Mechanical Engineering, The University of Kansas, Lawrence, Kansas 66045, United States
| |
Collapse
|
17
|
Del Real Mata C, Jeanne O, Jalali M, Lu Y, Mahshid S. Nanostructured-Based Optical Readouts Interfaced with Machine Learning for Identification of Extracellular Vesicles. Adv Healthc Mater 2023; 12:e2202123. [PMID: 36443009 DOI: 10.1002/adhm.202202123] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/14/2022] [Indexed: 11/30/2022]
Abstract
Extracellular vesicles (EVs) are shed from cancer cells into body fluids, enclosing molecular information about the underlying disease with the potential for being the target cancer biomarker in emerging diagnosis approaches such as liquid biopsy. Still, the study of EVs presents major challenges due to their heterogeneity, complexity, and scarcity. Recently, liquid biopsy platforms have allowed the study of tumor-derived materials, holding great promise for early-stage diagnosis and monitoring of cancer when interfaced with novel adaptations of optical readouts and advanced machine learning analysis. Here, recent advances in labeled and label-free optical techniques such as fluorescence, plasmonic, and chromogenic-based systems interfaced with nanostructured sensors like nanoparticles, nanoholes, and nanowires, and diverse machine learning analyses are reviewed. The adaptability of the different optical methods discussed is compared and insights are provided into prospective avenues for the translation of the technological approaches for cancer diagnosis. It is discussed that the inherent augmented properties of nanostructures enhance the sensitivity of the detection of EVs. It is concluded by reviewing recent integrations of nanostructured-based optical readouts with diverse machine learning models as novel analysis ventures that can potentially increase the capability of the methods to the point of translation into diagnostic applications.
Collapse
Affiliation(s)
| | - Olivia Jeanne
- McGill University, Department of Bioengineering, Montreal, QC, H3A 0E9, Canada
| | - Mahsa Jalali
- McGill University, Department of Bioengineering, Montreal, QC, H3A 0E9, Canada
| | - Yao Lu
- McGill University, Department of Bioengineering, Montreal, QC, H3A 0E9, Canada
| | - Sara Mahshid
- McGill University, Department of Bioengineering, Montreal, QC, H3A 0E9, Canada
| |
Collapse
|
18
|
Gao J, Li A, Hu J, Feng L, Liu L, Shen Z. Recent developments in isolating methods for exosomes. Front Bioeng Biotechnol 2023; 10:1100892. [PMID: 36714629 PMCID: PMC9879965 DOI: 10.3389/fbioe.2022.1100892] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/22/2022] [Indexed: 01/14/2023] Open
Abstract
Exosomes are the smallest extracellular vesicles that can be released by practically all cell types, and range in size from 30 nm to 150 nm. As the major marker of liquid biopsies, exosomes have great potential for disease diagnosis, therapy, and prognosis. However, their inherent heterogeneity, the complexity of biological fluids, and the presence of nanoscale contaminants make the isolation of exosomes a great challenge. Traditional isolation methods of exosomes are cumbersome and challenging with complex and time-consuming operations. In recent years, the emergence of microfluidic chips, nanolithography, electro-deposition, and other technologies has promoted the combination and innovation of the isolation methods. The application of these methods has brought very considerable benefits to the isolation of exosomes such as ultra-fast, portable integration, and low loss. There are significant functional improvements in isolation yield, isolation purity, and clinical applications. In this review, a series of methods for the isolation of exosomes are summarized, with emphasis on the emerging methods, and in-depth comparison and analysis of each method are provided, including their principles, merits, and demerits.
Collapse
Affiliation(s)
| | | | | | | | - Liu Liu
- *Correspondence: Zuojun Shen, ; Liu Liu,
| | | |
Collapse
|
19
|
Du X, Wei H, Zhang B, Wang B, Li Z, Pang LK, Zhao R, Yao W. Molecular mechanisms of osteosarcoma metastasis and possible treatment opportunities. Front Oncol 2023; 13:1117867. [PMID: 37197432 PMCID: PMC10183593 DOI: 10.3389/fonc.2023.1117867] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 04/18/2023] [Indexed: 05/19/2023] Open
Abstract
In osteosarcoma patients, metastasis of the primary cancer is the leading cause of death. At present, management options to prevent metastasis are limited and non-curative. In this study, we review the current state of knowledge on the molecular mechanisms of metastasis and discuss promising new therapies to combat osteosarcoma metastasis. Genomic and epigenomic changes, metabolic reprogramming, transcription factors, dysregulation of physiologic pathways, and alterations to the tumor microenvironment are some of the changes reportedly involved in the regulation of osteosarcoma metastasis. Key factors within the tumor microenvironment include infiltrating lymphocytes, macrophages, cancer-associated fibroblasts, platelets, and extracellular components such as vesicles, proteins, and other secreted molecules. We conclude by discussing potential osteosarcoma-limiting agents and their clinical studies.
Collapse
Affiliation(s)
- Xinhui Du
- Bone Soft Tissue Department, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Key Laboratory for Digital Assessment of Spinal-Pelvic Tumor and Surgical Aid Tools Design (Zhengzhou), Zhengzhou, Henan, China
- Key Laboratory for Perioperative Digital Assessment of Bone Tumors (Henan), Zhengzhou, Henan, China
- *Correspondence: Xinhui Du,
| | - Hua Wei
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Boya Zhang
- Bone Soft Tissue Department, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Key Laboratory for Digital Assessment of Spinal-Pelvic Tumor and Surgical Aid Tools Design (Zhengzhou), Zhengzhou, Henan, China
- Key Laboratory for Perioperative Digital Assessment of Bone Tumors (Henan), Zhengzhou, Henan, China
| | - Bangmin Wang
- Bone Soft Tissue Department, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Key Laboratory for Digital Assessment of Spinal-Pelvic Tumor and Surgical Aid Tools Design (Zhengzhou), Zhengzhou, Henan, China
- Key Laboratory for Perioperative Digital Assessment of Bone Tumors (Henan), Zhengzhou, Henan, China
| | - Zhehuang Li
- Bone Soft Tissue Department, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Key Laboratory for Digital Assessment of Spinal-Pelvic Tumor and Surgical Aid Tools Design (Zhengzhou), Zhengzhou, Henan, China
- Key Laboratory for Perioperative Digital Assessment of Bone Tumors (Henan), Zhengzhou, Henan, China
| | - Lon Kai Pang
- Baylor College of Medicine, Houston, TX, United States
| | - Ruiying Zhao
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Weitao Yao
- Bone Soft Tissue Department, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
- Key Laboratory for Digital Assessment of Spinal-Pelvic Tumor and Surgical Aid Tools Design (Zhengzhou), Zhengzhou, Henan, China
- Key Laboratory for Perioperative Digital Assessment of Bone Tumors (Henan), Zhengzhou, Henan, China
| |
Collapse
|
20
|
Wu Q, Fu S, Xiao H, Du J, Cheng F, Wan S, Zhu H, Li D, Peng F, Ding X, Wang L. Advances in Extracellular Vesicle Nanotechnology for Precision Theranostics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204814. [PMID: 36373730 PMCID: PMC9875626 DOI: 10.1002/advs.202204814] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/09/2022] [Indexed: 05/04/2023]
Abstract
Extracellular vesicles (EVs) have increasingly been recognized as important cell surrogates influencing many pathophysiological processes, including cellular homeostasis, cancer progression, neurologic disease, and infectious disease. These behaviors enable EVs broad application prospects for clinical application in disease diagnosis and treatment. Many studies suggest that EVs are superior to conventional synthetic carriers in terms of drug delivery and circulating biomarkers for early disease diagnosis, opening up new frontiers for modern theranostics. Despite these clinical potential, EVs containing diverse cellular components, such as nucleic acids, proteins, and metabolites are highly heterogeneous and small size. The limitation of preparatory, engineering and analytical technologies for EVs poses technical barriers to clinical translation. This article aims at present a critical overview of emerging technologies in EVs field for biomedical applications and challenges involved in their clinic translations. The current methods for isolation and identification of EVs are discussed. Additionally, engineering strategies developed to enhance scalable production and improved cargo loading as well as tumor targeting are presented. The superior clinical potential of EVs, particularly in terms of different cell origins and their application in the next generation of diagnostic and treatment platforms, are clarified.
Collapse
Affiliation(s)
- Qian Wu
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for BiosensorsInstitute of Advanced Materials (IAM)Nanjing University of Posts and TelecommunicationsNanjing210023China
| | - Siyuan Fu
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for BiosensorsInstitute of Advanced Materials (IAM)Nanjing University of Posts and TelecommunicationsNanjing210023China
| | - Hanyang Xiao
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for BiosensorsInstitute of Advanced Materials (IAM)Nanjing University of Posts and TelecommunicationsNanjing210023China
| | - Jiaxin Du
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for BiosensorsInstitute of Advanced Materials (IAM)Nanjing University of Posts and TelecommunicationsNanjing210023China
| | - Fang Cheng
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for BiosensorsInstitute of Advanced Materials (IAM)Nanjing University of Posts and TelecommunicationsNanjing210023China
| | - Shuangshuang Wan
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for BiosensorsInstitute of Advanced Materials (IAM)Nanjing University of Posts and TelecommunicationsNanjing210023China
| | - Houjuan Zhu
- A*STAR (Agency for ScienceTechnology and Research)Singapore138634Singapore
| | - Dan Li
- Department of DermatologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing210008China
| | - Fei Peng
- Wellman Center for PhotomedicineMassachusetts General HospitalHarvard Medical SchoolCharlestownMA02114USA
| | - Xianguang Ding
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for BiosensorsInstitute of Advanced Materials (IAM)Nanjing University of Posts and TelecommunicationsNanjing210023China
| | - Lianhui Wang
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for BiosensorsInstitute of Advanced Materials (IAM)Nanjing University of Posts and TelecommunicationsNanjing210023China
| |
Collapse
|
21
|
Mei R, Wan Z, Li Z, Wei M, Qin W, Yuan L, Liu L, Yang G. "All-in-One" Exosome Engineering Strategy for Effective Therapy of Familial Hypercholesterolemia. ACS APPLIED MATERIALS & INTERFACES 2022; 14:50626-50636. [PMID: 36342824 DOI: 10.1021/acsami.2c15785] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Exosomes serve as a promising therapeutic nanoplatform. However, the exosomes produced by donor cells are a heterogeneous group, with only a small portion having high therapeutic efficacy. Specific isolation of the subpopulation with high efficacy is important for lowering the dose and minimizing toxicity. In this study, we loaded target mRNA and displayed specific Flag in engineered exosomes simultaneously. Briefly, the donor cells were transfected with plasmid expressing a fusion protein Flag-TCS-PTGFRN-CTSL-MCP, namely, exosome sorter. During biogenesis, the RNA-binding motif MCP can specifically bind with MS2-containing RNA and sort the target RNA into the lumen of exosomes. Anti-Flag magnetic beads can capture and thus purify the engineered exosomes via recognition of the Flag on the surface of exosomes. After purification, the Flag could be cleaved by thrombin treatment while MCP can be separated from the fusion protein by CTSL autocleavage upon exosome acidification, minimizing the side effects and augmenting the therapeutic effects. By the proof-of-concept experiment, the exosome sorter-based "all-in-one" strategy was confirmed effective in both the encapsulation of therapeutic mRNA (Ldlr-MS2) into exosomes and the subsequent purification. The purified Ldlr-MS2-containing exosomes had much higher efficacy in alleviating atherosclerosis, in comparison with the bulk exosomes, confirming the advantage of the proposed "all-in-one" strategy.
Collapse
Affiliation(s)
- Ruiyan Mei
- Department of Hematology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, China
- Shaanxi Department of National Clinical Research Center for Hematological Diseases, Xi'an 710032, China
- Clinical Medical Research Center for Hematological Diseases of Shaanxi Province, Xi'an 710032, China
| | - Zhuo Wan
- Department of Hematology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, China
- Shaanxi Department of National Clinical Research Center for Hematological Diseases, Xi'an 710032, China
- Clinical Medical Research Center for Hematological Diseases of Shaanxi Province, Xi'an 710032, China
| | - Zhelong Li
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, China
| | - Mengying Wei
- The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, Shaanxi, China
| | - Weiwei Qin
- Department of Hematology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, China
- Shaanxi Department of National Clinical Research Center for Hematological Diseases, Xi'an 710032, China
- Clinical Medical Research Center for Hematological Diseases of Shaanxi Province, Xi'an 710032, China
| | - Lijun Yuan
- Department of Ultrasound Diagnostics, Tangdu Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, China
| | - Li Liu
- Department of Hematology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, China
- Shaanxi Department of National Clinical Research Center for Hematological Diseases, Xi'an 710032, China
- Clinical Medical Research Center for Hematological Diseases of Shaanxi Province, Xi'an 710032, China
| | - Guodong Yang
- The State Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an 710032, Shaanxi, China
| |
Collapse
|
22
|
Lak NSM, van der Kooi EJ, Enciso-Martinez A, Lozano-Andrés E, Otto C, Wauben MHM, Tytgat GAM. Extracellular Vesicles: A New Source of Biomarkers in Pediatric Solid Tumors? A Systematic Review. Front Oncol 2022; 12:887210. [PMID: 35686092 PMCID: PMC9173703 DOI: 10.3389/fonc.2022.887210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/12/2022] [Indexed: 12/12/2022] Open
Abstract
Virtually every cell in the body releases extracellular vesicles (EVs), the contents of which can provide a "fingerprint" of their cellular origin. EVs are present in all bodily fluids and can be obtained using minimally invasive techniques. Thus, EVs can provide a promising source of diagnostic, prognostic, and predictive biomarkers, particularly in the context of cancer. Despite advances using EVs as biomarkers in adult cancers, little is known regarding their use in pediatric cancers. In this review, we provide an overview of published clinical and in vitro studies in order to assess the potential of using EV-derived biomarkers in pediatric solid tumors. We performed a systematic literature search, which yielded studies regarding desmoplastic small round cell tumor, hepatoblastoma, neuroblastoma, osteosarcoma, and rhabdomyosarcoma. We then determined the extent to which the in vivo findings are supported by in vitro data, and vice versa. We also critically evaluated the clinical studies using the GRADE (Grading of Recommendations Assessment, Development, and Evaluation) system, and we evaluated the purification and characterization of EVs in both the in vivo and in vitro studies in accordance with MISEV guidelines, yielding EV-TRACK and PedEV scores. We found that several studies identified similar miRNAs in overlapping and distinct tumor entities, indicating the potential for EV-derived biomarkers. However, most studies regarding EV-based biomarkers in pediatric solid tumors lack a standardized system of reporting their EV purification and characterization methods, as well as validation in an independent cohort, which are needed in order to bring EV-based biomarkers to the clinic.
Collapse
Affiliation(s)
- Nathalie S M Lak
- Research Department, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Department of Experimental Immunohematology, Sanquin Research, Amsterdam, Netherlands
| | - Elvera J van der Kooi
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, Netherlands
| | | | - Estefanía Lozano-Andrés
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Cees Otto
- Medical Cell Biophysics Group, University of Twente, Enschede, Netherlands
| | - Marca H M Wauben
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Godelieve A M Tytgat
- Research Department, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands.,Department of Experimental Immunohematology, Sanquin Research, Amsterdam, Netherlands
| |
Collapse
|